Stimulation of Potent Antigen-Specific T-Cell Responses Using Semiallogenic Dendritic Cells Derived from Human Embryonic Stem Cells.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3704-3704
Author(s):  
Zhen Su ◽  
Fusheng Wei ◽  
Susan Fesperman ◽  
Siqing Wang ◽  
Philipp Dahm ◽  
...  

Abstract The major objective of this study is to develop a novel and broadly applicable immunotherapy platform against cancer and infectious diseases. We hypothesized that human embryonic stem cells (hESC) could serve as a source for generating dendritic cells (DC) with potent immunostimulatory function. One advantage of using hESC-derived DC in clinical settings is the ability to generate virtually unlimited amounts of antigen presenting cells for vaccination. Although hESC-derived DC are not genetically identical to the recipient patient, antigen processing and presentation can be facilitated by matching hESC to recipients that share HLA class I alleles. Another advantage of this technology is that hESC express highly polymorphic HLA class II molecules that serve as major rejection antigens, thereby augmenting the antigen-specific T-cell response in the cancer patient. In the current study, we have established a novel three-step method to differentiate hESC (H9 cell line) into mature DC sequentially through hematopoietic stem cell and myeloid precursor stages. During the first step, 10–15% CD34+ hematopoietic stem cells were generated by co-culturing hESCs with the bone marrow stromal cell line OP9. During the second step, these CD34+ hematopoietic stem cells were differentiated into CD45+CD33+ myeloid precursors and further expanded in the presence of GM-CSF. In the final step, all myeloid precursors were differentiated into mature DC using a cytokine cocktail including GM-CSF, Flt-3L and TNF-α. Using this method, we were able to generate approximately 1 × 108 mature hESC-derived DCs (hESDC) with ≥ 80% purity. These hESDC exhibited a similar phenotype than monocyte-derived DC with high expression of MHC class I, MHC class II, CD11c, CD54, CD40 and the co-stimulatory molecule CD86. Upon activation with proinflammatory cytokines, the hESDC secreted IL-12p70 and migrated in response to MIP-3β. In mixed lymphocyte reaction assays, hESDC exhibited strong allo-stimulatory capacity. Moreover, peptide-loaded mature hESDCs were able to stimulate antigen-specific CD8+ T-cell responses against the EBV peptide BMLF1280-288 and the MART-1 peptide (ELAGIGILTV) in a HLA-A2-restricted manner. Most importantly, hESDC stimulated HLA-A2+ MART-1 peptide-specific CD8+ T cells in vitro that were capable of recognizing and killing the HLA-A2+ melanoma cell line Malena-3M. These data suggest the development of a scalable DC platform that could be applied in clinical immunotherapy protocols.

Stem Cells ◽  
2014 ◽  
Vol 32 (12) ◽  
pp. 3137-3149 ◽  
Author(s):  
Lucy Leshansky ◽  
Daniel Aberdam ◽  
Joseph Itskovitz-Eldor ◽  
Sonia Berrih-Aknin

2009 ◽  
Vol 184 (3) ◽  
pp. 1300-1308 ◽  
Author(s):  
Nurit Yachimovich-Cohen ◽  
Sharona Even-Ram ◽  
Yoel Shufaro ◽  
Jacob Rachmilewitz ◽  
Benjamin Reubinoff

2006 ◽  
Vol 74 (4) ◽  
pp. 160-166 ◽  
Author(s):  
Shiro Iuchi ◽  
Meytha Marsch-Moreno ◽  
Cristina Velez-DelValle ◽  
Karen Easley ◽  
Walid Kuri-Harcuch ◽  
...  

2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Selami Demirci ◽  
Juan J. Haro-Mora ◽  
Alexis Leonard ◽  
Claire Drysdale ◽  
Daniela Malide ◽  
...  

Abstract Background Ex vivo production of hematopoietic stem/precursor cells (HSPCs) represents a promising versatile approach for blood disorders. Methods To derive definitive HSPCs from human embryonic stem cells (ESCs), we differentiated mesodermally specified embryoid bodies (EBs) on gelatin-coated plates in serum/feeder-free conditions. Results Seven-day EB maturation followed by an 8-day differentiation period on OP9 cells provided the highest number of definitive (CD34+ CD235a−, 69%, p < 0.01) and lowest number of primitive (CD34− CD235a+, 1.55%, p < 0.01) precursor cells along with the highest colony-forming units (149.8 ± 11.6, p < 0.01) in feeder-free conditions. Maximal HSPC fraction (CD34+ CD38− CD45RA− CD49f+ CD90+) was 7.6–8.9% after 10 days of hematopoietic differentiation with 14.5% adult β-globin expression following RBC differentiation. Myeloid and erythroid colonies were restricted strictly to the CD34+ CD43+ fraction (370.5 ± 65.7, p < 0.001), while the CD34− CD43+ fraction produced only a small number of colonies (21.6 ± 11.9). In addition, we differentiated the CD34+ CD43+ cells towards T-lymphocytes using the OP9/DLL1 co-culture system demonstrating double-positive T cells (CD4+ CD8+) with CD3+ expression displaying a broad T cell receptor (TCR) repertoire. Confocal imaging of organoid-like structures revealed a close association of CD31+ cells with CD34+ and CD43+ cells, suggesting a potential emergence of HSPCs through endothelial to hematopoietic transition. Furthermore, fluorescently labeled organoids exhibited the emergence of spherical non-attached cells from rare progenitors at the border of the organoid center. Conclusions In summary, definitive HSPCs can be derived from ESCs through a dynamic cellular process from an organoid-like structure, where erythroid progeny are capable of producing adult hemoglobin and lymphoid progeny shows a diverse TCR repertoire.


2013 ◽  
Vol 13 (2) ◽  
pp. 219-229 ◽  
Author(s):  
Audrey V. Parent ◽  
Holger A. Russ ◽  
Imran S. Khan ◽  
Taylor N. LaFlam ◽  
Todd C. Metzger ◽  
...  

Hematology ◽  
2007 ◽  
Vol 2007 (1) ◽  
pp. 11-16 ◽  
Author(s):  
Mickie Bhatia

Abstract The most common human cell-based therapy applied today is hematopoietic stem cell (HSC) transplantation. HSCs can be defined by two essential properties: self-renewal and multilineage hematopoietic differentiation. These combined HSC properties allow them to differentiate into all blood cell types (multilineage) in a sustained manner for the lifetime of the animal, which requires their ability to make cellular copies of themselves (self-renewal). These features can be tested by transplantation from donor to recipient and provide a functional basis to define and identify HSCs. Currently, human bone marrow (BM), mobilized peripheral blood, and umbilical cord blood (CB) represent the major sources of transplantable HSCs, but their availability for use is limited by both quantity and compatibility. Although increasing evidence suggests that somatic HSCs can be expanded to meet current needs, their in vivo potential is concomitantly compromised after ex vivo culture. Pluripotent human embryonic stem cells (hESCs) may provide an alternative. hESCs possess indefinite proliferative capacity in vitro, and have been shown to differentiate into the hematopoietic cell fate, giving rise to erythroid, myeloid, and lymphoid lineages using a variety of differentiation procedures. In most cases, hESC-derived hematopoietic cells show similar clonogenic progenitor capacity and primitive phenotype to somatic sources of hematopoietic progenitors, but possess limited in vivo repopulating capacity when transplanted into immunodeficient mice. Although this suggests HSC function can be derived from hESCs, the efficiency and quality of these cells must be characterized using surrogate models for potential clinical applications.


Sign in / Sign up

Export Citation Format

Share Document