Network


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

Hotspot


Dive into the research topics where Robert A. Preti is active.

Publication


Featured researches published by Robert A. Preti.


Journal of Clinical Oncology | 1998

CD34+CD33- cells influence days to engraftment and transfusion requirements in autologous blood stem-cell recipients.

Andrew Pecora; Robert A. Preti; Gilbert W. Gleim; Andrew Jennis; Kathleen Zahos; Susan Cantwell; Lisa Doria; Randi Isaacs; Alfred P. Gillio; Mary Ann Michelis; Joel A. Brochstein

PURPOSE To evaluate the reliability of CD34/CD33 subset enumeration as a predictor of hematopoietic repopulating potential in autologous blood stem-cell transplantation and to determine which patient and treatment-related factors affect the timing, quantity, and type of blood stem cells mobilized. PATIENTS AND METHODS We analyzed blood stem-cell collections from 410 consecutive cancer patients who received mobilization therapy and evaluated factors, including CD34+ subset quantities, that might influence engraftment kinetics and transfusion requirements in autologous blood stem-cell recipients. RESULTS The majority of patients (97%) mobilized CD34+33- cells, which were usually collected in the greatest quantity on the first day of apheresis. Patients who received only growth factor mobilized the highest percentage of CD34+33- cells. Extensive prior chemotherapy limited the collection of CD34+33- cells. In addition to patient diagnosis (P < .006) and total CD34+ cell dose (P = .0001), CD34+33- cell dose (P < .005) and percentage of CD34+33- cells (P < .005) were identified as independent factors significantly predictive of engraftment kinetics. CD34+33- cell dose (R2 < or = .177; P < .0001) was a strong and the only significant predictor of RBC and platelet transfusion requirements. Furthermore, independent of the total CD34+ cell dose, as the CD34+33- cell dose increased, days to neutrophil recovery, days to platelet recovery, and transfusion requirements decreased. CONCLUSION These findings show that CD34+33- cells are readily collected in most cancer patients and significantly influence engraftment kinetics and transfusion requirements in autologous blood stem-cell recipients. CD34+33- cell quantity of the blood stem-cell graft appears to be a more reliable predictor of hematopoietic recovery rates than total CD34+ cell quantity in this setting.


Circulation Research | 2017

PreSERVE-AMINovelty and Significance: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial of Intracoronary Administration of Autologous CD34+ Cells in Patients With Left Ventricular Dysfunction Post STEMI

Arshed A. Quyyumi; Alejandro Vasquez; Marc Klapholz; Gary L. Schaer; Ahmed Abdel-Latif; Stephen Frohwein; Timothy D. Henry; Richard A. Schatz; Nabil Dib; Catalin Toma; Charles J. Davidson; Gregory W. Barsness; David M. Shavelle; Martin H. Cohen; Joseph Poole; Thomas Moss; Pamela Hyde; Anna Maria Kanakaraj; Vitaly Druker; Amy Chung; Candice Junge; Robert A. Preti; Robin L. Smith; David J. Mazzo; Andrew Pecora; Douglas W. Losordo

Rationale: Despite direct immediate intervention and therapy, ST-segment–elevation myocardial infarction (STEMI) victims remain at risk for infarct expansion, heart failure, reinfarction, repeat revascularization, and death. Objective: To evaluate the safety and bioactivity of autologous CD34+ cell (CLBS10) intracoronary infusion in patients with left ventricular dysfunction post STEMI. Methods and Results: Patients who underwent successful stenting for STEMI and had left ventricular dysfunction (ejection fraction⩽48%) ≥4 days poststent were eligible for enrollment. Subjects (N=161) underwent mini bone marrow harvest and were randomized 1:1 to receive (1) autologous CD34+ cells (minimum 10 mol/L±20% cells; N=78) or (2) diluent alone (N=83), via intracoronary infusion. The primary safety end point was adverse events, serious adverse events, and major adverse cardiac event. The primary efficacy end point was change in resting myocardial perfusion over 6 months. No differences in myocardial perfusion or adverse events were observed between the control and treatment groups, although increased perfusion was observed within each group from baseline to 6 months (P<0.001). In secondary analyses, when adjusted for time of ischemia, a consistently favorable cell dose–dependent effect was observed in the change in left ventricular ejection fraction and infarct size, and the duration of time subjects was alive and out of hospital (P=0.05). At 1 year, 3.6% (N=3) and 0% deaths were observed in the control and treatment group, respectively. Conclusions: This PreSERVE-AMI (Phase 2, randomized, double-blind, placebo-controlled trial) represents the largest study of cell-based therapy for STEMI completed in the United States and provides evidence supporting safety and potential efficacy in patients with left ventricular dysfunction post STEMI who are at risk for death and major morbidity. Clinical Trial Registration: URL: http://www.clinicaltrials.gov. Unique identifier: NCT01495364.Rationale: Despite direct immediate intervention and therapy, ST-segment–elevation myocardial infarction (STEMI) victims remain at risk for infarct expansion, heart failure, reinfarction, repeat revascularization, and death. Objective: To evaluate the safety and bioactivity of autologous CD34+ cell (CLBS10) intracoronary infusion in patients with left ventricular dysfunction post STEMI. Methods and Results: Patients who underwent successful stenting for STEMI and had left ventricular dysfunction (ejection fraction≤48%) ≥4 days poststent were eligible for enrollment. Subjects (N=161) underwent mini bone marrow harvest and were randomized 1:1 to receive (1) autologous CD34+ cells (minimum 10 mol/L±20% cells; N=78) or (2) diluent alone (N=83), via intracoronary infusion. The primary safety end point was adverse events, serious adverse events, and major adverse cardiac event. The primary efficacy end point was change in resting myocardial perfusion over 6 months. No differences in myocardial perfusion or adverse events were observed between the control and treatment groups, although increased perfusion was observed within each group from baseline to 6 months ( P <0.001). In secondary analyses, when adjusted for time of ischemia, a consistently favorable cell dose–dependent effect was observed in the change in left ventricular ejection fraction and infarct size, and the duration of time subjects was alive and out of hospital ( P =0.05). At 1 year, 3.6% (N=3) and 0% deaths were observed in the control and treatment group, respectively. Conclusions: This PreSERVE-AMI (Phase 2, randomized, double-blind, placebo-controlled trial) represents the largest study of cell-based therapy for STEMI completed in the United States and provides evidence supporting safety and potential efficacy in patients with left ventricular dysfunction post STEMI who are at risk for death and major morbidity. Clinical Trial Registration: URL: . Unique identifier: [NCT01495364][1]. # Novelty and Significance {#article-title-42} [1]: /lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT01495364&atom=%2Fcircresaha%2F120%2F2%2F324.atom


Circulation Research | 2017

PreSERVE-AMINovelty and Significance

Arshed A. Quyyumi; Alejandro Vasquez; Marc Klapholz; Gary L. Schaer; Ahmed Abdel-Latif; Stephen Frohwein; Timothy D. Henry; Richard A. Schatz; Nabil Dib; Catalin Toma; Charles J. Davidson; Gregory W. Barsness; David M. Shavelle; Martin H. Cohen; Joseph Poole; Thomas Moss; Pamela Hyde; Anna Maria Kanakaraj; Vitaly Druker; Amy Chung; Candice Junge; Robert A. Preti; Robin L. Smith; David J. Mazzo; Andrew Pecora; Douglas W. Losordo

Rationale: Despite direct immediate intervention and therapy, ST-segment–elevation myocardial infarction (STEMI) victims remain at risk for infarct expansion, heart failure, reinfarction, repeat revascularization, and death. Objective: To evaluate the safety and bioactivity of autologous CD34+ cell (CLBS10) intracoronary infusion in patients with left ventricular dysfunction post STEMI. Methods and Results: Patients who underwent successful stenting for STEMI and had left ventricular dysfunction (ejection fraction⩽48%) ≥4 days poststent were eligible for enrollment. Subjects (N=161) underwent mini bone marrow harvest and were randomized 1:1 to receive (1) autologous CD34+ cells (minimum 10 mol/L±20% cells; N=78) or (2) diluent alone (N=83), via intracoronary infusion. The primary safety end point was adverse events, serious adverse events, and major adverse cardiac event. The primary efficacy end point was change in resting myocardial perfusion over 6 months. No differences in myocardial perfusion or adverse events were observed between the control and treatment groups, although increased perfusion was observed within each group from baseline to 6 months (P<0.001). In secondary analyses, when adjusted for time of ischemia, a consistently favorable cell dose–dependent effect was observed in the change in left ventricular ejection fraction and infarct size, and the duration of time subjects was alive and out of hospital (P=0.05). At 1 year, 3.6% (N=3) and 0% deaths were observed in the control and treatment group, respectively. Conclusions: This PreSERVE-AMI (Phase 2, randomized, double-blind, placebo-controlled trial) represents the largest study of cell-based therapy for STEMI completed in the United States and provides evidence supporting safety and potential efficacy in patients with left ventricular dysfunction post STEMI who are at risk for death and major morbidity. Clinical Trial Registration: URL: http://www.clinicaltrials.gov. Unique identifier: NCT01495364.Rationale: Despite direct immediate intervention and therapy, ST-segment–elevation myocardial infarction (STEMI) victims remain at risk for infarct expansion, heart failure, reinfarction, repeat revascularization, and death. Objective: To evaluate the safety and bioactivity of autologous CD34+ cell (CLBS10) intracoronary infusion in patients with left ventricular dysfunction post STEMI. Methods and Results: Patients who underwent successful stenting for STEMI and had left ventricular dysfunction (ejection fraction≤48%) ≥4 days poststent were eligible for enrollment. Subjects (N=161) underwent mini bone marrow harvest and were randomized 1:1 to receive (1) autologous CD34+ cells (minimum 10 mol/L±20% cells; N=78) or (2) diluent alone (N=83), via intracoronary infusion. The primary safety end point was adverse events, serious adverse events, and major adverse cardiac event. The primary efficacy end point was change in resting myocardial perfusion over 6 months. No differences in myocardial perfusion or adverse events were observed between the control and treatment groups, although increased perfusion was observed within each group from baseline to 6 months ( P <0.001). In secondary analyses, when adjusted for time of ischemia, a consistently favorable cell dose–dependent effect was observed in the change in left ventricular ejection fraction and infarct size, and the duration of time subjects was alive and out of hospital ( P =0.05). At 1 year, 3.6% (N=3) and 0% deaths were observed in the control and treatment group, respectively. Conclusions: This PreSERVE-AMI (Phase 2, randomized, double-blind, placebo-controlled trial) represents the largest study of cell-based therapy for STEMI completed in the United States and provides evidence supporting safety and potential efficacy in patients with left ventricular dysfunction post STEMI who are at risk for death and major morbidity. Clinical Trial Registration: URL: . Unique identifier: [NCT01495364][1]. # Novelty and Significance {#article-title-42} [1]: /lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT01495364&atom=%2Fcircresaha%2F120%2F2%2F324.atom


Journal of the American College of Cardiology | 2015

ONE YEAR FOLLOW-UP RESULTS FROM PRESERVE-AMI: A RANDOMIZED, DOUBLE-BLIND, PLACEBO CONTROLLED CLINICAL TRIAL OF INTRACORONARY INFUSION OF AUTOLOGOUS CD34+ CELLS IN PATIENTS WITH LEFT VENTRICULAR DYSFUNCTION POST STEMI

Arshed A. Quyyumi; David M. Shavelle; Timothy D. Henry; Ali E. Denktas; Ahmed Abdel-Latif; Catalin Toma; Gregory W. Barsness; Stephen Frohwein; Richard A. Schatz; Martin H. Cohen; Charles J. Davidson; Nabil Dib; Marc Klapholz; Gary L. Schaer; Alejandro Vasquez; Andrew Pecora; Thomas Moss; Pamela Hyde; Anna Maria Kanakaraj; Le Dich; Vitaly Druker; Candice Junge; Robert A. Preti; Douglas W. Losordo

ST segment Elevation Myocardial Infarction (STEMI) affects 160,000 annually in the US. Guidelines direct immediate revascularization and adjunctive medical therapies. Yet STEMI victims remain at risk for infarct expansion, heart failure, reinfarction, repeat revascularization and death. In pre-


Blood | 2002

Unusual viral infections (progressive multifocal leukoencephalopathy and cytomegalovirus disease) after high-dose chemotherapy with autologous blood stem cell rescue and peritransplantation rituximab

Stuart L. Goldberg; Andrew Pecora; Robert S. Alter; Mark S. Kroll; Scott D. Rowley; Stanley E. Waintraub; Kavita Imrit; Robert A. Preti


Circulation | 2014

Abstract 17457: PreSERVE-AMI: a Randomized, Double-Blind, Placebo-Controlled Clinical Trial of Intracoronary Administration of Autologous CD34+ Cells in Patients with Left Ventricular Dysfunction Post STEMI

Arshed A. Quyyumi; Alejandro Vasquez; Marc Klapholz; Gary L. Schaer; Ken Fujise; Ahmed Abdel-Latif; Robert S Iwaoka; Ali E. Denktas; Roger S. Gammon; Steve Frohwein; Vijaykumar S. Kasi; Michael R. Tamberella; Catalin Toma; Nabil Dib; Tanvir Bajwa; Richard A. Schatz; Timothy D. Henry; Martin Cohen; David M. Shavelle; Gregory W. Barsness; Charles Davidson; Thomas Moss; Pamela Hyde; AnnaMarie Kanakaraj; Vitaly Druker; Le Dich; Jonathan Sackner-Bernstein; Robert A. Preti; Douglas W. Losordo; Andrew Pecora


Journal of hematotherapy | 1997

An Intralaboratory Quality Control Program for Quantitation of CD34+ Cells by Flow Cytometry

Timothy J. Farley; William Rooney; Edward Kuhns; Tauseef Ahmed; Robert A. Preti


Journal of hematotherapy | 1997

Optimization of CD34+ Cell Selection Using Immunomagnetic Beads: Implications for Use in Cryopreserved Peripheral Blood Stem Cell Collections

Timothy J. Farley; Tauseef Ahmed; Maura Fitzgerald; Robert A. Preti


Archive | 2006

Compositions and methods of vascular injury repair

Andrew L. Pecora; Robert A. Preti


Biology of Blood and Marrow Transplantation | 2004

Unrelated donor hematopoietic stem cell transplantation for patients with hematologic malignancies using a nonmyeloablative conditioning regimen of fludarabine, low-dose total body irradiation, and rabbit antithymocyte globulin.

Scott D. Rowley; Stuart L. Goldberg; Andrew L. Pecora; Jack S. Hsu; Barbara Adler Brecher; Linda Butrin; Kelly Kobbe; Phyllis McKiernan; Robert A. Preti

Collaboration


Dive into the Robert A. Preti's collaboration.

Top Co-Authors

Avatar

Andrew L. Pecora

Hackensack University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Andrew Pecora

University of Medicine and Dentistry of New Jersey

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thomas Moss

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Stuart L. Goldberg

Hackensack University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Tauseef Ahmed

New York Medical College

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alejandro Vasquez

Huntsville Hospital System

View shared research outputs
Top Co-Authors

Avatar

Catalin Toma

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

David M. Shavelle

University of Southern California

View shared research outputs
Researchain Logo
Decentralizing Knowledge