scholarly journals Mobilized Mouse and Human Peripheral Blood Containing Elevated Numbers of Donor Treg Cells Ameliorates Pre-Clinical GvHD and GVL Is Maintained in an MHC-Matched Allogeneic Murine Model

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2767-2767
Author(s):  
Henry Barreras ◽  
Sabrina N. Copsel ◽  
Ying Ding ◽  
Charles J. Cash ◽  
Cameron S. Bader ◽  
...  

Abstract Mobilized hematopoietic stem / progenitor cells have become the primary option used for adult allogeneic hematopoietic stem cell transplants (aHSCTs) although GVHD remains a major immunologic complication (CIBMTR, 2017). The present studies are directed to developing a translational model for the application of donor regulatory T cells (Tregs) together with the use of mobilized peripheral blood (mPB) for more widespread application of these aHSCTs. We have previously reported that two-pathway Treg expansion strategy markedly elevates Treg numbers and function (Wolf, 2017; Copsel 2018). Additionally, this approach could be effectively applied in a concomitant strategy to amplify Treg suppressor activity in mPB to diminish pre-clinical GVHD (Barreras H, ASTCT Meeting S292 ,2019). Here, we present new findings using mPB transplants examining GVL reactivity in an MHC-matched minor antigen mismatched pre-clinical model and test the notion that human donor Tregs can be employed with mobilized healthy donor PB to effectively regulate xenogeneic GVHD. Donor C3H.SW (H2b) mice were treated with filgrastim for 4 days prior to use for transplants and PB analysis demonstrated significant elevations in c-kit + cells. Some PB donors were concurrently administered the two-pathway Treg expansion strategy (D-6 through D-1) consisting of TL1A-Ig, targeting TNFRSF25, and low dose huIL-2. A significant increase (up to 40%) in the frequency of CD4 +FoxP3 + Tregs occurred during the mobilization process. Pooled C3H.SW PB was collected from both mobilized and Treg unexpanded ("TrUM") or mobilized and Treg expanded ("TrEM") donors and transplanted into lethally irradiated, MHC-matched B6 (H2b) recipients. Recipients of TrEM donors exhibited significantly reduced weight loss and clinical GVHD scores compared to recipients of TrUM (Fig.1A). GVL responses were tested in animals administered B6-MLL-AF9 GFP leukemia cells administered at the time of transplant. Notably, recipients of TrEM exhibited comparable GVL activity to TrUM recipients evidenced by B6-MLL-AF9 GFP levels in bone marrow and spleen (Fig.1B). To begin to translate these findings, we tested the use of ex-vivo expanded human donor Tregs (huTregs) to ameliorate xenogeneic GVHD (xGVHD). First, sorted huTregs (CD4 +CD25 +CD127 lo) from a healthy donor were expanded ex-vivo using anti-CD3/anti-CD28 beads 1 week prior to transplant (Fig 2A). next, these expanded huTregs were combined with human mPB from the same healthy donor (6x10 6 PBMC) and transplanted into NSG/ NOD-scid IL2Rgamma null mice. We found that treatment with ex-vivo expanded huTregs resulted in significant reduction of mortality rate and clinical xGVHD (Fig 2B,C). Notably, 1 week post-transplant PB huTregs levels were still elevated and frequency of huCD4 +Tconv cells was diminished supporting xGVHD outcomes (Fig 2D). In total, these findings demonstrated that the use of mPB containing elevated Treg levels significantly reduced pre-clinical GVHD without loss of GVL activity in an MHC-matched allogeneic model. Moreover, utilizing ex-vivo expanded huTregs from a mPB donor and added back to the same donor at the time of transplant ameliorated xGVHD. The observations indicate that during the donor stem / progenitor cell mobilization process, manipulation of donor Tregs using our two-pathway strategy can be successfully accomplished in PB resulting in an effective and translational approach to ameliorate GVHD following aHSCT. In total, the present studies support the notion that in vivo or ex-vivo manipulation of donor tregs together with mobilized peripheral blood could provide therapeutic approaches to improve aHSCT outcomes. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.

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 ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3343-3343
Author(s):  
Matteo Maria Naldini ◽  
Gabriele Casirati ◽  
Erika Zonari ◽  
Giacomo Desantis ◽  
Andrea Cammarata ◽  
...  

Abstract Hematopoietic stem and progenitor cell (HSPC) expansion remains an important unmet goal for ex vivo gene therapy based on gene addition and editing to compensate for the negative impact of the gene transfer procedure enabling faster engraftment and less complications. Additionally, ex vivo expansion of corrected cells may improve efficacy at more sustainable manufacturing costs by downscaling transduction. To date, our knowledge of precise mechanisms of action of expansion compounds is limited, and it remains unclear whether cord blood expansion protocols also maintain stemness of mobilized peripheral blood CD34+ cells (mPB), the preferred HSPC source for gene therapy. We performed serial (day 0,4,8) droplet-based single cell RNA sequencing (scRNAseq) on lentivirally transduced mPB expanded with UM171 to dissect cellular heterogeneity, monitor population dynamics over time and identify a transcriptional profile of primitive cells in culture. By associating published HSPC gene expression profiles to our scRNAseq dataset from uncultured mPB, we found that 45% of cells harbored a myelo-lymphoid signature. Smaller cell clusters expressed a shared erythroid (ERY) and megakaryocytic (MK) signature (20%), or a more primitive multipotent HSC-like signature (15%) characterized by enhanced JAK/STAT signaling and expression of HSC associated genes (AVP, HOPX, ID3). Unsupervised ordering of cells within pseudotime separated emerging MK/ERYpoiesis (FCER1A, HBD) from lympho-myelopoiesis (CD52, JUN), with intermediate states of more primitive progenitors located in between. After 4 days in culture, we noted a general increase in nuclear and mitochondrial gene transcription with activation of oxidative metabolism, paralleled by cell cycle activation, as expected from cytokine stimulation. By d8 of culture these changes leveled off but remained higher than uncultured cells. Of note, cells at d8 revealed an activation of cellular stress response pathways (e.g. TNFa, IFNg) hinting towards a compromised culture that may eventually exhaust HSC. Unsupervised clustering of cultured mPB highlighted a dramatic expansion (70-80%) of MK/ERY progenitor cells with high cycling activity with only 20-30% cells showing myelo-lymphoid transcriptional features. In line, pseudotime analysis highlighted a main ERY and MK trajectory separated from that of cells characterized by the expression of HSPC genes (HOPX, SPINK2) and of an emerging myeloid trajectory (MPO). To profile HSC in culture, we sorted and sequenced CD34+90+201+ cells from d4 expansion culture (3% of total cells), which we show to contain >70% of SCID repopulating potential. ScRNAseq revealed transcriptional similarity with the myelo-lymphoid progenitor cluster identified in the unsorted d4 culture. Unsupervised clustering of the CD34+90+201+ population revealed cell cycle dependent heterogeneity, identifying a highly quiescent cluster with expression of HSC-like signatures. This cluster was also characterized by relatively low gene expression, possibly reflecting a non-activated cell state consistent with primitive HSPC. Pseudotime analysis produced a four-branched minimum spanning tree, which retained a clear cell cycle and metabolic effect. Top variable genes included cell cycle, glycolytic, mitochondrial and ribosomal genes, identifying different metabolic modules along the branched trajectory. These results highlight that cell heterogeneity within a purified, HSC-enriched population is driven mainly by metabolic activation and cell cycle status. As a complementary approach, we purified LT-HSC from uncultured mPB (CD34+38-90+45RA-49f+), marked them with CFSE and expanded them in UM171 culture. LT-HSCs expanded on average 3.5 fold in 7 days, with the following distribution: 0 divisions: 3%; 1: 26%; 2: 47%; 3: 21%; 4: 3%. We performed scRNAseq on LT-HSC pre culture and after 7d separating a highly proliferative (≥2 divisions) and quiescent (0 - 1 division) fraction, allowing us to obtain unprecedented insight into the response of engrafting cells to ex vivo culture and set a framework to dissect self-renewal (HSC expansion), HSC maintenance and loss through differentiation as potential culture outcomes. Our combined functional/transcriptomic approach will define new HSC markers in culture and greatly facilitate side-by-side comparison of different expansion protocols towards rapid clinical translation. Disclosures No relevant conflicts of interest to declare.


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 ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4416-4416
Author(s):  
Jennifer E Adair ◽  
Kevin G Haworth ◽  
Guy Sauvageau ◽  
Shelly Heimfeld ◽  
Jonah D Hocum ◽  
...  

Abstract Hematopoietic stem and progenitor cell (HSPC) gene therapy holds great promise but requires highly technical and dedicated facilities. Current state-of-the-art requires ex vivo HSPC gene transfer in dedicated Good Manufacturing Practices (GMP) facility infrastructure, limiting treatment to highly developed countries. A patient point-of-care strategy would therefore make HSPC gene therapy available to patients worldwide. We developed a short, semi-automated, mostly-closed platform for ex vivo isolation and lentivirus (LV) gene modification of CD34+ HSPCs from either bone marrow or mobilized peripheral blood sources using the CliniMACS Prodigy® (Figure 1). Experiments were performed with a biosafety cabinet and personal protective equipment to simulate anticipated conditions in clinical facilities of underdeveloped countries. A total of 7 custom programs were developed for bone marrow or mobilized peripheral blood CD34+ cell isolation and LV transduction. Addition of a pyrimidoindole derivative, UM 729, permitted efficient transduction of CD34+ HSPCs in <18 hours. Complete semi-automated production took <34 hours from collection to infusion of the gene modified cell product, requiring very little operator hands-on time. Autologous, LV gene-modified CD34+ HSPCs from two nonhuman primates produced using this platform engrafted and supported multilineage hematopoietic repopulation after myeloablative total body irradiation at 1020 cGy. Total cell doses achieved were 27 x 106 and 5.4 x 106 CD34+ cells/kg body weight, respectively. We observed stable, persistent and polyclonal gene marking in peripheral blood granulocytes up to 40% and lymphocytes up to 15% within 4 months after infusion. Neither animal displayed evidence for increased toxicity, including potential contamination from the cell products. We then validated processing of human bone marrow and mobilized apheresis products from healthy adult donors. We demonstrate efficient isolation of human CD34+ HSPCs and up to 60% transduction efficiency with a clinical-grade LV currently being tested in a phase I clinical trial for treatment of HIV-associated lymphoma. Products tested met current FDA approved specifications for infusion. Xenotransplantation of these products into immunodeficient mice resulted in polyclonal engraftment over 12 weeks. These data demonstrate preclinical safety and feasibility of a patient point-of-care strategy for ex vivo LV gene transfer into HSPCs. This platform represents a major advance in global portability of LV mediated HSPC gene therapy. Figure 1. Figure 1. Disclosures No relevant conflicts of interest to declare.


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.


Blood ◽  
1997 ◽  
Vol 89 (11) ◽  
pp. 3919-3924 ◽  
Author(s):  
Jean C.Y. Wang ◽  
Monica Doedens ◽  
John E. Dick

Abstract We have previously reported the development of in vivo functional assays for primitive human hematopoietic cells based on their ability to repopulate the bone marrow (BM) of severe combined immunodeficient (SCID) and nonobese diabetic/SCID (NOD/SCID) mice following intravenous transplantation. Accumulated data from gene marking and cell purification experiments indicate that the engrafting cells (defined as SCID-repopulating cells or SRC) are biologically distinct from and more primitive than most cells that can be assayed in vitro. Here we demonstrate through limiting dilution analysis that the NOD/SCID xenotransplant model provides a quantitative assay for SRC. Using this assay, the frequency of SRC in cord blood (CB) was found to be 1 in 9.3 × 105 cells. This was significantly higher than the frequency of 1 SRC in 3.0 × 106 adult BM cells or 1 in 6.0 × 106 mobilized peripheral blood (PB) cells from normal donors. Mice transplanted with limiting numbers of SRC were engrafted with both lymphoid and multilineage myeloid human cells. This functional assay is currently the only available method for quantitative analysis of human hematopoietic cells with repopulating capacity. Both CB and mobilized PB are increasingly being used as alternative sources of hematopoietic stem cells in allogeneic transplantation. Thus, the findings reported here will have important clinical as well as biologic implications.


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