Skip to main content
Antimicrobial Agents and Chemotherapy logoLink to Antimicrobial Agents and Chemotherapy
. 2013 Nov;57(11):5747–5754. doi: 10.1128/AAC.02446-12

In Vitro Effects of Novel Ruthenium Complexes in Neospora caninum and Toxoplasma gondii Tachyzoites

Fabienne Barna a, Karim Debache a, Carsten A Vock b,, Tatiana Küster a, Andrew Hemphill a,
PMCID: PMC3811262  PMID: 23979747

Abstract

Upon the screening of 16 antiproliferative compounds against Toxoplasma gondii and Neospora caninum, two hydrolytically stable ruthenium complexes (compounds 16 and 18) exhibited 50% inhibitory concentrations of 18.7 and 41.1 nM (T. gondii) and 6.7 and 11.3 nM (N. caninum). To achieve parasiticidal activity with compound 16, long-term treatment (22 to 27 days at 80 to 160 nM) was required. Transmission electron microscopy demonstrated the rapid impact on and ultrastructural alterations in both parasites. These preliminary findings suggest that the potential of ruthenium-based compounds should thus be further exploited.

TEXT

Toxoplasma gondii and Neospora caninum are cyst-forming apicomplexan parasites that infect a wide range of hosts. In an immunocompetent host, infection with either parasite does not cause disease (13). N. caninum has emerged as one of the most important infectious causes of bovine abortion (46). In contrast, T. gondii causes toxoplasmosis in humans and many animal species, either in chronically infected individuals during a decrease in immunoreactivity or if a seronegative mother acquires a primary infection during pregnancy, leading to abortion or serious fetal abnormalities (79). Toxoplasmosis treatment is based on only a few chemotherapeutics with considerable adverse effects (10, 11). In Neospora-seropositive cattle, pregnancy and the associated immunomodulation are already sufficient to cause recrudescence, fetal damage, and abortion (26). Chemotherapy has been considered a promising option if effective drugs can be identified (12, 13). Several compounds were investigated in vitro (1416), but only a few were evaluated in small-animal models (14, 1724).

We have evaluated compounds originally synthesized as anticancer drugs. Currently used metal complexes (2531) exhibit considerable toxicity. This has stimulated the interest in other compounds with more acceptable toxicity, such as ruthenium complexes (3236). Effects of ruthenium compounds on some bacteria and parasites have been studied (3746). “Classical ruthenium complexes” contain heteroatom ligands (e.g., azole derivatives), and NAMI-A and KP-1019 have been evaluated in phase I clinical trials for cancer treatment (4749). Organometallic complexes are defined by at least one metal-carbon bond. The η6-arene ruthenium(II) phosphite complexes 5, 6, 12, and 15 to 18 were characterized earlier (50), while [Ru(η6-p-cymene)(bipyridine)Cl][Cl] 11 was synthesized as shown previously (51). Based on our experiences in the design of selective inhibitors of CYP11B2 (53) and CYP11B1 (54), the pyridine-based compounds 4, 7 to 10, and 14 were from a small in-house library of CYP enzyme inhibitors. 2,2′-Bipyridine 3 was obtained from Joachim W. Heinicke, Ernst Moritz Arndt University of Greifswald, Greifswald, Germany. The cytotoxic lipophilic imidazolium salt 1,3-bis(2,4,6-trimethylphenyl)imidazolium chloride 3 was synthesized as described previously (5456). The arylimidamide DB745 2 (23) was kindly provided by David Boykin, Georgia State University, Atlanta, GA. The chemical structures and molecular masses of the drugs are shown in Fig. 1.

Fig 1.

Fig 1

Structures and molecular masses (M) of the compounds investigated in this study. Note that compound 2 (DB745) was used as a positive control in the assessment of parasite toxicity (see Fig. 2) and was replaced with Triton X-100 in the assessment of HFF host cell toxicity (see Fig. 3).

Maintenance of human foreskin fibroblasts (HFF) and Vero (African green monkey) cells and viability assessments by alamarBlue cytotoxicity assays were performed as described previously (50). Transgenic β-galactosidase-expressing T. gondii (RH) and N. caninum (Nc-1) tachyzoites (kindly provided by David Sibley, Washington University, St. Louis, MO) were maintained by serial passage in Vero cells (23, 24). Investigation of the inhibitory potential of the compounds was done as previously described (23, 24, 57). In short, confluent HFF grown in flat-bottom 96-well plates were infected with T. gondii or N. caninum tachyzoites at 103 parasites per well. After 2 h, compounds 2 to 18 (1 μM for initial screening and 0.5 nM to 1 μM for 50% inhibitory concentration [IC50] determinations) were added, and after 72 h of cultivation, parasite proliferation was assessed by the addition of chlorophenol red–β-d-galactopyranoside (Roche Diagnostics, Rotkreuz, Switzerland) in phosphate-buffered saline. A570 was measured in a VersaMax 96-well multiplate reader (Bucher Biotec, Basel, Switzerland) at various time points (23, 24).

Initial screening at 1 μM revealed that only ruthenium-based compounds 16 and 18 completely inhibited the proliferation of both T. gondii and N. caninum (Fig. 2A and B), exhibiting dose-dependent effects (Fig. 2C and D). IC50s (Table 1) show that N. caninum was slightly more susceptible. Treatment with all of the other compounds resulted in increased parasite proliferation (Fig. 2A and B), most likely because of nonlethal metabolic stress. At a concentration of 1 μM, neither of the compounds caused excessive cytotoxicity in noninfected HFF (Fig. 3A; Table 1). Exposure of extracellular T. gondii tachyzoites to 250 nM compound 16 for 1 to 2 h resulted in a pronounced (>90%) reduction of parasite numbers, while compound 18 had no effect (Fig. 4A). Both compounds had a severe impact on N. caninum infectivity (Fig. 4B). Pretreatment of host cells prior to infection was also investigated. Confluent HFF treated with compound 16 or 18 were washed, infected with tachyzoites, and cultured for 72 h. T. gondii proliferation was not affected (Fig. 5A), but N. caninum tachyzoites were severely impaired (Fig. 5B). This indicated that these compounds were taken up by the host cells as described earlier for other ruthenium-based drugs (58) and for dicationic pentamidine derivatives such as DB750 (59) and DB745 2 (23, 24).

Fig 2.

Fig 2

Proliferation-inhibitory effects upon T. gondii and N. caninum tachyzoites. Compounds 2 to 18 were added to transgenic T. gondii (A) and N. caninum (B) tachyzoites expressing β-galactosidase at 2 h postinfection of HFF monolayers at a concentration of 1 μM. Parasite proliferation was assessed by measurement of β-galactosidase activity after 72 h. Results are presented as percentages of β-galactosidase activity relative to that of a control containing the appropriate amount of dimethyl sulfoxide (c = 100%). (C, D) Compounds 16 and 18 were further assessed in dose-response experiments with T. gondii (C) and N. caninum (D), and measurements were done as described above.

Table 1.

IC50s of compounds 16 and 18 in noninfected HFF and in N. caninum and T. gondii tachyzoites expressing β-galactosidase grown in HFF monolayersa

Compound IC50b for:
N. caninum T. gondii HFF
16 6.7 18.7 2.4
18 11.3 41.1 6.95
a

Parasite proliferation was assessed by measuring β-galactosidase activity (23, 24, 45), and HFF cell vitality was assessed by alamarBlue assay (38).

b

IC50s are given in nM for parasites and in μM for HFF.

Fig 3.

Fig 3

Cytotoxicity assessment of compounds 3 to 18 in HFF monolayers. (A) HFF were exposed to the drugs at a concentration of 1 μM for 72 h and viability measurements were done by alamarBlue assay. Results are presented as percentages of fluorescence measured relative to that of a control containing the appropriate amount of dimethyl sulfoxide (c = 100%). Note that compound 2 represents the positive cytotoxicity control (addition of 1% Triton X-100). In panel B, HFF were exposed to different concentrations (0 to 20 μM) of compound 16 or 18 for 72 h and measurements were done as described above.

Fig 4.

Fig 4

Effects of preincubation of extracellular tachyzoites. Extracellular T. gondii (A) and N. caninum (B) tachyzoites were exposed to compound 16 or 18 for 1 or 2 h and then added to HFF for 2 h. Subsequently, cultures were further maintained in the absence of the drugs for 72 h and proliferation was assessed by β-galactosidase activity measurement. Note that compound 16 affected both T. gondii and N. caninum extracellular tachyzoites, while compound 18 was active only against N. caninum. Results are presented as percentages of β-galactosidase activity relative to that of a control containing the appropriate amount of dimethyl sulfoxide (control = 100%).

Fig 5.

Fig 5

Effects of preincubation of HFF host cell monolayers prior to infection on proliferation of T. gondii and N. caninum tachyzoites. HFF monolayers were exposed to compounds 16 and 18 for 1, 3, or 6 h; washed; and infected with T. gondii (A) or N. caninum (B) tachyzoites. Proliferation of parasites was assessed after 72 h by measurement of β-galactosidase activity. T. gondii proliferation (A) was not affected, while proliferation of N. caninum tachyzoites was impaired severely by compound 16 pretreatment and partially also by compound 18 (B). Results are presented as percentages of β-galactosidase relative to a control containing the appropriate amount of dimethyl sulfoxide (control = 100%).

The parasitostatic and/or parasiticidal activities of compounds 16 and 18 were studied as described previously (23, 24). In short-term experiments (Table 2), confluent HFF were infected with T. gondii or N. caninum, and at 2 h postinfection, 100, 250, or 500 nM compound 16 or 18 was added for 72 h of incubation, after which time the drug-containing medium was replaced with normal medium. Microscopy showed that both compounds failed to eliminate all of the tachyzoites, but compound 16 was more effective than compound 18. The abilities of T. gondii and N. caninum cells to adapt to compounds 16 and 18 were explored by slowly increasing the drug concentrations (Table 3). Infected HFF were initially cultured in the presence of compound 18 (50 nM for T. gondii; 12 nM for N. caninum) and compound 16 (20 nM and 10 nM, respectively), and drug levels were increased by 10 to 30 nM, typically every 3 to 4 days. Microscopy again demonstrated the higher efficacy of compound 16 (Table 3).

Table 2.

Short-term treatment of N. caninum and T. gondii tachyzoites grown in HFF is not parasiticidala

Parasite and compound Concn (nM) Posttreatment culture time (days)
N. caninum
    Control 3
    16 100 8
250 9
500 10
    18 100 6
250 7
500 8
T. gondii
    Control 2
    16 100 2
250 6
500 7
    18 100 2
250 2
500 3
a

T25 tissue culture flasks containing confluent HFF monolayers were infected with 8 × 105 N. caninum or T. gondii tachyzoites. At 2 h postinfection, compound 16 or 18 was added and cultivation continued for 72 h in the presence of each compound at 100, 250, or 500 nM. The cultures were then washed with medium to remove the drugs and then incubated further in the absence of the compounds as indicated in Table 3. Posttreatment culture time indicates the numbers of days of culture in the absence of drugs until the reemergence of parasite proliferation was detected by light microscopy.

Table 3.

T. gondii and N. caninum tachyzoites can adapt to increasing concentrations of compound 18 but not 16a

Drug treatment duration (days) Drug concn (nM)
T. gondii
N. caninum
Compound 18 Compound 16 Compound 18 Compound 16
0 50 20 12 8
3 70 40 20 16
6 90 40 40 20
9 110 60 60 30
12 130 80 80 40
16 150 100 80 40
22 170 130 80 Medium
27 190 160 100 Medium
31 210 Medium 120 Medium
35 250 Medium 150 Medium
39 270 Medium 170 Medium
42 300 Medium 200 Medium
45 330 Medium 240 Medium
a

T25 tissue culture flasks containing confluent HFF monolayers were infected and cultured initially in the presence of compound 16 or 18 at the concentrations indicated on day 0. Proliferation of parasites was monitored daily by light microscopy. At the time points indicated, the medium was replaced with new medium containing a slightly elevated concentration of the respective compound, the same concentration, or no drug at all. Every 6 to 10 days, cultures were trypsinized and seeded onto fresh HFF monolayers. The experiment was terminated on day 45.

Inspection of drug-treated infected HFF by transmission electron microscopy (TEM) revealed distinct ultrastructural alterations in both parasites (Fig. 6 and 7). Untreated T. gondii (Fig. 6A and B) and N. caninum (Fig. 7A and B) form parasitophorous vacuoles containing proliferating tachyzoites. In cultures treated with compound 16, the drug rapidly induced alterations. Obviously metabolically impaired tachyzoites with numerous empty or lipid-containing inclusions with electron-dense granular or amorphous material were visible after 12 h (Fig. 6C). The nuclear membrane had a fuzzy appearance, and chromatin appeared to be preferentially located at the nuclear periphery. At 36 h, most T. gondii tachyzoites exhibited a completely disorganized cytoplasm, organelles were hardly discernible, and many parasites were embedded in a granular matrix (Fig. 6D). Similar alterations were evident in N. caninum tachyzoites treated with compound 16 (Fig. 7C to F), with effects being most pronounced at 36 h of treatment (Fig. 7E and F). While these observations indicated a critical metabolic impairment of parasites, the alterations observed do not really point toward a defined mode of action.

Fig 6.

Fig 6

Effects of compound 16 on T. gondii ultrastructure. HFF monolayers grown to confluence in T25 culture flasks were infected with T. gondii tachyzoites, and after 48 h, they were treated with 100 nM compound 16 for 12 and 36 h, respectively; untreated infected cultures served as a control. Specimens were then processed for TEM as previously described (46, 47) and were viewed on a Philips 400T transmission electron microscope operating at 80 kV. (A, B) Numerous tachyzoites enclosed in a parasitophorous vacuole near the vicinity of the host cell nucleus (hcnuc), surrounded by a parasitophorous vacuole membrane (pvm). (B) Actively dividing tachyzoite with conoid (con) and rhoptry (rop) organelles. At 12 h after treatment started, clear alterations were observed (C). The tachyzoite cytoplasm is largely vacuolized, with vacuoles containing lipid droplets (ld) or membranous and electron-dense material. The white arrows point toward electron-dense chromatin deposits along the nuclear periphery of tachyzoites (nuc = nucleus). At 36 h of drug treatment (D), tachyzoites appear completely altered, exhibiting a disorganized cytoplasmic morphology, and intracellular parasites are often embedded in an electron-dense granular matrix (large white arrow in panel D). Bars: A, 1.4 μm; B, 0.8 μm; C, 0.5 μm; D, 0.8 μm.

Fig 7.

Fig 7

Effects of compound 16 on N. caninum ultrastructure. HFF monolayers were infected with T. gondii tachyzoites, and after 48 h, they were treated with 100 nM compound 16. (A, B) Nontreated N. caninum tachyzoites within a parasitophorous vacuole with intact nucleus (nuc), mitochondrion (mito), and anteriorly located conoid (con), micronemes (mic) and rhoptries (rop). After 12 h of treatment (C, D), the first alterations appear, such as the formation of cytoplasmic vacuoles (white arrows), and at 36 h, most tachyzoites appear completely altered (E), often embedded in granular material within the cytoplasm of host cells, but extracellular tachyzoite residues are also found (F). Bars: A, 0.8 μm; B, 0.6 μm; C, 1 μm; D, 0.3 μm; E, 0.8 μm; F, 1 μm.

How compound 16 exerts its parasiticidal action remains unknown. Earlier studies indicated that ruthenium compounds interact with DNA (25, 27). However, more recent investigations showed that ruthenium compounds bind more strongly to proteins (60, 61) and potential targets in cancer cells were postulated, including cathepsin B, P-glycoprotein, and glutathione S-transferase P1 (62). Exploitation of the wealth of available knowledge about ruthenium compounds could represent a starting point for the development of drugs with antiparasitic properties (63).

Of the eight ruthenium complexes investigated, the set of phosphite complexes can be divided into hydrolytically labile (compounds 5, 6, and 12) and hydrolytically stable complexes (compounds 16 to 18). Only compounds 16 to 18 exerted antiparasitic effects. We assume that the hydrocarbon substituents around the ruthenium center form a lipophilic sphere that facilitates the uptake of the compounds, though they should not be able to penetrate membranes because of their ionic nature. For compound 16, an optimum arrangement and the formation of a ball-shaped sphere around the ruthenium center can be considered to be of importance. The smaller the surface of the molecule, the lower the chance of interaction with other molecules, e.g., in cell membranes. An additional aspect could be the impossibility of ligand exchange on the ruthenium center when almost perfect coverage is provided. The combination of bulky isopropyl groups on the phosphite ligand, tBu residues on the 1,3-diketonate moiety, and a sterically demanding η6-arene unit should effectively prevent nucleophilic attack on the metal center, leading to very good stability even in the presence of strong nucleophiles. Presumably, an attack on the phenyl moieties in compound 17 is easier (for example, by protonation following an SEAr mechanism), which leads to decomposition of the triarylphosphite ligand and subsequent loss of the stability of the whole complex. In conclusion, the combination of reduced surface area and best shielding of the ruthenium center against nucleophilic attacks might explain the superior antiparasitic activity of 16.

ACKNOWLEDGMENTS

We thank David Boykin (Georgia State University, Atlanta, GA) and Chad E. Stephens (Augusta State University, Augusta, GA) for providing DB745, and David Sibley (Washington University, St. Louis, MO) and Sabrina Sonda (University of Zürich) are gratefully acknowledged for β-galactosidase-expressing T. gondii and N. caninum, respectively. Many thanks to Thierry Monney Institute of Parasitology, University of Bern) for help with cell cultures and Norbert Müller and Joachim Müller for technical advice and critical reading of the manuscript. For precious help with the synthesis of the compound library, we are grateful to Nadine Lense, Pauline Böhme, and Christian Nachtigal. Regarding the synthetic facilities, we acknowledge support of Joachim W. Heinicke (Ernst Moritz Arndt University of Greifswald).

This work was financed through the National Science Foundation (grant 31-127374).

Footnotes

Published ahead of print 26 August 2013

REFERENCES

  • 1.Reid AJ, Vermont SJ, Cotton JA, Harris D, Hill-Cawthorne GA, Könen-Waisman S, Latham SM, Mourier T, Norton R, Quail MA, Sanders M, Shanmugam D, Sohal A, Wasmuth JD, Brunk B, Grigg ME, Howard JC, Parkinson J, Roos DS, Trees AJ, Berriman M, Pain A, Wastling JM. 2012. Comparative genomics of the apicomplexan parasites Toxoplasma gondii and Neospora caninum: coccidia differing in host range and transmission strategy. PLoS Pathog. 8:e1002567. 10.1371/journal.ppat.1002567 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Hemphill A, Vonlaufen N, Naguleswaran A. 2006. Cellular and immunological basis of the host-parasite relationship during infection with Neospora caninum. Parasitology 133:261–278 [DOI] [PubMed] [Google Scholar]
  • 3.Innes EA, Vermeulen AN. 2006. Vaccination as a control strategy against the coccidial parasites Eimeria, Toxoplasma and Neospora. Parasitology 133(Suppl):S145–S168 [DOI] [PubMed] [Google Scholar]
  • 4.Dubey JP, Schares G, Ortega-Mora LM. 2007. Epidemiology and control of neosporosis and Neospora caninum. Clin. Microbiol. Rev. 20:323–367 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Monney T, Debache K, Hemphill A. 2011. Vaccines against a major cause of abortion in cattle, Neospora caninum infection. Animals 1:306–325 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Reichel MP, Ayanegui-Alcérrecac M, Gondim LF, Ellis JT. 2013. What is the global economic impact of Neospora caninum in cattle—the billion dollar question. Int. J. Parasitol. 43:133–142 [DOI] [PubMed] [Google Scholar]
  • 7.Saadatnia G, Golkar M. 2012. A review on human toxoplasmosis. Scand. J. Infect. Dis. 44:805–814 [DOI] [PubMed] [Google Scholar]
  • 8.Kortagere S. 2012. Screening for small molecule inhibitors of Toxoplasma gondii. Expert Opin. Drug Discov. 7:1193–1206 [DOI] [PubMed] [Google Scholar]
  • 9.Röse I. 1997. Morphology and diagnostics of human toxoplasmosis. Gen. Diagn. Pathol. 142:257–270 [PubMed] [Google Scholar]
  • 10.Dedicoat M, Livesley N. 2006. Management of toxoplasmic encephalitis in HIV-infected adults (with an emphasis on resource-poor settings). Cochrane Database Syst. Rev. 19:CD005420. 10.1002/14651858.CD005420.pub2 [DOI] [PubMed] [Google Scholar]
  • 11.Khalili H, Soudbakhsh A, Talasaz AH. 2011. Severe hepatotoxicity and probable hepatorenal syndrome associated with sulfadiazine. Am. J. Health Syst. Pharm. 68:888–892 [DOI] [PubMed] [Google Scholar]
  • 12.Häsler B, Regula G, Stärk KD, Sager H, Gottstein B, Reist M. 2006. Financial analysis of various strategies for the control of Neospora caninum in dairy cattle in Switzerland. Prev. Vet. Med. 77:230–253 [DOI] [PubMed] [Google Scholar]
  • 13.Häsler B, Stärk KD, Sager H, Gottstein B, Reist M. 2006. Simulating the impact of four control strategies on the population dynamics of Neospora caninum infection in Swiss dairy cattle. Prev. Vet. Med. 77:254–283 [DOI] [PubMed] [Google Scholar]
  • 14.Lindsay DS, Dubey JP. 1990. Effects of sulfadiazine and amprolium on Neospora caninum (Protozoa: Apicomplexa) infections in mice. J. Parasitol. 76:177–179 [PubMed] [Google Scholar]
  • 15.Lindsay DS, Dubey JP. 1989. Evaluation of anti-coccidial drugs' inhibition of Neospora caninum development in cell cultures. J. Parasitol. 75:990–992 [PubMed] [Google Scholar]
  • 16.Lindsay DS, Rippey NS, Cole RA, Parsons LC, Dubey JP, Tidwell RR, Blagburn BL. 1994. Examination of the activities of 43 chemotherapeutic agents against Neospora caninum tachyzoites in cultured cells. Am. J. Vet. Res. 55:976–981 [PubMed] [Google Scholar]
  • 17.Ammann P, Waldvogel A, Breyer I, Esposito M, Müller N, Gottstein B. 2004. The role of B- and T-cell immunity in toltrazuril-treated C57BL/6 WT, microMT and nude mice experimentally infected with Neospora caninum. Parasitol. Res. 93:178–187 [DOI] [PubMed] [Google Scholar]
  • 18.Gottstein B, Eperon S, Dai WJ, Cannas A, Hemphill A, Greif G. 2001. Efficacy of toltrazuril and ponazuril against experimental Neospora caninum infection in mice. Parasitol. Res. 87:43–48 [DOI] [PubMed] [Google Scholar]
  • 19.Gottstein B, Razmi GR, Ammann P, Sager H, Müller N. 2005. Toltrazuril treatment to control diaplacental Neospora caninum transmission in experimentally infected pregnant mice. Parasitology 130:41–48 [DOI] [PubMed] [Google Scholar]
  • 20.Debache K, Hemphill A. 2012. Effects of miltefosine treatment in fibroblast cell cultures and in mice experimentally infected with Neospora caninum tachyzoites. Parasitology 139:934–944 [DOI] [PubMed] [Google Scholar]
  • 21.Wang MZ, Zhu X, Srivastava A, Liu Q, Sweat JM, Pandharkar T, Stephens CE, Riccio E, Parman T, Munde M, Mandal S, Madhubala R, Tidwell RR, Wilson WD, Boykin DW, Hall JE, Kyle DE, Werbovetz KA. 2010. Novel arylimidamides for treatment of visceral leishmaniasis. Antimicrob. Agents Chemother. 54:2507–2516 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Debache K, Guionaud C, Kropf C, Boykin D, Stephens CE, Hemphill A. 2011. Experimental treatment of Neospora caninum-infected mice with the arylimidamide DB750 and the thiazolide nitazoxanide. Exp. Parasitol. 129:95–100 [DOI] [PubMed] [Google Scholar]
  • 23.Schorer M, Debache K, Barna F, Monney T, Müller J, Boykin D, Stephens CE, Hemphill A. 2012. Di-cationic arylimidamides act against Neospora caninum tachyzoites by interference in membrane structure and nucleolar integrity and are active against challenge infection in mice. Int. J. Parasitol. Drugs Drug Resist. 2:109–120 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Kropf C, Debache K, Rampa C, Barna F, Schorer M, Stephens CE, Ismail MA, Boykin DW, Hemphill A. 2012. The adaptive potential of a survival artist: characterization of the in vitro interactions of Toxoplasma gondii tachyzoites with di-cationic compounds in human fibroblast cell cultures. Parasitology. 139:208–220 [DOI] [PubMed] [Google Scholar]
  • 25.Schwietert CW, McCue JP. 1999. Coordination compounds in medicinal chemistry. Coord. Chem. Rev. 184:67–89 [Google Scholar]
  • 26.Bruijnincx PCA, Sadler PJ. 2008. New trends for metal complexes with anticancer activity. Curr. Opin. Chem. Biol. 12:197–206 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Gasser G, Ott I, Metzler-Nolte N. 2011. Organometallic anticancer compounds. J. Med. Chem. 54:3–25 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Garbutcheon-Singh KB, Grant MP, Harper BW, Krause-Heuer AM, Manohar M, Orkey N, Aldrich-Wright JR. 2011. Transition metal based anticancer drugs. Curr. Top. Med. Chem. 11:521–542 [DOI] [PubMed] [Google Scholar]
  • 29.Ang WH, Casini A, Sava G, Dyson PJ. 2011. Organometallic ruthenium-based antitumor compounds with novel modes of action. J. Organomet. Chem. 696:989–998 [Google Scholar]
  • 30.Komeda S, Casini A. 2012. Next-generation anticancer metallodrugs. Curr. Top. Med. Chem. 12:219–235 [DOI] [PubMed] [Google Scholar]
  • 31.Rosenberg B, Vancamp L, Krigas T. 1965. Inhibition of cell division in Escherichia coli by electrolysis products from a platinum electrode. Nature 205:698–699 [DOI] [PubMed] [Google Scholar]
  • 32.Kostova I. 2006. Ruthenium complexes as anticancer agents. Curr. Med. Chem. 13:1085–1107 [DOI] [PubMed] [Google Scholar]
  • 33.Peacock AF, Sadler PJ. 2008. Medicinal organometallic chemistry: designing metal arene complexes as anticancer agents. Chem. Asian J. 3:1890–1899 [DOI] [PubMed] [Google Scholar]
  • 34.Hartinger CG, Dyson PJ. 2009. Bioorganometallic chemistry—from teaching paradigms to medicinal applications. Chem. Soc. Rev. 38:391–401 [DOI] [PubMed] [Google Scholar]
  • 35.Levina A, Mitra A, Lay PA. 2009. Recent developments in ruthenium anticancer drugs. Metallomics 1:458–470 [DOI] [PubMed] [Google Scholar]
  • 36.Bergamo A, Sava G. 2011. Ruthenium anticancer compounds: myths and realities of the emerging metal-based drugs. Dalton Trans. 40:7817–7823 [DOI] [PubMed] [Google Scholar]
  • 37.Beckford FA, Thessing J, Shaloski M, Mbarushimana PC, Brock A, Didion J, Woods J, Gonzalez-Sarrías A, Seeram NP. 2011. Synthesis and characterization of mixed-ligand diimine-piperonal thiosemicarbazone complexes of ruthenium(II): biophysical investigations and biological evaluation as anticancer and antibacterial agents. J. Mol. Struct. 992:39–47 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Donnici CL, Araújo MG, Oliveira HS, Moreira DR, Pereira VR, de Assis Souza M, de Castro MC, Leite AC. 2009. Ruthenium complexes endowed with potent anti-Trypanosoma cruzi activity: synthesis, biological characterization and structure-activity relationships. Bioorg. Med. Chem. 17:5038–5043 [DOI] [PubMed] [Google Scholar]
  • 39.Otero L, Aguirre G, Boiani L, Denicola A, Rigol C, Olea-Azar C, Maya JD, Morello A, González M, Gambino D, Cerecetto H. 2006. Nitrofurylsemicarbazone rhenium and ruthenium complexes as anti-trypanosomal agents. Eur. J. Med. Chem. 41:1231–1239 [DOI] [PubMed] [Google Scholar]
  • 40.Beckford F, Dourth D, Shaloski M, Didion J, Thessing J, Woods J, Crowell V, Gerasimchuk N, Gonzalez-Sarrias A, Seeram NP. 2011. Half-sandwich ruthenium arene complexes with thiosemicarbazones: synthesis and biological evaluation of [(η6-p-cymene)Ru(piperonal thiosemicarbazones)Cl]Cl complexes. J. Inorg. Biochem. 105:1019–1029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Sathya N, Raja G, Priya NP, Jayabalakrishnan C. 2010. Ruthenium(II) complexes incorporating tridentate Schiff base ligands: synthesis, spectroscopic, redox, catalytic and biological properties. Appl. Organomet. Chem. 24:366–373 [Google Scholar]
  • 42.Silva JJ, Guedes PM, Zottis A, Balliano TL, Silva FO, Lopes LG, Ellena J, Oliva G, Andricopulo AD, Franco DW, Silva JS. 2010. Novel ruthenium complexes as potential drugs for Chagas's disease: enzyme inhibition and in vitro/in vivo trypanocidal activity. Br. J. Pharmacol. 160:260–269 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Silva JJ, Pavanelli WR, Pereira JC, Silva JS, Franco DW. 2009. Experimental chemotherapy against Trypanosoma cruzi infection using ruthenium nitric oxide donors. Antimicrob. Agents Chemother. 53:4414–4421 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Biot C, Nosten F, Fraisse L, Ter-Minassian D, Khalife J, Dive D. 2011. The antimalarial ferroquine: from bench to clinic. Parasite 18:207–214 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Sannella AR, Casini A, Gabbiani C, Messori L, Bilia AR, Vincieri FF, Majori G, Severini C. 2008. New uses for old drugs. Auranofin, a clinically established antiarthritic metallodrug, exhibits potent antimalarial effects in vitro: mechanistic and pharmacological implications. FEBS Lett. 582:844–847 [DOI] [PubMed] [Google Scholar]
  • 46.Caroli A, Simeoni S, Lepore R, Tramontano A, Via A. 2012. Investigation of a potential mechanism for the inhibition of SmTGR by auranofin and its implications for Plasmodium falciparum inhibition. Biochem. Biophys. Res. Commun. 417:576–581 [DOI] [PubMed] [Google Scholar]
  • 47.Rademaker-Lakhai JM, Van den Bongard D, Pluim D, Beijnen JH, Schellens JH. 2004. A phase I and pharmacological study with imidazolium-trans-DMSO-imidazole-tetrachlororuthenate, a novel ruthenium anticancer agent. Clin. Cancer Res. 10:3717–3727 [DOI] [PubMed] [Google Scholar]
  • 48.Hartinger CG, Zorbas-Seifried S, Jakupec MA, Kynast B, Zorbas H, Keppler BK. 2006. From bench to bedside—preclinical and early clinical development of the anticancer agent indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (KP1019 or FFC14A). J. Inorg. Biochem. 100:891–904 [DOI] [PubMed] [Google Scholar]
  • 49.Lentz F, Drescher A, Lindauer A, Henke M, Hilger RA, Hartinger CG, Scheulen ME, Dittrich C, Keppler BK, Jaehde U. 2009. Pharmacokinetics of a novel anticancer ruthenium complex (KP1019, FFC14A) in a phase I dose-escalation study. Anticancer Drugs 20:97–103 [DOI] [PubMed] [Google Scholar]
  • 50.Küster T, Lense N, Barna F, Hemphill A, Kindermann MK, Heinicke JW, Vock CA. 2012. A new promising application for highly cytotoxic metal compounds: η6-Arene ruthenium(II) phosphite complexes for the treatment of alveolar echinococcosis. J. Med. Chem. 55:4178–4188 [DOI] [PubMed] [Google Scholar]
  • 51.Lalrempuia R, Kollipara MR, Carroll PJ, Yap GPA, Kreisel KA. 2005. Synthesis and characterization of cyano-bridged homo and hetero bimetallic complexes containing h5- and h6-cyclic hydrocarbons. J. Organomet. Chem. 690:3990–3996 [Google Scholar]
  • 52.Zimmer C, Hafner M, Zender M, Ammann D, Hartmann RW, Vock CA. 2011. N-(Pyridin-3-yl)benzamides as selective inhibitors of human aldosterone synthase (CYP11B2). Bioorg. Med. Chem. Lett. 21:186–190 [DOI] [PubMed] [Google Scholar]
  • 53.Hille UE, Zimmer C, Vock CA, Hartmann RW. 2011. First selective CYP11B1 inhibitors for the treatment of cortisol-dependent diseases. ACS Med. Chem. Lett. 2:2–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Zhang C, Ding Z, Suhaimi NAM, Kng AL, Zhang Y, Zhuo L. 2009. A class of imidazolium salts is antioxidative and anti-fibrotic in hepatic stellate cells. Free Radic. Res. 43:899–912 [DOI] [PubMed] [Google Scholar]
  • 55.Gopalan B, Ke Z, Zhang C, Kng Y, Suhaimi NAM, Riduan SN, Zhang Y, Zhuo L. 2011. Metal-free imidazolium salts inhibit the growth of hepatocellular carcinoma in a mouse model. Lab. Invest. 91:744–751 [DOI] [PubMed] [Google Scholar]
  • 56.Hintermann L. 2007. Expedient syntheses of the N-heterocyclic carbene precursor imidazolium salts IPr.HCl, IMes.HCl and IXy.HCl. Beilstein J. Org. Chem. 3:22. 10.1186/1860-5397-3-22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.McFadden DC, Seeber F, Boothroyd JC. 1997. Use of Toxoplasma gondii expressing beta-galactosidase for colorimetric assessment of drug activity in vitro. Antimicrob. Agents Chemother. 41:1849–1853 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Puckett CA, Barton JK. 2007. Methods to explore cellular uptake of ruthenium complexes J. Am. Chem. Soc. 129:46–47 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Leepin A, Stüdli A, Brun R, Stephens CE, Boykin DW, Hemphill A. 2008. Host cells participate in the in vitro effects of novel diamidine analogues against tachyzoites of the intracellular apicomplexan parasites Neospora caninum and Toxoplasma gondii. Antimicrob. Agents Chemother. 52:1999–2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Scolaro C, Chaplin AB, Hartinger CG, Bergamo A, Cocchietto M, Keppler BK, Sava G, Dyson PJ. 2007. Tuning the hydrophobicity of ruthenium(II)-arene (RAPTA) drugs to modify uptake, biomolecular interactions and efficacy. Dalton Trans. 43:5065–5072 [DOI] [PubMed] [Google Scholar]
  • 61.Ravera M, Baracco S, Cassino C, Colangelo D, Bagni G, Sava G, Osella D. 2004. Electrochemical measurements confirm the preferential bonding of the antimetastatic complex [ImH][RuCl(4)(DMSO)(Im)] (NAMI-A) with proteins and the weak interaction with nucleobases. J. Inorg. Biochem. 98:984–990 [DOI] [PubMed] [Google Scholar]
  • 62.Casini A, Gabbiani C, Sorrentino F, Rigobello MP, Bindoli A, Geldbach TJ, Marrone A, Re N, Hartinger CG, Dyson PJ, Messori L. 2008. Emerging protein targets for anticancer metallodrugs: inhibition of thioredoxin reductase and cathepsin B by antitumor ruthenium(II)-arene compounds. J. Med. Chem. 51:6773–6781 [DOI] [PubMed] [Google Scholar]
  • 63.Klinkert MQ, Heussler V. 2006. The use of anticancer drugs in antiparasitic chemotherapy. Mini Rev. Med. Chem. 6:131–143 [DOI] [PubMed] [Google Scholar]

Articles from Antimicrobial Agents and Chemotherapy are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES