Zinc Complexes as Anticancer Agents: Comparison
Please note this is a comparison between Version 1 by Carlo Santini and Version 3 by Marina Porchia.

The search for anticancer metal-based drugs alternative to platinum derivatives could not exclude zinc derivatives due to the importance of this metal for the correct functioning of the human body. Zinc, the second most abundant trace element in the human body, is one of the most important micro-elements essential for human physiology. Its ubiquity in thousands of proteins and enzymes is related to its chemical features, in particular, its lack of redox activity and its ability to support different coordination geometries and to promote fast ligands exchange. Analogously to other trace elements, the impairment of its homeostasis can lead to various diseases and in some cases can be also related to cancer development. However, zinc complexes generally exert lower toxicity in comparison to other metal-based drugs and many zinc derivatives have been proposed as antitumor agents. Among them zinc complexes  comprising  N-donor ligands have been surveyed and analyzed. 

  • Zinc(II)
  • Zinc(II) complexes
  • N-donor ligands
  • metal-based drugs
  • medicinal chemistry
  • antitumor agents

Note: The following contents are extract from your paper. The entry will be online only after author check and submit it.

1. Introduction

Zinc is among the few transition metals, namely Mn, Fe, Co, Cu, Zn and Mo, which, together with the first and second series metals Na, K, Mg and Ca, are essential for human physiology. In the human body, zinc, after iron, is the second most abundant trace element. About 3 g of zinc, mostly localized in testicles, muscles, liver, and brain, are present in an average adult provided by a daily intake of 8–11 mg.[1] At the physiological concentration, zinc is crucial for increasing cell survival and protecting tissues against damages. Zinc concentration (about 0.6 mM) is regulated by specific homeostasis and, similarly to the other micro-elements, either a deficiency or an overload can lead to toxic effects to the organism.[2][3][4][5] Zinc deficiency can be related to inadequate zinc intake due to nutritional or absorption problems, ageing (several data showed that 35–45% of adults over 60 have a Zn intake below the required estimated average), Zn losses from the body or deregulation of zinc homeostasis. Zinc deficiency can depress immune function as Zn plays a crucial role in the immune system through cellular proliferation and RNA and DNA synthesis and is necessary for T-lymphocyte development. It also can determine other effects including growth retardation, impotence, and hypogonadism. Many symptoms due to Zn deficiency are not specific and can be related to other health conditions so that the diagnosis is not always straightforward. Zinc excess, however, is less frequent and most often occurs via excess supplementation. Most toxic effects due to a chronic high Zn intake (e.g., myeloneuropathy) are mainly related to the inhibition of copper absorption, and hence are secondary to a zinc-induced copper deficiency.[6]

The vital importance of zinc can be easily understood considering that this metal is present in more than 3000 human proteins including nucleic acid binding proteins; is involved in the catalytic activity of thousands of enzymes; plays a role in DNA synthesis, protein synthesis and immune functions.[7][8] The binding of Zn2+ with catalytic and/or structural sites of a large number of proteins is a key-factor in determining their conformations.[2] All in all, zinc is essential for virtually all cellular functions and also for the growth and development of all forms of life, not only human.[9] The majority of Zn in the human body (95%) is intracellular and the lack of specialized zinc storage systems makes a suitable daily intake necessary for maintaining a steady concentration in the organism. In biological systems the concentration of free Zn2+ ions is extremely low (pM-nM), i.e., it is not a relevant pool for trafficking, transport and cellular actions of zinc, so that these processes occur by a direct exchange of the metal from donor to acceptor Zn ligands.[10] Specific Zn transporters (ZIP and ZnT proteins) regulate Zn homeostasis and control its efflux via plasma membranes when the concentration of intracellular Zn is too high or when it must be transferred to other organs. But, whereas zinc coordination and its role in proteins and enzymes have been clarified and extensively reviewed, further studies are still necessary to completely explain the mechanisms of the exchange processes between intra- and extra-cellular space.[11]

The importance of zinc in biological systems is definitively related to its unique chemical features: Zn2+ is redox inactive, is a strong Lewis acid, has a d10 configuration, is diamagnetic, can support a variable coordination geometry and is prone to a fast exchange of ligands. Its electron affinity resembles that of copper or nickel, but the lack of redox activity of divalent zinc ion, differently from copper or iron, eliminates any chances of free radical reactions and makes it crucial for the body’s antioxidant protection system.

The Zn2+ d10 configuration, and the consequent absence of d-d transition, could be seen as a limit for the spectroscopic characterization of Zn derivatives, together with their diamagnetism and white colour, but on the other hand, the absence of ligand field stabilization can guarantee highly flexible coordination geometry determined only by the charge and steric hindrance of the ligands.[12][13] In biological systems zinc can be tetra-, penta-, or hexacoordinated to N, O or S donor atoms comprised in histidine, glutamate/aspartate, and cysteine residues, or to water molecules with a tetrahedral, pyramidal, or octahedral coordination geometry. In proteins, the most frequent geometry is tetrahedral, with few examples of distorted trigonal bipyramidal. In proteins, also multiple zinc clusters, comprising from two to four metal ions, can be found in the metal intrasphere binding geometry.

In addition to its physiological role, zinc can have beneficial therapeutic and preventive effects on infectious diseases and, compared to other metal-based drugs, Zn(II) complexes generally exert lower toxicity and have fewer side effects. An example of a worldwide commercial Zn-derivative is pyrithione zinc, first described in 1930 and used as a topical antimicrobial to treat fungal or bacterial infections of skin and hair. In the years different classes of zinc coordination complexes have shown a good potential in different applications, among which as radioprotective agents,[14] tumor photosensitizer,[15] antidiabetic,[16][17][18] anticonvulsant,[19] anti-inflammatory,[20] antimicrobial,[21][22][23][24][25][26] antioxidant,[27][28] antiproliferative/antitumor,[29][30][31] anti-Alzheimer’s disease[32] and in several neglected diseases.[33]

On the other hand, deregulation of zinc homeostasis can determine cell apoptosis and hence trigger cancer progress.[34] The relationship between zinc deficiency and cancer has been recognized in human, animal, and cell culture studies[35][36] and zinc-containing metalloenzymes have been identified as alternative targets for metal-based anticancer agents.[37] Zinc deficiency causes oxidative DNA damage,[38][39] and chromosome breaks have been reported in zinc-deficient diet-fed animals [40] In addition, zinc is useful in reducing cardio and hepatotoxicity caused by some anticancer drugs.[41]

The relationship between Zn deficiency and prostate cancer has been deeply analysed,[42] as well as the effect that Zn imbalance can have on the genesis and development of different forms of leukaemia.[43]

Examples of the detrimental effects of both excess or depletion of Zn in tumoral pathologies have been faced with opposite approaches: on one hand, a chelation therapy approach based on depletion of excess cellular Zn by the use of suitable chelating ligands,[44][45] on the other hand, the use of ionophore systems such as clioquinol.[46]

Another approach consists in using zinc complexes as metal-based antitumor drugs. This approach is very promising due to the fact that (1) having a specific homeostasis zinc metal ion could be better managed by human physiology and cause fewer side effects in comparison to non-essential metal-based compounds;[47][48] (2) zinc is significantly non-toxic even at higher doses than other metals (Fe, Cu, Hg, etc.), with obvious advantages for bio-compatibility;[47][48][49][50][51] (3) Zn(II) complexes probably have targets and mechanisms of action different from the classical platinum-based drugs;[52][53][54][55] (4) zinc is one of the most studied metals in the coordination of photosensitive systems for Photo Dynamic Therapy (PDT),[56][57][58] and (5) due to their ability to assist Lewis activation, nucleophile formation and rapid ligand exchange, zinc compounds can be employed as catalysts of hydrolytic reactions, such as hydrolysis and DNA cleavage, thus making anti-tumor activity possible.[59][60] Recent studies have confirmed the above assumptions showing that Zn(II) derivatives could be potential anticancer agents with low toxicity in vivo, low side effects and probably different cellular targets and modes of action when compared with classical metal-based drugs.[53][55][61][62][63][64][65][66]

2. Nitrogen Ligands in Zn Complexes

As reported in the Introduction, Zn(II) has a very versatile chemistry. It can adopt a range of coordination numbers giving rise to different geometry, even though especially in solution octahedral stereochemistries dominate. Zinc can coordinate various donor atoms, especially the first-row donor atoms oxygen or nitrogen rather than second-row sulphur or phosphorus, according to its hard acid nature. Accordingly, N-donor ligands are almost the most representative category. Homoleptic and mixed-ligand complexes have been reported and, due to the variety of accessible arrangements, a great assortment of frameworks (from mono- to hexadentate chelates) have been employed.

The sOurveyed complexes were organized on the basis of classification is based on the ligand nature according to the following classification:

  1. Quinoline and Diimine Systems   and  2,2'-Bipyridine and 1,10-Phenanthroline Systems  (24 Zn Complexes)
  2. Terpyridine and Pyridine-Based Systems  (23 Zn complexes)
  3. Imidazoles and Analogous Imidazole-Based Systems  (36 Zn complexes)
  4. Schiff Base Systems (9 Zn complexes)

. Planar aromatic quinoline, 2,2′-bipyridine and 1,10-phenanthroline ligands have often been the ligands of choice for medicinal chemists, due to their DNA intercalation properties and often to their intrinsic toxicity, which could enhance the metal effect. On the other hand, it’s known that several diimines have low specificity for tumor cell lines and can be genotoxic.[67] Here we report on 24 Zn complexes with these ligands (cap. 3 and 4). Terpyridine metal complexes are able to intercalate into DNA showing inhibitory effects on tumor cells and possess photoluminescence properties.[68][69][70][71] Here we report on 23 Zn complexes with terpyridine and pyridine-based systems (cap. 5). Imidazolyl derivatives are among the most utilized N-donor ligands due to their excellent coordination ability,[72][73] different hapticity and possibility to be derivatized or conjugated to active moieties. Within this class, benzoimidazolyl derivatives are the most representative (22 out of 36 Zn complexes, cap. 6), mainly thanks to the accessibility of phenyl ring substitution, which in turn allows SAR determination for different families of Zn complexes. Schiff bases are generally one of the most representative classes of ligands, mainly due to their easy way of synthesis. N-donor Schiff bases have been surveyed on the basis of their different hapticity (9 Zn complexes, cap. 7), whereas some examples of N,O-coordination are reported in the miscellanea (10 Zn complexes, cap. 8).

An important ther application of Zn derivatives in medicinal chemistry is Photo Dynamic Therapy and, among many metals utilized in the coordination of photosensitive systems, such as phthalocyanines, zinc is one of the most studied. Zinc complexes with photo-activable N-donor ligands, such as porphyrins and phthalocyanines, used in PDT, have been extensively reviewed in the last years [56,57] and are not treated in this surveypaper. Zinc-phthalocyanine complexes generally show low toxicity, high chemical and photochemical stability [56]. Anyway, low dark cytotoxicity is generally a prerequisite for photosensitizers in biological applications, even though chemotoxicity is sometimes associated to some Zn derivatives. The phototoxicity of the reported complexes is generally very high (IC50 values in micro-nanomolar range) and cannot be compared to the toxicity showed by the other families of zinc complexes surveyed in this paper, as the mechanism of action is not relied upon a biological involvement of the metal.

Considering the antiproliferative activity of the reviewed complexes, we found out that 43 out of 102 derivatives exhibited an antitumoral activity with IC50 values ≤10 micromolar against one or more cancer cell lines, reaching in some cases the nanomolar range. In addition to the complexes showing micromolar or sub-micromolar anticancer activity, 10 compounds exhibited a noticeable antitumor activity with  IC50 values of 10–20 μM, whereas the remaining ones showed a moderate/low activity.  Considering that antiproliferative activity in vitro is not predictive of an activity in vivo, it would be important to have more data of in vivo experiments with the most promising candidates to effectively evaluate the potential of Zn-based anticancer agents, as only few examples of in vivo studies have been reported. Looking critically at these data, without making any considerations on action mechanisms and proposed targets, some general considerations can be made trying to find a correlation between structure and activity. As far as the chemical structure is concerned, among the different geometries which Zn can adopt, hexa- and penta-coordination are by far the most common situations for active compounds, differently from zinc proteins where tetrahedral coordination, frequently slightly distorted, is the preferred geometry. The hapticity of the ligands is not decisive for the activity of the final complex, whereas the frequent presence of water molecules in the coordination sphere can allow an easy exchange with biological substrates. The use of active ligands does not always determine an increase of cytotoxicity upon coordination. From studies carried on with analogous complexes of different bivalent metal (such as Ni(II), Cu(II), Co(II), Fe(II), Mn(II)), it very often came out that zinc derivatives were less active, suggesting a minor effect of the metal compared to other metals. It seems that, excluding the use of hypotoxic Zn as a carrier of photoactive species for PDT or of active ligands, the antitumor efficacy of Zn-coordination complexes is not so appealing in comparison to other metal-based derivatives. The concentration of Zn in cells is probably so (relatively) high, that small variations do not induce an antiproliferative action of the metal unless specific mechanisms are involved. Probably an interesting application could be the use the low active, but at the same time low toxic zinc derivatives in combination with other chemotherapeutic agents to reduce their side effects.

Anyway this analysis has to be extended to all the classes of zinc coordination complexes, not only those with N-donor ligands, to confirm the above general considerations.

 

 

References

  1. Zinc . National Institutes of Health. Retrieved 2020-12-19
  2. Crichton, R.. Chapter 12: Zinc – Lewis Acid and Gene Regulator. In Biological Inorganic Chemistry (Second Edition); Crichton, R., Eds.; Elsevier: Amsterdam, 2012; pp. 229-247.
  3. Crichton, R.. Chapter 22: Metals in Medicine and Metals as Drugs. In Biological Inorganic Chemistry (Second Edition).; Crichton, R., Eds.; Elsevier: Amsterdam, 2012; pp. 415-432.
  4. Lydia A. Finney; Transition Metal Speciation in the Cell: Insights from the Chemistry of Metal Ion Receptors. Science 2003, 300, 931-936, 10.1126/science.1085049.
  5. Iztok Turel; Jakob Kljun; Interactions of metal ions with DNA, its constituents and derivatives, which may be relevant for anticancer research.. Current Topics in Medicinal Chemistry 2011, 11, 2661-2687, 10.2174/156802611798040787.
  6. Vineeth Tatineni; Julie Y. An; Matthew R. Leffew; Sameer A. Mahesh; Anemia from A to zinc: Hypocupremia in the setting of gastric bypass and zinc excess. Clinical Case Reports 2020, 8, 745-750, 10.1002/ccr3.2741.
  7. Hajo Haase; Lothar Rink; Multiple impacts of zinc on immune function. Metallomics 2014, 6, 1175-1180, 10.1039/c3mt00353a.
  8. Vladimir M. Kolenko; Ervin Teper; Alexander Kutikov; Robert G Uzzo; Zinc and zinc transporters in prostate carcinogenesis. Nature Reviews Urology 2013, 10, 219-226, 10.1038/nrurol.2013.43.
  9. B. L. Vallee; K. H. Falchuk; The biochemical basis of zinc physiology. Physiological Reviews 1993, 73, 79-118, 10.1152/physrev.1993.73.1.79.
  10. Leslie C. Costello; Catherine Fenselau; Renty B. Franklin; Evidence for operation of the direct zinc ligand exchange mechanism for trafficking, transport, and reactivity of zinc in mammalian cells. Journal of Inorganic Biochemistry 2011, 105, 589-599, 10.1016/j.jinorgbio.2011.02.002.
  11. Artur Krężel; Wolfgang Maret; The biological inorganic chemistry of zinc ions. Archives of Biochemistry and Biophysics 2016, 611, 3-19, 10.1016/j.abb.2016.04.010.
  12. C. Pettinari; A. Lorenzotti; M. Pellei; Carlo Santini; Zinc(II), cadmium(II) and mercury(II) derivatives of bis(4-halopyrazol-1-yl)alkanes: synthesis, spectroscopic characterization and behaviour in solution. Polyhedron 1997, 16, 3435-3445, 10.1016/s0277-5387(97)00089-2.
  13. Suman Adhikari; Tirtha Bhattacharjee; Raymond J. Butcher; Marina Porchia; Michele De Franco; Cristina Marzano; Valentina Gandin; Francesco Tisato; Synthesis and characterization of mixed-ligand Zn(II) and Cu(II) complexes including polyamines and dicyano-dithiolate(2-): In vitro cytotoxic activity of Cu(II) compounds. Inorganica Chimica Acta 2019, 498, 119098, 10.1016/j.ica.2019.119098.
  14. Saeed Emami; Hossein Asgarian-Omran; Seyed Mohammad Taghdisi; Shahram Akhlaghpoor; Kojic acid and its manganese and zinc complexes as potential radioprotective agents. Bioorganic & Medicinal Chemistry Letters 2007, 17, 45-48, 10.1016/j.bmcl.2006.09.097.
  15. Zhou Jiang; Jingwei Shao; Tingting Yang; Jian Wang; Lee Jia; Pharmaceutical development, composition and quantitative analysis of phthalocyanine as the photosensitizer for cancer photodynamic therapy. Journal of Pharmaceutical and Biomedical Analysis 2014, 87, 98-104, 10.1016/j.jpba.2013.05.014.
  16. Akihiro Nakayama; Makoto Hiromura; Yusuke Adachi; Hiromu Sakurai; Molecular mechanism of antidiabetic zinc–allixin complexes: regulations of glucose utilization and lipid metabolism. JBIC Journal of Biological Inorganic Chemistry 2008, 13, 675-684, 10.1007/s00775-008-0352-0.
  17. Hiromu Sakurai; Yutaka Yoshikawa; Hiroyuki Yasui; Current state for the development of metallopharmaceutics and anti-diabetic metal complexes. Chemical Society Reviews 2008, 37, 2383-2392, 10.1039/b710347f.
  18. Hiromu Sakurai; Yoshitane Kojima; Yutaka Yoshikawa; Kenji Kawabe; Hiroyuki Yasui; Antidiabetic vanadium(IV) and zinc(II) complexes. Coordination Chemistry Reviews 2002, 226, 187-198, 10.1016/s0010-8545(01)00447-7.
  19. Jean D'angelo; Georges Morgant; Nour Eddine Ghermani; Didier Desmaële; Bernard Fraisse; François Bonhomme; Emma Dichi; Mehrez Sghaier; Yanling Li; Yves Journaux; et al.John R.J Sorenson Crystal structures and physico-chemical properties of Zn(II) and Co(II) tetraaqua(3-nitro-4-hydroxybenzoato) complexes: Their anticonvulsant activities as well as related (5-nitrosalicylato)–metal complexes. Polyhedron 2008, 27, 537-546, 10.1016/j.poly.2007.10.006.
  20. Qingdi Zhou; Trevor W. Hambley; Brendan J. Kennedy; Peter A. Lay; Peter Turner; Barry Warwick; John R. Biffin; Hubertus L. Regtop; Syntheses and Characterization of Anti-inflammatory Dinuclear and Mononuclear Zinc Indomethacin Complexes. Crystal Structures of [Zn2(Indomethacin)4(L)2] (L =N,N-Dimethylacetamide, Pyridine, 1-Methyl-2-pyrrolidinone) and [Zn(Indomethacin)2(L1)2] (L1= Ethanol, Methanol). Inorganic Chemistry 2000, 39, 3742-3748, 10.1021/ic991477i.
  21. Noriko Chikaraishi Kasuga; Kiyoshi Sekino; Motoki Ishikawa; Ayano Honda; Masaki Yokoyama; Saori Nakano; Nobuhiro Shimada; Chisa Koumo; Kenji Nomiya; Synthesis, structural characterization and antimicrobial activities of 12 zinc(II) complexes with four thiosemicarbazone and two semicarbazone ligands. Journal of Inorganic Biochemistry 2003, 96, 298-310, 10.1016/s0162-0134(03)00156-9.
  22. Ze-Quan Li; Feng-Jing Wu; Yun Gong; Changwen Hu; Yun-Huai Zhang; Meng-Yu Gan; Synthesis, Characterization and Activity againstStaphylococcus of Metal(II)-Gatifloxacin Complexes. Chinese Journal of Chemistry 2007, 25, 1809-1814, 10.1002/cjoc.200790334.
  23. Zhen-Feng Chen; Ren-Gen Xiong; Jing Zhang; Xue-Tai Chen; Zi-Ling Xue; Xiao-Zeng You; 2D molecular square grid with strong blue fluorescent emission: a complex of norfloxacin with zinc(II).. Inorganic Chemistry 2001, 40, 4075-4077, 10.1021/ic001470x.
  24. M. Pilar López-Gresa; R Ortiz; L Perelló; J Latorre; M Liu-González; S Garcı́a-Granda; M Pérez-Priede; E Cantón; Interactions of metal ions with two quinolone antimicrobial agents (cinoxacin and ciprofloxacin). Journal of Inorganic Biochemistry 2002, 92, 65-74, 10.1016/s0162-0134(02)00487-7.
  25. Dong-Rong Xiao; En-Bo Wang; Hai-Yan An; Zhong-Min Su; Yangguang Li; Lei Gao; Chun-Yan Sun; Lin Xu; Rationally Designed, Polymeric, Extended Metal-Ciprofloxacin Complexes. Chemistry - A European Journal 2005, 11, 6673-6686, 10.1002/chem.200500548.
  26. Alketa Tarushi; Kostas Lafazanis; Jakob Kljun; Iztok Turel; Anastasia A. Pantazaki; George Psomas; Dimitris P. Kessissoglou; First- and second-generation quinolone antibacterial drugs interacting with zinc(II): Structure and biological perspectives. Journal of Inorganic Biochemistry 2013, 121, 53-65, 10.1016/j.jinorgbio.2012.12.009.
  27. Alketa Tarushi; Zoi Karaflou; Jakob Kljun; Iztok Turel; George Psomas; Athanasios N. Papadopoulos; Dimitris P. Kessissoglou; Antioxidant capacity and DNA-interaction studies of zinc complexes with a non-steroidal anti-inflammatory drug, mefenamic acid. Journal of Inorganic Biochemistry 2013, 128, 85-96, 10.1016/j.jinorgbio.2013.07.013.
  28. Alketa Tarushi; Xanthippi Totta; Athanasios Papadopoulos; Jakob Kljun; Iztok Turel; Dimitris P. Kessissoglou; George Psomas; Antioxidant activity and interaction with DNA and albumins of zinc–tolfenamato complexes. Crystal structure of [Zn(tolfenamato)2(2,2′-dipyridylketoneoxime)2]. European Journal of Medicinal Chemistry 2014, 74, 187-198, 10.1016/j.ejmech.2013.12.019.
  29. Dimitra Kovala-Demertzi; Paras Nath Yadav; Joanna Wiecek; Stauroula Skoulika; Tatiana Varadinova; Mavroudis A. Demertzis; Zinc(II) complexes derived from pyridine-2-carbaldehyde thiosemicarbazone and (1E)-1-pyridin-2-ylethan-1-one thiosemicarbazone. Synthesis, crystal structures and antiproliferative activity of zinc(II) complexes. Journal of Inorganic Biochemistry 2006, 100, 1558-1567, 10.1016/j.jinorgbio.2006.05.006.
  30. Marisa Belicchi Ferrari; Franco Bisceglie; Giorgio Pelosi; Pieralberto Tarasconi; Roberto Albertini; Silvana Pinelli; New methyl pyruvate thiosemicarbazones and their copper and zinc complexes: synthesis, characterization, X-ray structures and biological activity. Journal of Inorganic Biochemistry 2001, 87, 137-147, 10.1016/s0162-0134(01)00321-x.
  31. Zdeněk Trávníček; Vladimír Kryštof; Michal Šipl; Zinc(II) complexes with potent cyclin-dependent kinase inhibitors derived from 6-benzylaminopurine: Synthesis, characterization, X-ray structures and biological activity. Journal of Inorganic Biochemistry 2006, 100, 214-225, 10.1016/j.jinorgbio.2005.07.006.
  32. Massimo Di Vaira; Carla Bazzicalupi; Pierluigi Orioli; Luigi Messori; Bruno Bruni; Paolo Zatta; Clioquinol, a Drug for Alzheimer's Disease Specifically Interfering with Brain Metal Metabolism: Structural Characterization of Its Zinc(II) and Copper(II) Complexes. Inorganic Chemistry 2004, 43, 3795-3797, 10.1021/ic0494051.
  33. Yih Ching Ong; Saonli Roy; Philip C Andrews; Gilles Gasser; Metal Compounds against Neglected Tropical Diseases. Chemical Reviews 2018, 119, 730-796, 10.1021/acs.chemrev.8b00338.
  34. Renty B. Franklin; Leslie C. Costello; The important role of the apoptotic effects of zinc in the development of cancers. Journal of Cellular Biochemistry 2009, 106, 750-757, 10.1002/jcb.22049.
  35. Alessandro Federico; P Iodice; A Del Río; M C Mellone; G Catalano; P Federico; Effects of selenium and zinc supplementation on nutritional status in patients with cancer of digestive tract. European Journal of Clinical Nutrition 2001, 55, 293-297, 10.1038/sj.ejcn.1601157.
  36. Ananda S. Prasad; Frances W.J. Beck; Timothy D. Doerr; Falah H. Shamsa; Hayward S. Penny; Steven C. Marks; Joseph Kaplan; Omer Kucuk; Robert H. Mathog; Nutritional and zinc status of head and neck cancer patients: an interpretive review.. Journal of the American College of Nutrition 1998, 17, 409-418, 10.1080/07315724.1998.10718787.
  37. Ruirong Ye; Caiping Tan; Bichun Chen; Rongtao Li; Zongwan Mao; Zinc-Containing Metalloenzymes: Inhibition by Metal-Based Anticancer Agents. Frontiers in Chemistry 2020, 8, 402, 10.3389/fchem.2020.00402.
  38. Patricia I Oteiza; Michael S Clegg; M.Paola Zago; Carl L Keen; Zinc deficiency induces oxidative stress and AP-1 activation in 3T3 cells. Free Radical Biology and Medicine 2000, 28, 1091-1099, 10.1016/s0891-5849(00)00200-8.
  39. Emily Ho; Zinc deficiency, DNA damage and cancer risk. The Journal of Nutritional Biochemistry 2004, 15, 572-578, 10.1016/j.jnutbio.2004.07.005.
  40. M S Golub; M E Gershwin; L S Hurley; A G Hendrickx; W Y Saito; Studies of marginal zinc deprivation in rhesus monkeys: Infant behavior. The American Journal of Clinical Nutrition 1985, 42, 1229-1239, 10.1093/ajcn/42.6.1229.
  41. Mamdouh M Ali; Eva Frei; Josef Straub; Andrea Breuer; Manfred Wiessler; Mamdouh M. Ali; Induction of metallothionein by zinc protects from daunorubicin toxicity in rats. Toxicology 2002, 179, 85-93, 10.1016/s0300-483x(02)00322-0.
  42. Leslie C Costello; Renty B. Franklin; A comprehensive review of the role of zinc in normal prostate function and metabolism; and its implications in prostate cancer. Archives of Biochemistry and Biophysics 2016, 611, 100-112, 10.1016/j.abb.2016.04.014.
  43. Alexey P. Orlov; Marina A. Orlova; Tatiana P. Trofimova; Stepan N. Kalmykov; Dmitry A. Kuznetsov; The role of zinc and its compounds in leukemia. JBIC Journal of Biological Inorganic Chemistry 2018, 23, 347-362, 10.1007/s00775-018-1545-9.
  44. Mohammad Hashemi; Saeid Ghavami; Mehdi Eshraghi; Evan P. Booy; Marek Jan Los; Cytotoxic effects of intra and extracellular zinc chelation on human breast cancer cells. European Journal of Pharmacology 2007, 557, 9-19, 10.1016/j.ejphar.2006.11.010.
  45. Muralidhar L. Hegde; P. Bharathi; Anitha Suram; Chitra Venugopal; Ramya Jagannathan; Pankaj Poddar; Pullabhatla Srinivas; Kumar Sambamurti; Kosagisharaf Jagannatha Rao; Janez Scancar; et al.Luigi MessoriLuigi ZeccaPaolo Zatta Challenges Associated with Metal Chelation Therapy in Alzheimer's Disease. Journal of Alzheimer's Disease 2009, 17, 457-468, 10.3233/jad-2009-1068.
  46. Renty B. Franklin; Jing Zou; Yao Zheng; Michael J. Naslund; Leslie C Costello; Zinc Ionophore (Clioquinol) Inhibition of Human ZIP1-Deficient Prostate Tumor Growth in the Mouse Ectopic Xenograft Model: A Zinc Approach for the Efficacious Treatment of Prostate Cancer. International Journal of Cancer and Clinical Research 2016, 3, 037.
  47. Sudipta Bhattacharyya; Amrita X. Sarkar; Suman Kr Dey; Arindam Mukherjee; Effect of glucosamine conjugation to zinc(II) complexes of a bis-pyrazole ligand: Syntheses, characterization and anticancer activity. Journal of Inorganic Biochemistry 2014, 140, 131-142, 10.1016/j.jinorgbio.2014.07.009.
  48. Ananda S. Prasad; Frances W. J. Beck; Diane C. Snell; Omer Kucuk; Zinc in Cancer Prevention. Nutrition and Cancer 2009, 61, 879-887, 10.1080/01635580903285122.
  49. Heloisa Beraldo; Dinorah Gambino; The Wide Pharmacological Versatility of Semicarbazones, Thiosemicarbazones and Their Metal Complexes. Mini-Reviews in Medicinal Chemistry 2004, 4, 31-39, 10.2174/1389557043487484.
  50. Shuang-Qing Zhang; Xue-Feng Yu; Hai-Bo Zhang; Ning Peng; Zhi-Xian Chen; Qian Cheng; Xiao-Li Zhang; Sui-Han Cheng; Yan Zhang; Comparison of the Oral Absorption, Distribution, Excretion, and Bioavailability of Zinc Sulfate, Zinc Gluconate, and Zinc-Enriched Yeast in Rats. Molecular Nutrition & Food Research 2018, 62, e1700981, 10.1002/mnfr.201700981.
  51. Sebastien A. Rider; S. J. Davies; Awadhesh N. Jha; R. Clough; J. W. Sweetman; Bioavailability of co-supplemented organic and inorganic zinc and selenium sources in a white fishmeal-based rainbow trout (Oncorhynchus mykiss) diet. Journal of Animal Physiology and Animal Nutrition 2009, 94, 99-110, 10.1111/j.1439-0396.2008.00888.x.
  52. Alessio Terenzi; Mirco Fanelli; Gianluca Ambrosi; Stefano Amatori; Vieri Fusi; Luca Giorgi; Vincenzo Turco Liveri; Giampaolo Barone; DNA binding and antiproliferative activity toward human carcinoma cells of copper(ii) and zinc(ii) complexes of a 2,5-diphenyl[1,3,4]oxadiazole derivative. Dalton Transactions 2012, 41, 4389-4395, 10.1039/c2dt11759b.
  53. Paola F. Liguori; Alessandra Valentini; Mariagrazia Palma; Anna Bellusci; Sergio Bernardini; Mauro Ghedini; Maria Luisa Panno; Riccardo Pettinari; Fabio Marchetti; Alessandra Crispini; et al.Daniela Pucci Non-classical anticancer agents: synthesis and biological evaluation of zinc(ii) heteroleptic complexes. Dalton Transactions 2010, 39, 4205-4212, 10.1039/b922101h.
  54. Qin Jiang; Jianhui Zhu; Yangmiao Zhang; Nan Xiao; Zijian Guo; DNA binding property, nuclease activity and cytotoxicity of Zn(II) complexes of terpyridine derivatives. BioMetals 2008, 22, 297-305, 10.1007/s10534-008-9166-3.
  55. Barbara Sanz Mendiguchia; Daniela Pucci; Teresa F. Mastropietro; Mauro Ghedini; Alessandra Crispini; Non-classical anticancer agents: on the way to water soluble zinc(ii) heteroleptic complexes. Dalton Transactions 2013, 42, 6768-6774, 10.1039/c3dt50367d.
  56. Leonor P. Roguin; Nicolás Chiarante; María C. García Vior; Julieta Marino; Zinc(II) phthalocyanines as photosensitizers for antitumor photodynamic therapy. The International Journal of Biochemistry & Cell Biology 2019, 114, 105575, 10.1016/j.biocel.2019.105575.
  57. Weronika Kuzyniak; Eugeny A. Ermilov; Devrim Atilla; Ayşe Gül Gürek; Bianca Nitzsche; Katja Derkow; Björn Hoffmann; Gustav Steinemann; Vefa Ahsen; Michael Hopfner; et al. Tetra-triethyleneoxysulfonyl substituted zinc phthalocyanine for photodynamic cancer therapy. Photodiagnosis and Photodynamic Therapy 2016, 13, 148-157, 10.1016/j.pdpdt.2015.07.001.
  58. Işik Didem Karagöz; Yusuf Yilmaz; Kayode Sanusi; Anticancer Activity Study and Density Functional/Time-Dependent Density Functional Theory (DFT/TD-DFT) Calculations of 2(3),9(10),16(17),23(24)-Tetrakis-(6-Methylpyridin-2-Yloxy)Phthalocyaninato Zn(II). Journal of Fluorescence 2020, 30, 1151-1160, 10.1007/s10895-020-02584-1.
  59. Jing Qian; Liping Wang; Wen Gu; Xin Liu; Jinlei Tian; Shiping Yan; Efficient double-strand cleavage of DNA mediated by Zn(ii)-based artificial nucleases. Dalton Transactions 2011, 40, 5617-5624, 10.1039/c0dt01659d.
  60. Elisa Boseggia; Maddalena Gatos; Lorena Lucatello; Fabrizio Mancin; Stefano Moro; Manlio Palumbo; Claudia Sissi; Paolo Tecilla; Umberto Tonellato; Giuseppe Zagotto; et al. Toward Efficient Zn(II)-Based Artificial Nucleases. Journal of the American Chemical Society 2004, 126, 4543-4549, 10.1021/ja039465q.
  61. Daniela Pucci; Alessandra Crispini; Bárbara Sanz Mendiguchía; Sante Pirillo; Mauro Ghedini; Sabrina Morelli; Loredana De Bartolo; Improving the bioactivity of Zn(ii)-curcumin based complexes. Dalton Transactions 2013, 42, 9679-9687, 10.1039/c3dt50513h.
  62. Sze Koon Lee; Kong Wai Tan; Seik Weng Ng; Zinc, copper and nickel derivatives of 2-[2-bromoethyliminomethyl]phenol as topoisomerase inhibitors exhibiting anti-proliferative and anti-metastatic properties. RSC Advances 2014, 4, 60280-60292, 10.1039/c4ra09256b.
  63. Zahra Jannesari; Hassan Hadadzadeh; Zahra Amirghofran; Jim Simpson; Taghi Khayamian; Batool Maleki; A mononuclear zinc(II) complex with piroxicam: Crystal structure, DNA- and BSA-binding studies; in vitro cell cytotoxicity and molecular modeling of oxicam complexes. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy 2015, 136, 1119-1133, 10.1016/j.saa.2014.09.136.
  64. Jun Tan; Bochu Wang; Liancai Zhu; DNA binding, cytotoxicity, apoptotic inducing activity, and molecular modeling study of quercetin zinc(II) complex. Bioorganic & Medicinal Chemistry 2009, 17, 614-620, 10.1016/j.bmc.2008.11.063.
  65. Ruchi Singh; Mohd. Afzal; Mehvash Zaki; Musheer Ahmad; Sartaj Tabassum; Parimal K. Bharadwaj; Synthesis, structure elucidation and DFT studies of a new coumarin-derived Zn(ii) complex: in vitro DNA/HSA binding profile and pBR322 cleavage pathway. RSC Adv. 2014, 4, 43504-43515, 10.1039/c4ra05637j.
  66. Chun-Yan Gao; Xin Qiao; Zhong-Ying Ma; Zhi-Gang Wang; Jing Lu; Jin-Lei Tian; Jing-Yuan Xu; Shi-Ping Yan; Synthesis, characterization, DNA binding and cleavage, BSA interaction and anticancer activity of dinuclear zinc complexes. Dalton Transactions 2012, 41, 12220-12232, 10.1039/c2dt31306e.
  67. Valentina Gandin; Marina Porchia; Francesco Tisato; Alessandro Zanella; E. Severin; Alessandro Dolmella; Cristina Marzano; Novel Mixed-Ligand Copper(I) Complexes: Role of Diimine Ligands on Cytotoxicity and Genotoxicity. Journal of Medicinal Chemistry 2013, 56, 7416-7430, 10.1021/jm400965m.
  68. Andreas Winter; Michael Gottschaldt; George R. Newkome; Ulrich S. Schubert; Terpyridines and their complexes with first row transition metal ions: cytotoxicity, nuclease activity and self-assembly of biomacromolecules.. Current Topics in Medicinal Chemistry 2012, 12, 158-175, 10.2174/156802612799078919.
  69. Elaine A. Medlycott; Garry S. Hanan; Designing tridentate ligands for ruthenium(ii) complexes with prolonged room temperature luminescence lifetimes. Chemical Society Reviews 2005, 34, 133-142, 10.1039/b316486c.
  70. Vaidyanathan Ganesan Vaidyanathan; Balachandran Unni Nair; Nucleobase Oxidation of DNA by (Terpyridyl)chromium(III) Derivatives. European Journal of Inorganic Chemistry 2004, 2004, 1840-1846, 10.1002/ejic.200300718.
  71. V. M. Manikandamathavan; T. Weyhermüller; R. P. Parameswari; M. Sathishkumar; V. Subramanian; Balachandran Unni Nair; DNA/protein interaction and cytotoxic activity of imidazole terpyridine derived Cu(ii)/Zn(ii) metal complexes. Dalton Transactions 2014, 43, 13018-13031, 10.1039/c4dt01378f.
  72. Maura Pellei; Claudio Pettinari; Augusto Cingolani; Andrea Lacche; On the interaction between imidazoles and zinc salts. The role of counterions and of substituents. Main Group Metal Chemistry 2000, 23, 673-682, 10.1515/mgmc.2000.23.11.673.
  73. Maura Pellei; Claudio Pettinari; On the interaction between imidazoles and cadmium salts. Comparison between cadmium and zinc complexes of imidazoles. Main Group Metal Chemistry 2001, 24, 43-52, 10.1515/mgmc.2001.24.1.43.
More