Network


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

Hotspot


Dive into the research topics where Vikas Shinde is active.

Publication


Featured researches published by Vikas Shinde.


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.


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.


Antimicrobial Agents and Chemotherapy | 2014

Assessment of Mycobacterium tuberculosis Pantothenate Kinase Vulnerability through Target Knockdown and Mechanistically Diverse Inhibitors

B. K. Kishore Reddy; Sudhir Landge; Sudha Ravishankar; Vikas Patil; Vikas Shinde; Subramanyam J. Tantry; Manoj Kale; Anandkumar Raichurkar; Sreenivasaiah Menasinakai; Naina Vinay Mudugal; Anisha Ambady; Anirban Ghosh; Ragadeepthi Tunduguru; Parvinder Kaur; Ragini Singh; Naveen Kumar; Aishwarya Sundaram; Jyothi Bhat; Vasan K. Sambandamurthy; Christofer Björkelid; T. Alwyn Jones; Kaveri Das; Balachandra Bandodkar; Krishnan Malolanarasimhan; Kakoli Mukherjee

ABSTRACT Pantothenate kinase (PanK) catalyzes the phosphorylation of pantothenate, the first committed and rate-limiting step toward coenzyme A (CoA) biosynthesis. In our earlier reports, we had established that the type I isoform encoded by the coaA gene is an essential pantothenate kinase in Mycobacterium tuberculosis, and this vital information was then exploited to screen large libraries for identification of mechanistically different classes of PanK inhibitors. The present report summarizes the synthesis and expansion efforts to understand the structure-activity relationships leading to the optimization of enzyme inhibition along with antimycobacterial activity. Additionally, we report the progression of two distinct classes of inhibitors, the triazoles, which are ATP competitors, and the biaryl acetic acids, with a mixed mode of inhibition. Cocrystallization studies provided evidence of these inhibitors binding to the enzyme. This was further substantiated with the biaryl acids having MIC against the wild-type M. tuberculosis strain and the subsequent establishment of a target link with an upshift in MIC in a strain overexpressing PanK. On the other hand, the ATP competitors had cellular activity only in a M. tuberculosis knockdown strain with reduced PanK expression levels. Additionally, in vitro and in vivo survival kinetic studies performed with a M. tuberculosis PanK (MtPanK) knockdown strain indicated that the target levels have to be significantly reduced to bring in growth inhibition. The dual approaches employed here thus established the poor vulnerability of PanK in M. tuberculosis.


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.


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 Chemical Research-s | 2013

A convenient synthesis of 5-substituted 2-amino-1,3,4-oxadiazoles from corresponding acylthiosemicarbazides using iodine and Oxone®

Vikas Shinde; Bheemarao G. Ugarkar; Sandeep R. Ghorpade

A convenient methodology has been developed for the synthesis of substituted 2-amino-1,3,4-oxadiazoles from corresponding acylthiosemicarbazides using catalytic amount of iodine/KI in the presence of Oxone® as a bulk oxidant. This offers the advantage of short reaction times and substrate variability which is suitable for the rapid production of analogues required for lead optimisation programmes. The method could also be useful for large-scale synthesis because of the mild reaction conditions and the use commercially inexpensive and safe reagents.


Synthetic Communications | 2013

Short and Efficient Synthesis of Oxazinone- and Thiazinone-Containing Bicyclic Heteroaromatic Aldehydes

P Shahul Hameed; Gajanan Shanbhag; Vikas Shinde; Murugan Chinnapattu; Praveena Manjrekar; Jayashree Puttur; Vikas Patil; Bheemarao G. Ugarkar

Abstract Short and efficient route for the synthesis of oxazinone- and thiazinone-containing bicyclic heteroaromatic aldehydes, which involves the key step of palladium-catalyzed reductive carbonylation, is described. Overall routes for the synthesis of these aldehydes are short, versatile, and scalable with good yields of the product. [Supplementary materials are available for this article. Go to the publishers online edition of Synthetic Communications® for the following free supplemental resource(s): Full experimental and spectral details.] GRAPHICAL ABSTRACT


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 Vikas Shinde's collaboration.

Researchain Logo
Decentralizing Knowledge