A stromal-free, serum-free system to expand ex vivo hematopoietic stem cells from mobilized peripheral blood of patients with hematologic malignancies and healthy donors

Cytotherapy ◽  
2013 ◽  
Vol 15 (9) ◽  
pp. 1126-1135 ◽  
Author(s):  
Chao-Ling Yao ◽  
Shu-Ching Hsu ◽  
Shiaw-Min Hwang ◽  
Wei-Chi Lee ◽  
Tzeon-Jye Chiou
Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 482-482
Author(s):  
Holger Karsunky ◽  
Robert J. Tressler ◽  
Joy Chananukul

Abstract Abstract 482 Thrombocytopenia is a common side effect of high-dose chemotherapy that can compromise cancer treatment by requiring treatment delay and/or dose reduction for the patient. Platelet transfusion is typically given to prevent severe hemorrhage. However, several factors including acquisition, banking, and associated risks of bacterial infections and alloimmunization are hampering reliance on platelet transfusion. Growth factors are also used to stimulate proliferation and differentiation of megakaryocytes to increase platelet production, but in severely myelosuppressed patients these have only had modest benefit. The limitations of these two modalities for the treatment of chemotherapy-induced thrombocytopenia indicates that additional treatment approaches are needed. We have developed a novel approach to reconstitute megakaryocytes and platelets in thrombocytopenic patients which is presented here. We have identified a scalable culture system using serum-free medium and a defined cytokine cocktail free of animal products to expand CD34+ hematopoietic stem cells from G-CSF mobilized peripheral blood donors in vitro and direct their development to the megakaryocyte lineage to yield committed human megakaryocyte progenitors (MKPs). These MKPs can be readily cryopreserved while retaining their capacity to generate CFU-MK and platelets in vitro. When infused into NSG mice, ex vivo expanded MKP generate clinically relevant platelet levels of platelets in blood within a few days with sustained platelet levels for several weeks. The platelets generated from MKP in vivo are also functional as assessed by CD62P expression in responses to ADP stimulation in vitro. Our results present a compelling approach for the development of off-the-shelf storable MKPs for the treatment of thrombocytopenia. Disclosures: Karsunky: Cellerant Therapeutics Inc.: Employment, Patents & Royalties. Tressler:Cellerant Therapeutics, Inc.: Employment, Equity Ownership. Chananukul:Cellerant Therapeutics Inc.: Employment, Patents & Royalties.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 371-371
Author(s):  
HakMo Lee ◽  
Wan Wu ◽  
Marcin Wysoczynski ◽  
Magdalena Kucia ◽  
Mary J. Laughlin ◽  
...  

Abstract Abstract 371 Current strategies to accelerate hematopoietic reconstitution after transplantation, include transplantation of greater numbers of HSC or ex vivo expansion of harvested HSC before transplant. However, the number of HSC availabel for allogeneic or autologous transplantation can be low (e.g., umbilical cord blood, poor mobilizers) and strategies to expand HSC and maintain equivalent engraftment capability ex vivo are limited. We reported that some compounds present in leucopheresis products [(e.g., platelet-derived microparticles (Blood 2001, 98: 3143)] and some complement cascade cleavage fragments, e.g., anaphylatoxin C3a (Blood 2005, 101, 3784), enhance the homing responses of HSC to SDF-1 gradient. We recently noted that small cationic peptides released from activated granulocytes (beta2-defensin and cathelicidin) positively prime responsiveness of murine and human HSC to SDF-1 gradient (Leukemia 2009; in press). Accordingly, both compounds enhanced transwell migration of HSC to low threshold doses of SDF-1. This phenomenon was not receptor-dependent, as agonists of membrane receptors that may bind beta2-defensin (FPRL-1), cathelicidin (CCR6) - FPRL-1 agonist, and MIP-3alpha, respectively, did not show similar priming effects. This could be explained by affected distribution of membrane lipids by cationic peptides. In support of this notion, an inhibitor of cell membrane raft formation (methyl-b-cyclodextran) inhibited the priming effect of both compounds, indicating this effect is dependent on CXCR4 incorporation into lipid rafts. Direct confocal analysis of CXCR4 and lipid raft colocalization in the presence or absence of cationic peptides confirmed these findings. Because leucopheresis products are enriched in activated granulocytes that release beta2-defensin and cathelicidin, we tested whether this may explain why mobilized peripheral blood stem cells (PBSC) engraft faster compared to HSC isolated directly from bone marrow (BM) in a murine BM transplant model. Accordingly, syngeneic BMMNCs were exposed ex vivo to beta2-defensin or cathelicidin for 30 minutes and subsequently transplanted into lethally irradiated recipients. We noted that animals transplanted with BM cells primed by those cationic peptides showed accelerated recovery of platelets and neutrophils by ∼3-5 days compared to unprimed control cells. We envision that small cationic peptides, which primarily possess antimicrobial functions and are harmless to mammalian cells, could be clinically applied to prime human HSC before transplantation. This novel approach would be particularly important in cord blood transplantation, where the number of HSC availabel for transplantation is usually limited. We postulate that this promising strategy warrants further investigations. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 105 (10) ◽  
pp. 3848-3854 ◽  
Author(s):  
Rong Hu ◽  
Galina L. Mukhina ◽  
Steven Piantadosi ◽  
Jamie P. Barber ◽  
Richard J. Jones ◽  
...  

AbstractParoxysmal nocturnal hemoglobinuria (PNH) is caused by phosphatidylinositol glycan–class A (PIG-A) mutations in hematopoietic stem cells (HSCs). PIG-A mutations have been found in granulocytes from most healthy individuals, suggesting that these spontaneous PIG-A mutations are important in the pathogenesis of PNH. It remains unclear if these PIG-A mutations have relevance to those found in PNH. We isolated CD34+ progenitors from 4 patients with PNH and 27 controls. The frequency of PIG-A mutant progenitors was determined by assaying for colony-forming cells (CFCs) in methylcellulose containing toxic doses of aerolysin (1 × 10-9 M). Glycosylphosphatidylinositol (GPI)–anchored proteins serve as receptors for aerolysin; thus, PNH cells are resistant to aerolysin. The frequency of aerolysin resistant CFC was 14.7 ± 4.0 × 10-6 in the bone marrow of healthy donors and was 57.0 ± 6.7 × 10-6 from mobilized peripheral blood. DNA was extracted from individual day-14 aerolysin-resistant CFCs and the PIG-A gene was sequenced to determine clonality. Aerolysin-resistant CFCs from patients with PNH exhibited clonal PIG-A mutations. In contrast, PIG-A mutations in the CFCs from controls were polyclonal, and did not involve T cells. Our data confirm the finding that PIG-A mutations are relatively common in normal hematopoiesis; however, the finding suggests that these mutations occur in differentiated progenitors rather than HSCs.


2020 ◽  
Vol 15 (3) ◽  
pp. 250-262
Author(s):  
Maryam Islami ◽  
Fatemeh Soleimanifar

Transplantation of hematopoietic stem cells (HSCs) derived from umbilical cord blood (UCB) has been taken into account as a therapeutic approach in patients with hematologic malignancies. Unfortunately, there are limitations concerning HSC transplantation (HSCT), including (a) low contents of UCB-HSCs in a single unit of UCB and (b) defects in UCB-HSC homing to their niche. Therefore, delays are observed in hematopoietic and immunologic recovery and homing. Among numerous strategies proposed, ex vivo expansion of UCB-HSCs to enhance UCB-HSC dose without any differentiation into mature cells is known as an efficient procedure that is able to alter clinical treatments through adjusting transplantation-related results and making them available. Accordingly, culture type, cytokine combinations, O2 level, co-culture with mesenchymal stromal cells (MSCs), as well as gene manipulation of UCB-HSCs can have effects on their expansion and growth. Besides, defects in homing can be resolved by exposing UCB-HSCs to compounds aimed at improving homing. Fucosylation of HSCs before expansion, CXCR4-SDF-1 axis partnership and homing gene involvement are among strategies that all depend on efficiency, reasonable costs, and confirmation of clinical trials. In general, the present study reviewed factors improving the expansion and homing of UCB-HSCs aimed at advancing hematopoietic recovery and expansion in clinical applications and future directions.


Blood ◽  
2004 ◽  
Vol 103 (10) ◽  
pp. 3710-3716 ◽  
Author(s):  
Peter A. Horn ◽  
Kirsten A. Keyser ◽  
Laura J. Peterson ◽  
Tobias Neff ◽  
Bobbie M. Thomasson ◽  
...  

Abstract The use of lentiviral vectors for the transduction of hematopoietic stem cells has evoked much interest owing to their ability to stably integrate into the genome of nondividing cells. However, published large animal studies have reported highly variable gene transfer rates of typically less than 1%. Here we report the use of lentiviral vectors for the transduction of canine CD34+ hematopoietic repopulating cells using a very short, 18-hour transduction protocol. We compared lentiviral transduction of hematopoietic repopulating cells from either stem cell factor (SCF)– and granulocyte-colony stimulating factor (G-CSF)–primed marrow or mobilized peripheral blood in a competitive repopulation assay in 3 dogs. All dogs engrafted rapidly within 9 days. Transgene expression was detected in all lineages (B cells, T cells, granulocytes, and red blood cells as well as platelets) indicating multilineage engraftment of transduced cells, with overall long-term marking levels of up to 12%. Gene transfer levels in mobilized peripheral blood cells were slightly higher than in primed marrow cells. In conclusion, we show efficient lentiviral transduction of canine repopulating cells using an overnight transduction protocol. These results have important implications for the design of stem cell gene therapy protocols, especially for those diseases in which the maintenance of stem cells in culture is a major limitation.


2003 ◽  
Vol 14 (17) ◽  
pp. 1683-1686 ◽  
Author(s):  
Bobbie Thomasson ◽  
Laura Peterson ◽  
Jesse Thompson ◽  
Martin Goerner ◽  
Hans-Peter Kiem

Sign in / Sign up

Export Citation Format

Share Document