Network


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

Hotspot


Dive into the research topics where K. R. Prabhakar is active.

Publication


Featured researches published by K. R. Prabhakar.


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

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.


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.


Journal of Medicinal Chemistry | 2014

Aminoazabenzimidazoles, a Novel Class of Orally Active Antimalarial Agents

Shahul Hameed P; Murugan Chinnapattu; Gajanan Shanbag; Praveena Manjrekar; Krishna Koushik; Anandkumar Raichurkar; Vikas Patil; Sandesh Jatheendranath; Suresh Rudrapatna; Shubhada Pramod Barde; Nikhil Rautela; Disha Awasthy; Sapna Morayya; Chandan Narayan; Stefan Kavanagh; Ramanatha Saralaya; Pavithra Viswanath; Kakoli Mukherjee; Balachandra Bandodkar; Abhishek Srivastava; Jitender Reddy; K. R. Prabhakar; Achyut Sinha; María Belén Jiménez-Díaz; María Santos Martínez; Iñigo Angulo-Barturen; Santiago Ferrer; Laura Sanz; Francisco Javier Gamo; Sandra Duffy

Whole-cell high-throughput screening of the AstraZeneca compound library against the asexual blood stage of Plasmodium falciparum (Pf) led to the identification of amino imidazoles, a robust starting point for initiating a hit-to-lead medicinal chemistry effort. Structure-activity relationship studies followed by pharmacokinetics optimization resulted in the identification of 23 as an attractive lead with good oral bioavailability. Compound 23 was found to be efficacious (ED90 of 28.6 mg·kg(-1)) in the humanized P. falciparum mouse model of malaria (Pf/SCID model). Representative compounds displayed a moderate to fast killing profile that is comparable to that of chloroquine. This series demonstrates no cross-resistance against a panel of Pf strains with mutations to known antimalarial drugs, thereby suggesting a novel mechanism of action for this chemical class.


Journal of Medicinal Chemistry | 2017

Discovery of Imidazo[1,2-a]pyridine Ethers and Squaramides as Selective and Potent Inhibitors of Mycobacterial Adenosine Triphosphate (ATP) Synthesis

Subramanyam J. Tantry; Shankar D. Markad; Vikas Shinde; Jyothi Bhat; Gayathri Balakrishnan; Amit K. Gupta; Anisha Ambady; Anandkumar Raichurkar; Chaitanyakumar Kedari; Sreevalli Sharma; Naina V. Mudugal; Ashwini Narayan; C. N. Naveen Kumar; Robert Nanduri; Jitendar Reddy; K. R. Prabhakar; Karthikeyan Kandaswamy; Ramanatha Saralaya; Parvinder Kaur; Neela Dinesh; Supreeth Guptha; Kirsty Rich; David Murray; Helen Plant; Marian Preston; Helen Ashton; Darren Plant; Jarrod Walsh; Peter Alcock; Kathryn Naylor

The approval of bedaquiline to treat tuberculosis has validated adenosine triphosphate (ATP) synthase as an attractive target to kill Mycobacterium tuberculosis (Mtb). Herein, we report the discovery of two diverse lead series imidazo[1,2-a]pyridine ethers (IPE) and squaramides (SQA) as inhibitors of mycobacterial ATP synthesis. Through medicinal chemistry exploration, we established a robust structure-activity relationship of these two scaffolds, resulting in nanomolar potencies in an ATP synthesis inhibition assay. A biochemical deconvolution cascade suggested cytochrome c oxidase as the potential target of IPE class of molecules, whereas characterization of spontaneous resistant mutants of SQAs unambiguously identified ATP synthase as its molecular target. Absence of cross resistance against bedaquiline resistant mutants suggested a different binding site for SQAs on ATP synthase. Furthermore, SQAs were found to be noncytotoxic and demonstrated efficacy in a mouse model of tuberculosis infection.


MedChemComm | 2016

Scaffold morphing leading to evolution of 2,4-diaminoquinolines and aminopyrazolopyrimidines as inhibitors of the ATP synthesis pathway

Subramanyam J. Tantry; Vikas Shinde; Gayathri Balakrishnan; Shankar D. Markad; Amit K. Gupta; Jyothi Bhat; Ashwini Narayan; Anandkumar Raichurkar; Lalit kumar Jena; Sreevalli Sharma; Naveen Kumar; Robert Nanduri; Jitendar Reddy; K. R. Prabhakar; Karthikeyan Kandaswamy; Parvinder Kaur; Neela Dinesh; Supreeth Guptha; Ramanatha Saralaya; Manoranjan Panda; Suresh Rudrapatna; Meenakshi Mallya; Harvey Rubin; Takahiro Yano; Khisi Mdluili; Christopher B. Cooper; V. Balasubramanian; Vasan K. Sambandamurthy; Radha Shandil; Stefan Kavanagh

The success of bedaquiline as an anti-tubercular agent for the treatment of multidrug-resistant tuberculosis has validated the ATP synthesis pathway and in particular ATP synthase as an attractive target. However, limitations associated with its use in the clinic and the drug–drug interactions with rifampicin have prompted research efforts towards identifying alternative ATP synthesis inhibitors with differentiated mechanisms of action. A biochemical assay was employed to screen AstraZenecas corporate compound collection to identify the inhibitors of mycobacterial ATP synthesis. The high-throughput screening resulted in the identification of 2,4-diaminoquinazolines as inhibitors of the ATP synthesis pathway. A structure–activity relationship for the quinazolines was established and the knowledge was utilized to morph the quinazoline core into quinoline and pyrazolopyrimidine to expand the scope of chemical diversity. The morphed scaffolds exhibited a 10-fold improvement in enzyme potency and over 100-fold improvement in selectivity against inhibition of mammalian mitochondrial ATP synthesis. These novel compounds were bactericidal and demonstrated growth retardation of Mycobacterium tuberculosis in the acute mouse model of tuberculosis infection.


Antimicrobial Agents and Chemotherapy | 2015

In Silico-Based High-Throughput Screen for Discovery of Novel Combinations for Tuberculosis Treatment

Ragini Singh; Radha Shandil; Sreevalli Sharma; Swati Khandelwal; Malancha Karmarkar; Naveen Kumar; Suresh Solapure; Ramanatha Saralaya; Robert Nanduri; Jitendar Reddy; K. R. Prabhakar; Swaminathan Rajagopalan; Narasimha Rao; Shridhar Narayanan; Anand Anandkumar; V. Balasubramanian; Santanu Datta

ABSTRACT There are currently 18 drug classes for the treatment of tuberculosis, including those in the development pipeline. An in silico simulation enabled combing the innumerably large search space to derive multidrug combinations. Through the use of ordinary differential equations (ODE), we constructed an in silico kinetic platform in which the major metabolic pathways in Mycobacterium tuberculosis and the mechanisms of the antituberculosis drugs were integrated into a virtual proteome. The optimized model was used to evaluate 816 triplets from the set of 18 drugs. The experimentally derived cumulative fractional inhibitory concentration (∑FIC) value was within twofold of the model prediction. Bacterial enumeration revealed that a significant number of combinations that were synergistic for growth inhibition were also synergistic for bactericidal effect. The in silico-based screen provided new starting points for testing in a mouse model of tuberculosis, in which two novel triplets and five novel quartets were significantly superior to the reference drug triplet of isoniazid, rifampin, and ethambutol (HRE) or the quartet of HRE plus pyrazinamide (HREZ).


Antimicrobial Agents and Chemotherapy | 2014

UDP-N-Acetylmuramic Acid l-Alanine Ligase (MurC) Inhibition in a tolC Mutant Escherichia coli Strain Leads to Cell Death

Vaishali Humnabadkar; K. R. Prabhakar; Ashwini Narayan; Sreevalli Sharma; Supreeth Guptha; Praveena Manjrekar; Murugan Chinnapattu; Shahul P. Hameed; Sudha Ravishankar; Monalisa Chatterji

ABSTRACT The Mur ligases play an essential role in the biosynthesis of bacterial peptidoglycan and hence are attractive antibacterial targets. A screen of the AstraZeneca compound library led to the identification of compound A, a pyrazolopyrimidine, as a potent inhibitor of Escherichia coli and Pseudomonas aeruginosa MurC. However, cellular activity against E. coli or P. aeruginosa was not observed. Compound A was active against efflux pump mutants of both strains. Experiments using an E. coli tolC mutant revealed accumulation of the MurC substrate and a decrease in the level of product upon treatment with compound A, indicating inhibition of MurC enzyme in these cells. Such a modulation was not observed in the E. coli wild-type cells. Further, overexpression of MurC in the E. coli tolC mutant led to an increase in the compound A MIC by ≥16-fold, establishing a correlation between MurC inhibition and cellular activity. In addition, estimation of the intracellular compound A level showed an accumulation of the compound over time in the tolC mutant strain. A significant compound A level was not detected in the wild-type E. coli strain even upon treatment with high concentrations of the compound. Therefore, the lack of MIC and absence of MurC inhibition in wild-type E. coli were possibly due to suboptimal compound concentration as a consequence of a high efflux level and/or poor permeativity of compound A.


Xenobiotica | 2017

Evaluation of the metabolism, bioactivation and pharmacokinetics of triaminopyrimidine analogs toward selection of a potential candidate for antimalarial therapy.

Abhishek Srivastava; Ramanatha Saralaya; K. R. Prabhakar; Shahul P. Hameed; Suresh Solapure; Vinayak Hosagrahara

Abstract 1. During the course of metabolic profiling of lead Compound 1, glutathione (GSH) conjugates were detected in rat bile, suggesting the formation of reactive intermediate precursor(s). This was confirmed by the identification of GSH and N-acetylcysteine (NAC) conjugates in microsomal incubations. 2. It was proposed that bioactivation of Compound 1 occurs via the formation of a di-iminoquinone reactive intermediate through the involvement of the C-2 and C-5 nitrogens of the pyrimidine core. 3. To further investigate this hypothesis, structural analogs with modifications at the C-5 nitrogen were studied for metabolic activation in human liver microsomes supplemented with GSH/NAC. 4. Compounds 1 and 2, which bear secondary nitrogens at the C-5 of the pyrimidine core, were observed to form significant amounts of GSH/NAC-conjugates in vitro, whereas compounds with tertiary nitrogens at C-5 (Compound 3 and 4) formed no such conjugates. 5. These observations provide evidence that electron/hydrogen abstraction is required for the bioactivation of the triaminopyrimidines, potentially via a di-iminoquinone intermediate. The lack of a hydrogen and/or steric hindrance rendered Compound 3 and 4 incapable of forming thiol conjugates. 6. This finding enabled advancement of compound 4, with a desirable potency, safety and PK profile, as a lead candidate for further development in the treatment of malaria.


Journal of Medicinal Chemistry | 2013

Azaindoles: Noncovalent DprE1 Inhibitors from Scaffold Morphing Efforts, Kill Mycobacterium tuberculosis and Are Efficacious in Vivo

Pravin S. Shirude; Radha Shandil; Claire Sadler; Maruti Naik; Vinayak Hosagrahara; Shahul P. Hameed; Vikas Shinde; Chandramohan Bathula; Vaishali Humnabadkar; Naveen Kumar; Jitendar Reddy; Sreevalli Sharma; Anisha Ambady; Naina Hegde; James Whiteaker; Robert E. McLaughlin; Humphrey Gardner; Prashanti Madhavapeddi; Parvinder Kaur; Ashwini Narayan; Supreeth Guptha; Disha Awasthy; Chandan Narayan; Jyothi Mahadevaswamy; K. G. Vishwas; Vijaykamal Ahuja; Abhishek Srivastava; K. R. Prabhakar; Ramesh R. Kale; Manjunatha Ramaiah

Collaboration


Dive into the K. R. Prabhakar's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge