Fexinidazole – A New Oral Nitroimidazole Drug Candidate Entering Clinical Development for the Treatment of Sleeping Sickness

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 Cmax 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.

[1]  M. Boelaert,et al.  High failure rates of melarsoprol for sleeping sickness, Democratic Republic of Congo. , 2008, Emerging infectious diseases.

[2]  D. Murphy Safety pharmacology of the respiratory system: Techniques and study design , 1994 .

[3]  F. Chappuis,et al.  Human African Trypanosomiasis in Areas without Surveillance , 2010, Emerging infectious diseases.

[4]  J. Frearson,et al.  HTS and hit finding in academia – from chemical genomics to drug discovery , 2009, Drug discovery today.

[5]  J. Périé,et al.  Effect of megazol on Trypanosoma brucei brucei acute and subacute infections in Swiss mice. , 1995, Acta tropica.

[6]  R Fautz,et al.  In vitro approaches to develop weight of evidence (WoE) and mode of action (MoA) discussions with positive in vitro genotoxicity results. , 2007, Mutagenesis.

[7]  P. Kennedy,et al.  Human African trypanosomiasis of the CNS: current issues and challenges. , 2004, The Journal of clinical investigation.

[8]  C. Barry,et al.  Prospects for clinical introduction of nitroimidazole antibiotics for the treatment of tuberculosis. , 2004, Current pharmaceutical design.

[9]  W. Raether,et al.  Chemotherapeutically active nitro compounds. 4.5-Nitroimidazoles (Part II). , 1978, Arzneimittel-Forschung.

[10]  T. Baltz,et al.  Cultivation in a semi‐defined medium of animal infective forms of Trypanosoma brucei, T. equiperdum, T. evansi, T. rhodesiense and T. gambiense. , 1985, The EMBO journal.

[11]  Jimmy Opigo,et al.  NECT trial: more than a small victory over sleeping sickness , 2009, The Lancet.

[12]  Nihal Ahmad,et al.  Dose translation from animal to human studies revisited , 2007, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[13]  R. Kaminsky,et al.  Melarsoprol refractory T. b. gambiense from Omugo, north‐western Uganda , 2001, Tropical medicine & international health : TM & IH.

[14]  F. Nesslany,et al.  In vitro and in vivo chromosomal aberrations induced by megazol. , 2004, Mutation research.

[15]  B. Ames,et al.  Revised methods for the Salmonella mutagenicity test. , 1983, Mutation research.

[16]  S. Irwin,et al.  Comprehensive observational assessment: Ia. A systematic, quantitative procedure for assessing the behavioral and physiologic state of the mouse , 1968, Psychopharmacologia.

[17]  D. Boykin,et al.  New Treatment Option for Second-Stage African Sleeping Sickness: In Vitro and In Vivo Efficacy of Aza Analogs of DB289 , 2009, Antimicrobial Agents and Chemotherapy.

[18]  The effect of reducing the number of cells scored on the performance of the in vivo rat liver UDS assay. , 1995, Mutation research.

[19]  F. Jennings,et al.  The use of the 2 substituted 5-nitroimidazole, fexinidazole (Hoe 239) in the treatment of chronicT. brucei infections in mice , 2004, Zeitschrift für Parasitenkunde.

[20]  A. Basu,et al.  Mutagenicity of nitroaromatic compounds. , 2000, Chemical research in toxicology.

[21]  G. Duménil,et al.  Evaluation of the mutagenic and genotoxic activities of 48 nitroimidazoles and related imidazole derivatives by the Ames test and the SOS Chromotest , 1992, Environmental and molecular mutagenesis.

[22]  Salah Ghabri,et al.  Nifurtimox-eflornithine combination therapy for second-stage African Trypanosoma brucei gambiense trypanosomiasis: a multicentre, randomised, phase III, non-inferiority trial , 2009, The Lancet.

[23]  L. Johnson,et al.  Protein Kinase Inhibitors: Insights into Drug Design from Structure , 2004, Science.

[24]  R. Brun,et al.  Efficacy of the diamidine DB75 and its prodrug DB289, against murine models of human African trypanosomiasis. , 2008, Acta tropica.

[25]  Anika Ashok,et al.  ICH Harmonised Tripartite Guideline , 2009 .

[26]  C. E. Voogd On the mutagenicity of nitroimidazoles. , 1981, Mutation research.

[27]  R. Kaminsky,et al.  In Vitro and In Vivo Activities of Trybizine Hydrochloride against Various Pathogenic Trypanosome Species , 1998, Antimicrobial Agents and Chemotherapy.

[28]  M. Matsumoto,et al.  OPC-67683, a Nitro-Dihydro-Imidazooxazole Derivative with Promising Action against Tuberculosis In Vitro and In Mice , 2006, PLoS medicine.

[29]  Human African trypanosomiasis (sleeping sickness): epidemiological update. , 2006, Releve epidemiologique hebdomadaire.

[30]  The nonclinical Evaluation of the Potential for delayed Ventricular Repolarization (QT Interval Prolongation) by Human Pharmaceuticals Step 5 NOTE FOR GUIDANCE ON THE NONCLINICAL EVALUATION OF THE POTENTIAL FOR DELAYED VENTRICULAR REPOLARIZATION (QT INTERVAL PROLONGATION) BY HUMAN PHARMACEUTICALS (C , 2004 .

[31]  F. Chappuis,et al.  Eflornithine is safer than melarsoprol for the treatment of second-stage Trypanosoma brucei gambiense human African trypanosomiasis. , 2005, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America.

[32]  M. Hayashi,et al.  In vivo rodent micronucleus assay: protocol, conduct and data interpretation. , 2000, Mutation research.

[33]  H. Rosenkranz,et al.  Evidence for the existence of a family of bacterial nitroreductases capable of activating nitrated polycyclics to mutagens. , 1981, Environmental mutagenesis.

[34]  D. Horn,et al.  A mechanism for cross-resistance to nifurtimox and benznidazole in trypanosomes , 2008, Proceedings of the National Academy of Sciences.

[35]  P. Hoffmann,et al.  Genotoxicity assessment of the antiepileptic drug AMP397, an Ames-positive aromatic nitro compound. , 2002, Mutation research.

[36]  M K Pugsley,et al.  Principles of Safety Pharmacology , 2008, Handbook of Experimental Pharmacology.

[37]  D J Rance,et al.  The prediction of human pharmacokinetic parameters from preclinical and in vitro metabolism data. , 1997, The Journal of pharmacology and experimental therapeutics.

[38]  P. Simarro,et al.  Eliminating Human African Trypanosomiasis: Where Do We Stand and What Comes Next> , 2008, PLoS medicine.

[39]  K. Lamp,et al.  Metronidazole. A therapeutic review and update. , 1997, Drugs.

[40]  GUIDANCE DOCUMENT,et al.  Guidance on Genotoxicity Testing and Data Interpretation for Pharmaceuticals Intended for Human Use , 2008 .

[41]  M. Barrett,et al.  Human African trypanosomiasis: pharmacological re‐engagement with a neglected disease , 2007, British journal of pharmacology.

[42]  M. Fenech Cytokinesis-block micronucleus cytome assay , 2007, Nature Protocols.

[43]  M. Barrett,et al.  Activity of Megazol, a Trypanocidal Nitroimidazole, Is Associated with DNA Damage , 2003, Antimicrobial Agents and Chemotherapy.

[44]  I. Hidalgo,et al.  Evaluation of the MDR-MDCK cell line as a permeability screen for the blood-brain barrier. , 2005, International journal of pharmaceutics.

[45]  C Helma,et al.  Predictive Models for Carcinogenicity and Mutagenicity: Frameworks, State-of-the-Art, and Perspectives , 2009, Journal of environmental science and health. Part C, Environmental carcinogenesis & ecotoxicology reviews.

[46]  Michael Snowden,et al.  The impact of diversity-based, high-throughput screening on drug discovery: "chance favours the prepared mind". , 2008, Current opinion in drug discovery & development.

[47]  Honglu Zhang,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. , 2001, Drug metabolism and disposition: the biological fate of chemicals.

[48]  Clifton E. Barry,et al.  A small-molecule nitroimidazopyran drug candidate for the treatment of tuberculosis , 2000, Nature.

[49]  W. Raether,et al.  The activity of fexinidazole (HOE 239) against experimental infections with Trypanosoma cruzi, trichomonads and Entamoeba histolytica. , 1983, Annals of tropical medicine and parasitology.

[50]  R. Kaminsky,et al.  The Alamar Blue assay to determine drug sensitivity of African trypanosomes (T.b. rhodesiense and T.b. gambiense) in vitro. , 1997, Acta tropica.

[51]  P. Wardman,et al.  Reduction Potentials of One-Electron Couples Involving Free Radicals in Aqueous Solution , 1989 .

[52]  C. Burri,et al.  Treatment of human African trypanosomiasis--present situation and needs for research and development. , 2002, The Lancet. Infectious diseases.

[53]  Pranav Shah,et al.  Role of Caco‐2 Cell Monolayers in Prediction of Intestinal Drug Absorption , 2006, Biotechnology progress.