Nitroheterocyclic compounds are more efficacious than CYP51 inhibitors against Trypanosoma cruzi: implications for Chagas disease drug discovery and development

Carolina B Moraes, Miriam A Giardini, Hwayoung Kim, Caio H Franco, Adalberto M Araujo-Junior, Sergio Schenkman, Eric Chatelain, Lucio H Freitas-Junior, Carolina B Moraes, Miriam A Giardini, Hwayoung Kim, Caio H Franco, Adalberto M Araujo-Junior, Sergio Schenkman, Eric Chatelain, Lucio H Freitas-Junior

Abstract

Advocacy for better drugs and access to treatment has boosted the interest in drug discovery and development for Chagas disease, a chronic infection caused by the genetically heterogeneous parasite, Trypanosoma cruzi. In this work new in vitro assays were used to gain a better understanding of the antitrypanosomal properties of the most advanced antichagasic lead and clinical compounds, the nitroheterocyclics benznidazole, nifurtimox and fexinidazole sulfone, the oxaborole AN4169, and four ergosterol biosynthesis inhibitors--posaconazole, ravuconazole, EPL-BS967 and EPL-BS1246. Two types of assays were developed: one for evaluation of potency and efficacy in dose-response against a panel of T. cruzi stocks representing all current discrete typing units (DTUs), and a time-kill assay. Although less potent, the nitroheterocyclics and the oxaborole showed broad efficacy against all T. cruzi tested and were rapidly trypanocidal, whilst ergosterol biosynthesis inhibitors showed variable activity that was both compound- and strain-specific, and were unable to eradicate intracellular infection even after 7 days of continuous compound exposure at most efficacious concentrations. These findings contest previous reports of variable responses to nitroderivatives among different T. cruzi strains and further challenge the introduction of ergosterol biosynthesis inhibitors as new single chemotherapeutic agents for the treatment of Chagas disease.

Figures

Figure 1. The nitroheterocyclics benznidazole and nifurtimox…
Figure 1. The nitroheterocyclics benznidazole and nifurtimox are the most efficacious compounds against a phylogenetically broad panel of Trypanosoma cruzi.
Tissue cultures in 384-well plates were infected with one of the following T. cruzi: Dm28c (DTU I, purple), Y (DTU II, red), ARMA13 cl1 (DTU III, orange), ERA cl2 (DTU IV, light green), 92-80 cl2 (DTU V, light blue), CL Brener (DTU VI, dark blue), and Tulahuen (DTU VI, dark green). Compounds were 2-fold diluted and were added to infected cultures 24 h after T. cruzi plating and incubated for 72 (Dm28c) or 96 h (all others) prior to assessment of antiparasitic activity by high content analysis. Dose-response curves of compounds antiparasitic activity normalized to infected and non-infected controls are shown. The X-axis shows log of compound molar (M) concentrations and Y-axis shows the normalized activity, based on the measurement of ratio of infected cells. Data refers to mean values of at least two independent experiments.
Figure 2. Benznidazole, nifurtimox, fexinidazole sulfone, and…
Figure 2. Benznidazole, nifurtimox, fexinidazole sulfone, and AN4169 are fast trypanocidal compounds that can eliminate intracellular T. cruzi within 96 h of continuous exposure in vitro.
U2OS cells tissue cultures in 384-well plates were infected with the Y strain for 24 hours prior to addition of compounds in serial dilution by the factor of 2-fold, as indicated the by colors according to the compound gradient (right, top). The following concentrations are shown: 400, 200, 100, 50, 25, 12.5, 6.25, 3.125, 1.56 and 0.78 μM for benznidazole and fexinidazole sulfone; 100, 50, 25, 12.5, 6.25, 3.125, 1.56, 0.78, 0.39, and 0.20 μM for nifurtimox; and 40, 20, 10, 5, 2.5, 1.25, 0.625, 0.31, 0.16 and 0.078 μM for AN4169. Antiparasitic activity was analyzed every 24 h, starting right after compound addition (time point 0 h), up to 96 h. Data points are means and standard deviations of at least two independent experiments.
Figure 3. TcCYP51 inhibitors are slow trypanocidal…
Figure 3. TcCYP51 inhibitors are slow trypanocidal compounds that after prolonged exposure can greatly reduce but not eliminate intracellular infection.
U2OS cells tissue cultures in 384-well plates were infected with the Y strain for 24 hours prior to addition of compounds in serial dilution by the factor of 2-fold and as indicated the by the colors according to compound gradient (right, top). The following concentrations are shown: 80, 40, 20, 10, 5, 2.5, 1.25, 0.625, 0.3125 and 0.156 nM for posaconazole and ravuconazole; and 2000, 1000, 500, 250, 125, 62.5, 31.25, 15.62, 7.81 and 3.91 nM for EPL-BS967 and EPL-BS1246. Antiparasitic activity was analyzed every 24 h, starting right after compound addition (time point 0 h), up to 144 h. Data points are means and standard deviations of three independent experiments.

References

    1. WHO. Working to overcome the global impact of neglected tropical diseases. Available from (2010) (Accessed on 18/02/2014).
    1. Gascon J., Bern C. & Pinazo M.-J. Chagas disease in Spain, the United States and other non-endemic countries. Acta Trop. 115, 22–27 (2010).
    1. Bern C. & Montgomery S. P. An estimate of the burden of Chagas disease in the United States. Clin. Infec. Dis. 49, e52–4 (2009).
    1. Lee B. Y., Bacon K. M., Bottazzi M. E. & Hotez P. J. Global economic burden of Chagas disease: a computational simulation model. Lancet Infect. Dis. 13, 342–348 (2013).
    1. Rassi A. J., Rassi A. & Marcondes de Rezende J. American trypanosomiasis (Chagas disease). Infect. Dis. Clin. North Am. 26, 275–291 (2012).
    1. Coura J. R. & de Castro S. L. A critical review on Chagas disease chemotherapy. Mem. Inst. Oswaldo Cruz 97, 3–24 (2002).
    1. Coura J. R. & Borges-Pereira J. Chronic phase of Chagas disease: why should it be treated? A comprehensive review. Mem. Inst. Oswaldo Cruz 106, 641–645 (2011).
    1. Marin-Neto J. A. et al. Rationale and design of a randomized placebo-controlled trial assessing the effects of etiologic treatment in Chagas' cardiomyopathy: the BENznidazole Evaluation For Interrupting Trypanosomiasis (BENEFIT). Am. Heart J. 156, 37–43 (2008).
    1. Marin-Neto J. A. et al. The BENEFIT trial: testing the hypothesis that trypanocidal therapy is beneficial for patients with chronic Chagas heart disease. Mem. Inst. Oswaldo Cruz 104 Suppl 1, 319–324 (2009).
    1. Castro J. A., de Mecca M. M. & Bartel L. C. Toxic side effects of drugs used to treat Chagas' disease (American trypanosomiasis). Hum. Exp. Toxicol. 25, 471–479 (2006).
    1. Viotti R. et al. Side effects of benznidazole as treatment in chronic Chagas disease: fears and realities. Expert Rev. Anti Infect. Ther. 7, 157–163 (2009).
    1. Yun O. et al. Feasibility, drug safety, and effectiveness of etiological treatment programs for Chagas disease in Honduras, Guatemala, and Bolivia: 10-year experience of Medecins Sans Frontieres. PLoS Negl. Trop. Dis. 3, e488 (2009).
    1. Sosa Estani S. et al. Efficacy of chemotherapy with benznidazole in children in the indeterminate phase of Chagas' disease. Am. J. Trop. Med. Hyg. 59, 526–529 (1998).
    1. Sosa-Estani S., Colantonio L. & Segura E. L. Therapy of Chagas disease: implications for levels of prevention. J. Trop. Med. 2012, (2012).
    1. Urbina J. A. et al. Antiproliferative effects and mechanism of action of SCH 56592 against Trypanosoma (Schizotrypanum) cruzi: in vitro and in vivo studies. Antimicrob. Agents Chemother. 42, 1771–1777 (1998).
    1. Molina J. et al. Activities of the triazole derivative SCH 56592 (posaconazole) against drug-resistant strains of the protozoan parasite Trypanosoma (Schizotrypanum) cruzi in immunocompetent and immunosuppressed murine hosts. Antimicrob. Agents Chemother. 44, 150–155 (2000).
    1. Urbina J. A., Payares G., Sanoja C., Lira R. & Romanha A. J. In vitro and in vivo activities of ravuconazole on Trypanosoma cruzi, the causative agent of Chagas disease. Int. J. Antimicrob. Agents 21, 27–38 (2003).
    1. Ferraz M. L., Gazzinelli R. T., Alves R. O., Urbina J. A. & Romanha A. J. The Anti-Trypanosoma cruzi activity of posaconazole in a murine model of acute Chagas' disease is less dependent on gamma interferon than that of benznidazole. Antimicrob. Agents Chemother. 51, 1359–1364 (2007).
    1. Olivieri B. P. et al. A comparative study of posaconazole and benznidazole in the prevention of heart damage and promotion of trypanocidal immune response in a murine model of Chagas disease. Int. J. Antimicrob. Agents 36, 79–83 (2010).
    1. Diniz L. et al. Effects of ravuconazole treatment on parasite load and immune response in dogs experimentally infected with Trypanosoma cruzi. Antimicrob. Agents. Chemother. 54, 2979–2986 (2010).
    1. Veiga-Santos P. et al. Effects of amiodarone and posaconazole on the growth and ultrastructure of Trypanosoma cruzi. Int. J. Antimicrob. Agents 40, 61–71 (2012).
    1. Buckner F. S. Experimental chemotherapy and approaches to drug discovery for Trypanosoma cruzi infection. Adv. Parasitol. 75, 89 (2011).
    1. Merck. STOPCHAGAS: a Study of the Use of Oral Posaconazole (POS) in the Treatment of Asymptomatic Chronic Chagas Disease. In: [Internet]. Bethesda (MD): National Library of Medicine (US). 2011 May 13 (cited 2013 December 07). Available from <> Identifier NCT 01377480.
    1. Drugs for Neglected Diseases initiative. Proof of Concept Study of E1224 to Treat Adults Patients with Chagas Disease. In: [Internet]. Bethesda (MD): National Library of Medicine (US). 2011 November 24 (cited 2013 December 07). Available from <> Identifier NCT 01489228.
    1. Hospital Universitari Vall d'Hebron Research Institute. Clinical Trial For The Treatment Of Chronic Chagas Disease With Posaconazole And Benznidazole (CHAGASAZOL). In: [Internet]. Bethesda (MD): National Library of Medicine (US). 2011 November 24 (cited 2013 December 07). Available from <> Identifier NCT 01162967.
    1. Bahia M. T. et al. Fexinidazole: A potential new drug candidate for Chagas disease. PLoS Negl. Trop. Dis. 6, e1870 (2012).
    1. Bustamante J. M., Craft J. M., Crowe B. D., Ketchie S. A. & Tarleton R. L. New, combined, and reduced dosing treatment protocols cure Trypanosoma cruzi infection in mice. J. Infect. Dis. 10.1093/infdis/jit420 (2013).
    1. Keenan M. et al. Analogues of fenarimol are potent inhibitors of Trypanosoma cruzi and are efficacious in a murine model of Chagas disease. J. Med. Chem. 55, 4189–4204 (2012).
    1. Hargrove T. Y. et al. Complexes of Trypanosoma cruzi sterol 14alpha-demethylase (CYP51) with two pyridine-based drug candidates for Chagas disease: structural basis for pathogen-selectivity. J. Biol. Chem. 10.1074/jbc.M113.497990 (2013).
    1. Keenan M. et al. Design, structure-activity relationship and in vivo efficacy of piperazine analogues of fenarimol as inhibitors of Trypanosoma cruzi. Bioorg. Med. Chem. 21, 1756–1763 (2013).
    1. Zingales B. et al. A new consensus for Trypanosoma cruzi intraspecific nomenclature: second revision meeting recommends TcI to TcVI. Mem. Inst. Oswaldo Cruz 104, 1051–1054 (2009).
    1. Miles M. A. et al. The molecular epidemiology and phylogeography of Trypanosoma cruzi and parallel research on Leishmania: looking back and to the future. Parasitology 136, 1509 (2009).
    1. Marcili A. et al. Comparative phylogeography of Trypanosoma cruzi TCIIc: New hosts, association with terrestrial ecotopes, and spatial clustering. Infect. Genet. Evol. 9, 1265–1274 (2009).
    1. Llewellyn M. S. et al. Genome-scale multilocus microsatellite typing of Trypanosoma cruzi discrete typing unit I reveals phylogeographic structure and specific genotypes linked to human infection. PLoS Pathog. 5, e1000410 (2009).
    1. Lewis M. D. et al. Genotyping of Trypanosoma cruzi: systematic selection of assays allowing rapid and accurate discrimination of all known lineages. Am. J. Trop. Med. Hyg. 81, 1041–1049 (2009).
    1. Carrasco H. J. et al. Geographical distribution of Trypanosoma cruzi genotypes in Venezuela. PLoS Negl. Trop. Dis. 6, e1707 EP– (2012).
    1. Burgos J. M. et al. Molecular identification of Trypanosoma cruzi discrete typing units in end-stage chronic Chagas heart disease and reactivation after heart transplantation. Clin. Infect. Dis. 51, 485–495 (2010).
    1. Cura C. I. et al. Trypanosoma cruzi discrete typing units in Chagas disease patients from endemic and non-endemic regions of Argentina. Parasitology 139, 516–521 (2012).
    1. Filardi L. S. & Brener Z. Susceptibility and natural resistance of Trypanosoma cruzi strains to drugs used clinically in Chagas disease. Trans. R. Soc. Trop. Med. Hyg. 81, 755–759 (1987).
    1. Neal R. A. & van Bueren J. Comparative studies of drug susceptibility of five strains of Trypanosoma cruzi in vivo and in vitro. Trans. R. Soc. Trop. Med. Hyg. 82, 709–714 (1988).
    1. Veloso V. M. et al. Variation in susceptibility to benznidazole in isolates derived from Trypanosoma cruzi parental strains. Mem. Inst. Oswaldo Cruz 96, 1005–1011 (2001).
    1. Camandaroba E. L. P., Reis E. A. G., Gonçalves M. S., Reis M. G. & Andrade S. G. Trypanosoma cruzi: susceptibility to chemotherapy with benznidazole of clones isolated from the highly resistant Colombian strain. Rev. Soc. Bras. Med. Trop. 36, 201–209 (2003).
    1. Moreno M. et al. Trypanosoma cruzi benznidazole susceptibility in vitro does not predict the therapeutic outcome of human Chagas disease. Mem. Inst. Oswaldo Cruz 105, 918–924 (2010).
    1. Teston A. P. et al. In vivo susceptibility to benznidazole of Trypanosoma cruzi strains from the western Brazilian Amazon. Trop. Med. Int. Health 18, 85–95 (2013).
    1. Nare B. et al. Discovery of novel orally bioavailable oxaborole 6-carboxamides that demonstrate cure in a murine model of late-stage central nervous system African trypanosomiasis. Antimicrob. Agents Chemother. 54, 4379–4388 (2010).
    1. Genovesio A. et al. Visual genome-wide RNAi screening to identify human host factors required for Trypanosoma cruzi infection. PLoS ONE 6, e19733 (2011).
    1. Heise N., Raper J., Buxbaum L. U., Peranovich T. M. & de Almeida M. L. Identification of complete precursors for the glycosylphosphatidylinositol protein anchors of Trypanosoma cruzi. J. Biol. Chem. 271, 16877–16887 (1996).
    1. Moon S. et al. An image-based algorithm for precise and accurate high throughput assessment of drug activity against the human parasite Trypanosoma cruzi. PLoS ONE 9, e87188 (2014).
    1. Lepesheva G. I. et al. Structural insights into inhibition of sterol 14alpha-demethylase in the human pathogen Trypanosoma cruzi. J. Biol. Chem. 285, 25582–25590 (2010).
    1. Dvorak J. A. The natural heterogeneity of Trypanosoma cruzi: biological and medical implications. J. Cell. Biochem. 24, 357–371 (1984).
    1. Engel J. C., Doyle P. S. & Dvorak J. A. Trypanosoma cruzi: biological characterization of clones derived from chronic chagasic patients. II. Quantitative analysis of the intracellular cycle. J Protozool 32, 80–83 (1985).
    1. Siqueira-Neto J. L. et al. An image-based high-content screening assay for compounds targeting intracellular Leishmania donovani amastigotes in human macrophages. PLoS Negl. Trop. Dis. 6, e1671 (2012).
    1. Villarreal D., Barnabé C., Sereno D. & Tibayrenc M. Lack of correlation between in vitro susceptibility to benznidazole and phylogenetic diversity of Trypanosoma cruzi, the agent of Chagas disease. Exp. Parasitol. 108, 24–31 (2004).
    1. Buckner F. S., Wilson A. J., White T. C. & Van Voorhis W. C. Induction of resistance to azole drugs in Trypanosoma cruzi. Antimicrob. Agents. Chemother. 42, 3245–3250 (1998).
    1. Wilkinson S. R., Taylor M. C., Horn D., Kelly J. M. & Cheeseman I. A mechanism for cross-resistance to nifurtimox and benznidazole in trypanosomes. Proc. Natl. Acad. Sci. U.S.A. 105, 5022–5027 (2008).
    1. Mejia A. M. et al. Benznidazole-resistance in Trypanosoma cruzi is a readily acquired trait that can arise independently in a single population. J. Infect. Dis. 206, 220–228 (2012).
    1. Akama T. et al. Linking phenotype to kinase: Identification of a novel benzoxaborole hinge binding motif for kinase inhibition and the development of high potency rho-kinase inhibitors. J. Pharmacol. Exp. Ther. 10.1124/jpet.113.207662 (2013).
    1. Shapiro A. B., Gao N., Hajec L. & McKinney D. C. Time-dependent, reversible, oxaborole inhibition of Escherichia coli leucyl-tRNA synthetase measured with a continuous fluorescence assay. Anal. Biochem. 431, 48–53 (2012).
    1. Hu Q.-H. et al. Discovery of a potent benzoxaborole-based anti-pneumococcal agent targeting leucyl-tRNA synthetase. Sci. Rep. 3, 2475 (2013).
    1. Drugs for Neglected Diseases initiative. Human African Trypanosomiasis: First in Man Clinical Trial of a New Medicinal Product, the SCYX-7158. In: ClinicalTrialsgov [Internet] Bethesda (MD): National Library of Medicine (US). 2012 February 13 (cited 2013 December 07). Available from <> Identifier NCT01533961.
    1. Buckner F. S. & Urbina J. A. Recent developments in sterol 14-demethylase inhibitors for Chagas disease. Int. J. Parasitol. Drugs Drug Resist. 2, 236–242 (2012).
    1. Lepesheva G. I., Villalta F. & Waterman M. R. Targeting Trypanosoma cruzi sterol 14α-demethylase (CYP51). Adv. Parasitol. 75, 65–87 (2011).
    1. Chen C.-K. et al. Structural characterization of CYP51 from Trypanosoma cruzi and Trypanosoma brucei bound to the antifungal drugs posaconazole and fluconazole. PLoS Negl. Trop. Dis. 4, e651 (2010).
    1. Urbina J. A. Specific chemotherapy of Chagas disease: relevance, current limitations and new approaches. Acta Trop. 115, 55–68 (2010).
    1. Buckner F. S. Sterol 14-demethylase inhibitors for Trypanosoma cruzi infections. Adv. Exp. Med. Biol. 625, 61–80 (2008).
    1. Jacobs R. T. et al. SCYX-7158, an orally-active benzoxaborole for the treatment of stage 2 human African trypanosomiasis. PLoS Negl. Trop. Dis. 5, e1151 (2011).
    1. Pinazo M.-J. et al. Benznidazole-related adverse drug reactions and their relationship in patients with chronic Chagas disease. Antimicrob. Agents Chemother. 57, 390–395 (2013).

Source: PubMed

3
Abonnere