Lemuel A. Moyé
University of Texas Health Science Center at Houston
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Lemuel A. Moyé.
JAMA | 2012
Emerson C. Perin; James T. Willerson; Carl J. Pepine; Timothy D. Henry; Stephen G. Ellis; David Zhao; Guilherme V. Silva; Dejian Lai; James D. Thomas; Marvin W. Kronenberg; A. Daniel Martin; R. David Anderson; Jay H. Traverse; Marc S. Penn; Saif Anwaruddin; Antonis K. Hatzopoulos; Adrian P. Gee; Doris A. Taylor; Christopher R. Cogle; Deirdre Smith; Lynette Westbrook; James Chen; Eileen Handberg; Rachel E. Olson; Carrie Geither; Sherry Bowman; Judy Francescon; Sarah Baraniuk; Linda B. Piller; Lara M. Simpson
CONTEXTnPrevious studies using autologous bone marrow mononuclear cells (BMCs) in patients with ischemic cardiomyopathy have demonstrated safety and suggested efficacy.nnnOBJECTIVEnTo determine if administration of BMCs through transendocardial injections improves myocardial perfusion, reduces left ventricular end-systolic volume (LVESV), or enhances maximal oxygen consumption in patients with coronary artery disease or LV dysfunction, and limiting heart failure or angina.nnnDESIGN, SETTING, AND PATIENTSnA phase 2 randomized double-blind, placebo-controlled trial of symptomatic patients (New York Heart Association classification II-III or Canadian Cardiovascular Society classification II-IV) with a left ventricular ejection fraction of 45% or less, a perfusion defect by single-photon emission tomography (SPECT), and coronary artery disease not amenable to revascularization who were receiving maximal medical therapy at 5 National Heart, Lung, and Blood Institute-sponsored Cardiovascular Cell Therapy Research Network (CCTRN) sites between April 29, 2009, and April 18, 2011.nnnINTERVENTIONnBone marrow aspiration (isolation of BMCs using a standardized automated system performed locally) and transendocardial injection of 100 million BMCs or placebo (ratio of 2 for BMC group to 1 for placebo group).nnnMAIN OUTCOME MEASURESnCo-primary end points assessed at 6 months: changes in LVESV assessed by echocardiography, maximal oxygen consumption, and reversibility on SPECT. Phenotypic and functional analyses of the cell product were performed by the CCTRN biorepository core laboratory.nnnRESULTSnOf 153 patients who provided consent, a total of 92 (82 men; average age: 63 years) were randomized (n = 61 in BMC group and n = 31 in placebo group). Changes in LVESV index (-0.9 mL/m(2) [95% CI, -6.1 to 4.3]; P = .73), maximal oxygen consumption (1.0 [95% CI, -0.42 to 2.34]; P = .17), and reversible defect (-1.2 [95% CI, -12.50 to 10.12]; P = .84) were not statistically significant. There were no differences found in any of the secondary outcomes, including percent myocardial defect, total defect size, fixed defect size, regional wall motion, and clinical improvement.nnnCONCLUSIONnAmong patients with chronic ischemic heart failure, transendocardial injection of autologous BMCs compared with placebo did not improve LVESV, maximal oxygen consumption, or reversibility on SPECT.nnnTRIAL REGISTRATIONnclinicaltrials.gov Identifier: NCT00824005.
JAMA | 2011
Jay H. Traverse; Timothy D. Henry; Stephen G. Ellis; Carl J. Pepine; James T. Willerson; David Zhao; John R. Forder; Barry J. Byrne; Antonis K. Hatzopoulos; Marc S. Penn; Emerson C. Perin; Kenneth W. Baran; Jeffrey W. Chambers; Charles R. Lambert; Ganesh Raveendran; Daniel I. Simon; Douglas E. Vaughan; Lara M. Simpson; Adrian P. Gee; Doris A. Taylor; Christopher R. Cogle; James D. Thomas; Guilherme V. Silva; Beth C. Jorgenson; Rachel E. Olson; Sherry Bowman; Judy Francescon; Carrie Geither; Eileen Handberg; Deirdre Smith
CONTEXTnClinical trial results suggest that intracoronary delivery of autologous bone marrow mononuclear cells (BMCs) may improve left ventricular (LV) function when administered within the first week following myocardial infarction (MI). However, because a substantial number of patients may not present for early cell delivery, the efficacy of autologous BMC delivery 2 to 3 weeks post-MI warrants investigation.nnnOBJECTIVEnTo determine if intracoronary delivery of autologous BMCs improves global and regional LV function when delivered 2 to 3 weeks following first MI.nnnDESIGN, SETTING, AND PATIENTSnA randomized, double-blind, placebo-controlled trial (LateTIME) of the National Heart, Lung, and Blood Institute-sponsored Cardiovascular Cell Therapy Research Network of 87 patients with significant LV dysfunction (LV ejection fraction [LVEF] ≤45%) following successful primary percutaneous coronary intervention (PCI) between July 8, 2008, and February 28, 2011.nnnINTERVENTIONSnIntracoronary infusion of 150 × 10(6) autologous BMCs (total nucleated cells) or placebo (BMC:placebo, 2:1) was performed within 12 hours of bone marrow aspiration after local automated cell processing.nnnMAIN OUTCOME MEASURESnChanges in global (LVEF) and regional (wall motion) LV function in the infarct and border zone between baseline and 6 months, measured by cardiac magnetic resonance imaging. Secondary end points included changes in LV volumes and infarct size.nnnRESULTSnA total of 87 patients were randomized (mean [SD] age, 57 [11] years; 83% men). Harvesting, processing, and intracoronary delivery of BMCs in this setting was feasible. Change between baseline and 6 months in the BMC group vs placebo for mean LVEF (48.7% to 49.2% vs 45.3% to 48.8%; between-group mean difference, -3.00; 95% CI, -7.05 to 0.95), wall motion in the infarct zone (6.2 to 6.5 mm vs 4.9 to 5.9 mm; between-group mean difference, -0.70; 95% CI, -2.78 to 1.34), and wall motion in the border zone (16.0 to 16.6 mm vs 16.1 to 19.3 mm; between-group mean difference, -2.60; 95% CI, -6.03 to 0.77) were not statistically significant. No significant change in LV volumes and infarct volumes was observed; both groups decreased by a similar amount at 6 months vs baseline.nnnCONCLUSIONnAmong patients with MI and LV dysfunction following reperfusion with PCI, intracoronary infusion of autologous BMCs vs intracoronary placebo infusion, 2 to 3 weeks after PCI, did not improve global or regional function at 6 months.nnnTRIAL REGISTRATIONnclinicaltrials.gov Identifier: NCT00684060.
Circulation Research | 2015
Mariann Gyöngyösi; Wojciech Wojakowski; Patricia Lemarchand; Ketil Lunde; Michal Tendera; Jozef Bartunek; Eduardo Marbán; Birgit Assmus; Timothy D. Henry; Jay H. Traverse; Lemuel A. Moyé; Daniel Sürder; Roberto Corti; Heikki V. Huikuri; Johanna A. Miettinen; Jochen Wöhrle; Slobodan Obradovic; Jérôme Roncalli; Konstantinos Malliaras; Evgeny Pokushalov; Alexander Romanov; Jens Kastrup; Martin W. Bergmann; Douwe E. Atsma; Axel Cosmus Pyndt Diederichsen; István Édes; Imre Benedek; Theodora Benedek; Hristo Pejkov; Noemi Nyolczas
RATIONALEnThe meta-Analysis of Cell-based CaRdiac study is the first prospectively declared collaborative multinational database, including individual data of patients with ischemic heart disease treated with cell therapy.nnnOBJECTIVEnWe analyzed the safety and efficacy of intracoronary cell therapy after acute myocardial infarction (AMI), including individual patient data from 12 randomized trials (ASTAMI, Aalst, BOOST, BONAMI, CADUCEUS, FINCELL, REGENT, REPAIR-AMI, SCAMI, SWISS-AMI, TIME, LATE-TIME; n=1252).nnnMETHODS AND RESULTSnThe primary end point was freedom from combined major adverse cardiac and cerebrovascular events (including all-cause death, AMI recurrance, stroke, and target vessel revascularization). The secondary end point was freedom from hard clinical end points (death, AMI recurrence, or stroke), assessed with random-effects meta-analyses and Cox regressions for interactions. Secondary efficacy end points included changes in end-diastolic volume, end-systolic volume, and ejection fraction, analyzed with random-effects meta-analyses and ANCOVA. We reported weighted mean differences between cell therapy and control groups. No effect of cell therapy on major adverse cardiac and cerebrovascular events (14.0% versus 16.3%; hazard ratio, 0.86; 95% confidence interval, 0.63-1.18) or death (1.4% versus 2.1%) or death/AMI recurrence/stroke (2.9% versus 4.7%) was identified in comparison with controls. No changes in ejection fraction (mean difference: 0.96%; 95% confidence interval, -0.2 to 2.1), end-diastolic volume, or systolic volume were observed compared with controls. These results were not influenced by anterior AMI location, reduced baseline ejection fraction, or the use of MRI for assessing left ventricular parameters.nnnCONCLUSIONSnThis meta-analysis of individual patient data from randomized trials in patients with recent AMI revealed that intracoronary cell therapy provided no benefit, in terms of clinical events or changes in left ventricular function.nnnCLINICAL TRIAL REGISTRATIONnURL: http://www.clinicaltrials.gov. Unique identifier: NCT01098591.
Circulation | 2014
Deborah D. Ascheim; Annetine C. Gelijns; Daniel R. Goldstein; Lemuel A. Moyé; Nicholas G. Smedira; S. Lee; Charles T. Klodell; Anita Szady; Michael K. Parides; Neal Jeffries; Donna Skerrett; Doris A. Taylor; J. Eduardo Rame; Carmelo A. Milano; Joseph G. Rogers; Janine Lynch; Todd M. Dewey; Eric J. Eichhorn; Benjamin Sun; David S. Feldman; Robert D. Simari; Patrick T. O’Gara; Wendy C. Taddei-Peters; Marissa A. Miller; Yoshifumi Naka; Emilia Bagiella; Eric A. Rose; Y. Joseph Woo
Background— Allogeneic mesenchymal precursor cells (MPCs) injected during left ventricular assist device (LVAD) implantation may contribute to myocardial recovery. This trial explores the safety and efficacy of this strategy. Methods and Results— In this multicenter, double-blind, sham-procedure controlled trial, 30 patients were randomized (2:1) to intramyocardial injection of 25 million MPCs or medium during LVAD implantation. The primary safety end point was incidence of infectious myocarditis, myocardial rupture, neoplasm, hypersensitivity reaction, and immune sensitization (90 days after randomization). Key efficacy end points were functional status and ventricular function while temporarily weaned from LVAD support (90 days after randomization). Patients were followed up until transplant or 12 months after randomization, whichever came first. Mean age was 57.4 (±13.6) years, mean left ventricular ejection fraction was 18.1%, and 66.7% were destination therapy LVADs. No safety events were observed. Successful temporary LVAD weaning was achieved in 50% of MPC and 20% of control patients at 90 days (P=0.24); the posterior probability that MPCs increased the likelihood of successful weaning was 93%. At 90 days, 3 deaths (30%) occurred in control patients, and none occurred in MPC patients. Mean left ventricular ejection fraction after successful wean was 24.0% (MPC=10) and 22.5% (control=2; P=0.56). At 12 months, 30% of MPC patients and 40% of control patients were successfully temporarily weaned from LVAD support (P=0.69), and 6 deaths (30%) occurred in MPC patients. Donor-specific HLA sensitization developed in 2 MPC and 3 control patients and resolved by 12 months. Conclusions— In this preliminary trial, administration of MPCs appeared to be safe, and there was a potential signal of efficacy. Future studies will evaluate the potential for higher or additional doses to enhance the ability to wean LVAD recipients off support. Clinical Trial Registration— URL: http://www.clinicaltrials.gov. Unique identifier: NCT01442129.
Circulation | 2013
Joshua M. Hare; Roberto Bolli; John P. Cooke; David J. Gordon; Timothy D. Henry; Emerson C. Perin; Keith L. March; Michael P. Murphy; Carl J. Pepine; Robert D. Simari; Sonia I. Skarlatos; Jay H. Traverse; James T. Willerson; Anita Szady; Doris A. Taylor; Rachel W. Vojvodic; Phillip C. Yang; Lemuel A. Moyé
Clinical trials demonstrate the best of medical expertise and epidemiological elegance. From the simple building blocks of contemporaneous control groups, randomization, and blinding, they assemble a clear picture of the nature of the treatment-effect relationship. This accomplishment has earned them the star ascendant position in cardiovascular research.nnTheir advantage was demonstrated with Bradford Hill’s work on streptomycin, and as knowledge of the pathogenesis of atherosclerotic disease produced possibilities for new treatments, cardiovascular researchers applied this new research tool to identify effective therapies in a sequential approach (Figure 1).nnnnFigure 1. nThe traditional phases of clinical trials. *The study takes place before Food and Drug Administration approval.nnnnThis work accelerated with clinical trials demonstrating treatment benefits for chronic diseases such as hypertension,1,2 lipid abnormalities,3,4 and heart failure,5,6 cementing their role in identifying new therapies to prevent the sequela of cardiovascular disease in vulnerable populations.nnHowever, the limitations of early clinical trial interpretation also appeared. The findings of the Multiple Risk Factor Intervention Trial (MRFIT),7 the International Verapamil SR/Trandolapril Study (INVEST),8,9 the Early Versus Late Intervention Trial With Estradiol (ELITE),7,10 the Cardiac Arrhythmia Suppression Trial (CAST),11,12 and the US Carvedilol program controversy13–19 together served to undermine the confidence of cardiologists in the interpretation of clinical trial results. Thought leaders in the field identified the interpretation problem (ie, the hyperreliance on P values to reflect positive results for any analysis in a clinical trial) and called for a fundamental change in the design and analysis of clinical trials.nnSubsequent work produced the following clinical trials principles: (1)There should be clear and prospective declaration in the written protocol about all important aspects of the clinical trial with particular emphasis on the end points …
American Heart Journal | 2010
James T. Willerson; Emerson C. Perin; Stephen G. Ellis; Carl J. Pepine; Timothy D. Henry; David Zhao; Dejian Lai; Marc S. Penn; Barry J. Byrne; Guilherme Silva; Adrian P. Gee; Jay H. Traverse; Antonis K. Hatzopoulos; John R. Forder; Daniel Martin; Marvin Kronenberg; Doris A. Taylor; Christopher R. Cogle; Sarah Baraniuk; Lynette Westbrook; Shelly L. Sayre; Rachel W. Vojvodic; David J. Gordon; Sonia I. Skarlatos; Lemuel A. Moyé; Robert D. Simari
BACKGROUNDnThe increasing worldwide prevalence of coronary artery disease (CAD) continues to challenge the medical community. Management options include medical and revascularization therapy. Despite advances in these methods, CAD is a leading cause of recurrent ischemia and heart failure, posing significant morbidity and mortality risks along with increasing health costs in a large patient population worldwide.nnnTRIAL DESIGNnThe Cardiovascular Cell Therapy Research Network (CCTRN) was established by the National Institutes of Health to investigate the role of cell therapy in the treatment of chronic cardiovascular disease. FOCUS is a CCTRN-designed randomized, phase II, placebo-controlled clinical trial that will assess the effect of autologous bone marrow mononuclear cells delivered transendocardially to patients with left ventricular (LV) dysfunction and symptomatic heart failure or angina. All patients need to have limiting ischemia by reversible ischemia on single-photon emission computed tomography assessment.nnnRESULTSnAfter thoughtful consideration of both statistical and clinical principles, we will recruit 87 patients (58 cell treated and 29 placebo) to receive either bone marrow-derived stem cells or placebo. Myocardial perfusion, LV contractile performance, and maximal oxygen consumption are the primary outcome measures.nnnCONCLUSIONSnThe designed clinical trial will provide a sound assessment of the effect of autologous bone marrow mononuclear cells in improving blood flow and contractile function of the heart. The target population is patients with CAD and LV dysfunction with limiting angina or symptomatic heat failure. Patient safety is a central concern of the CCTRN, and patients will be followed for at least 5 years.
Circulation Research | 2014
Robert D. Simari; Carl J. Pepine; Jay H. Traverse; Timothy D. Henry; Roberto Bolli; Daniel B. Spoon; Ed Yeh; Joshua M. Hare; Ivonne Hernandez Schulman; R. David Anderson; Charles R. Lambert; Shelly L. Sayre; Doris A. Taylor; Ray F. Ebert; Lemuel A. Moyé
To understand the role of bone marrow mononuclear cells in the treatment of acute myocardial infarction, this overview offers a retrospective examination of strengths and limitations of 3 contemporaneous trials with attention to critical design features and provides an analysis of the combined data set and implications for future directions in cell therapy for acute myocardial infarction.
Cytotherapy | 2010
Adrian P. Gee; Sara Richman; April G. Durett; David H. McKenna; Jay H. Traverse; Timothy D. Henry; Diann Fisk; Carl J. Pepine; Jeannette Bloom; James T. Willerson; Karen Prater; David Zhao; Jane Reese Koç; Steven Ellis; Doris A. Taylor; Christopher R. Cogle; Lemuel A. Moyé; Robert D. Simari; Sonia I. Skarlatos
Abstract Background aims. Multicenter cellular therapy clinical trials require the establishment and implementation of standardized cell-processing protocols and associated quality control (QC) mechanisms. The aims here were to develop such an infrastructure in support of the Cardiovascular Cell Therapy Research Network (CCTRN) and to report on the results of processing for the first 60 patients. Methods. Standardized cell preparations, consisting of autologous bone marrow (BM) mononuclear cells, prepared using a Sepax device, were manufactured at each of the five processing facilities that supported the clinical treatment centers. Processing staff underwent centralized training that included proficiency evaluation. Quality was subsequently monitored by a central QC program that included product evaluation by the CCTRN biorepositories. Results. Data from the first 60 procedures demonstrated that uniform products, that met all release criteria, could be manufactured at all five sites within 7 h of receipt of BM. Uniformity was facilitated by use of automated systems (the Sepax for processing and the Endosafe device for endotoxin testing), standardized procedures and centralized QC. Conclusions. Complex multicenter cell therapy and regenerative medicine protocols can, where necessary, successfully utilize local processing facilities once an effective infrastructure is in place to provide training and QC.
Journal of Cardiovascular Translational Research | 2010
Robert D. Simari; Lemuel A. Moyé; Sonia I. Skarlatos; Stephen G. Ellis; David Zhao; James T. Willerson; Timothy D. Henry; Carl J. Pepine
The emerging sciences of stem cell biology and cellular plasticity have led to the development of cell-based therapies for advanced human disease. Pre-clinical studies which defined the potential of bone marrow-derived mononuclear cells to repair damaged and dysfunctional myocardium led to the rapid advancement of these strategies to the clinic. Such rapid advancement has led to controversy regarding the appropriate conduct of such studies. In the United States, the National Heart, Lung, and Blood Institute established the Cardiovascular Cell Therapy Research Network (CCTRN) to facilitate the early translation of clinical trials of cell therapy for left ventricular dysfunction. The premise upon which the CCTRN was established was that multiple clinical trial sites would interact effectively with a Data Coordinating Center to perform early phase 1 and 2 clinical trials within a highly coordinated network structure. In order to develop this network, the unmet needs of the community needed to be defined, the clinical trials identified, and the structure to perform the studies needed to be established. This manuscript highlights the challenges in the development of the CCTRN and the approaches faced to define a network to perform clinical trials in human cell therapy of cardiovascular disease.
Annals of Oncology | 2015
Gael Deplanque; Martin Demarchi; Mohamed Hebbar; Patrick J. Flynn; Bohuslav Melichar; James M. Atkins; E. Nowara; Lemuel A. Moyé; D. Piquemal; D. Ritter; Patrice Dubreuil; Colin Mansfield; Y. Acin; Alain Moussy; Olivier Hermine; Pascal Hammel
Two subgroups showing significantly poor survival when treated with single-agent gemcitabine have been identified; one is defined by an overexpression of ACOX1 in blood, and the other via a pain intensity threshold. Masitinib in combination with gemcitabine appears to have a positive benefit–risk ratio in both subgroups. These findings could influence future trial design in pancreatic cancer.BACKGROUNDnMasitinib is a selective oral tyrosine-kinase inhibitor. The efficacy and safety of masitinib combined with gemcitabine was compared against single-agent gemcitabine in patients with advanced pancreatic ductal adenocarcinoma (PDAC).nnnPATIENTS AND METHODSnPatients with inoperable, chemotherapy-naïve, PDAC were randomized (1: 1) to receive gemcitabine (1000 mg/m2) in combination with either masitinib (9 mg/kg/day) or a placebo. The primary endpoint was overall survival (OS) in the modified intent-to-treat population. Secondary OS analyses aimed to characterize subgroups with poor survival while receiving single-agent gemcitabine with subsequent evaluation of masitinib therapeutic benefit. These prospectively declared subgroups were based on pharmacogenomic data or a baseline characteristic.nnnRESULTSnThree hundred and fifty-three patients were randomly assigned to receive either masitinib plus gemcitabine (N = 175) or placebo plus gemcitabine (N = 178). Median OS was similar between treatment-arms for the overall population, at respectively, 7.7 and 7.1 months, with a hazard ratio (HR) of 0.89 (95% CI [0.70; 1.13]. Secondary analyses identified two subgroups having a significantly poor survival rate when receiving single-agent gemcitabine; one defined by an overexpression of acyl-CoA oxidase-1 (ACOX1) in blood, and another via a baseline pain intensity threshold (VAS > 20 mm). These subgroups represent a critical unmet medical need as evidenced from median OS of 5.5 months in patients receiving single-agent gemcitabine, and comprise an estimated 63% of patients. A significant treatment effect was observed in these subgroups for masitinib with median OS of 11.7 months in the ACOX1 subgroup [HR = 0.23 (0.10; 0.51), P = 0.001], and 8.0 months in the pain subgroup [HR = 0.62 (0.43; 0.89), P = 0.012]. Despite an increased toxicity of the combination as compared with single-agent gemcitabine, side-effects remained manageable.nnnCONCLUSIONSnThe present data warrant initiation of a confirmatory study that may support the use of masitinib plus gemcitabine for treatment of PDAC patients with overexpression of ACOX1 or baseline pain (VAS > 20mm). Masitinibs effect in these subgroups is also supported by biological plausibility and evidence of internal clinical validation.nnnTRIAL REGISTRATIONnClinicalTrials.gov:NCT00789633.