Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Stem Cell Reports ; 10(5): 1429-1431, 2018 May 08.
Article in English | MEDLINE | ID: mdl-29742388

ABSTRACT

Stem cell-based clinical interventions are increasingly advancing through preclinical testing and approaching clinical trials. The complexity and diversity of these approaches, and the confusion created by unproven and untested stem cell-based "therapies," create a growing need for a more comprehensive review of these early-stage human trials to ensure they place the patients at minimal risk of adverse events but are also based on solid evidence of preclinical efficacy with a clear scientific rationale for that effect. To address this issue and supplement the independent review process, especially that of the ethics and institutional review boards who may not be experts in stem cell biology, the International Society for Stem Cell Research (ISSCR) has developed a set of practical questions to cover the major issues for which clear evidence-based answers need to be obtained before approving a stem cell-based trial.


Subject(s)
Clinical Trials as Topic/ethics , Ethics Committees, Research , Stem Cell Transplantation/ethics , Stem Cells/cytology , Humans , Surveys and Questionnaires , Translational Research, Biomedical
2.
Prog Brain Res ; 230: 151-163, 2017.
Article in English | MEDLINE | ID: mdl-28552227

ABSTRACT

The development of pluripotent stem cell (PSC) therapies is rapidly advancing, and a number of PSC-derived cell products are currently being tested in clinical trials. The biological complexity of these therapies results in specific challenges in complying with regulatory guidelines. This includes the choice of starting material, reproducible and consistent manufacturing, and preclinical safety and efficacy assessment of the PSC-derived product. This review discusses current US cell therapy regulations and strategies for compliance with these regulations when developing PSC-derived products.


Subject(s)
Cell- and Tissue-Based Therapy , Drug Industry/legislation & jurisprudence , Government Regulation , Pluripotent Stem Cells/cytology , United States Food and Drug Administration , Cell- and Tissue-Based Therapy/standards , Humans , United States
4.
Stem Cells Transl Med ; 4(10): 1097-100, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26285658

ABSTRACT

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.


Subject(s)
Guidelines as Topic , Human Embryonic Stem Cells , Stem Cell Research/legislation & jurisprudence , Tissue Donors/legislation & jurisprudence , United States Food and Drug Administration/legislation & jurisprudence , Biological Products/isolation & purification , Donor Selection/legislation & jurisprudence , Donor Selection/standards , Guideline Adherence , Humans , Patient Safety , United States , United States Food and Drug Administration/standards
5.
Stem Cells Transl Med ; 4(4): 381-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25722426

ABSTRACT

The field of pluripotent stem cells (PSCs) is in a state of dynamic flux driven by significant advances in the derivation of specific phenotypes from embryonic stem cells, breakthroughs in somatic cell nuclear transfer, and dramatic improvements in generating induced PSCs using zero footprint methods. Spurred by these technological advances, companies have begun to plan clinical studies using human PSC derivatives manufactured in current Good Manufacturing Practice-compliant conditions. In the present review, we discuss the challenges in making these biological products, starting from tissue sourcing to the processes involved in manufacture, storage, and distribution. Additional challenges exist to meeting the regulatory requirements and keeping costs affordable. A model is described that has been proposed by the U.S. National Institutes of Health for reducing the costs and permitting flexibility and innovation by individual investigators. This model, combined with small adjustments in the regulatory processes tailored to address the unique properties of PSCs, has the potential of significantly accelerating the implementation of PSC-based cell therapy.


Subject(s)
Cell Differentiation/genetics , Cell- and Tissue-Based Therapy , Pluripotent Stem Cells/cytology , Stem Cell Transplantation , Embryonic Stem Cells/cytology , Humans , United States
6.
Stem Cell Res ; 8(3): 388-402, 2012 May.
Article in English | MEDLINE | ID: mdl-22459095

ABSTRACT

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.


Subject(s)
Cell Culture Techniques/methods , Cell Culture Techniques/standards , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Bioreactors/standards , Cell Differentiation , Cells, Cultured , Cryopreservation , Culture Media, Serum-Free , Humans , Karyotyping , Myocytes, Cardiac/cytology
7.
Nat Biotechnol ; 29(8): 750-6, 2011 Jul 31.
Article in English | MEDLINE | ID: mdl-21804561

ABSTRACT

Using a flow cytometry-based screen of commercial antibodies, we have identified cell-surface markers for the separation of pancreatic cell types derived from human embryonic stem (hES) cells. We show enrichment of pancreatic endoderm cells using CD142 and of endocrine cells using CD200 and CD318. After transplantation into mice, enriched pancreatic endoderm cells give rise to all the pancreatic lineages, including functional insulin-producing cells, demonstrating that they are pancreatic progenitors. In contrast, implanted, enriched polyhormonal endocrine cells principally give rise to glucagon cells. These antibodies will aid investigations that use pancreatic cells generated from pluripotent stem cells to study diabetes and pancreas biology.


Subject(s)
Antigens, CD/metabolism , Biomarkers/metabolism , Cell Separation/methods , Embryonic Stem Cells/cytology , Pancreas/cytology , Animals , Antibodies/metabolism , Cells, Cultured , Embryonic Stem Cells/metabolism , Endoderm/cytology , Flow Cytometry , Humans , Mice , Mice, SCID , Microscopy, Fluorescence , Transplantation, Heterologous
8.
PLoS One ; 6(8): e23018, 2011.
Article in English | MEDLINE | ID: mdl-21857983

ABSTRACT

Human pluripotent stem cell (hPSC) lines have been considered to be homogeneously euploid. Here we report that normal hPSC--including induced pluripotent--lines are karyotypic mosaics of euploid cells intermixed with many cells showing non-clonal aneuploidies as identified by chromosome counting, spectral karyotyping (SKY) and fluorescent in situ hybridization (FISH) of interphase/non-mitotic cells. This mosaic aneuploidy resembles that observed in progenitor cells of the developing brain and preimplantation embryos, suggesting that it is a normal, rather than pathological, feature of stem cell lines. The karyotypic heterogeneity generated by mosaic aneuploidy may contribute to the reported functional and phenotypic heterogeneity of hPSCs lines, as well as their therapeutic efficacy and safety following transplantation.


Subject(s)
Aneuploidy , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Cell Culture Techniques , Cell Line , Female , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Mice , Spectral Karyotyping
9.
Regen Med ; 5(4): 569-79, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20632860

ABSTRACT

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.


Subject(s)
Cell- and Tissue-Based Therapy , Health Policy , Induced Pluripotent Stem Cells/transplantation , Tissue Engineering , United States Food and Drug Administration/legislation & jurisprudence , Cell- and Tissue-Based Therapy/ethics , Cell- and Tissue-Based Therapy/standards , Cell- and Tissue-Based Therapy/trends , Drug Industry/legislation & jurisprudence , Guidelines as Topic , Humans , United States
12.
Nat Biotechnol ; 26(4): 443-52, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18288110

ABSTRACT

Development of a cell therapy for diabetes would be greatly aided by a renewable supply of human beta-cells. Here we show that pancreatic endoderm derived from human embryonic stem (hES) cells efficiently generates glucose-responsive endocrine cells after implantation into mice. Upon glucose stimulation of the implanted mice, human insulin and C-peptide are detected in sera at levels similar to those of mice transplanted with approximately 3,000 human islets. Moreover, the insulin-expressing cells generated after engraftment exhibit many properties of functional beta-cells, including expression of critical beta-cell transcription factors, appropriate processing of proinsulin and the presence of mature endocrine secretory granules. Finally, in a test of therapeutic potential, we demonstrate that implantation of hES cell-derived pancreatic endoderm protects against streptozotocin-induced hyperglycemia. Together, these data provide definitive evidence that hES cells are competent to generate glucose-responsive, insulin-secreting cells.


Subject(s)
Cell Culture Techniques/trends , Embryonic Stem Cells/cytology , Glucose/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Tissue Engineering/trends , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/transplantation , Endoderm/cytology , Endoderm/metabolism , Humans , Insulin-Secreting Cells/transplantation , Mice , Pancreas, Artificial/trends
13.
Nat Biotechnol ; 24(11): 1392-401, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17053790

ABSTRACT

Of paramount importance for the development of cell therapies to treat diabetes is the production of sufficient numbers of pancreatic endocrine cells that function similarly to primary islets. We have developed a differentiation process that converts human embryonic stem (hES) cells to endocrine cells capable of synthesizing the pancreatic hormones insulin, glucagon, somatostatin, pancreatic polypeptide and ghrelin. This process mimics in vivo pancreatic organogenesis by directing cells through stages resembling definitive endoderm, gut-tube endoderm, pancreatic endoderm and endocrine precursor--en route to cells that express endocrine hormones. The hES cell-derived insulin-expressing cells have an insulin content approaching that of adult islets. Similar to fetal beta-cells, they release C-peptide in response to multiple secretory stimuli, but only minimally to glucose. Production of these hES cell-derived endocrine cells may represent a critical step in the development of a renewable source of cells for diabetes cell therapy.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/metabolism , Enteroendocrine Cells/physiology , Islets of Langerhans/growth & development , Pancreatic Hormones/biosynthesis , Peptide Hormones/biosynthesis , Cells, Cultured , Ghrelin , Humans , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Pancreas/cytology , Pancreatic Hormones/isolation & purification
14.
Stem Cells Dev ; 15(6): 931-41, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17253954

ABSTRACT

Current procedures for the maintenance of cardiomyocytes from human embryonic stem (hES) cells rely on either co-culture with mouse cells or medium containing fetal bovine serum (FBS). Due to exposure to animal products, these methods carry the risk of potential pathogen contamination and increased immunogenicity. Additionally, FBS introduces inherent variability in the cultures due to the inevitable differences in serum lots. Here we investigated whether a defined serum-free medium containing creatine, carnitine, taurine, and insulin (CCTI) could maintain hES cell-derived cardiomyocytes. We show that hES cell-derived cardiomyocytes maintained in the CCTI medium in the absence of any feeders exhibit similar phenotypes to those maintained in serum, as indicated by the following observations: (1) comparable levels of cardiac gene transcription were found in cells grown in serum-containing medium versus those in the CCTI medium; (2) cardiomyocyte-associated proteins were expressed in cells cultured in the CCTI medium; (3) beating cells in the CCTI medium responded to pharmacological agents in a dose-dependent manner; and (4) the vast majority of the beating embryoid bodies displayed ventricular-like action potentials (APs), and the ventricular cells in serum-containing medium and the CCTI medium had indistinguishable AP properties. Therefore, culturing hES cell-derived cardiomyocytes in serum-free medium as described here should facilitate the use of the cells for in vitro and in vivo applications.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Heart/physiology , Muscle Cells/physiology , Myocardium/cytology , Carnitine , Cell Culture Techniques/methods , Creatine , Culture Media, Serum-Free , Embryonic Stem Cells/physiology , Humans , Insulin , Muscle Cells/cytology , Taurine
15.
Stem Cells ; 23(10): 1468-78, 2005.
Article in English | MEDLINE | ID: mdl-16123389

ABSTRACT

Human embryonic stem cells (hESCs) derived from human blastocysts have an apparently unlimited proliferative capacity and can differentiate into ectoderm, mesoderm, and endoderm. As such, hESC lines have enormous potential for use in cell replacement therapies. It must first be demonstrated, however, that hESCs maintain a stable karyotype and phenotype and that gene expression is appropriately regulated. To date, different hESC lines exhibit similar patterns of expression of markers associated with pluripotent cells. However, the evaluation of epigenetic status of hESC lines has only recently been initiated. One example of epigenetic gene regulation is dosage compensation of the X chromosome in mammalian females. This is achieved through an epigenetic event referred to as X-chromosome inactivation (XCI), an event initiated upon cellular differentiation. We provide the first evidence that undifferentiated hESC lines exhibit different patterns of XCI.


Subject(s)
Stem Cells/metabolism , X Chromosome Inactivation/physiology , Aneuploidy , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line , Chromatin/genetics , Chromatin/metabolism , Decitabine , Embryo Research , Female , Gene Expression Profiling , Gene Silencing , Humans , RNA, Long Noncoding , RNA, Untranslated/drug effects , RNA, Untranslated/metabolism , Stem Cells/cytology , Teratogens/pharmacology
16.
Nat Biotechnol ; 23(6): 699-708, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15940242

ABSTRACT

Human embryonic stem cells have been defined as self-renewing cells that can give rise to many types of cells of the body. How and whether these cells can be manipulated to replace cells in diseased tissues, used to screen drugs and toxins, or studied to better understand normal development, however, depends on knowing more about their fundamental properties. Many different human embryonic stem cell lines--which are pluripotent, proliferate indefinitely in vitro and maintain a normal, euploid karyotype over extended culture--have now been derived, but whether these cell lines are in fact equivalent remains unclear. It will therefore be important to define robust criteria for the assessment of both existing and newly derived cell lines and for the validation of new culture conditions.


Subject(s)
Cell Culture Techniques , Stem Cells/physiology , Biomarkers , Cell Differentiation , Cell Proliferation , Cytogenetic Analysis , Embryo, Mammalian/cytology , Gene Expression Profiling , Humans
17.
Stem Cells ; 23(3): 315-23, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15749926

ABSTRACT

Previous studies have shown that prolonged propagation of undifferentiated human embryonic stem cells (hESCs) requires conditioned medium from mouse embryonic feeders (MEF-CM) as well as matrix components. Because hESCs express growth factor receptors, including those for basic fibroblast growth factor (bFGF), stem cell factor (SCF), and fetal liver tyrosine kinase-3 ligand (Flt3L), we evaluated these and other growth factors for their ability to maintain undifferentiated hESCs in the absence of conditioned medium. We found cultures maintained in bFGF alone or in combination with other factors showed characteristics similar to MEF-CM control cultures, including morphology, surface marker and transcription factor expression, telomerase activity, differentiation, and karyotypic stability. In contrast, cells in media containing Flt-3L, thrombopoietin, and SCF, individually or in combination, showed almost complete differentiation after 6 weeks in culture. These data demonstrate that hESCs can be maintained in nonconditioned medium using growth factors.


Subject(s)
Cell Proliferation/drug effects , Embryo, Mammalian/cytology , Fibroblast Growth Factor 2/pharmacology , Pluripotent Stem Cells/cytology , Animals , Antigens, CD/metabolism , Antigens, Surface , Cell Differentiation/physiology , Cell Line , Cell Survival/drug effects , Culture Media, Conditioned/pharmacology , DNA-Binding Proteins/genetics , Epidermal Growth Factor/genetics , Flow Cytometry , GPI-Linked Proteins , Gene Expression/genetics , Glycoproteins/metabolism , Glycosphingolipids/metabolism , Humans , Intercellular Signaling Peptides and Proteins , Karyotyping , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, SCID , Neoplasm Proteins/genetics , Octamer Transcription Factor-3 , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Proteoglycans , Stage-Specific Embryonic Antigens , Telomerase/genetics , Telomerase/metabolism , Teratoma/pathology , Tetraspanin 29 , Transcription Factors/genetics
18.
Stem Cell Rev ; 1(2): 139-44, 2005.
Article in English | MEDLINE | ID: mdl-17142848

ABSTRACT

Human embryonic stem cells (hESCs) are derived from human preimplantation embryos, and exhibit the defining characteristics of immortality and pluripotency. Indeed, these cell populations can be maintained for several years in continuous culture, and undergo hundreds of population doublings. hESCs are thus likely candidates for source of cells for cell replacement therapies. Although hESC lines appear stable in their expression of cytokine markers, expression of telomerase, ability to differentiate, and maintenance of a stable karyotype, several other aspects of stability have not yet been addressed, including mitochondrial sequencing, methylation patterns, and fine resolution cytogenetic analysis. Because of the potential utility of hESCs, it will be of utmost importance to evaluate the stability of these aspects of ESC biology.


Subject(s)
Embryonic Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Differentiation , Embryonic Stem Cells/physiology , Humans , Telomere/physiology
19.
Glia ; 49(3): 385-96, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15538751

ABSTRACT

Human embryonic stem cells (hESCs) demonstrate remarkable proliferative and developmental capacity. Clinical interest arises from their ability to provide an apparently unlimited cell supply for transplantation, and from the hope that they can be directed to desirable phenotypes in high purity. Here we present for the first time a method for obtaining oligodendrocytes and their progenitors in high yield from hESCs. We expanded hESCs, promoted their differentiation into oligodendroglial progenitors, amplified those progenitors, and then promoted oligodendroglial differentiation using positive selection and mechanical enrichment. Transplantation into the shiverer model of dysmyelination resulted in integration, differentiation into oligodendrocytes, and compact myelin formation, demonstrating that these cells display a functional phenotype. This differentiation protocol provides a means of generating human oligodendroglial lineage cells in high purity, for use in studies of lineage development, screening assays of oligodendroglial-specific compounds, and treating neurodegenerative diseases and traumatic injuries to the adult CNS.


Subject(s)
Cell Differentiation/physiology , Embryo, Mammalian , Myelin Sheath/physiology , Myelin Sheath/transplantation , Oligodendroglia/cytology , Spinal Cord/cytology , Stem Cell Transplantation/methods , Animals , Cell Line , Demyelinating Diseases/embryology , Demyelinating Diseases/pathology , Demyelinating Diseases/surgery , Humans , Mice , Mice, Neurologic Mutants , Oligodendroglia/transplantation , Spinal Cord/embryology , Spinal Cord/transplantation
20.
Stem Cells ; 22(6): 972-80, 2004.
Article in English | MEDLINE | ID: mdl-15536188

ABSTRACT

Human embryonic stem cells (hESCs) have the potential to generate multiple cell types and hold promise for future therapeutic applications. Although undifferentiated hESCs can proliferate indefinitely, hESC derivatives significantly downregulate telomerase and have limited replication potential. In this study we examine whether the replicative lifespan of hESC derivatives can be extended by ectopic expression of human telomerase reverse transcriptase (hTERT), the catalytic component of the telomerase complex. To this end, we have derived HEF1 cells, a fibroblast-like cell type, differentiated from hESCs. Infection of HEF1 cells with a retrovirus expressing hTERT extends their replicative capacity, resulting in immortal human HEF1-hTERT cells. HEF1-hTERT cells can be used to produce conditioned medium (CM) capable of supporting hESC growth under feeder-free conditions. Cultures maintained in HEF1-CM show characteristics similar to mouse embryonic fibroblast CM control cultures, including morphology, surface marker and transcription factor expression, telomerase activity, differentiation, and karyotypic stability. In addition, HEF1-hTERT cells have the capacity to differentiate into cells of the osteogenic lineage. These results suggest that immortalized cell lines can be generated from hESCs and that cells derived from hESCs can be used to support their own growth, creating a genotypically homogeneous system for the culture of hESCs.


Subject(s)
Embryo, Mammalian/cytology , Fibroblasts/metabolism , Stem Cells/cytology , Adipocytes/metabolism , Alkaline Phosphatase/metabolism , Animals , Anthraquinones/pharmacology , Catalysis , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Cellular Senescence , Chondrocytes/metabolism , Culture Media, Conditioned/pharmacology , Down-Regulation , Flow Cytometry , Humans , Immunohistochemistry , Karyotyping , Mice , Osteoblasts/metabolism , Retroviridae/genetics , Reverse Transcriptase Polymerase Chain Reaction , Telomerase/metabolism , Time Factors , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...