Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Laryngoscope ; 132(3): 523-527, 2022 03.
Article in English | MEDLINE | ID: mdl-33988246

ABSTRACT

OBJECTIVES/HYPOTHESIS: To evaluate the safety and potential efficacy of autologous muscle-derived cells (AMDCs) for the treatment of swallowing impairment following treatment for oropharynx cancer. STUDY DESIGN: Prospective, phase I, open label, clinical trial. METHODS: Oropharynx cancer survivors disease free ≥2 years post chemoradiation were recruited. All patients had swallowing impairment but were not feeding tube dependent (Functional Oral Intake Scale [FOIS] ≥ 5). Muscle tissue (50-250 mg) was harvested from the vastus lateralis and 150 × 106 AMDCs were prepared (Cook MyoSite Inc., Pittsburgh, PA). The cells were injected into four sites throughout the intrinsic tongue musculature. Participants were followed for 24 months. The primary outcome measure was safety. Secondary endpoints included objective measures on swallowing fluoroscopy, oral and pharyngeal pressure, and changes in patient-reported outcomes. RESULTS: Ten individuals were enrolled. 100% (10/10) were male. The mean age of the cohort was 65 (±8.87) years. No serious adverse event occurred. Mean tongue pressure increased significantly from 26.3 (±11.1) to 31.8 (±9.5) kPa (P = .017). The mean penetration-aspiration scale did not significantly change from 5.6 (±2.1) to 6.8 (±1.8), and the mean FOIS did not significantly change from 5.4 (±0.5) to 4.6 (±0.7). The incidence of pneumonia was 30% (3/10) and only 10% (1/10) experienced deterioration in swallowing function throughout 2 years of follow-up. The mean eating assessment tool (EAT-10) did not significantly change from 24.1 (±5.57) to 21.3 (±6.3) (P = .12). CONCLUSION: Results of this phase I clinical trial demonstrate that injection of 150 × 106 AMDCs into the tongue is safe and may improve tongue strength, which is durable at 2 years. A blinded placebo-controlled trial is warranted. LEVEL OF EVIDENCE: 3 Laryngoscope, 132:523-527, 2022.


Subject(s)
Cell Transplantation/methods , Deglutition Disorders/therapy , Head and Neck Neoplasms/complications , Muscle Cells/transplantation , Aged , Deglutition Disorders/etiology , Fluoroscopy/methods , Humans , Male , Manometry , Prospective Studies
2.
World J Urol ; 38(11): 2881-2889, 2020 Nov.
Article in English | MEDLINE | ID: mdl-31938840

ABSTRACT

PURPOSE: The primary objective was to evaluate the effect of autologous muscle-derived cell (AMDC) injections on the urethral sphincter morphometry compared to placebo injections. Secondary aims were to explore the reduction of stress incontinence episode frequency (IEF) and factors associated with the efficacy of AMDC. METHODS: This prospective randomized-controlled study compared the urethral sphincter volumes of participants who had received either an intra-sphincteric injection of 4 cc AMDC in injection media or 4 cc placebo solution, using a transperineal 3D/4D ultrasound at baseline and at 12 months. The reduction of stress IEF on 3-day bladder diary and potential predictors at baseline for response to AMDC were assessed. RESULTS: Fifty-eight participants were included in the study. Compared to baseline, the mean total and external sphincter volumes increased significantly in both groups (respectively, p = 0.001 and p < 0.001 in the AMDC group, p < 0.001 and p = 0.005 in the placebo group) at 12 months. Both groups showed a significant reduction of stress IEF compared to baseline (p = 0.03 and p ≤ 0.001 for AMDC and placebo groups, respectively). There were no between-group differences regarding total and external sphincter volumes and reduction of stress IEF. A longer urethral length (p ≤ 0.001) and a larger external sphincter volume (p ≤ 0.05) were significantly associated with lower stress IEF. CONCLUSION: Significant increases of sphincter volumes as well as reduction of stress IEF occurred among the AMDC and placebo injection groups with no between-group differences at 12 months. A longer urethral length and a larger external sphincter volume at baseline were identified as potential predictors of AMDC injection response.


Subject(s)
Muscle Cells/transplantation , Urethra/diagnostic imaging , Urinary Incontinence, Stress/therapy , Aged , Autografts , Double-Blind Method , Humans , Imaging, Three-Dimensional , Injections , Male , Middle Aged , Prospective Studies , Ultrasonography/methods , Urethra/anatomy & histology
3.
Int Urol Nephrol ; 50(12): 2153-2165, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30324580

ABSTRACT

PURPOSE: The purpose of the study was to assess safety and efficacy of autologous muscle derived cells for urinary sphincter repair (AMDC-USR) in female subjects with predominant stress urinary incontinence. METHODS: A randomized, double-blind, multicenter trial examined intra-sphincteric injection of 150 × 106 AMDC-USR versus placebo in female subjects with stress or stress predominant, mixed urinary incontinence. AMDC-USR products were generated from vastus lateralis needle biopsies. Subjects were randomized 2:1 to receive AMDC-USR or placebo and 1:1 to receive 1 or 2 treatments (6 months after the first). Primary outcome was composite of ≥ 50% reduction in stress incontinence episode frequency (IEF), 24-h or in-office pad weight tests at 12 months. Other outcome data included validated subject-recorded questionnaires. Subjects randomized to placebo could elect to receive open-label AMDC-USR treatment after 12 months. Subject follow-up was up to 2 years. RESULTS: AMDC-USR was safe and well-tolerated with no product-related serious adverse events or discontinuations due to adverse events. Interim analysis revealed an unexpectedly high placebo response rate (90%) using the composite primary outcome which prevented assessment of treatment effect as designed and thus enrollment was halted at 61% of planned subjects. Post hoc analyses suggested that more stringent endpoints lowered placebo response rates and revealed a possible treatment effect. CONCLUSIONS: Although the primary efficacy finding was inconclusive, these results inform future trial design of AMDC-USR to identify clinically meaningful efficacy endpoints based on IEF reduction, understanding of placebo response rate, and refinement of subject selection criteria to more appropriately align with AMDC-USR's proposed mechanism of action.


Subject(s)
Muscle Cells/transplantation , Urethra/surgery , Urinary Incontinence, Stress/therapy , Adult , Aged , Aged, 80 and over , Double-Blind Method , Dysuria/etiology , Female , Humans , Middle Aged , Nausea/etiology , Pain/etiology , Quadriceps Muscle/cytology , Severity of Illness Index , Surveys and Questionnaires , Transplantation, Autologous/adverse effects , Treatment Outcome , Urinary Tract Infections/etiology , Young Adult
4.
Stem Cell Res Ther ; 9(1): 239, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30241573

ABSTRACT

BACKGROUND: Cell therapy constitutes an attractive alternative to treat stress urinary incontinence. Although promising results have been demonstrated in this field, the procedure requires further optimization. The most commonly proposed cell types for intraurethral injections are muscle derived cells (MDCs) and mesenchymal stem/stromal cell (MSCs). The aim of this study was to evaluate the effects of MDC-MSC co-transplantation into the urethra. METHODS: Autologous transplantation of labeled MDCs, bone marrow MSCs or co-transplantation of MDC-MSC were performed in aged multiparous female goats (n = 6 in each group). The mean number of cells injected per animal was 29.6 × 106(± 4.3 × 106). PBS-injected animals constituted the control group (n = 5). Each animal underwent urethral pressure profile (UPP) measurements before and after the injection procedure. The maximal urethral closure pressure (MUCP) and functional area (FA) of UPPs were calculated. The urethras were collected at the 28th or the 84th day after transplantation. The marker fluorochrome (DID) was visualized and quantified using in vivo imaging system in whole explants. Myogenic differentiation of the graft was immunohistochemically evaluated. RESULTS: The grafted cells were identified in all urethras collected at day 28 regardless of injected cell type. At this time point the strongest DID-derived signal (normalized to the number of injected cells) was noted in the co-transplanted group. There was a distinct decline in signal intensity between day 28 and day 84 in all types of transplantation. Both MSCs and MDCs contributed to striated muscle formation if transplanted directly to the external urethral sphincter. In the MSC group those events were rare. If cells were injected into the submucosal region they remained undifferentiated usually packed in clearly distinguishable depots. The mean increase in MUCP after transplantation in comparison to the pre-transplantation state in the MDC, MSC and MDC-MSC groups was 12.3% (± 11.2%, not significant (ns)), 8.2% (± 9.6%, ns) and 24.1% (± 3.1%, p = 0.02), respectively. The mean increase in FA after transplantation in the MDC, MSC and MDC-MSC groups amounted to 17.8% (± 15.4%, ns), 15.2% (± 12.9%, ns) and 17.8% (± 2.5%, p = 0.04), respectively. CONCLUSIONS: The results suggest that MDC-MSC co-transplantation provides a greater chance of improvement in urethral closure than transplantation of each population alone.


Subject(s)
Mesenchymal Stem Cell Transplantation/methods , Muscle Cells/transplantation , Urinary Incontinence, Stress/therapy , Urinary Incontinence, Stress/veterinary , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Count , Cell Differentiation , Female , Goats , Graft Survival/physiology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Microscopy, Fluorescence , Muscle Cells/cytology , Muscle Cells/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Transplantation, Autologous , Treatment Outcome , Urethra/physiopathology , Urinary Incontinence, Stress/physiopathology
6.
Lipids Health Dis ; 16(1): 189, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28969646

ABSTRACT

BACKGROUND: Lipoprotein lipase (LPL) deficiency is an autosomal recessive genetic disorder characterized by extreme hypertriglyceridemia, with no cure presently available. The purpose of this study was to test the possibility of using cell therapy to alleviate LPL deficiency. METHODS: The LPL coding sequence was cloned into the MSCV retrovirus vector, after which MSCV-hLPL and MSCV (empty construct without LPL coding sequence) virion suspensions were made using the calcium chloride method. A muscle cell line (C2C12), kidney cell line (HEK293T) and pre-adipocyte cell line (3 T3-L1) were transfected with the virus in order to express recombinant LPL in vitro. Finally, each transfected cell line was injected subcutaneously into nude mice to identify the cell type which could secret recombinant LPL in vivo. Control cells were transfected with the MSCV empty vector. LPL activity was analyzed using a radioimmunoassay. RESULTS: After virus infection, the LPL activity at the cell surface of each cell type was significantly higher than in the control cells, which indicates that all three cell types can be used to generate functional LPL. The transfected cells were injected subcutaneously into nude mice, and the LPL activity of the nearby muscle tissue at the injection site in mice injected with 3 T3-L1 cells was more than 5 times higher at the injection sites than at non-injected control sites. The other two types of cells did not show this trend. CONCLUSION: The subcutaneous injection of adipocytes overexpressing LPL can improve the LPL activity of the adjacent tissue of nude mice. This is a ground-breaking preliminary study for the treatment of LPL deficiency, and lays a good foundation for using cell therapy to correct LPL deficiency.


Subject(s)
Adipocytes/transplantation , Cell- and Tissue-Based Therapy/methods , Hyperlipoproteinemia Type I/therapy , Hypertriglyceridemia/therapy , Lipoprotein Lipase/genetics , Muscle Cells/transplantation , Adipocytes/cytology , Adipocytes/metabolism , Adipocytes/virology , Animals , Cell Line , Disease Models, Animal , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Hyperlipoproteinemia Type I/genetics , Hyperlipoproteinemia Type I/metabolism , Hyperlipoproteinemia Type I/pathology , Hypertriglyceridemia/genetics , Hypertriglyceridemia/metabolism , Hypertriglyceridemia/pathology , Injections, Subcutaneous , Lipoprotein Lipase/metabolism , Mice , Mice, Nude , Muscle Cells/cytology , Muscle Cells/metabolism , Muscle Cells/virology , NIH 3T3 Cells , Retroviridae/genetics , Retroviridae/metabolism , Transfection , Triglycerides/metabolism
7.
Biomed Res Int ; 2017: 1972608, 2017.
Article in English | MEDLINE | ID: mdl-28758111

ABSTRACT

Muscle-derived stem cells (MDSCs) possess multipotent differentiation and self-renewal capacities; however, the effects and mechanism in neuron injury remain unclear. The aim of this study was to investigate the effects of MDSCs on neuron secondary injury, oxidative stress-induced apoptosis. An in vivo study showed the Basso, Beattie, and Bresnahan (BBB) score and number of neurons significantly increased after MDSCs' transplantation in spinal cord injury (SCI) rats. An in vitro study demonstrated that MDSCs attenuated neuron apoptosis, and the expression of antioxidants was upregulated as well as the ratio of Bcl-2 and Bax in the MNT (MDSCs cocultured with injured neurons) group compared with the NT (injured neurons) group. Both LC3II/LC3I and ß-catenin were enhanced in the MNT group, while XAV939 (a ß-catenin inhibitor) decreased the expression of nuclear erythroid-related factor 2 (Nrf2) and LC3II/LC3I. Moreover, MDSCs became NSE- (neuron-specific enolase-) positive neuron-like cells with brain-derived neurotrophic factor (BDNF) treatment. The correlation analysis indicated that there was a significant relation between the level of BDNF and neuron injury. These findings suggest that MDSCs may protect the spinal cord from injury by inhibiting apoptosis and replacing injured neurons, and the increased BDNF and ß-catenin could contribute to MDSCs' effects.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Multipotent Stem Cells/metabolism , Muscle Cells/transplantation , Neuroprotection , Spinal Cord Injuries/therapy , Stem Cell Transplantation , Animals , Male , Multipotent Stem Cells/pathology , Muscle Cells/metabolism , Muscle Cells/pathology , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology
8.
Stem Cell Reports ; 9(1): 12-22, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28528701

ABSTRACT

Recent reports have documented the differentiation of human pluripotent stem cells toward the skeletal myogenic lineage using transgene- and cell purification-free approaches. Although these protocols generate myocytes, they have not demonstrated scalability, safety, and in vivo engraftment, which are key aspects for their future clinical application. Here we recapitulate one prominent protocol, and show that it gives rise to a heterogeneous cell population containing myocytes and other cell types. Upon transplantation, the majority of human donor cells could not contribute to myofiber formation. As a proof-of-principle, we incorporated the inducible PAX7 lentiviral system into this protocol, which then enabled scalable expansion of a homogeneous population of skeletal myogenic progenitors capable of forming myofibers in vivo. Our findings demonstrate the methods for scalable expansion of PAX7+ myogenic progenitors and their purification are critical for practical application to cell replacement treatment of muscle degenerative diseases.


Subject(s)
Cell Culture Techniques/methods , Muscle Cells/cytology , Muscle Cells/transplantation , Muscle Development , PAX7 Transcription Factor/genetics , Pluripotent Stem Cells/cytology , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Cells, Cultured , Humans , Lentivirus/genetics , Male , Mice , Muscle Cells/metabolism , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/physiology , Pluripotent Stem Cells/metabolism , Regeneration , Transgenes
9.
J Biomed Mater Res A ; 104(12): 3107-3114, 2016 12.
Article in English | MEDLINE | ID: mdl-27506966

ABSTRACT

Scaffolds are three-dimensional structures used for tissue regeneration being the base in tissue engineering. These scaffolds are obtained from natural and/or synthetic polymers and they should satisfy some specific requirements such as biocompatibility, suitable mechanical, and microstructural properties to favor cellular adhesion and neovascularization. This work shows a preclinic study about the production of low and medium molecular weight alginate through the use of calcium salts (calcium glutamate). The results showed prove that better structures, distribution, and pore sizes as well as better mechanical properties correspond to medium molecular weight alginate and higher calcium salts concentration. This type of scaffold, after muscular cells cultivation, has been proved as an excellent material for muscle growth. The histopathological analysis shows a low inflammatory response, without a foreign body reaction, suitable neovascularization and good fibroblasts incorporation. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 3107-3114, 2016.


Subject(s)
Alginates/chemistry , Biocompatible Materials/chemistry , Muscle Cells/cytology , Tissue Scaffolds/chemistry , Animals , Calcium/chemistry , Cell Line , Compressive Strength , Female , Glucuronic Acid/chemistry , Glutamic Acid/chemistry , Hexuronic Acids/chemistry , Humans , Materials Testing , Muscle Cells/transplantation , Porosity , Rats , Rats, Wistar , Tensile Strength , Tissue Engineering
10.
Proteomics ; 16(14): 2028-42, 2016 07.
Article in English | MEDLINE | ID: mdl-27246553

ABSTRACT

Proteomic profiling plays a decisive role in the elucidation of molecular signatures representative of a specific clinical context. MuStem cell based therapy represents a promising approach for clinical applications to cure Duchenne muscular dystrophy (DMD). To expand our previous studies collected in the clinically relevant DMD animal model, we decided to investigate the skeletal muscle proteome 4 months after systemic delivery of allogenic MuStem cells. Quantitative proteomics with isotope-coded protein labeling was used to compile quantitative changes in the protein expression profiles of muscle in transplanted Golden Retriever muscular dystrophy (GRMD) dogs as compared to Golden Retriever muscular dystrophy dogs. A total of 492 proteins were quantified, including 25 that were overrepresented and 46 that were underrepresented after MuStem cell transplantation. Interestingly, this study demonstrates that somatic stem cell therapy impacts on the structural integrity of the muscle fascicle by acting on fibers and its connections with the extracellular matrix. We also show that cell infusion promotes protective mechanisms against oxidative stress and favors the initial phase of muscle repair. This study allows us to identify putative candidates for tissue markers that might be of great value in objectively exploring the clinical benefits resulting from our cell-based therapy for DMD. All MS data have been deposited in the ProteomeXchange with identifier PXD001768 (http://proteomecentral.proteomexchange.org/dataset/PXD001768).


Subject(s)
Cell- and Tissue-Based Therapy/methods , Muscle Cells/transplantation , Muscular Dystrophy, Animal/therapy , Proteome/genetics , Stem Cell Transplantation , Stem Cells/metabolism , Animals , Dogs , Gene Expression Profiling , Gene Expression Regulation , Gene Ontology , Internet , Molecular Sequence Annotation , Muscle Cells/cytology , Muscle Cells/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/pathology , Oxidative Stress , Proteome/metabolism , Proteomics/methods , Software , Stem Cells/cytology , Treatment Outcome
11.
BMC Musculoskelet Disord ; 17: 209, 2016 05 11.
Article in English | MEDLINE | ID: mdl-27170302

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy (DMD) is an X-linked muscle disease that leads to fibre necrosis and progressive paralysis. At present, DMD remains a lethal disease without any effective treatment, requiring a better understanding of the pathophysiological processes and comprehensive assessment of the newly identified therapeutic strategies. MicroRNAs including members of the muscle-specific myomiR family have been identified as being deregulated in muscle of DMD patients and in mdx mice used as a model for DMD. In recent years, the Golden Retriever muscular dystrophy (GRMD) dog has appeared as the crucial animal model for objectively assessing the potential of new innovative approaches. Here, we first aim at establishing the muscle expression pattern of five selected miRNAs in this clinically relevant model to determine if they are similarly affected compared with other DMD contexts. Second, we attempt to show whether these miRNAs could be impacted by the systemic delivery of a promising stem cell candidate (referred to as MuStem cells) to implement our knowledge on its mode of action and/or identify markers associated with cell therapy efficacy. METHODS: A comparative study of miRNAs expression levels and cellular localization was performed on 9-month-old healthy dogs, as well as on three sub-sets of GRMD dog (without immunosuppression or cell transplantation, with continuous immunosuppressive regimen and with MuStem cell transplantation under immunosuppression), using RT-qPCR and in situ hybridization. RESULTS: We find that miR-222 expression is markedly up-regulated in GRMD dog muscle compared to healthy dog, while miR-486 tends to be down-expressed. Intriguingly, the expression of miR-1, miR-133a and miR-206 does not change. In situ hybridization exploration reveals, for the first time, that miR-486 and miR-206 are mainly localized in newly regenerated fibres in GRMD dog muscle. In addition, we show that cyclosporine-based immunosuppression, classically used in allogeneic cell transplantation, exclusively impacts the miR-206 expression. Finally, we demonstrate that intra-arterial administration of MuStem cells results in up-regulation of miR-133a and miR-222 concomitantly with a down-expression of two sarcomeric proteins corresponding to miR-222 targets. CONCLUSION: We point out a differential muscle expression of miR-222 and miR-486 associated with the pathophysiology of the clinically relevant GRMD dog model with a tissue localization focused on regenerated fibres. We also establish a modified expression of miR-133a and miR-222 subsequent to MuStem cell infusion.


Subject(s)
MicroRNAs/metabolism , Muscle Cells/transplantation , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/therapy , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Stem Cell Transplantation/methods , Animals , Biomarkers/metabolism , Cyclosporine/pharmacology , Cyclosporine/therapeutic use , Disease Models, Animal , Dogs , Down-Regulation , Fluorescent Antibody Technique , Humans , Immunosuppression Therapy/methods , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , In Situ Hybridization , Injections, Intra-Arterial , Mice , Mice, Inbred mdx , MicroRNAs/drug effects , Muscle Cells/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/pathology , Myosin Heavy Chains/metabolism , Stem Cells/metabolism , Up-Regulation
12.
Muscle Nerve ; 54(4): 763-8, 2016 10.
Article in English | MEDLINE | ID: mdl-26998682

ABSTRACT

INTRODUCTION: Duchenne muscular dystrophy (DMD) is caused by loss of the structural protein, dystrophin, resulting in muscle fragility. Muscle stem cell (MuSC) transplantation is a potential therapy for DMD. It is unknown whether donor-derived muscle fibers are structurally innervated. METHODS: Green fluorescent protein (GFP)-expressing MuSCs were transplanted into the tibials anterior of adult dystrophic mdx/mTR mice. Three weeks later the neuromuscular junction was labeled by immunohistochemistry. RESULTS: The percent overlap between pre- and postsynaptic immunolabeling was greater in donor-derived GFP(+) myofibers, and fewer GFP(+) myofibers were identified as denervated compared with control GFP(-) fibers (P = 0.001 and 0.03). GFP(+) fibers also demonstrated acetylcholine receptor fragmentation and expanded endplate area, indicators of muscle reinnervation (P = 0.008 and 0.033). CONCLUSION: It is unclear whether GFP(+) fibers are a result of de novo synthesis or fusion with damaged endogenous fibers. Either way, donor-derived fibers demonstrate clear histological innervation. Muscle Nerve 54: 763-768, 2016.


Subject(s)
Muscle Cells/transplantation , Muscle, Skeletal/innervation , Muscular Dystrophy, Duchenne/physiopathology , Muscular Dystrophy, Duchenne/therapy , Stem Cell Transplantation/methods , Animals , Mice , Mice, Inbred mdx , Mice, Transgenic , Muscle Cells/chemistry , Muscle Fibers, Skeletal/chemistry , Muscle Fibers, Skeletal/transplantation , Muscle, Skeletal/chemistry , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Animal/therapy
15.
J Urol ; 192(2): 469-76, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24582537

ABSTRACT

PURPOSE: We assess the 12-month safety and potential efficacy of autologous muscle derived cells for urinary sphincter repair (Cook MyoSite Incorporated, Pittsburgh, Pennsylvania) in women with stress urinary incontinence. MATERIALS AND METHODS: Pooled data from 2 phase I/II studies with identical patient selection criteria and outcome measures were analyzed. Enrolled patients had stress urinary incontinence refractory to prior treatment and no symptom improvement during the last 6 months. Patients received intrasphincter injection of 10 (16), 50 (16), 100 (24) or 200×10(6) (24) autologous muscle derived cells for urinary sphincter repair, derived from biopsies of each patient's quadriceps femoris. The primary outcome measure was safety, determined by incidence and severity of adverse events. Potential efficacy was measured by changes in 3-day voiding diaries, 24-hour pad tests, and UDI-6 and IIQ-7 scores. RESULTS: A total of 80 patients underwent injection of autologous muscle derived cells for urinary sphincter repair, and 72 completed diaries and pad tests at 12-month followup. No adverse events attributed to autologous muscle derived cells for urinary sphincter repair were reported. Higher dose groups tended to have greater percentages of patients with at least a 50% reduction in stress leaks and pad weight at 12-month followup. All dose groups had statistically significant improvement in UDI-6 and IIQ-7 scores at 12-month followup compared to baseline. CONCLUSIONS: Autologous muscle derived cells for urinary sphincter repair at doses of 10, 50, 100 and 200×10(6) cells appears safe. Efficacy data suggest a potential dose response with a greater percentage of patients responsive to higher doses.


Subject(s)
Cell- and Tissue-Based Therapy , Muscle Cells/transplantation , Urinary Incontinence, Stress/surgery , Adult , Aged , Aged, 80 and over , Female , Humans , Middle Aged , Treatment Outcome
16.
Curr Protoc Stem Cell Biol ; Chapter 2: Unit 2C.4, 2013.
Article in English | MEDLINE | ID: mdl-23661246

ABSTRACT

Studies in mice showed tremendous promise for the eventual clinical utility of myoblast transplantation to treat human muscular dystrophies. Initial attempts to translate the murine studies to humans, however, were not successful, due in part to limited engraftability of expanded donor myoblasts. Conventionally, muscle cells have been cultured on collagen-coated tissue culture-treated polystyrene. However, this promotes lineage progression and differentiation of cells, which limits engraftment potential. This unit describes the isolation of canine muscle-derived cells, ex vivo expansion of cells on plates coated with a modified Notch ligand, and the xenotransplant method used to evaluate engraftment potential. Activation of Notch signaling in freshly isolated canine muscle-derived cells with Delta-1(ext)-IgG inhibits myogenic differentiation, and maintains cells earlier in myogenic lineage progression. Delta-1(ext)-IgG-expanded cells engraft into the regenerating muscle of NOD/SCID mice more effectively than control cells expanded on human IgG, as evidenced by a significant increase in the number of muscle fibers expressing canine dystrophin in recipient murine muscle. Therefore, this protocol provides the basis for further developing culture conditions for ex vivo expansion of donor muscle cells for transplant.


Subject(s)
Cell Culture Techniques/methods , Muscle Cells/cytology , Muscle Cells/transplantation , Animals , Cell Count , Cell Proliferation , Cell Separation , Cells, Cultured , Cryoultramicrotomy , Dogs , Humans , Immunoglobulin G/metabolism , Injections , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/transplantation , Mice , Mice, SCID
17.
BJU Int ; 111(7): 1105-16, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23470219

ABSTRACT

UNLABELLED: WHAT'S KNOWN ON THE SUBJECT? AND WHAT DOES THE STUDY ADD?: Cell therapy using muscle precursor cell (MPC) injections has shown promise for urinary incontinence due to intrinsic sphincter deficiency (ISD), but the cell-preparation process is complex and costly. Implantation of freshly isolated myofibres carrying MPCs, mainly satellite cells, was very efficient in repairing muscle damage in recent animal experiments. In a phase I clinical trial, we investigated whether periurethral myofibre implantation generated local myogenesis and improved continence in 10 patients (five men and five women) with ISD. We found that myofibre implantation increased intraurethral pressure and periurethral electromyographic activity in patients with ISD. There were no serious side-effects. OBJECTIVES: To assess the safety of periurethral myofibre implantation in patients with urinary incontinence due to intrinsic sphincter deficiency (ISD) To assess the resulting myogenic process and effects on urinary continence. PATIENTS AND METHODS: An open-label non-randomised phase I clinical trial was conducted in five men and five women with ISD (mean age, 62.5 years). A free muscle strip from the patient's gracilis muscle was implanted around the urethra as a means to deliver locally myofibres and muscle precursor cells (MPCs). Patients were assessed for collection formation and incomplete bladder emptying. The maximum urethral closure pressure (MUCP) and concomitant periurethral electromyographic (EMG) activity were recorded before surgery and 1 and 3 months after surgery. Continence was assessed using the 24-h pad test and self-completed questionnaires, for 12 months. RESULTS: There were no serious side-effects. Continence improved significantly during the 12-month follow-up in four of the five women, including two who recovered normal continence. In the women, MUCP increased two-fold and de novo EMG periurethral activity was recorded. In the men, MUCP and EMG recordings showed similar improvements but the effect on continence was moderate. The few patients enrolled could affect these results. CONCLUSIONS: This is the first report of a one-step procedure for transferring autologous MPCs via myofibre implantation in patients with ISD. EMG and urodynamic assessments showed improvement of periurethral muscle activity. Further work is needed to confirm and improve the therapeutic efficiency of this procedure.


Subject(s)
Muscle Cells/transplantation , Muscle, Smooth/transplantation , Urethra/physiopathology , Urinary Incontinence/surgery , Urologic Surgical Procedures/methods , Animals , Female , Humans , Injections , Male , Middle Aged , Patient Satisfaction , Recovery of Function , Surveys and Questionnaires , Treatment Outcome , Urinary Incontinence/physiopathology , Urodynamics
18.
Laryngoscope ; 122(11): 2482-96, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22965802

ABSTRACT

OBJECTIVES/HYPOTHESIS: To determine if the spontaneous reinnervation that characteristically ensues after recurrent laryngeal nerve (RLN) injury could be selectively promoted and directed to certain laryngeal muscles with the use of neurotrophic factor (NF)-secreting muscle stem cell (MSC) vectors while antagonistic reinnervation is inhibited with vincristine (VNC). STUDY DESIGN: Basic science investigation involving primary cell cultures, gene cloning/transfer, and animal experiments. METHODS: MSC survival assays were used to test multiple individual NFs in vitro. Motoneuron outgrowth assays assessed the trophic effects of identified NF on cranial nerve X (CNX)-derived motoneurons in vitro. Therapeutic NF was cloned into a lentiviral vector, and MSCs were transduced to secrete NF. Sixty rats underwent left RLN transection injury, and at 3 weeks received injections of either MSCs (n = 24), MSCs secreting NF (n = 24), or saline (n = 12) into the left thyroarytenoid muscle complex; half of the animals in the MSC groups simultaneously received left posterior cricoarytenoid injections of VNC, whereas half of the animals received saline. RESULTS: Ciliary neurotrophic factor (CNTF) had the greatest survival-promoting effect on MSCs in culture. The addition of CNTF (50 ng/mL) to CNX motoneuron cultures resulted in enhanced neurite outgrowth and branching. In the animal model, the injected MSCs fused with the denervated myofibers, immunohistochemistry demonstrated enhanced reinnervation based on motor endplate to nerve contact, and reverse transcriptase-polymerase chain reaction confirmed stable CNTF expression at longest follow-up (4 months) in the CNTF-secreting MSC treated groups. CONCLUSIONS: MSC therapy may have a future role in selectively promoting and directing laryngeal reinnervation after RLN injury.


Subject(s)
Muscle Cells/transplantation , Nerve Growth Factors/pharmacology , Nerve Regeneration/drug effects , Recurrent Laryngeal Nerve Injuries/therapy , Stem Cell Transplantation , Animals , Cell Survival , Cells, Cultured , Ciliary Neurotrophic Factor/pharmacology , Disease Models, Animal , Electromyography , Genetic Vectors , Immunohistochemistry , Laryngoscopy , Lentivirus , Plasmids , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Autologous , Vincristine/administration & dosage , Vincristine/pharmacology
19.
Stem Cells ; 30(10): 2212-20, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22865615

ABSTRACT

Transplantation of myogenic stem cells possesses great potential for long-term repair of dystrophic muscle. However, a single donor muscle biopsy is unlikely to provide enough cells to effectively transplant the muscle mass of a patient affected by muscular dystrophy. Expansion of cells ex vivo using traditional culture techniques significantly reduces engraftment potential. We hypothesized that activation of Notch signaling during ex vivo expansion would maintain donor cell engraftment potential. In this study, we expanded freshly isolated canine muscle-derived cells on tissue culture plates coated with Delta-1(ext) -IgG to activate Notch signaling or with human IgG as a control. A model of canine-to-murine xenotransplantation was used to quantitatively compare canine muscle cell engraftment and determine whether engrafted donor cells could function as satellite cells in vivo. We show that Delta-1(ext) -IgG inhibited differentiation of canine muscle-derived cells and increased the level of genes normally expressed in myogenic precursors. Moreover, cells expanded on Delta-1(ext) -IgG resulted in a significant increase in the number of donor-derived fibers, as compared to cells expanded on human IgG, reaching engraftment levels similar to freshly isolated cells. Importantly, cells expanded on Delta-1(ext) -IgG engrafted to the recipient satellite cell niche and contributed to further regeneration. A similar strategy of expanding human muscle-derived cells on Notch ligand might facilitate engraftment and muscle regeneration for patients affected with muscular dystrophy.


Subject(s)
Graft Survival , Muscle Cells/metabolism , Muscle, Skeletal/metabolism , Receptors, Notch/metabolism , Stem Cells/metabolism , Animals , Cell Communication , Cell Differentiation , Cell Proliferation , Cells, Cultured , Dogs , Humans , Immunoglobulin G/pharmacology , Mice , Mice, SCID , Muscle Cells/cytology , Muscle Cells/transplantation , Muscle, Skeletal/cytology , Regeneration , Signal Transduction , Species Specificity , Stem Cell Transplantation , Stem Cells/cytology , Transplantation, Heterologous
20.
Curr Opin Neurol ; 25(5): 597-603, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22918488

ABSTRACT

PURPOSE OF REVIEW: This review focuses on stem cell-based therapies to treat skeletal muscle disorders, with a special emphasis on muscular dystrophies. RECENT FINDINGS: We briefly review previous attempts at cell therapy by the use of donor myoblasts, explaining the likely reasons for the poor clinical results; we then describe the use of the same cells in current promising trials for localized treatments of different diseases of skeletal muscle. Moreover, we discuss important novel findings on muscle stem/progenitor cell biology and their promise for future clinical translation. Preclinical and clinical applications of novel myogenic stem/progenitor cells are also described. SUMMARY: We summarize several ongoing clinical trials for different muscle disorders and the advances in the understanding of the biology of the myogenic progenitors used in such trials. On the basis of the currently available information, a prediction of developments in the field is proposed.


Subject(s)
Muscular Diseases/therapy , Stem Cell Transplantation , Animals , Humans , Muscle Cells/transplantation , Myoblasts/transplantation , Pluripotent Stem Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...