12 research outputs found

    From Bench to Bedside: Review of Gene and Cell-Based Therapies and the Slow Advancement into Phase 3 Clinical Trials, with a Focus on Aastrom’s Ixmyelocel-T

    Get PDF
    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

    Culturing Keratinocytes and Fibroblasts in a Three-Dimensional Mesh Results in Epidermal Differentiation and Formation of a Basal Lamina-Anchoring Zone

    Get PDF
    The purpose of this study was to characterize an in vitro co-culture model in which fibroblasts grown in a three-dimensional nylon mesh were recombined with human keratinocytes. The cultures were kept for 3 and 5 weeks and then processed for electron microscopy. Keratinocytes showed reconstruction of an epidermis consisting of a basal layer with hemidesmosome's, a stratified epithelium with tonofilament's and desmosomes, a granular layer with keratinosomes and keratohyaline granules, and a transitional stratum corneum. Anchoring filaments, lamina densa, anchoring fibrils, bundles of elastin-associated microfibrils (diameters 10 nm) and fine collagen fibrils were formed. Collagen fibrils near the epidermis were much thinner than those in the lower levels.The present study shows that the dermal model containing metabolically active fibroblasts in their natural environment will support epidermal morphogenesis and differentiation including the formation of a basal lamina and anchoring zone

    Cellular Therapy With Ixmyelocel-T to Treat Critical Limb Ischemia: The Randomized, Double-blind, Placebo-controlled RESTORE-CLI Trial

    Get PDF
    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

    Interim analysis results from the RESTORE-CLI, a randomized, double-blind multicenter phase II trial comparing expanded autologous bone marrow-derived tissue repair cells and placebo in patients with critical limb ischemia

    Get PDF
    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).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 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.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, = .0053) and amputation-free survival in patients receiving TRC treatment, (log-rank test, = .038). Major amputation occurred in 19% of TRC-treated patients compared to 43% of controls ( = .14, Fisher exact test). There was evidence of improved wound healing in the TRC-treated patients when compared with controls at 12 months.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

    Characterization, Barrier Function, and Drug Metabolism of an In Vitro Skin Model

    Get PDF
    We have characterized an in vitro skin model consisting of neonatal keratinocytes and fibroblasts grown on a nylon mesh. To produce a dermal model, fibroblasts were seeded onto nylon mesh and grown for 4 weeks until a physiologic dermal-like matrix was formed. This matrix was found to consist of collagens I and III, fibronectin, and glycosaminoglycans. Keratinocytes were then seeded onto the dermal model and the co-culture was grown at the air/liquid interface. A differentiated epidermis with distinct basal, spinous, granular, and stratum corneum layers was formed. When incubated in the presence of keratinocytes, fibronectin immunofluorescence increased throughout the dermis compared to cultures incubated similarly in the absence of keratinocytes. A basement membrane zone rich in laminin, collagen IV, and heparan sulfate proteoglycan was detected. The epidermis, isolated from the co-culture by thermolysin digestion, was analyzed for differentiation markers. K1 keratin (67-kDa) and involucrin were detected by immunologic techniques. Ceramide lipids (types III and IV), thought to be important in barrier function, were detected by thin-layer chromatography. The permeability of the co-culture to a panel of compounds, including [3H]-water, was determined using Franz and side-by-side diffusion cells. The permeability coefficient for water was of the same order of magnitude as that determined for neonatal foreskin. The co-culture also showed selective permeability to a panel of compounds of differing lipid solubility. This co-culture metabolized [3H]-testosterone to a profile of metabolites similar to that of neonatal foreskin. We believe that this in vitro skin model will be useful for the study of drug permeability and metabolism

    Angiogenic and Osteogenic Potential of Bone Repair Cells for Craniofacial Regeneration

    Full text link
    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.Peer Reviewedhttp://deepblue.lib.umich.edu/bitstream/2027.42/85090/1/ten_tea_2010_0079.pd
    corecore