Thierry T. Diagana
Novartis
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Thierry T. Diagana.
Science | 2010
Matthias Rottmann; Case W. McNamara; Bryan K. S. Yeung; Marcus C. S. Lee; Bin Zou; Bruce Russell; Patrick Seitz; David Plouffe; Neekesh V. Dharia; Jocelyn Tan; Steven B. Cohen; Kathryn R. Spencer; Gonzalo E. González-Páez; Suresh B. Lakshminarayana; Anne Goh; Rossarin Suwanarusk; Timothy Jegla; Esther K. Schmitt; Hans-Peter Beck; Reto Brun; François Nosten; Laurent Rénia; Véronique Dartois; Thomas H. Keller; David A. Fidock; Elizabeth A. Winzeler; Thierry T. Diagana
Antimalarial Drug Candidate Spiroindolones were discovered as promising antimalarial drug candidates through a high-throughput screening approach that should be applicable to a range of neglected infectious diseases. Rottmann et al. (p. 1175; see the Perspective by Wells) present the preclinical profile for an optimized spiroindolone drug candidate, NITD609. They obtained evidence for a decrease in drug sensitivity in strains of the malaria parasite Plasmodium falciparum bearing amino acid mutations in the P-type ATPase, indicating possible mechanisms of action and/or resistance. High-throughput screening has offered up an oral antimalarial drug and pointers to its mechanism of action. Recent reports of increased tolerance to artemisinin derivatives—the most recently adopted class of antimalarials—have prompted a need for new treatments. The spirotetrahydro-β-carbolines, or spiroindolones, are potent drugs that kill the blood stages of Plasmodium falciparum and Plasmodium vivax clinical isolates at low nanomolar concentration. Spiroindolones rapidly inhibit protein synthesis in P. falciparum, an effect that is ablated in parasites bearing nonsynonymous mutations in the gene encoding the P-type cation-transporter ATPase4 (PfATP4). The optimized spiroindolone NITD609 shows pharmacokinetic properties compatible with once-daily oral dosing and has single-dose efficacy in a rodent malaria model.
Journal of Medicinal Chemistry | 2010
Bryan K. S. Yeung; Bin Zou; Matthias Rottmann; Suresh B. Lakshminarayana; Shi Hua Ang; Seh Yong Leong; Jocelyn Tan; Josephine Wong; Sonja Keller-Maerki; Christoph Fischli; Anne Goh; Esther K. Schmitt; Philipp Krastel; Eric Francotte; Kelli Kuhen; David Plouffe; Kerstin Henson; Trixie Wagner; Elizabeth A. Winzeler; Frank Petersen; Reto Brun; Véronique Dartois; Thierry T. Diagana; Thomas H. Keller
The antiplasmodial activity of a series of spirotetrahydro β-carbolines is described. Racemic spiroazepineindole (1) was identified from a phenotypic screen on wild type Plasmodium falciparum with an in vitro IC50 of 90 nM. Structure−activity relationships for the optimization of 1 to compound 20a (IC50 = 0.2 nM) including the identification of the active 1R,3S enantiomer and elimination of metabolic liabilities is presented. Improvement of the pharmacokinetic profile of the series translated to exceptional oral efficacy in the P. berghei infected malaria mouse model where full cure was achieved in four of five mice with three daily doses of 30 mg/kg.
Science | 2011
Stephan Meister; David Plouffe; Kelli Kuhen; Ghislain M. C. Bonamy; Tao Wu; S. Whitney Barnes; Selina Bopp; Rachel Borboa; A. Taylor Bright; Jianwei Che; Steve Cohen; Neekesh V. Dharia; Kerstin Gagaring; Montip Gettayacamin; Perry Gordon; Todd Groessl; Nobutaka Kato; Marcus C. S. Lee; Case W. McNamara; David A. Fidock; Advait Nagle; Tae-gyu Nam; Wendy Richmond; Jason Roland; Matthias Rottmann; Bin Zhou; Patrick Froissard; Richard Glynne; Dominique Mazier; Jetsumon Sattabongkot
Imidazolopiperazine compounds inhibit liver-stage malaria parasites with one oral dose in mice. Most malaria drug development focuses on parasite stages detected in red blood cells, even though, to achieve eradication, next-generation drugs active against both erythrocytic and exo-erythrocytic forms would be preferable. We applied a multifactorial approach to a set of >4000 commercially available compounds with previously demonstrated blood-stage activity (median inhibitory concentration < 1 micromolar) and identified chemical scaffolds with potent activity against both forms. From this screen, we identified an imidazolopiperazine scaffold series that was highly enriched among compounds active against Plasmodium liver stages. The orally bioavailable lead imidazolopiperazine confers complete causal prophylactic protection (15 milligrams/kilogram) in rodent models of malaria and shows potent in vivo blood-stage therapeutic activity. The open-source chemical tools resulting from our effort provide starting points for future drug discovery programs, as well as opportunities for researchers to investigate the biology of exo-erythrocytic forms.
Nature | 2013
Case W. McNamara; Marcus C. S. Lee; Chek Shik Lim; Siau Hoi Lim; Jason Roland; Advait Nagle; Oliver Simon; Bryan K. S. Yeung; Arnab K. Chatterjee; Susan McCormack; Micah J. Manary; Anne-Marie Zeeman; Koen J. Dechering; T. R. Santha Kumar; Philipp P. Henrich; Kerstin Gagaring; Maureen Ibanez; Nobutaka Kato; Kelli Kuhen; Christoph Fischli; Matthias Rottmann; David Plouffe; Badry Bursulaya; Stephan Meister; Lucia E. Rameh; Joerg Trappe; Dorothea Haasen; Martijn Timmerman; Robert W. Sauerwein; Rossarin Suwanarusk
Achieving the goal of malaria elimination will depend on targeting Plasmodium pathways essential across all life stages. Here we identify a lipid kinase, phosphatidylinositol-4-OH kinase (PI(4)K), as the target of imidazopyrazines, a new antimalarial compound class that inhibits the intracellular development of multiple Plasmodium species at each stage of infection in the vertebrate host. Imidazopyrazines demonstrate potent preventive, therapeutic, and transmission-blocking activity in rodent malaria models, are active against blood-stage field isolates of the major human pathogens P. falciparum and P. vivax, and inhibit liver-stage hypnozoites in the simian parasite P. cynomolgi. We show that imidazopyrazines exert their effect through inhibitory interaction with the ATP-binding pocket of PI(4)K, altering the intracellular distribution of phosphatidylinositol-4-phosphate. Collectively, our data define PI(4)K as a key Plasmodium vulnerability, opening up new avenues of target-based discovery to identify drugs with an ideal activity profile for the prevention, treatment and elimination of malaria.Summary Achieving the goal of malaria elimination will depend on targeting Plasmodium pathways essential across all life stages. Here, we identify a lipid kinase, phosphatidylinositol 4-kinase (PI4K), as the target of imidazopyrazines, a novel antimalarial compound class that inhibits the intracellular development of multiple Plasmodium species at each stage of infection in the vertebrate host. Imidazopyrazines demonstrate potent preventive, therapeutic, and transmission-blocking activity in rodent malaria models, are active against blood-stage field isolates of the major human pathogens, P. falciparum and P. vivax, and inhibit liver stage hypnozoites in the simian parasite P. cynomolgi. We show that imidazopyrazines exert their effect through inhibitory interaction with the ATP-binding pocket of PI4K, altering the intracellular distribution of phosphatidylinositol 4-phosphate. Collectively, our data define PI4K as a key Plasmodium vulnerability, opening up new avenues of target-based discovery to identify drugs with an ideal activity profile for the prevention, treatment and elimination of malaria.
The New England Journal of Medicine | 2014
Nicholas J. White; Sasithon Pukrittayakamee; Aung Pyae Phyo; Ronnatrai Rueangweerayut; François Nosten; Podjanee Jittamala; Atthanee Jeeyapant; Jay Prakash Jain; Gilbert Lefèvre; Ruobing Li; Baldur Magnusson; Thierry T. Diagana; Leong Fj
BACKGROUND KAE609 (cipargamin; formerly NITD609, Novartis Institute for Tropical Diseases) is a new synthetic antimalarial spiroindolone analogue with potent, dose-dependent antimalarial activity against asexual and sexual stages of Plasmodium falciparum. METHODS We conducted a phase 2, open-label study at three centers in Thailand to assess the antimalarial efficacy, safety, and adverse-event profile of KAE609, at a dose of 30 mg per day for 3 days, in two sequential cohorts of adults with uncomplicated P. vivax malaria (10 patients) or P. falciparum malaria (11). The primary end point was the parasite clearance time. RESULTS The median parasite clearance time was 12 hours in each cohort (interquartile range, 8 to 16 hours in patients with P. vivax malaria and 10 to 16 hours in those with P. falciparum malaria). The median half-lives for parasite clearance were 0.95 hours (range, 0.68 to 2.01; interquartile range, 0.85 to 1.14) in the patients with P. vivax malaria and 0.90 hours (range, 0.68 to 1.64; interquartile range, 0.78 to 1.07) in those with P. falciparum malaria. By comparison, only 19 of 5076 patients with P. falciparum malaria (<1%) who were treated with oral artesunate in Southeast Asia had a parasite clearance half-life of less than 1 hour. Adverse events were reported in 14 patients (67%), with nausea being the most common. The adverse events were generally mild and did not lead to any discontinuations of the drug. The mean terminal half-life for the elimination of KAE609 was 20.8 hours (range, 11.3 to 37.6), supporting a once-daily oral dosing regimen. CONCLUSIONS KAE609, at dose of 30 mg daily for 3 days, cleared parasitemia rapidly in adults with uncomplicated P. vivax or P. falciparum malaria. (Funded by Novartis and others; ClinicalTrials.gov number, NCT01524341.).
Nature Medicine | 2014
Laurent Dembélé; Jean-François Franetich; Audrey Lorthiois; Audrey Gego; Anne-Marie Zeeman; Clemens H. M. Kocken; Roger Le Grand; Nathalie Dereuddre-Bosquet; Geert-Jan van Gemert; Robert W. Sauerwein; Jean-Christophe Vaillant; Laurent Hannoun; Matthew J. Fuchter; Thierry T. Diagana; Nicholas A. Malmquist; Artur Scherf; Georges Snounou; Dominique Mazier
Malaria relapses, resulting from the activation of quiescent hepatic hypnozoites of Plasmodium vivax and Plasmodium ovale, hinder global efforts to control and eliminate malaria. As primaquine, the only drug capable of eliminating hypnozoites, is unsuitable for mass administration, an alternative drug is needed urgently. Currently, analyses of hypnozoites, including screening of compounds that would eliminate them, can only be made using common macaque models, principally Macaca rhesus and Macaca fascicularis, experimentally infected with the relapsing Plasmodium cynomolgi. Here, we present a protocol for long-term in vitro cultivation of P. cynomolgi–infected M. fascicularis primary hepatocytes during which hypnozoites persist and activate to resume normal development. In a proof-of-concept experiment, we obtained evidence that exposure to an inhibitor of histone modification enzymes implicated in epigenetic control of gene expression induces an accelerated rate of hypnozoite activation. The protocol presented may further enable investigations of hypnozoite biology and the search for compounds that kill hypnozoites or disrupt their quiescence.
Antimicrobial Agents and Chemotherapy | 2012
J.C. van Pelt-Koops; Helmi Pett; Wouter Graumans; M.G. van de Vegte-Bolmer; G.J.A. van Gemert; Matthias Rottmann; Bryan K. S. Yeung; Thierry T. Diagana; Robert W. Sauerwein
ABSTRACT The global malaria agenda has undergone a reorientation from control of clinical cases to entirely eradicating malaria. For that purpose, a key objective is blocking transmission of malaria parasites from humans to mosquito vectors. The new antimalarial drug candidate NITD609 was evaluated for its transmission-reducing potential and compared to a few established antimalarials (lumefantrine, artemether, primaquine), using a suite of in vitro assays. By the use of a microscopic readout, NITD609 was found to inhibit the early and late development of Plasmodium falciparum gametocytes in vitro in a dose-dependent fashion over a range of 5 to 500 nM. In addition, using the standard membrane feeding assay, NITD609 was also found to be a very effective drug in reducing transmission to the Anopheles stephensi mosquito vector. Collectively, our data suggest a strong transmission-reducing effect of NITD609 acting against different P. falciparum transmission stages.
Journal of Medicinal Chemistry | 2011
Tao Wu; Advait Nagle; Kelli Kuhen; Kerstin Gagaring; Rachel Borboa; Caroline Francek; Zhong Chen; David Plouffe; Anne Goh; Suresh B. Lakshminarayana; Jeanette Wu; Hui Qing Ang; Peiting Zeng; Min Low Kang; William Tan; Maria Tan; Nicole Ye; Xuena Lin; Christopher Caldwell; Jared Ek; Suzanne Skolnik; Fenghua Liu; Jianling Wang; Jonathan Chang; Chun Li; Thomas Hollenbeck; Tove Tuntland; John Isbell; Christoph Fischli; Reto Brun
Starting from a hit series from a GNF compound library collection and based on a cell-based proliferation assay of Plasmodium falciparum, a novel imidazolopiperazine scaffold was optimized. SAR for this series of compounds is discussed, focusing on optimization of cellular potency against wild-type and drug resistant parasites and improvement of physiochemical and pharmacokinetic properties. The lead compounds in this series showed good potencies in vitro and decent oral exposure levels in vivo. In a Plasmodium berghei mouse infection model, one lead compound lowered the parasitemia level by 99.4% after administration of 100 mg/kg single oral dose and prolonged mice survival by an average of 17.0 days. The lead compounds were also well-tolerated in the preliminary in vitro toxicity studies and represents an interesting lead for drug development.
Antimicrobial Agents and Chemotherapy | 2014
Kelli Kuhen; Arnab K. Chatterjee; Matthias Rottmann; Kerstin Gagaring; Rachel Borboa; Jennifer Buenviaje; Zhong Chen; Carolyn Francek; Tao Wu; Advait Nagle; S. Whitney Barnes; David Plouffe; Marcus C. S. Lee; David A. Fidock; Wouter Graumans; Marga van de Vegte-Bolmer; Geert Jan van Gemert; Grennady Wirjanata; Boni F. Sebayang; Jutta Marfurt; Bruce Russell; Rossarin Suwanarusk; Ric N. Price; François Nosten; Anchalee Tungtaeng; Montip Gettayacamin; Jetsumon Sattabongkot; Jennifer Taylor; John R. Walker; David C. Tully
ABSTRACT Renewed global efforts toward malaria eradication have highlighted the need for novel antimalarial agents with activity against multiple stages of the parasite life cycle. We have previously reported the discovery of a novel class of antimalarial compounds in the imidazolopiperazine series that have activity in the prevention and treatment of blood stage infection in a mouse model of malaria. Consistent with the previously reported activity profile of this series, the clinical candidate KAF156 shows blood schizonticidal activity with 50% inhibitory concentrations of 6 to 17.4 nM against P. falciparum drug-sensitive and drug-resistant strains, as well as potent therapeutic activity in a mouse models of malaria with 50, 90, and 99% effective doses of 0.6, 0.9, and 1.4 mg/kg, respectively. When administered prophylactically in a sporozoite challenge mouse model, KAF156 is completely protective as a single oral dose of 10 mg/kg. Finally, KAF156 displays potent Plasmodium transmission blocking activities both in vitro and in vivo. Collectively, our data suggest that KAF156, currently under evaluation in clinical trials, has the potential to treat, prevent, and block the transmission of malaria.
Journal of Medicinal Chemistry | 2012
Advait Nagle; Tao Wu; Kelli Kuhen; Kerstin Gagaring; Rachel Borboa; Caroline Francek; Zhong Chen; David Plouffe; Xuena Lin; Christopher Caldwell; Jared Ek; Suzanne Skolnik; Fenghua Liu; Jianling Wang; Jonathan Chang; Chun Li; Bo Liu; Thomas Hollenbeck; Tove Tuntland; John Isbell; Tiffany Chuan; Philip B. Alper; Christoph Fischli; Reto Brun; Suresh B. Lakshminarayana; Matthias Rottmann; Thierry T. Diagana; Elizabeth A. Winzeler; Richard Glynne; David C. Tully
On the basis of the initial success of optimization of a novel series of imidazolopiperazines, a second generation of compounds involving changes in the core piperazine ring was synthesized to improve antimalarial properties. These changes were carried out to further improve the potency and metabolic stability of the compounds by leveraging the outcome of a set of in vitro metabolic identification studies. The optimized 8,8-dimethyl imidazolopiperazine analogues exhibited improved potency, in vitro metabolic stability profile and, as a result, enhanced oral exposure in vivo in mice. The optimized compounds were found to be more efficacious than the current antimalarials in a malaria mouse model. They exhibit moderate oral exposure in rat pharmacokinetic studies to achieve sufficient multiples of the oral exposure at the efficacious dose in toxicology studies.