Network


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

Hotspot


Dive into the research topics where Andrew L. Pecora is active.

Publication


Featured researches published by Andrew L. Pecora.


American Heart Journal | 2011

CD34+ cell infusion after ST elevation myocardial infarction is associated with improved perfusion and is dose dependent

Arshed A. Quyyumi; Edmund K. Waller; Jonathan R. Murrow; Fabio Esteves; James R. Galt; John N. Oshinski; Stamatios Lerakis; Salman Sher; Douglas E. Vaughan; Emerson C. Perin; James T. Willerson; Bernard J. Gersh; Douglas Gregory; Astrid Werner; Thomas J. Moss; Wai Shun Chan; Robert A. Preti; Andrew L. Pecora

BACKGROUND the objective of the study was to determine whether the effects of infarct-related artery (IRA) infusion of autologous bone marrow-derived CD34(+) cells after ST elevation myocardial infarction (STEMI) are dependent on the dose (quantity and mobility) of the cells infused. Beneficial effects of IRA infusion of mononuclear cells after STEMI have been inconsistent, possibly because of differences in timing, cell type, quantity, and mobility of infused cells. METHODS patients were randomized to bone marrow harvest (n = 16) or control (n = 15). At a median of 8.3 days after coronary stenting for STEMI, CD34(+) cells were infused via the IRA at 3 dose levels (5, 10, and 15 × 10(6)) in cohorts of 5 patients each. Baseline and follow-up imaging and ex vivo CD34(+) cell mobility were performed. RESULTS Cell harvest and infusion were safe. Quantitative rest hypoperfusion score measured by single-photon emission computed tomography improved at 6 months in the ≥ 10 million cohorts compared with controls (-256 vs +14, P = .02). There was a trend toward improved ejection fraction at 6 months (+4.5%) in the ≥ 10 million cohorts compared with no change in the controls and 5 million cohort (+0.7%). Improved perfusion and infarct size reduction correlated with the quantity and mobility of the infused CD34(+) cells. CONCLUSIONS the effects of CD34(+) cell IRA infusion during the repair phase after STEMI are dose dependent and, at a threshold dose of 10 million CD34(+) cells, associated with a significant improvement in perfusion that may limit deterioration in cardiac function (IRA infusion of CD34(+) cells in patients with acute myocardial infarction [AMR-01] NCT00313339).


Journal of Clinical Bioinformatics | 2015

Clinical decision support systems for improving diagnostic accuracy and achieving precision medicine.

Christian Castaneda; Kip Nalley; Ciaran Mannion; Pritish K. Bhattacharyya; Patrick Blake; Andrew L. Pecora; Andre Goy; K. Stephen Suh

As research laboratories and clinics collaborate to achieve precision medicine, both communities are required to understand mandated electronic health/medical record (EHR/EMR) initiatives that will be fully implemented in all clinics in the United States by 2015. Stakeholders will need to evaluate current record keeping practices and optimize and standardize methodologies to capture nearly all information in digital format. Collaborative efforts from academic and industry sectors are crucial to achieving higher efficacy in patient care while minimizing costs. Currently existing digitized data and information are present in multiple formats and are largely unstructured. In the absence of a universally accepted management system, departments and institutions continue to generate silos of information. As a result, invaluable and newly discovered knowledge is difficult to access. To accelerate biomedical research and reduce healthcare costs, clinical and bioinformatics systems must employ common data elements to create structured annotation forms enabling laboratories and clinics to capture sharable data in real time. Conversion of these datasets to knowable information should be a routine institutionalized process. New scientific knowledge and clinical discoveries can be shared via integrated knowledge environments defined by flexible data models and extensive use of standards, ontologies, vocabularies, and thesauri. In the clinical setting, aggregated knowledge must be displayed in user-friendly formats so that physicians, non-technical laboratory personnel, nurses, data/research coordinators, and end-users can enter data, access information, and understand the output. The effort to connect astronomical numbers of data points, including ‘-omics’-based molecular data, individual genome sequences, experimental data, patient clinical phenotypes, and follow-up data is a monumental task. Roadblocks to this vision of integration and interoperability include ethical, legal, and logistical concerns. Ensuring data security and protection of patient rights while simultaneously facilitating standardization is paramount to maintaining public support. The capabilities of supercomputing need to be applied strategically. A standardized, methodological implementation must be applied to developed artificial intelligence systems with the ability to integrate data and information into clinically relevant knowledge. Ultimately, the integration of bioinformatics and clinical data in a clinical decision support system promises precision medicine and cost effective and personalized patient care.


Cancer | 2012

Post‐treatment (not interim) positron emission tomography‐computed tomography scan status is highly predictive of outcome in mantle cell lymphoma patients treated with R‐HyperCVAD

Anthony R Mato; Jakub Svoboda; Tatyana Feldman; Tania Zielonka; Harry Agress; David Panush; Mitchell Miller; Patrick Toth; Paul M. Lizotte; Sunita D. Nasta; Stuart L. Goldberg; Emeline Chong; Steven Schuster; Andrew L. Pecora; Andre Goy

Although convincing data exist regarding the prognostic utility of positron emission tomographic (PET)‐computed tomographic (CT) imaging in Hodgkin lymphoma and diffuse large B‐cell lymphoma, its prognostic utility both during treatment and immediately after treatment have not been systematically evaluated in a large mantle cell lymphoma (MCL) patient cohort to support its use in clinical practice.


Bone Marrow Transplantation | 1999

Impact of stem cell dose on hematopoietic recovery in autologous blood stem cell recipients.

Andrew L. Pecora

Hematopoietic recovery rates following high-dose chemotherapy in autologous blood stem cell recipients have been shown, in part, to be dependent on the source and quantity of hematopoietic stem cells infused. Mobilized blood stem cell quantity (identified by the surface expression of the CD34 antigen) has been demonstrated in multiple studies to be the strongest predictor of days to hematopoietic recovery (ie platelets and neutrophils) in autologous blood stem cell recipients. Additionally, data from four large studies confirm that prompt and sustained hematopoietic recovery will occur in the majority of patients treated with high-dose chemotherapy if a stem cell dose 5 × 106/kg is administered. However, multiple aphereses are needed in the majority of patients to achieve these optimal stem cell doses. Problems associated with multiple aphereses procedures include hypocalcemia, anemia, thrombocytopenia, infection, thrombosis, increased costs, and malignant cell contamination of the apheresis product. Recent data indicate that less differentiated (eg CD33−) stem and progenitor cells result in both early and sustained hematopoietic recovery in bone marrow and blood stem cell recipients. Future trials using new growth factors such as stem cell factor (which has been shown to increase CD34+/CD33− cell mobilization), as well as improvements in purging strategies are needed to ensure prompt, sustained, and malignant-cell-free engraftment for the majority of autologous blood stem cell recipients.


Blood | 2013

Phase 2 clinical trial of rapamycin-resistant donor CD4+ Th2/Th1 (T-Rapa) cells after low-intensity allogeneic hematopoietic cell transplantation

Daniel H. Fowler; Miriam E. Mossoba; Seth M. Steinberg; David Halverson; David F. Stroncek; Hahn Khuu; Frances T. Hakim; Luciano Castiello; Marianna Sabatino; Susan F. Leitman; Jacopo Mariotti; Juan Gea-Banacloche; Claude Sportes; Nancy M. Hardy; Dennis D. Hickstein; Steven Z. Pavletic; Scott D. Rowley; Andre Goy; Michele L. Donato; Robert Korngold; Andrew L. Pecora; Bruce L. Levine; Carl H. June; Ronald E. Gress; Michael R. Bishop

In experimental models, ex vivo induced T-cell rapamycin resistance occurred independent of T helper 1 (Th1)/T helper 2 (Th2) differentiation and yielded allogeneic CD4(+) T cells of increased in vivo efficacy that facilitated engraftment and permitted graft-versus-tumor effects while minimizing graft-versus-host disease (GVHD). To translate these findings, we performed a phase 2 multicenter clinical trial of rapamycin-resistant donor CD4(+) Th2/Th1 (T-Rapa) cells after allogeneic-matched sibling donor hematopoietic cell transplantation (HCT) for therapy of refractory hematologic malignancy. T-Rapa cell products, which expressed a balanced Th2/Th1 phenotype, were administered as a preemptive donor lymphocyte infusion at day 14 post-HCT. After T-Rapa cell infusion, mixed donor/host chimerism rapidly converted, and there was preferential immune reconstitution with donor CD4(+) Th2 and Th1 cells relative to regulatory T cells and CD8(+) T cells. The cumulative incidence probability of acute GVHD was 20% and 40% at days 100 and 180 post-HCT, respectively. There was no transplant-related mortality. Eighteen of 40 patients (45%) remain in sustained complete remission (range of follow-up: 42-84 months). These results demonstrate the safety of this low-intensity transplant approach and the feasibility of subsequent randomized studies to compare T-Rapa cell-based therapy with standard transplantation regimens.


British Journal of Haematology | 2014

Addition of lenalidomide to rituximab, ifosfamide, carboplatin, etoposide (RICER) in first-relapse/primary refractory diffuse large B-cell lymphoma

Tatyana Feldman; Anthony R Mato; Kar F. Chow; Ewelina A Protomastro; Kara Yannotti; Pritish K. Bhattacharyya; Xiao Yang; Michele L. Donato; Scott D. Rowley; Carolanne Carini; Marisa Valentinetti; Judith Smith; Gabriella Gadaleta; Coleen Bejot; Susan Stives; Mary Timberg; Sabrina Kdiry; Andrew L. Pecora; Anne W. Beaven; Andre Goy

Relapsed/refractory diffuse large B‐cell lymphoma (DLBCL) is associated with a poor prognosis. Outcomes are particularly poor following immunochemotherapy failure or relapse within 12 months of induction. We conducted a Phase I/II trial of lenalidomide plus RICE (rituximab, ifosfamide, carboplatin, and etoposide) (RICER) as a salvage regimen for first‐relapse or primary refractory DLBCL. Dose‐escalated lenalidomide was combined with RICE every 14 d. After three cycles of RICER, patients with chemosensitive disease underwent stem cell collection and consolidation with BEAM [BCNU (carmustine), etoposide, cytarabine, melphalan] followed by autologous stem cell transplantation (autoSCT). Patients who recovered from autoSCT toxicities within 90 d initiated maintenance treatment with lenalidomide 25 mg daily for 21 d every 28 d for 12 months. No dose‐limiting or unexpected toxicities occurred with lenalidomide 25 mg plus RICE. Grade 3/4 haematological toxicities resolved appropriately, and planned dose density and dose intensity of RICER were preserved. No lenalidomide or RICE dose reductions were required in any of the three cycles. After two cycles of RICER, nine of 15 patients (60%) achieved a complete response, and two achieved a partial response (13%). Combining lenalidomide with RICE is feasible, and results in promising response rates (particularly complete response rates) in high‐risk DLBCL patients.


Circulation Research | 2017

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; 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 Cohen; Joseph Poole; Thomas J. Moss; Pamela Hyde; Anna Maria Kanakaraj; Vitaly Druker; Amy Chung; Candice Junge; Robert A. Preti; Robin L. Smith; David J. Mazzo; Andrew L. 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


Leukemia & Lymphoma | 2011

A phase III study of anti-B4-blocked ricin as adjuvant therapy post-autologous bone marrow transplant: CALGB 9254

Richard R. Furman; Michael L. Grossbard; Jeffrey L. Johnson; Andrew L. Pecora; Peter A. Cassileth; Sin-Ho Jung; Bruce A. Peterson; Lee M. Nadler; Arnold S. Freedman; Ruthee-Lu Bayer; Nancy L. Bartlett; David D. Hurd; Bruce D. Cheson

Anti-B4-blocked ricin (anti-B4-bR) is a potent immunotoxin directed against the CD19 antigen. Previous phase I and II studies suggested a possible role for anti-B4-bR as consolidation after high-dose chemotherapy and autologous stem cell transplant. Cancer and Leukemia Group B (CALGB) 9254 is a phase III study which randomized 157 patients with B-cell lymphoma in complete remission following autologous transplant to treatment with anti-B4-bR or observation. With a median follow-up time for patients of 5.8 years, the median event-free survival for protocol treatment and observation are 2.1 and 2.9 years, respectively (p = 0.275). The median overall survival for treatment and observation are 6.1 years and not reached, respectively (p = 0.063). Therefore, no differences were found in event-free survival and overall survival between protocol treatment and observation, although there was a trend toward improved survival with observation. These data fail to support a role for anti-B4-bR as consolidative therapy after bone marrow transplant in patients with B-cell lymphoma.


Cytotherapy | 2001

Tumor cell depletion of peripheral blood progenitor cells using positive and positive/negative selection in metastatic breast cancer

Ra Preti; Hillard M. Lazarus; Jane N. Winter; Edward A. Stadtmauer; S. Nadasi; J. McMannis; S. Karandish; Andrew Jennis; S.L. Goldberg; Andrew L. Pecora

BACKGROUND The clinical relevance of tumor cell purging of hematopoietic progenitor cell grafts has yet to be conclusively determined. Therefore, in addition to the demonstration that a method for graft purification is capable of removing an adequate number of tumor cells, it is critical that the procedure has as benign an impact upon the hematopoietic repopulating potential of the graft as possible. We evaluated tumor cell depletion, recovery of CD34(+) cells and post transplant engraftment kinetics as accepted measures of the effectiveness of an immunomagnetic bead (positive and positive/negative) purging methodology. METHODS The patients received either positive selection (CD34 selection alone) or a combination of positive and negative (CD34 selection followed by breast cancer cell depletion) using the Isolex 300 (automated and semiautomated) devices. Immunocytochemistry was used to determine the degree of breast cancer cell contamination before and after the selection procedures to determine the efficacy of the procedure. CD34 enumeration was employed to evaluate the recovery and purity of the CD34-selected cellular products and engraftment indices (days to absolute neutrophil count (ANC) recovery and platelet count (Plt) recovery and transfusion requirements) were evaluated to determine the safety of the procedure. RESULTS A total of 130 aphereses was performed on 101 patients. Ten pairs of collections were pooled before selection to increase the likelihood of achieving CD34 dose goals after selection. In all, 100 positive selections and 20 positive/negative selections were performed. Of the 10 (10.4%) ICC-positive preselection samples, 2 products showed persistent contamination after processing. The majority of patients (85.4%) required one selection procedure to achieve an adequate CD34(+) selected cell dose. Median CD34(+) cell recovery was > 50% for positive selection procedures and > 60% for the positive/negative procedures. The dose of CD34(+) cells infused ranged from 0.76 x 10(6) CD34(+) cells/kg to 27.7 x 106 CD34(+) cells/kg. There were no significant delays in neutrophil or platelet recovery or infections between any of the treatment groups. DISCUSSION CD34 selection alone or in combination with negative selection can result in a significant reduction of contaminating tumor cells in the peripheral blood progenitor cell autograft. Although there was one engraftment failure with the CD34-positive selected cells, transplantation of the selected products after high-dose chemotherapy for metastatic breast cancer did not result in a clinically significant delay in the hematopoietic reconstitutive capacity of the autografts.


Journal of Hematology & Oncology | 2013

Fibroblast growth factor-2 (FGF2) and syndecan-1 (SDC1) are potential biomarkers for putative circulating CD15+/CD30+ cells in poor outcome Hodgkin lymphoma patients.

Rajendra Gharbaran; Andre Goy; Takemi Tanaka; Jongwhan Park; Chris Kim; Nafis Hasan; Swathi Vemulapalli; Sreeja Sarojini; Madalina Tuluc; Kip Nalley; Pritish K. Bhattacharyya; Andrew L. Pecora; K. Stephen Suh

BackgroundHigh risk, unfavorable classical Hodgkin lymphoma (cHL) includes those patients with primary refractory or early relapse, and progressive disease. To improve the availability of biomarkers for this group of patients, we investigated both tumor biopsies and peripheral blood leukocytes (PBL) of untreated (chemo-naïve, CN) Nodular Sclerosis Classic Hodgkin Lymphoma (NS-cHL) patients for consistent biomarkers that can predict the outcome prior to frontline treatment.Methods and materialsBioinformatics data mining was used to generate 151 candidate biomarkers, which were screened against a library of 10 HL cell lines. Expression of FGF2 and SDC1 by CD30+ cells from HL patient samples representing good and poor outcomes were analyzed by qRT-PCR, immunohistochemical (IHC), and immunofluorescence analyses.ResultsTo identify predictive HL-specific biomarkers, potential marker genes selected using bioinformatics approaches were screened against HL cell lines and HL patient samples. Fibroblast Growth Factor-2 (FGF2) and Syndecan-1 (SDC1) were overexpressed in all HL cell lines, and the overexpression was HL-specific when compared to 116 non-Hodgkin lymphoma tissues. In the analysis of stratified NS-cHL patient samples, expression of FGF2 and SDC1 were 245 fold and 91 fold higher, respectively, in the poor outcome (PO) group than in the good outcome (GO) group. The PO group exhibited higher expression of the HL marker CD30, the macrophage marker CD68, and metastatic markers TGFβ1 and MMP9 compared to the GO group. This expression signature was confirmed by qualitative immunohistochemical and immunofluorescent data. A Kaplan-Meier analysis indicated that samples in which the CD30+ cells carried an FGF2+/SDC1+ immunophenotype showed shortened survival. Analysis of chemo-naive HL blood samples suggested that in the PO group a subset of CD30+ HL cells had entered the circulation. These cells significantly overexpressed FGF2 and SDC1 compared to the GO group. The PO group showed significant down-regulation of markers for monocytes, T-cells, and B-cells. These expression signatures were eliminated in heavily pretreated patients.ConclusionThe results suggest that small subsets of circulating CD30+/CD15+ cells expressing FGF2 and SDC1 represent biomarkers that identify NS-cHL patients who will experience a poor outcome (primary refractory and early relapsing).

Collaboration


Dive into the Andrew L. Pecora's collaboration.

Top Co-Authors

Avatar

Scott D. Rowley

Hackensack University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Andre Goy

Hackensack University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Michele L. Donato

Hackensack University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Stuart L. Goldberg

Hackensack University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Tatyana Feldman

Hackensack University Medical Center

View shared research outputs
Top Co-Authors

Avatar

David H. Vesole

Hackensack University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Anthony R Mato

Hackensack University Medical Center

View shared research outputs
Top Co-Authors

Avatar

David Siegel

Hackensack University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Phyllis McKiernan

Hackensack University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge