Network


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

Hotspot


Dive into the research topics where Srinivasan Thangathirupathy is active.

Publication


Featured researches published by Srinivasan Thangathirupathy.


Neuropsychopharmacology | 2016

Negative Allosteric Modulators Selective for The NR2B Subtype of The NMDA Receptor Impair Cognition in Multiple Domains

Michael R. Weed; Mark Bookbinder; Joseph Polino; Deborah Keavy; Rudolf N. Cardinal; Jean Simmermacher-Mayer; Fu Ni L Cometa; Dalton King; Srinivasan Thangathirupathy; John E. Macor; Linda J. Bristow

Antidepressant activity of N-methyl-D-aspartate (NMDA) receptor antagonists and negative allosteric modulators (NAMs) has led to increased investigation of their behavioral pharmacology. NMDA antagonists, such as ketamine, impair cognition in multiple species and in multiple cognitive domains. However, studies with NR2B subtype-selective NAMs have reported mixed results in rodents including increased impulsivity, no effect on cognition, impairment or even improvement of some cognitive tasks. To date, the effects of NR2B-selective NAMs on cognitive tests have not been reported in nonhuman primates. The current study evaluated two selective NR2B NAMs, CP101,606 and BMT-108908, along with the nonselective NMDA antagonists, ketamine and AZD6765, in the nonhuman primate Cambridge Neuropsychological Test Automated Battery (CANTAB) list-based delayed match to sample (list-DMS) task. Ketamine and the two NMDA NR2B NAMs produced selective impairments in memory in the list-DMS task. AZD6765 impaired performance in a non-specific manner. In a separate cohort, CP101,606 impaired performance of the nonhuman primate CANTAB visuo-spatial Paired Associates Learning (vsPAL) task with a selective impairment at more difficult conditions. The results of these studies clearly show that systemic administration of a selective NR2B NAM can cause transient cognitive impairment in multiple cognitive domains.


PLOS ONE | 2016

The qEEG Signature of Selective NMDA NR2B Negative Allosteric Modulators; A Potential Translational Biomarker for Drug Development.

Deborah Keavy; Linda J. Bristow; Digavalli V. Sivarao; Margaret Batchelder; Dalton King; Srinivasan Thangathirupathy; John E. Macor; Michael R. Weed

The antidepressant activity of the N-methyl-D-aspartate (NMDA) receptor channel blocker, ketamine, has led to the investigation of negative allosteric modulators (NAMs) selective for the NR2B receptor subtype. The clinical development of NR2B NAMs would benefit from a translational pharmacodynamic biomarker that demonstrates brain penetration and functional inhibition of NR2B receptors in preclinical species and humans. Quantitative electroencephalography (qEEG) is a translational measure that can be used to demonstrate pharmacodynamic effects across species. NMDA receptor channel blockers, such as ketamine and phencyclidine, increase the EEG gamma power band, which has been used as a pharmacodynamic biomarker in the development of NMDA receptor antagonists. However, detailed qEEG studies with ketamine or NR2B NAMs are lacking in nonhuman primates. The aim of the present study was to determine the effects on the qEEG power spectra of the NR2B NAMs traxoprodil (CP-101,606) and BMT-108908 in nonhuman primates, and to compare them to the NMDA receptor channel blockers, ketamine and lanicemine. Cynomolgus monkeys were surgically implanted with EEG radio-telemetry transmitters, and qEEG was measured after vehicle or drug administration. The relative power for a number of frequency bands was determined. Ketamine and lanicemine increased relative gamma power, whereas the NR2B NAMs traxoprodil and BMT-108908 had no effect. Robust decreases in beta power were elicited by ketamine, traxoprodil and BMT-108908; and these agents also produced decreases in alpha power and increases in delta power at the doses tested. These results suggest that measurement of power spectra in the beta and delta bands may represent a translational pharmacodynamic biomarker to demonstrate functional effects of NR2B NAMs. The results of these studies may help guide the selection of qEEG measures that can be incorporated into early clinical evaluation of NR2B NAMs in healthy humans.


British Journal of Pharmacology | 2017

Modulation of Kv11.1 (hERG) channels by 5‐(((1H–indazol‐5‐yl)oxy)methyl)‐N‐(4‐(trifluoromethoxy)phenyl)pyrimidin‐2‐amine (ITP‐2), a novel small molecule activator

Harinath Sale; Samrat Roy; Jayakumar Sankara Warrier; Srinivasan Thangathirupathy; Yoganand Vadari; Shruthi K Gopal; Prasad Krishnamurthy; Manjunath Ramarao

Activators of Kv11.1 (hERG) channels have potential utility in the treatment of acquired and congenital long QT (LQT) syndrome. Here, we describe a new hERG channel activator, 5‐(((1H–indazol‐5‐yl)oxy)methyl)‐N‐(4‐(trifluoromethoxy)phenyl)pyrimidin‐2‐amine (ITP‐2), with a chemical structure distinct from previously reported compounds.


ACS Medicinal Chemistry Letters | 2018

BMS-986163, a Negative Allosteric Modulator of GluN2B with Potential Utility in Major Depressive Disorder

Jayakumar Sankara Warrier; Srinivasan Thangathirupathy; Jianliang Shi; George N. Karageorge; Bradley C. Pearce; Alicia Ng; Hyunsoo Park; James Kempson; Jianqing Li; Huiping Zhang; Arvind Mathur; Aliphedi B. Reddy; G. Nagaraju; Gopikishan Tonukunuru; Grandhi V. R.K. M. Gupta; Manjunatha Kamble; Raju Mannoori; Srinivas Cheruku; Srinivas Jogi; Jyoti Gulia; Tanmaya Bastia; Charulatha Sanmathi; Jayant Aher; Rajareddy Kallem; Bettadapura N. Srikumar; Kumar Kuchibhotla Vijaya; Pattipati S. Naidu; Mahesh Paschapur; Narasimharaju Kalidindi; Reeba K. Vikramadithyan

There is a significant unmet medical need for more efficacious and rapidly acting antidepressants. Toward this end, negative allosteric modulators of the N-methyl-d-aspartate receptor subtype GluN2B have demonstrated encouraging therapeutic potential. We report herein the discovery and preclinical profile of a water-soluble intravenous prodrug BMS-986163 (6) and its active parent molecule BMS-986169 (5), which demonstrated high binding affinity for the GluN2B allosteric site (Ki = 4.0 nM) and selective inhibition of GluN2B receptor function (IC50 = 24 nM) in cells. The conversion of prodrug 6 to parent 5 was rapid in vitro and in vivo across preclinical species. After intravenous administration, compounds 5 and 6 have exhibited robust levels of ex vivo GluN2B target engagement in rodents and antidepressant-like activity in mice. No significant off-target activity was observed for 5, 6, or the major circulating metabolites met-1 and met-2. The prodrug BMS-986163 (6) has demonstrated an acceptable safety and toxicology profile and was selected as a preclinical candidate for further evaluation in major depressive disorder.


Journal of Pharmacology and Experimental Therapeutics | 2017

Preclinical Characterization of (R)-3-((3S,4S)-3-fluoro-4-(4-hydroxyphenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one (BMS-986169), a Novel, Intravenous, Glutamate N-Methyl-D-Aspartate 2B (GluN2B) Receptor Negative Allosteric Modulator with Potential in Major Depressive Disorder

Linda J. Bristow; Jyoti Gulia; Michael R. Weed; Bettadapura N. Srikumar; Yu-Wen Li; John D. Graef; Pattipati S. Naidu; Charulatha Sanmathi; Jayant Aher; Tanmaya Bastia; Mahesh Paschapur; Narasimharaju Kalidindi; Kuchibhotla Vijaya Kumar; Thaddeus F. Molski; Rick L. Pieschl; Alda Fernandes; Jeffrey M. Brown; Digavalli V. Sivarao; Kimberly Newberry; Mark Bookbinder; Joseph Polino; Deborah Keavy; Amy Newton; Eric Shields; Jean Simmermacher; James Kempson; Jianqing Li; Huiping Zhang; Arvind Mathur; Raja Reddy Kallem

(R)-3-((3S,4S)-3-fluoro-4-(4-hydroxyphenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one (BMS-986169) and the phosphate prodrug 4-((3S,4S)-3-fluoro-1-((R)-1-(4-methylbenzyl)-2-oxopyrrolidin-3-yl)piperidin-4-yl)phenyl dihydrogen phosphate (BMS-986163) were identified from a drug discovery effort focused on the development of novel, intravenous glutamate N-methyl-d-aspartate 2B receptor (GluN2B) negative allosteric modulators (NAMs) for treatment-resistant depression (TRD). BMS-986169 showed high binding affinity for the GluN2B subunit allosteric modulatory site (Ki = 4.03–6.3 nM) and selectively inhibited GluN2B receptor function in Xenopus oocytes expressing human N-methyl-d-aspartate receptor subtypes (IC50 = 24.1 nM). BMS-986169 weakly inhibited human ether-a-go-go–related gene channel activity (IC50 = 28.4 μM) and had negligible activity in an assay panel containing 40 additional pharmacological targets. Intravenous administration of BMS-986169 or BMS-986163 dose-dependently increased GluN2B receptor occupancy and inhibited in vivo [3H](+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine ([3H]MK-801) binding, confirming target engagement and effective cleavage of the prodrug. BMS-986169 reduced immobility in the mouse forced swim test, an effect similar to intravenous ketamine treatment. Decreased novelty suppressed feeding latency, and increased ex vivo hippocampal long-term potentiation was also seen 24 hours after acute BMS-986163 or BMS-986169 administration. BMS-986169 did not produce ketamine-like hyperlocomotion or abnormal behaviors in mice or cynomolgus monkeys but did produce a transient working memory impairment in monkeys that was closely related to plasma exposure. Finally, BMS-986163 produced robust changes in the quantitative electroencephalogram power band distribution, a translational measure that can be used to assess pharmacodynamic activity in healthy humans. Due to the poor aqueous solubility of BMS-986169, BMS-986163 was selected as the lead GluN2B NAM candidate for further evaluation as a novel intravenous agent for TRD.


Archive | 2015

Selective NR2B antagonists

Dalton King; Lorin A. Thompson; Jianliang Shi; Srinivasan Thangathirupathy; Jayakumar Sankara Warrier; Imadul Islam; John E. Macor


Archive | 2016

INHIBITORS OF INDOLEAMINE 2,3-DIOXYGENASE FOR THE TREATMENT OF CANCER

James Aaron Balog; Emily Charlotte Cherney; Weiwei Guo; Audris Huang; Jay A. Markwalder; Steven P. Seitz; Weifang Shan; David K. Williams; Natesan Murugesan; Susheel Jethanand Nara; Saumya Roy; Soodamani Thangavel; Ramesh Sistla; Srinivas Cheruku; Srinivasan Thangathirupathy; Yadagiri Kanyaboina; Nagalakshmi Pulicharla


Archive | 2016

INHIBIDORES DE LA FUNCIÓN ENZIMÁTICA DE LA INDOLAMINA-2,3-DIOXIGENASA (IDO) Y COMPOSICIONES FARMACÉUTICAS QUE LOS CONTIENEN

Natesan Murugesan; Srinivasan Thangathirupathy; Balog James Aaron; Cherney Emily Charlotte; Weiwei Guo; Audris Huang; Markwalder Jay A; Seitz Steven P; Weifang Shan; Williams David K; Nara Susheel Jethanand; Saumya Roy; Soodamani Thangavel; Sistla Ramesh Kumar; Srinivas Cheruku; Yadagiri Kanyaboina; Nagalakshmi Pulicharla


Archive | 2016

Inhibiteurs de l'indoléamine 2,3-dioxygénase pour le traitement du cancer

James Aaron Balog; Emily Charlotte Cherney; Jay A. Markwalder; Steven P. Seitz; Weifang Shan; David K. Williams; Susheel Jethanand Nara; Saumya Roy; Soodamani Thangavel; Ramesh Sistla; Srinivas Cheruku; Srinivasan Thangathirupathy; Yadagiri Kanyaboina; Nagalakshmi Pulicharla; Huang Audris


Archive | 2015

COMPUESTOS ANTAGONISTAS SELECTIVOS DE N-METIL-D-ASPARTATO (NMDA) SUBTIPO NR2B Y COMPOSICIONES QUE LOS CONTIENEN

Iii Lorin A Thompson; Macor John E; Dalton King; Janliang Shi; Srinivasan Thangathirupathy; Warrier Jayakumar Sankara; Imadul Islam

Collaboration


Dive into the Srinivasan Thangathirupathy'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge