Network


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

Hotspot


Dive into the research topics where Prashanti Madhavapeddi is active.

Publication


Featured researches published by Prashanti Madhavapeddi.


ACS Chemical Biology | 2013

Aminopyrazinamides: Novel and Specific GyrB Inhibitors that Kill Replicating and Nonreplicating Mycobacterium tuberculosis

Pravin S. Shirude; Prashanti Madhavapeddi; Julie Tucker; Kannan Murugan; Vikas Patil; Halesha D. Basavarajappa; Anandkumar Raichurkar; Vaishali Humnabadkar; Syeed Hussein; Sreevalli Sharma; V. K. Ramya; Chandan Narayan; Tanjore S. Balganesh; Vasan K. Sambandamurthy

Aminopyrazinamides originated from a high throughput screen targeting the Mycobacterium smegmatis (Msm) GyrB ATPase. This series displays chemical tractability, robust structure-activity relationship, and potent antitubercular activity. The crystal structure of Msm GyrB in complex with one of the aminopyrazinamides revealed promising attributes of specificity against other broad spectrum pathogens and selectivity against eukaryotic kinases due to novel interactions at hydrophobic pocket, unlike other known GyrB inhibitors. The aminopyrazinamides display excellent mycobacterial kill under in vitro, intracellular, and hypoxic conditions.


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.


Bioorganic & Medicinal Chemistry Letters | 2014

Thiazolopyridone ureas as DNA gyrase B inhibitors: optimization of antitubercular activity and efficacy.

Ramesh R. Kale; Manoj Kale; David Waterson; Anandkumar Raichurkar; Shahul P. Hameed; M. R. Manjunatha; B. K. Kishore Reddy; Krishnan Malolanarasimhan; Vikas Shinde; Krishna Koushik; Lalit kumar Jena; Sreenivasaiah Menasinakai; Vaishali Humnabadkar; Prashanti Madhavapeddi; Halesha D. Basavarajappa; Sreevalli Sharma; Radha Nandishaiah; K.N. Mahesh Kumar; Samit Ganguly; Vijaykamal Ahuja; Sheshagiri Gaonkar; C. N. Naveen Kumar; Derek Ogg; P. Ann Boriack-Sjodin; Vasan K. Sambandamurthy; Sunita M. de Sousa; Sandeep R. Ghorpade

Scaffold hopping from the thiazolopyridine ureas led to thiazolopyridone ureas with potent antitubercular activity acting through inhibition of DNA GyrB ATPase activity. Structural diversity was introduced, by extension of substituents from the thiazolopyridone N-4 position, to access hydrophobic interactions in the ribose pocket of the ATP binding region of GyrB. Further optimization of hydrogen bond interactions with arginines in site-2 of GyrB active site pocket led to potent inhibition of the enzyme (IC50 2 nM) along with potent cellular activity (MIC=0.1 μM) against Mycobacterium tuberculosis (Mtb). Efficacy was demonstrated in an acute mouse model of tuberculosis on oral administration.


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.


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.


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.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Discovery of cofactor-specific, bactericidal Mycobacterium tuberculosis InhA inhibitors using DNA-encoded library technology

Holly H. Soutter; Paolo A. Centrella; Matthew A. Clark; John W. Cuozzo; Christoph E. Dumelin; Marie-Aude Guie; Sevan Habeshian; Anthony D. Keefe; Kaitlyn M. Kennedy; Eric A. Sigel; Dawn M. Troast; Ying Zhang; Andrew D. Ferguson; Gareth Davies; Eleanor R. Stead; Jason Breed; Prashanti Madhavapeddi; Jon Read

Significance The increasing prevalence of multidrug-resistant strains of tuberculosis has created an urgent need for novel therapies to treat tuberculosis infections. Here we have demonstrated the successful utilization of the DNA-encoded X-Chem technology for the discovery inhibitors of Mycobacterium tuberculosis enoyl–acyl-carrier protein (ACP) reductase, InhA, a validated target for the treatment of tuberculosis. The identified inhibitors are cofactor specific and have activity in multiple cellular assays. Crystal structures of representative compounds from five chemical series revealed that the compounds bind adjacent to the NADH cofactor and adopt a variety of conformations, including two previously unreported binding modes. The compounds identified may serve as useful leads in the development of new antibacterial drugs with efficacy against multidrug-resistant tuberculosis. Millions of individuals are infected with and die from tuberculosis (TB) each year, and multidrug-resistant (MDR) strains of TB are increasingly prevalent. As such, there is an urgent need to identify novel drugs to treat TB infections. Current frontline therapies include the drug isoniazid, which inhibits the essential NADH-dependent enoyl–acyl-carrier protein (ACP) reductase, InhA. To inhibit InhA, isoniazid must be activated by the catalase-peroxidase KatG. Isoniazid resistance is linked primarily to mutations in the katG gene. Discovery of InhA inhibitors that do not require KatG activation is crucial to combat MDR TB. Multiple discovery efforts have been made against InhA in recent years. Until recently, despite achieving high potency against the enzyme, these efforts have been thwarted by lack of cellular activity. We describe here the use of DNA-encoded X-Chem (DEX) screening, combined with selection of appropriate physical properties, to identify multiple classes of InhA inhibitors with cell-based activity. The utilization of DEX screening allowed the interrogation of very large compound libraries (1011 unique small molecules) against multiple forms of the InhA enzyme in a multiplexed format. Comparison of the enriched library members across various screening conditions allowed the identification of cofactor-specific inhibitors of InhA that do not require activation by KatG, many of which had bactericidal activity in cell-based assays.


ACS Medicinal Chemistry Letters | 2015

Left-Hand Side Exploration of Novel Bacterial Topoisomerase Inhibitors to Improve Selectivity against hERG Binding

Shahul Hameed P; Praveena Manjrekar; Anandkumar Raichurkar; Vikas Shinde; Jayashree Puttur; Gajanan Shanbhag; Murugan Chinnapattu; Vikas Patil; Suresh Rudrapatana; Sreevalli Sharma; C. N. Naveen Kumar; Radha Nandishaiah; Prashanti Madhavapeddi; Dharmarajan Sriram; Suresh Solapure; Vasan K. Sambandamurthy

Structure-activity relationship (SAR) exploration on the left-hand side (LHS) of a novel class of bacterial topoisomerase inhibitors led to a significant improvement in the selectivity against hERG cardiac channel binding with concomitant potent antimycobacterial activity. Bulky polar substituents at the C-7 position of the naphthyridone ring did not disturb its positioning between two base pairs of DNA. Further optimization of the polar substituents on the LHS of the naphthyridone ring led to potent antimycobacterial activity (Mtb MIC = 0.06 μM) against Mycobacterium tuberculosis (Mtb). Additionally, this knowledge provided a robust SAR understanding to mitigate the hERG risk. This compound class inhibits Mtb DNA gyrase and retains its antimycobacterial activity against moxifloxacin-resistant strains of Mtb. Finally, we demonstrate in vivo proof of concept in an acute mouse model of TB following oral administration of compound 19.


Journal of Biomolecular Screening | 2015

Assays, Surrogates, and Alternative Technologies for a TB Lead Identification Program Targeting DNA Gyrase ATPase

Vaishali Humnabadkar; Prashanti Madhavapeddi; Halesha D. Basavarajappa; Md. Gulebahar Sheikh; Rajendra Rane; Reetobrata Basu; Prateek Verma; Aishwarya Sundaram; Kakoli Mukherjee; Sunita M. de Sousa

Mycobacterium tuberculosis (Mtb) DNA gyrase ATPase was the target of a tuberculosis drug discovery program. The low specific activity of the Mtb ATPase prompted the use of Mycobacterium smegmatis (Msm) enzyme as a surrogate for lead generation, since it had 20-fold higher activity. Addition of GyrA or DNA did not significantly increase the activity of the Msm GyrB ATPase, and an assay was developed using GyrB alone. Inhibition of the Msm ATPase correlated well with inhibition of Mtb DNA gyrase supercoiling across three chemical scaffolds, justifying its use. As the IC50 of compounds approached the enzyme concentration, surrogate assays were used to estimate potencies (e.g., the shift in thermal melt of Mtb GyrB, which correlated well with IC50s >10 nM). Analysis using the Morrison equation enabled determination of K i app s in the sub-nanomolar range. Surface plasmon resonance was used to confirm these IC50s and measure the Kds of binding, but a fragment of Mtb GyrB had to be used. Across three scaffolds, the dissociation half life, t1/2, of the inhibitor-target complex was ≤8 min. This toolkit of assays was developed to track the potency of enzyme inhibition and guide the chemistry for progression of compounds in a lead identification program.


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 Prashanti Madhavapeddi's collaboration.

Researchain Logo
Decentralizing Knowledge