Network


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

Hotspot


Dive into the research topics where Sunil Kumar Panigrahi is active.

Publication


Featured researches published by Sunil Kumar Panigrahi.


Bioorganic & Medicinal Chemistry Letters | 2013

Design and synthesis of 2-substituted benzoxazoles as novel PTP1B inhibitors.

Arun P. Chandrasekharappa; Sangamesh E. Badiger; Pramod Kumar Dubey; Sunil Kumar Panigrahi; Sekhar Reddy V.V.V. Manukonda

A series of benzoxazole compounds containing oxamic acid were synthesized and screened for the PTP1B inhibition. Compound 31d showed best biochemical potency (Ki) of 6.7 μM. Structure-activity relationship were explained with the help of molecular modeling approach.


Bioorganic & Medicinal Chemistry Letters | 2013

Discovery of 7-azaindole based anaplastic lymphoma kinase (ALK) inhibitors: Wild type and mutant (L1196M) active compounds with unique binding mode.

Venkateshwar Rao Gummadi; Sujatha Rajagopalan; Chung Yeng Looi; Mohammadjavad Paydar; Girish Aggunda Renukappa; Bharathi Raja Ainan; Narasimha R. Krishnamurthy; Sunil Kumar Panigrahi; Kumari Mahasweta; Sangeetha Raghuramachandran; Manoj Rajappa; Anuradha Ramanathan; Anirudha Lakshminarasimhan; Murali Ramachandra; Pooi-Fong Wong; Mohammad Rais Mustafa; Srinivas Nanduri; Subramanya Hosahalli

We have identified a novel 7-azaindole series of anaplastic lymphoma kinase (ALK) inhibitors. Compounds 7b, 7 m and 7 n demonstrate excellent potencies in biochemical and cellular assays. X-ray crystal structure of one of the compounds (7 k) revealed a unique binding mode with the benzyl group occupying the back pocket, explaining its potency towards ALK and selectivity over tested kinases particularly Aurora-A. This binding mode is in contrast to that of known ALK inhibitors such as Crizotinib and NVP-TAE684 which occupy the ribose binding pocket, close to DFG motif.


European Journal of Medicinal Chemistry | 2014

Discovery of azetidine based ene-amides as potent bacterial enoyl ACP reductase (FabI) inhibitors

Mohamed Takhi; Kandepu Sreenivas; Chandrashekar K. Reddy; Mahadari Munikumar; Kolakota Praveena; Pabolu Sudheer; Bandaru N.V.M. Rao; Gollamudi Ramakanth; Jampala Sivaranjani; Shardaprasad Mulik; Yeruva R. Reddy; Krishnamurthy N. Rao; Rentala Pallavi; Anirudha Lakshminarasimhan; Sunil Kumar Panigrahi; Thomas Antony; Iskandar Abdullah; Yean K. Lee; Murali Ramachandra; Rohana Yusof; Noorsaadah Abd. Rahman; Hosahalli Subramanya

A novel and potent series of ene-amides featuring azetidines has been developed as FabI inhibitors active against drug resistant Gram-positive pathogens particularly staphylococcal organisms. Most of the compounds from the series possessed excellent biochemical inhibition of Staphylococcus aureus FabI enzyme and whole cell activity against clinically relevant MRSA, MSSA and MRSE organisms which are responsible for significant morbidity and mortality in community as well as hospital settings. The binding mode of one of the leads, AEA16, in Escherichia coli FabI enzyme was determined unambiguously using X-ray crystallography. The lead compounds displayed good metabolic stability in mice liver microsomes and pharmacokinetic profile in mice. The in vivo efficacy of lead AEA16 has been demonstrated in a lethal murine systemic infection model.


Bioorganic & Medicinal Chemistry Letters | 2017

Novel highly selective peroxisome proliferator-activated receptor δ (PPARδ) modulators with pharmacokinetic properties suitable for once-daily oral dosing

Bharat Lagu; Arthur F. Kluge; Ross Fredenburg; Effie Tozzo; Ramesh S. Senaiar; Mahaboobi Jaleel; Sunil Kumar Panigrahi; Nirbhay Kumar Tiwari; Narasimha R. Krishnamurthy; Taisuke Takahashi; Michael A. Patane

Optimization of benzamide PPARδ modulator 1 led to (E)-6-(2-((4-(furan-2-yl)-N-methylbenzamido)methyl)phenoxy)-4-methylhex-4-enoic acid (18), a potent selective PPARδ modulator with significantly improved exposure in multiple species following oral administration.


Molecular Cancer Research | 2016

Abstract A72: Novel NAMPT inhibitors for the treatment of Pancreatic cancer

Dinesh Chikanna; Anirudha Lakshminarasimhan; Vinayak Khairnar; Sunil Kumar Panigrahi; Anuradha Ramanathan; Narasimha Rao; Kishore Narayanan; Sreevalsam Gopinath; Raghuveer Ramachandra; Shekar Chelur; Chetan Pandit; Murali Ramachandra

Pancreatic cancer is reported to be dependent on NAD salvage pathway for its growth and survival. Nicotinamide phosphoribosyl transferase (NAMPT), an enzyme that catalyzes the rate limiting step of NAD biosynthesis is over expressed in a number of cancers. Inhibition of NAMPT with first generation inhibitors has been demonstrated to result in anti-tumor efficacy in preclinical models. Clinical development of first generation NAMPT inhibitors has been hindered because of their poor pharmacological profile, high cytochrome inhibition and possibly mechanism-based toxicities. Therefore, our objective was to develop NAMPT inhibitors with the “best-in-class” profile with strategies for overcoming mechanism-based toxicities. Utilizing structure-guided drug design including determination of co-crystal structures and SAR-based approaches, we have identified a novel chemical series of inhibitors of NAMPT. Optimization of the series for transient target inhibition as a result of reduced binding strength coupled with desirable pharmacokinetic profile to minimize mechanism based toxicity resulted in identification of AU-4869 as the lead compound. AU-4869 showed potent cross-species activity and reduced strength of binding in comparison with first generation NAMPT inhibitors. Anti-proliferative activity of AU-4869 correlated well with NAD depletion in a pancreatic cancer cell line. The anti-proliferative activities were rescued in NAPRT-proficient cell lines with the addition of nicotinic acid due to the NAMPT independent salvage pathway for biosynthesis of NAD, confirming the mechanism of action through NAD depletion. AU-4869 exhibited desirable drug-like properties including solubility, permeability, metabolic stability, lack of CYP & hERG inhibition and pharmacokinetic exposure upon oral dosing. At well-tolerated doses, AU-4869 exhibited superior efficacy at MTD doses in mice xenograft models as compared to first generation inhibitors. Addition of nicotinic acid improved the tolerability of AU-4869 and reversed the effects of mechanism based toxicity in Rodents. Anti-tumor activities of AU-4869 in the presence of nicotinic acid in NAPRT-deficient pancreatic cancer models are currently being evaluated in preclinical models. Citation Format: Dinesh Chikanna, Anirudha Lakshminarasimhan, Vinayak Khairnar, Sunil Panigrahi, Anuradha Ramanathan, Narasimha Rao, Kishore Narayanan, Sreevalsam Gopinath, Raghuveer Ramachandra, Shekar Chelur, Chetan Pandit, Murali Ramachandra. Novel NAMPT inhibitors for the treatment of Pancreatic cancer. [abstract]. In: Proceedings of the AACR Special Conference: Metabolism and Cancer; Jun 7-10, 2015; Bellevue, WA. Philadelphia (PA): AACR; Mol Cancer Res 2016;14(1_Suppl):Abstract nr A72.


Cancer Research | 2016

Abstract 339: Identification of novel covalent inhibitors of K-Ras G12C that are efficacious in a xenograft model of NSCLC

Leena K. Satyam; Dinesh Chikkanna; Aswani K. G; Vinayak Khairnar; Sreekanth Reddy; Vakkapatla Durgaprasad; Kowju Radhakrishna; Sunil Kumar Panigrahi; Anuradha Ramanathan; Kumari Mahasweta; Anirudha Lakshminarasimhan; Narasimha R. K; Vinutha R; Sreevalsam Gopinath; Suryakant Kumar; Mubarak H. Shah; Raghuveer Ramachandra; Kiran A. B; Chetan Pandit; Murali Ramachandra

KRAS is the frequently mutated isoform in RAS driven cancers. The G12C mutation is more predominantly associated with various tumor types over other changes in K-Ras. Although direct targeting of RAS is very challenging, it is possible to selectively target G12C mutant K-Ras using a covalent approach. Mutant specific covalent inhibitors with high selectivity against wild type K-Ras and other GTPases are expected to lead to efficacy with a very high degree of tolerability. Here, we report identification of lead compounds from two distinct chemical series that selectively target K-Ras G12C. Molecular modeling based on the reported crystal structures aided in the identification of these compounds. Covalent binding of the lead compounds to K-Ras G12C was demonstrated by MALDI-TOF. Lead compounds were potent in selectively inhibiting proliferation of cell lines with K-Ras G12C mutation but not with wild type K-Ras. The anti-proliferative activity of the lead compounds correlated well with their potency in a cellular mechanistic assay. Lead compounds from both series exhibited excellent drug-like properties including solubility, metabolic stability, permeability lack of CYP inhibition and desired exposure in pharmacokinetic studies. In a xenograft model of NSCLC, the lead compound demonstrated dose-dependent tumor growth inhibition with excellent tolerability upon oral dosing. In summary, we have identified a novel, potent and selective K-Ras G12C inhibitor with optimized drug-like properties including oral bioavailability and efficacy in a NSCLC derived xenograft model. Toxicity evaluation is ongoing towards progressing the lead compound to the clinic. Citation Format: Leena Khare Satyam, Dinesh Chikkanna, Aswani K. G, Vinayak V. Khairnar, Sreekanth Reddy, Vakkapatla Durgaprasad, Kowju Radhakrishna, Sunil K. Panigrahi, Anuradha Ramanathan, Kumari Mahasweta, Anirudha Lakshminarasimhan, Narasimha R. K, Vinutha R, Sreevalsam Gopinath, Suryakant Kumar, Mubarak H. Shah, Raghuveer Ramachandra, Kiran A. B, Chetan Pandit, Murali Ramachandra. Identification of novel covalent inhibitors of K-Ras G12C that are efficacious in a xenograft model of NSCLC. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 339.


Protein and Peptide Letters | 2013

X-ray structure of PTP1B in complex with a new PTP1B inhibitor.

M.V.V.V. reddy; Chakshumathi Ghadiyaram; Sunil Kumar Panigrahi; Narasimha R. Krishnamurthy; Subramanya Hosahalli; Arun P. Chandrasekharappa; Deepankar Manna; Sangamesh E. Badiger; Pramod Kumar Dubey; Lakshmi Narasu Mangamoori

Protein tyrosine phosphatase 1B (PTP1B) is a prototype non receptor cytoplasmic PTPase enzyme that has been implicated in regulation of insulin and leptin signaling pathways. Studies on PTP1B knockout mice and PTP1B antisense treated mice suggested that inhibition of PTP1B would be an effective strategy for the treatment of type II diabetes and obesity. Here we report the X-ray structure of PTP1B in complex with compound IN1834-146C (PDB ID 4I8N). The crystals belong to P3121 space group with cell dimensions (a = b = 87.89 Å, c = 103.68 Å) diffracted to 2.5 Å. The crystal structure contained one molecule of protein in the asymmetric unit and was solved by molecular replacement method. The compound engages both catalytic site and allosteric sites of PTP1B protein. We described the molecular interaction of the compound with the active site residues of PTP1B in this crystal structure report.


ACS Medicinal Chemistry Letters | 2018

Selective PPARδ Modulators Improve Mitochondrial Function: Potential Treatment for Duchenne Muscular Dystrophy (DMD)

Bharat R. Lagu; Arthur F. Kluge; Effie Tozzo; Ross Fredenburg; Eric L. Bell; Matthew M. Goddeeris; Peter Dwyer; Andrew Basinski; Ramesh S. Senaiar; Mahaboobi Jaleel; Nirbhay Kumar Tiwari; Sunil Kumar Panigrahi; Narasimha R. Krishnamurthy; Taisuke Takahashi; Michael A. Patane

The X-ray structure of the previously reported PPARδ modulator 1 bound to the ligand binding domain (LBD) revealed that the amide moiety in 1 exists in the thermodynamically disfavored cis-amide orientation. Isosteric replacement of the cis-amide with five-membered heterocycles led to the identification of imidazole 17 (MA-0204), a potent, selective PPARδ modulator with good pharmacokinetic properties. MA-0204 was tested in vivo in mice and in vitro in patient-derived muscle myoblasts (from Duchenne Muscular Dystrophy (DMD) patients); 17 altered the expression of PPARδ target genes and improved fatty acid oxidation, which supports the therapeutic hypothesis for the study of MA-0204 in DMD patients.


Cancer Research | 2017

Abstract 5108: Potent small molecule compounds that selectively inhibit proliferation of ABC-DLBCL cell lines

Leena K. Satyam; Dinesh Chikkanna; Vinayak Khairnar; Manoj Kumar Pothuganti; Sunil Kumar Panigrahi; Anirudha Lakshminarasimhan; Narasimha Rao; Wesley Roy Balasubramanian; Sandeep Patil; Sreevalsam Gopinath; Gunta Upendra; Jwala Nagaraj; Kiran Aithal; Vijay Ahuja; Sanjeev Giri; Chetan Pandit; Murali Ramachandra

Diffuse large B cell lymphoma (DLBCL), which accounts for 25% of all lymphomas cases, has been classified into molecular subtypes including germinal center B cell like (GCB) DLBCL, activated B cell-like (ABC) DLBCL, and primary mediastinal B cell lymphoma (PMBL). Among these subtypes, patients with ABC-DLBCLs have the worst prognosis because of the high chemo-resistance, and require effective therapies. Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 (MALT1) protease activity is linked to the pathogenesis of ABC-DLBCL. Therefore, a focused library of covalent compounds selected based on molecular docking on the reported crystal structure was screened for selective sensitivity to ABC-DLBCL, but not GCB-DLBCL cell lines. Optimization of initial hits resulted in the identification of lead compounds with an anti-proliferative EC50 of 10 over cellular EC50 for up to 8 hours. The lead compounds showed dose-dependent tumor growth inhibition in a xenograft model upon oral dosing. In summary, we have identified novel and potent MALT1 inhibitors capable of selectively inhibiting proliferation of DLBCL cell lines with optimized drug-like properties including oral bioavailability. The data presented here strongly support further development of these compounds for DLBCL and other indications. Citation Format: Leena Khare Satyam, Dinesh Chikkanna, Vinayak Khairnar, Manoj Pothuganti, Sunil Panigrahi, Anirudha Lakshminarasimhan, Narasimha Rao, Wesley Balasubramanian, Sandeep Patil, Sreevalsam Gopinath, Gunta Upendra, Jwala Nagaraj, Kiran Aithal, Vijay Ahuja, Sanjeev Giri, Chetan Pandit, Murali Ramachandra. Potent small molecule compounds that selectively inhibit proliferation of ABC-DLBCL cell lines [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 5108. doi:10.1158/1538-7445.AM2017-5108


Cancer Research | 2015

Abstract 5367: Discovery of dihydro-isoxazole derivatives as novel inhibitors of NAMPT for the treatment of multiple myeloma

Dinesh Chikkanna; Anirudha Lakshminarasimhan; Vinayak Khairnar; Sunil Kumar Panigrahi; Anuradha Ramanathan; Sumalatha Rani; Narasimha Rao; S Karthikeyan; Kishore Narayanan; Sreevalsam Gopinath; Raghuveer Ramachandra; Charamanna K B; Shekar Chelur; Chetan Pandit; Murali Ramachandra

Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA Nicotinamide phosphoribosyl transferase (NAMPT) is the enzyme that catalyzes the rate-limiting step in the salvage pathway of Nicotinamide Adenine Dinucleotide (NAD) biosynthesis. NAMPT is overexpressed in a number of cancers, and inhibition of NAMPT has been shown to result in anti-tumor efficacy in preclinical models. Clinical development to first generation NAMPT inhibitors has been hindered because of their poor pharmacological profile, high cytochrome inhibition and possibly mechanism-based toxicities. Therefore, we sought to develop NAMPT inhibitors with the “best-in-class” profile for overcoming mechanism-based toxicities and/or resistance to current therapies. Utilizing structure-guided drug design including determination of co-crystal structures and SAR-based approaches, we have identified a novel chemical series of inhibitors of NAMPT. Optimization of the series for transient target inhibition as a result of reduced binding strength coupled with desirable pharmacokinetic profile to minimize mechanism based toxicity resulted in identification of AU-4869 as the Lead compound.AU-4869 showed potent cross-species activity and reduced strength of binding in comparison with first generation NAMPT inhibitors. Anti-proliferative activity of AU-4869 correlated well with NAD depletion in multiple cell lines derived from multiple myeloma and Pancreatic cancers. The anti-proliferative activities were rescued in NAPRT-proficient cell lines with the addition of NA due to the NAMPT independent salvage pathway for biosynthesis of NAD, confirming the mechanism of action through NAD depletion. AU-4869 exhibited desirable drug-like properties including solubility, permeability, metabolic stability, lack of CYP & hERG inhibition and pharmacokinetic exposure upon oral dosing. At well-tolerated doses, AU-4869 exhibited superior efficacy at MTD doses in mice xenograft models (pancreatic cancer and multiple myeloma) as compared to FK866 and GNE-617. In view of the ability of NAMPT inhibitors to enhance sensitivity to a number of targeted agents and overcome resistance to available therapies such as bortezomib in multiple myeloma, further pre-clinical development of AU-4869 for multiple myeloma is ongoing. Citation Format: Dinesh Chikkanna, Anirudha Lakshminarasimhan, Vinayak Khairnar, Sunil panigrahi, Anuradha Ramanathan, Sumalatha Rani, Narasimha Rao, Karthikeyan S, Kishore Narayanan, Sreevalsam Gopinath, Raghuveer Ramachandra, Charamanna K B, Shekar Chelur, Chetan Pandit, Murali Ramachandra. Discovery of dihydro-isoxazole derivatives as novel inhibitors of NAMPT for the treatment of multiple myeloma. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 5367. doi:10.1158/1538-7445.AM2015-5367

Collaboration


Dive into the Sunil Kumar Panigrahi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge