Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Tove Tuntland is active.

Publication


Featured researches published by Tove Tuntland.


Nature | 2010

Targeting Bcr-Abl by combining allosteric with ATP-binding-site inhibitors

Jianming Zhang; Francisco Adrian; Wolfgang Jahnke; Sandra W. Cowan-Jacob; Allen Li; Roxana E. Iacob; Taebo Sim; John T. Powers; Christine Dierks; Fangxian Sun; Gui Rong Guo; Qiang Ding; Barun Okram; Yongmun Choi; Amy Wojciechowski; Xianming Deng; Guoxun Liu; Gabriele Fendrich; André Strauss; Navratna Vajpai; Stephan Grzesiek; Tove Tuntland; Yi Liu; Badry Bursulaya; Mohammad Azam; Paul W. Manley; John R. Engen; George Q. Daley; Markus Warmuth; Nathanael S. Gray

In an effort to find new pharmacological modalities to overcome resistance to ATP-binding-site inhibitors of Bcr–Abl, we recently reported the discovery of GNF-2, a selective allosteric Bcr–Abl inhibitor. Here, using solution NMR, X-ray crystallography, mutagenesis and hydrogen exchange mass spectrometry, we show that GNF-2 binds to the myristate-binding site of Abl, leading to changes in the structural dynamics of the ATP-binding site. GNF-5, an analogue of GNF-2 with improved pharmacokinetic properties, when used in combination with the ATP-competitive inhibitors imatinib or nilotinib, suppressed the emergence of resistance mutations in vitro, displayed additive inhibitory activity in biochemical and cellular assays against T315I mutant human Bcr–Abl and displayed in vivo efficacy against this recalcitrant mutant in a murine bone-marrow transplantation model. These results show that therapeutically relevant inhibition of Bcr–Abl activity can be achieved with inhibitors that bind to the myristate-binding site and that combining allosteric and ATP-competitive inhibitors can overcome resistance to either agent alone.


Journal of Medicinal Chemistry | 2013

Synthesis, structure-activity relationships, and in vivo efficacy of the novel potent and selective anaplastic lymphoma kinase (ALK) inhibitor 5-chloro-N2-(2-isopropoxy-5-methyl-4-(piperidin-4-yl)phenyl)-N4-(2-(isopropylsulfonyl)phenyl)pyrimidine-2,4-diamine (LDK378) currently in phase 1 and phase 2 clinical trials.

Thomas H. Marsilje; Wei Pei; Bei Chen; Wenshuo Lu; Tetsuo Uno; Yunho Jin; Tao Jiang; Sung Joon Kim; Nanxin Li; Markus Warmuth; Yelena Sarkisova; Frank Sun; Auzon Steffy; AnneMarie C. Pferdekamper; Allen Li; Sean B. Joseph; Young Chul Kim; Bo Liu; Tove Tuntland; Xiaoming Cui; Nathanael S. Gray; Ruo Steensma; Yongqin Wan; Jiqing Jiang; Greg Chopiuk; Jie Li; W. Perry Gordon; Wendy Richmond; Kevin Johnson; Jonathan Chang

The synthesis, preclinical profile, and in vivo efficacy in rat xenograft models of the novel and selective anaplastic lymphoma kinase inhibitor 15b (LDK378) are described. In this initial report, preliminary structure-activity relationships (SARs) are described as well as the rational design strategy employed to overcome the development deficiencies of the first generation ALK inhibitor 4 (TAE684). Compound 15b is currently in phase 1 and phase 2 clinical trials with substantial antitumor activity being observed in ALK-positive cancer patients.


Science | 2011

Imaging of Plasmodium liver stages to drive next-generation antimalarial drug discovery

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 Chemical Biology | 2008

Gene expression signatures and small-molecule compounds link a protein kinase to Plasmodium falciparum motility.

Nobutaka Kato; Tomoyo Sakata; Ghislain Breton; Karine G. Le Roch; Advait Nagle; Carsten B Andersen; Badry Bursulaya; Kerstin Henson; Jeffrey R. Johnson; Kota Arun Kumar; Felix Marr; Daniel E. Mason; Case W. McNamara; David Plouffe; Muriel Spooner; Tove Tuntland; Yingyao Zhou; Eric C. Peters; Arnab K. Chatterjee; Peter G. Schultz; Gary E. Ward; Nathanael S. Gray; Jeffrey F. Harper; Elizabeth A. Winzeler

Calcium-dependent protein kinases play a crucial role in intracellular calcium signaling in plants, some algae and protozoa. In Plasmodium falciparum, calcium-dependent protein kinase 1 (PfCDPK1) is expressed during schizogony in the erythrocytic stage as well as in the sporozoite stage. It is coexpressed with genes that encode the parasite motor complex, a cellular component required for parasite invasion of host cells, parasite motility and potentially cytokinesis. A targeted gene-disruption approach demonstrated that pfcdpk1 seems to be essential for parasite viability. An in vitro biochemical screen using recombinant PfCDPK1 against a library of 20,000 compounds resulted in the identification of a series of structurally related 2,6,9-trisubstituted purines. Compound treatment caused sudden developmental arrest at the late schizont stage in P. falciparum and a large reduction in intracellular parasites in Toxoplasma gondii, which suggests a possible role for PfCDPK1 in regulation of parasite motility during egress and invasion.


Bioorganic & Medicinal Chemistry Letters | 2006

Design, synthesis, and biological evaluations of novel oxindoles as HIV-1 non-nucleoside reverse transcriptase inhibitors. Part 2.

Tao Jiang; Kelli Kuhen; Karen Wolff; Hong Yin; Kimberly Bieza; Jeremy S. Caldwell; Badry Bursulaya; Tove Tuntland; Kanyin Zhang; Donald S. Karanewsky; Yun He

A series of heterocycle-containing oxindoles was synthesized and their HIV antiviral activities were assessed. Some of these analogs exhibited potent inhibitory activities against both wild-type virus and a number of drug-resistant mutant viruses. In addition, oxindole 9z also showed promising pharmacokinetics.


Proceedings of the National Academy of Sciences of the United States of America | 2010

A small molecule accelerates neuronal differentiation in the adult rat.

Heiko Wurdak; Shoutian Zhu; Kyung-Hoon Min; Lindsey Aimone; Luke L. Lairson; James E. M. Watson; Gregory Chopiuk; James Demas; Bradley D. Charette; Rajkumar Halder; Eranthie Weerapana; Benjamin F. Cravatt; Hollis T. Cline; Eric C. Peters; Jay Zhang; John R. Walker; Chunlei Wu; Jonathan Chang; Tove Tuntland; Charles Y. Cho; Peter G. Schultz

Adult neurogenesis occurs in mammals and provides a mechanism for continuous neural plasticity in the brain. However, little is known about the molecular mechanisms regulating hippocampal neural progenitor cells (NPCs) and whether their fate can be pharmacologically modulated to improve neural plasticity and regeneration. Here, we report the characterization of a small molecule (KHS101) that selectively induces a neuronal differentiation phenotype. Mechanism of action studies revealed a link of KHS101 to cell cycle exit and specific binding to the TACC3 protein, whose knockdown in NPCs recapitulates the KHS101-induced phenotype. Upon systemic administration, KHS101 distributed to the brain and resulted in a significant increase in neuronal differentiation in vivo. Our findings indicate that KHS101 accelerates neuronal differentiation by interaction with TACC3 and may provide a basis for pharmacological intervention directed at endogenous NPCs.


Nature | 2016

Proteasome inhibition for treatment of leishmaniasis, Chagas disease and sleeping sickness

Shilpi Khare; Advait Nagle; Agnes Biggart; Yin H. Lai; Fang Liang; Lauren C. Davis; S. Whitney Barnes; Casey J. N. Mathison; Elmarie Myburgh; Mu-Yun Gao; J. Robert Gillespie; Xianzhong Liu; Jocelyn L. Tan; Monique Stinson; Ianne Rivera; Jaime Ballard; Vince Yeh; Todd Groessl; Hazel X. Y. Koh; John D. Venable; Badry Bursulaya; Michael B. Shapiro; Pranab Mishra; Glen Spraggon; Ansgar Brock; Jeremy C. Mottram; Frederick S. Buckner; Srinivasa P. S. Rao; Ben G. Wen; John R. Walker

Chagas disease, leishmaniasis and sleeping sickness affect 20 million people worldwide and lead to more than 50,000 deaths annually. The diseases are caused by infection with the kinetoplastid parasites Trypanosoma cruzi, Leishmania spp. and Trypanosoma brucei spp., respectively. These parasites have similar biology and genomic sequence, suggesting that all three diseases could be cured with drugs that modulate the activity of a conserved parasite target. However, no such molecular targets or broad spectrum drugs have been identified to date. Here we describe a selective inhibitor of the kinetoplastid proteasome (GNF6702) with unprecedented in vivo efficacy, which cleared parasites from mice in all three models of infection. GNF6702 inhibits the kinetoplastid proteasome through a non-competitive mechanism, does not inhibit the mammalian proteasome or growth of mammalian cells, and is well-tolerated in mice. Our data provide genetic and chemical validation of the parasite proteasome as a promising therapeutic target for treatment of kinetoplastid infections, and underscore the possibility of developing a single class of drugs for these neglected diseases.


Journal of Medicinal Chemistry | 2011

Imidazolopiperazines: hit to lead optimization of new antimalarial agents.

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.


Journal of Medicinal Chemistry | 2012

Imidazolopiperazines:Lead Optimization of the Second-Generation Antimalarial Agents

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.


Drug Discovery Today | 2008

Snapshot PK: a rapid rodent in vivo preclinical screening approach

Bo Liu; Jonathan Chang; William Perry Gordon; John Isbell; Yingyao Zhou; Tove Tuntland

Described in this article are strategies implemented to increase the throughput of in vivo rodent pharmacokinetic (PK) studies using the snapshot PK study design and automated methods for compound submission, sample processing, data analysis and reporting. Applying snapshot PK studies to categorize the oral exposure of >1300 discovery compounds as low, moderate or high resulted in an attrition rate of 86%. The follow up full PK studies on the remaining compounds found that 98% of the compounds were predicted in the correct (69%) or adjacent (29%) oral exposure category by the snapshot PK studies. These results demonstrate that the snapshot PK screen in rodents can serve as an effective and efficient in vivo tool in the compound selection process in drug discovery.

Collaboration


Dive into the Tove Tuntland's collaboration.

Top Co-Authors

Avatar

Jonathan Chang

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Bo Liu

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Arnab K. Chatterjee

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

David C. Tully

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Jennifer L. Harris

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Donald S. Karanewsky

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

H. Martin Seidel

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Thomas Hollenbeck

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge