Network


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

Hotspot


Dive into the research topics where Richard E. Lee is active.

Publication


Featured researches published by Richard E. Lee.


Advanced Drug Delivery Reviews | 2016

New agents for the treatment of drug-resistant Mycobacterium tuberculosis.

Daniel T. Hoagland; Jiuyu Liu; Robin B. Lee; Richard E. Lee

Inadequate dosing and incomplete treatment regimens, coupled with the ability of the tuberculosis bacilli to cause latent infections that are tolerant of currently used drugs, have fueled the rise of multidrug-resistant tuberculosis (MDR-TB). Treatment of MDR-TB infections is a major clinical challenge that has few viable or effective solutions; therefore patients face a poor prognosis and years of treatment. This review focuses on emerging drug classes that have the potential for treating MDR-TB and highlights their particular strengths as leads including their mode of action, in vivo efficacy, and key medicinal chemistry properties. Examples include the newly approved drugs bedaquiline and delaminid, and other agents in clinical and late preclinical development pipeline for the treatment of MDR-TB. Herein, we discuss the challenges to developing drugs to treat tuberculosis and how the field has adapted to these difficulties, with an emphasis on drug discovery approaches that might produce more effective agents and treatment regimens.


Science | 2012

Catalysis and sulfa drug resistance in dihydropteroate synthase.

Mi-Kyung Yun; Yinan Wu; Zhenmei Li; Ying Zhao; M.B Waddell; A.M Ferreira; Richard E. Lee; Donald Bashford; Stephen W. White

Sulfas Crystal View The sulfonamide antibiotics (sulfa drugs) have been used to treat infections for over 70 years; however, emerging resistance has eroded their clinical utility. Sulfa drugs target dihydropteroate synthase, a key enzyme in the bacterial folate pathway. By performing the reaction in the crystalline form of the enzyme, Yun et al. (p. 1110) have characterized the key structural intermediates. In combining structural data with theoretical and mutagenesis studies, they propose a detailed mechanism for dihydropteroate synthase catalysis. By resolving this structure with a sulfa drug bound to the enzyme, they showed how inhibition occurred and indicated how resistance could emerge. Structures of a target enzyme in the bacteria that cause anthrax and bubonic plague may lead to effective drugs. The sulfonamide antibiotics inhibit dihydropteroate synthase (DHPS), a key enzyme in the folate pathway of bacteria and primitive eukaryotes. However, resistance mutations have severely compromised the usefulness of these drugs. We report structural, computational, and mutagenesis studies on the catalytic and resistance mechanisms of DHPS. By performing the enzyme-catalyzed reaction in crystalline DHPS, we have structurally characterized key intermediates along the reaction pathway. Results support an SN1 reaction mechanism via formation of a novel cationic pterin intermediate. We also show that two conserved loops generate a substructure during catalysis that creates a specific binding pocket for p-aminobenzoic acid, one of the two DHPS substrates. This substructure, together with the pterin-binding pocket, explains the roles of the conserved active-site residues and reveals how sulfonamide resistance arises.


PLOS ONE | 2013

Pantothenamides are potent, on-target inhibitors of plasmodium falciparum growth when serum pantetheinase is inactivated

Christina Spry; Zhiyang Lin; Kristopher G. Virga; Richard E. Lee; Erick Strauss; Kevin J. Saliba

Growth of the virulent human malaria parasite Plasmodium falciparum is dependent on an extracellular supply of pantothenate (vitamin B5) and is susceptible to inhibition by pantothenate analogues that hinder pantothenate utilization. In this study, on the hunt for pantothenate analogues with increased potency relative to those reported previously, we screened a series of pantothenamides (amide analogues of pantothenate) against P. falciparum and show for the first time that analogues of this type possess antiplasmodial activity. Although the active pantothenamides in this series exhibit only modest potency under standard in vitro culture conditions, we show that the potency of pantothenamides is selectively enhanced when the parasite culture medium is pre-incubated at 37°C for a prolonged period. We present evidence that this finding is linked to the presence in Albumax II (a serum-substitute routinely used for in vitro cultivation of P. falciparum) of pantetheinase activity: the activity of an enzyme that hydrolyzes the pantothenate metabolite pantetheine, for which pantothenamides also serve as substrates. Pantetheinase activity, and thereby pantothenamide degradation, is reduced following incubation of Albumax II-containing culture medium for a prolonged period at 37°C, revealing the true, sub-micromolar potency of pantothenamides. Importantly we show that the potent antiplasmodial effect of pantothenamides is attenuated with pantothenate, consistent with the compounds inhibiting parasite proliferation specifically by inhibiting pantothenate and/or CoA utilization. Additionally, we show that the pantothenamides interact with P. falciparum pantothenate kinase, the first enzyme involved in converting pantothenate to coenzyme A. This is the first demonstration of on-target antiplasmodial pantothenate analogues with sub-micromolar potency, and highlights the potential of pantetheinase-resistant pantothenamides as antimalarial agents.


Bioorganic & Medicinal Chemistry Letters | 2011

Antibacterial and Antitubercular Activity of Fosmidomycin, FR900098, and their Lipophilic Analogs

Eugene Uh; Emily R. Jackson; Géraldine San Jose; Marcus M. Maddox; Robin E. B. Lee; Richard E. Lee; Helena I. Boshoff; Cynthia S. Dowd

The nonmevalonate pathway (NMP) of isoprene biosynthesis is an exciting new route toward novel antibiotic development. Inhibitors against several enzymes in this pathway are currently under examination. A significant liability of many of these agents is poor cell penetration. To overcome and improve our understanding of this problem, we have synthesized a series of lipophilic, prodrug analogs of fosmidomycin and FR900098, inhibitors of the NMP enzyme Dxr. Several of these compounds show improved antibacterial activity against a panel of organisms relative to the parent compound, including activity against Mycobacterium tuberculosis (Mtb). Our results show that this strategy can be an effective way for improving whole cell activity of NMP inhibitors.


Antimicrobial Agents and Chemotherapy | 2016

Therapeutic potential of the Mycobacterium tuberculosis mycolic acid transporter, MmpL3

Wei Li; Andrés Obregón-Henao; Joshua B. Wallach; E. Jeffrey North; Richard E. Lee; Mercedes Gonzalez-Juarrero; Dirk Schnappinger; Mary Jackson

ABSTRACT In recent years, whole-cell-based screens for novel small molecule inhibitors active against Mycobacterium tuberculosis in culture followed by the whole-genome sequencing of spontaneous resistant mutants have identified multiple chemical scaffolds thought to kill the bacterium through the inactivation of the mycolic acid transporter, MmpL3. Consistent with the fact that MmpL3 is required for the formation of the mycobacterial outer membrane, we have conclusively shown in this study, using conditionally regulated knockdown mutants, that mmpL3 is required for the replication and viability of M. tuberculosis, both under standard laboratory growth conditions and during the acute and chronic phases of infection in mice. Speaking for the vulnerability of this target, silencing mmpL3 had a rapid bactericidal effect on actively replicating cells in vitro and reduced by 3 to 5 logs in less than 4 weeks the bacterial loads of acutely and chronically infected mouse lungs, respectively. Depletion of MmpL3 further rendered M. tuberculosis hypersusceptible to MmpL3 inhibitors. The exquisite vulnerability of MmpL3 at all stages of the infection establishes this transporter as an attractive new target with the potential to improve and shorten current drug-susceptible and drug-resistant tuberculosis chemotherapies.


Bioorganic & Medicinal Chemistry | 2012

Antitubercular nitrofuran isoxazolines with improved pharmacokinetic properties.

Rakesh; David F. Bruhn; Dora B. Madhura; Marcus M. Maddox; Robin B. Lee; Ashit Trivedi; Lei Yang; Michael S. Scherman; Janet C. Gilliland; Veronica Gruppo; Michael R. McNeil; Anne J. Lenaerts; Bernd Meibohm; Richard E. Lee

A series of tetracyclic nitrofuran isoxazoline anti-tuberculosis agents was designed and synthesized to improve the pharmacokinetic properties of an initial lead compound, which had potent anti-tuberculosis activity but suffered from poor solubility, high protein binding and rapid metabolism. In this study, structural modifications were carried on the outer phenyl and piperidine rings to introduce solubilizing and metabolically blocking functional groups. The compounds generated were evaluated for their in vitro antitubercular activity, bacterial spectrum of activity, solubility, permeability, microsomal stability and protein binding. Pharmacokinetic profiles for the most promising candidates were then determined. Compounds with phenyl morpholine and pyridyl morpholine outer rings were found to be the most potent anti-tuberculosis agents in the series. These compounds retained a narrow antibacterial spectrum of activity, with weak anti-Gram positive and no Gram negative activity, as well as good activity against non-replicating Mycobacterium tuberculosis in a low oxygen model. Overall, the addition of solubilizing and metabolically blocked outer rings did improve solubility and decrease protein binding as designed. However, the metabolic stability for compounds in this series was generally lower than desired. The best three compounds selected for in vivo pharmacokinetic testing all showed high oral bioavailability, with one notable compound showing a significantly longer half-life and good tolerability supporting its further advancement.


ChemMedChem | 2012

Structure-Based Design of Novel Pyrimido[4,5-c]pyridazine Derivatives as Dihydropteroate Synthase Inhibitors with Increased Affinity.

Ying Zhao; Dalia Hammoudeh; Mi-Kyung Yun; Jianjun Qi; Stephen W. White; Richard E. Lee

Dihydropteroate synthase (DHPS) is the validated drug target for sulfonamide antimicrobial therapy. However, due to widespread drug resistance and poor tolerance, the use of sulfonamide antibiotics is now limited. The pterin binding pocket in DHPS is highly conserved and is distinct from the sulfonamide binding site. It therefore represents an attractive alternative target for the design of novel antibacterial agents. We previously carried out the structural characterization of a known pyridazine inhibitor in the Bacillus anthracis DHPS pterin site and identified a number of unfavorable interactions that appear to compromise binding. With this structural information, a series of 4,5‐dioxo‐1,4,5,6‐tetrahydropyrimido[4,5‐c]pyridazines were designed to improve binding affinity. Most importantly, the N‐methyl ring substitution was removed to improve binding within the pterin pocket, and the length of the side chain carboxylic acid was optimized to fully engage the pyrophosphate binding site. These inhibitors were synthesized and evaluated by an enzyme activity assay, X‐ray crystallography, isothermal calorimetry, and surface plasmon resonance to obtain a comprehensive understanding of the binding interactions from structural, kinetic, and thermodynamic perspectives. This study clearly demonstrates that compounds lacking the N‐methyl substitution exhibit increased inhibition of DHPS, but the beneficial effects of optimizing the side chain length are less apparent.


Journal of Antimicrobial Chemotherapy | 2011

Reutericyclin and related analogues kill stationary phase Clostridium difficile at achievable colonic concentrations

Julian G. Hurdle; Amy E. Heathcott; Lei Yang; Bing Yan; Richard E. Lee

OBJECTIVES The stationary phase of Clostridium difficile, which is associated with the symptoms of the diarrhoeal disease, is refractory to antibiotic killing. The aim of this study was to explore whether probiotic-derived reutericyclin and related synthetic analogues could kill stationary phase C. difficile at concentrations achievable in the gastrointestinal tract. METHODS The bactericidal activities of reutericyclin and lead compound derivatives were examined against logarithmic and stationary phase cultures of different C. difficile strains. The absorption of compounds across the intestinal epithelia was tested using the Caco-2 permeability model. RESULTS Unlike vancomycin and metronidazole, reutericyclins demonstrated concentration-dependent killing, being rapidly bactericidal against both logarithmic and stationary phase cells, at low concentrations (0.09-2 mg/L). The intestinal absorption of unmodified reutericyclin was poor and comparable to that of vancomycin. However, this property varied significantly for the synthetic reutericyclin analogues, ranging from well absorbed to non-absorbed. The non-absorbable compounds were highly effluxed, suggesting this parameter could be modulated to obtain agents with superior efficacy. CONCLUSIONS Reutericyclins showed excellent potency against the lethal non-growing stage of C. difficile at concentrations that may be attained in the gastrointestinal tract. Since these agents represent novel potential treatments for C. difficile infection, further development of this compound class is warranted.


ChemMedChem | 2012

Evaluation of Flavonoid and Resveratrol Chemical Libraries Reveals Abyssinone II as a Promising Antibacterial Lead

Dianqing Sun; Julian G. Hurdle; Robin E. B. Lee; Richard E. Lee; Mark Cushman; John M. Pezzuto

Lead on! In the course of screening flavonoid and resveratrol libraries, abyssinone II, a naturally occurring prenylated flavonoid, was found to exhibit relatively good antitubercular and antibacterial activity. Preliminary mechanistic studies revealed that abyssinone II hyperpolarizes the bacterial membrane potential and inhibits the biosynthesis of key cellular macromolecules (DNA, RNA, and protein).


PLOS ONE | 2010

Crystal Structure of the 6-Hydroxymethyl-7,8-Dihydropterin Pyrophosphokinase•Dihydropteroate Synthase Bifunctional Enzyme from Francisella tularensis

Charles W. Pemble; Perdeep K. Mehta; Smriti Mehra; Zhenmei Li; Amanda Nourse; Richard E. Lee; Stephen W. White

The 6-hydroxymethyl-7,8-dihydropterin pyrophosphokinase (HPPK) and dihydropteroate synthase (DHPS) enzymes catalyze sequential metabolic reactions in the folate biosynthetic pathway of bacteria and lower eukaryotes. Both enzymes represent validated targets for the development of novel anti-microbial therapies. We report herein that the genes which encode FtHPPK and FtDHPS from the biowarfare agent Francisella tularensis are fused into a single polypeptide. The potential of simultaneously targeting both modules with pterin binding inhibitors prompted us to characterize the molecular details of the multifunctional complex. Our high resolution crystallographic analyses reveal the structural organization between FtHPPK and FtDHPS which are tethered together by a short linker. Additional structural analyses of substrate complexes reveal that the active sites of each module are virtually indistinguishable from those of the monofunctional enzymes. The fused bifunctional enzyme therefore represents an excellent vehicle for finding inhibitors that engage the pterin binding pockets of both modules that have entirely different architectures. To demonstrate that this approach has the potential of producing novel two-hit inhibitors of the folate pathway, we identify and structurally characterize a fragment-like molecule that simultaneously engages both active sites. Our study provides a molecular framework to study the enzyme mechanisms of HPPK and DHPS, and to design novel and much needed therapeutic compounds to treat infectious diseases.

Collaboration


Dive into the Richard E. Lee's collaboration.

Top Co-Authors

Avatar

David F. Bruhn

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Jiuyu Liu

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Aman P. Singh

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Julian G. Hurdle

University of Texas at Arlington

View shared research outputs
Top Co-Authors

Avatar

Stephen W. White

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Charles O. Rock

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Mi-Kyung Yun

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bernd Meibohm

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge