Network


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

Hotspot


Dive into the research topics where Jianjun Qi is active.

Publication


Featured researches published by Jianjun Qi.


Nature Medicine | 2014

Spectinamides: a new class of semisynthetic antituberculosis agents that overcome native drug efflux

Richard E. Lee; Julian Gregston Hurdle; Jiuyu Liu; David F. Bruhn; Tanja Matt; Michael S. Scherman; Pavan K. Vaddady; Zhong Zheng; Jianjun Qi; Rashid Akbergenov; Sourav Das; Dora B. Madhura; Chetan Rathi; Ashit Trivedi; Cristina Villellas; Robin B. Lee; Samanthi L. Waidyarachchi; Dianqing Sun; Michael R. McNeil; José A. Aínsa; Helena I. Boshoff; Mercedes Gonzalez-Juarrero; Bernd Meibohm; Erik C. Böttger; Anne J. Lenaerts

Although the classical antibiotic spectinomycin is a potent bacterial protein synthesis inhibitor, poor antimycobacterial activity limits its clinical application for treating tuberculosis. Using structure-based design, we generated a new semisynthetic series of spectinomycin analogs with selective ribosomal inhibition and excellent narrow-spectrum antitubercular activity. In multiple murine infection models, these spectinamides were well tolerated, significantly reduced lung mycobacterial burden and increased survival. In vitro studies demonstrated a lack of cross resistance with existing tuberculosis therapeutics, activity against multidrug-resistant (MDR) and extensively drug-resistant tuberculosis and an excellent pharmacological profile. Key to their potent antitubercular properties was their structural modification to evade the Rv1258c efflux pump, which is upregulated in MDR strains and is implicated in macrophage-induced drug tolerance. The antitubercular efficacy of spectinamides demonstrates that synthetic modifications to classical antibiotics can overcome the challenge of intrinsic efflux pump-mediated resistance and expands opportunities for target-based tuberculosis drug discovery.


Journal of Antimicrobial Chemotherapy | 2008

A microbiological assessment of novel nitrofuranylamides as anti-tuberculosis agents

Julian G. Hurdle; Robin B. Lee; Nageshwar Budha; Elizabeth I. Carson; Jianjun Qi; Michael S. Scherman; Sang Hyun Cho; Michael R. McNeil; Anne J. Lenaerts; Scott G. Franzblau; Bernd Meibohm; Richard E. Lee

OBJECTIVES Nitrofuranylamides (NFAs) are nitroaromatic compounds that have recently been discovered and have potent anti-tuberculosis (TB) activity. A foundational study was performed to evaluate whether this class of agents possesses microbiological properties suitable for future antimycobacterial therapy. METHODS Five representative compounds of the NFA series were evaluated by standard microbiological assays to determine MICs, MBCs, activity against anaerobic non-replicating persistent Mycobacterium tuberculosis, post-antibiotic effects (PAEs), antibiotic synergy and the basis for resistance. RESULTS The antimicrobial activity of these compounds was restricted to bacteria of the M. tuberculosis complex, and all compounds were highly active against drug-susceptible and -resistant strains of M. tuberculosis, with MICs 0.0004-0.05 mg/L. Moreover, no antagonism was observed with front-line anti-TB drugs. Activity was also retained against dormant bacilli in two in vitro low-oxygen models for M. tuberculosis persistence. A long PAE was observed, which was comparable to that of rifampicin, but superior to isoniazid and ethambutol. Spontaneous NFA-resistant mutants arose at a frequency of 10(-5)-10(-7), comparable to that for isoniazid (10(-5)-10(-6)). Some of these mutants exhibited cross-resistance to one or both of the nitroimidazoles PA-824 and OPC-67683. Cross-resistance was associated with inactivation of the reduced F(420)-deazaflavin cofactor pathway and not with inactivation of the Rv3547, the nitroreductase for PA-824 and OPC-67683. CONCLUSIONS Based on these studies, NFAs have many useful antimycobacterial properties applicable to TB chemotherapy and probably possess a unique mode of action that results in good activity against active and dormant M. tuberculosis. Therefore, the further development of lead compounds in this series is warranted.


Journal of Medicinal Chemistry | 2010

Structural studies of pterin-based inhibitors of dihydropteroate synthase.

Kirk E. Hevener; Mi Kyung Yun; Jianjun Qi; Iain D. Kerr; Kerim Babaoglu; Julian G. Hurdle; Kanya Balakrishna; Stephen W. White; Richard E. Lee

Dihydropteroate synthase (DHPS) is a key enzyme in bacterial folate synthesis and the target of the sulfonamide class of antibacterials. Resistance and toxicities associated with sulfonamides have led to a decrease in their clinical use. Compounds that bind to the pterin binding site of DHPS, as opposed to the p-amino benzoic acid (pABA) binding site targeted by the sulfonamide agents, are anticipated to bypass sulfonamide resistance. To identify such inhibitors and map the pterin binding pocket, we have performed virtual screening, synthetic, and structural studies using Bacillus anthracis DHPS. Several compounds with inhibitory activity have been identified, and crystal structures have been determined that show how the compounds engage the pterin site. The structural studies identify the key binding elements and have been used to generate a structure-activity based pharmacophore map that will facilitate the development of the next generation of DHPS inhibitors which specifically target the pterin site.


Aaps Journal | 2008

Pharmacokinetically-Guided Lead Optimization of Nitrofuranylamide Anti-Tuberculosis Agents

Nageshwar Budha; Nitin Mehrotra; Rajendra P. Tangallapally; Jianjun Qi; Antwan J. Daniels; Richard E. Lee; Bernd Meibohm

In an effort to develop novel and more potent therapies to treat tuberculosis, a new class of chemical agents, nitrofuranylamides, is being developed. The present study examines biopharmaceutic properties and preclinical pharmacokinetics of nitrofuranylamides at early stages of drug discovery to accelerate the optimization of leads into development candidates. The first tested compound, Lee 562, had high anti-tuberculosis activity in vitro, but exhibited poor metabolic stability resulting in a high systemic clearance, a short elimination half-life and low oral bioavailability in vivo in rats. Thus, two follow-up compounds were designed and tested that included structural modifications for increased metabolic stability. Both compounds showed improved metabolic stability compared to Lee 562, with Lee 878 being much more stable than Lee 952. As a consequence, the oral bioavailability of Lee 878 reached ~27% compared to 16% for the other two compounds. This observation prompted us to select compounds based on metabolic stability screening and a new set of nine compounds with high in vitro activity were tested for metabolic stability. The most stable compound in the assay, Lee 1106 was selected for further pharmacokinetic evaluation in rats. Surprisingly, Lee 1106 exhibited poor oral bioavailability, 4.6%. Biopharmaceutic evaluation of the compound showed that the compound has poor aqueous solubility and a high clogP. Based on these results, a screening paradigm was developed for optimization of the nitrofuranylamide lead compounds in a timely and cost-effective manner that might also be applicable to other classes of anti-infective drugs.


ChemMedChem | 2008

Novel acyl phosphate mimics that target PlsY, an essential acyltransferase in gram-positive bacteria.

Kimberly D. Grimes; Ying Jie Lu; Yong Mei Zhang; Vicki A. Luna; Julian G. Hurdle; Elizabeth I. Carson; Jianjun Qi; Sucheta Kudrimoti; Charles O. Rock; Richard E. Lee

PlsY is a recently discovered acyltransferase that executes an essential step in membrane phospholipid biosynthesis in Gram‐ positive bacteria. By using a bioisosteric replacement approach to generate substrate‐based inhibitors of PlsY as potential novel antibacterial agents, a series of stabilized acyl phosphate mimetics, including acyl phosphonates, acyl α,α‐difluoromethyl phosphonates, acyl phosphoramides, reverse amide phosphonates, acyl sulfamates, and acyl sulfamides were designed and synthesized. Several acyl phosphonates, phosphoramides, and sulfamates were identified as inhibitors of PlsY from Streptococcus pneumoniae and Bacillus anthracis. As anticipated, these inhibitors were competitive inhibitors with respect to the acyl phosphate substrate. Antimicrobial testing showed the inhibitors to have generally weak activity against Gram‐positive bacteria with the exception of some acyl phosphonates, reverse amide phosphonates, and acyl sulfamates, which had potent activity against multiple strains of B. anthracis.


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.


Bioorganic & Medicinal Chemistry Letters | 2016

Pterin-sulfa conjugates as dihydropteroate synthase inhibitors and antibacterial agents.

Ying Zhao; William R. Shadrick; Miranda J. Wallace; Yinan Wu; Elizabeth Griffith; Jianjun Qi; Mi-Kyung Yun; Stephen W. White; Richard E. Lee

The sulfonamide class of antibiotics has been in continuous use for over 70years. They are thought to act by directly inhibiting dihydropteroate synthase (DHPS), and also acting as prodrugs that sequester pterin pools by forming dead end pterin-sulfonamide conjugates. In this study, eight pterin-sulfonamide conjugates were synthesized using a novel synthetic strategy and their biochemical and microbiological properties were investigated. The conjugates were shown to competitively inhibit DHPS, and inhibition was enhanced by the presence of pyrophosphate that is crucial to catalysis and is known to promote an ordering of the DHPS active site. The co-crystal structure of Yersinia pestis DHPS bound to one of the more potent conjugates revealed a mode of binding that is similar to that of the enzymatic product analog pteroic acid. The antimicrobial activities of the pterin-sulfonamide conjugates were measured against Escherichia coli in the presence and absence of folate precursors and dependent metabolites. These results show that the conjugates have appreciable antibacterial activity and act by an on target, anti-folate pathway mechanism rather than as simple dead end products.


Bioorganic & Medicinal Chemistry | 2011

Synthesis of bi-substrate state mimics of dihydropteroate synthase as potential inhibitors and molecular probes.

Jianjun Qi; Kristopher G. Virga; Sourav Das; Ying Zhao; Mi-Kyung Yun; Stephen W. White; Richard E. Lee

The increasing emergence of resistant bacteria drives us to design and develop new antimicrobial agents. Pursuant to that goal, a new targeting approach of the dihydropteroate synthase enzyme, which serves as the site of action for the sulfonamide class of antimicrobial agents, is being explored. Using structural information, a new class of transition state mimics has been designed and synthesized that have the capacity to bind to the pterin, phosphate and para-amino binding sites. The design, synthesis and evaluation of these compounds as inhibitors of Bacillusanthracis dihydropteroate synthase is described herein. Outcomes from this work have identified the first trivalent inhibitors of dihydropteroate synthase whose activity displayed slow binding inhibition. The most active compounds in this series contained an oxidized pterin ring. The binding of these inhibitors was modeled into the dihydropteroate synthase active site and demonstrated a good correlation with the observed bioassay data, as well as provided important insight for the future design of higher affinity transition state mimics.


Molecular Cancer Therapeutics | 2006

Development of an etoposide prodrug for dual prodrug-enzyme antitumor therapy

K. Jin Yoon; Jianjun Qi; Joanna S. Remack; Kristopher G. Virga; M. Jason Hatfield; Philip M. Potter; Richard E. Lee; Mary K. Danks

Enzyme-prodrug approaches to cancer therapy, theoretically, have the potential to mediate tumor-selective cytotoxicity. However, even if tumor-specific prodrug activation is achieved, enzyme-prodrug systems investigated thus far comprised a single enzyme and a specific prodrug. Although targeted, such systems constitute single-agent therapy, which may be ineffective and/or may promote development of drug resistance. Therefore, a goal of our laboratories was to design and characterize a novel dipiperidinyl derivative of etoposide [1,4′-dipiperidine-1′-carboxylate-etoposide (dp-VP16)] that would act as a prodrug. We envisioned that dp-VP16 would be converted to the active chemotherapeutic agent VP-16 by the same rabbit carboxylesterase (rCE) that we have previously shown to efficiently activate the prodrug irinotecan (CPT-11). This dp-VP16 prodrug might then be used in combination with CPT-11, with both drugs activated by a single enzyme. We evaluated the ability of pure rCE and two human carboxylesterases, hCE1 and hiCE (hCE2), to activate dp-VP16 in vitro, and in neuroblastoma cell lines designed to express/overexpress each enzyme. In SK-N-AS neuroblastoma cell transfectants, expression of rCE or hiCE decreased the IC50 of dp-VP16 as a single agent by 8.3- and 3.4-fold, respectively, in growth inhibition assays. Purified hCE1 did not metabolize dp-VP16 in vitro and did not affect its IC50 in intact cells. The combination indices of sequential exposure to CPT-11 followed by dp-VP16 ranged from ∼0.4 to 0.6, suggesting that this combination produced greater-than-additive cytotoxicity in neuroblastoma cells expressing rCE. These data provide proof-of-principle that enzyme-prodrug therapy approaches comprised of prodrugs with complementary mechanisms of cytotoxicity that are activated by a single enzyme can be developed. [Mol Cancer Ther 2006;5(6):1577–84]


Molecular Cell | 2006

Acyl-phosphates initiate membrane phospholipid synthesis in Gram-positive pathogens.

Ying-Jie Lu; Yong-Mei Zhang; Kimberly D. Grimes; Jianjun Qi; Richard E. Lee; Charles O. Rock

Collaboration


Dive into the Jianjun Qi's collaboration.

Top Co-Authors

Avatar

Richard E. Lee

University of Tennessee Health Science Center

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

Bernd Meibohm

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Julian G. Hurdle

University of Texas at Arlington

View shared research outputs
Top Co-Authors

Avatar

Kerim Babaoglu

University of Tennessee Health Science Center

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

Ying Zhao

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Charles O. Rock

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Elizabeth I. Carson

University of Tennessee Health Science Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge