Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Circ Res ; 120(2): 324-331, 2017 Jan 20.
Article in English | MEDLINE | ID: mdl-27821724

ABSTRACT

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.


Subject(s)
Antigens, CD34/administration & dosage , Bone Marrow Transplantation/methods , ST Elevation Myocardial Infarction/diagnostic imaging , ST Elevation Myocardial Infarction/therapy , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/therapy , Aged , Coronary Vessels/diagnostic imaging , Coronary Vessels/drug effects , Double-Blind Method , Female , Humans , Infusions, Intra-Arterial/methods , Male , Middle Aged , ST Elevation Myocardial Infarction/complications , Transplantation, Autologous/methods , Treatment Outcome , Ventricular Dysfunction, Left/etiology
2.
Cytotherapy ; 16(11): 1558-1574, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24927716

ABSTRACT

BACKGROUND AIMS: Evaluation of the BD Stem Cell Enumeration Kit was conducted at four clinical sites with flow cytometry CD34(+) enumeration to assess agreement between two investigational methods: (i) the BD FACSCanto II and BD FACSCalibur systems and (ii) the predicate method (Beckman Coulter StemKit and StemTrol, Immunotech SAS, Beckman Coulter, Marseille Cedex 9, France). METHODS: Leftover and delinked specimens (n = 1032) from clinical flow cytometry testing were analyzed on the BD FACSCanto II (n = 918) and BD FACSCalibur (n = 905) in normal and mobilized blood, frozen and thawed bone marrow and leucopheresis and cord blood anticoagulated with citrate phosphate dextrose, anticoagulant citrate dextrose-solution A, heparin and ethylenediaminetetraacetate, alone or in combination. Fresh leucopheresis analysis addressed site equivalency for sample preparation, testing and analysis. RESULTS: The mean relative bias showed agreement within predefined parameters for the BD FACSCanto II (-2.81 to 4.31 ±7.1) and BD FACSCalibur (-2.69 to 5.2 ±7.9). Results are reported as absolute and relative differences compared with the predicate for viable CD34(+), percentage of CD34(+) in CD45(+) and viable CD45(+) populations (or gates). Bias analyses of the distribution of the predicate low, mid and high bin values were done using BD FACSCanto II optimal gating and BD FACSCalibur manual gating for viable CD34(+), percentage of CD34(+) in CD45(+) and viable CD45(+). Bias results from both investigational methods show agreement. Deming regression analyses showed a linear relationship with R(2) > 0.92 for both investigational methods. DISCUSSION: In conclusion, the results from both investigational methods demonstrated agreement and equivalence with the predicate method for enumeration of absolute viable CD34(+), percentage of viable CD34(+) in CD45(+) and absolute viable CD45(+) populations.


Subject(s)
Antigens, CD34/metabolism , Flow Cytometry/methods , Stem Cell Transplantation , Stem Cells/cytology , Antigens, CD34/immunology , Cell Count , Cell Lineage/genetics , Fetal Blood/cytology , Fetal Blood/immunology , Flow Cytometry/instrumentation , Humans , Stem Cells/immunology
3.
Am Heart J ; 161(1): 98-105, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21167340

ABSTRACT

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).


Subject(s)
Antigens, CD34 , Bone Marrow Cells/immunology , Bone Marrow Transplantation/methods , Coronary Circulation/physiology , Electrocardiography , Myocardial Infarction/therapy , Coronary Vessels , Female , Follow-Up Studies , Humans , Infusions, Intra-Arterial , Magnetic Resonance Imaging , Male , Middle Aged , Myocardial Infarction/diagnosis , Myocardial Infarction/physiopathology , Tomography, Emission-Computed, Single-Photon , Treatment Outcome
4.
J Nucl Cardiol ; 18(5): 912-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21717276

ABSTRACT

BACKGROUND: Left ventricular dyssynchrony is an adverse consequence of ST-elevation myocardial infarction (STEMI) and bears an unfavorable prognosis. Mechanical dyssynchrony as measured by phase analysis from gated single photon emission computed tomography (GSPECT) correlates well with other imaging methods of assessing dyssynchrony but has not been studied in STEMI. We hypothesized that systolic dyssynchrony as measured by GSPECT would correlate with adverse remodeling after STEMI. METHODS: In 28 subjects suffering STEMI, GSPECT with technetium-99m sestamibi was performed immediately after presentation (day 5) and remotely (6 months). Parameters of left ventricular dyssynchrony (QRS width, histogram bandwidth (HBW) and phase standard deviation (PSD)) were measured from GSPECT using the Emory Cardiac Toolbox. Left ventricular volumes, ejection fraction (LVEF) and infarct size were also assessed. RESULTS: After successful primary percutaneous coronary intervention to the infarct-related artery, subjects had an LVEF of 46.4% ± 11% and a resting perfusion defect of 27.4% ± 16% at baseline. Baseline QRS width was normal (91.5 ± 17.5 ms). Subjects with STEMI had dyssynchrony compared with a cohort of 22 normal subjects (age 57.2 ± 10.6 years, <5% perfusion defect) by both HBW (100.3° ± 70.7° vs 26.5° ± 5.3°, P < .0001) and PSD (35.3° ± 16.9° vs 7.9° ± 2.1°, P < .0001). Baseline HBW correlated with resting perfusion defect size (r = 0.67, P < .001), end-systolic volume (r = 0.72, P < .001), end-diastolic volume (r = 0.63, P = .001), and inversely with LVEF (r = -0.74, P < .001). HBW and PSD improved over the follow-up period (-24.1 ± 35.9 degrees, P = .003 and -8.7° ± 14.6°, P = .006, respectively), and improvement in HBW correlated with reduction in LV end-systolic volumes (r = 0.43, P = .034). Baseline HBW and PSD, however, did not independently predict LVEF at 6 months follow-up. CONCLUSIONS: After STEMI, subjects exhibit mechanical dyssynchrony as measured by GSPECT phase analysis without evidence of electrical dyssynchrony. Improvement in mechanical dyssynchrony correlates with beneficial ventricular remodeling. The full predictive value of this measure in post-infarct patients warrants further study.


Subject(s)
Myocardial Infarction/diagnostic imaging , Myocardial Infarction/physiopathology , Tomography, Emission-Computed, Single-Photon/methods , Adult , Aged , Electrocardiography , Female , Humans , Male , Middle Aged , Systole , Technetium Tc 99m Sestamibi , Ventricular Function, Left
6.
Nat Rev Drug Discov ; 19(2): 93-111, 2020 02.
Article in English | MEDLINE | ID: mdl-31836861

ABSTRACT

Most rare diseases still lack approved treatments despite major advances in research providing the tools to understand their molecular basis, as well as legislation providing regulatory and economic incentives to catalyse the development of specific therapies. Addressing this translational gap is a multifaceted challenge, for which a key aspect is the selection of the optimal therapeutic modality for translating advances in rare disease knowledge into potential medicines, known as orphan drugs. With this in mind, we discuss here the technological basis and rare disease applicability of the main therapeutic modalities, including small molecules, monoclonal antibodies, protein replacement therapies, oligonucleotides and gene and cell therapies, as well as drug repurposing. For each modality, we consider its strengths and limitations as a platform for rare disease therapy development and describe clinical progress so far in developing drugs based on it. We also discuss selected overarching topics in the development of therapies for rare diseases, such as approval statistics, engagement of patients in the process, regulatory pathways and digital tools.


Subject(s)
Drug Approval , Drug Development , Drug Repositioning/statistics & numerical data , Orphan Drug Production/statistics & numerical data , Rare Diseases/drug therapy , Humans
7.
Nat Biomed Eng ; 2(6): 362-376, 2018 06.
Article in English | MEDLINE | ID: mdl-31011198

ABSTRACT

The achievements of cell-based therapeutics have galvanized efforts to bring cell therapies to the market. To address the demands of the clinical and eventual commercial-scale production of cells, and with the increasing generation of large clinical datasets from chimeric antigen receptor T-cell immunotherapy, from transplants of engineered haematopoietic stem cells and from other promising cell therapies, an emphasis on biomanufacturing requirements becomes necessary. Robust infrastructure should address current limitations in cell harvesting, expansion, manipulation, purification, preservation and formulation, ultimately leading to successful therapy administration to patients at an acceptable cost. In this Review, we highlight case examples of cutting-edge bioprocessing technologies that improve biomanufacturing efficiency for cell therapies approaching clinical use.


Subject(s)
Biotechnology , Cell- and Tissue-Based Therapy , Immunotherapy , Humans
9.
Biol Blood Marrow Transplant ; 10(11): 784-93, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15505609

ABSTRACT

Hematopoietic stem cell chimerism can be established after low-dose conditioning regimens, although the risk of donor cell rejection increases for unrelated donor transplantations. We added pretransplantation rabbit antithymocyte globulin (6 mg/kg) to an established conditioning regimen of fludarabine (90 mg/m2) and single-fraction total body irradiation (200 cGy) followed by postgrafting immunosuppression with cyclosporine A and mycophenolate mofetil for 22 patients with hematologic malignancies. One patient rejected the graft and successfully underwent transplantation with cells from a second donor by using the same conditioning regimen. The actuarial probability of developing acute graft-versus-host disease grade II to IV before day 100 was 40%, although 9 of 14 patients who survived beyond 100 days developed chronic graft-versus-host disease. These data support a hypothesis that the addition of antithymocyte globulin decreases the risk of graft-versus-host and host-versus-graft reactions when combined with a nonmyeloablative conditioning regimen of fludarabine and total body irradiation.


Subject(s)
Graft vs Host Disease/prevention & control , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation , Transplantation Conditioning/methods , Vidarabine/analogs & derivatives , Adult , Aged , Antilymphocyte Serum/administration & dosage , Female , Graft Rejection/prevention & control , Humans , Immunosuppressive Agents/administration & dosage , Male , Middle Aged , Vidarabine/administration & dosage , Whole-Body Irradiation
10.
Blood ; 99(4): 1486-8, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11830505

ABSTRACT

Efforts to reduce relapse of non-Hodgkin lymphoma after autologous transplantation have included ex vivo stem cell selection and/or peritransplantation immunotherapy. The late infectious and immunologic consequences of these maneuvers are not well understood, although an increase in early cytomegaloviral disease after CD34(+) stem cell selection and an alteration in immunoglobulin and T-cell recovery after peritransplantation rituximab has been noted. We report the first 2 cases of progressive multifocal leukoencephalopathy caused by JC papovavirus after autologous peripheral blood stem cell transplantation and a case each of cytomegalovirus retinitis and pneumonitis. All 4 patients experienced significant impairment of CD4 T-cell recovery, placing them at risk for these unusual viral infections. The clustering of cases is concerning because all occurred shortly after the introduction of peritransplantation rituximab into treatment protocols (4 of 62 immunotherapy recipients compared with 0 of 276 without; z = 3.595; P <.001), although a direct association with this CD20 B-cell-directed therapy remains speculative.


Subject(s)
Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/adverse effects , Virus Diseases/chemically induced , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Murine-Derived , Antineoplastic Agents/administration & dosage , Cytomegalovirus Infections/chemically induced , Cytomegalovirus Infections/etiology , Female , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Leukoencephalopathy, Progressive Multifocal/chemically induced , Leukoencephalopathy, Progressive Multifocal/virology , Lymphoma/complications , Lymphoma/therapy , Male , Retrospective Studies , Rituximab , Transplantation, Autologous/adverse effects , Virus Diseases/etiology
11.
Biol Blood Marrow Transplant ; 8(10): 536-43, 2002.
Article in English | MEDLINE | ID: mdl-12434948

ABSTRACT

The incidence and clinical relevance of tumor cells contaminating the stem cell products of patients with advanced breast cancer treated with high-dose chemotherapy is uncertain because prior studies used small sample sizes and lacked standardization of the immunocytochemistry (ICC) detection method used. We evaluated blood stem cell and bone marrow samples obtained from 535 women with metastatic breast cancer who received high-dose chemotherapy and unmanipulated mobilized blood stem cell support. Of the patients tested, 20.6% and 26.3% had blood stem cell and bone marrow contamination, respectively. Blood stem cell contamination was significantly more frequent in patients with marrow involvement than in patients without marrow involvement (35% versus 18.4%, respectively; P = .009). In fact, according to multivariate analysis results, marrow involvement was the only significant predictor for blood stem cell product contamination. Patients without marrow involvement who had fewer apheresis procedures were also observed to have a significantly lower incidence rate of blood stem cell contamination than patients who had more procedures (P < or = .008), and patients who received combined chemotherapy and cytokine mobilization therapy had less contamination than patients who received cytokine alone (P = .0001). Combined mobilization therapy appears to be associated with a lower incidence of contamination as a result of fewer apheresis procedures rather than through an antitumor effect of chemotherapy (P < or = .001). Patients with ICC-negative blood stem cell products had significantly longer progression-free survival (PFS) and overall survival (OS) than did patients with ICC-positive blood stem cell products (median PFS, 401 versus 291 days, respectively, P = .007; median OS, 1060 versus 697 days, P = .009) . However, multivariate analysis did not reveal any significant independent predictors of survival outcomes. Thus, further study is needed to determine if contaminating tumor cells in the stem cell products of breast cancer patients ever directly impact survival outcomes or are only indicative of residual in vivo disease in high-dose chemotherapy recipients.


Subject(s)
Breast Neoplasms/pathology , Breast Neoplasms/therapy , Neoplastic Cells, Circulating/pathology , Peripheral Blood Stem Cell Transplantation/standards , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow Neoplasms/pathology , Bone Marrow Neoplasms/secondary , Breast Neoplasms/mortality , Female , Hematopoietic Stem Cell Mobilization/adverse effects , Humans , Middle Aged , Peripheral Blood Stem Cell Transplantation/mortality , Risk Factors , Survival Analysis , Transplantation, Autologous/mortality , Transplantation, Autologous/standards , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL