Characterization and Function of Cryopreserved Bone Marrow from Deceased Organ Donors: A Potential Viable Alternative Graft Source

Brian H Johnstone, John R Woods, W Scott Goebel, Dongsheng Gu, Chieh-Han Lin, Hannah M Miller, Kelsey G Musall, Aubrey M Sherry, Barbara J Bailey, Emily Sims, Anthony L Sinn, Karen E Pollok, Stephen Spellman, Jeffery J Auletta, Erik J Woods, Brian H Johnstone, John R Woods, W Scott Goebel, Dongsheng Gu, Chieh-Han Lin, Hannah M Miller, Kelsey G Musall, Aubrey M Sherry, Barbara J Bailey, Emily Sims, Anthony L Sinn, Karen E Pollok, Stephen Spellman, Jeffery J Auletta, Erik J Woods

Abstract

Despite the readily available graft sources for allogeneic hematopoietic cell transplantation (alloHCT), a significant unmet need remains in the timely provision of suitable unrelated donor grafts. This shortage is related to the rarity of certain HLA alleles in the donor pool, nonclearance of donors owing to infectious disease or general health status, and prolonged graft procurement and processing times. An alternative hematopoietic progenitor cell (HPC) graft source obtained from the vertebral bodies (VBs) of deceased organ donors could alleviate many of the obstacles associated with using grafts from healthy living donors or umbilical cord blood (UCB). Deceased organ donor-derived bone marrow (BM) can be preemptively screened, cryogenically banked for on-demand use, and made available in adequate cell doses for HCT. We have developed a good manufacturing practice (GMP)-compliant process to recover and cryogenically bank VB-derived HPCs from deceased organ donor (OD) BM. Here we present results from an analysis of HPCs from BM obtained from 250 deceased donors to identify any substantial difference in composition or quality compared with HPCs from BM aspirated from the iliac crests of healthy living donors. BM from deceased donor VBs was processed in a central GMP facility and packaged for cryopreservation in 5% DMSO/2.5% human serum albumin. BM aspirated from living donor iliac crests was obtained and used for comparison. A portion of each specimen was analyzed before and after cryopreservation by flow cytometry and colony-forming unit potential. Bone marrow chimerism potential was assessed in irradiated immunocompromised NSG mice. Analysis of variance with Bonferroni correction for multiple comparisons was used to determine how cryopreservation affects BM cells and to evaluate indicators of successful engraftment of BM cells into irradiated murine models. The t test (with 95% confidence intervals [CIs]) was used to compare cells from deceased donors and living donors. A final dataset of complete clinical and matched laboratory data from 226 cryopreserved samples was used in linear regressions to predict outcomes of BM HPC processing. When compared before and after cryopreservation, OD-derived BM HPCs were found to be stable, with CD34+ cells maintaining high viability and function after thawing. The yield from a single donor is sufficient for transplantation of an average of 1.6 patients (range, 1.2 to 7.5). CD34+ cells from OD-derived HPCs from BM productively engrafted sublethally irradiated immunocompromised mouse BM (>44% and >67% chimerism at 8 and 16 weeks, respectively). Flow cytometry and secondary transplantation confirmed that OD HPCs from BM is composed of long-term engrafting CD34+CD38-CD45RA-CD90+CD49f+ HSCs. Linear regression identified no meaningful predictive associations between selected donor-related characteristics and OD BM HPC quality or yield. Collectively, these data demonstrate that cryopreserved BM HPCs from deceased organ donors is potent and functionally equivalent to living donor BM HPCs and is a viable on-demand graft source for clinical HCT. Prospective clinical trials will soon commence in collaboration with the Center for International Blood and Marrow Research to assess the feasibility, safety, and efficacy of Ossium HPCs from BM (ClinicalTrials.gov identifier NCT05068401).

Keywords: Bone Marrow; CD34; Cryopreservation; Murine Engraftment; Organ Donor; Vertebral Body.

Copyright © 2022 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1.
Figure 1.
Comparison of deceased OD VB-derived OD HPC, marrow and LD-IC-aspirated BM. (A) Viability of total CD45+WBCs. (B) Viability of CD34+HSPCs. (C) Viability of CD3+ T cells. (D) Percentage of TNCs that are CD34+ HSPCs. (E) Percentage of TNCs that are CD3+ T cells. (F) Numbers of granulocyte macrophage colony-forming unit (CFU-GM) progenitors in 105 whole BM cells. (G) Numbers of total CFU progenitors in 105 whole BM cells. *P < .05, Welch 2-tailed ttest.
Figure 2.
Figure 2.
Comparison of HSPC populations in the CD34+ cell fraction of OD HPC, marrow and living donor BM. (A) Representative gating strategy to define and enumerate HSPCs. (B) Long-term repopulating HSC populations, defined as CD34+CD38−CD45RA−CD90+CD49f+, in CD34+ populations selected from OD-VB and LD-IC BM. (C) Multipotent progenitor (MPP) populations (CD34+CD38+) in CD34+ populations selected from OD-VB and LD-IC BM. (D) CFU progenitor populations in selected CD34+ cells from OD-VB (black bars) and LD-IC (gray bars) BM. CFU-E, CFU-erythroid; BFU-E, burst forming unit-erythroid; CFU-GEMM, CFU-granulocyte, erythroid, macrophage, megakaryocyte; CFU-total, the sum of individual progenitors.
Figure 3.
Figure 3.
Characterization of fresh (green circles) and post-cryopreserved (blue boxes) OD HPC, marrow. (A) TNC and CD45+WBC counts/mL. (B) Total and viable CD34+ HSPCs/mL. (C) Viable CD34+ cells as a percentage of WBCs. (D) Total and viable CD3+ T cells/mL. (E) Viable percentages of CD45+ WBCs, CD3+ T cells, and CD34+ HSPCs. (F) Mean ± SD and range CD34+ HSPC counts. Viable CD34+ cells/unit is the number of HSPCs in 65 mL stored in 250-mL blood bags. Units/donor is the number of bags obtained from a donor. Viable CD34+ cells/donor is the total yield of HSPCs per donor. Grafts/70 kg patient is the number of transplantations at 2 × 106 CD34+ cells/kg that can be performed with the total CD34 cells yielded from each donor. (G) Frequency of viable WBCs positive for markers defining the indicated lymphocyte subsets. (H) Fractions of CFU subsets in fresh and cryopreserved HPC, marrow. The average total CFUs per 105 BM cells plated is indicated. *P < .05; **P < .01; ***P < .001; ****P < 0.0001, 2-way analysis of variance with Sidak’s multiple comparison test. The total number of fresh samples was 250, of which 226 were analysed post-thaw. Six samples were analyzed in (G).
Figure 4.
Figure 4.
Transplantation of cryopreserved human immunomagnetically selected CD34+ cells from OD HPC, marrow recovered from 2 donors (BM1 and BM2). Immunocompromised NSG mice were irradiated at 300 cGy, followed by injection of CD34+ cells at a dose of 5 × 105 through the tail vein. (A) Percentage of human CD45+ cells in mouse peripheral blood at 8 weeks. (B-F)Analysis of BM at 16 weeks for percentage of cells expressing human surface epitopes for CD45 (B), CD34 (C), CD38 (D), CD33 (E), and CD19 (F). (G and H) Percentage of human CD45+ cells in peripheral blood (PB) (G) and spleen (H) at 16 weeks. (I) Comparison of total CFU in cryopreserved selected CD34+ cells. (J) Comparison of total human CFU in BM of mice at 16 weeks. (K and L) Secondary transplantations: human CD45+ cells in peripheral blood (K) and BM (L) at 16 weeks following irradiation and injection with whole BM from mice receiving transplantation with HPC, marrow CD34+ cells (10 NSG mice per group). *P < .05; **P < .01, ***P < .001, analysis of variance with Tukey’s multiple comparisons test.
Figure 5.
Figure 5.
Summary results from linear regression analyses showing the donor-related variables found to be significantly associated with variation in OD HPC, marrow composition. A complete list of the donor-related variables tested in the regressions is provided in Supplementary Table S4. Means (intercept values) are represented by vertical lines. The left columns identify the donor-related variables that were significantly associated with processed cell outcomes. The right columns show the values of regression coefficients associated with those donor-related variables. The bars indicate whether a donor-related variable had a negative (red) or positive (green) impact (ie, raised or lowered the slope, respectively) on the outcome. The range is represented by the horizontal bar. Donor-related variables contributed significantly to OD HPC, marrow variation in CD34+ cell viability (A), CD3+ cell viability (B), and total CD3+ cell count/mL (C).

References

    1. Auletta JJ, Kou J, Chen M, Shaw BE. Current use and outcome of hematopoietic stem cell transplantation: CIBMTR US summary slides, 2021. Available at: . Accessed XXX.
    1. Ciurea SO, Zhang MJ, Bacigalupo AA, et al. Haploidentical transplant with posttransplant cyclophosphamide vs matched unrelated donor transplant for acute myeloid leukemia. Blood. 2015;126:1033–1040.
    1. Kasamon YL, Ambinder RF, Fuchs EJ, et al. Prospective study of nonmyeloablative, HLA-mismatched unrelated BMT with high-dose posttransplantation cyclophosphamide. Blood Adv. 2017;1:288–292.
    1. Gragert L, Eapen M, Williams E, et al. HLA match likelihoods for hematopoietic stem-cell grafts in the U.S. registry. N Engl J Med. 2014;371:339–348.
    1. Dehn J, Chitphakdithai P, Shaw BE, et al. Likelihood of proceeding to allogeneic hematopoietic cell transplantation in the United States after search activation in the National Registry: impact of patient age, disease, and search prognosis. Transplant Cell Ther. 2021;27. 184.e1–184.e13.
    1. Alessandrino EP, Della Porta MG, Malcovati L, et al. Optimal timing of allogeneic hematopoietic stem cell transplantation in patients with myelodysplastic syndrome. Am J Hematol. 2013;88:581–588.
    1. Koreth J, Pidala J, Perez WS, et al. Role of reduced-intensity conditioning allogeneic hematopoietic stem-cell transplantation in older patients with de novo myelodysplastic syndromes: an international collaborative decision analysis. J Clin Oncol. 2013;31:2662–2670.
    1. Ustun C, Lazarus HM, Weisdorf D. To transplant or not: a dilemma fortreatment of elderly AML patients in the twenty-first century. Bone Marrow Transplant. 2013;48:1497–1505.
    1. Ferrebee JW, Atkins L, Lochte HL Jr, et al. The collection, storage and preparation of viable cadaver marrow for intravenous use. Blood. 1959;14:140–147.
    1. Burke GW, Ricordi C, Karatzas T, et al. Donor bone marrow infusion in simultaneous pancreas/kidney transplant recipients: a preliminary study. Transplant Proc. 1995;27:3121–3122.
    1. Burke GW, Ricordi C, Karatzas T, et al. Donor bone marrow infusion insimultaneous pancreas/kidney transplantation with OKT3 induction: evidence for augmentation of chimerism. Transplant Proc. 1997;29:1207–1208.
    1. Carroll PB, Fontes P, Rao AS, et al. Simultaneous solid organ, bone marrow, and islet allotransplantation in type I diabetic patients. Transplant Proc. 1994;26:3523–3524.
    1. Ciancio G. Donor bone marrow infusion in cadaveric renal transplantation. Transplant Proc. 2003;35:871–872.
    1. Corry RJ, Chakrabarti PK, Shapiro R, et al. Simultaneous administration of adjuvant donor bone marrow in pancreas transplant recipients. Ann Surg. 1999;230:372–379. [discussion: 379–381].
    1. De Pauw L, Abramowicz D, Donckier V, et al. Isolation and infusion of donor CD34+ bone marrow cells in cadaver kidney transplantation. Nephrol Dial Transplant. 1998;13:34–36.
    1. Fontes P, Rao AS, Demetris AJ, et al. Bone marrow augmentation of donor-cell chimerism in kidney, liver, heart, and pancreas islet transplantation. Lancet. 1994;344:151–155.
    1. Pham SM, Rao AS, Zeevi A, et al. A clinical trial combining donor bone marrow infusion and heart transplantation: intermediate-term results. J Thorac Cardiovasc Surg. 2000;119(4 Pt 1):673–681.
    1. Pham SM, Rao AS, Zeevi A, et al. Effects of donor bone marrow infusion in clinical lung transplantation. Ann Thorac Surg. 2000;69:345–350.
    1. Rao AS, Fontes P, Iyengar A, et al. Augmentation of chimerism with perioperative donor bone marrow infusion in organ transplant recipients: a 44-month follow-up. Transplant Proc. 1997;29:1184–1185.
    1. Rao AS, Fontes P, Zeevi A, et al. Enhancement of donor cell chimerism in whole organ allograft recipients by adjuvant bone marrow transplantation. Transplant Proc. 1995;27:3387–3388.
    1. Rao AS, Fontes P, Zeevi A, et al. Augmentation of chimerism in whole organ recipients by simultaneous infusion of donor bone marrow cells. Transplant Proc. 1995;27:210–212.
    1. Schneeberger S, Gorantla VS, Brandacher G, et al. Upper-extremity transplantation using a cell-based protocol to minimize immunosuppression. Ann Surg. 2013;257:345–351.
    1. Woods EJ, Sherry AM, Woods JR, et al. Ischemia considerations for the development of an organ and tissue donor derived bone marrow bank. J Transl Med. 2020;18:300.
    1. Acker JP, Bondarovych M, Brunotte R, et al. Preservation and storage of cells for therapy: current applications and protocols. In: Gimble JM, ed. Cell Engineering and Regeneration. Reference Series in Biomedical Engineering. Cham, Switzerland: Springer; 2022:27–30.
    1. Sutherland DR, Anderson L, Keeney M, Nayar R, Chin-Yee I. The ISHAGE guidelines for CD34+ cell determination by flow cytometry. International Society of Hematotherapy and Graft Engineering. J Hematother. 1996;5:213–226.
    1. Shultz LD, Lyons BL, Burzenski LM, et al. Human lymphoid and myeloid cell development in NOD/LtSz-scid IL2R gamma null mice engrafted with mobilized human hemopoietic stem cells. J Immunol. 2005;174:6477–6489.
    1. Gelman A. Scaling regression inputs by dividing by two standard deviations. Stat Med. 2008;27:2865–2873.
    1. Arnaud F, Yang H, McGann LE. Freezing injury of granulocytes during slow cooling: role of the granules. Cryobiology. 1996;33:391–403.
    1. Shultz LD, Saito Y, Najima Y, et al. Generation of functional human T-cell subsets with HLA-restricted immune responses in HLA class I expressing NOD/SCID/IL2r gamma(null) humanized mice. Proc Natl Acad Sci U S A.. 2010;107:13022–13027.
    1. Carpenter DJ, Granot T, Matsuoka N, et al. Human immunology studies using organ donors: impact of clinical variations on immune parameters in tissues and circulation. Am J Transplant. 2018;18:74–88.
    1. Blazar BR, Lasky LC, Perentesis JP, et al. Successful donor cell engraftment in a recipient of bone marrow from a cadaveric donor. Blood. 1986;67:1655–1660.
    1. Pham SM, Keenan RJ, Rao AS, et al. Perioperative donor bone marrow infusion augments chimerism in heart and lung transplant recipients. Ann Thorac Surg. 1995;60:1015–1020.
    1. Biernacki MA, Sheth VS, Bleakley M. T cell optimization for graft-versus-leukemia responses. JCI Insight. 2020;5: e134939.
    1. Elmariah H. Commentary: Target CD34 cell dose for allogeneic hematopoietic cell transplantation: can we finally agree? Transplant Cell Ther. 2022;28:59–60.
    1. Saad A, Lamb L, Wang T, et al. Impact of T cell dose on outcome of T cell-replete HLA-matched allogeneic peripheral blood stem cell transplantation. Biol Blood Marrow Transplant. 2019;25:1875–1883.
    1. Alousi A, Wang T, Hemmer MT, et al. Peripheral blood versus bone marrow from unrelated donors: bone marrow allografts have improved long-term overall and graft-versus-host disease-free, relapse-free survival. Biol Blood Marrow Transplant. 2019;25:270–278.
    1. Fernandez-Sojo J, Azqueta C, Valdivia E, et al. Cryopreservation of unrelated donor hematopoietic stem cells: the right answer for transplantations during the COVID-19 pandemic? Bone Marrow Transplant. 2021;56:2489–2496.
    1. Kanda Y, Inoue M, Uchida N, et al. Cryopreservation of unrelated hematopoietic stem cells from a blood and marrow donor bank during the COVID-19 pandemic: a nationwide survey by the Japan Marrow Donor Program. Transplant Cell Ther. 2021;27. 664.e1–664.e6.
    1. Bankova AK, Caveney J, Yao B, et al. Real-world experience of cryopreserved allogeneic hematopoietic grafts during the COVID-19 pandemic: a single-center report. Transplant Cell Ther. 2022;28. 215.e1–215.e10.
    1. Valentini CG, Chiusolo P, Bianchi M, et al. Coronavirus disease 2019 pandemic and allogeneic hematopoietic stem cell transplantation: a single center reappraisal. Cytotherapy. 2021;23:635–640.
    1. Mfarrej B, Lemarie C, Granata A, et al. Related versus unrelated allogeneic HPC graft cryopreservation: a single-center experience in the context of the global COVID-19 pandemic. Bone Marrow Transplant. 2021;56:2013–2015.
    1. Alotaibi AS, Prem S, Chen S, et al. Fresh vs. frozen allogeneic peripheral blood stem cell grafts: a successful timely option. Am J Hematol. 2021;96:179–187.
    1. Aziz J, Morris G, Rizk M, et al. Cryopreservation of adult unrelated donor products in hematopoietic cell transplantation: the OneMatch experience and systematic review of the literature. Transfusion. 2017;57:2782–2789.
    1. Hsu JW, Farhadfar N, Murthy H, et al. The effect of donor graft cryopreservation on allogeneic hematopoietic cell transplantation outcomes: a Center for International Blood and Marrow Transplant Research analysis. Implications during the COVID-19 pandemic. Transplant Cell Ther. 2021;27:507–516.
    1. Hamadani M, Zhang MJ, Tang XY, et al. Graft cryopreservation does not impact overall survival after allogeneic hematopoietic cell transplantation using post-transplantation cyclophosphamide for graft-versus-host disease prophylaxis. Biol Blood Marrow Transplant. 2020;26:1312–1317.
    1. Wiercinska E, Schlipfenbacher V, Bug G, et al. Allogeneic transplant procurement in the times of COVID-19: quality report from the central European cryopreservation site. J Transl Med. 2021;19:145.
    1. Knebel AR, Coleman CN, Cliffer KD, et al. Allocation of scarce resources after a nuclear detonation: setting the context. Disaster Med Public Health Prep. 2011:(5 suppl 1):S20–S31.
    1. Waselenko JK, MacVittie TJ, Blakely WF, et al. Medical management of the acute radiation syndrome: recommendations of the Strategic National Stockpile Radiation Working Group. Ann Intern Med. 2004;140:1037–1051.

Source: PubMed

3
Subskrybuj