Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Nov 1.
Published in final edited form as: Curr Opin Hematol. 2022 Aug 29;29(6):317–326. doi: 10.1097/MOH.0000000000000732

Umbilical Cord Blood: An Undervalued and Underutilized Resource in Allogeneic Hematopoietic Stem Cell Transplant and Novel Cell Therapy Applications

Patricia A Shi 1, Larry L Luchsinger 1, John M Greally 2, Colleen S Delaney 3,4
PMCID: PMC9547826  NIHMSID: NIHMS1819150  PMID: 36066376

Abstract

Purpose of review:

The purpose of this review is to primarily discuss the unwarranted decline in the use of umbilical cord blood (UCB) as a source of donor hematopoietic stem cells (HSC) for hematopoietic cell transplantation (HCT) and the resulting important implications in addressing health care inequities, and secondly to highlight the incredible potential of UCB and related birthing tissues for the development of a broad range of therapies to treat human disease including but not limited to oncology, neurologic, cardiac, orthopedic and immunologic conditions.

Recent findings:

When current best practices are followed, unrelated donor umbilical cord blood transplant (CBT) can provide superior quality of life-related survival compared to other allogeneic HSC donor sources (sibling, matched or mismatched unrelated, and haploidentical) through decreased risks of relapse and chronic graft versus host disease. Current best practices include improved UCB donor selection criteria with consideration of higher resolution HLA typing and CD34+ cell dose, availability of newer myeloablative but reduced toxicity conditioning regimens, and rigorous supportive care in the early post-transplant period with monitoring for known complications, especially related to viral and other infections that may require intervention. Emerging best practice may include the use of ex vivo expanded single-unit CBT rather than double-unit CBT (dCBT) or “haplo-cord” transplant, and the incorporation of post-transplant cyclophosphamide as with haploidentical transplant and/or incorporation of novel post-transplant therapies to reduce the risk of relapse, such as NK cell adoptive transfer. Novel, non-HCT uses of UCB and birthing tissue include the production of UCB-derived immune effector cell therapies such as unmodified NK cells, CAR-NK cells and immune T-cell populations, the isolation of mesenchymal stem cells for immune modulatory treatments and derivation of iPSC haplobanks for regenerative medicine development and population studies to facilitate exploration of drug development through functional genomics.

Summary:

The potential of allogeneic UCB for HCT and novel cell-based therapies is undervalued and underutilized. The inventory of high-quality UCB units available from public cord blood banks (CBB) should be expanding rather than contracting in order to address ongoing health care inequities and to maintain a valuable source of cellular starting material for cell and gene therapies and regenerative medicine approaches. The expertise in GMP-grade manufacturing provided by CBB should be supported to effectively partner with groups developing UCB for novel cell-based therapies.

Keywords: allogeneic, umbilical cord blood, transplant, haplobank, gene therapy, immune effector cells, cord blood expansion, haplo-cord, immunotherapy, HLA, induced pluripotent stem cells, functional genomics, haplobank

Introduction

UCB is a well-established source of allogeneic donor HSCs for hematopoietic cell transplantation (HCT) (in this case, referred to as cord blood transplant (CBT)). However, despite known advantages of UCB as a donor source for HCT and the availability of optimal practice guidelines for unrelated donor CBT [15] as well as the equivalency and even potential superiority of some outcomes observed after CBT compared to convention donor HCT (peripheral blood (PB) or bone marrow (BM))[68],** the number of CBTs performed annually has been declining in favor of haploidentical transplant [9,10] when a suitably matched related or unrelated adult volunteer donor cannot be identified. In addition, UCB and related birthing tissue are invaluable as starting cellular material for the generation of innovative cell therapies and regenerative medicines across multiple indications. This paper describes ways in which UCB is an undervalued resource for both standard hematopoietic stem cell (HSC) allogeneic transplant and for novel cell therapy applications.

Overview of HSC sources for Allogeneic Transplant

In allogeneic HCT for both malignant and non-malignant diseases, the optimal donor source depends on level of HLA-matching between the donor and recipient, cell dose, urgency of the transplant (i.e., underlying disease), and donor availability. An 8/8 HLA-matched sibling donor (MSD)-sourced from peripheral blood (PB), bone marrow (BM), generally remains the standard of care [11] because of its ability to rapidly meet all the above donor selection criteria without concern for cell dose and with reduced risk of graft versus host disease (GVHD). However, only approximately 30% of allogeneic transplant candidates have an HLA-matched sibling [12]. An 8/8 or 10/10 matched unrelated donor (MUD) is typically the next choice in line [13] due to meeting HLA and cell dose requirements, although center transplant experience may favor UCB [14] and HLA matches for ethnicities other than whites of European descent [12], donor availability, and time to transplant can be limiting factors.

Next in line for consideration are typically HLA-haploidentical relatives or banked unrelated UCB over an HLA-mismatched unrelated donor [6**,15]. Both are rapidly available sources of HSC typically, but have their relative advantages and disadvantages. In terms of HLA matching, the sources are thought to be similar, as UCB has relaxed HLA-matching requirements of 4/6 (at least one allele matched at HLA-A, -B, and -DRB1) without increased risk of GVHD due to fewer CD3+ T-cells and their immunologic naivete[16,17]. Use of haploidentical relatives is possible due to removal of CD3+ T-cells from the graft through in vitro positive or negative selection or more commonly now in vivo post-transplant cyclophosphamide [18]. In terms of HSC dose, which affects the kinetics of hematopoietic recovery (engraftment) [19,20], haploidentical transplant is the superior choice, and is one reason for the declining use of UCB transplant in favor of haploidentical transplant [11]. Nevertheless, it is worth considering that UCB HSCs compared to adult HSCs have longer telomeres, less DNA damage and higher proliferative and hematopoietic reconstitutive potential [21,22], which may have safety implications in regard to the replicative stress of HSC transplant[23]*. Finally, in regard to urgency of transplant and donor availability, UCB is already banked and HLA-typed and is thus the most readily available donor source and enabling of a flexible transplantation date. Furthermore, ~12% of patients have no HLA-haploidentical relative [24] and suitable relatives may be unviable donors due to the presence of anti-HLA antibodies in the recipient [24,25], their ineligibility or refusal to donate[26], or medical or psychological conditions that preclude donation[27]. Such considerations likely contribute to the continued demand for UCB units, particularly for pediatric patients and with inherited metabolic conditions, congenital leukodystrophies, and immune deficiency syndromes, where age at the time of transplant and time to transplant affect long-term outcomes. Furthermore, relatives may be carriers of the genetic disorder[2831].

Special Immune Characteristics of UCB HSCs for Allogeneic Transplant

When HCT is being utilized as the curative approach for hematologic malignancies, HLA-mismatched UCB donor T-cells have characteristics that may lead to a reduced risk of relapse as compared to MSD [32], MUD [6],** or haploidentical transplant [33], related to enhanced anti-tumor ability and cytotoxic CD8+ and CD4+ T-helper (Th1) profiles[34]. A higher CD8+ UCB content is associated with faster and higher myeloid and platelet engraftment, lower non-relapse mortality (NRM), lower relapse, and increased overall survival (OS).[35] A recent publication by Hiwarkar, et al., reports an important and very interesting finding wherein the normally delayed CD8+ T-cell recovery with UCB transplant can be reversed with granulocyte transfusion and was associated with eradication of refractory leukemia[36];** the mechanism is thought to be neutrophil cross-priming of naïve CD8+ T-cells that is as efficient as that by macrophages[37]. UCB units also have increased immature NK cells which are hyper-responsive, with similar secretion levels of perforin and granzyme B as mature NK cells[38].

Relevant in either malignant or non-malignant disease, UCB has characteristics which contribute to a relatively low incidence of chronic graft versus host disease (cGVHD) compared to haploidentical [39], MUD [40,41] and even MSD [7*,32] HSC sources which should favorably impact long-term quality of life and survival. For example, UCB is enriched in plasmacytoid dendritic cells (pDC), which unlike PB pDC, express low levels of TNFα [42]. Also possibly contributing to the low incidence of cGVHD is more rapid and higher recovery of naïve B, IgM and switched memory B cells in comparison to BM and PB, as naïve B cell recovery is critical for deletion of self-reactive B cells [38].

Immune reconstitution, however, can be delayed due to the immunologic naivete of UCB T-cells, which do not immediately confer protective T-cell memory function, and the high number of regulatory T-cells[17], resulting in increased risk of post-transplant infectious complications, especially viral. In this regard, it is best to avoid ATG conditioning as it damages immune reconstitution of both CD4+ and CD8+ T-cells, which in turn worsens NRM and relapse[43,44]. ATG omission also simplifies considerations with KIR ligand incompatibility [45]and is associated with rapid early thymus-independent CD4+ T-cell recovery in both children and adults without increase in aGVHD [46,47]. ATG omission is also associated with decreased cGVHD compared to ≥ 8/10 HLA-matched unrelated donors [48] and decreased risk of relapse[49].

The intricacies of UCB as the donor HSC source in Allogeneic Transplant

Given the need to choose between unrelated UCB versus haploidentical related donor sources, it is worthwhile to compare the safety and efficacy of their HSC recipient outcomes. There is no prospective randomized comparison in non-malignant disease, but in hematologic malignancy (leukemia and lymphoma) a prospective multi-center Phase III trial, BMT CTN 1101, randomized patients between reduced-intensity dCBT and haploidentical BM transplant. It found CBT recipients to have higher NRM (18% vs 11%, p=0.04) leading to lower 2-year overall survival (OS) (46% vs 57%, p=0.04) [26]. This NRM rate is similar to that reported at a highly experienced UCB transplant center [8]. Higher NRM in CBT vs haploidentical transplant recipients was also observed in a small (n=45) but randomized trial of myeloablative transplant for leukemia/MDS[24], a retrospective CIBMTR data comparison of 708 hematologic malignancy patients who had myeloablative conditioning [33], a large retrospective study of non-myeloablative transplant for lymphoma [50], and a recent meta-analysis[39]. BMT CTN 1101 found no differences in acute graft versus host disease (aGVHD), 2-year chronic graft versus host disease (cGVHD), 2-year relapse/progression, or 2-year progression free survival [26], but the meta-analysis [39] found a significant reduction in cGVHD with CBT vs haploidentical transplant.

There are various issues that need to be considered when interpreting the higher NRM with UCB. First, as previously discussed, ATG affects NRM and was used in one trial[24]. Second, dCBT predominated in the other studies and it must be recognized that single-donor CBT with adequate cell dose (e.g. ≥2.5 × 107 total nucleated cells (TNC)/kg and ≥1.5–2.0 × 105 CD34+ cells/kg [4]) remains the standard of care, with double-UCB transplantation only appropriate for patients who lack a single unit with adequate cell dose [5,29,51]. This is due to higher rates of Grade III-IV aGVHD and Grade III-IV cGVHD and delayed platelet recovery with double versus single CBT [5254].** In this light, it is worth considering that for overweight and obese patients, a recent study of single-unit CBT found comparable hematopoietic recovery and overall survival when dosing CD34+ cells/kg by ideal body weight rather than actual body weight [55].

Third, although increasing CD34+ cell dose may overcome the problem [8], HLA mismatch contributes to NRM in CBT. Indeed, the greater the allele level matching at HLA-A, -B, -C, and -DRB1, the lower the NRM and graft failure for malignant and non-malignant disease, respectively, and the higher the OS [56*,57], with >2 of 8 HLA-allele mismatch associated with significantly prolonged neutrophil recovery independent of cell dose [56].* Only 2 HLA mismatches or single-allele mismatches at HLA-A, -B, and -DRB1 can increase the risk of Grade III-IV aGVHD [58,59]. Furthermore, mismatch at HLA locus C is associated with higher NRM [60]. Thus, it is worth considering that, although the aforementioned trials provided adequate cell doses (e.g. in BMT CTN 1101, each UCB unit had to contain at least 1.5 × 107 TNC/recipient kg), the HLA matching requirement was the minimum 4/6, and not all trials required high resolution typing at all loci [4], although BMT CTN 1101 required at least one allele match at HLA-A, HLA-B, and HLA-DRB1. It is therefore not surprising that these trials found higher NRM. For example, in the BMT CTN 1101 trial mortality from pulmonary or intracranial hemorrhage and viral or bacterial infection was higher in the UCB group, as would be expected from a platelet recovery of a median 42 versus 28 days and incidence of neutrophil recovery at day 56 of 95% versus 99% (p=0.05).

Fourth, thymic-dependent de novo production of T-cells is critical for long-term reconstitution of a broad and diverse T-cell repertoire as measured by recent thymic emigrants and T-cell receptor excision circles. [17,61] HLA mismatch can lead to donor-T cell induced thymic dysfunction [62,63], as suggested by better T-cell diversity after MSD than MUD transplant [64].

The data presented suggests that using more closely HLA-matched UCB units is likely to assuage the increased risk of NRM with CBT. However, this must be balanced with the positive effect of mismatched UCB donors and reduced risk of relapse [65]. Considering this, a 7/8 HLA match may be best over an 8/8 HLA-matched UCB donor, especially in patients with high risk leukemia placing them at higher risk of post-transplant relapse Importantly however, there is no clear evidence in UCB transplant that 6–8/8 HLA matching increases relapse risk compared to ≤ 5/8 HLA matching [8,33]. Furthermore, although the frequency of NIMA matching is estimated at < 10% [66], when the recipient’s mismatched HLA antigen matches the non-inherited maternal HLA antigens (NIMA) of the UCB donor, such NIMA-matched recipients have greater neutrophil recovery, reduced NRM and improved overall survival compared to non-NIMA matched UBC recipients [67].

Thus, rather than transplant centers replacing CBT with haploidentical donor transplant, which will undoubtedly reduce critical physician training and transplant center experience with CBT, not to mention endangering the survival of public CBBs and the availability of high-quality UCB units, UCB collection should be expanded and diversified in order to provide the closer recipient HLA matching needed to improve their hematopoietic recovery and immune reconstitution. Importantly, ongoing UCB collection and banking is absolutely essential to establish and maintain availability of HSC donors that reflect the increasing genetic diversity and racial admixture of the population. Without this, future patients requiring HCT as a curative approach may not have any suitable unrelated donors available. It is also worth considering that the greater a center’s experience with CBT, the lower the NRM [58,68,69], with at least 10 CBT per year qualifying as sufficient expertise. Indeed, in the hands of experienced CBT centers, GVHD-free and relapse free survival, which is a better measure of quality of life than OS [70], is better as compared to MUD or even MSD transplant[7*,14,32].

Solutions to the higher NRM with CBT

The issue of the low cell dose in UCB units needs to be addressed herein [71]. Low cell dose, especially CD34+ cell dose, which can result in delayed kinetics of hematopoietic recovery [19,20] and thus NRM and OS[8], has resulted in the common use of dCBT to ensure adequate cell dose and engraftment [66]. This is especially critical given that ACOG has also made clear that delayed CB clamping should be routine standard of care [72] which will impact the numer of collected UCB units that will meet cell dose thresholds for processing and storage in the CBBs. Yet, importantly, there has been significant improvement in time to hematopoietic recovery observed following CBT simply due to higher quality CBUs available in the CBBs, the consideration of high-resolution HLA-typing and CD34+ cell dose[3,56*,73]

Another solution may be the “haplo-cord” approach, in which mobilized CD34+ cells isolated from a haploidentical donor, who may be related or unrelated[74], are infused with a single UCB unit in order to provide faster neutrophil and platelet recovery than is typically obtained from a single UCB unit transplant alone. ATG conditioning, however, is required for in-vivo T-cell depletion of the UCB to prevent rejection of the CD34+ selected (and therefore T-cell depleted) haploidentical HSCs. [75,76] A retrospective study comparing this approach to MUD transplants showed no significant differences in transplant outcomes [77]. A retrospective study comparing it to haploidentical BM, however, showed haplo-cord to have faster neutrophil and platelet recovery and lower cGVHD at 1 year (4% versus 16%, p < .0001); there were no differences in OS, progression-free survival (PFS), relapse, or NRM including Grade III-IV aGVHD [78]. These findings support the previous meta-analysis showing reduction in cGVHD with UCB [39]. An important study will be prospective comparison of haploidentical PB to haplo-cord transplant, since haploidentical mobilized PB versus haploidentical BM may lower relapse risk but possibly at the cost of higher Grade III-IV aGVHD and cGVHD[33,7981].

A potential impediment to the “haplo-cord” approach is the cost associated with haploidentical CD34+ selection. A different “haplo-cord” approach is to infuse unmanipulated mobilized PB from a haploidentical donor along with a low-dose UCB unit (median UCB MNC= 1.8 × 107/kg)[82]. Median time to engraftment and 30 day engraftment rate showed no differences compared to a concurrent MUD cohort. This approach resulted in engraftment of the haploidentical donor HSCs rather than the UCB unit, with a lower relapse risk than in the MUD cohort. Yet another important and cost-saving study to reduce NRM with UCB transplant is the effect of using, as with haploidentical donor transplant, post-transplant cyclophosphamide (PT-CY) (NCT03802773) for aGVHD prophylaxis [83], given the immune reconstitution issues with ATG. [43,44] Use of PT-CY may also help retain anti-viral immunity, which is mediated by persisting virus-specific recipient T-cells [84].

Although also likely expensive and with increased logistical difficulty and potential delays in time to transplant, infusion of ex-vivo expanded UCB CD34+ cells from one UCB unit along with an unmanipulated UCB unit or as a stand alone graft also appears to be an effective method to address delayed engraftment with UCB and can be done by several methods, such as co-culture with Notch ligand [85], mesenchymal stromal cells [86], aryl hydrocarbon receptor antagonist [87], or nicotinamide. [87] Recently, both nicotinamide-expanded and UM171-expanded grafts were successfully used as stand-alone single UCB unit transplants. [8890] This is possible as the negative fraction obtained when isolating the CD34+, which contains the donor T-cells, is re-cryopreserved, and infused at the time of transplant with the expanded cell graft. A randomized Phase III trial of nicotinamide-expanded versus standard unmanipulated 1–2 unit UCB transplant using myeloablative conditioning for heme malignancy found the nicotinamide transplant to result in faster neutrophil and platelet recovery and reduced grade 2–3 bacterial or invasive fungal infection[89].* A helpful follow-up trial would be to compare this product to haploidentical transplant or the haplo-cord approach which does not require manipulation/ex vivo expansion and therefore does not delay time to transplant. Finally, co-infusion of additional UCB cell types has been explored, such as UCB-derived and expanded regulatory T-cells to reduce the risk of aGVHD and cGVHD[91], and in a pre-clinical model, UCB mesenchymal stem cells (MSC) to promote engraftment of non-expanded UCB HSCs.[92]

UCB as a source material for generation of iPSC haplobanks

Pluripotent stem cells (PSCs), including embryonic stem cells (ESCs), have the ability to self-renew and give rise to all types of cells in the body. The discovery of factors that can reprogram adult cells into induced pluripotent stem cells (iPSCs) sidesteps the ethical controversy surrounding ESCs and has therefore made an invaluable contribution to the field of regenerative medicine. In theory, sourcing and scaling of iPSCs for mass production of cellular teraputics for clinical trials makes the potential clinical utility virtually unlimited. Furthermore, since sampling of primary tissues from living donors at the scale necessary to study large cohorts of patients is not tenable, in vitro differentiation of iPSCs into mature cell types have the potential to generate in vitro representations for disease modeling and drug validation.[93] Ultimately, the use of iPSCs for tissue engineering and cellular replacement therapies could revolutionize regenerative medicine. The first published clinical trial utilizing iPSCs explored the safety and efficacy of iPSC-derived mesenchymal stromal cells in reducing inflammation associated with graft-versus-host disease.[94]* Currently, 19 clinical trials are testing the therapeutic value of iPSC-derived cells worldwide, including a trial for iPSC-derived platelets for treatment of thrombocytopenia in alloimmune patients with aplastic anemia.[95]

The success of iPSC therapies relies on identifying high quality adult somatic cells as well as the maintaining the reprogamming and directed differentiation of iPSCs in a fully Good Manufacturing Practice (GMP)-compliant environment such that they can be used safely and effectively in the clinical setting. Despite the potential of iPSCs to generate cellular therapeutics, practical issues related to the time and expense needed to generate autologous (i.e., patient-derived) iPSCs have cast uncertainty as to their practicality in the clinical environment. Alternatively, allogeneic (i.e., unrelated) iPSC ‘haplobanks’ could overcome the challenges of quality control and availability[96]. However, the complexity of immune matching allogeneic iPSCs to a diverse population is a major challenge.

UCB units are an excellent source of somatic cells for creation of allogeneic iPSCs lines for a number of reasons. First, HLA typing of UCB allows for identification of units with optimal immunotolerance properties best suited for allogeneic therapeutics: mainly O blood type and homozygous HLA haplotypes. In this way, HLA homozygote donors can match a significant numbers of recipients using a relatively small number of well selected donors that are representative of the population. Second, CBUs are manufactured under GMP-conditions and have been screened for infectious diseases, which ensures the source of the somatic cells are of the highest quality. Third, UCB cells have less somatic mutation and exposure to environmental damage, and can be reprogrammed into iPSCs with high efficiency[97].* In fact, precedent for the repurposing of CBU for iPSC line derivation exists and several iPSC haplobanks have been successfully created using UCB. [98,99] Thus, UCB cells represent a novel and attractive resource for the purposes of iPSC production towards platforms for drug discovery as well as regenerative and cellular therapy manufacturing.

Importance of the racial-ethnic diversity of UCB to health care equity

The American College of Obstetricians and Gynecologists (ACOG) has emphasized the importance of public over private CBB and contribution from all ethnicities and races.[72] Even though racial and ethnic minority groups are underrepresented in UCB registries [100],* they are still better represented than in volunteer adult unrelated donor registries despite the expansion of such registries. [12,101103] Public cord blood banks are therefore critical resources for health equity. UCB is a critical option for ethnic minorities and mixed race individuals even with a haplo-identical relative, as 65% of recipients of African ancestry have a suitable UCB graft versus 44% having a suitable haplo donor.[27] Even though UCB inventories should be expanded and diversified to provide closer HLA matching, the less stringent HLA matching requirement compared to other HSC sources allows for ethnicity difference between donor and recipient. Indeed, a person of mixed race appears to be cured of HIV using a single partially matched UCB unit from a donor with a CCR5 co-receptor variant, which is more common in people of European heritage; furthermore, the patient has had no GVHD in the 4 years since the transplant, despite HLA mismatch. [104,105]

In the setting of autologous gene therapy for sickle cell disease (SCD), collection of autologous UCB is a potential critical resource for health care equity because HSC mobilization and collection is currently not feasible in much of Africa, where SCD is most prevalent. Exploring this potential resource is critical because in allogeneic transplant for SCD, HSC donor sources other than the rare 8/8 HLA-matched sibling are plagued by high rates of graft failure and cGVHD. [106,107]

In the setting of genetic studies into the biology of complex traits and drug development, genetic variation between ethnicities contributes significantly to disease manifestations [108] and response to potential treatments. Thus, efforts to remediate the disproportionate focus in genetic research on individuals with Northern European white ancestry, including the creation of iPSC from ethnic minority CB units, are critical.[109112]* This is a pressing need, as iPSCs are a platform for modeling disease and drug discovery, so including diverse and under-represented groups in iPSC research enables the application of genetic findings equally across all races and ancestries. For example, only rarely do genome-wide association studies (GWAS) deliver a result indicating a sequence change within a gene as the association with the disease. Instead, GWAS has revealed the importance of sequence variation at the non-coding regulatory loci between genes or within introns. Understanding which how these non-coding variants influence gene expression is the focus of the field of functional genomics. Indeed, it has already been shown that the use of samples from individuals with diverse ancestries in functional genomics studies improves our ability to identify functional variants in the non-coding majority of the genome.[113115*]

Finally, for the purpose of iPSC haplobank creation, unsurprisingly the coverage provided by homozygous HLA haplobanks across a particular population is proportional to the diversity of the CB bank demographic.[98] Thus, rigorous cross-referencing of HLA haplotypes present in the intended target population with chosen CBUs for reprogramming will improve the feasibility of homozygous HLA haplobanks for clinical application.

Importance of UCB and Cord Blood Banks for Other Innovative Cell Therapies

UCB is not just a source of stem cells for HCT and iPSC haplobanks, but UCB and birthing tissue are also an important source of starting material for other cellular therapy applications. For example, UCB-derived HLA-mismatched CAR NK cells are an effective alternative to CAR NK cells due to their better side effect profile. [116]** UCB-derived polyclonal multivirus-specific cytotoxic T-cells are being developed for viral infections in immunosuppressed states.[117] In neonates, UCB and CB tissue is being explored in lung, brain, and cardiac injury and disease.[118] UCB-derived myeloid-derived suppressor cells, which have immune suppressive effects, are being explored for treatment of feto-maternal intolerance, GVHD, and autoimmunity.[119] These are just a few examples of why CBB must be supported as a key foundation for innovative cell therapies. CBB are regulated and have all the required infrastructure to manufacture high quality starting material, and to lose these invaluable resources would impact innovation and slow progress.

Conclusion

UCB is an undervalued and underutilized source for both standard allogeneic HSC transplant and novel cell therapy indications. Compared to adult HSC sources, it is the most readily available HSC source with unique immune properties and the least replicative stress damage, which can lead to the lowest risks of relapse, cGVHD, and possibly post-transplant hematologic malignancy. The higher risk of non-relapse mortality observed in allogeneic HSC transplant can be overcome by considerations such as ATG alternatives, better HLA matching, use of expanded single UCB units, and center experience. UCB is also an ideal source for iPSC generation for functional genomic studies and haplobanks for regenerative medicine. Last but not least, it is a critical resource for health care equity in regard to standard allogeneic transplant, autologous gene therapy, regenerative medicine applications, and drug development through functional genomics.

Key points.

  1. UCB compared to adult HSC sources have the advantage of having undergone less replicative stress and are already banked and HLA-typed, and are thus the least age-affected and most readily available and transplant date-flexible HSC source.

  2. HLA-mismatched UCB transplant can result in lower relapse and lower chronic GVHD compared to adult HSC sourced from matched sibling donors, matched unrelated donors, and haploidentical donors.

  3. The observed increase in non-relapse mortality with UCB transplant must be considered in the light of the increased non-relapse mortality risks with ATG conditioning with its suboptimal risk-to benefit-ratio; two versus one (expanded or not) UCB unit; and previous minimum requirements of only a 4 out of 6 HLA match.

  4. In regard to the contribution of UCB cell dose to non-relapse mortality, the future is bright with multiple existing and evolving solutions including haplo-cord options, post-transplantation cyclophosphamide, UCB unit expansion, and co-infusion of other CB-derived cell populations.

  5. Its immunotolerant properties, high iPSC reprogramming efficiency, and easy availability make UCB an ideal source for haplobank creation.

  6. Health care equity demands that public cord blood bank inventory be expanded and diversified in order to provide higher degrees of HLA matching for allogeneic transplant and iPSCs for functional genomics studies and haplobanks.

Acknowledgements

All authors collaboratively drafted and made final edits to the manuscript.

Financial support and sponsorship:

This work was supported in part by the National Institutes of Health, National Heart, Lung, and Blood Institute R01HL155574 (L.L.).

Footnotes

Conflicts of interest: New York Blood Center has an FDA-licensed cord blood bank. Deverra Therapeutics is currently developing UCB-derived allogeneic cell therapies.

References

  • 1.Barker JN, Kurtzberg J, Ballen K, Boo M, Brunstein C, Cutler C, Horwitz M, Milano F, Olson A, Spellman S, et al. : Optimal Practices in Unrelated Donor Cord Blood Transplantation for Hematologic Malignancies. Biol Blood Marrow Transplant 2017, 23:882–896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Little AM, Akbarzad-Yousefi A, Anand A, Diaz Burlinson N, Dunn PPJ, Evseeva I, Latham K, Poulton K, Railton D, Vivers S, et al. : BSHI guideline: HLA matching and donor selection for haematopoietic progenitor cell transplantation. Int J Immunogenet 2021, 48:75–109. [DOI] [PubMed] [Google Scholar]
  • 3.Fatobene G, Volt F, Moreira F, Mariano L, Chevallier P, Furst S, Labussiere-Wallet H, de la Tour RP, Deconinck E, Cluzeau T, et al. : Optimizing selection of double cord blood units for transplantation of adult patients with malignant diseases. Blood Adv 2020, 4:6327–6335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Dehn J, Spellman S, Hurley CK, Shaw BE, Barker JN, Burns LJ, Confer DL, Eapen M, Fernandez-Vina M, Hartzman R, et al. : Selection of unrelated donors and cord blood units for hematopoietic cell transplantation: guidelines from the NMDP/CIBMTR. Blood 2019, 134:924–934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Hough R, Danby R, Russell N, Marks D, Veys P, Shaw B, Wynn R, Vora A, Mackinnon S, Peggs KS, et al. : Recommendations for a standard UK approach to incorporating umbilical cord blood into clinical transplantation practice: an update on cord blood unit selection, donor selection algorithms and conditioning protocols. Br J Haematol 2016, 172:360–370. [DOI] [PubMed] [Google Scholar]
  • **6.Milano F, Gooley T, Wood B, Woolfrey A, Flowers ME, Doney K, Witherspoon R, Mielcarek M, Deeg JH, Sorror M, et al. : Cord-Blood Transplantation in Patients with Minimal Residual Disease. N Engl J Med 2016, 375:944–953. [DOI] [PMC free article] [PubMed] [Google Scholar]; This retrospective analysis comparing UCB, MUD, and mimatched unrelated donors found that in acute leukemia or MDS patients with minimal residual disease undergoing myeloalative transplant, UCB compared to MUD lowered the risk of relapse and trended towards increased OS. UCB was also superior to mismatched unrelated donors in these respects.
  • 7*.Sharma P, Purev E, Haverkos B, Pollyea DA, Cherry E, Kamdar M, Mark T, Forsberg P, Sherbenou D, Hammes A, et al. : Adult cord blood transplant results in comparable overall survival and improved GRFS vs matched related transplant. Blood Adv 2020, 4:2227–2235. [DOI] [PMC free article] [PubMed] [Google Scholar]; This retrospective single center comparison of UCB versus MSD transplant, avoiding ATG conditioning with UCB, found GRFS, a more relevant measure than OS, to be superior with UCB in the myeloablative setting, and to be similar in the nonmyeloablative setting. Both decreased cGVHD and decreased relapse was observed, and NRM was similar.
  • 8.Barker JN, Devlin SM, Naputo KA, Skinner K, Maloy MA, Flynn L, Anagnostou T, Avecilla ST, Scaradavou A, Cho C, et al. : High progression-free survival after intermediate intensity double unit cord blood transplantation in adults. Blood Adv 2020, 4:6064–6076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Passweg JR, Baldomero H, Chabannon C, Basak GW, de la Camara R, Corbacioglu S, Dolstra H, Duarte R, Glass B, Greco R, et al. : Hematopoietic cell transplantation and cellular therapy survey of the EBMT: monitoring of activities and trends over 30 years. Bone Marrow Transplant 2021, 56:1651–1664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Liu JH, Kanakry CG, Luznik L: Have haploidentical transplants replaced umbilical cord transplants for acute leukemias? Curr Opin Hematol 2018, 25:103–111. [DOI] [PubMed] [Google Scholar]
  • 11.Farhadfar N, Burns LJ, Mupfudze T, Shaw BE, Bollard CM, Devine SM, Horowitz MM, Jones RJ, Murthy HS, Wingard JR, et al. : Hematopoietic Cell Transplantation: Practice Predictions for the Year 2023. Transplant Cell Ther 2021, 27:183 e181–183 e187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Gragert L, Eapen M, Williams E, Freeman J, Spellman S, Baitty R, Hartzman R, Rizzo JD, Horowitz M, Confer D, et al. : HLA match likelihoods for hematopoietic stem-cell grafts in the U.S. registry. N Engl J Med 2014, 371:339–348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Kekre N, Antin JH: Hematopoietic stem cell transplantation donor sources in the 21st century: choosing the ideal donor when a perfect match does not exist. Blood 2014, 124:334–343. [DOI] [PubMed] [Google Scholar]
  • 14.Keating AK, Langenhorst J, Wagner JE, Page KM, Veys P, Wynn RF, Stefanski H, Elfeky R, Giller R, Mitchell R, et al. : The influence of stem cell source on transplant outcomes for pediatric patients with acute myeloid leukemia. Blood Adv 2019, 3:1118–1128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Mehta RS, Holtan SG, Wang T, Hemmer MT, Spellman SR, Arora M, Couriel DR, Alousi AM, Pidala J, Abdel-Azim H, et al. : GRFS and CRFS in alternative donor hematopoietic cell transplantation for pediatric patients with acute leukemia. Blood Adv 2019, 3:1441–1449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Yun HD, Varma A, Hussain MJ, Nathan S, Brunstein C: Clinical Relevance of Immunobiology in Umbilical Cord Blood Transplantation. J Clin Med 2019, 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Politikos I, Boussiotis VA: The role of the thymus in T-cell immune reconstitution after umbilical cord blood transplantation. Blood 2014, 124:3201–3211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kanakry CG, Fuchs EJ, Luznik L: Modern approaches to HLA-haploidentical blood or marrow transplantation. Nat Rev Clin Oncol 2016, 13:10–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Migliaccio AR, Adamson JW, Stevens CE, Dobrila NL, Carrier CM, Rubinstein P: Cell dose and speed of engraftment in placental/umbilical cord blood transplantation: graft progenitor cell content is a better predictor than nucleated cell quantity. Blood 2000, 96:2717–2722. [PubMed] [Google Scholar]
  • 20.Rafii H, Garnier F, Ruggeri A, Ionescu I, Ballot C, Bensoussan D, Chabannon C, Dazey B, De Vos J, Gautier E, et al. : Umbilical cord blood transplants facilitated by the French cord blood banks network. On behalf of the Agency of Biomedicine, Eurocord and the French society of bone marrow transplant and cell therapy (SFGM-TC). Bone Marrow Transplant 2021, 56:2497–2509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Mayani H, Wagner JE, Broxmeyer HE: Cord blood research, banking, and transplantation: achievements, challenges, and perspectives. Bone Marrow Transplant 2020, 55:48–61. [DOI] [PubMed] [Google Scholar]
  • 22.Christen F, Hablesreiter R, Hoyer K, Hennch C, Maluck-Bottcher A, Segler A, Madadi A, Frick M, Bullinger L, Briest F, et al. : Modeling clonal hematopoiesis in umbilical cord blood cells by CRISPR/Cas9. Leukemia 2022, 36:1102–1110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23*.Watts KL, Beard BC, Wood BL, Trobridge GD, Humphries RK, Adams AB, Nelson V, Kiem HP: No evidence of clonal dominance after transplant of HOXB4-expanded cord blood cells in a nonhuman primate model. Exp Hematol 2014, 42:497–504. [DOI] [PMC free article] [PubMed] [Google Scholar]; HOXB4-expanded bone marrow but not cord blood HSCs led to clonal dominance and leukemia, which has implications for the occurrence of post-transplant hematologic malignancy in the setting of post-transplant replicative stress.
  • 24.Sanz J, Montoro J, Solano C, Valcarcel D, Sampol A, Ferra C, Parody R, Lorenzo I, Montesinos P, Orti G, et al. : Prospective Randomized Study Comparing Myeloablative Unrelated Umbilical Cord Blood Transplantation versus HLA-Haploidentical Related Stem Cell Transplantation for Adults with Hematologic Malignancies. Biol Blood Marrow Transplant 2020, 26:358–366. [DOI] [PubMed] [Google Scholar]
  • 25.Gladstone DE, Bettinotti MP: HLA donor-specific antibodies in allogeneic hematopoietic stem cell transplantation: challenges and opportunities. Hematology Am Soc Hematol Educ Program 2017, 2017:645–650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Fuchs EJ, O’Donnell PV, Eapen M, Logan B, Antin JH, Dawson P, Devine S, Horowitz MM, Horwitz ME, Karanes C, et al. : Double unrelated umbilical cord blood vs HLA-haploidentical bone marrow transplantation: the BMT CTN 1101 trial. Blood 2021, 137:420–428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kosuri S, Wolff T, Devlin SM, Byam C, Mazis CM, Naputo K, Davis E, Paulson J, Nhaissi M, Wells DS, et al. : Prospective Evaluation of Unrelated Donor Cord Blood and Haploidentical Donor Access Reveals Graft Availability Varies by Patient Ancestry: Practical Implications for Donor Selection. Biol Blood Marrow Transplant 2017, 23:965–970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Allan DS, Green M, Morris G, Weiss J, Dibdin N, Mercer D, Seftel M: Demand and usage of unrelated donor products for allogeneic haematopoietic cell transplantation during the COVID-19 pandemic: A Canadian Blood Services Stem Cell Registry analysis. Vox Sang 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Wynn R, Nataraj R, Nadaf R, Poulton K, Logan A: Strategies for Success With Umbilical Cord Haematopoietic Stem Cell Transplantation in Children With Malignant and Non-Malignant Disease Indications. Front Cell Dev Biol 2022, 10:836594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Page KM, Ream MA, Rangarajan HG, Galindo R, Mian AY, Ho ML, Provenzale J, Gustafson KE, Rubin J, Shenoy S, et al. : Benefits of newborn screening and hematopoietic cell transplant in infantile Krabbe disease. Blood Adv 2022, 6:2947–2956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Orchard PJ, Gupta A, Eisengart JB, Polgreen LE, Pollard LM, Braunlin E, Pasquali M, Lund T: Hematopoietic stem cell transplant for Hurler Syndrome: does using bone marrow or umbilical cord blood make a difference? Blood Adv 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Zheng CC, Zhu XY, Tang BL, Zhang XH, Zhang L, Geng LQ, Liu HL, Sun ZM: Clinical separation of cGvHD and GvL and better GvHD-free/relapse-free survival (GRFS) after unrelated cord blood transplantation for AML. Bone Marrow Transplant 2017, 52:88–94. [DOI] [PubMed] [Google Scholar]
  • 33.Wagner JE, Ballen KK, Zhang MJ, Allbee-Johnson M, Karanes C, Milano F, Verneris MR, Eapen M, Brunstein CG: Comparison of haploidentical and umbilical cord blood transplantation after myeloablative conditioning. Blood Adv 2021, 5:4064–4072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Hiwarkar P, Qasim W, Ricciardelli I, Gilmour K, Quezada S, Saudemont A, Amrolia P, Veys P: Cord blood T cells mediate enhanced antitumor effects compared with adult peripheral blood T cells. Blood 2015, 126:2882–2891. [DOI] [PubMed] [Google Scholar]
  • 35.Moscardo F, Sanz J, Carbonell F, Sanz MA, Larrea L, Montesinos P, Lorenzo I, Vera B, Boluda B, Salazar C, et al. : Effect of CD8(+) cell content on umbilical cord blood transplantation in adults with hematological malignancies. Biol Blood Marrow Transplant 2014, 20:1744–1750. [DOI] [PubMed] [Google Scholar]
  • 36.**.Hiwarkar P, Adams S, Gilmour K, Nataraj R, Bonney D, Poulton K, Wynn R: Cord blood CD8+ T-cell expansion following granulocyte transfusions eradicates refractory leukemia. Blood Adv 2020, 4:4165–4174. [DOI] [PMC free article] [PubMed] [Google Scholar]; Transfusion of granulocytes can cross-prime naïve CD8+ T-cells, whose numbers in UCB units are associated with improved NRM and OS, and led to the eradication of refractory leukemia. This is a relatively simple treatment intervention that should be further tested.
  • 37.Beauvillain C, Delneste Y, Scotet M, Peres A, Gascan H, Guermonprez P, Barnaba V, Jeannin P: Neutrophils efficiently cross-prime naive T cells in vivo. Blood 2007, 110:2965–2973. [DOI] [PubMed] [Google Scholar]
  • 38.Elfeky R, Lazareva A, Qasim W, Veys P: Immune reconstitution following hematopoietic stem cell transplantation using different stem cell sources. Expert Rev Clin Immunol 2019, 15:735–751. [DOI] [PubMed] [Google Scholar]
  • 39.Wu R, Ma L: Haploidentical Hematopoietic Stem Cell Transplantation Versus Umbilical Cord Blood Transplantation in Hematologic Malignancies: A Systematic Review and Meta-Analysis. Cell Transplant 2020, 29:963689720964771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Gutman JA, Ross K, Smith C, Myint H, Lee CK, Salit R, Milano F, Delaney C, Gao D, Pollyea DA: Chronic graft versus host disease burden and late transplant complications are lower following adult double cord blood versus matched unrelated donor peripheral blood transplantation. Bone Marrow Transplant 2016, 51:1588–1593. [DOI] [PubMed] [Google Scholar]
  • 41.Eapen M, Rocha V, Sanz G, Scaradavou A, Zhang MJ, Arcese W, Sirvent A, Champlin RE, Chao N, Gee AP, et al. : Effect of graft source on unrelated donor haemopoietic stem-cell transplantation in adults with acute leukaemia: a retrospective analysis. Lancet Oncol 2010, 11:653–660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Crespo I, Paiva A, Couceiro A, Pimentel P, Orfao A, Regateiro F: Immunophenotypic and functional characterization of cord blood dendritic cells. Stem Cells Dev 2004, 13:63–70. [DOI] [PubMed] [Google Scholar]
  • 43.Admiraal R, van Kesteren C, Jol-van der Zijde CM, Lankester AC, Bierings MB, Egberts TC, van Tol MJ, Knibbe CA, Bredius RG, Boelens JJ: Association between anti-thymocyte globulin exposure and CD4+ immune reconstitution in paediatric haemopoietic cell transplantation: a multicentre, retrospective pharmacodynamic cohort analysis. Lancet Haematol 2015, 2:e194–203. [DOI] [PubMed] [Google Scholar]
  • 44.Castillo N, Garcia-Cadenas I, Barba P, Canals C, Diaz-Heredia C, Martino R, Ferra C, Badell I, Elorza I, Sierra J, et al. : Early and Long-Term Impaired T Lymphocyte Immune Reconstitution after Cord Blood Transplantation with Antithymocyte Globulin. Biol Blood Marrow Transplant 2017, 23:491–497. [DOI] [PubMed] [Google Scholar]
  • 45.Tanaka J, Morishima Y, Takahashi Y, Yabe T, Oba K, Takahashi S, Taniguchi S, Ogawa H, Onishi Y, Miyamura K, et al. : Effects of KIR ligand incompatibility on clinical outcomes of umbilical cord blood transplantation without ATG for acute leukemia in complete remission. Blood Cancer J 2013, 3:e164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Politikos I, Lavery JA, Hilden P, Cho C, Borrill T, Maloy MA, Giralt SA, van den Brink MRM, Perales MA, Barker JN: Robust CD4+ T-cell recovery in adults transplanted with cord blood and no antithymocyte globulin. Blood Adv 2020, 4:191–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Chiesa R, Gilmour K, Qasim W, Adams S, Worth AJ, Zhan H, Montiel-Equihua CA, Derniame S, Cale C, Rao K, et al. : Omission of in vivo T-cell depletion promotes rapid expansion of naive CD4+ cord blood lymphocytes and restores adaptive immunity within 2 months after unrelated cord blood transplant. Br J Haematol 2012, 156:656–666. [DOI] [PubMed] [Google Scholar]
  • 48.Tong J, Xuan L, Sun Y, Huang D, Liu H, Zheng C, Zhu X, Tang B, Song K, Zhang X, et al. : Umbilical Cord Blood Transplantation without Antithymocyte Globulin Results in Similar Survival but Better Quality of Life Compared with Unrelated Peripheral Blood Stem Cell Transplantation for the Treatment of Acute Leukemia-A Retrospective Study in China. Biol Blood Marrow Transplant 2017, 23:1541–1548. [DOI] [PubMed] [Google Scholar]
  • 49.Zheng C, Luan Z, Fang J, Sun X, Chen J, Li CK, Hu S, Zhu Y, Sun Z: Comparison of conditioning regimens with or without antithymocyte globulin for unrelated cord blood transplantation in children with high-risk or advanced hematological malignancies. Biol Blood Marrow Transplant 2015, 21:707–712. [DOI] [PubMed] [Google Scholar]
  • 50.Fatobene G, Rocha V, St Martin A, Hamadani M, Robinson S, Bashey A, Boumendil A, Brunstein C, Castagna L, Dominietto A, et al. : Nonmyeloablative Alternative Donor Transplantation for Hodgkin and Non-Hodgkin Lymphoma: From the LWP-EBMT, Eurocord, and CIBMTR. J Clin Oncol 2020, 38:1518–1526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Bhella S, Majhail NS, Betcher J, Costa LJ, Daly A, Dandoy CE, DeFilipp Z, Doan V, Gulbis A, Hicks L, et al. : Choosing Wisely BMT: American Society for Blood and Marrow Transplantation and Canadian Blood and Marrow Transplant Group’s List of 5 Tests and Treatments to Question in Blood and Marrow Transplantation. Biol Blood Marrow Transplant 2018, 24:909–913. [DOI] [PubMed] [Google Scholar]
  • 52.Michel G, Galambrun C, Sirvent A, Pochon C, Bruno B, Jubert C, Loundou A, Yakoub-Agha I, Milpied N, Lutz P, et al. : Single- vs double-unit cord blood transplantation for children and young adults with acute leukemia or myelodysplastic syndrome. Blood 2016, 127:3450–3457. [DOI] [PubMed] [Google Scholar]
  • 53.**.Wagner JE Jr., Eapen M, Carter S, Wang Y, Schultz KR, Wall DA, Bunin N, Delaney C, Haut P, Margolis D, et al. : One-unit versus two-unit cord-blood transplantation for hematologic cancers. N Engl J Med 2014, 371:1685–1694. [DOI] [PMC free article] [PubMed] [Google Scholar]; This multi-center trial randomizing between one-unit and two-unit cord blood transplant found improved platelet recovery and lower incidences of grade III and IV acute and extensive chronic GVHD in the one-unit group.
  • 54.Zheng CC, Zhu XY, Tang BL, Zhang XH, Zhang L, Geng LQ, Liu HL, Sun ZM: Double vs. single cord blood transplantation in adolescent and adult hematological malignancies with heavier body weight (>/=50 kg). Hematology 2018, 23:96–104. [DOI] [PubMed] [Google Scholar]
  • 55.Okada Y, Nakasone H, Konuma T, Uchida N, Tanaka M, Sugio Y, Aotsuka N, Nishijima A, Katsuoka Y, Ara T, et al. : Ideal body weight is useful for predicting neutrophil engraftment and platelet recovery for overweight and obese recipients in single-unit cord blood transplantation. Transplant Cell Ther 2022. [DOI] [PubMed] [Google Scholar]
  • 56.*.Eapen M, Klein JP, Ruggeri A, Spellman S, Lee SJ, Anasetti C, Arcese W, Barker JN, Baxter-Lowe LA, Brown M, et al. : Impact of allele-level HLA matching on outcomes after myeloablative single unit umbilical cord blood transplantation for hematologic malignancy. Blood 2014, 123:133–140. [DOI] [PMC free article] [PubMed] [Google Scholar]; This retrospective registry data analysis in heme malignancy found that even single allele-level HLA mismatch at HLA-A, -C, or -DRB1 was associated with a 3-fold increase in NRM risk.
  • 57.Eapen M, Wang T, Veys PA, Boelens JJ, St Martin A, Spellman S, Bonfim CS, Brady C, Cant AJ, Dalle JH, et al. : Allele-level HLA matching for umbilical cord blood transplantation for non-malignant diseases in children: a retrospective analysis. Lancet Haematol 2017, 4:e325–e333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Kanda J, Hayashi H, Ruggeri A, Kimura F, Volt F, Takahashi S, Labopin M, Kako S, Tozatto-Maio K, Yano S, et al. : Prognostic factors for adult single cord blood transplantation among European and Japanese populations: the Eurocord/ALWP-EBMT and JSHCT/JDCHCT collaborative study. Leukemia 2020, 34:128–137. [DOI] [PubMed] [Google Scholar]
  • 59.Kanda J, Hirabayashi S, Yokoyama H, Kawase T, Tanaka H, Uchida N, Taniguchi S, Takahashi S, Onizuka M, Tanaka M, et al. : Effect of multiple HLA-locus mismatches on outcomes after single cord blood transplantation. Transplant Cell Ther 2022. [DOI] [PubMed] [Google Scholar]
  • 60.Eapen M, Klein JP, Sanz GF, Spellman S, Ruggeri A, Anasetti C, Brown M, Champlin RE, Garcia-Lopez J, Hattersely G, et al. : Effect of donor-recipient HLA matching at HLA A, B, C, and DRB1 on outcomes after umbilical-cord blood transplantation for leukaemia and myelodysplastic syndrome: a retrospective analysis. Lancet Oncol 2011, 12:1214–1221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Velardi E, Clave E, Arruda LCM, Benini F, Locatelli F, Toubert A: The role of the thymus in allogeneic bone marrow transplantation and the recovery of the peripheral T-cell compartment. Semin Immunopathol 2021, 43:101–117. [DOI] [PubMed] [Google Scholar]
  • 62.Muller AMS, Min D, Wernig G, Levy RB, Perez VL, Herretes S, Florek M, Burnett C, Weinberg K, Shizuru JA: Modeling Chronic Graft-versus-Host Disease in MHC-Matched Mouse Strains: Genetics, Graft Composition, and Tissue Targets. Biol Blood Marrow Transplant 2019, 25:2338–2349. [DOI] [PubMed] [Google Scholar]
  • 63.Krenger W, Hollander GA: The immunopathology of thymic GVHD. Semin Immunopathol 2008, 30:439–456. [DOI] [PubMed] [Google Scholar]
  • 64.Eyrich M, Leiler C, Lang P, Schilbach K, Schumm M, Bader P, Greil J, Klingebiel T, Handgretinger R, Niethammer D, et al. : A prospective comparison of immune reconstitution in pediatric recipients of positively selected CD34+ peripheral blood stem cells from unrelated donors vs recipients of unmanipulated bone marrow from related donors. Bone Marrow Transplant 2003, 32:379–390. [DOI] [PubMed] [Google Scholar]
  • 65.Yokoyama H, Morishima Y, Fuji S, Uchida N, Takahashi S, Onizuka M, Tanaka M, Yuju O, Eto T, Ozawa Y, et al. : Impact of HLA Allele Mismatch at HLA-A, -B, -C, and -DRB1 in Single Cord Blood Transplantation. Biol Blood Marrow Transplant 2020, 26:519–528. [DOI] [PubMed] [Google Scholar]
  • 66.Zhu X, Tang B, Sun Z: Umbilical cord blood transplantation: Still growing and improving. Stem Cells Transl Med 2021, 10 Suppl 2:S62–S74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Rocha V, Spellman S, Zhang MJ, Ruggeri A, Purtill D, Brady C, Baxter-Lowe LA, Baudoux E, Bergamaschi P, Chow R, et al. : Effect of HLA-matching recipients to donor noninherited maternal antigens on outcomes after mismatched umbilical cord blood transplantation for hematologic malignancy. Biol Blood Marrow Transplant 2012, 18:1890–1896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Abou-Ismail MY, Fraser R, Allbee-Johnson M, Metheny L 3rd, Ravi G, Ahn KW, Bhatt NS, Lazarus HM, de Lima M, El Jurdy N, et al. : Does recipient body mass index inform donor selection for allogeneic haematopoietic cell transplantation? Br J Haematol 2022, 197:326–338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Brunstein CG, O’Donnell PV, Logan B, Dawson P, Costa L, Cutler C, Craig M, Hogan W, Horowitz MM, Horwitz ME, et al. : Impact of Center Experience with Donor Type on Outcomes: A Secondary Analysis, Blood and Marrow Transplant Clinical Trials Network 1101Open for Accrual June 2012Open for Accrual June 2012. Transplant Cell Ther 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Holtan SG, DeFor TE, Lazaryan A, Bejanyan N, Arora M, Brunstein CG, Blazar BR, MacMillan ML, Weisdorf DJ: Composite end point of graft-versus-host disease-free, relapse-free survival after allogeneic hematopoietic cell transplantation. Blood 2015, 125:1333–1338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Beksac M, Yurdakul P: How to Improve Cord Blood Engraftment? Front Med (Lausanne) 2016, 3:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.ACOG Committee Opinion No. 771 Summary: Umbilical Cord Blood Banking. Obstet Gynecol 2019, 133:604–606. [DOI] [PubMed] [Google Scholar]
  • 73.Milano FRA, Kurtzberg J, Karanes C, Gutman JA, Duncan C, Keyzner A, Hanna R, Marmon TK, Dahlberg A, Delaney C.: No Engraftment Advantage after Single or Double Umbilical Cord Blood Transplant (CBT) with the Addition of a Non-HLA Matched Off-the-Shelf Expanded Cord Blood Unit Compared to Conventional CBT: Results of a Randomized Trial. In American Society of Hematology Annual Meeting. Edited by: Blood 2019:146. vol 134.] [Google Scholar]
  • 74.Arteaga A, Orfali N, Guarneri D, Cushing MM, Gergis U, Hsu J, Hsu YS, Mayer SA, Phillips AA, Chase SA, et al. : Cord blood transplants supported by unrelated donor CD34(+) progenitor cells. Bone Marrow Transplant 2020, 55:2298–2307. [DOI] [PubMed] [Google Scholar]
  • 75.Lindemans CA, Te Boome LC, Admiraal R, Jol-van der Zijde EC, Wensing AM, Versluijs AB, Bierings MB, Kuball J, Boelens JJ: Sufficient Immunosuppression with Thymoglobulin Is Essential for a Successful Haplo-Myeloid Bridge in Haploidentical-Cord Blood Transplantation. Biol Blood Marrow Transplant 2015, 21:1839–1845. [DOI] [PubMed] [Google Scholar]
  • 76.Politikos I, Devlin SM, Arcila ME, Barone JC, Maloy MA, Naputo KA, Ruiz JD, Mazis CM, Scaradavou A, Avecilla ST, et al. : Engraftment kinetics after transplantation of double unit cord blood grafts combined with haplo-identical CD34+ cells without antithymocyte globulin. Leukemia 2021, 35:850–862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Tsai SB, Rhodes J, Liu H, Shore T, Bishop M, Cushing MM, Gergis U, Godley L, Kline J, Larson RA, et al. : Reduced-Intensity Allogeneic Transplant for Acute Myeloid Leukemia and Myelodysplastic Syndrome Using Combined CD34-Selected Haploidentical Graft and a Single Umbilical Cord Unit Compared with Matched Unrelated Donor Stem Cells in Older Adults. Biol Blood Marrow Transplant 2018, 24:997–1004. [DOI] [PubMed] [Google Scholar]
  • 78.van Besien K, Artz A, Champlin RE, Guarneri D, Bishop MR, Chen J, Gergis U, Shore T, Liu H, Rondon G, et al. : Haploidentical vs haplo-cord transplant in adults under 60 years receiving fludarabine and melphalan conditioning. Blood Adv 2019, 3:1858–1867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Bashey A, Zhang MJ, McCurdy SR, St Martin A, Argall T, Anasetti C, Ciurea SO, Fasan O, Gaballa S, Hamadani M, et al. : Mobilized Peripheral Blood Stem Cells Versus Unstimulated Bone Marrow As a Graft Source for T-Cell-Replete Haploidentical Donor Transplantation Using Post-Transplant Cyclophosphamide. J Clin Oncol 2017, 35:3002–3009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Yu X, Liu L, Xie Z, Dong C, Zhao L, Zhang J, Gu J, Zhu HH: Bone marrow versus peripheral blood as a graft source for haploidentical donor transplantation in adults using post-transplant cyclophosphamide-A systematic review and meta-analysis. Crit Rev Oncol Hematol 2019, 133:120–128. [DOI] [PubMed] [Google Scholar]
  • 81.Mariotti J, Devillier R, Bramanti S, Giordano L, Sarina B, Furst S, Granata A, Maisano V, Pagliardini T, De Philippis C, et al. : Peripheral Blood Stem Cells versus Bone Marrow for T Cell-Replete Haploidentical Transplantation with Post-Transplant Cyclophosphamide in Hodgkin Lymphoma. Biol Blood Marrow Transplant 2019, 25:1810–1817. [DOI] [PubMed] [Google Scholar]
  • 82.Lyu H, Lu W, Yao J, Xiao X, Li Q, Wang J, Mu J, Qi Y, Zhu H, Jiang Y, et al. : Comparison of outcomes of haploidentical donor hematopoietic stem cell transplantation supported by third-party cord blood with HLA-matched unrelated donor transplantation. Leuk Lymphoma 2020, 61:840–847. [DOI] [PubMed] [Google Scholar]
  • 83.Bacigalupo A, Sica S, Laurenti L, Sora F, Giammarco S, Metafuni E, Innocenti I, Autore F, Teofili L, Bianchi M, et al. : Unrelated cord blood transplantation and post-transplant cyclophosphamide. Haematologica 2019, 104:e77–e78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Sellar RS, Vargas FA, Henry JY, Verfuerth S, Charrot S, Beaton B, Chakraverty R, Quezada SA, Mackinnon S, Thomson KJ, et al. : CMV promotes recipient T-cell immunity following reduced-intensity T-cell-depleted HSCT, significantly modulating chimerism status. Blood 2015, 125:731–739. [DOI] [PubMed] [Google Scholar]
  • 85.Delaney C, Heimfeld S, Brashem-Stein C, Voorhies H, Manger RL, Bernstein ID: Notch-mediated expansion of human cord blood progenitor cells capable of rapid myeloid reconstitution. Nat Med 2010, 16:232–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.de Lima M, McNiece I, Robinson SN, Munsell M, Eapen M, Horowitz M, Alousi A, Saliba R, McMannis JD, Kaur I, et al. : Cord-blood engraftment with ex vivo mesenchymal-cell coculture. N Engl J Med 2012, 367:2305–2315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Wagner JE Jr., Brunstein CG, Boitano AE, DeFor TE, McKenna D, Sumstad D, Blazar BR, Tolar J, Le C, Jones J, et al. : Phase I/II Trial of StemRegenin-1 Expanded Umbilical Cord Blood Hematopoietic Stem Cells Supports Testing as a Stand-Alone Graft. Cell Stem Cell 2016, 18:144–155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Horwitz ME, Chao NJ, Rizzieri DA, Long GD, Sullivan KM, Gasparetto C, Chute JP, Morris A, McDonald C, Waters-Pick B, et al. : Umbilical cord blood expansion with nicotinamide provides long-term multilineage engraftment. J Clin Invest 2014, 124:3121–3128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.*.Horwitz ME, Stiff PJ, Cutler C, Brunstein C, Hanna R, Maziarz RT, Rezvani AR, Karris NA, McGuirk J, Valcarcel D, et al. : Omidubicel vs standard myeloablative umbilical cord blood transplantation: results of a phase 3 randomized study. Blood 2021, 138:1429–1440. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study randomizing patients with heme malignancy between products from a single UCB unit (expanded and unexpanded cell types) versus standard UBC transplant found faster hematopoietic recovery and lower infection in the manipulated UCB group.
  • 90.Cohen S, Roy J, Lachance S, Delisle JS, Marinier A, Busque L, Roy DC, Barabe F, Ahmad I, Bambace N, et al. : Hematopoietic stem cell transplantation using single UM171-expanded cord blood: a single-arm, phase 1–2 safety and feasibility study. Lancet Haematol 2020, 7:e134–e145. [DOI] [PubMed] [Google Scholar]
  • 91.Brunstein CG, Miller JS, McKenna DH, Hippen KL, DeFor TE, Sumstad D, Curtsinger J, Verneris MR, MacMillan ML, Levine BL, et al. : Umbilical cord blood-derived T regulatory cells to prevent GVHD: kinetics, toxicity profile, and clinical effect. Blood 2016, 127:1044–1051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Huang Z, Xiao Y, Chen X, Li H, Gao J, Wei W, Zhang X, Feng X: Cotransplantation of Umbilical Cord Mesenchymal Stem Cells Promotes the Engraftment of Umbilical Cord Blood Stem Cells in Iron Overload NOD/SCID Mice. Transplant Cell Ther 2021, 27:230 e231–230 e237. [DOI] [PubMed] [Google Scholar]
  • 93.Sharma A, Sances S, Workman MJ, Svendsen CN: Multi-lineage Human iPSC-Derived Platforms for Disease Modeling and Drug Discovery. Cell Stem Cell 2020, 26:309–329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.*.Bloor AJC, Patel A, Griffin JE, Gilleece MH, Radia R, Yeung DT, Drier D, Larson LS, Uenishi GI, Hei D, et al. : Production, safety and efficacy of iPSC-derived mesenchymal stromal cells in acute steroid-resistant graft versus host disease: a phase I, multicenter, open-label, dose-escalation study. Nat Med 2020, 26:1720–1725. [DOI] [PubMed] [Google Scholar]; This study represents the first completed Phase I FDA clinical trial for the use of an iPSC-derived cell therapy for the treatment of acute graft-versus-host disease.
  • 95.Kim JY, Nam Y, Rim YA, Ju JH: Review of the Current Trends in Clinical Trials Involving Induced Pluripotent Stem Cells. Stem Cell Rev Rep 2022, 18:142–154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Shin S, Song EY, Kwon YW, Oh S, Park H, Kim NH, Roh EY: Usefulness of the Hematopoietic Stem Cell Donor Pool as a Source of HLA-Homozygous Induced Pluripotent Stem Cells for Haplobanking: Combined Analysis of the Cord Blood Inventory and Bone Marrow Donor Registry. Biol Blood Marrow Transplant 2020, 26:e202–e208. [DOI] [PubMed] [Google Scholar]
  • 97.*.Tian P, Elefanty A, Stanley EG, Durnall JC, Thompson LH, Elwood NJ: Creation of GMP-Compliant iPSCs From Banked Umbilical Cord Blood. Front Cell Dev Biol 2022, 10:835321. [DOI] [PMC free article] [PubMed] [Google Scholar]; This article highlights the efficiency and robustness of creating of clinical-grade iPSC cell lines from small volumes of cryopreserved CB cells.
  • 98.Lee S, Huh JY, Turner DM, Lee S, Robinson J, Stein JE, Shim SH, Hong CP, Kang MS, Nakagawa M, et al. : Repurposing the Cord Blood Bank for Haplobanking of HLA-Homozygous iPSCs and Their Usefulness to Multiple Populations. Stem Cells 2018, 36:1552–1566. [DOI] [PubMed] [Google Scholar]
  • 99.Alvarez-Palomo B, Garcia-Martinez I, Gayoso J, Raya A, Veiga A, Abad ML, Eiras A, Guzman-Fulgencio M, Luis-Hidalgo M, Eguizabal C, et al. : Evaluation of the Spanish population coverage of a prospective HLA haplobank of induced pluripotent stem cells. Stem Cell Res Ther 2021, 12:233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.*.Barker JN, Mazis CM, Devlin SM, Davis E, Maloy MA, Naputo K, Nhaissi M, Wells D, Scaradavou A, Politikos I: Evaluation of Cord Blood Total Nucleated and CD34(+) Cell Content, Cell Dose, and 8-Allele HLA Match by Patient Ancestry. Biol Blood Marrow Transplant 2020, 26:734–744. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study of units selected for UCB transplant by recipient ancestry found markedly lower total nucleated cell counts, CD34+ counts, and HLA matching in units selected for people of non-European versus European ancestry.
  • 101.Viljoen IM, Hendricks CL, Mellet J, Pepper MS: Perspectives on establishing a public cord blood inventory in South Africa. Cytotherapy 2021, 23:548–557. [DOI] [PubMed] [Google Scholar]
  • 102.Greco-Stewart V, Kiernan J, Killeen D, Haun S, Mercer D, Young K, Liwski RS, Allan DS: Unrelated donor choices for allogeneic hematopoietic cell transplantation in Canada: an evaluation of factors influencing donor selection. Transfusion 2018, 58:718–725. [DOI] [PubMed] [Google Scholar]
  • 103.Barker JN, Boughan K, Dahi PB, Devlin SM, Maloy MA, Naputo K, Mazis CM, Davis E, Nhaissi M, Wells D, et al. : Racial disparities in access to HLA-matched unrelated donor transplants: a prospective 1312-patient analysis. Blood Adv 2019, 3:939–944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Hsu JM vB K, Glesby MJ, Coletti A, Pahwa SG, Warshaw M, Golner A, Bone F, Tobin N, Riches M, Mellors JW, Browining R, Persaud D, Bryson Y=: HIV-1 REMISSION WITH CCR5 MUTANT HAPLO-CORD TRANSPLANT IN A US WOMAN: IMPAACT P1107. In Conference on Retroviruses and Opportunistic Infections: 2022. [Google Scholar]
  • 105.Graham F: Daily briefing: Woman cleared of HIV after transplant with umbilical-cord blood. Nature 2022. [DOI] [PubMed] [Google Scholar]
  • 106.Gluckman E, Cappelli B, Scigliuolo GM, De la Fuente J, Corbacioglu S: Alternative donor hematopoietic stem cell transplantation for sickle cell disease in Europe. Hematol Oncol Stem Cell Ther 2020, 13:181–188. [DOI] [PubMed] [Google Scholar]
  • 107.Eapen M, Brazauskas R, Walters MC, Bernaudin F, Bo-Subait K, Fitzhugh CD, Hankins JS, Kanter J, Meerpohl JJ, Bolanos-Meade J, et al. : Effect of donor type and conditioning regimen intensity on allogeneic transplantation outcomes in patients with sickle cell disease: a retrospective multicentre, cohort study. Lancet Haematol 2019, 6:e585–e596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Wojcik GL, Graff M, Nishimura KK, Tao R, Haessler J, Gignoux CR, Highland HM, Patel YM, Sorokin EP, Avery CL, et al. : Genetic analyses of diverse populations improves discovery for complex traits. Nature 2019, 570:514–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Martin AR, Kanai M, Kamatani Y, Okada Y, Neale BM, Daly MJ: Clinical use of current polygenic risk scores may exacerbate health disparities. Nat Genet 2019, 51:584–591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.All of Us Research Program I, Denny JC, Rutter JL, Goldstein DB, Philippakis, Smoller JW, Jenkins G, Dishman E: The “All of Us” Research Program. N Engl J Med 2019, 381:668–676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.*.Nehme R, Barrett LE: Using human pluripotent stem cell models to study autism in the era of big data. Mol Autism 2020, 11:21. [DOI] [PMC free article] [PubMed] [Google Scholar]; This article discusses four issues which influence the efficacy of hPSC models for studying disease, including (i) sources of variance, (ii) scale and format of study design, (iii) divergence from the human brain in vivo, and (iv) regulatory policies overseeing the use of hPSCs.
  • 112.Horwitz R, Riley EAU, Millan MT, Gunawardane RN: It’s time to incorporate diversity into our basic science and disease models. Nat Cell Biol 2021, 23:1213–1214. [DOI] [PubMed] [Google Scholar]
  • 113.Zhong Y, De T, Alarcon C, Park CS, Lec B, Perera MA: Discovery of novel hepatocyte eQTLs in African Americans. PLoS Genet 2020, 16:e1008662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Chen W, Brehm JM, Lin J, Wang T, Forno E, Acosta-Perez E, Boutaoui N, Canino G, Celedon JC: Expression quantitative trait loci (eQTL) mapping in Puerto Rican children. PLoS One 2015, 10:e0122464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.*.Bonder MJ, Smail C, Gloudemans MJ, Fresard L, Jakubosky D, D’Antonio M, Li X, Ferraro NM, Carcamo-Orive I, Mirauta B, et al. : Identification of rare and common regulatory variants in pluripotent cells using population-scale transcriptomics. Nat Genet 2021, 53:313–321. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study collates data from 1,367 human iPSC lines to map regulatory variants and demonstrates how iPSCs enable the identification of causal genes for rare diseases.
  • 116.**.Liu E, Marin D, Banerjee P, Macapinlac HA, Thompson P, Basar R, Nassif Kerbauy L, Overman B, Thall P, Kaplan M, et al. : Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. N Engl J Med 2020, 382:545–553. [DOI] [PMC free article] [PubMed] [Google Scholar]; In this Phase I-II trial, 8 of 11 patients with relapsed or refractory CD19+ tumors treated with HLA-mismatched anti-CD19 CAR-NK cells derived from cord blood responded and there was no major side effect toxicity.
  • 117.Dave H, Luo M, Blaney JW, Patel S, Barese C, Cruz CR, Shpall EJ, Bollard CM, Hanley PJ: Toward a Rapid Production of Multivirus-Specific T Cells Targeting BKV, Adenovirus, CMV, and EBV from Umbilical Cord Blood. Mol Ther Methods Clin Dev 2017, 5:13–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Zhou L, McDonald C, Yawno T, Jenkin G, Miller S, Malhotra A: Umbilical Cord Blood and Cord Tissue-Derived Cell Therapies for Neonatal Morbidities: Current Status and Future Challenges. Stem Cells Transl Med 2022, 11:135–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Bizymi N, Georgopoulou A, Mastrogamvraki N, Matheakakis A, Gontika I, Fragiadaki I, Mavroudi I, Papadaki HA: Myeloid-Derived Suppressor Cells (MDSC) in the Umbilical Cord Blood: Biological Significance and Possible Therapeutic Applications. J Clin Med 2022, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES