Use of Third Party Ancillary Cells for Enhancement of Full Donor Chimerism and Immunomodulation in Cord Blood Transplants.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 335-335
Author(s):  
Manuel N. Fernandez ◽  
Jose R. Cabrera ◽  
Carmen Regidor ◽  
Guiomar Bautista ◽  
Emilio Ojeda ◽  
...  

Abstract We have pioneered co-infusion of a low number of T-cell highly depleted mobilized hematopoietic stem cells (MHSC) from a third party donor (TPD) as a tool to increase rates of cord blood transplant (CBT) engraftment and full chimerism in adults with high risk hematologic malignancies (“dual transplant”, Haematologica2006; 9:640–8). The conditioning regimes used have been myeloablative although of reduced extra-hematological toxicity. After achieving very favourable results regarding both engraftment and full chimerism, we have started using this approach to evaluate the addition of other TPD cells to the purpose of optimizing CBT immune reconstitution. Results on CBT engraftment, chimerism and survival are available for analysis at this time on 53 patients (M/F 33/20, median age 35 years, range 16–60) who received units with a total cell count (TCC) of 1.1 to 4.3 x 107/Kg (median 2.3) and 0–3/6 HLA disparities, who have received TPD MHSC: 38 from an haploidentical donor, 15 from a related or unrelated donor not sharing an HLA haplotype. Days to ANC>500/uL ranged 9–36 (P50: 11; P90: 20). Initially most of the ANC was predominantly from the TPD with increasing proportions of granulocytes of CB source. Days to full CB chimerisms ranged from 11 to 97 (P50: 37; P90: 93). With a median follow up of 15 months, 3 year OS and DFS of these 53 patients are 60% and 53%. OS for patients older and younger than 40 years are 50% and 64% respectively (p= 0,37). Five patients relapsed, 2 of them achieved new complete remission maintaining full CB chimerism. Acute GVHD occurred in 19 patients, most of them grade I-II with favorable response to treatment. Four cases were grade III-IV, causing death to 3 patients. Other were toxic (VOD 2, MOF 3 and cerebral hemorrhage 1) or infections (all but one CMV). These have been the main cause of morbidity after post-transplant neutropenia and were favoured by a slow recovery of the protective immunity. Reconstitution of lymphocyte subpopulations (detailed data available for 31) is similar to what has been described for single unit CBT: prompt recovery of NK cells (1 month), followed by recovery of B cells (3 months) and slower recovery of T cells subpopulations: T8 in about one year, T4 and T-regs within the second year. No adverse effects due to the co-infusion of the TPD MHSC have been observed. Ex-vivo expanded MSC from the same TPD have been co-infused to 8 patients in addition to the MHSC. The number of co-infused MSC has ranged 1.16–3.24 x 106 cells/kg (median 1.38) without adverse effects observed so far: ANC has occurred as in the other patients and only one had signs of aGVHD, who did nor achieve stable response to antivirals for CMV, achieving continued control of both after the infusion of a new dose of 1.12 x106 cells/kg. Conclusion: These results consolidate our previous description of the “dual transplant” strategy as an approach that may allow high rates of engraftment, full chimerisms and survival of HLA mismatch CBT of relatively low cell content for adults of a wide age range with haematological malignancies, with the possibility of adding other subpopulations of the same TPD as cell therapy tools.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4106-4106
Author(s):  
Simon N. Robinson ◽  
Paul J. Simmons ◽  
Nathalie Brouard ◽  
Shannon Kidd ◽  
Hong Yang ◽  
...  

Abstract INTRODUCTION: Our previous studies have shown that clinically-relevant levels of hematopoietic stem and progenitor cell (HSPC) expansion are possible by ex vivo co-culture of cord blood (CB) mononuclear cells (MNC) with third-party bone marrow (BM)-derived mesenchymal stem cells (MSC) and growth factors.1 A recently activated M. D. Anderson protocol requires that BM from a haplo-identical family member be used for the de novo generation of sufficient MSC for subsequent co-culture, a process requiring ∼3 weeks. Time constraints, uncertainties associated with the identification of a suitable BM donor and potential variation in MSC performance make logistical execution of this strategy difficult. We therefore investigated the potential efficacy of ‘off-the-shelf’ commercially-available sources of MSC. Since MSC do not express HLA-II (DR) they are non-immunogenic, suggesting that this might be a valuable alternative strategy. We compared ex vivo CB HSPC expansion obtained following CB MNC co-culture with 2 commercially-available research-grade MSC isolated by density separation and plastic adherence (MSC#1, Cambrex, Walkersville, MD and MSC#2, Allcells, Emeryville, CA). A third MSC, isolated by Stro-12 selection (MSC#3, supplied by PJS) was also evaluated. METHODS: Two MDACC frozen CB units (CB#1&2) were thawed, washed and co-cultured with adherent monolayers from each MSC. Total nucleated cell (TNC) and HSPC (CD34+ cells and colony-forming units, CFU) numbers were measured at input (Day 0) and output (Day 14). RESULTS: TNC and HSPC numbers revealed that the 2 commercially-available research-grade MSC (MSC#1&2) supported ex vivo CB HSPC expansion. MSC TNC CB34+ CFU n/a - not available CB#1 #1 x 6 x23 n/a #2 x 3 x 8 x15 #3 x 6 x16 x23 CB#2 #1 x 7 x20 x31 #2 x 5 x10 x20 #3 x10 x16 x34 1 Robinson et al. x13 x14 x25 MSC#2 performed less well than MSC#1 for both CB units suggesting that variation may exist between individual MSC. These data suggest that the screening of clinical-grade MSC that perform optimally during ex vivo expansion co-culture might be warranted to best utilize this ‘off-the-shelf’ strategy. Data were similar to previous reports where TNC, CD34+ and CFU numbers were shown to increase approximately 13, 14 and 25-fold, respectively.1 Data were also similar for MSC#3, suggesting that the method used to isolate MSC does not appear to be an important variable for effective CB MNC/MSC co-culture. CONCLUSION: Although research-grade MSC were compared from different commercial sources, these data suggest that, in principle, commercially-available clinical-grade MSC might prove a valuable ‘off-the-shelf’ option, potentially reducing the time to therapy and addressing concerns associated with identifying a BM donor and variation in MSC performance. Future studies will evaluate FDA-compliant MSC that could be used clinically.


F1000Research ◽  
2019 ◽  
Vol 8 ◽  
pp. 1833 ◽  
Author(s):  
Xinxin Huang ◽  
Bin Guo ◽  
Maegan Capitano ◽  
Hal E. Broxmeyer

Cord blood (CB) has been used as a viable source of hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) in over 35,000 clinical hematopoietic cell transplantation (HCT) efforts to treat the same variety of malignant and non-malignant disorders treated by bone marrow (BM) and mobilized peripheral blood (mPB) using HLA-matched or partially HLA-disparate related or unrelated donor cells for adult and children recipients. This review documents the beginning of this clinical effort that started in the 1980’s, the pros and cons of CB HCT compared to BM and mPB HCT, and recent experimental and clinical efforts to enhance the efficacy of CB HCT. These efforts include means for increasing HSC numbers in single CB collections, expanding functional HSCs ex vivo, and improving CB HSC homing and engraftment, all with the goal of clinical translation. Concluding remarks highlight the need for phase I/II clinical trials to test the experimental procedures that are described, either alone or in combination.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3643-3643 ◽  
Author(s):  
Thomas A. Davis ◽  
Fred Gage

Abstract Umbilical cord blood (UCB) is an attractive source of hematopoietic stem cells (HSC) because of greater availability, less stringent HLA matching requirements, and lower incidence and severity of GVHD. Currently, UCB transplant procedures in adults are limited by low collection volumes of total nucleated cells and CD34+ HSC. Approaches to ex vivo expand long-term engraftable HSC have been widely unsuccessful. Recent studies have clearly demonstrated that infusion of a greater number of cells UCB cells enhances the rate of engraftment and lowers the risk of transplantation-related mortality. Machine pulsatile perfusion (MPP) has been successfully used to select cadaveric renal allografts for transplantation, to isolate human islets from pancreata and shown to be a useful cardiac preservation technique in canine heart transplant studies. In this study, the feasibility of using machine pulsatile perfusion to collect human UCB total nucleated cells and CD34+ HSCs was evaluated using placentas designated for research purposes. Immediately following delivery UCB (65 ± 15 mL, n=5) was first collected by needle aspiration from the umbilical cord vein in accordance with standard procedures then followed by MPP (~500 ml) of the placental arteries within 2–3 hours of delivery. Clinically total nucleated cells count (TNC), CD34+ cell numbers and myeloid, erythroid and multipotent CFU progenitor cell content of UCB units are used as predictive measurements of hematopoietic/engraftment potential. Low-density cells (<1.077 g/mL) were isolated by density centrifugation. The median number of viable low density cells obtained was 488 × 106 (range, 240–652 × 106), and 1541 × 106 (range, 888–1800 × 106) for UCB and MPP collections, respectively. MMP low density cell preparations contained significantly more mature segmented neutrophils with a low percentage (<0.1%) of sheared-off vessel wall endothelial cells. Both UCB and MPP low density cells collections showed similar number of assayable CFU-GEMM, CFU-GM, CFU-M, and CFU-G progenitor cells. In contrast, MMP collected cells contained 2–3 times more erythroid BFU-E colonies than UCB collections. Equivalent numbers of CD34+ HSC were enumerated by FACS analysis and subsequently isolated by positive immunomagnetic MACS selection from MPP and UCB collections. Likewise, the progenitor cell content (CFU-GEMM, CFU-GM, CFU-M, CFU-G and BFU-e) of the isolated CD34+ cell populations derived from each cell collection were very similar. These results demonstrate that pulsatile perfusion can be performed easily after traditional UCB collection procedures. This technique effectively recovers on average twice as many TNC and multilineage CD34+ HSC cells when compared to traditional UCB collection procedures. Altogether these results are particular promising since increased numbers of UCB HSC available for infusion should result in accelerated hematopoietic recovery. Moreover, the demonstrated enhanced HSC cell yield together with the simplicity of collection could potentially widen the clinical applicability of UCB transplants in adults.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3032-3032
Author(s):  
Elizabeth Hexner ◽  
Selina M. Luger ◽  
James K. Mangan ◽  
Noelle V. Frey ◽  
Grace R Jeschke ◽  
...  

Abstract Abstract 3032 Successful outcomes following umbilical cord blood transplantation (UCBT) are limited in large part by delayed engraftment, impaired immune reconstitution and an inability to give donor lymphocyte infusions (DLI) in the event of relapse or graft failure. Recent studies suggest double UCBT enhances hematopoietic recovery and may improve leukemia free survival, despite the engraftment of only one unit. Our previous work in a preclinical (xenograft) model showed that T cell activation can enhance hematopoietic recovery after single UCBT. Thus we performed a phase 1 study testing safety and defining the maximum tolerated dose (MTD) of ex vivo CD3/CD28 costimulated UCB-derived T cells co-infused with single UCB grafts in patients with advanced hematologic malignancies. A second objective was to test the feasibility of ex vivo expansion and cryopreservation of UCB T cells for administration as DLI in the event of disease relapse. Eligible subjects had no suitable related or unrelated donor, and had a single 4/6 (or better) HLA-matched UCB graft containing at least 2.5 × 107 nucleated cells/kg. Single umbilical cord blood units stored in 2 fractions were eligible for the intervention. The smaller fraction was thawed 10–14 days prior to infusion and cultured with magnetic beads conjugated to antibodies directed against CD3 and CD28. After myeloablative conditioning, the larger unmanipulated UCB fraction was infused, followed immediately by a fixed dose of the expanded CD3/CD28 costimulated T cells. The remainder of the costimulated T cells were cryopreserved for potential future use as DLI. Four dose levels of initial costimulated T cells (105-108 T cells/kg) were planned. 5 subjects enrolled on the trial; 4 underwent UCBT all of whom were treated at the first dose level (105cells/kg). There were no infusion related adverse events; the dose limiting toxicity (DLT) was conservative and defined as grade 3 or grade 4 GVHD within the first 90 days following UCBT. An MTD was reached at the 105 cells/kg dose level with two subjects experiencing grade 3 GVHD of the gut on days +40 and +27 respectively. For the first 3 subjects enrolled on study, neutrophil engraftment occurred on days +20, +12, and +17, while the fourth subject experienced primary graft failure and received a second mismatched unrelated donor graft. One subject experienced platelet engraftment on day +23. Early (day +11) donor T cell trafficking was documented in this subject's skin using fluorescence in situ hybridization directed at the Y chromosome, and one year post-transplant bone marrow morphologic findings were notable for an exuberant expansion (20% of cellularity) of physiologic precursor B lymphoblasts (hematogones) with a maturing B cell phenotype which correlated with CD4+ immune reconstitution in peripheral blood. Cytokines were measured in the supernatants from expanded T cells and in serum from all subjects. Supernatants contained supraphysiologic levels of cytokines important for engraftment/progenitor/dendritic cell development (GM-CSF, IL-3, FLT-3L) as well as T and B cell differentiation/function (IL-2, IL-4, IL-10, IFN- γ, BAFF). Serum cytokine measurements in recipients were notable for measurable increases in IL-10 following the infusion of expanded T cells for all subjects, with absolute levels lower in the two subjects with DLTs. 3 of 4 expansions yielded adequate numbers of cells for cryopreservation as future use for DLI. Taken together, these preliminary data are consistent with our preclinical observations of rapid engraftment in recipients of a single UCBT combined with relatively low doses of activated T cells. Additional safety studies are needed to determine the optimal T cell dose. If confirmed in larger numbers of patients, this represents an attractive strategy for improving engraftment, immune reconstitution, as well as a method to enable DLI following UCBT. Disclosures: Off Label Use: Investigational cellular therapy product tested under an IND.


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.


Author(s):  
Valentina Orticelli ◽  
Andrea Papait ◽  
Elsa Vertua ◽  
Patrizia Bonassi Signoroni ◽  
Pietro Romele ◽  
...  

Blood ◽  
2003 ◽  
Vol 101 (12) ◽  
pp. 5061-5067 ◽  
Author(s):  
Jennifer Jaroscak ◽  
Kristin Goltry ◽  
Alan Smith ◽  
Barbara Waters-Pick ◽  
Paul L. Martin ◽  
...  

AbstractAllogeneic stem cell transplantation with umbilical cord blood (UCB) cells is limited by the cell dose a single unit provides recipients. Ex vivo expansion is one strategy to increase the number of cells available for transplantation. Aastrom Biosciences developed an automated continuous perfusion culture device for expansion of hematopoietic stem cells (HSCs). Cells are expanded in media supplemented with fetal bovine serum, horse serum, PIXY321, flt-3 ligand, and erythropoietin. We performed a phase 1 trial augmenting conventional UCB transplants with ex vivo–expanded cells. The 28 patients were enrolled on the trial between October 8, 1997 and September 30, 1998. UCB cells were expanded in the device, then administered as a boost to the conventional graft on posttransplantation day 12. While expansion of total cells and colony-forming units (CFUs) occurred in all cases, the magnitude of expansion varied considerably. The median fold increase was 2.4 (range, 1.0-8.5) in nucleated cells, 82 (range, 4.6-266.4) in CFU granulocyte-macrophages, and 0.5 (range, 0.09-2.45) in CD34+ lineage negative (lin–) cells. CD3+ cells did not expand under these conditions. Clinical-scale ex vivo expansion of UCB is feasible, and the administration of ex vivo–expanded cells is well tolerated. Augmentation of UCB transplants with ex vivo–expanded cells did not alter the time to myeloid, erythroid, or platelet engraftment in 21 evaluable patients. Recipients of ex vivo–expanded cells continue to have durable engraftment with a median follow-up of 47 months (range, 41-51 months). A randomized phase 2 study will determine whether augmenting UCB transplants with ex vivo–expanded UCB cells is beneficial.


Sign in / Sign up

Export Citation Format

Share Document