Network


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

Hotspot


Dive into the research topics where Madhu Chintala is active.

Publication


Featured researches published by Madhu Chintala.


Blood Coagulation & Fibrinolysis | 2013

Differential profiles of thrombin inhibitors (heparin, hirudin, bivalirudin, and dabigatran) in the thrombin generation assay and thromboelastography in vitro.

Yiming Xu; Weizhen Wu; Liangsu Wang; Madhu Chintala; Andrew S. Plump; Martin L. Ogletree; Zhu Chen

Thrombin is a central enzyme in hemostasis and thrombosis, and a proven target for anticoagulant therapies. We compared four marketed and representative thrombin inhibitors, heparin, hirudin, bivalirudin, and dabigatran, in in-vitro spike-in assays that covered their therapeutic ranges. The assays employed were low tissue factor (1u200apmol/l)-triggered thrombin generation assay (TGA) with plasma and 1u200a:u200a8000 Recombiplastin-triggered thromboelastography (TEG) with whole blood, with or without tissue plasminogen activator (tPA)-induced fibrinolysis. The three direct thrombin inhibitors (DTIs) prolonged TGA lag time and TEG clotting time (R) with a potency stack-ranking of hirudinu200a>u200adabigatranu200a≅u200abivalirudin. Heparin had the most steep concentration–response curve for both parameters. In TGA, 1–2u200a&mgr;mol/l dabigatran or hirudin resulted in complete inhibition on peak, slope, and endogenous thrombin potential, whereas bivalirudin had no effect on these parameters up to 10u200a&mgr;mol/l. All three DTIs, but not heparin, displayed a paradoxical increase in peak and slope in the low concentration range. In TEG, whereas all four agents reduced clot strength (maximal amplitude) in synergy with tPA, hirudin was the only DTI that reduced maximal amplitude appreciably without tPA. Dabigatran had the strongest potentiating effect on tPA-induced fibrinolytic activity (Ly30). With regard to the effects on coagulation and clot strength (lag time, R, and maximal amplitude) in the respective therapeutic range, dabigatran elicited the most modest changes. In summary, our observations highlight the distinct features of each agent in thrombin generation, coagulation, and fibrinolysis. The contrasts between the agents are consistent with their known properties and are informative on efforts to define the optimal profiles of new anticoagulants.


Bioorganic & Medicinal Chemistry Letters | 2010

Discovery of a vorapaxar analog with increased aqueous solubility.

Yan Xia; Samuel Chackalamannil; William J. Greenlee; Yuguang Wang; Zhiyong Hu; Yuriko Root; Jesse Wong; Jianshe Kong; Ho-Sam Ahn; George Boykow; Yunsheng Hsieh; Stan Kurowski; Madhu Chintala

An analog of the thrombin receptor antagonist vorapaxar (SCH 530348) with increased aqueous solubility, compound 9c (SCH 602539), was discovered through incorporation of polar substituents on the pyridine ring of the himbacine-derived lead series. This analog retained the excellent potency, pharmacokinetic and safety properties of vorapaxar while increasing the aqueous solubility by 20-fold. Also presented are in vivo evaluations of this compound in a cynomolgus monkey platelet aggregation assay and in a Folts model of thrombosis in anesthetized monkeys.


European Journal of Pharmacology | 2015

Platelet transfusion reverses bleeding evoked by triple anti-platelet therapy including vorapaxar, a novel platelet thrombin receptor antagonist

Tian-Quan Cai; L. Alexandra Wickham; Gary R. Sitko; Maria S. Michener; Richard Raubertas; Larry Handt; Madhu Chintala; Dietmar Seiffert; Michael J. Forrest

Vorapaxar is a novel protease-activated receptor-1 (PAR-1) antagonist recently approved for the reduction of thrombotic cardiovascular events in patients with a history of myocardial infarction or with peripheral arterial disease. Patients who received vorapaxar in addition to standard of care antiplatelet therapy had an increased incidence of major bleeding events compared with placebo. To assess whether platelet transfusion can restore hemostasis in primates on triple antiplatelet therapy, template bleeding times were assessed concurrently in the buccal mucosa, finger pad, and distolateral tail of anesthetized cynomolgus macaques to evaluate bleeding with vorapaxar as either monotherapy or in combination with aspirin or aspirin and clopidogrel. Aspirin (5mg/kg, IV) or vorapaxar (1mg/kg, PO) alone had no significant effect on bleeding times in the three vascular beds examined. A modest (<2-fold) increase in bleeding time was achieved in the three beds with the dual combination of aspirin and vorapaxar. Major increases in bleeding time were achieved in the three beds with the triple combination of aspirin (5mg/kg, IV), vorapaxar (1mg/kg, PO), and clopidogrel (1mg/kg, PO). Transfusion of fresh human platelet rich plasma, but not platelet poor plasma, reversed the increase in bleeding time in the triple therapy group. Transfusion of human platelets may be a viable approach in situations requiring a rapid reversal of platelet function in individuals treated with triple anti-platelet therapy that includes vorapaxar.


Thrombosis Research | 2014

Zucker Diabetic Fatty rats exhibit hypercoagulability and accelerated thrombus formation in the Arterio-Venous shunt model of thrombosis

Jin Shang; Zhu Chen; Min Wang; Qiu Li; Wen Feng; Yangsong Wu; Weizhen Wu; Michael P. Graziano; Madhu Chintala

INTRODUCTIONnDiabetes is a significant risk factor for thrombosis. The present study aimed at assessing coagulability, platelet reactivity, and thrombogenicity of the diabetic female Zucker Diabetic Fatty (ZDF) rat model and its relevance in studying antithrombotic mechanisms.nnnMATERIALS AND METHODSnThe basal coagulant state in ZDF rats was evaluated by clotting times, thromboelastography, and thrombin generation assay. A 14-day treatment with dapagliflozin in ZDF rats was pursued to investigate if glycemic control can improve coagulability. Thrombus formation in the Arterio-Venous (A-V) shunt model and the FeCl3-induced arterial thrombosis model was studied, with the antithrombotic effect of apixaban in the former model further investigated.nnnRESULTSnZDF rats exhibited significantly shortened clotting times, enhanced thrombin generation, and decreased fibrinolysis at baseline. Effective glycemic control achieved with dapagliflozin did not improve any of these parameters. ZDF rats displayed accelerated thrombus formation and were amenable to apixaban treatment in the A-V shunt model albeit with less sensitivity than normal rats. ZDF rats exhibited less platelet aggregation in response to ADP, collagen and PAR-4, and attenuated thrombotic response in the FeCl3 model.nnnCONCLUSIONSnZDF rats are at a chronic hypercoagulable and hypofibrinolytic state yet with compromised platelet reactivity. They display accelerated and attenuated thrombosis in the A-V shunt and FeCl3 model of thrombosis, respectively. Results shed new light on the pathophysiology of the ZDF rat model and illustrate its potential value in translational research on anticoagulant agents in diabetics. Caution needs to be exerted in utilizing this model in assessing antiplatelet mechanisms in diabetes-associated atherothrombosis.


Bioorganic & Medicinal Chemistry Letters | 2012

Discovery of nor-seco himbacine analogs as thrombin receptor antagonists.

Mariappan V. Chelliah; Samuel Chackalamannil; Yan Xia; Keith Eagen; William J. Greenlee; Ho-Sam Ahn; Jacqueline Agans-Fantuzzi; George Boykow; Yunsheng Hsieh; Matthew Bryant; Tze-Ming Chan; Madhu Chintala

Discovery of a novel nor-seco himbacine analog as potent thrombin receptor (PAR-1) antagonist is described. Despite low plasma level, these new analogs showed excellent ex vivo efficacy in the monkey platelet aggregation assay. A potent hydroxy metabolite generated in vivo was identified as the agent responsible for the ex vivo efficacy. Following this discovery, the metabolite series was optimized to obtain analogs that showed very good ex vivo efficacy along with excellent pharmacokinetic profile in c. monkey.


European Journal of Pharmacology | 2015

In vitro pharmacological characterization of vorapaxar, a novel platelet thrombin receptor antagonist

Brian Hawes; Ying Zhai; David Hesk; Mark Wirth; Huijun Wei; Madhu Chintala; Dietmar Seiffert

Vorapaxar is a novel protease-activated receptor-1 (PAR1) antagonist recently approved for the reduction of thrombotic cardiovascular events in patients with a history of myocardial infarction or with peripheral arterial disease. The present study provides a comprehensive in vitro pharmacological characterization of vorapaxar interaction with the PAR1 receptor on human platelets. Similar studies were performed with a metabolite of vorapaxar (M20). Vorapaxar and M20 were competitive PAR1 antagonists that demonstrated concentration-dependent, saturable, specific, and slowly reversible binding to the receptor present on intact human platelets. The affinities of vorapaxar and M20 for the PAR1 receptor were in the low nanomolar range, as determined by saturation-, kinetic- and competitive binding studies. The calculated Kd and Ki values for vorapaxar increased in the presence of plasma, indicating a decrease in the free fraction available for binding to the PAR1 receptor on human platelets. Vorapaxar was also evaluated in functional assays using thrombin or a PAR1 agonist peptide (SFLLRN). Vorapaxar and M20 completely blocked thrombin-stimulated PAR1/β-arrestin association in recombinant cells and abolished thrombin-stimulated calcium influx in washed human platelets and vascular smooth muscle cells. Moreover, vorapaxar and M20 inhibited PAR1 agonist peptide-mediated platelet aggregation in human platelet rich plasma with a steep concentration response relationship. Vorapaxar exhibited high selectivity for inhibition of PAR1 over other platelet GPCRs. In conclusion, vorapaxar is a potent PAR1 antagonist exhibiting saturable, reversible, selective binding with slow off-rate kinetics and effectively inhibits thrombins PAR1-mediated actions on human platelets.


Bioorganic & Medicinal Chemistry Letters | 2015

Iminopyrimidinones: A novel pharmacophore for the development of orally active renin inhibitors

Brian A. McKittrick; John P. Caldwell; Thomas Bara; George Boykow; Madhu Chintala; John W. Clader; Michael Czarniecki; Brandy Courneya; Ruth Duffy; Linda Fleming; Rachel Giessert; William J. Greenlee; Charles R. Heap; Liwu Hong; Ying Huang; Ulrich Iserloh; Hubert Josien; Tanweer Khan; Walter A. Korfmacher; Xian Liang; Robert Mazzola; Soumya Mitra; Kristina Moore; Peter Orth; Murali Rajagopalan; Sudipta Roy; Samuel A. Sakwa; Corey Strickland; Henry M. Vaccaro; Johannes Voigt

The development of renin inhibitors with favorable oral pharmacokinetic profiles has been a longstanding challenge for the pharmaceutical industry. As part of our work to identify inhibitors of BACE1, we have previously developed iminopyrimidinones as a novel pharmacophore for aspartyl protease inhibition. In this letter we describe how we modified substitution around this pharmacophore to develop a potent, selective and orally active renin inhibitor.


Bioorganic & Medicinal Chemistry Letters | 2012

SAR studies of C2 ethers of 2H-pyrano[2,3-d]pyrimidine-2,4,7(1H,3H)-triones as nicotinic acid receptor (NAR) agonist

Xianhai Huang; Jing Su; Ashwin U. Rao; Haiqun Tang; Wei Zhou; Xiaohong Zhu; Xiao Chen; Zhidan Liu; Ying Huang; Sylvia Degrado; Dong Xiao; Jun Qin; Robert Aslanian; Brian A. McKittrick; Scott Greenfeder; Margaret van Heek; Madhu Chintala; Anandan Palani

Based on in house screening lead compound 1 for the NAR project, SAR studies have been focused on the modification of the C2 ethers of the pyrimidinedione core structure. In this effort, an unpredictable SAR trend was overcome in the alkyl ether and arylalkyl ether series to identify compound 24 with improved in vitro activity compared to nicotinic acid. More consistent and predictable SAR was achieved in the propargyl ether series. Lead compound 41 was identified with good in vitro and in vivo activity in rat, and much improved rat PK profile.


Archive | 2005

Crystalline polymorph of a bisulfate salt of a thrombin receptor antagonist

Tiruvettipuram K. Thiruvengadam; Wenxue Wu; Tao Wang; John S. Chiu; Susan Bogdanowich-Knipp; Anastasia Pavlovsky; William J. Greenlee; Michael P. Graziano; Teddy Kosoglou; Madhu Chintala; Samuel Chackalamannil


Arteriosclerosis, Thrombosis, and Vascular Biology | 2012

Abstract 264: Differential Profiles of Thrombin Inhibitors (Dabigatran, Hirudin, Bivalirudin and Heparin) in the Thrombin Generation Assay (TGA) and Thromboelastography (TEG) in Vitro

Yiming Xu; Weizhen Wu; Andrew S. Plump; Madhu Chintala; Martin L. Ogletree; Zhu Chen

Researchain Logo
Decentralizing Knowledge