Network


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

Hotspot


Dive into the research topics where Frederick S. Buckner is active.

Publication


Featured researches published by Frederick S. Buckner.


Nature Reviews Drug Discovery | 2008

Genomic-scale prioritization of drug targets: the TDR Targets database

Fernán Agüero; Bissan Al-Lazikani; Martin Aslett; Matthew Berriman; Frederick S. Buckner; Robert K. Campbell; Santiago J. Carmona; Ian M. Carruthers; A.W. Edith Chan; Feng Chen; Gregory J. Crowther; Maria A. Doyle; Christiane Hertz-Fowler; Andrew L. Hopkins; Gregg McAllister; Solomon Nwaka; John P. Overington; Arnab Pain; Gaia V. Paolini; Ursula Pieper; Stuart A. Ralph; Aaron Riechers; David S. Roos; Andrej Sali; Dhanasekaran Shanmugam; Takashi Suzuki; Wesley C. Van Voorhis; Christophe L. M. J. Verlinde

The increasing availability of genomic data for pathogens that cause tropical diseases has created new opportunities for drug discovery and development. However, if the potential of such data is to be fully exploited, the data must be effectively integrated and be easy to interrogate. Here, we discuss the development of the TDR Targets database (http://tdrtargets.org), which encompasses extensive genetic, biochemical and pharmacological data related to tropical disease pathogens, as well as computationally predicted druggability for potential targets and compound desirability information. By allowing the integration and weighting of this information, this database aims to facilitate the identification and prioritization of candidate drug targets for pathogens.


Molecular and Biochemical Parasitology | 2003

Protein farnesyl and N-myristoyl transferases: piggy-back medicinal chemistry targets for the development of antitrypanosomatid and antimalarial therapeutics

Michael H. Gelb; Wesley C. Van Voorhis; Frederick S. Buckner; Kohei Yokoyama; Richard T. Eastman; Elisabeth P. Carpenter; Chrysoula Panethymitaki; Katherine A. Brown; Deborah F. Smith

To accelerate progress in the development of therapeutics for protozoan parasitic diseases, we are studying enzymes active in co- and post-translational protein modification that are already the focus of drug development in other eukaryotic systems. Inhibitors of the protein farnesyltransferases (PFT) are well-established antitumour agents of low cytotoxicity and known pharmokinetic properties, while inhibitors of N-myristoyl transferase show both selectivity and specificity in the treatment of fungal infections. Here, we summarise the current evidence that supports the targeting of these ubiquitous eukaryotic enzymes for drug development against trypanosomatid infections and malaria.


Nature Structural & Molecular Biology | 2010

Toxoplasma gondii calcium-dependent protein kinase 1 is a target for selective kinase inhibitors.

Kayode K. Ojo; Eric T. Larson; Katelyn R. Keyloun; Lisa J. Castaneda; Amy E. DeRocher; Krishna K Inampudi; Jessica E. Kim; Tracy L. Arakaki; Ryan C. Murphy; Li Zhang; Alberto J. Napuli; Dustin J. Maly; Christophe L. M. J. Verlinde; Frederick S. Buckner; Marilyn Parsons; Wim G. J. Hol; Ethan A. Merritt; Wesley C. Van Voorhis

New drugs are needed to treat toxoplasmosis. Toxoplasma gondii calcium-dependent protein kinases (TgCDPKs) are attractive targets because they are absent in mammals. We show that TgCDPK1 is inhibited by low nanomolar levels of bumped kinase inhibitors (BKIs), compounds inactive against mammalian kinases. Cocrystal structures of TgCDPK1 with BKIs confirm that the structural basis for selectivity is due to the unique glycine gatekeeper residue in the ATP-binding site. We show that BKIs interfere with an early step in T. gondii infection of human cells in culture. Furthermore, we show that TgCDPK1 is the in vivo target of BKIs because T. gondii expressing a glycine to methionine gatekeeper mutant enzyme show significantly decreased sensitivity to BKIs. Thus, design of selective TgCDPK1 inhibitors with low host toxicity may be achievable.


Journal of Medicinal Chemistry | 2009

Identification of a metabolically stable triazolopyrimidine-based dihydroorotate dehydrogenase inhibitor with antimalarial activity in mice.

Ramesh Gujjar; Alka Marwaha; Farah El Mazouni; John Kenneth White; Karen L. White; Sharon A. Creason; David M. Shackleford; Jeffrey Baldwin; William N. Charman; Frederick S. Buckner; Susan A. Charman; Pradip Rathod; Margaret A. Phillips

Plasmodium falciparum causes 1-2 million deaths annually. Yet current drug therapies are compromised by resistance. We previously described potent and selective triazolopyrimidine-based inhibitors of P. falciparum dihydroorotate dehydrogenase (PfDHODH) that inhibited parasite growth in vitro; however, they showed no activity in vivo. Here we show that lack of efficacy against P. berghei in mice resulted from a combination of poor plasma exposure and reduced potency against P. berghei DHODH. For compounds containing naphthyl (DSM1) or anthracenyl (DSM2), plasma exposure was reduced upon repeated dosing. Phenyl-substituted triazolopyrimidines were synthesized leading to identification of analogs with low predicted metabolism in human liver microsomes and which showed prolonged exposure in mice. Compound 21 (DSM74), containing p-trifluoromethylphenyl, suppressed growth of P. berghei in mice after oral administration. This study provides the first proof of concept that DHODH inhibitors can suppress Plasmodium growth in vivo, validating DHODH as a new target for antimalarial chemotherapy.


Journal of Lipid Research | 2006

Thematic review series: Lipid Posttranslational Modifications. Fighting parasitic disease by blocking protein farnesylation

Richard T. Eastman; Frederick S. Buckner; Kohei Yokoyama; Michael H. Gelb; Wesley C. Van Voorhis

Protein farnesylation is a form of posttranslational modification that occurs in most, if not all, eukaryotic cells. Inhibitors of protein farnesyltransferase (PFTIs) have been developed as anticancer chemotherapeutic agents. Using the knowledge gained from the development of PFTIs for the treatment of cancer, researchers are currently investigating the use of PFTIs for the treatment of eukaryotic pathogens. This “piggy-back” approach not only accelerates the development of a chemotherapeutic agent for protozoan pathogens but is also a means of mitigating the costs associated with de novo drug design. PFTIs have already been shown to be efficacious in the treatment of eukaryotic pathogens in animal models, including both Trypanosoma brucei, the causative agent of African sleeping sickness, and Plasmodium falciparum, one of the causative agents of malaria. Here, current evidence and progress are summarized that support the targeting of protein farnesyltransferase for the treatment of parasitic diseases.


Journal of Medicinal Chemistry | 2009

Rational Modification of a Candidate Cancer Drug for Use Against Chagas Disease

James M. Kraus; Christophe L. M. J. Verlinde; Mandana Karimi; Galina I. Lepesheva; Michael H. Gelb; Frederick S. Buckner

Chagas disease is one of the major neglected diseases of the world. Existing drug therapies are limited, ineffective, and highly toxic. We describe a novel strategy of drug discovery of adapting an existing clinical compound with excellent pharmaceutical properties to target a pathogenic organism. The protein farnesyltransferase (PFT) inhibitor tipifarnib, now in phase III anticancer clinical trials, was previously found to kill Trypanosoma cruzi by blocking sterol 14 alpha-demethylase (14DM). We rationally developed tipifarnib analogues that display reduced affinity for human PFT to reduce toxicity while increasing affinity for parasite 14DM. The lead compound has picomolar activity against cultured T. cruzi and is efficacious in a mouse model of acute Chagas disease.


Journal of Medicinal Chemistry | 2011

Lead-optimization of aryl and aralkyl amine based triazolopyrimidine inhibitors of Plasmodium falciparum dihydroorotate dehydrogenase with antimalarial activity in mice

Ramesh Gujjar; Farah El Mazouni; Karen L. White; John Kenneth White; Sharon A. Creason; David M. Shackleford; Xiaoyi Deng; William N. Charman; Ian Bathurst; Jeremy N. Burrows; David M. Floyd; David Matthews; Frederick S. Buckner; Susan A. Charman; Margaret A. Phillips; Pradipsinh K. Rathod

Malaria is one of the leading causes of severe infectious disease worldwide; yet, our ability to maintain effective therapy to combat the illness is continually challenged by the emergence of drug resistance. We previously reported identification of a new class of triazolopyrimidine-based Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) inhibitors with antimalarial activity, leading to the discovery of a new lead series and novel target for drug development. Active compounds from the series contained a triazolopyrimidine ring attached to an aromatic group through a bridging nitrogen atom. Herein, we describe systematic efforts to optimize the aromatic functionality with the goal of improving potency and in vivo properties of compounds from the series. These studies led to the identification of two new substituted aniline moieties (4-SF(5)-Ph and 3,5-Di-F-4-CF(3)-Ph), which, when coupled to the triazolopyrimidine ring, showed good plasma exposure and better efficacy in the Plasmodium berghei mouse model of the disease than previously reported compounds from the series.


Chemical Reviews | 2014

Recent Developments in Drug Discovery for Leishmaniasis and Human African Trypanosomiasis

Advait Nagle; Shilpi Khare; Arun Babu Kumar; Frantisek Supek; Andriy Buchynskyy; Casey J. N. Mathison; Naveen Kumar Chennamaneni; Nagendar Pendem; Frederick S. Buckner; Michael H. Gelb

Leishmaniasis is a parasitic disease that presents four main clinical syndromes: cutaneous leishmaniasis (CL), mucocutaneous leishmaniasis (MCL), visceral leishmaniasis/kala azar (VL), and post kala azar dermal leishmaniasis (PKDL). Causative Leishmania are protozoan parasites that are transmitted among mammalian hosts by phlebotomine sandflies. In mammalian hosts, parasite cells proliferate inside the host phagocytic cells as round amastigotes. Infection of sandflies with Leishmania occurs during insect feeding on infected mammalian hosts. After introduction into the insect gut together with the blood meal, Leishmania amastigotes transform into elongated flagellated promastigotes that propagate in the insect gut. A new round of infection is initiated after the infected sandfly takes a blood meal from a naive mammalian host and introduces Leishmania parasites into the bite wound in the host dermis (Scheme 1). More than 20 different Leishmania species have been found to cause human leishmaniasis (Table 1). Leishmaniasis is endemic in 98 countries and is closely associated with poverty. More than a million new cases are reported per year and 350 million people are at risk of contracting the infection. For the most severe form of leishmaniasis, VL, ∼300 000 new cases are estimated to occur annually resulting in ∼40 000 deaths. Approximately 90% of all VL cases occur in 3 endemic foci: 1. India, Bangladesh, and Nepal; 2. East Africa; and 3. Brazil. In spite of the high prevalence, currently available treatments for leishmaniasis are inadequate. Pentavalent antimonials, the standard treatment for leishmaniasis for many decades, are not efficacious in Bihar (∼60% of VL cases worldwide) any longer due to widespread resistance to the drug in this region. Several new VL treatments have emerged during the past 10–15 years, but each has serious shortcomings (summarized in Table 2). These include paromomycin (injectable, long treatment, region-dependent efficacy), miltefosine (cost, teratogenicity, long treatment), and liposomal amphotericin B (cost, hospitalization, region-dependent efficacy). An additional challenge is represented by patients with HIV/VL coinfections who are more difficult to cure (lower initial and final cure rates), have greater susceptibility to drug toxicity, and have higher rates of death and relapse. Due to the limitations of the existing treatments, better drugs are urgently needed. Ideally, new VL drugs would be efficacious across all endemic regions, would affect cure in ≤10 days, and would cost <


Molecular and Biochemical Parasitology | 1998

The effects of protein farnesyltransferase inhibitors on trypanosomatids: inhibition of protein farnesylation and cell growth

Kohei Yokoyama; Patty Trobridge; Frederick S. Buckner; Jeffrey D. Scholten; Kenneth Stuart; Wesley C. Van Voorhis; Michael H. Gelb

10 per course (for a complete target product profile for new VL drugs, which was formulated by DNDi, see Table 4).1 Here we describe the disease history and parasite biology followed by a summary of the currently available treatments and, finally, review reports of novel small molecules with antileishmanial activity.


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

Attachment of the prenyl groups farnesyl and geranylgeranyl to specific eukaryotic cell proteins by protein prenyltransferases is required for the functioning of a number of cellular processes including signal transduction. In this study it was found that previously reported inhibitors of mammalian protein farnesyltransferase (PFT) [those that mimic the substrate farnesyl pyrophosphate and those that mimic the protein acceptor of the farnesyl group (CaaX mimetic)] inhibit in vitro farnesylation catalyzed by partially purified Trypanosoma brucei (T. brucei) PFT. The most potent PFT inhibitors at concentrations of 3-10 microM inhibit the growth of insect (procyclic) and bloodstream forms of T. brucei. One of the PFT inhibitors was found to block the incorporation of radiolabeled mevalonic acid (the precursor of prenyl groups) into specific T. brucei proteins. This study also shows that protein prenylation occurs in the protozoan parasites Trypanosoma cruzi (T. cruzi) and Leishmania mexicana (L. mexicana). The growth of T. cruzi intracellular form (amastigote) is also sensitive to PFT inhibitors, whereas the insect form (epimastigote) is considerably more resistant to inhibition of protein farnesylation. On the other hand, growth of 3T3 fibroblast cells (host cells for amastigote growth) was not affected by up to 100 microM PFT inhibitors. The growth of L. mexicana insect form (promastigote) is modestly inhibited by protein farnesyltransferase inhibitors. These results suggest the potential for the development of PFT inhibitors for treating trypanosomiasis and leishmaniasis.

Collaboration


Dive into the Frederick S. Buckner's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wim G. J. Hol

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Erkang Fan

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Frank Zucker

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kohei Yokoyama

University of Washington

View shared research outputs
Researchain Logo
Decentralizing Knowledge