Network


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

Hotspot


Dive into the research topics where Manoranjan Panda is active.

Publication


Featured researches published by Manoranjan Panda.


Journal of Computational Chemistry | 2008

Explicit ion, implicit water solvation for molecular dynamics of nucleic acids and highly charged molecules

Ninad V. Prabhu; Manoranjan Panda; Qingyi Yang; Kim A. Sharp

An explicit ion, implicit water solvent model for molecular dynamics was developed and tested with DNA and RNA simulations. The implicit water model uses the finite difference Poisson (FDP) model with the smooth permittivity method implemented in the OpenEye ZAP libraries. Explicit counter‐ions, co‐ions, and nucleic acid were treated with a Langevin dynamics molecular dynamics algorithm. Ion electrostatics is treated within the FDP model when close to the solute, and by the Coulombic model when far from the solute. The two zone model reduces computation time, but retains an accurate treatment of the ion atmosphere electrostatics near the solute. Ion compositions can be set to reproduce specific ionic strengths. The entire ion/water treatment is interfaced with the molecular dynamics package CHARMM. Using the CHARMM‐ZAPI software combination, the implicit solvent model was tested on A and B form duplex DNA, and tetraloop RNA, producing stable simulations with structures remaining close to experiment. The model also reproduced the A to B duplex DNA transition. The effect of ionic strength, and the structure of the counterion atmosphere around B form duplex DNA were also examined.


Bioorganic & Medicinal Chemistry Letters | 2013

Synthesis and structure activity relationship of imidazo[1,2-a]pyridine-8-carboxamides as a novel antimycobacterial lead series.

Manoranjan Panda; Kakoli Mukherjee; Nilanjana Roy Choudhury; Subramanyam J. Tantry; Chaitanya Kumar Kedari; Sreevalli Sharma; V. K. Ramya; Supreeth Guptha; Vasan K. Sambandamurthy

Imidazo[1,2-a]pyridine-8-carboxamides as a novel antimycobacterial lead were generated by whole cell screening of a focused library against Mycobacterium tuberculosis. Herein, we describe the synthesis and structure activity relationship evaluation of this class of inhibitors and the optimization of physicochemical properties. These are selective inhibitors of Mycobacterium tuberculosis with no activity on either gram positive or gram negative pathogens.


Journal of Medicinal Chemistry | 2014

Diarylthiazole: an antimycobacterial scaffold potentially targeting PrrB-PrrA two-component system.

Eknath Bellale; Maruti Naik; Varun Vb; Anisha Ambady; Ashwini Narayan; Sudha Ravishankar; Parvinder Kaur; Robert E. McLaughlin; James Whiteaker; Sapna Morayya; Supreeth Guptha; Sreevalli Sharma; Anandkumar Raichurkar; Disha Awasthy; Vijayshree Achar; Prakash Vachaspati; Balachandra Bandodkar; Manoranjan Panda; Monalisa Chatterji

Diarylthiazole (DAT), a hit from diversity screening, was found to have potent antimycobacterial activity against Mycobacterium tuberculosis (Mtb). In a systematic medicinal chemistry exploration, we demonstrated chemical opportunities to optimize the potency and physicochemical properties. The effort led to more than 10 compounds with submicromolar MICs and desirable physicochemical properties. The potent antimycobacterial activity, in conjunction with low molecular weight, made the series an attractive lead (antibacterial ligand efficiency (ALE)>0.4). The series exhibited excellent bactericidal activity and was active against drug-sensitive and resistant Mtb. Mutational analysis showed that mutations in prrB impart resistance to DAT compounds but not to reference drugs tested. The sensor kinase PrrB belongs to the PrrBA two component system and is potentially the target for DAT. PrrBA is a conserved, essential regulatory mechanism in Mtb and has been shown to have a role in virulence and metabolic adaptation to stress. Hence, DATs provide an opportunity to understand a completely new target system for antimycobacterial drug discovery.


Journal of Medicinal Chemistry | 2015

Structure Guided Lead Generation for M. Tuberculosis Thymidylate Kinase (Mtb Tmk): Discovery of 3-Cyanopyridone and 1,6-Naphthyridin-2-One as Potent Inhibitors.

Maruti Naik; Anandkumar Raichurkar; Balachandra Bandodkar; Begur V. Varun; Shantika Bhat; Rajesh Kalkhambkar; Kannan Murugan; Rani Menon; Jyothi Bhat; Beena Paul; Harini Iyer; Syeed Hussein; Julie Tucker; Martin Vogtherr; Kevin J. Embrey; Helen McMiken; Swati Prasad; Adrian Liam Gill; Bheemarao G. Ugarkar; Janani Venkatraman; Jon Read; Manoranjan Panda

M. tuberculosis thymidylate kinase (Mtb TMK) has been shown in vitro to be an essential enzyme in DNA synthesis. In order to identify novel leads for Mtb TMK, we performed a high throughput biochemical screen and an NMR based fragment screen through which we discovered two novel classes of inhibitors, 3-cyanopyridones and 1,6-naphthyridin-2-ones, respectively. We describe three cyanopyridone subseries that arose during our hit to lead campaign, along with cocrystal structures of representatives with Mtb TMK. Structure aided optimization of the cyanopyridones led to single digit nanomolar inhibitors of Mtb TMK. Fragment based lead generation, augmented by crystal structures and the SAR from the cyanopyridones, enabled us to drive the potency of our 1,6-naphthyridin-2-one fragment hit from 500 μM to 200 nM while simultaneously improving the ligand efficiency. Cyanopyridone derivatives containing sulfoxides and sulfones showed cellular activity against M. tuberculosis. To the best of our knowledge, these compounds are the first reports of non-thymidine-like inhibitors of Mtb TMK.


ACS Medicinal Chemistry Letters | 2014

2-Phenylindole and Arylsulphonamide: Novel Scaffolds Bactericidal against Mycobacterium tuberculosis.

Maruti Naik; Sandeep R. Ghorpade; Lalit kumar Jena; Gopinath Gorai; Ashwini Narayan; Supreeth Guptha; Sreevalli Sharma; Neela Dinesh; Parvinder Kaur; Radha Nandishaiah; Jyothi Bhat; Gayathri Balakrishnan; Vaishali Humnabadkar; Lava Naviri; Pallavi Khadtare; Manoranjan Panda; Pravin S. Iyer; Monalisa Chatterji

A cellular activity-based screen on Mycobacterium tuberculosis (Mtb) H37Rv using a focused library from the AstraZeneca corporate collection led to the identification of 2-phenylindoles and arylsulphonamides, novel antimycobacterial scaffolds. Both the series were bactericidal in vitro and in an intracellular macrophage infection model, active against drug sensitive and drug resistant Mtb clinical isolates, and specific to mycobacteria. The scaffolds showed promising structure-activity relationships; compounds with submicromolar cellular potency were identified during the hit to lead exploration. Furthermore, compounds from both scaffolds were tested for inhibition of known target enzymes or pathways of antimycobacterial drugs including InhA, RNA polymerase, DprE1, topoisomerases, protein synthesis, and oxidative-phosphorylation. Compounds did not inhibit any of the targets suggesting the potential of a possible novel mode of action(s). Hence, both scaffolds provide the opportunity to be developed further as leads and tool compounds to uncover novel mechanisms for tuberculosis drug discovery.


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.


Medicinal Chemistry Research | 2015

Novel lead generation of an anti-tuberculosis agent active against non-replicating mycobacteria: exploring hybridization of pyrazinamide with multiple fragments

Shankar D. Markad; Parvinder Kaur; B. K. Kishore Reddy; Murugan Chinnapattu; Anandkumar Raichurkar; Radha Nandishaiah; Manoranjan Panda; Pravin S. Iyer

The key to shortening tuberculosis (TB) drug regimen lies in eliminating the reservoir of non-replicating persistent (NRP) Mycobacterium tuberculosis (Mtb). Pyrazinamide (PZA) is the only known drug used as part of a combination therapy that is believed to kill NRP Mtb and achieve sterilization. PZA is active only under low pH screening conditions. Screening and identification of NRP-active anti-TB compounds are severely limited because compounds are usually inactive under regular assay conditions. In an effort to design novel NRP-active anti-TB compounds, we used pyrazinamide as a core and hybridized it with the fragments derived from marketed drugs. One of these designs, compound 8, was a hybrid with fluoroquinolone. This compound exhibited >10 fold improvement in NRP activity under low pH condition as compared to pyrazinamide and a modest activity (0.8 log10 kill) under nutritionally starved NRP condition. Furthermore, compound 8 was active against fluoroquinolone-resistant strains and did not show any activity in a DNA supercoiling assay (gyrase inhibition), suggesting that its mechanism of action is not that of the parent fluoroquinolone. These results provide a novel avenue in the exploration of new chemotypes that are active against non-replicating Mtb.


ACS Chemical Biology | 2014

Pyrazolopyrimidines Establish MurC as a Vulnerable Target in Pseudomonas aeruginosa and Escherichia coli

Shahul Hameed P; Praveena Manjrekar; Murugan Chinnapattu; Vaishali Humnabadkar; Gajanan Shanbhag; Chaitanyakumar Kedari; Naina Vinay Mudugal; Anisha Ambady; Boudewijn L. M. de Jonge; Claire Sadler; Beena Paul; Shubha Sriram; Parvinder Kaur; Supreeth Guptha; Anandkumar Raichurkar; Paul R. Fleming; Charles J. Eyermann; David C. McKinney; Vasan K. Sambandamurthy; Manoranjan Panda; Sudha Ravishankar

The bacterial peptidoglycan biosynthesis pathway provides multiple targets for antibacterials, as proven by the clinical success of β-lactam and glycopeptide classes of antibiotics. The Mur ligases play an essential role in the biosynthesis of the peptidoglycan building block, N-acetyl-muramic acid-pentapeptide. MurC, the first of four Mur ligases, ligates l-alanine to UDP-N-acetylmuramic acid, initiating the synthesis of pentapeptide precursor. Therefore, inhibiting the MurC enzyme should result in bacterial cell death. Herein, we report a novel class of pyrazolopyrimidines with subnanomolar potency against both Escherichia coli and Pseudomonas aeruginosa MurC enzymes, which demonstrates a concomitant bactericidal activity against efflux-deficient strains. Radio-labeled precursor incorporation showed these compounds selectively inhibited peptidoglycan biosynthesis, and genetic studies confirmed the target of pyrazolopyrimidines to be MurC. In the presence of permeability enhancers such as colistin, pyrazolopyrimidines exhibited low micromolar MIC against the wild-type bacteria, thereby, indicating permeability and efflux as major challenges for this chemical series. Our studies provide biochemical and genetic evidence to support the essentiality of MurC and serve to validate the attractiveness of target for antibacterial discovery.


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.


ChemMedChem | 2016

Nitroarenes as Antitubercular Agents: Stereoelectronic Modulation to Mitigate Mutagenicity

Sudhir Landge; Anupriya Kumar; João Neres; Kannan Murugan; Claire Sadler; Mick D. Fellows; Vaishali Humnabadkar; Prakash Vachaspati; Anandkumar Raichurkar; Sreevalli Sharma; Sudha Ravishankar; Supreeth Guptha; Vasan K. Sambandamurthy; Tanjore S. Balganesh; Bheemarao G. Ugarkar; V. Balasubramanian; Balachandra Bandodkar; Manoranjan Panda

Nitroarenes are less preferred in drug discovery due to their potential to be mutagenic. However, several nitroarenes were shown to be promising antitubercular agents with specific modes of action, namely, nitroimidazoles and benzothiazinones. The nitro group in these compounds is activated through different mechanisms, both enzymatic and non‐enzymatic, in mycobacteria prior to binding to the target of interest. From a whole‐cell screening program, we identified a novel lead nitrobenzothiazole (BT) series that acts by inhibition of decaprenylphosphoryl‐β‐d‐ribose 2′‐epimerase (DprE1) of Mycobacterium tuberculosis (Mtb). The lead was found to be mutagenic to start with. Our efforts to mitigate mutagenicity resulted in the identification of 6‐methyl‐7‐nitro‐5‐(trifluoromethyl)‐1,3‐benzothiazoles (cBTs), a novel class of antitubercular agents that are non‐mutagenic and exhibit an improved safety profile. The methyl group ortho to the nitro group decreases the electron affinity of the series, and is hence responsible for the non‐mutagenic nature of these compounds. Additionally, the co‐crystal structure of cBT in complex with Mtb DprE1 established the mode of binding. This investigation led to a new non‐mutagenic antitubercular agent and demonstrates that the mutagenic nature of nitroarenes can be solved by modulation of stereoelectronic properties.

Collaboration


Dive into the Manoranjan Panda's collaboration.

Researchain Logo
Decentralizing Knowledge