Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Circ Cardiovasc Interv ; 15(2): e010802, 2022 02.
Article in English | MEDLINE | ID: mdl-35067072

ABSTRACT

BACKGROUND: Coronary microvascular dysfunction results in angina and adverse outcomes in patients with evidence of ischemia and nonobstructive coronary artery disease; however, no specific therapy exists. CD34+ cell therapy increases microvasculature in preclinical models and improves symptoms, exercise tolerance, and mortality in refractory angina patients with obstructive coronary artery disease. The objective of this research was to evaluate the safety, tolerability, and efficacy of intracoronary CD34+ cell therapy in patients with coronary microvascular dysfunction. METHODS: We conducted a 2-center, 20-participant trial of autologous CD34+ cell therapy (protocol CLBS16-P01; NCT03508609) in patients with ischemia and nonobstructive coronary artery disease with persistent angina and coronary flow reserve ≤2.5. Efficacy measures included coronary flow reserve, angina frequency, Canadian Cardiovascular Society angina class, Seattle Angina Questionnaire, SF-36, and modified Bruce exercise treadmill test obtained at baseline and 6 months after treatment. Autologous CD34+ cells (CLBS16) were mobilized by administration of granulocyte-colony stimulating factor 5µg/kg/day for 5 days and collected by leukapheresis. Participants received a single intracoronary left anterior descending infusion of isolated CD34+ cells in medium that enhances cell function. RESULTS: Coronary flow reserve improved from 2.08±0.32 at baseline to 2.68±0.79 at 6 months after treatment (P<0.005). Angina frequency decreased (P<0.004), Canadian Cardiovascular Society class improved (P<0.001), and quality of life improved as assessed by the Seattle Angina Questionnaire (P≤0.03, all scales) and SF-36 (P≤0.04, all scales). There were no cell-related serious adverse events. CONCLUSIONS: In this pilot clinical trial of microvascular angina, patients with ischemia and nonobstructive coronary artery disease receiving intracoronary infusion of CD34+ cell therapy had higher coronary flow reserve, less severe angina, and better quality of life at 6 months. The current study supports a potential therapeutic role for CD34+ cells in patients with microvascular angina. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT03508609.


Subject(s)
Coronary Artery Disease , Microvascular Angina , Myocardial Ischemia , Antigens, CD34 , Canada , Cell- and Tissue-Based Therapy , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/therapy , Humans , Ischemia , Quality of Life , Treatment Outcome
2.
Stem Cell Res Ther ; 6: 25, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25889271

ABSTRACT

INTRODUCTION: Bone marrow derived cellular therapies are an emerging approach to promoting therapeutic angiogenesis in ischemic cardiovascular disease. However, the percentage of regenerative cells in bone marrow mononuclear cells (BMMNCs) is small, and large amounts of BMMNCs are required. Ixmyelocel-T, an expanded autologous multicellular therapy, is manufactured from a small sample of bone marrow aspirate. Ixmyelocel-T contains expanded populations of mesenchymal stromal cells (MSCs) and M2-like macrophages, as well as many of the CD45+ cells found in the bone marrow. It is hypothesized that this expanded multi-cellular therapy would induce angiogenesis and endothelial repair. METHODS: A rat model of hind limb ischemia was used to determine the effects of ixmyelocel-T on blood flow recovery. To further determine the effects on endothelial cells, ixmyelocel-T was co-cultured with human umbilical vein endothelial cells (HUVEC) in non-contacting Transwell® inserts. RESULTS: Co-culture of HUVECs with ixmyelocel-T resulted secretion of a variety of pro-angiogenic factors. HUVECs stimulated by ixmyelocel-T exhibited enhanced migration, proliferation, and branch formation. Ixmyelocel-T co-culture also resulted in increased endothelial nitric oxide synthase (eNOS) expression and nitric oxide (NO) production. In tumor necrosis factor alpha (TNFα)-stimulated HUVECs, ixmyelocel-T co-culture decreased apoptosis and reactive oxygen species generation, increased super oxide dismutase activity, and decreased nuclear factor kappa B (NFκB) activation. Treatment with ixmyelocel-T in a rat model of hind limb ischemia resulted in significantly increased blood flow perfusion and capillary density, gene expression and plasma levels of the anti-inflammatory cytokine interleukin (IL)-10, plasma nitrates, plasma platelet-derived growth factor (PDGF)-BB, vascular endothelial growth factor (VEGF) expression, and significantly decreased plasma thiobarbituric acid reactive substances (TBARS). CONCLUSIONS: This work demonstrates that ixmyelocel-T interacts with endothelial cells in a paracrine manner, resulting in angiogenesis and endothelial protection. This data suggests that ixmyelocel-T could be useful for promoting of angiogenesis and tissue repair in ischemic cardiovascular diseases. In conclusion, ixmyelocel-T therapy may provide a new aspect of therapeutic angiogenesis in this patient population where expanded populations of regenerative cells might be required.


Subject(s)
Bone Marrow Transplantation , Cell- and Tissue-Based Therapy/methods , Hindlimb/blood supply , Mesenchymal Stem Cell Transplantation , Neovascularization, Physiologic/physiology , Animals , Apoptosis/drug effects , Becaplermin , Bone Marrow Cells/physiology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Enzyme Activation/drug effects , Hindlimb/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukin-10/blood , Ischemia/pathology , Ischemia/therapy , Leukocyte Common Antigens/metabolism , Macrophages/transplantation , Mesenchymal Stem Cells/physiology , NF-kappa B/metabolism , Nitrates/blood , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/biosynthesis , Paracrine Communication/physiology , Proto-Oncogene Proteins c-sis/biosynthesis , Proto-Oncogene Proteins c-sis/blood , Rats , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/blood , Wound Healing/drug effects , Wound Healing/physiology
3.
Stem Cell Rev Rep ; 9(3): 373-83, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23456574

ABSTRACT

There is a large body of preclinical research demonstrating the efficacy of gene and cellular therapy for the potential treatment of severe (limb-threatening) peripheral arterial disease (PAD), including evidence for growth and transcription factors, monocytes, and mesenchymal stem cells. While preclinical research has advanced into early phase clinical trials in patients, few late-phase clinical trials have been conducted. The reasons for the slow progression of these therapies from bench to bedside are as complicated as the fields of gene and cellular therapies. The variety of tissue sources of stem cells (embryonic, adult bone marrow, umbilical cord, placenta, adipose tissue, etc.); autologous versus allogeneic donation; types of cells (hematopoietic, mesenchymal stromal, progenitor, and mixed populations); confusion and stigmatism by the public and patients regarding gene, protein, and stem cell therapy; scaling of manufacturing; and the changing regulatory environment all contribute to the small number of late phase (Phase 3) clinical trials and the lack of Food and Drug Administration (FDA) approvals. This review article provides an overview of the progression of research from gene therapy to the cellular therapy field as it applies to peripheral arterial disease, as well as the position of Aastrom's cellular therapy, ixmyelocel-T, within this field.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Genetic Therapy/methods , Peripheral Arterial Disease/therapy , Stem Cell Transplantation , Extremities/physiopathology , Humans , Neovascularization, Physiologic , Stem Cells
4.
Cell Transplant ; 22(5): 767-77, 2013.
Article in English | MEDLINE | ID: mdl-22776413

ABSTRACT

Stem cell therapy offers potential in the regeneration of craniofacial bone defects; however, it has not been studied clinically. Tissue repair cells (TRCs) isolated from bone marrow represent a mixed stem and progenitor population enriched in CD90- and CD14-positive cells. In this phase I/II, randomized, controlled feasibility trial, we investigated TRC cell therapy to reconstruct localized craniofacial bone defects. Twenty-four patients requiring localized reconstruction of jawbone defects participated in this longitudinal trial. For regenerative therapy, patients were randomized to receive either guided bone regeneration (GBR) or TRC transplantation. At 6 or 12 weeks following treatment, clinical and radiographic assessments of bone repair were performed. Bone biopsies were harvested and underwent quantitative micro-computed tomographic (µCT) and bone histomorphometric analyses. Oral implants were installed, subsequently restored, and functionally loaded with tooth restorations. Reconstructed sites were assessed for 1 year following therapy. No study-related, serious adverse events were reported. Following therapy, clinical, radiographic, tomographic, and histological measures demonstrated that TRC therapy accelerated alveolar bone regeneration compared to GBR therapy. Additionally, TRC treatment significantly reduced the need for secondary bone grafting at the time of oral implant placement with a five fold decrease in implant bony dehiscence exposure (residual bone defects) as compared to GBR-treated sites(p < 0.01). Transplantation of TRCs for treatment of alveolar bone defects appears safe and accelerates bone regeneration, enabling jawbone reconstruction with oral implants. The results from this trial support expanded studies of TRC therapy in the treatment of craniofacial deformities (ClinicalTrials.gov number CT00755911).


Subject(s)
Bone Regeneration , Mesenchymal Stem Cells/cytology , Stem Cell Transplantation , Adult , Aged , Bone Density , Bone Marrow Cells/cytology , Female , Guided Tissue Regeneration, Periodontal , Histocompatibility , Humans , Jaw/diagnostic imaging , Jaw/pathology , Lipopolysaccharide Receptors/metabolism , Longitudinal Studies , Male , Mesenchymal Stem Cells/metabolism , Middle Aged , Thy-1 Antigens/metabolism , Tomography, X-Ray Computed
5.
Stem Cell Res Ther ; 4(6): 134, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24405629

ABSTRACT

INTRODUCTION: M2 macrophages promote tissue repair and regeneration through various mechanisms including immunomodulation and scavenging of tissue debris. Delivering increased numbers of these cells to ischemic tissues may limit tissue injury and promote repair. Ixmyelocel-T is an expanded, autologous multicellular therapy cultured from bone-marrow mononuclear cells (BMMNCs). The purpose of this study was to characterize further a unique expanded population of M2-like macrophages, generated in ixmyelocel-T therapy. METHODS: Approximately 50 ml of whole bone marrow was obtained from healthy donors and shipped overnight. BMMNCs were produced by using density-gradient separation and cultured for approximately 12 days to generate ixmyelocel-T. CD14+ cells were isolated from ixmyelocel-T with positive selection for analysis. Cell-surface phenotype was examined with flow cytometry and immunofluorescence, and expression of cytokines and chemokines was analyzed with enzyme-linked immunosorbent assay (ELISA). Quantitative real-time PCR was used to analyze expression of genes in BMMNCs, ixmyelocel-T, the CD14+ population from ixmyelocel-T, and M1 and M2 macrophages. Ixmyelocel-T was cultured with apoptotic BMMNCs, and then visualized under fluorescence microscopy to assess efferocytosis. RESULTS: Macrophages in ixmyelocel-T therapy expressed surface markers of M2 macrophages, CD206, and CD163. These cells were also found to express several M2 markers, and few to no M1 markers. After stimulation with lipopolysaccharide (LPS), they showed minimal secretion of the proinflammatory cytokines interleukin-12 (IL-12) and tumor necrosis factor alpha (TNF-α) compared with M1 and M2 macrophages. Ixmyelocel-T macrophages efficiently ingested apoptotic BMMNCs. CONCLUSIONS: Ixmyelocel-T therapy contains a unique population of M2-like macrophages that are characterized by expression of M2 markers, decreased secretion of proinflammatory cytokines after inflammatory stimuli, and efficient removal of apoptotic cells. This subpopulation of cells may have a potential role in tissue repair and regeneration.


Subject(s)
Bone Marrow Cells/cytology , Macrophages/cytology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Cells, Cultured , Humans , Interleukin-12/metabolism , Lectins, C-Type/metabolism , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/toxicity , Macrophages/drug effects , Macrophages/metabolism , Mannose Receptor , Mannose-Binding Lectins/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/metabolism , Tumor Necrosis Factor-alpha/metabolism , c-Mer Tyrosine Kinase
6.
Stem Cell Res Ther ; 4(6): 135, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24405662

ABSTRACT

INTRODUCTION: Advanced atherosclerotic lesions are characterized by lipid accumulation, inflammation, and defective efferocytosis. An ideal therapy should address all aspects of this multifactorial disease. Ixmyelocel-T therapy, an expanded autologous multicellular therapy showing clinical promise in the treatment of diseases associated with advanced atherosclerosis, includes a novel population of M2-like macrophages. Here, we examine the macrophages of ixmyelocel-T and determine their ability to influx modified cholesterol in an atheroprotective manner, maintaining cholesterol homeostasis and preventing cellular dysfunction and death, ultimately promoting reverse cholesterol efflux. METHODS: Approximately 50 ml of whole bone marrow was obtained from healthy donors and shipped overnight. Bone marrow mononuclear cells (BMMNCs) were produced by using density gradient separation and cultured for approximately 12 days to generate ixmyelocel-T. CD14+ cells were isolated from ixmyelocel-T via positive selection for analysis. Ixmyelocel-T and human leukemia monocyte (THP-1) cells were loaded with acetylated low-density lipoprotein (Ac-LDL) for analysis. Flow cytometry and immunofluorescence were used to examine Ac-LDL uptake, expression of cytokines was analyzed by enzyme-linked immunofluorescence assay (ELISA), and quantitative real-time PCR was used to analyze expression of cholesterol-transport genes. Both the in vitro cholesterol efflux assay and in vivo reverse cholesterol transport assay were used to examine cholesterol transport. RESULTS: Ixmyelocel-T macrophages take up acetylated low-density lipoprotein and express the scavenger receptors CD36 and scavenger receptor-B1 (SR-B1). Ixmyelocel-T did not become apoptotic or proinflammatory after lipid loading. The cholesterol transporter genes ABAC1 and ABCG1 were both statistically significantly upregulated when ixmyelocel-T macrophages were loaded with cholesterol. Ixmyelocel-T also exhibited enhanced apolipoprotein A-I (ApoAI)-mediated cholesterol efflux. In addition, in vivo reverse cholesterol-transport assay demonstrated that ixmyelocel-T was able to efflux cholesterol in this model. CONCLUSIONS: Ixmyelocel-T macrophages influx modified cholesterol, remained anti-inflammatory in the face of lipid loading and inflammatory challenge, and displayed enhanced cholesterol efflux capabilities. These combined features suggest that this autologous multicellular therapy may exert beneficial effects in atherosclerotic diseases.


Subject(s)
Macrophages/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/metabolism , Apolipoprotein A-I/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/surgery , Bone Marrow Cells/cytology , CD36 Antigens/metabolism , Cells, Cultured , Cholesterol/pharmacology , Cytokines/metabolism , Humans , Lipoproteins, LDL/toxicity , Macrophages/drug effects , Macrophages/transplantation , Scavenger Receptors, Class B/metabolism
7.
Stem Cell Res Ther ; 3(4): 26, 2012 Jul 09.
Article in English | MEDLINE | ID: mdl-22776246

ABSTRACT

Aastrom Biosciences has developed a proprietary cell-processing technology that enables the manufacture of ixmyelocel-T, a patient-specific multicellular therapy expanded from a small sample of a patient's own bone marrow. Ixmyelocel-T is produced under current good manufacturing practices (cGMP) in a fully closed, automated system that expands mesenchymal stem cells (MSCs) and macrophages. While the cell types in ixmyelocel-T are the same as those found in the bone marrow, the numbers of MSCs and alternative macrophages are greater in ixmyelocel-T. We propose that the mixture of expanded MSCs and alternatively activated macrophages promote long-term tissue repair of ischemic tissue. The multiple cell types in ixmyelocel-T have a range of biological activities that are likely to contribute to a complex mechanism of action. Clinical trial data collected to date support the potential for ixmyelocel-T as an efficacious and safe treatment for ischemic cardiovascular indications, including critical limb ischemia (CLI) and a severe form of heart failure, dilated cardiomyopathy (DCM). The CLI clinical program has completed phase 2 and has reached concurrence with the Food and Drug Administration (FDA) on a phase 3 study (REVIVE) through the Special Protocol Assessment (SPA) process. The phase 3 study began screening patients in February 2012. The DCM clinical program will initiate phase 2b in 2012.


Subject(s)
Drug Industry , Antigens, CD/metabolism , Cardiovascular Diseases/mortality , Cardiovascular Diseases/therapy , Clinical Trials as Topic , Humans , Kaplan-Meier Estimate , Macrophages/cytology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Phenotype
8.
Mol Ther ; 20(6): 1280-6, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22453769

ABSTRACT

Ixmyelocel-T is a patient-specific, expanded, multicellular therapy evaluated in patients with lower extremity critical limb ischemia (CLI) with no options for revascularization. This randomized, double-blind, placebo-controlled, phase 2 trial (RESTORE-CLI) compared the efficacy and safety of intramuscular injections of ixmyelocel-T with placebo. Patients received one-time injections over 20 locations in a single leg and were followed for 12 months. Safety assessments included occurrence of adverse events. Efficacy assessments included time to first occurrence of treatment failure (TTF; major amputation of injected leg; all-cause mortality; doubling of total wound surface area from baseline; de novo gangrene) and amputation-free survival (AFS; major amputation of injected leg; all-cause mortality). A total of 77 patients underwent bone marrow or sham aspiration; 72 patients received ixmyelocel-T (48 patients) or placebo (24 patients). Adverse event rates were similar. Ixmyelocel-T treatment led to a significantly prolonged TTF (P = 0.0032, logrank test). AFS had a clinically meaningful 32% reduction in event rate that was not statistically significant (P = 0.3880, logrank test). Treatment effect in post hoc analyses of patients with baseline wounds was more pronounced (TTF: P < 0.0001, AFS: P = 0.0802, logrank test). Ixmyelocel-T treatment was well tolerated and may offer a potential new treatment option.


Subject(s)
Ischemia/therapy , Lower Extremity/blood supply , Macrophages/transplantation , Mesenchymal Stem Cell Transplantation , Aged , Aged, 80 and over , Amputation, Surgical , Cell Transplantation/adverse effects , Female , Humans , Ischemia/mortality , Ischemia/surgery , Male , Middle Aged , Survival Analysis , Treatment Failure , Treatment Outcome
9.
J Vasc Surg ; 54(4): 1032-41, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21684715

ABSTRACT

OBJECTIVES: Cell therapy is a novel experimental treatment modality for patients with critical limb ischemia (CLI) of the lower extremities and no other established treatment options. This study was conducted to assess the safety and clinical efficacy of intramuscular injection of autologous tissue repair cells (TRCs). METHODS: A prospective, randomized double-blinded, placebo controlled, multicenter study (RESTORE-CLI) was conducted at 18 centers in the United States in patients with CLI and no option for revascularization. Enrollment of 86 patients began in April 2007 and ended in February 2010. For the prospectively planned interim analysis, conducted in February 2010, 33 patients had the opportunity to complete the trial (12 months of follow-up), and 46 patients had completed at least 6 months of follow-up. The interim analysis included analysis of both patient populations. An independent physician performed the bone marrow or sham control aspiration. The aspirate was processed in a closed, automated cell manufacturing system for approximately 12 days to generate the TRC population of stem and progenitor cells. An average of 136 ± 41 × 10(6) total viable cells or electrolyte (control) solution were injected into 20 sites in the ischemic lower extremity. The primary end point was safety as evaluated by adverse events, and serious adverse events as assessed at multiple follow-up time points. Clinical efficacy end points included major amputation-free survival and time to first occurrence of treatment failure (defined as any of the following: major amputation, death, de novo gangrene, or doubling of wound size), as well as major amputation rate and measures of wound healing. RESULTS: There was no difference in adverse or serious adverse events between the two groups. Statistical analysis revealed a significant increase in time to treatment failure (log-rank test, P = .0053) and amputation-free survival in patients receiving TRC treatment, (log-rank test, P = .038). Major amputation occurred in 19% of TRC-treated patients compared to 43% of controls (P = .14, Fisher exact test). There was evidence of improved wound healing in the TRC-treated patients when compared with controls at 12 months. CONCLUSIONS: Intramuscular injection of autologous bone marrow-derived TRCs is safe and decreases the occurrence of clinical events associated with disease progression when compared to placebo in patients with lower extremity CLI and no revascularization options.


Subject(s)
Bone Marrow Transplantation , Ischemia/surgery , Lower Extremity/blood supply , Stem Cell Transplantation , Aged , Aged, 80 and over , Amputation, Surgical , Bone Marrow Transplantation/adverse effects , Bone Marrow Transplantation/mortality , Cells, Cultured , Double-Blind Method , Female , Humans , Injections, Intramuscular , Ischemia/mortality , Ischemia/pathology , Ischemia/physiopathology , Kaplan-Meier Estimate , Limb Salvage , Male , Middle Aged , Prospective Studies , Reoperation , Risk Assessment , Risk Factors , Stem Cell Transplantation/adverse effects , Stem Cell Transplantation/mortality , Time Factors , Transplantation, Autologous , Treatment Outcome , United States , Wound Healing
10.
Tissue Eng Part A ; 16(9): 2809-20, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20412009

ABSTRACT

There has been increased interest in the therapeutic potential of bone marrow derived cells for tissue engineering applications. Bone repair cells (BRCs) represent a unique cell population generated via an ex vivo, closed-system, automated cell expansion process, to drive the propagation of highly osteogenic and angiogenic cells for bone engineering applications. The aims of this study were (1) to evaluate the in vitro osteogenic and angiogenic potential of BRCs, and (2) to evaluate the bone and vascular regenerative potential of BRCs in a craniofacial clinical application. BRCs were produced from bone marrow aspirates and their phenotypes and multipotent potential characterized. Flow cytometry demonstrated that BRCs were enriched for mesenchymal and vascular phenotypes. Alkaline phosphatase and von Kossa staining were performed to assess osteogenic differentiation, and reverse transcriptase-polymerase chain reaction was used to determine the expression levels of bone specific factors. Angiogenic differentiation was determined through in vitro formation of tube-like structures and fluorescent labeling of endothelial cells. Finally, 6 weeks after BRC transplantation into a human jawbone defect, a biopsy of the regenerated site revealed highly vascularized, mineralized bone tissue formation. Taken together, these data provide evidence for the multilineage and clinical potential of BRCs for craniofacial regeneration.


Subject(s)
Bone Marrow Cells/cytology , Facial Bones/surgery , Neovascularization, Physiologic/physiology , Osteogenesis/physiology , Tissue Engineering/methods , Alkaline Phosphatase/metabolism , Bone Marrow Cells/metabolism , Cell Differentiation/physiology , Cells, Cultured , Facial Bones/metabolism , Facial Bones/pathology , Flow Cytometry , Humans , Jaw/pathology , Orthognathic Surgical Procedures , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...