Network


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

Hotspot


Dive into the research topics where William A. Schumacher is active.

Publication


Featured researches published by William A. Schumacher.


Journal of Thrombosis and Haemostasis | 2005

Effects of factor IX or factor XI deficiency on ferric chloride‐induced carotid artery occlusion in mice

Xinkang Wang; Qiufang Cheng; Xu L; Giora Z. Feuerstein; Hsu My; Smith Pl; Seiffert Da; William A. Schumacher; Martin L. Ogletree; David Gailani

Summary.  Factor XI (FXI) and factor IX (FIX) are zymogens of plasma serine proteases required for normal hemostasis. The purpose of this work was to evaluate FXI and FIX as potential therapeutic targets by means of a refined ferric chloride (FeCl3)‐induced arterial injury model in factor‐deficient mice. Various concentrations of FeCl3 were used to establish the arterial thrombosis model in C57BL/6 mice. Carotid artery blood flow was completely blocked within 10 min in C57BL/6 mice by application of 3.5% FeCl3. In contrast, FXI‐ and FIX‐deficient mice were fully protected from occlusion induced by 5% FeCl3, and were partially protected against the effect of 7.5% FeCl3. The protective effect was comparable to very high doses of heparin (1000 units kg−1) and substantially more effective than aspirin. While FXI and FIX deficiencies were indistinguishable in the carotid artery injury model, there was a marked difference in a tail‐bleeding‐time assay. FXI‐deficient and wild‐type mice have similar bleeding times, while FIX deficiency was associated with severely prolonged bleeding times (>5.8‐fold increase, P < 0.01). Given the relatively mild bleeding diathesis associated with FXI deficiency, therapeutic inhibition of FXI may be a reasonable strategy for treating or preventing thrombus formation.


Journal of Thrombosis and Haemostasis | 2006

Effects of factor XI deficiency on ferric chloride‐induced vena cava thrombosis in mice

Xinkang Wang; Patricia L. Smith; Mei-Yin Hsu; David Gailani; William A. Schumacher; Martin L. Ogletree; Seiffert Da

Summary.  Background: Increased plasma levels of coagulation factor (F) XI are a risk factor for venous thrombosis. Objective: To further explore the relationship between FXI and venous thrombosis, we evaluated FXI‐deficient and wild‐type mice in a ferric chloride (FeCl3)‐induced vena cava thrombosis model. Methods and Results: Thrombosis was induced by 3‐min topical application of filter papers containing increasing concentrations of FeCl3 and the thrombus was measured at 30 min. In contrast to wild‐type mice, FXI‐deficient mice failed to form a thrombus with 5% FeCl3, and were partially protected against 7.5% and 10% FeCl3, respectively. The protective effect was substantially stronger than a high dose of heparin (1000 units kg−1, i.v.), clopidogrel (30 mg kg−1, p.o.) or argatroban (30 mg kg−1, i.p.). These antithrombotic agents resulted in off‐scale bleeding in a tail bleeding time assay, whereas the bleeding time of FXI‐deficient mice was unchanged compared to wild‐type mice. In addition to its known effect on the coagulation cascade, enhanced clot lysis was demonstrated in FXI‐deficient mouse and human plasma compared to those supplemented with FXIa. Conclusion: Given the strong antithrombotic efficacy (possibly contributed by strong anticoagulant activity associated with increased fibrinolytic activity) and mild bleeding diathesis associated with FXI deficiency, therapeutic inhibition of FXI may be a reasonable therapeutic strategy to treat or prevent venous thrombosis.


Journal of Thrombosis and Haemostasis | 2006

Murine model of ferric chloride‐induced vena cava thrombosis: evidence for effect of potato carboxypeptidase inhibitor

Xinkang Wang; Patricia L. Smith; Mei-Yin Hsu; Martin L. Ogletree; William A. Schumacher

Summary.  Background/objective: Thrombin‐activatable fibrinolysis inhibitor (TAFI) is a plasma carboxypeptidase that renders a fibrin‐containing thrombus less sensitive to lysis. In the present study, we describe the development of a murine model of vena cava thrombosis and its use to characterize the antithrombotic activity of potato carboxypeptidase inhibitor (PCI) of TAFIa (activated TAFI) in mice. Methods/results: Vena cava thrombosis was induced by various concentrations of FeCl3 in C57BL/6 mice. A relatively mild stimulus (3.5% FeCl3) induced thrombosis that was consistent and sensitive to reference antithrombotic agents such as clopidogrel and heparin. Dose–response studies identified a PCI dose (5 mg kg−1 bolus plus 5 mg kg−1 h−1, i.v.) that produced a maximum 45% decrease in vena cava thrombus mass as assessed by protein content (n = 8, P < 0.01 compared to vehicle) in the 3.5% FeCl3‐induced model without exogenous tissue plasminogen activator administration. In contrast, PCI had no effect on 3.5% FeCl3‐induced carotid artery thrombosis in mice. In a tail transection bleeding model, the 5 mg kg−1 bolus plus 5 mg kg−1 h−1 dose of PCI increased tail‐bleeding time up to 3.5 times control (n = 8, P < 0.05). The ex vivo activity of antithrombotic doses of PCI was also demonstrated by the enhanced lysis of whole blood clots formed in a thrombelastograph with the addition of a sub‐threshold concentration of tPA. Conclusion: These studies provide evidence for a role of TAFIa in venous thrombosis in mice, and describe an optimized vena cava injury model appropriate for the evaluation of antithrombotic drugs and the characterization of novel therapeutic targets.


Bioorganic & Medicinal Chemistry Letters | 2002

Synthesis and SAR of 4-carboxy-2-azetidinone mechanism-based tryptase inhibitors

James C. Sutton; Scott A. Bolton; Karen S. Hartl; Ming-Hsing Huang; Glenn Anthony Jacobs; Wei Meng; Martin L. Ogletree; Zulan Pi; William A. Schumacher; Steven M. Seiler; William A. Slusarchyk; Uwe D. Treuner; Robert Zahler; Guohua Zhao; Gregory S. Bisacchi

A series of N1-activated C4-carboxy azetidinones was prepared and tested as inhibitors of human tryptase. The key stereochemical and functional features required for potency, serine protease specificity and aqueous stability were determined. From these studies compound 2, BMS-262084, was identified as a potent and selective tryptase inhibitor which, when dosed intratracheally in ovalbumin-sensitized guinea pigs, reduced allergen-induced bronchoconstriction and inflammatory cell infiltration into the lung.


Bioorganic & Medicinal Chemistry Letters | 2002

Synthesis of potent and highly selective inhibitors of human tryptase

William A. Slusarchyk; Scott A. Bolton; Karen S. Hartl; Ming-Hsing Huang; Glenn Anthony Jacobs; Wei Meng; Martin L. Ogletree; Zulan Pi; William A. Schumacher; Steven M. Seiler; James C. Sutton; Uwe D. Treuner; Robert Zahler; Guohua Zhao; Gregory S. Bisacchi

The serine protease tryptase has been implicated in allergic and inflammatory diseases and associated with asthma. The synthesis and SAR of a series of N1-activated-4-carboxy azetidinones are described, resulting in identification of BMS-363131 (2) as a potent inhibitor of human tryptase (IC(50)<1.7 nM) with high selectivity (>3000-fold) for tryptase versus related serine proteases including trypsin.


Journal of Pharmacological and Toxicological Methods | 1996

A FERRET MODEL OF ELECTRICAL-INDUCTION OF ARTERIAL THROMBOSIS THAT IS SENSITIVE TO ASPIRIN

William A. Schumacher; Thomas E. Steinbacher; John R. Megill; Stephen K. Durham

An experimental model of acute thrombosis was developed in pentobarbital- anesthetized ferrets. A 10-min anodal electrical stimulation of 1 mA was delivered to the external surface of the carotid artery while measuring carotid blood flow (CBF). This produced an occlusive thrombus in all vehicle-treated ferrets within 41 +/- 3 min with an average weight of 8 +/- 1 mg (n = 7). These thrombi were enriched in both platelets and fibrin and were adherent at the site of transmural vascular injury as determined by light and electron microscopy. To determine the models sensitivity to antiplatelet drugs, aspirin or a thromboxane (TxA2) receptor antagonist (ifetroban) were administered 15 min before electrical stimulation. Thrombus weight was reduced 58% by aspirin (10 mg/kg, i.v.) and 74% by ifetroban (1 mg/kg + 1 mg/kg per hr, i.v.). Both drugs also improved CBF and decreased vascular occlusion. Ferrets were more sensitive than rats to aspirins inhibition of collagen-induced platelet aggregation as determined ex vivo in whole blood. Separate in vitro platelet aggregation studies revealed species differences in reactivity to U-46619 (TxA2 receptor agonist) and collagen in the order of human > ferret > rat, with relatively lesser variations in ADP responses. These studies identify the ferret as a useful species for evaluating antithrombotic drugs in a model in which aspirin is efficacious.


Thrombosis Research | 2011

Bleeding response induced by anti-thrombotic doses of a phosphoinositide 3-kinase (PI3K)-β inhibitor in mice☆

J.Eileen Bird; Patricia L. Smith; Jeffrey S. Bostwick; Petia Shipkova; William A. Schumacher

INTRODUCTION Published evidence suggests that phosphoinositide 3 kinase-β (PI3K-β) plays an important role in platelet aggregation and shear activation. TGX-221 is a selective PI3K-β inhibitor with a good separation of anti-thrombotic efficacy and bleeding (therapeutic index) in rats. Our goal was to further evaluate potential of a PI3K-β inhibitor as an anti-thrombotic agent by determining the therapeutic index in another species and efficacy model. Reported effects of TGX-221 in rats were also confirmed. MATERIALS AND METHODS TGX-221 (0.3 + 0.3, 1 + 1, 3 + 3 mg/kg + mg/kg/hr, i.v.) or vehicle was given to mice starting 15 min prior to FeCl(3) arterial thrombosis (AT), tail or kidney bleeding time (BT) procedures. RESULTS Integrated blood flow over 30 min (%baseline mean ± SEM) improved (p < 0.05) with TGX-221 doses 1 + 1 (49 ± 13.9%) and 3+3 (88 ± 10.6%) versus 0.3 + 0.3 (10 ± 0.8%) and vehicle (10 ± 0.6%). Vascular patency (non-occluded/total arteries) improved (p < 0.01) with TGX-221 doses of 3 + 3 (7/8), but not 0.3 + 0.3 (0/8) or 1 + 1 (4/8) versus vehicle (0/8). Tail BT (sec) increased (p < 0.05) with TGX-221 doses of 3 + 3 (median 1560) and 1 + 1 (1305) versus vehicle (225). Mean renal BT (sec) increased (p < 0.05) in all TGX-221 groups (3 + 3: 510 + 26; 1 + 1: 478 + 41; 0.3 + 0.3: 246 + 37) versus vehicle (123 + 9). For comparison, a reference agent, aspirin (30 mpk, i.p.) increased tail BT 1.9X and renal BT 2.6X. CONCLUSIONS The novel finding of a clear impact on hemostasis by TGX-221 was demonstrated by increased bleeding in two models in mice at anti-thrombotic doses. The results suggest a narrower therapeutic index for this PI3K-β inhibitor than previously recognized, at least for this species.


Science Translational Medicine | 2017

Blockade of protease-activated receptor-4 (PAR4) provides robust antithrombotic activity with low bleeding

Pancras C. Wong; Dietmar Seiffert; Bird Je; Carol A. Watson; Bostwick Js; Giancarli M; Allegretto N; Hua J; Harden D; Guay J; Callejo M; Miller Mm; Lawrence Rm; Jacques Banville; Guy J; Maxwell Bd; Priestley Es; Anne Marinier; Ruth R. Wexler; Michel Bouvier; David A. Gordon; William A. Schumacher; Jing Yang

PAR4 inhibition with a reversible small-molecule antagonist is a promising antiplatelet therapy. This antiplatelet agent is just right Antiplatelet agents are common drugs that are used to prevent thrombotic events such as stroke in patients at high risk. Unfortunately, a frequent side effect of these drugs is excessive bleeding because they indiscriminately suppress thrombotic function. To improve on the safety margin of antiplatelet agents, Wong et al. identified a new target for treatment, a platelet receptor called PAR4. The authors developed a small-molecule drug against this target and evaluated its efficacy and safety in animal models. In head-to-head comparisons, the new drug was no less effective than clopidogrel, an antiplatelet agent widely used in the clinic, but it had a much larger therapeutic window. Antiplatelet agents are proven efficacious treatments for cardiovascular and cerebrovascular diseases. However, the existing drugs are compromised by unwanted and sometimes life-threatening bleeding that limits drug usage or dosage. There is a substantial unmet medical need for an antiplatelet drug with strong efficacy and low bleeding risk. Thrombin is a potent platelet agonist that directly induces platelet activation via the G protein (heterotrimeric guanine nucleotide–binding protein)–coupled protease-activated receptors PAR1 and PAR4. A PAR1 antagonist is approved for clinical use, but its use is limited by a substantial bleeding risk. Conversely, the potential of PAR4 as an antiplatelet target has not been well characterized. Using anti-PAR4 antibodies, we demonstrated a low bleeding risk and an effective antithrombotic profile with PAR4 inhibition in guinea pigs. Subsequently, high-throughput screening and an extensive medicinal chemistry effort resulted in the discovery of BMS-986120, an orally active, selective, and reversible PAR4 antagonist. In a cynomolgus monkey arterial thrombosis model, BMS-986120 demonstrated potent and highly efficacious antithrombotic activity. BMS-986120 also exhibited a low bleeding liability and a markedly wider therapeutic window compared to the standard antiplatelet agent clopidogrel tested in the same nonhuman primate model. These preclinical findings define the biological role of PAR4 in mediating platelet aggregation. In addition, they indicate that targeting PAR4 is an attractive antiplatelet strategy with the potential to treat patients at a high risk of atherothrombosis with superior safety compared with the current standard of care.


Bioorganic & Medicinal Chemistry Letters | 2014

Identification of 1-{2-[4-chloro-1′-(2,2-dimethylpropyl)-7-hydroxy-1,2-dihydrospiro[indole-3,4′-piperidine]-1-yl]phenyl}-3-{5-chloro-[1,3]thiazolo[5,4-b]pyridin-2-yl}urea, a potent, efficacious and orally bioavailable P2Y1 antagonist as an antiplatelet agent

Yoon T. Jeon; Wu Yang; Jennifer X. Qiao; Ling Li; Rejean Ruel; Carl Thibeault; Sheldon Hiebert; Tammy C. Wang; Yufeng Wang; Yajun Liu; Charles G. Clark; Henry S. Wong; Juliang Zhu; Dauh-Rurng Wu; Dawn Sun; Bang-Chi Chen; Arvind Mathur; Silvi A. Chacko; Mary F. Malley; Xue-Qing Chen; Hong Shen; Christine Huang; William A. Schumacher; Jeffrey S. Bostwick; Anne B. Stewart; Laura A. Price; Ji Hua; Danshi Li; Paul Levesque; Dietmar Seiffert

Spiropiperidine indoline-substituted diaryl ureas had been identified as antagonists of the P2Y1 receptor. Enhancements in potency were realized through the introduction of a 7-hydroxyl substitution on the spiropiperidinylindoline chemotype. SAR studies were conducted to improve PK and potency, resulting in the identification of compound 3e, a potent, orally bioavailable P2Y1 antagonist with a suitable PK profile in preclinical species. Compound 3e demonstrated a robust antithrombotic effect in vivo and improved bleeding risk profile compared to the P2Y12 antagonist clopidogrel in rat efficacy/bleeding models.


Prostaglandins | 1992

Antiplatelet activity of the long-acting thromboxane receptor antagonist BMS 180,291 in monkeys

William A. Schumacher; Thomas E. Steinbacher; Sonia Youssef; Martin L. Ogletree

The effects of the novel TxA2/prostaglandin endoperoxide (TP) receptor antagonist BMS 180,291 on platelet reactivity was determined ex vivo in conscious African green monkeys. Platelet aggregation responses to U-46,619 were decreased 50% and 100% at 23 to 24 hrs after BMS 180,291 oral doses of 1 and 3 mg/kg, respectively. In addition to inhibiting aggregation, a 3 mg/kg oral dose of BMS 180,291 also produced an 11 +/- 3-fold shift to the right in the U-46,619 concentration-response relationship for platelet shape change at 24 hrs after dosing. When the 3 mg/kg oral dose was continued for 11 days, the shift in this concentration-response relationship increased to 26 +/- 10- and 93 +/- 30-fold at 24 hrs after the 8th and 11th doses, respectively. This progressive inhibition corresponds to 93 +/- 3 and 99 +/- 1% blockade of platelet TP-receptors responsible for shape change, respectively. Comparable levels of TP-receptor blockade have been previously correlated with antithrombotic and antiischemic activities of TP-receptor antagonists in vivo. Platelet reactivity to U-46,619 had completely recovered on the 7th day after the final dose of BMS 180,291, indicating effective elimination from the circulation over this interval. In separate experiments, a 3-mg/kg i.v. dose of BMS 180,291 produced only marginal and transient hemodynamic effects in anesthetized African green monkeys. Overall, these data demonstrate that BMS 180,291 given orally once a day produces a sustained and therapeutically-relevant level of TP-receptor antagonism.

Collaboration


Dive into the William A. Schumacher'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
Researchain Logo
Decentralizing Knowledge