Fexinidazole--a new oral nitroimidazole drug candidate entering clinical development for the treatment of sleeping sickness

Els Torreele, Bernadette Bourdin Trunz, David Tweats, Marcel Kaiser, Reto Brun, Guy Mazué, Michael A Bray, Bernard Pécoul, Els Torreele, Bernadette Bourdin Trunz, David Tweats, Marcel Kaiser, Reto Brun, Guy Mazué, Michael A Bray, Bernard Pécoul

Abstract

Background: Human African trypanosomiasis (HAT), also known as sleeping sickness, is a fatal parasitic disease caused by trypanosomes. Current treatment options for HAT are scarce, toxic, no longer effective, or very difficult to administer, in particular for the advanced, fatal stage of the disease (stage 2, chronic HAT). New safe, effective and easy-to-use treatments are urgently needed. Here it is shown that fexinidazole, a 2-substituted 5-nitroimidazole rediscovered by the Drugs for Neglected Diseases initiative (DNDi) after extensive compound mining efforts of more than 700 new and existing nitroheterocycles, could be a short-course, safe and effective oral treatment curing both acute and chronic HAT and that could be implemented at the primary health care level. To complete the preclinical development and meet the regulatory requirements before initiating human trials, the anti-parasitic properties and the pharmacokinetic, metabolic and toxicological profile of fexinidazole have been assessed.

Methods and findings: Standard in vitro and in vivo anti-parasitic activity assays were conducted to assess drug efficacy in experimental models for HAT. In parallel, a full range of preclinical pharmacology and safety studies, as required by international regulatory guidelines before initiating human studies, have been conducted. Fexinidazole is moderately active in vitro against African trypanosomes (IC₅₀ against laboratory strains and recent clinical isolates ranged between 0.16 and 0.93 µg/mL) and oral administration of fexinidazole at doses of 100 mg/kg/day for 4 days or 200 mg/kg/day for 5 days cured mice with acute and chronic infection respectively, the latter being a model for the advanced and fatal stage of the disease when parasites have disseminated into the brain. In laboratory animals, fexinidazole is well absorbed after oral administration and readily distributes throughout the body, including the brain. The absolute bioavailability of oral fexinidazole was 41% in mice, 30% in rats, and 10% in dogs. Furthermore, fexinidazole is rapidly metabolised in vivo to at least two biologically active metabolites (a sulfoxide and a sulfone derivative) that likely account for a significant portion of the therapeutic effect. Key pharmacokinetic parameter after oral absorption in mice for fexinidazole and its sulfoxide and sulfone metabolites are a C(max) of 500, 14171 and 13651 ng/mL respectively, and an AUC₀₋₂₄ of 424, 45031 and 96286 h.ng/mL respectively. Essentially similar PK profiles were observed in rats and dogs. Toxicology studies (including safety pharmacology and 4-weeks repeated-dose toxicokinetics in rat and dog) have shown that fexinidazole is well tolerated. The No Observed Adverse Event Levels in the 4-weeks repeated dose toxicity studies in rats and dogs was 200 mg/kg/day in both species, with no issues of concern identified for doses up to 800 mg/kg/day. While fexinidazole, like many nitroheterocycles, is mutagenic in the Ames test due to bacterial specific metabolism, it is not genotoxic to mammalian cells in vitro or in vivo as assessed in an in vitro micronucleus test on human lymphocytes, an in vivo mouse bone marrow micronucleus test, and an ex vivo unscheduled DNA synthesis test in rats.

Conclusions: The results of the preclinical pharmacological and safety studies indicate that fexinidazole is a safe and effective oral drug candidate with no untoward effects that would preclude evaluation in man. The drug has entered first-in-human phase I studies in September 2009. Fexinidazole is the first new clinical drug candidate with the potential for treating advanced-stage sleeping sickness in thirty years.

Conflict of interest statement

DT, GM and MAB are paid consultants to DNDi. All other authors declare that they have no competing interests.

Figures

Figure 1. Effect of fexinidazole and its…
Figure 1. Effect of fexinidazole and its two main metabolites on T. b. rhodesiense (STIB 900).
Parasite viability was measured in vitro after 72-h drug exposure. Fexinidazole - open circles (n = 11). Fexinidazole sulfoxide - open squares (n = 4). Fexinidazole sulfone - open diamonds (n = 4).
Figure 2. Chemical structure of fexinidazole and…
Figure 2. Chemical structure of fexinidazole and its main metabolites , including 14C-labeled fexinidazole indicating which carbon atom was labelled.
Figure 3. Plasma concentrations of fexinidazole and…
Figure 3. Plasma concentrations of fexinidazole and its two main metabolites after 5 days of oral administration.
200 mg/kg fexinidazole was administered to mice (n = 3). Fexinidazole - open circles. Fexinidazole sulfoxide - open squares. Fexinidazole sulfone - open diamonds.
Figure 4. Mutagenic activity of fexinidazole in…
Figure 4. Mutagenic activity of fexinidazole in the Ames test.
Salmonella typhymurium strains TA98 (A) and TA100 (B) and their nitroreductase-deficient variants TA98NR and TA100NR were used, in the presence and absence of metabolic activation (+/− S9). A: Solid circles: TA98 +S9; Open circles: TA98 -S9; Solid squares: TA98NR +S9; Open squares: TA98NR -S9; Negative control: Mean number of revertants per plate were TA98 (−S9): 21; TA98 (+S9): 34; TA98NR (−S9): 29; TA98 (+S9): 18. B: Solid circles: TA100 +S9; Open circles: TA100 −S9; Solid squares: TA100NR +S9; Open squares: TA100NR −S9; Negative control: Mean number of revertants per plate were TA100 (−S9): 104; TA100 (+S9): 116; TA100NR (−S9): 90; TA100NR (+S9): 111.

References

    1. Snowden M, Green DV. The impact of diversity-based, high-throughput screening on drug discovery: “chance favours the prepared mind”. Curr Opin Drug Discov Devel. 2008;11:553–558.
    1. Frearson JA, Collie IT. HTS and hit finding in academia–from chemical genomics to drug discovery. Drug Discov Today 14: 1150-1158. Epub 2009 Sep. 2009;28
    1. Noble ME, Endicott JA, Johnson LN. Protein kinase inhibitors: insights into drug design from structure. Science. 2004;303:1800–1805.
    1. WHO fact sheet. 28 . Accessed 2010, July.
    1. WHO Human African trypanosomiasis (sleeping sickness): epidemiological update. Weekly Epidemiological Record. 2006;8:71–80.
    1. Simarro PP, Jannin J, Cattand P. Eliminating human African trypanosomiasis: where do we stand and what comes next? PLoS Med. 2008;5:e55.
    1. Brun R, Blum J, Chappuis F, Burri C. Human African trypanosomiasis. Lancet. 2010;375:148–159.
    1. Barrett MP, Boykin DW, Brun R, Tidwell RR. Human African trypanosomiasis: pharmacological re-engagement with a neglected disease. Br J Pharmacol. 2007;152:1155–1171.
    1. Kennedy PG. Human African trypanosomiasis of the CNS: current issues and challenges. J Clin Invest. 2004;113:496–504.
    1. Legros D, Ollivier G, Gastellu-Etchegorry M, Paquet C, Burri C, et al. Treatment of human African trypanosomiasis: present situation and needs for research and development. Lancet Infect Dis. 2002;2:437–440.
    1. Robays J, Nyamowala G, Sese C, Betu Ku Mesu Kande V, Lutumba P, et al. High failure rates of melarsoprol for sleeping sickness, Democratic Republic of Congo. Emerg Infect Dis. 2008;14:966–967.
    1. Matovu E, Enyaru JC, Legros D, Schmid C, Seebeck T, et al. Melarsoprol refractory T. b. gambiense from Omugo, northwestern Uganda. Trop Med Int Health. 2001;6:407–411.
    1. Chappuis F, Udayraj N, Stietenroth K, Meussen A, Bovier PA. Eflornithine is safer than melarsoprol for the treatment of second-stage Trypanosoma brucei gambiense human African trypanosomiasis. Clin Infect Dis. 2005;41:748–751.
    1. Priotto G, Kasparian S, Mutombo W, Ngouma D, Ghorashian S, et al. Nifurtimox-eflornithine combination therapy for second-stage African Trypanosoma brucei gambiense trypanosomiasis: a multicentre, randomised phase III, non-inferiority trial. Lancet. 2009;374:56–64.
    1. Opigo J, Woodrow C. NECT trial: more than a small victory over sleeping sickness. Lancet. 2009;374:7–9.
    1. . Accessed 2010, July 28.
    1. Chappuis F, Lima MA, Flevaud L, Ritmeijer K. Human African trypanosomiasis in areas without surveillance. Emerg Infect Dis. 2010;16:354–356.
    1. Winkelmann E, Raether W, Gebert U, Sinharay A. Chemotherapeutically active nitro compounds. 4. 5-Nitroimidazoles (Part I-IV). Arzneimittelforschung. 1977;27-28
    1. Marie-Daragon A, Rouillard MC, Bouteille B, Bisser S, de Albuquerque C, et al. An efficacy trial on Trypanosoma brucei brucei of molecules permeating the blood-brain barrier and of megazol. Bull Soc Pathol Exot. 1994;87:347–352.
    1. Bouteille B, Marie-Daragon A, Chauviere G, de Albuquerque C, Enanga B, et al. Effect of megazol on Trypanosoma brucei brucei acute and subacute infections in Swiss mice. Acta Trop. 1995;60:73–80.
    1. Nesslany F, Brugier S, Mouries MA, Le Curieux F, Marzin D. In vitro and in vivo chromosomal aberrations induced by megazol. Mutat Res. 2004;560:147–158.
    1. Enanga B, Ariyanayagam MR, Stewart ML, Barrett MP. Activity of megazol, a trypanocidal nitroimidazole, is associated with DNA damage. Antimicrob Agents Chemother. 2003;47:3368–3370.
    1. De Meo M, Vanelle P, Bernadini E, Laget M, Maldonado J, et al. Evaluation of the mutagenic and genotoxic activities of 48 nitroimidazoles and related imidazole derivatives by the Ames test and the SOS chromotest. Environ Mol Mutagen. 1992;19:167–181.
    1. Voogd CE. On the mutagenicity of nitroimidazoles. Mutat Res. 1981;86:243–277.
    1. Freeman CD, Klutman NE, Lamp KC. Metronidazole. A therapeutic review and update. Drugs. 1997;54:679–708.
    1. Winkelmann E, Raether W. New chemotherapeutically active nitroimidazoles. Curr Chemother Infect Dis, Proc Int Congr Chemother. 1980;11th 2:969–970.
    1. Raether W, Seidenath H. The activity of fexinidazole (HOE 239) against experimental infections with Trypanosoma cruzi, trichomonads and Entamoeba histolytica. Ann Trop Med Parasitol. 1983;77:13–26.
    1. Jennings FW, Urquhart GM. The use of the 2 substituted 5-nitroimidazole, Fexinidazole (Hoe 239) in the treatment of chronic T. brucei infections in mice. Z Parasitenkd. 1983;69:577–581.
    1. Räz B, Iten M, Grether-Bühler Y, Kaminsky R, Brun R. The Alamar Blue assay to determine drug sensitivity of African trypanosomes in vitro. Acta Trop. 1997;68:139–147.
    1. Baltz T, Baltz D, Giroud C, Crockett J. Cultivation in a semi-defined medium of animal infective forms of Trypanosoma brucei, T. equiperdum, T. evansi, T. rhodesiense and T. gambiense. EMBO J. 1985;4:1273–1277.
    1. Wenzler T, Boykin DW, Ismail MA, Hall JE, Tidwell RR, et al. New treatment option for second-stage African sleeping sickness: in vitro and in vivo efficacy of aza analogs of DB289. Antimicrob Agents Chemother. 2009;53:4185–4192.
    1. Kaminsky R, Brun R. In Vitro and In Vivo Activities of Trybizine Hydrochloride against Various Pathogenic Trypanosome Species. Antimicrob. Agents and Chemother. 1998;42:2858–2862.
    1. Thuita JK, Karanja SM, Wenzler T, Mdachi RE, Ngotho JM, et al. Efficacy of the diamidine DB75 and its prodrug DB289, against murine models of human African trypanosomiasis. Acta Trop. 2008;108:6–10.
    1. Obach RS, Baxter JG, Liston TE, Silber BM, Jones BC, et al. The prediction of human pharmacokinetic parameters from preclinical and in vitro metabolism data. J Pharmacol Exp Ther. 1997;283:46–58.
    1. Dierks EA, Stams KR, Lim H-K, Cornelius G, Zhang H, et al. A Method for the Simultaneous Evaluation of the Activities of Seven Major Human Drug-Metabolizing Cytochrome P450s Using an in Vitro Cocktail of Probe Substrates and Fast Gradient Liquid Chromatography Tandem Mass Spectrometry. Drug Metab Dispos. 2001;29:23–29.
    1. Shah P, Jogani V, Bagchi T, Misra A. Role of Caco-2 cell monolayers in prediction of intestinal drug absorption, Biotechnol Prog. 2006;22:186–198.
    1. Wang Q, Rager JD, Weinstein K, Kardos PS, Glenn L, et al. Evaluation of the MDR-MDCK cell line as a permeability screen for the blood-brain barrier. Int J Pharm. 2005;288:349–359.
    1. European Agency for the Evaluation of Medicinal Products. ICH Harmonised Tripartite Guideline; 2001. ICH Topic S 7 A. “Safety Pharmacology Studies for Human Pharmaceuticals”.
    1. Pugsley MK, Authier S, Curtis MJ. Principles of safety pharmacology. Br J Pharmacol. 2008;154:1382–1399. Epub 2008 Jul 7.
    1. European Agency for the Evaluation of Medicinal Products. ICH Harmonised Tripartite Guideline; 2005. ICH Topic S 7 B. “The nonclinical Evaluation of the Potential for delayed Ventricular Repolarization (QT Interval Prolongation) by Human Pharmaceuticals”.
    1. Irwin S. Comprehensive observational assessment: 1a. A systematic, quantitative procedure for assessing the behavioural and physiologic state of the mouse. Psychopharmacologia (Berl) 1968;13:222–257.
    1. Murphy DJ. Safety Pharmacology of the Respiratory System: Techniques and Study Design. Drug Dev Res. 1994;32:237–246.
    1. European Agency for the Evaluation of Medicinal Products. ICH Topic M 3 (R2). “Non-Clinical Safety Studies for the Conduct of Human Clinical Trials and Marketing Authorization for Pharmaceuticals” 2009.
    1. European Agency for the Evaluation of Medicinal Products. ICH Harmonised Tripartite Guideline; 1995. ICH Topic S 3 A. “Toxicokinetics: A Guidance for Assessing Systemic Exposure in Toxicology Studies”.
    1. European Agency for the Evaluation of Medicinal Products. ICH Topic S 5A Reproductive toxicology: detection of toxicity to reproduction for medicinal products including toxicity to male fertility (CPMP/ICH/386/95). 1994
    1. European Agency for the Evaluation of Medicinal Products. ICH Topic S2 (R1). Guidance on Genotoxicity Testing and Data Interpretation for Pharmaceuticals Intended for Human Use. 2008 International Conference on Harmonisation of Technical Requirements for the Registration of Pharmaceuticals for Human Use. Note for Guidance on Genotoxicity : A standard battery for genotoxicity testing of pharmaceuticals; International Conference on Harmonisation of Technical Requirements for the Registration of Pharmaceuticals for Human Use. Note for Guidance on Genotoxicity: Guidance on specific aspects of regulatory genotoxicity tests of pharmaceuticals, . Accessed 2010 July 28.
    1. McCoy EC, Rosenkranz HS, Mermelstein R. Evidence for the existence of a family of bacterial nitroreductases capable of activating nitrated polycyclics to mutagens. Environ Mutagen. 1981;3:421–427.
    1. Purohit V, Basu AK. Mutagenicity of nitroaromatic compounds. Chem Res Toxicol. 2000;13:673–692.
    1. Maron DM, Ames BN. Revised methods for the Salmonella mutagenicity test. Mutat Res. 1983;113:173–215.
    1. Fenech M. Cytokinesis-block micronucleus cytome assay. Nature Protocols. 2007;2:1084–1104.
    1. Krishna G, Hayashi M. In vivo micronucleus assay: protocol, conduct and interpretation. Mutat Res. 2000;455:155–166.
    1. Kennelly JC, Waters R, Ashby J, Lefevre PA, Burlinson B, et al. In vivo rat liver UDS assay. In: Supplementary Mutagenicity Tests UKEMS Recommended Procedures. Kirkland DJ, Fox M editors. Cambridge: Cambridge University Press. pp. 1993;52-77
    1. Wardman P. Reduction potentials of one-electron couples involving free radicals in aqueous solution. J Phys Chem Ref Data. 1989;18:1637–1755.
    1. Barry CE, III, Boshoff HI, Dowd CS. Prospects for Clinical Introduction of Nitroimidazole Antibiotics for the Treatment of Tuberculosis. Curr Pharm Des. 2004;10:3239–3262.
    1. Wilkinson SR, Taylor MC, Horn D, Kelly JM, Cheeseman I. A mechanism for cross-resistance to nifurtimox and benzidazole in trypanosomes, Proc Nat Acad Sci USA. 2008;105:5022–5027.
    1. Reagan-Shaw S, Nihal M, Ahmad N. Dose translation from animal to human studies revisited. FASEB J. 2008;22:659–661.
    1. Kirkland DJ, Aardema M, Banduhn N, Carmichael P, Fautz R, et al. In vitro approaches to develop weight of evidence (W0E) and mode of action (MoA) discussions with positive in vitro genotoxicity results. Mutagenesis. 2007;22:161–175.
    1. Suter W, Hartmann A, Poetter F, Sagelsdorff P, Hoffmann P. Genotoxicity assessment of the antiepileptic drug AMP397, an Ames-positive aromatic nitro compound. Mutat Res. 2002;518:181–194.
    1. Stover CK, Warrener P, VanDevanter DR, Sherman DR, Arain TM, et al. A small-molecule nitroimidazopyran drug candidate for the treatment of tuberculosis. Nature. 2000;405:962–966.
    1. Matsumoto M, Hashizume H, Tomishige T, Kawasaki M, Tsubouchi H, et al. OPC-67683, a nitro-dihydro-imidazooxazole derivative with promising action against tuberculosis in vitro and in mice. PLoS Med. 2006;3:e466.
    1. Benfenati E, Benigni R, Demarini DM, Helma C, Kirkland D, et al. Predictive models for carcinogenicity and mutagenicity frameworks, state-of-the-art and perspectives. J Environ Sci Health C Eviron Carcinog Ecotoxicol Rev. 2009;27:57–90.
    1. , Identifier: NCT00982904. Accessed 2010 July 28.

Source: PubMed

3
Tilaa