Network


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

Hotspot


Dive into the research topics where S. Betty Yan is active.

Publication


Featured researches published by S. Betty Yan.


Critical Care Medicine | 2001

Safety and dose relationship of recombinant human activated protein C for coagulopathy in severe sepsis.

Gordon R. Bernard; E. Wesley Ely; Theressa J. Wright; Joseph Fraiz; Jerome E. Stasek; James A. Russell; Irvin Mayers; Brian A. Rosenfeld; Peter E. Morris; S. Betty Yan; Jeffery D. Helterbrand

Objectives To assess the safety and effect on coagulopathy of a range of doses of recombinant human activated protein C (rhAPC). To determine an effective dose and duration of rhAPC for use in future clinical trials. Design Double-blind, randomized, placebo-controlled, multicenter, dose-ranging (sequential), phase II clinical trial. Setting Forty community or academic medical institutions in United States and Canada. Patients One hundred thirty-one adult patients with severe sepsis. Interventions Intravenous infusion of rhAPC (12, 18, 24, or 30 &mgr;g/kg/hr) or placebo for 48 or 96 hrs. Measurements and Main Results No significant differences in incidence of serious bleeding events (4% rhAPC, 5% placebo, p > .999) or incidence of serious adverse events (39% rhAPC, 46% placebo, p = 0.422) between rhAPC- and placebo-treated patients were observed. One of 53 rhAPC-treated patients with suitable immunogenicity samples had a low level, transient, non-neutralizing anti-APC antibody response not associated with any clinical adverse event. Significant dose-dependent decreases in both D-dimer (p <0.001) and end of infusion interleukin 6 levels (p = .021) were demonstrated. No statistically significant effects on fibrinogen or platelet counts were observed. A nonstatistically significant 15% relative risk reduction in 28-day all-cause mortality was observed between rhAPC- and placebo-treated patients. Conclusions rhAPC was safe and well-tolerated and demonstrated a dose-dependent reduction in D-dimer and interleukin 6 levels relative to placebo. The dose of 24 &mgr;g/kg/hr for 96 hrs was selected for use in future clinical studies.


Critical Care Medicine | 2000

Prognostic value of protein C concentrations in neutropenic patients at high risk of severe septic complications.

Rolf M. Mesters; Jeff Helterbrand; Barbara G. Utterback; S. Betty Yan; Y. Bernice Chao; José A. Fernández; Daniel Hartman

Objective To assess the prognostic value of protein C, endogenous activated protein C, and d-dimer concentrations in patients at high risk of developing severe septic complications secondary to cytostatic chemotherapy. Design Prospective, comparative, single-center study. Setting Specialized ward for treating patients with acute leukemia and associated intensive care unit at a university hospital. Subjects Twenty-six consecutive patients who developed either severe sepsis (n = 13) or septic shock (n = 13) during chemotherapy-induced neutropenia (leukocytes <1000/&mgr;L). Intervention None, other than standard care. Measurements and Main Results Baseline blood samples were obtained from 97 adult patients treated with intensive cytostatic chemotherapy. Serial blood sampling was performed in 62 of 97 patients who developed fever (>38.3°C). Thirteen patients progressed to severe sepsis and 13 patients to septic shock. Protein C, endogenous activated protein C, and d-dimer were measured in these 26 patients. At fever onset, protein C concentrations decreased from normal baseline concentrations and were significantly lower in the group of patients who progressed to septic shock compared with those who developed severe sepsis (medians for protein C activity: 23.1% vs. 69.5%;p = .0003). The median elapsed time between detection of fever and the diagnosis of severe sepsis or septic shock was 16 hrs and 12 hrs, respectively. All septic shock patients died, whereas patients who progressed only to severe sepsis survived. Conclusions Septic shock in neutropenic patients is associated with increased protein C consumption. The data demonstrate that the coagulation cascade is activated and produces a hypercoagulable state before the onset of clinical symptoms of severe sepsis and septic shock. Low protein C concentrations at the onset of fever and before the onset of clinical symptoms of severe sepsis or septic shock may have prognostic value in predicting an unfavorable outcome. Protein C measurements may help identify patients at risk in an early phase of neutropenic sepsis. It is also attractive to speculate that because low protein C concentrations were seen in these patients, protein C replacement may be beneficial in sepsis.


Clinical Infectious Diseases | 2003

Systemic Host Responses in Severe Sepsis Analyzed by Causative Microorganism and Treatment Effects of Drotrecogin Alfa (Activated)

Steven M. Opal; Gary Garber; Steven P. LaRosa; Dennis G. Maki; Ross Freebairn; Gary T. Kinasewitz; Jean-François Dhainaut; S. Betty Yan; Mark D. Williams; Delores E. Graham; David R. Nelson; Howard Levy; Gordon R. Bernard

Clinical trials with novel therapeutic agents for severe sepsis have suggested that patients might respond differently depending on causative microorganism. Data from a large, placebo-controlled trial of recombinant human drotrecogin alfa (activated) (DrotAA) were analyzed by type of causative microorganism for treatment-associated differences in mortality, coagulopathy, and inflammatory response. Compared with placebo, mortality rates associated with DrotAA were consistently reduced for each microorganism group (gram-positive bacteria, gram-negative bacteria, mixed bacteria, fungi, other, and unknown microbial etiology), with a stratified relative risk (RR) of 0.80 (95% confidence interval [CI], 0.69-0.94). The greatest reduction in the mortality rate was for Streptococcus pneumoniae infection (RR, 0.56; 95% CI, 0.35-0.88). Levels of coagulation and inflammation biomarkers varied with different pathogens at study entry. Results demonstrate that DrotAA, administered as an adjunct to standard anti-infective therapy, can improve the rate of survival for patients who develop severe sepsis regardless of causative microorganism.


Thrombosis and Haemostasis | 2003

Drotrecogin alfa (activated) (recombinant human activated protein C) reduces host coagulopathy response in patients with severe sepsis

Jean-François Dhainaut; S. Betty Yan; Benjamin D. Margolis; José A. Lorente; James A. Russell; Ross Freebairn; Herbert D. Spapen; Hanno Riess; Bruce R. Basson; Gerald Johnson; Gary T. Kinasewitz

Drotrecogin alfa (activated) improved survival in patients with severe sepsis in PROWESS, a double-blind, study of 1690 adult patients randomized to drotrecogin alfa (activated) at 24 microg/kg/h (N=850) or placebo (N=840) infused for 96 hours. Pharmacodynamic effects of drotrecogin alfa (activated) were assessed with 15 prospectively defined systemic biomarkers of hemostasis, inflammation and endothelial injury. The last-observation-carried-forward (LOCF) method of imputation for missing observations was the prospectively defined statistical method. The results were also analyzed with only the observed values without imputation for missing data (repeated measures analysis). With both statistical methods, drotrecogin alfa (activated)-treated patients demonstrated antithrombotic (reduced markers of thrombin generation and accelerated normalization of anticoagulant factor, protein C and fibrinolytic factors) and anticoagulant (prolonged PT and APTT) effects compared with placebo. A profibrinolytic (reduction in plasminogen activator inhibitor-1) effect was significant only with the LOCF imputation method in observed case and percent change from baseline analyses. An anti-inflammatory (reduction in interleukin-6) effect was significant only with the LOCF imputation method in change from baseline and percent change from baseline analyses. Drotrecogin alfa (activated) is a new and promising agent for treatment of patients with severe sepsis. The extensive analysis of systemic biomarkers confirms the previously published antithrombotic effects. However, the present results using different statistical methods do not provide a strong basis for systemic anti-inflammatory or pro-fibrinolytic effects. These latter two effects may occur at the local or cellular level. The systemic biomarkers reported here might not be the most appropriate approach to demonstrate these potential effects of drotrecogin alfa (activated).


Critical Care Medicine | 2001

Activated protein C versus protein C in severe sepsis

S. Betty Yan; Jean-François Dhainaut

ObjectiveTo delineate critical differences between activated protein C (APC) and its precursor, protein C, with regard to plasma levels in health and in severe sepsis, and to discuss the implications of these differences as they relate to treatment strategies in patients with severe sepsis. Data Source/Study Selection Published literature including abstracts, manuscripts, and review articles reporting studies in both experimental animal models and humans that provide an understanding of the relationship and the critical differences between circulating levels of APC and protein C. Data Extraction and Synthesis The protein C pathway represents one of the major regulatory systems of hemostasis, exhibiting antithrombotic, profibrinolytic and anti-inflammatory properties. This pathway also plays a critical role in the pathophysiology of severe sepsis. Central to this pathway is the vitamin K-dependent serine protease, APC, and its precursor, protein C. The conversion of protein C to APC is dependent on the complex of thrombin and thrombomodulin, an integral endothelial surface receptor. The conversion of protein C to APC is further augmented by another endothelial surface protein, the endothelial protein C receptor. There are limited published data on APC levels in health and disease, probably due to the complexity of the assay methodology for measuring APC and the absence of commercially available diagnostic kits. In animals and humans with normal functioning endothelium, circulating levels of APC (1–3 ng/mL) are positively correlated with protein C (4000–5000 ng/mL) concentration and the amount of thrombin generated. In patients with severe sepsis, there is a generalized endothelial dysfunction, contributing to multiple organ failure with increased morbidity and mortality. Persistently low protein C levels are related to poor prognosis. Key to understanding the treatment strategy with APC or protein C is knowledge of the functional status of the endothelium and, specifically, whether the microvasculature in patients with severe sepsis can support the conversion of protein C to APC. To date, only APC (drotrecogin alfa [activated]) has been shown to reduce mortality in severe sepsis in a large, phase 3, placebo-controlled, double-blind international trial. In contrast, no data, other than open-label case studies, are available for evaluation of the effects of protein C in the treatment of severe sepsis. ConclusionThe limited data available indicate that lower levels of protein C in sepsis occur in the absence of appreciable conversion to APC. These observations indicate that treatment with APC may be more efficacious than protein C in severe sepsis, where generalized endothelial dysfunction may impair conversion of protein C to APC. Additional research is required to confirm these observations.


Critical Care | 2005

New insights into the protein C pathway: potential implications for the biological activities of drotrecogin alfa (activated)

William L. Macias; S. Betty Yan; Mark D. Williams; Suzane L. Um; George E. Sandusky; Darryl Ballard; Jean Michel S Planquois

It has been hypothesized that the protein C pathway is a pivotal link between the inflammation and coagulation cascades. The demonstration that a survival benefit is associated with administration of drotrecogin alfa (activated) (recombinant human activated protein C [APC]) in severe sepsis patients has provided new insights into the protein C pathway. APC was originally identified based on its antithrombotic properties, which result from the inhibition of activated Factors V and VIII. In the early 1990s, any potential anti-inflammatory properties of APC were thought to relate primarily to its inhibition of thrombin generation. However, the mid-1990s saw the identification of the endothelial protein C receptor (EPCR), which has subsequently been shown to be neither endothelial specific nor protein C specific, but has a primary function as a cofactor for enhancing the generation of APC or behaving as an APC receptor. Thus, the potential biologic activities of APC can be classed into two categories related either to the limiting of thrombin generation or to cellular effects initiated by binding to the EPCR. Intracellular signaling initiated by binding of APC to its receptor appears to be mediated by interaction with an adjacent protease-activated receptor (PAR), or by indirect activation of the sphingosine 1-phosphate pathway. Based mostly on in vitro studies, binding of APC to its receptor on endothelial cells leads to a decrease in thrombin-induced endothelial permeability injury, while such binding on blood cells, epithelial cells, and neurons has been shown to inhibit chemotaxis, be anti-apoptotic, and be neuroprotective, respectively. In the Recombinant Human Activated Protein C Worldwide Evaluation in Severe Sepsis (PROWESS) study, drotrecogin alfa (activated) was associated with improved cardiovascular function, respiratory function, and a prevention of hematologic dysfunction. This article discusses the way in which the interactions of APC may alter the microcirculation.


Journal of Critical Care | 2010

Troponin-I as a prognosticator of mortality in severe sepsis patients ☆ ☆☆ ★

Jijo John; D Bradley Woodward; Yanping Wang; S. Betty Yan; Diana Fisher; Gary T. Kinasewitz; Darell Heiselman

PURPOSE The purpose of this retrospective study was to evaluate cardiac troponin-I (cTnI) as a 28-day mortality prognosticator and predictor for a drotrecogin alfa (activated) (DrotAA) survival benefit in recombinant human activated Protein C Worldwide Evaluation in Severe Sepsis patients. METHODS Cardiac troponin-I was measured using the Access AccuTnI Troponin I assay (Beckman Coulter, Fullerton, CA). There were 598 patients (305 DrotAA, 293 placebo) with baseline cTnI data (cTnI negative [<0.06 ng/mL], n = 147; cTnI positive [>or=0.06 ng/mL], n = 451). RESULTS Cardiac troponin-I-positive patients were older (mean age, 61 vs 56 years; P = .002), were sicker (mean Acute Physiology and Chronic Health Evaluation II, 26.1 vs 22.3; P < .001), had lower baseline protein C levels (mean level, 49% vs 56%; P = .017), and had higher 28-day mortality (32% vs 14%, P < .0001) than cTnI-negative patients. Elevated cTnI was an independent prognosticator of mortality (odds ratio, 2.020; 95% confidence interval, 1.153-3.541) after adjusting for other significant variables. Breslow-Day interaction test between cTnI levels and treatment was not significant (P = .65). CONCLUSION This is the largest severe sepsis study reporting an association between elevated cTnI and higher mortality. Cardiac troponin-I elevation was not predictive of a survival benefit with DrotAA treatment.


Clinical Cancer Research | 2014

LY2875358, a Neutralizing and Internalizing Anti-MET Bivalent Antibody, Inhibits HGF-Dependent and HGF-Independent MET Activation and Tumor Growth

Ling Liu; Wei Zeng; Mark Wortinger; S. Betty Yan; Paul Cornwell; Victoria L. Peek; Jennifer R. Stephens; Jonathan Tetreault; Jinqi Xia; Jason Manro; Kelly M. Credille; Darryl Ballard; Patricia Brown-Augsburger; Volker Wacheck; Chi-Kin Chow; Lihua Huang; Yong Wang; Irene Denning; Julian Davies; Ying Tang; Peter Edward Vaillancourt; Jirong Lu

Purpose: MET, the receptor for hepatocyte growth factor (HGF), has been implicated in driving tumor proliferation and metastasis. High MET expression is correlated with poor prognosis in multiple cancers. Activation of MET can be induced either by HGF-independent mechanisms such as gene amplification, specific genetic mutations, and transcriptional upregulation or by HGF-dependent autocrine or paracrine mechanisms. Experimental Design/Results: Here, we report on LY2875358, a novel humanized bivalent anti-MET antibody that has high neutralization and internalization activities, resulting in inhibition of both HGF-dependent and HGF-independent MET pathway activation and tumor growth. In contrast to other bivalent MET antibodies, LY2875358 exhibits no functional agonist activity and does not stimulate biologic activities such as cell proliferation, scattering, invasion, tubulogenesis, or apoptosis protection in various HGF-responsive cells and no evidence of inducing proliferation in vivo in a monkey toxicity study. LY2875358 blocks HGF binding to MET and HGF-induced MET phosphorylation and cell proliferation. In contrast to the humanized one-armed 5D5 anti-MET antibody, LY2875358 induces internalization and degradation of MET that inhibits cell proliferation and tumor growth in models where MET is constitutively activated. Moreover, LY2875358 has potent antitumor activity in both HGF-dependent and HGF-independent (MET-amplified) xenograft tumor models. Together, these findings indicate that the mechanism of action of LY2875358 is different from that of the one-armed MET antibody. Conclusions: LY2875358 may provide a promising therapeutic strategy for patients whose tumors are driven by both HGF-dependent and HGF-independent MET activation. LY2875358 is currently being investigated in multiple clinical studies. Clin Cancer Res; 20(23); 6059–70. ©2014 AACR.


Critical Care Medicine | 2002

Soluble thrombomodulin, plasma-derived unactivated protein C, and recombinant human activated protein C in sepsis.

Jean-François Dhainaut; S. Betty Yan; Alain Cariou; Jean-Paul Mira

ObjectiveTo review the physiologic and biochemical mechanisms and the rationale for the use of soluble thrombomodulin, plasma-derived protein C, and recombinant human activated protein C in sepsis. Data Sources and Study SelectionResearch and review articles related to the protein C pathway published in English from 1960 to present. Data Extraction and SynthesisThe protein C anticoagulant pathway plays a major role in controlling microvascular coagulation and inflammation. Protein C is the zymogen of the vitamin K–dependent serine protease activated protein C. Protein C is converted to activated protein C when thrombin complexes with thrombomodulin, an endothelial surface transmembrane glycoprotein. Activated protein C inactivates factors Va and VIIIa and effectively limits further thrombin generation. This protein also enhances endogenous fibrinolytic activity and modulates the inflammatory response. A rapid depletion of protein C occurs in sepsis, which contributes to sepsis-induced coagulopathy and correlates with a poor prognosis. The decrease in tissue levels of thrombomodulin in patients with meningococcemia suggests that the ability to convert protein C to activated protein C may also be compromised. The ability of soluble thrombomodulin to block fibrinogen clotting and cell activation, to activate protein C, and to promote thrombin inhibition in different animal models suggests that soluble thrombomodulin could be a useful therapeutic agent in sepsis. However, soluble thrombomodulin is less effective in blocking fibrinogen and platelet activation and in promoting thrombin inhibition than endothelial surface membrane-bound thrombomodulin. Only activated protein C, and not protein C, has clearly shown a reduction in mortality in experimental animal models of sepsis and in humans. ConclusionsThe multipotent pharmacodynamic effects (antithrombotic, profibrinolytic, and anti-inflammatory) of activated protein C may explain why recombinantly derived human activated protein C is the first experimental agent to demonstrate a significant survival benefit in patients with severe sepsis.


Clinical Cancer Research | 2013

Inhibition of Tumor Growth and Metastasis in Non–Small Cell Lung Cancer by LY2801653, an Inhibitor of Several Oncokinases, Including MET

Wenjuan Wu; Chen Bi; Kelly M. Credille; Jason Manro; Victoria L. Peek; Gregory P. Donoho; Lei Yan; John A. Wijsman; S. Betty Yan; Richard A. Walgren

Purpose: Lung cancer is the leading cause of cancer-related death worldwide. Sustained activation, overexpression, or mutation of the MET pathway is associated with a poor prognosis in a variety of tumors, including non–small cell lung cancer (NSCLC), implicating the MET pathway as a potential therapeutic target for lung cancer. Previously, we reported on the development of LY2801653: a novel, orally bioavailable oncokinase inhibitor with MET as one of its targets. Here, we discuss the evaluation of LY2801653 in both preclinical in vitro and in vivo NSCLC models. Experimental Design/Results: Treatment with LY2801653 showed tumor growth inhibition in tumor cell lines and patient-derived tumor xenograft models as a single agent (37.4%–90.0% inhibition) or when used in combination with cisplatin, gemcitabine, or erlotinib (66.5%–86.3% inhibition). Mechanistic studies showed that treatment with LY2801653 inhibited the constitutive activation of MET pathway signaling and resulted in inhibition of NCI-H441 cell proliferation, anchorage-independent growth, migration, and invasion. These in vitro findings were confirmed in the H441 orthotopic model where LY2801653 treatment significantly inhibited both primary tumor growth (87.9% inhibition) and metastasis (64.5% inhibition of lymph node and 67.7% inhibition of chest wall). Tumor-bearing animals treated with LY2801653 had a significantly greater survival time (87% increase compared with the vehicle-treated mice). In the MET-independent NCI-H1299 orthotopic model, treatment with LY2801653 showed a significant inhibition of primary tumor growth but not metastasis. Conclusions: Collectively, these results support clinical evaluation of LY2801653 in NSCLCs and suggest that differences in the MET activation of tumors may be predictive of response. Clin Cancer Res; 19(20); 5699–710. ©2013 AACR.

Collaboration


Dive into the S. Betty Yan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lei Yan

Eli Lilly and Company

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wei Zeng

Eli Lilly and Company

View shared research outputs
Top Co-Authors

Avatar

Gary T. Kinasewitz

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge