Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Stem Cells ; 36(2): 265-277, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29086457

RESUMEN

The ability to differentiate human pluripotent stem cells (hPSCs) into cardiomyocytes (CMs) makes them an attractive source for repairing injured myocardium, disease modeling, and drug testing. Although current differentiation protocols yield hPSC-CMs to >90% efficiency, hPSC-CMs exhibit immature characteristics. With the goal of overcoming this limitation, we tested the effects of varying passive stretch on engineered heart muscle (EHM) structural and functional maturation, guided by computational modeling. Human embryonic stem cells (hESCs, H7 line) or human induced pluripotent stem cells (IMR-90 line) were differentiated to hPSC-derived cardiomyocytes (hPSC-CMs) in vitro using a small molecule based protocol. hPSC-CMs were characterized by troponin+ flow cytometry as well as electrophysiological measurements. Afterwards, 1.2 × 106 hPSC-CMs were mixed with 0.4 × 106 human fibroblasts (IMR-90 line) (3:1 ratio) and type-I collagen. The blend was cast into custom-made 12-mm long polydimethylsiloxane reservoirs to vary nominal passive stretch of EHMs to 5, 7, or 9 mm. EHM characteristics were monitored for up to 50 days, with EHMs having a passive stretch of 7 mm giving the most consistent formation. Based on our initial macroscopic observations of EHM formation, we created a computational model that predicts the stress distribution throughout EHMs, which is a function of cellular composition, cellular ratio, and geometry. Based on this predictive modeling, we show cell alignment by immunohistochemistry and coordinated calcium waves by calcium imaging. Furthermore, coordinated calcium waves and mechanical contractions were apparent throughout entire EHMs. The stiffness and active forces of hPSC-derived EHMs are comparable with rat neonatal cardiomyocyte-derived EHMs. Three-dimensional EHMs display increased expression of mature cardiomyocyte genes including sarcomeric protein troponin-T, calcium and potassium ion channels, ß-adrenergic receptors, and t-tubule protein caveolin-3. Passive stretch affects the structural and functional maturation of EHMs. Based on our predictive computational modeling, we show how to optimize cell alignment and calcium dynamics within EHMs. These findings provide a basis for the rational design of EHMs, which enables future scale-up productions for clinical use in cardiovascular tissue engineering. Stem Cells 2018;36:265-277.


Asunto(s)
Biología Computacional/métodos , Miocardio/citología , Línea Celular , Citometría de Flujo , Humanos , Miocardio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Ingeniería de Tejidos/métodos
2.
Circulation ; 135(19): 1832-1847, 2017 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-28167635

RESUMEN

BACKGROUND: Advancing structural and functional maturation of stem cell-derived cardiomyocytes remains a key challenge for applications in disease modeling, drug screening, and heart repair. Here, we sought to advance cardiomyocyte maturation in engineered human myocardium (EHM) toward an adult phenotype under defined conditions. METHODS: We systematically investigated cell composition, matrix, and media conditions to generate EHM from embryonic and induced pluripotent stem cell-derived cardiomyocytes and fibroblasts with organotypic functionality under serum-free conditions. We used morphological, functional, and transcriptome analyses to benchmark maturation of EHM. RESULTS: EHM demonstrated important structural and functional properties of postnatal myocardium, including: (1) rod-shaped cardiomyocytes with M bands assembled as a functional syncytium; (2) systolic twitch forces at a similar level as observed in bona fide postnatal myocardium; (3) a positive force-frequency response; (4) inotropic responses to ß-adrenergic stimulation mediated via canonical ß1- and ß2-adrenoceptor signaling pathways; and (5) evidence for advanced molecular maturation by transcriptome profiling. EHM responded to chronic catecholamine toxicity with contractile dysfunction, cardiomyocyte hypertrophy, cardiomyocyte death, and N-terminal pro B-type natriuretic peptide release; all are classical hallmarks of heart failure. In addition, we demonstrate the scalability of EHM according to anticipated clinical demands for cardiac repair. CONCLUSIONS: We provide proof-of-concept for a universally applicable technology for the engineering of macroscale human myocardium for disease modeling and heart repair from embryonic and induced pluripotent stem cell-derived cardiomyocytes under defined, serum-free conditions.


Asunto(s)
Células Madre Embrionarias/trasplante , Insuficiencia Cardíaca/terapia , Células Madre Pluripotentes Inducidas/trasplante , Miocitos Cardíacos/trasplante , Ingeniería de Tejidos/métodos , Remodelación Ventricular/fisiología , Animales , Diferenciación Celular/fisiología , Células Madre Embrionarias/fisiología , Insuficiencia Cardíaca/patología , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Miocardio/citología , Miocardio/patología , Miocitos Cardíacos/fisiología , Impresión Tridimensional , Ratas , Ratas Desnudas
3.
Circ Res ; 117(8): 720-30, 2015 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-26291556

RESUMEN

RATIONALE: Tissue engineering approaches may improve survival and functional benefits from human embryonic stem cell-derived cardiomyocyte transplantation, thereby potentially preventing dilative remodeling and progression to heart failure. OBJECTIVE: Assessment of transport stability, long-term survival, structural organization, functional benefits, and teratoma risk of engineered heart muscle (EHM) in a chronic myocardial infarction model. METHODS AND RESULTS: We constructed EHMs from human embryonic stem cell-derived cardiomyocytes and released them for transatlantic shipping following predefined quality control criteria. Two days of shipment did not lead to adverse effects on cell viability or contractile performance of EHMs (n=3, P=0.83, P=0.87). One month after ischemia/reperfusion injury, EHMs were implanted onto immunocompromised rat hearts to simulate chronic ischemia. Bioluminescence imaging showed stable engraftment with no significant cell loss between week 2 and 12 (n=6, P=0.67), preserving ≤25% of the transplanted cells. Despite high engraftment rates and attenuated disease progression (change in ejection fraction for EHMs, -6.7±1.4% versus control, -10.9±1.5%; n>12; P=0.05), we observed no difference between EHMs containing viable and nonviable human cardiomyocytes in this chronic xenotransplantation model (n>12; P=0.41). Grafted cardiomyocytes showed enhanced sarcomere alignment and increased connexin 43 expression at 220 days after transplantation. No teratomas or tumors were found in any of the animals (n=14) used for long-term monitoring. CONCLUSIONS: EHM transplantation led to high engraftment rates, long-term survival, and progressive maturation of human cardiomyocytes. However, cell engraftment was not correlated with functional improvements in this chronic myocardial infarction model. Most importantly, the safety of this approach was demonstrated by the lack of tumor or teratoma formation.


Asunto(s)
Células Madre Embrionarias/trasplante , Supervivencia de Injerto , Trasplante de Corazón/métodos , Infarto del Miocardio/cirugía , Miocitos Cardíacos/trasplante , Músculos Papilares/trasplante , Ingeniería de Tejidos/métodos , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Línea Celular , Supervivencia Celular , Conexina 43/metabolismo , Modelos Animales de Enfermedad , Células Madre Embrionarias/inmunología , Células Madre Embrionarias/metabolismo , Trasplante de Corazón/efectos adversos , Xenoinjertos , Humanos , Inmunosupresores/farmacología , Masculino , Contracción Miocárdica , Infarto del Miocardio/inmunología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Músculos Papilares/inmunología , Músculos Papilares/metabolismo , Músculos Papilares/patología , Músculos Papilares/fisiopatología , Ratas Desnudas , Ratas Sprague-Dawley , Volumen Sistólico , Factores de Tiempo , Transfección
4.
Proc Natl Acad Sci U S A ; 109(31): 12544-9, 2012 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-22802633

RESUMEN

The stability of human embryonic stem cells (hESCs) is of critical importance for both experimental and clinical applications. We find that as an initial response to altered culture conditions, hESCs change their transcription profile for hundreds of genes and their DNA methylation profiles for several genes outside the core pluripotency network. After adaption to conditions of feeder-free defined and/or xeno-free culture systems, expression and DNA methylation profiles are quite stable for additional passaging. However, upon reversion to the original feeder-based culture conditions, numerous transcription changes are not reversible. Similarly, although the majority of DNA methylation changes are reversible, highlighting the plasticity of DNA methylation, a few are persistent. Collectively, this indicates these cells harbor a memory of culture history. For culture-induced DNA methylation changes, we also note an intriguing correlation: hypomethylation of regions 500-2440 bp upstream of promoters correlates with decreased expression, opposite to that commonly seen at promoter-proximal regions. Lastly, changes in regulation of G-coupled protein receptor pathways provide a partial explanation for many of the unique transcriptional changes observed during hESC adaptation and reverse adaptation.


Asunto(s)
Metilación de ADN/fisiología , Células Madre Embrionarias/metabolismo , Epigénesis Genética/fisiología , Regiones Promotoras Genéticas/fisiología , Transcripción Genética/fisiología , Línea Celular , Células Madre Embrionarias/citología , Células Nutrientes/citología , Células Nutrientes/metabolismo , Humanos
5.
Cytotherapy ; 15(8): 999-1010, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23664011

RESUMEN

BACKGROUND AIMS: We have previously described a xeno-free scalable system to generate transplantable dopaminergic neurons from human pluripotent stem cells. However, several important questions remain to be answered about our cell therapy efforts. These include determining the exact time at which cells should be transplanted and whether cells at this stage can be frozen, shipped, thawed and injected without compromising their ability to mature and survive the transplantation procedure. We also needed to determine whether further optimization of the culture process could shorten the development time and reduce variability and whether a current Good Manufacture Practice (CGMP) facility could manufacture cells with fidelity. METHODS: We developed an optimized protocol that included modulating the sonic hedgehog homolog gradient with bone morphogenetic proteins (BMP2) and addition of activin to the culture medium, which shortened the time to generate Lmx1A and FoxA2 immunoreactive cells by 4-6 days. RESULTS: We showed that cells at this stage could be safely frozen and thawed while retaining an excellent ability to continue to mature in vitro and survive transplant in vivo. Importantly, we successfully adapted this process to a CGMP facility and manufactured two lots of transplant-ready dopaminergic neurons (>250 vials) under CGMP-compatible conditions. In vitro characterization, including viability/recovery on thawing, whole genome expression as well as expression of midbrain/dopaminergic markers, showed that the cells manufactured under GMP-compatible conditions were similar to cells produced at lab scale. CONCLUSIONS: Our results suggest that this optimized protocol can be used to generate dopaminergic neurons for Investigational New Drug enabling studies.


Asunto(s)
Técnicas de Cultivo de Célula , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/trasplante , Células Madre Pluripotentes Inducidas/metabolismo , Neurogénesis , Células Madre Pluripotentes/citología , Activinas , Animales , Proteína Morfogenética Ósea 2/metabolismo , Proliferación Celular , Supervivencia Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Células Cultivadas , Criopreservación/métodos , Dopamina/análisis , Dopamina/biosíntesis , Descubrimiento de Drogas/métodos , Proteínas Hedgehog/metabolismo , Factor Nuclear 3-beta del Hepatocito/inmunología , Humanos , Proteínas con Homeodominio LIM/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción/inmunología
6.
Mol Ther ; 19(3): 557-66, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21179010

RESUMEN

Lentiviral vectors are now in clinical trials for a variety of inherited and acquired disorders. A challenge for moving any viral vector into the clinic is the ability to screen the vector product for the presence of replication-competent virus. Assay development for replication-competent lentivirus (RCL) is particularly challenging because recombination of vector packaging plasmids and cellular DNA leading to RCL has not been reported with the current viral vector systems. Therefore, the genomic structure of a RCL remains theoretical. In this report, we describe a highly sensitive RCL assay suitable for screening vector product and have screened large-scale vector supernatant, cells used in vector production, and cells transduced with clinical grade vector. We discuss the limitations and challenges of the current assay, and suggest modifications that may improve the suitability of this assay for screening US Food and Drug Administration (US FDA)-licensed products.


Asunto(s)
Vectores Genéticos/aislamiento & purificación , Vectores Genéticos/normas , Lentivirus/aislamiento & purificación , Replicación Viral , Línea Celular , Ensayo de Inmunoadsorción Enzimática/métodos , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Células HEK293 , Proteína p24 del Núcleo del VIH/inmunología , Humanos , Lentivirus/genética , Control de Calidad , Recombinación Genética , Estándares de Referencia , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
8.
EBioMedicine ; 4: 74-85, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26981572

RESUMEN

The directed differentiation of human cardiomyocytes (CMs) from pluripotent cells provides an invaluable model for understanding mechanisms of cell fate determination and offers considerable promise in cardiac regenerative medicine. Here, we utilize a human embryonic stem cell suspension bank, produced according to a good manufacturing practice, to generate CMs using a fully defined and small molecule-based differentiation strategy. Primitive and cardiac mesoderm purification was used to remove non-committing and multi-lineage populations and this significantly aided the identification of key transcription factors, lncRNAs, and essential signaling pathways that define cardiomyogenesis. Global methylation profiles reflect CM development and we report on CM exon DNA methylation "memories" persisting beyond transcription repression and marking the expression history of numerous developmentally regulated genes, especially transcription factors.


Asunto(s)
Metilación de ADN , Células Madre Embrionarias/citología , Epigénesis Genética , Exones , Miocitos Cardíacos/citología , Células Madre Pluripotentes/citología , Transcriptoma , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Células Madre Embrionarias/metabolismo , Humanos , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes/metabolismo
9.
Methods Mol Biol ; 1283: 13-21, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25537838

RESUMEN

Suspension cell culture systems with superior scalability, controllability, and monitoring options are an attractive alternative to static adherent culture methods for expansion and production of human pluripotent stem cells (hPSCs), including embryonic stem cells (ESCs) and induced pluripotent cells (iPSCs). In this chapter, we describe a scalable suspension culture system using serum-free, feeder-free, matrix-free, and defined culture conditions with spinner flasks for hPSC maintenance and expansion. This suspension culture system provides an efficient and GMP-compatible process for large-scale manufacture of hPSCs.


Asunto(s)
Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Células Madre Pluripotentes/citología , Diferenciación Celular , Criopreservación/métodos , Células Madre Embrionarias/citología , Humanos , Células Madre Pluripotentes Inducidas/citología
10.
Stem Cells Transl Med ; 4(10): 1097-100, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26285658

RESUMEN

UNLABELLED: Several human embryonic stem cell (hESC)-derived cell therapeutics have entered clinical testing and more are in various stages of preclinical development. The U.S. Food and Drug Administration (FDA) regulates these products under existing regulations and has stated that these products do not constitute a new class of biologic. However, as human tissue, hESCs are subject to regulations that were developed before hESCs were first described. The regulations have not been revised since 2005, well before the first hESC-derived product entered clinical studies. The current regulations require donors of hESCs to be tested in the same manner as donors of tissues intended for transplantation. However, because hESC-derived cell products are more than minimally manipulated, they are also subject to the same end-of-production release testing as most other biologic agents. In effect, this makes hESC products subject to redundant testing. No other biologic is subject to a similar testing requirement. Furthermore, the regulations that require donor testing are specifically applicable to hESC cells harvested from donors after a date in 2005. It is unclear which regulations cover hESCs harvested before 2005. Ambiguity in the guidelines and redundant testing requirements have unintentionally created a burdensome regulatory paradigm for these products and reluctance on the part of developers to invest in these promising therapeutics. We propose a simple solution that would address FDA safety concerns, eliminate regulatory uncertainty and risk, and provide flexibility for the FDA in the regulation of hESC-derived cell therapies. SIGNIFICANCE: Regulatory ambiguity concerning donor eligibility screening and testing requirements for human embryonic stem cell lines, in particular those lines created before 2005, are causing significant concern for drug developers. Technically, most of these lines fail to meet eligibility under U.S. Food and Drug Administration (FDA) rules for product licensure, and many developers are unaware that FDA approval to begin trials under an exemption is not an assurance that the FDA will grant licensure of the product. This Perspective outlines the ambiguity and the problem it has caused and proposes a workable solution. The intent is to generate stakeholder and FDA discussion on this issue.


Asunto(s)
Guías como Asunto , Células Madre Embrionarias Humanas , Investigación con Células Madre/legislación & jurisprudencia , Donantes de Tejidos/legislación & jurisprudencia , United States Food and Drug Administration/legislación & jurisprudencia , Productos Biológicos/aislamiento & purificación , Selección de Donante/legislación & jurisprudencia , Selección de Donante/normas , Adhesión a Directriz , Humanos , Seguridad del Paciente , Estados Unidos , United States Food and Drug Administration/normas
11.
J Clin Invest ; 125(7): 2551-7, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26132109

RESUMEN

Human pluripotent stem cells are known to have the capacity to renew indefinitely, being intrinsically able to differentiate into many different cell types. These characteristics have generated tremendous enthusiasm about the potential applications of these cells in regenerative medicine. However, major challenges remain with the development and testing of novel experimental stem cell therapeutics in the field. In this Review, we focus on the nature of the preclinical challenges and discuss potential solutions that could help overcome them. Furthermore, we discuss the use of allogeneic versus autologous stem cell products, including a review of their respective advantages and disadvantages, major clinical requirements, quality standards, time lines, and costs of clinical grade development.


Asunto(s)
Células Madre Pluripotentes Inducidas/trasplante , Animales , Bancos de Muestras Biológicas/normas , Diferenciación Celular , Prueba de Histocompatibilidad , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/inmunología , Medicina Regenerativa/tendencias , Seguridad , Trasplante de Células Madre/legislación & jurisprudencia , Trasplante de Células Madre/normas , Trasplante de Células Madre/tendencias , Investigación Biomédica Traslacional/tendencias , Trasplante Autólogo , Trasplante Homólogo , Estados Unidos , United States Food and Drug Administration
12.
Stem Cell Res ; 15(2): 365-75, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26318718

RESUMEN

To meet the need of a large quantity of hPSC-derived cardiomyocytes (CM) for pre-clinical and clinical studies, a robust and scalable differentiation system for CM production is essential. With a human pluripotent stem cells (hPSC) aggregate suspension culture system we established previously, we developed a matrix-free, scalable, and GMP-compliant process for directing hPSC differentiation to CM in suspension culture by modulating Wnt pathways with small molecules. By optimizing critical process parameters including: cell aggregate size, small molecule concentrations, induction timing, and agitation rate, we were able to consistently differentiate hPSCs to >90% CM purity with an average yield of 1.5 to 2×10(9) CM/L at scales up to 1L spinner flasks. CM generated from the suspension culture displayed typical genetic, morphological, and electrophysiological cardiac cell characteristics. This suspension culture system allows seamless transition from hPSC expansion to CM differentiation in a continuous suspension culture. It not only provides a cost and labor effective scalable process for large scale CM production, but also provides a bioreactor prototype for automation of cell manufacturing, which will accelerate the advance of hPSC research towards therapeutic applications.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes/citología , Actinina/metabolismo , Técnicas de Cultivo de Célula , Línea Celular , Regulación de la Expresión Génica , Humanos , Microscopía Fluorescente , Miocitos Cardíacos/citología , Células Madre Pluripotentes/metabolismo , ARN/química , ARN/aislamiento & purificación , Análisis de Secuencia de ARN , Troponina I/metabolismo , Troponina T/metabolismo , Vía de Señalización Wnt
13.
Mol Ther Methods Clin Dev ; 1: 14015, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-26015959

RESUMEN

Broader implementation of cell-based therapies has been hindered by the logistics associated with the expansion of clinically relevant cell numbers ex vivo. To overcome this limitation, Wilson Wolf Manufacturing developed the G-Rex, a cell culture flask with a gas-permeable membrane at the base that supports large media volumes without compromising gas exchange. Although this culture platform has recently gained traction with the scientific community due to its superior performance when compared with traditional culture systems, the limits of this technology have yet to be explored. In this study, we investigated multiple variables including optimal seeding density and media volume, as well as maximum cell output per unit of surface area. Additionally, we have identified a novel means of estimating culture growth kinetics. All of these parameters were subsequently integrated into a novel G-Rex "M" series, which can accommodate these optimal conditions. A multicenter study confirmed that this fully optimized cell culture system can reliably produce a 100-fold cell expansion in only 10 days using 1L of medium. The G-Rex M series is linearly scalable and adaptable as a closed system, allowing an easy translation of preclinical protocols into the good manufacturing practice.

14.
Stem Cell Res Ther ; 4(2): 25, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23672848

RESUMEN

Parkinson's disease (PD) is a common debilitating neurodegenerative disease. The motor symptoms of PD are caused mainly by a progressive loss of dopaminergic neurons from the substania nigra, resulting in a loss of dopamine production. Current therapies are palliative and, in the long term, ineffective. In addition, some can result in significant clinical side effects. The relatively localized pathology of PD makes it an ideal candidate for cell replacement therapy. Initial efforts focused on fetal cell transplantation, and significant clinical benefit lasting more than 10 years has been reported in some cases. However, the approach is controversial and results have been inconsistent. Inherent limitations of this approach for widespread use are the limited availability and variability of transplant material. In contrast, the self-renewal and differentiation potential of human pluripotent stem cells (hPSCs) make them a promising alternative cell source for cell replacement therapy for PD. Efforts in the past decade have demonstrated that hPSCs can be induced to differentiate in culture to functional dopaminergic neurons. Studies in delivering these cells into PD animal models have demonstrated survival, engraftment, and behavioral deficit improvements. Several groups are developing these cells with clinical trials in mind. Here, we review the state of the technology and consider the suitability of current manufacturing processes, cell purity, and tumorgenicity for clinical testing.


Asunto(s)
Enfermedad de Parkinson/terapia , Células Madre Pluripotentes/trasplante , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/trasplante , Enfermedad de Parkinson/patología , Células Madre Pluripotentes/citología
15.
Sci Transl Med ; 5(184): 184ra59, 2013 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-23658244

RESUMEN

High-grade gliomas are extremely difficult to treat because they are invasive and therefore not curable by surgical resection; the toxicity of current chemo- and radiation therapies limits the doses that can be used. Neural stem cells (NSCs) have inherent tumor-tropic properties that enable their use as delivery vehicles to target enzyme/prodrug therapy selectively to tumors. We used a cytosine deaminase (CD)-expressing clonal human NSC line, HB1.F3.CD, to home to gliomas in mice and locally convert the prodrug 5-fluorocytosine to the active chemotherapeutic 5-fluorouracil. In vitro studies confirmed that the NSCs have normal karyotype, tumor tropism, and CD expression, and are genetically and functionally stable. In vivo biodistribution studies demonstrated NSC retention of tumor tropism, even in mice pretreated with radiation or dexamethasone to mimic clinically relevant adjuvant therapies. We evaluated safety and toxicity after intracerebral administration of the NSCs in non-tumor-bearing and orthotopic glioma-bearing immunocompetent and immunodeficient mice. We detected no difference in toxicity associated with conversion of 5-fluorocytosine to 5-fluorouracil, no NSCs outside the brain, and no histological evidence of pathology or tumorigenesis attributable to the NSCs. The average tumor volume in mice that received HB1.F3.CD NSCs and 5-fluorocytosine was about one-third that of the average volume in control mice. On the basis of these results, we conclude that combination therapy with HB1.F3.CD NSCs and 5-fluorocytosine is safe, nontoxic, and effective in mice. These data have led to approval of a first-in-human study of an allogeneic NSC-mediated enzyme/prodrug-targeted cancer therapy in patients with recurrent high-grade glioma.


Asunto(s)
Glioma/tratamiento farmacológico , Glioma/terapia , Células-Madre Neurales/citología , Profármacos/uso terapéutico , Animales , Línea Celular , Citosina Desaminasa/metabolismo , Femenino , Citometría de Flujo , Flucitosina/metabolismo , Flucitosina/uso terapéutico , Fluorouracilo/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Células-Madre Neurales/metabolismo , Profármacos/metabolismo
16.
Stem Cell Res ; 8(3): 388-402, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22459095

RESUMEN

Suspension bioreactors are an attractive alternative to static culture of human embryonic stem cells (hESCs) for the generation of clinically relevant cell numbers in a controlled system. In this study, we have developed a scalable suspension culture system using serum-free defined media with spinner flasks for hESC expansion as cell aggregates. With optimized cell seeding density and splitting interval, we demonstrate prolonged passaging and expansion of several hESC lines with overall expansion, yield, viability and maintenance of pluripotency equivalent to adherent culture. Human ESCs maintained in suspension as aggregates can be passaged at least 20 times to achieve over 1×10(13) fold calculated expansion with high undifferentiation rate and normal karyotype. Furthermore, the aggregates are able to differentiate to cardiomyocytes in a directed fashion. Finally, we show that the cells can be cryopreserved in serum-free medium and thawed into adherent or suspension cultures to continue passaging and expansion. We have successfully used this method under cGMP or cGMP-equivalent conditions to generate cell banks of several hESC lines. Taken together, our suspension culture system provides a powerful approach for scale-up expansion of hESCs under defined and serum-free conditions for clinical and research applications.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Técnicas de Cultivo de Célula/normas , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Reactores Biológicos/normas , Diferenciación Celular , Células Cultivadas , Criopreservación , Medio de Cultivo Libre de Suero , Humanos , Cariotipificación , Miocitos Cardíacos/citología
17.
Methods Mol Biol ; 767: 147-59, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21822873

RESUMEN

Human pluripotent stem cells (PSCs), which include human embryonic stem cells (ESCs) as well as induced pluripotent stem cells (iPSCs), represent an important source of cellular therapies in regenerative medicine and the study of early human development. As such, it is becoming increasingly important to develop methods for the large-scale banking of human PSC lines. There are several well-established methods for the propagation of human PSCs. The key to development of a good manufacturing practice (GMP) bank is to determine a manufacturing method that is amenable to large-scale production using materials that are fully documented. We have developed several banks of hESCs using animal feeder cells, animal-based matrices, or animal-free matrices. Protocols for growing hESCs on mouse embryonic fibroblasts (MEFs) are well established and are very helpful for producing research grade banks of cells. As most human ESCs cultured by research laboratories have been exposed to xenogeneic reagents, it is not imperative that all materials used in the production of a master cell bank be animal-free in origin. Nevertheless, as the field develops, it will no doubt become increasingly important to produce a bank of cells for clinical use without xenogeneic reagents, particularly nonhuman feeder cells which might harbor viruses with potential risk to human health or cell product integrity. Thus, even for cell lines previously exposed to xenogeneic reagents, it is important to minimize any subsequent exposure of the cell lines to additional adventitious agents. We have specifically described procedures for the growth of hESCs on Matrigel, an animal-matrix, and CELLstart, an animal-free matrix, and these can be used to produce hESCs as part of a clinical manufacturing process.


Asunto(s)
Bancos de Muestras Biológicas , Técnicas de Cultivo de Célula/métodos , Técnicas de Cultivo de Célula/normas , Células Madre Pluripotentes/citología , Trasplante de Células Madre/métodos , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Colágeno/farmacología , Criopreservación , Combinación de Medicamentos , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Humanos , Laminina/farmacología , Ratones , Células Madre Pluripotentes/efectos de los fármacos , Proteoglicanos/farmacología
18.
Regen Med ; 5(4): 569-79, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20632860

RESUMEN

Induced pluripotent stem (iPS) cells offer tremendous opportunity for the creation of autologous cellular therapies, in which gene correction or the avoidance of immune response issues are desirable. In addition, iPS cells avoid the ethical concerns raised by the sourcing of human embryonic stem cells (hESCs) from embryos. iPS cells share many characteristics with hESCs and it is anticipated that existing experience with hESCs will translate to rapid progress in moving iPS cell-derived products toward clinical trials. While the potential clinical value for these products is considerable, the nature of current manufacturing paradigms for autologous iPS cell products raises considerable regulatory concerns. Here, the regulatory challenges posed by autologous iPS cell-derived products are examined. We conclude that there will be considerable regulatory concerns primarily relating to reproducibility of the manufacturing process and safety testing within clinically limited time constraints. Demonstrating safety of the final cell product in an autologous setting will be the single greatest obstacle to progressing autologous iPS cell-based therapies into the clinic.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Política de Salud , Células Madre Pluripotentes Inducidas/trasplante , Ingeniería de Tejidos , United States Food and Drug Administration/legislación & jurisprudencia , Tratamiento Basado en Trasplante de Células y Tejidos/ética , Tratamiento Basado en Trasplante de Células y Tejidos/normas , Tratamiento Basado en Trasplante de Células y Tejidos/tendencias , Industria Farmacéutica/legislación & jurisprudencia , Guías como Asunto , Humanos , Estados Unidos
19.
J Clin Oncol ; 27(8): 1235-42, 2009 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-19204206

RESUMEN

PURPOSE: Squamous cell carcinoma of the head and neck (SCCHN) is characterized by upregulation of the epidermal growth factor receptor (EGFR). We developed a novel strategy to target EGFR by using a therapeutic gene that consisted of an EGFR antisense (AS) gene sequence under U6 promoter control. A phase I clinical trial was conducted to evaluate the safety and biologic effects of EGFR AS. PATIENTS AND METHODS: Patients with advanced SCCHN who were refractory to standard therapies and who had at least one assessable and accessible lesion were enrolled. The EGFR AS dose was escalated in successive cohorts (six dose levels; 60 to 1,920 microg/injection). Patients received four weekly intratumoral EGFR AS injections. Tumor biopsies were performed before and after completion of therapy. Treatment response was assessed by tumor volume measurements (positron emission tomography/computed tomography), and levels of target proteins were assessed by immunohistochemistry. RESULTS: Seventeen assessable patients were treated. No grades 3 to 4 or dose-limiting toxicities were noted, and a maximum-tolerated dose was not reached. Five patients (29%) achieved a clinical response, which included two complete responses (CRs) and three partial responses (PRs); two additional patients had stable disease (SD) as the best response. Patients with disease control (CR + PR + SD) had tumors with higher EGFR and lower STAT3 expression at baseline compared with patients who had progressive disease (P = .0312 and P = .095, respectively). CONCLUSION: Intratumoral EGFR AS was safe and resulted in antitumor activity in patients with advanced SCCHN. Baseline levels of high EGFR and low STAT3 may be associated with antitumor effects.


Asunto(s)
Carcinoma de Células Escamosas/terapia , ADN sin Sentido/uso terapéutico , Receptores ErbB/antagonistas & inhibidores , Neoplasias de Cabeza y Cuello/terapia , Anciano , Carcinoma de Células Escamosas/química , Receptores ErbB/análisis , Receptores ErbB/genética , Femenino , Fluorodesoxiglucosa F18 , Terapia Genética , Neoplasias de Cabeza y Cuello/química , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Tomografía de Emisión de Positrones , Proteínas Proto-Oncogénicas c-akt/análisis , Factor de Transcripción STAT3/análisis
20.
Vaccine ; 24(3): 293-303, 2006 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-16115700

RESUMEN

Prostatic acid phosphatase (PAP) is a prostate tumor antigen currently being investigated as a target antigen in several human vaccine trials, some with evidence of clinical benefit. We have previously demonstrated that plasmid DNA vaccines encoding either human or rat PAP can elicit antigen-specific cellular and humoral immunity in rat models. The current study was performed to determine the safety and potential immunological efficacy in rodents of large and repetitive doses of a GMP-grade plasmid DNA vaccine encoding human PAP, pTVG-HP. Fifty-four male Lewis rats were immunized intradermally at 2-week intervals with 100, 500, or 1,500 microg pTVG-HP with 5 microg recombinant rat GM-CSF protein given as a vaccine adjuvant. An additional 12 male Lewis rats served as controls with groups immunized with 1,500 microg of a parental DNA vector not encoding human PAP, and a group that received GM-CSF protein only without plasmid DNA. Groups of animals (n=3-6) were euthanized after two, four, or six immunizations with collections of tissues and blood for toxicity assessment and immunological analysis. No significant toxicities were observed in terms of animal weights, histopathology, hematological changes, or changes in serum chemistries. Six of fifty-four were found to have subtle evidence of possible renal toxicity, however these findings were not statistically different from control animals. The vaccine was found to be effective in eliciting PAP-specific CD4 and CD8 T cells, predominantly Th1 in type, in all immunized animals at all doses and numbers of immunizations. PAP-specific IgG were detected in a dose-dependent fashion, with titers increasing after multiple immunizations. These studies demonstrate that, in rats, immunization with the pTVG-HP vaccine is safe and effective in eliciting PAP-specific cellular and humoral immune responses. These findings support the further clinical evaluation of pTVG-HP in patients with prostate cancer.


Asunto(s)
Fosfatasa Ácida/inmunología , Vacunas contra el Cáncer/inmunología , Próstata/enzimología , Próstata/inmunología , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/prevención & control , Animales , Formación de Anticuerpos/inmunología , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/toxicidad , Ensayo de Inmunoadsorción Enzimática , Inmunidad Celular/inmunología , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/inmunología , Masculino , Plásmidos/inmunología , Ratas , Ratas Endogámicas Lew , Bazo/inmunología , Linfocitos T/inmunología , Vacunas de ADN/efectos adversos , Vacunas de ADN/inmunología , Vacunas de ADN/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA