Network


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

Hotspot


Dive into the research topics where Jitendar Reddy is active.

Publication


Featured researches published by Jitendar Reddy.


Journal of Medicinal Chemistry | 2013

Thiazolopyridine Ureas as Novel Antitubercular Agents Acting through Inhibition of DNA Gyrase B.

Manoj Kale; Anandkumar Raichurkar; Shahul Hameed P; David Waterson; David C. McKinney; M. R. Manjunatha; Usha Kranthi; Krishna Koushik; Lalit kumar Jena; Vikas Shinde; Suresh Rudrapatna; Shubhada Barde; Vaishali Humnabadkar; Prashanti Madhavapeddi; Halesha D. Basavarajappa; Anirban Ghosh; V. K. Ramya; Supreeth Guptha; Sreevalli Sharma; Prakash Vachaspati; K.N. Mahesh Kumar; Jayashree Giridhar; Jitendar Reddy; Samit Ganguly; Vijaykamal Ahuja; Sheshagiri Gaonkar; C. N. Naveen Kumar; Derek Ogg; Julie Tucker; P. Ann Boriack-Sjodin

A pharmacophore-based search led to the identification of thiazolopyridine ureas as a novel scaffold with antitubercular activity acting through inhibition of DNA Gyrase B (GyrB) ATPase. Evaluation of the binding mode of thiazolopyridines in a Mycobacterium tuberculosis (Mtb) GyrB homology model prompted exploration of the side chains at the thiazolopyridine ring C-5 position to access the ribose/solvent pocket. Potent compounds with GyrB IC50 ≤ 1 nM and Mtb MIC ≤ 0.1 μM were obtained with certain combinations of side chains at the C-5 position and heterocycles at the C-6 position of the thiazolopyridine core. Substitutions at C-5 also enabled optimization of the physicochemical properties. Representative compounds were cocrystallized with Streptococcus pneumoniae (Spn) ParE; these confirmed the binding modes predicted by the homology model. The target link to GyrB was confirmed by genetic mapping of the mutations conferring resistance to thiazolopyridine ureas. The compounds are bactericidal in vitro and efficacious in vivo in an acute murine model of tuberculosis.


Antimicrobial Agents and Chemotherapy | 2013

In Vitro and In Vivo Efficacy of β-Lactams against Replicating and Slowly Growing/Nonreplicating Mycobacterium tuberculosis

Suresh Solapure; Neela Dinesh; Radha Shandil; Sreevalli Sharma; Deepa Bhattacharjee; Samit Ganguly; Jitendar Reddy; Vijaykamal Ahuja; Manish Parab; K. G. Vishwas; Naveen Kumar; Meenakshi Balganesh; V. Balasubramanian

ABSTRACT Beta-lactams, in combination with beta-lactamase inhibitors, are reported to have activity against Mycobacterium tuberculosis bacteria growing in broth, as well as inside the human macrophage. We tested representative beta-lactams belonging to 3 different classes for activity against replicating M. tuberculosis in broth and nonreplicating M. tuberculosis under hypoxia, as well as against streptomycin-starved M. tuberculosis strain 18b (ss18b) in the presence or absence of clavulanate. Most of the combinations showed bactericidal activity against replicating M. tuberculosis, with up to 200-fold improvement in potency in the presence of clavulanate. None of the combinations, including those containing meropenem, imipenem, and faropenem, killed M. tuberculosis under hypoxia. However, faropenem- and meropenem-containing combinations killed strain ss18b moderately. We tested the bactericidal activities of meropenem-clavulanate and amoxicillin-clavulanate combinations in the acute and chronic aerosol infection models of tuberculosis in BALB/c mice. Based on pharmacokinetic/pharmacodynamic indexes reported for beta-lactams against other bacterial pathogens, a cumulative percentage of a 24-h period that the drug concentration exceeds the MIC under steady-state pharmacokinetic conditions (%TMIC) of 20 to 40% was achieved in mice using a suitable dosing regimen. Both combinations showed marginal reduction in lung CFU compared to the late controls in the acute model, whereas both were inactive in the chronic model.


Antimicrobial Agents and Chemotherapy | 2014

1,4-Azaindole, a Potential Drug Candidate for Treatment of Tuberculosis

Monalisa Chatterji; Radha Shandil; M. R. Manjunatha; Suresh Solapure; Naveen Kumar; Ramanatha Saralaya; Jitendar Reddy; K. R. Prabhakar; Sreevalli Sharma; Claire Sadler; Christopher B. Cooper; Khisi Mdluli; Pravin S. Iyer; Shridhar Narayanan; Pravin S. Shirude

ABSTRACT New therapeutic strategies against multidrug-resistant (MDR) and extensively drug-resistant (XDR) Mycobacterium tuberculosis are urgently required to combat the global tuberculosis (TB) threat. Toward this end, we previously reported the identification of 1,4-azaindoles, a promising class of compounds with potent antitubercular activity through noncovalent inhibition of decaprenylphosphoryl-β-d-ribose 2′-epimerase (DprE1). Further, this series was optimized to improve its physicochemical properties and pharmacokinetics in mice. Here, we describe the short-listing of a potential clinical candidate, compound 2, that has potent cellular activity, drug-like properties, efficacy in mouse and rat chronic TB infection models, and minimal in vitro safety risks. We also demonstrate that the compounds, including compound 2, have no antagonistic activity with other anti-TB drugs. Moreover, compound 2 shows synergy with PA824 and TMC207 in vitro, and the synergy effect is translated in vivo with TMC207. The series is predicted to have a low clearance in humans, and the predicted human dose for compound 2 is ≤1 g/day. Altogether, our data suggest that a 1,4-azaindole (compound 2) is a promising candidate for the development of a novel anti-TB drug.


Journal of Medicinal Chemistry | 2014

Novel N-Linked Aminopiperidine-Based Gyrase Inhibitors with Improved hERG and in Vivo Efficacy against Mycobacterium tuberculosis

Shahul Hameed P; Vikas Patil; Suresh Solapure; Umender Sharma; Prashanti Madhavapeddi; Anandkumar Raichurkar; Murugan Chinnapattu; Praveena Manjrekar; Gajanan Shanbhag; Jayashree Puttur; Vikas Shinde; Sreenivasaiah Menasinakai; Suresh Rudrapatana; Vijayashree Achar; Disha Awasthy; Radha Nandishaiah; Vaishali Humnabadkar; Anirban Ghosh; Chandan Narayan; V. K. Ramya; Parvinder Kaur; Sreevalli Sharma; Jim Werngren; Sven Hoffner; C. N. Naveen Kumar; Jitendar Reddy; Mahesh Kumar Kn; Samit Ganguly; Ugarkar Bheemarao; Kakoli Mukherjee

DNA gyrase is a clinically validated target for developing drugs against Mycobacterium tuberculosis (Mtb). Despite the promise of fluoroquinolones (FQs) as anti-tuberculosis drugs, the prevalence of pre-existing resistance to FQs is likely to restrict their clinical value. We describe a novel class of N-linked aminopiperidinyl alkyl quinolones and naphthyridones that kills Mtb by inhibiting the DNA gyrase activity. The mechanism of inhibition of DNA gyrase was distinct from the fluoroquinolones, as shown by their ability to inhibit the growth of fluoroquinolone-resistant Mtb. Biochemical studies demonstrated this class to exert its action via single-strand cleavage rather than double-strand cleavage, as seen with fluoroquinolones. The compounds are highly bactericidal against extracellular as well as intracellular Mtb. Lead optimization resulted in the identification of potent compounds with improved oral bioavailability and reduced cardiac ion channel liability. Compounds from this series are efficacious in various murine models of tuberculosis.


Antimicrobial Agents and Chemotherapy | 2012

Effect of Coadministration of Moxifloxacin and Rifampin on Mycobacterium tuberculosis in a Murine Aerosol Infection Model

V. Balasubramanian; Suresh Solapure; Sheshagiri Gaonkar; K.N. Mahesh Kumar; Radha Shandil; Abhijeet Deshpande; Naveen Kumar; K. G. Vishwas; Jitendar Reddy; Samit Ganguly; Arnold Louie; George L. Drusano

ABSTRACT Coadministration of moxifloxacin and rifampin was evaluated in a murine model of Mycobacterium tuberculosis pulmonary infection to determine whether the finding of antagonism documented in a hollow-fiber infection model could be recapitulated in vivo. Colony counts were followed in a no-treatment control group, groups administered moxifloxacin or rifampin monotherapy, and a group administered a combination of the two agents. Following 18 days of once-daily oral administration to mice infected with M. tuberculosis, there was a reduction in the plasma exposure to rifampin that decreased further when rifampin was coadministered with moxifloxacin. Pharmacodynamic analysis demonstrated a mild antagonistic interaction between moxifloxacin and rifampin with respect to cell kill in the mouse model for tuberculosis (TB). No emergence of resistance was noted over 28 days of therapy, even with monotherapy. This was true even though one of the agents in the combination (moxifloxacin) induces error-prone replication. The previously noted antagonism with respect to cell kill shown in the hollow-fiber infection model was recapitulated in the murine TB lung model, although to a lesser extent.


Journal of Medicinal Chemistry | 2014

N-Aryl-2-aminobenzimidazoles: Novel, Efficacious, Antimalarial Lead Compounds

P Shahul Hameed; Abhishek Srivastava; Gajanan Shanbhag; Sapna Morayya; Nikhil Rautela; Disha Awasthy; Stefan Kavanagh; Jitendar Reddy; K. R. Prabhakar; Ramanatha Saralaya; Robert Nanduri; Anandkumar Raichurkar; Sreenivasaiah Menasinakai; Vijayashree Achar; María Belén Jiménez-Díaz; María Santos Martínez; Iñigo Angulo-Barturen; Santiago Ferrer; Laura Sanz; Francisco Javier Gamo; Sandra Duffy; Vicky M. Avery; David Waterson; Marcus C. S. Lee; Olivia Coburn-Flynn; David A. Fidock; Pravin S. Iyer; Shridhar Narayanan; Vinayak Hosagrahara; Vasan K. Sambandamurthy

From the phenotypic screening of the AstraZeneca corporate compound collection, N-aryl-2-aminobenzimidazoles have emerged as novel hits against the asexual blood stage of Plasmodium falciparum (Pf). Medicinal chemistry optimization of the potency against Pf and ADME properties resulted in the identification of 12 as a lead molecule. Compound 12 was efficacious in the P. berghei (Pb) model of malaria. This compound displayed an excellent pharmacokinetic profile with a long half-life (19 h) in rat blood. This profile led to an extended survival of animals for over 30 days following a dose of 50 mg/kg in the Pb malaria model. Compound 12 retains its potency against a panel of Pf isolates with known mechanisms of resistance. The fast killing observed in the in vitro parasite reduction ratio (PRR) assay coupled with the extended survival highlights the promise of this novel chemical class for the treatment of malaria.


ACS Medicinal Chemistry Letters | 2014

Benzimidazoles: novel mycobacterial gyrase inhibitors from scaffold morphing.

Shahul Hameed P; Anandkumar Raichurkar; Prashanti Madhavapeddi; Sreenivasaiah Menasinakai; Sreevalli Sharma; Parvinder Kaur; Radha Nandishaiah; Jitendar Reddy; Vasan K. Sambandamurthy; Dharmarajan Sriram

Type II topoisomerases are well conserved across the bacterial species, and inhibition of DNA gyrase by fluoroquinolones has provided an attractive option for treatment of tuberculosis (TB). However, the emergence of fluoroquinolone-resistant strains of Mycobacterium tuberculosis (Mtb) poses a threat for its sustainability. A scaffold hopping approach using the binding mode of novel bacterial topoisomerase inhibitors (NBTIs) led to the identification of a novel class of benzimidazoles as DNA gyrase inhibitors with potent anti-TB activity. Docking of benzimidazoles to a NBTI bound crystal structure suggested that this class of compound makes key contacts in the enzyme active site similar to the reported NBTIs. This observation was further confirmed through the measurement of DNA gyrase inhibition, and activity against Mtb strains harboring mutations that confer resistance to aminopiperidines based NBTIs and Mtb strains resistant to moxifloxacin. Structure-activity relationship modification at the C-7 position of the left-hand side ring provided further avenue to improve hERG selectivity for this chemical series that has been the major challenges for NBTIs.


Nature Communications | 2015

Triaminopyrimidine is a fast-killing and long-acting antimalarial clinical candidate

Shahul Hameed P; Suresh Solapure; Vikas Patil; Philipp P. Henrich; Pamela Magistrado; Kannan Murugan; Pavithra Viswanath; Jayashree Puttur; Abhishek Srivastava; Eknath Bellale; Gajanan Shanbag; Disha Awasthy; Sudhir Landge; Sapna Morayya; Krishna Koushik; Ramanatha Saralaya; Anandkumar Raichurkar; Nikhil Rautela; Nilanjana Roy Choudhury; Anisha Ambady; Radha Nandishaiah; Jitendar Reddy; K. R. Prabhakar; Sreenivasaiah Menasinakai; Suresh Rudrapatna; Monalisa Chatterji; María Belén Jiménez-Díaz; María Santos Martínez; Laura Sanz; Olivia Coburn-Flynn

The widespread emergence of Plasmodium falciparum (Pf) strains resistant to frontline agents has fuelled the search for fast-acting agents with novel mechanism of action. Here, we report the discovery and optimization of novel antimalarial compounds, the triaminopyrimidines (TAPs), which emerged from a phenotypic screen against the blood stages of Pf. The clinical candidate (compound 12) is efficacious in a mouse model of Pf malaria with an ED99 <30 mg kg−1 and displays good in vivo safety margins in guinea pigs and rats. With a predicted half-life of 36 h in humans, a single dose of 260 mg might be sufficient to maintain therapeutic blood concentration for 4–5 days. Whole-genome sequencing of resistant mutants implicates the vacuolar ATP synthase as a genetic determinant of resistance to TAPs. Our studies highlight the potential of TAPs for single-dose treatment of Pf malaria in combination with other agents in clinical development.


Antimicrobial Agents and Chemotherapy | 2014

Optimization of Pyrrolamides as Mycobacterial GyrB ATPase Inhibitors: Structure-Activity Relationship and In Vivo Efficacy in a Mouse Model of Tuberculosis

Shahul Hameed P; Suresh Solapure; Kakoli Mukherjee; Vrinda Nandi; David Waterson; Radha Shandil; Meenakshi Balganesh; Vasan K. Sambandamurthy; Anand Kumar V. Raichurkar; Abhijeet Deshpande; Anirban Ghosh; Disha Awasthy; Gajanan Shanbhag; Gulebahar Sheikh; Helen McMiken; Jayashree Puttur; Jitendar Reddy; Jim Werngren; Jon Read; Mahesh Kumar; Manjunatha R; Murugan Chinnapattu; Prashanti Madhavapeddi; Praveena Manjrekar; Reetobrata Basu; Sheshagiri Gaonkar; Sreevalli Sharma; Sven Hoffner; Vaishali Humnabadkar; Venkita Subbulakshmi

ABSTRACT Moxifloxacin has shown excellent activity against drug-sensitive as well as drug-resistant tuberculosis (TB), thus confirming DNA gyrase as a clinically validated target for discovering novel anti-TB agents. We have identified novel inhibitors in the pyrrolamide class which kill Mycobacterium tuberculosis through inhibition of ATPase activity catalyzed by the GyrB domain of DNA gyrase. A homology model of the M. tuberculosis H37Rv GyrB domain was used for deciphering the structure-activity relationship and binding interactions of inhibitors with mycobacterial GyrB enzyme. Proposed binding interactions were later confirmed through cocrystal structure studies with the Mycobacterium smegmatis GyrB ATPase domain. The most potent compound in this series inhibited supercoiling activity of DNA gyrase with a 50% inhibitory concentration (IC50) of <5 nM, an MIC of 0.03 μg/ml against M. tuberculosis H37Rv, and an MIC90 of <0.25 μg/ml against 99 drug-resistant clinical isolates of M. tuberculosis. The frequency of isolating spontaneous resistant mutants was ∼10−6 to 10−8, and the point mutation mapped to the M. tuberculosis GyrB domain (Ser208 Ala), thus confirming its mode of action. The best compound tested for in vivo efficacy in the mouse model showed a 1.1-log reduction in lung CFU in the acute model and a 0.7-log reduction in the chronic model. This class of GyrB inhibitors could be developed as novel anti-TB agents.


European Journal of Pharmaceutical Sciences | 2013

Effect of repeated dosing on rifampin exposure in BALB/c mice.

Vinayak Hosagrahara; Jitendar Reddy; Samit Ganguly; Vijaykamal Ahuja; Manish Parab; Jayashree Giridhar

The discovery of novel therapeutics for the treatment of tuberculosis involves routine testing in a mouse model over four weeks of daily dosing with test compounds. In this model, daily oral administration of rifampin (10 mg/kg) showed significantly lower plasma exposure on day 5 compared to day 1. The absence of PXR-mediated induction of mouse Cyp3a isoforms was confirmed in the present study by incubating liver microsomes prepared from control and rifampin treated mice with probe substrates of CYP3A. To test whether the reduction in exposure was due to Pgp-mediated efflux, verapamil, a known Pgp inhibitor, was dosed to the rifampin pre-treated mice which led to an increase in exposure to that obtained after a single dose of rifampin, suggesting the role of Pgp induction in reducing exposure to rifampin. To further confirm Pgp induction in rifampin treated mice, digoxin, a known substrate of Pgp, was administered to the rifampin pre-treated mice, and a significant drop in the digoxin exposure was observed compared to the control group. Collectively, our results show that repeated administration of rifampin in mice leads to a reduction in oral exposure due to induction of Pgp-mediated efflux of rifampin, and not via induction of CYP3A isoforms.

Collaboration


Dive into the Jitendar Reddy's collaboration.

Researchain Logo
Decentralizing Knowledge