Next Article in Journal
Association between Serum Uric Acid and Liver Enzymes in Adults Aged 20 Years and Older in the United States: NHANES 2005–2012
Next Article in Special Issue
Comparison of Different Rabbit Anti-Thymocyte Globulin Formulations in the Prophylaxis of Graft-Versus-Host Disease: A Systematic Review
Previous Article in Journal
Factors Associated with Postpartum Maternal Functioning in Black Women: A Secondary Analysis
Previous Article in Special Issue
Handheld Ultrasound or Conventional Ultrasound Devices in Patients Undergoing HCT: A Validation Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Human Leucocyte Antigen System and Selection of Unrelated Hematopoietic Stem Cell Donors: Impact of Patient–Donor (Mis)matching and New Challenges with the Current Technologies

by
Roberto Crocchiolo
1,* and
Gianni Rombolà
2
1
Servizio di Immunoematologia e Medicina Trasfusionale, ASST Grande Ospedale Metropolitano Niguarda, Piazza dell’Ospedale Maggiore, 3, 20162 Milano, Italy
2
Laboratory of Immunogenetics and Transplant Immunology, Azienda Ospedaliero-Universitaria Careggi, 50134 Firenze, Italy
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(2), 646; https://doi.org/10.3390/jcm12020646
Submission received: 11 December 2022 / Revised: 9 January 2023 / Accepted: 11 January 2023 / Published: 13 January 2023

Abstract

:
The selection of hematopoietic stem cell donors for allogeneic transplantation (allo-HSCT) is mainly driven by human leucocyte antigen (HLA) matching between patient and donor, with HLA-identical matched siblings being the preferred choice in most situations. Although other clinical and demographical variables matter, especially, donor age, which is unequivocally associated with better transplant outcomes, the histocompatibility criteria have a central role in the search for the best donor, particularly in the setting of unrelated allo-HSCT where HLA disparities between patient and donor are frequent. The present review is focused on the role of HLA incompatibilities on patient outcome according to the most recent literature, in an attempt to guide transplant physicians and search coordinators during the process of adult unrelated-donor selection. The technological progresses in HLA typing, i.e., with next-generation sequencing (NGS), now allow disclosing a growing number of HLA incompatibilities associated with a heterogeneous and sometimes unknown spectrum of clinical severity. Their immunogenic characteristics, i.e., their position inside or outside the antigen recognition domain (ARD), their permissiveness, their intronic or exonic nature and even the expected expression of the HLA loci where those mismatches occur, will be presented and discussed here, integrating the advances in the immunobiology of transplantation with survival and toxicity outcomes reported in the most relevant studies, within the perspective of improving donor selection in the current practice.

1. Introduction

The selection of hematopoietic stem cell donors for allogeneic transplantation (allo-HSCT) is mainly driven by human leucocyte antigen (HLA) matching between patient and donor, with HLA-identical matched siblings being the preferred choice in most situations [1]. The increase of unrelated donations for allo-HSCT in last years [2] and the technological progresses in HLA typing [3] entail today a growing number of HLA incompatibilities disclosed between patients and their respective donors, with a heterogeneous and sometimes undefined spectrum of clinical significance. The immunogenic characteristics of the incompatibilities, i.e., their position inside or outside the antigen recognition domain (ARD) or their immunogenicity, the permissiveness with respect to transplant outcomes, their intronic or exonic nature and the expression of the HLA locus where those mismatches occur, are all elements to be taken into account before allo-HSCT is performed, in order to eventually adapt graft-versus-host disease (GvHD) prophylaxis strategies and optimize patient outcome. Nowadays, over 40,000,000 unrelated donors are potentially available from worldwide registries [4], and sources of HLA-mismatched hematopoietic stem cells like cord blood or haploidentical donors are readily available for allo-HSCT; therefore, the donor selection process requires up-to-date knowledge and cooperation between HLA laboratories, search coordinators and transplant physicians. As a consequence of the evolution of the transplantation procedures, HLA typing technologies and immunogenetics progresses, the HLA incompatibilities between patients and donors should be considered today from a renewed perspective, going beyond the “old” definitions of antigenic or allelic, to now include concepts of permissiveness and immunological risk.
We present here the most recent literature on HLA and allo-HSCT from adult unrelated donors by providing evidence about the immunological and clinical meaning of HLA incompatibilities between patients and their respective donors. Finally, integrating the advances in the immunobiology of transplantation with clinical experience is warranted to sustain and improve patient survival after allo-HSCT.

2. Definitions of HLA Mismatch in allo-HSCT

Except for HLA-identical siblings, all unrelated stem cell donors present some degree of HLA incompatibilities with their patients even in the best HLA-matched situation, due to the fact that the HLA haplotypes are not those inherited from the parents. The comparable clinical outcomes after allo-HSCT from a well-matched unrelated donor and an HLA-identical sibling [5,6,7] are explained by the fact that the above-cited HLA incompatibilities do not involve the ARD of the loci known to affect survival, being classically HLA-A, -B, -C and DRB1 [8] as well as, to some extent, DQB1 [9]. Of course, factors other than HLA do impact patient survival after allo-HSCT, among which, age matters most [10], suggesting the superiority of a younger well-matched unrelated donor upon an older HLA-identical sibling [11].
Beyond the cited HLA loci, DPB1 matching has been extensively studied in allo-HSCT, and some combinations (referred to as permissive) demonstrated to be better tolerated than others, based on the immunogenicity of the associated T-cell epitopes [12,13,14] and HLA expression [15,16]. Due to weak linkage disequilibrium, indeed, DPB1 is frequently mismatched in 10/10 HLA-matched patient–donor pairs [17]; therefore, matching at DPB1 (12/12 HLA-matched) is achievable in a lower proportion of pairs with respect to 10/10 HLA matching and permissiveness [17].
Noteworthy, the recent introduction of second-generation sequencing technologies for routine HLA typing (next-generation sequencing, NGS) has allowed for the detection of incompatibilities occurring beyond the second field [3], whose clinical significance is less defined than that of incompatibilities within the ARD, albeit under investigation. Recently, improved survival was observed among patients receiving allo-HSCT from donors matched at ultra-high resolution [18]; nonetheless, the current standards contemplate the first two fields for clinical significance of HLA mismatches [19]. Future refinements cannot be excluded, as growing data will allow for further association analyses. A recent national study [20] found lower acute GvHD when patient and donor shared the same geographical origin, possibly related to higher HLA similarity beyond the second field.
The so-called “non-classical” HLA loci (i.e., HLA-G and -E) might also have a clinical impact [21,22], although this is beyond the scope of the present article.
All these elements together demonstrate that the definition of clinically meaningful HLA incompatibilities in allo-HSCT are not fixed but change over time according to the technological advances in HLA typing coupled to clinical studies.
Based on the most prominent current literature and respective evidence, we will focus here on the clinical relevance of HLA incompatibilities within the ARD. The effect of such incompatibilities on the physico-chemical structure of the HLA molecules and their bound peptides (immunopeptidome), the direction of mismatches and the expression of the loci involved have to be taken into account for a proper interpretation [19] and to finally deal with permissiveness and immunological risk.

3. Methodology of Most HLA Studies in the HSCT Setting

The most relevant studies reporting the clinical impact of HLA matching between transplanted patients and their adult unrelated donors are retrospective, mono- or multicenter and even registry-based, using univariate and multivariate regression analyses for any association between HLA matching and clinical outcomes. As a consequence, the evidence generated is limited due to the lack of prospective interventional trials that are unfeasible in this setting. Nevertheless, the current recommendations and up-to-date daily practice among search coordinators and transplant physicians refer to such level of evidence, particularly appreciated when huge numbers of patient–donor pairs are reported, as it occurs in registry studies. Besides this, two meta-analyses have been published so far to our knowledge [23,24], showing similar results.
The immunological risk associated with the presence of any HLA incompatibility must be firstly assessed by the morbidity and mortality outcomes, mostly, acute GvHD and overall survival, although secondary outcomes such as progression-free survival, non-relapse mortality, relapse incidence, chronic GvHD or even composite endpoints including GvHD-free, relapse-free survival are also reported but with potential discrepancies among studies, due to differences in the definitions of the endpoints or due to reporting bias especially in multicenter or registry analyses. Despite the fact that relapse is a problematic issue after allo-HSCT for malignancies, there is no current evidence that any HLA mismatch confers a reduction of the relapse risk without increasing non-relapse mortality, consistently translating into a lack of survival benefit among the published studies. In addition, relapse and non-relapse mortality are competing risks, meaning that patients with one event are no longer at risk for the competing one [25].
Acute GvHD, especially of grade II or higher [26], that is, when systemic immunosuppressive treatment is needed, is probably the most suitable marker of morbidity among the above-cited studies, mainly because of its easily reproducible evaluation and definition, providing low expected discrepancies between the reported studies; moreover, any reported additional acute GvHD risk associated with any HLA mismatch might be the basis for a reinforced GvHD prophylaxis in the presence of such incompatibility [27]. Secondly, the multivariate hazard ratio together with the 95% confidence interval (CI) provides an estimate of the risk carried on by any mismatch(es) when compared with matched pairs, thus giving valuable information about the respective clinical importance. Noteworthy, the amplitude of the 95% CI often reflects the number of pairs analyzed, an important factor to be taken into account when interpreting the reported results.
While the immunological risk carried on by HLA incompatibilities between patients and their respective donors might be quantified, the concept of permissiveness is more related to the extent transplant physicians and patients accept for the immunological risk in the presence of any HLA mismatch. Of course, this risk acceptance depends on several factors, including disease prognosis, patient wishes and available therapies. Nevertheless, in most current literature, the permissive HLA mismatches usually refer to incompatibilities that do not significantly impair the transplant outcomes [13,28,29].

4. Locus-Specific HLA Incompatibilities

Several studies reported significant associations between the HLA matching of patient–donor pairs and transplant outcomes, and the most relevant among them provided the basis for the unrelated donor search and selection over time [8,13,30,31,32,33]. However, as transplantation practices change and evolve [34], many studies reappraised the question in recent years and provided updated results in an allo-HSCT setting where a predominant proportion of peripheral blood stem cell grafts vs. bone marrow, less toxic conditioning regimens, improved GvHD prophylaxis and higher median patient age is generally observed when compared to the picture of the previous studies published in the first decade of the years 2000, referring to allo-HSCTs performed in the late 1980s and 1990s.

4.1. HLA-A

The first among the class I alleles, HLA-A, shows the highest expression, together with HLA-B [35]. When dissecting recent studies (published after 2010) with a relevant number of transplants (>1000), the hazard ratio of mortality is between 1.17 and 1.43, and the grade II–IV acute GvHD risk is from 1.18 to 1.34 times higher in HLA-A mismatched vs. matched pairs (Table 1). There is no significant differential impact on the outcome depending on whether the mismatch is antigenic or allelic; thus, they are comparable. This finding is consistent across all classical HLA loci, with the exception of HLA-C (see below).

4.2. HLA-B

As shown in Table 1, the risks of overall mortality and grade II–IV acute GvHD in the presence of one incompatibility at locus B are 1.20–1.52 and 1.11–2.02 times higher, respectively, when compared to those of matched patient–donor pairs. A recent classification using data from the HLA-B Exon 1 (“B-leader”), proposed by Petersdorf and colleagues [41], informs on the acute GvHD risk and may be considered whenever two or more unrelated donors are B-mismatched with a patient and are potentially suitable for selection.

4.3. HLA-C

With the exception of the C*03:03/C*03:04 mismatch [36], all other incompatibilities at this locus represent a significant risk factor for both morbidity and mortality, to a similar extent as those regarding HLA-A and -B, despite the lower surface expression, which is nevertheless heterogeneous across distinct HLA-C allotypes [37]. While the C*03:03/C*03:04 mismatch is not associated with impaired survival and can be considered permissive, both allelic and antigen incompatibilities at locus C are similarly detrimental for transplant outcomes [36] and should be considered equivalent in terms of donor choice. Due to lower surface expression levels, mismatches for the allotypes C*03, C*07 or C*17 might be better tolerated than others [37].

4.4. HLA-DRB1

Being the most expressed among all class II alleles [42], HLA-DRB1 is rarely mismatched in unrelated allo-HSCT, as shown in early studies that reported a significant negative impact of any mismatch at this locus [43], driving the donor search, still focused on DRB1-matched unrelated donor first. As a consequence, the cohorts of DRB1-mismatched patient–donor pairs are quite small, and evidence on any effect is rather low. Only one study [32] found higher mortality in the presence of a DRB1 mismatch, without increase in acute GvHD (Table 1). According to one [24] of the two previously cited meta-analyses, significant higher mortality is associated with mismatch at DRB1 (HR 1.19, 95% CI: 1.07–1.32) but appears to be better tolerated than class I incompatibilities, these latter being associated with hazard ratios ranging from 1.23 to 1.33 [24].

4.5. HLA-DRB3/4/5

Less investigated and not routinely typed, the HLA-DRB3/4/5 genes appear to be relevant for transplant outcomes. Indeed, two studies so far demonstrated significant impaired survival [39] and increased acute GvHD [38] when mismatches at these genes are present. A previous collaborative analysis suggested a significant role for the DRB4 locus [44]. Moreover, mismatches at HLA-DRB3/4/5 should be avoided in the presence of multiple other incompatibilities (see below, “Low-expression loci”).

4.6. HLA-DQB1

Incompatibilities at HLA-DQB1 seem to be less relevant than the other mismatches [20,31,32], possibly due to either a lower expression vs. that of HLA-DRB1 or few patient–donor pairs that are DQB1-mismatched and DRB1-matched (strong linkage), preventing robust findings. The available data suggest that mismatches at DQB1 might be preferred over those at DRB1 or at class I alleles.

4.7. HLA-DPB1

Consistent evidence, accumulated over years and by different groups, supports the inclusion of DPB1 in the selection process [45], taking into account the permissiveness of mismatches at DPB1 according to the T-cell epitope or the expression model [46]. The last CIBMTR guidelines [19] include the algorithm based on the T-cell epitope model in order to avoid a non-permissive unrelated donor, currently avoidable in at least 71% of cases [47], although a higher fraction is expected whenever HLA-DPB1 is typed upfront for all potential unrelated donors, as it is the case with NGS typing. Notably, although the benefit of avoiding a non-permissive donor is less pronounced for one-locus-mismatched allo-HSCT [14], this selection is still recommended when two or more 9/10 (or 7/8) HLA-matched donors are potentially suitable in the absence of a 10/10 (8/8) HLA-matched one [19].

4.8. Low-Expression Loci (DQ, DRB3/4/5, DP)

In addition to studies on the “highly expressed” HLA-A, -B, -C and -DRB1 loci, a CIBMTR study addressed the question of whether cumulative mismatches at the “low-expression” loci DRB3/4/5, DQB1 and DPB1 matter [40]. Interestingly, higher morbidity and mortality risks are associated with three or more DRB3/4/5, DQB1 or DPB1 incompatibilities if one A, B, C or DRB1 incompatibility is present (7/8 HLA-matched donor). In the 8/8 HLA-matched group, mismatches at the low-expression loci were not associated with any adverse outcome [40].

5. Directionality

The vector of an HLA mismatch is bidirectional unless the patient or the donor are homozygous for the mismatched locus; then, host-versus-graft (HvG) or graft-versus-host (GvH) directions characterize the mismatch, respectively. A single study, conducted on myeloablative allo-HSCTs [48], found that unidirectional GvH vector mismatches are comparable to bidirectional ones in terms of clinical risk, which is higher than for unidirectional HvG, thus suggesting that for an HLA homozygous patient, a donor with a mismatch at this homozygous locus (HvG vector) is preferable over a donor with a mismatch at other loci [48].

6. HLA Allele Combinations

Other approaches have been applied in the allo-HSCT setting in the attempt of identifying combinations of HLA mismatches having different immunogenic implications and thus hypothetically carrying distinct immunological risks [49,50,51,52,53]. These approaches, mainly based on the characteristics of the epitope mismatches or on the similarity of the haplotypes from both individuals, stand on the assumption that the more the HLA haplotypes are similar (in both evolutionary and immunological terms), the less alloreactivity is expected after allo-HSCT, finally resulting in better prognosis [54]. Such models would be intended to be more useful in supporting the unrelated donor selection as the number of HLA incompatibilities increases, as it occurs for organ transplantation that is the setting where most models come from. However, none of them has been included in the current search recommendations to date. Very recently, HLA evolutionary divergence, a measure of haplotype difference based on evolutionary distance, was shown to predict the prognosis of pediatric and young adult patients undergoing allo-HSCT from matched and mismatched unrelated donors [55], strengthening the concept that not all HLA mismatches are equals and opening to innovative ways to look at HLA incompatibilities.

7. Mismatches Outside the ARD

NGS allelic resolution typing provides the full nucleotide sequence of all exons and, for class I genes, noncoding regions. Incompatibilities outside the ARD could potentially affect the allele expression (clinically relevant null alleles) and the indirect presentation of HLA-derived peptides; moreover, the regions outside the ARD are informative for haplotype assignment. A report from the National Marrow Donor Program network showed that diversities (including null alleles) outside the ARD are not frequent in fully matched donor–recipient pairs [56]. The impact of these incompatibilities is now under investigation. Another collaborative study found that DRB1*14:01 vs. *14:54 and DRB3*02:01 vs. *02:02 (two common pairs of alleles belonging to the same P-group) mismatches do not affect a transplant outcome [57]. Similarly, the B*44:02 vs. B*44:27 (B*44:02P) mismatch does not induce alloreactivity in vitro and could be permissive [58]. In a retrospective study on 10/10 matched unrelated transplants, fully matched pairs (i.e., 12/12) confirmed by ultra-high resolution (a technique which allows long-range extensive typing) showed a better survival with respect to pairs with any degree of mismatch at this level of resolution [18]. Most of the discrepant findings referred to mismatches in ARD previously not detected, and only 13 out of 810 carried diversity only in non-ARD exons (n = 3) and introns (n = 10) [59]. A validation multicenter study confirmed the detrimental impact of incompatibilities outside the ARD only on acute GvHD [60]. Taken together, all these data support the principle that the donor selection process should focus only on the sequence of ARD (match at P-group level) and include the common and well-defined null alleles [61].

8. HLA and Disease Relapse

Currently, there is no consistent evidence that HLA incompatibilities confer a reduction of the relapse risk without increasing non-relapse mortality; therefore, it is not routinely recommended to prefer a mismatched unrelated donor over a matched one in an attempt to enhance the graft-versus-malignancy effect, which instead has shown to be elicited by minor histocompatibility antigens [62]. However, a timely available donor, even if HLA-mismatched, may be a suitable choice for patients with high-risk disease and who are in need of urgent allo-HSCT. The impact of HLA mismatch on the outcome, indeed, is less pronounced when the disease risk is higher [63].
Conversely, an HLA mismatch between patient and donor is expected to provide a clinical benefit when KIR alloreactivity is present; therefore, it can be exploited as an adoptive immunotherapeutic strategy both in the unrelated and in the haploidentical setting [64,65].
It is worth mentioning the phenomenon of “HLA loss” in disease relapse after allo-HSCT from family haploidentical donors [66], leading to the selection of a new donor carrying a distinct HLA haplotype for further transplantation in order to provide post-transplant immunological pressure against those relapsed leukemic clones no longer expressing or downregulating the surface HLA molecules [67]. The downregulation of surface HLA proteins may also occur in relapsed leukemic cells after unrelated allo-HSCT [68,69].

9. Translation into Daily Practice

A pragmatic interpretation of the data from the reported studies allows for their application to the adult unrelated donors selection, a process requiring up-to-date knowledge and a tight cooperation between HLA laboratories, search coordinators and transplant physicians. At present, if multiple 10/10 HLA-matched unrelated donors are potentially available during the search, the donor selection should take into account the DPB1 and DRB3/4/5 loci. Whenever multiple 9/10 HLA-matched unrelated donors are potentially available without any 10/10 HLA-matched one, the selection process should aim at choosing those donors with the less detrimental incompatibilities with respect to clinical outcomes. These principles are summarized in Table 2.
In the setting of cord blood transplantation, the recommended HLA matching between patient and donor(s) is less stringent, and the number of HLA alleles usually typed is lower, as well as the respective resolution of the allowed incompatibilities. Unit–unit HLA match is not required for double cord transplantation [19].
Given the rapid advances in HLA typing and the major improvements in GvHD prophylaxis, as observed with high-dose post-transplant cyclophosphamide [70,71], for example, the current indications will probably evolve in the next future, driven by the impressive amount of HLA data provided by NGS and the constant increase of allo-HSCTs from unrelated and HLA-mismatched donors. The experience acquired with high-dose post-transplant cyclophosphamide for haploidentical transplants has prompted transplant physicians to enhance its administration in the HLA-mismatched unrelated setting in recent years [72,73], under the hypothesis that such GvHD prophylaxis would be superior to the “conventional” one based on calcineurin inhibitors, in the presence of one or more HLA incompatibilities. Similarly, considerations about the intensity of the conditioning regimen are warranted, since the expected organ toxicity is tightly linked to the risk of acute GvHD, acting as a potential trigger, enhanced by HLA incompatibilities.
Beyond HLA, the donor age has proven to be the most relevant factor. Although, to our knowledge, there is no consistent study suggesting a better outcome with younger HLA-mismatched donors over older well-matched ones, patient survival after allo-HSCTs has been reported to be higher with young unrelated donors than with older HLA-identical siblings [11]. As a consequence, donor age should be taken into great consideration during the search process, due to its robust and well-documented impact on patient outcome.
Finally, besides the fact that the technological improvements are expected to allow for NGS allelic resolution typing of patients and donors in all settings, including allo-HSCT from relative and cord blood units, it is worth noting that its use is particularly helpful during the search of adult unrelated donors, where all the above-cited considerations apply and where the large amount of HLA data obtained provide the basis for a potential improvement of patient prognosis.

10. Conclusions

The HLA system has proven to be crucial for the success of allo-HSCT; therefore, an appropriate donor selection process based on up-to-date knowledge and the cooperation between HLA laboratories, search coordinators and transplant physicians is of paramount importance. Here, we provided the most relevant evidence on the role of HLA incompatibilities according to the recent literature in the adult unrelated donor setting, in an attempt to guide all the involved stakeholders during the unrelated donor selection. The technological progresses in HLA typing together with the advances in the immunobiology of transplantation and the significant improvements in GvHD prophylaxis will probably lead this field to a rapidly evolution in the next future, with new and exciting perspectives on the horizon.

Author Contributions

R.C. performed the literature search and wrote the manuscript. G.R. supervised the literature search and provided scientific overview. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We thank all members of the directory board of the Associazione Italiana di Immunogenetica e Biologia dei Trapianti (AIBT). Part of this review was presented at the XVIII annual congress of AIBT in Parma (Italy), October 2022.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Shouval, R.; Fein, J.A.; Labopin, M.; Kröger, N.; Duarte, R.F.; Bader, P.; Chabannon, C.; Kuball, J.; Basak, G.W.; Dufour, C.; et al. Outcomes of allogeneic haematopoietic stem cell transplantation from HLA-matched and alternative donors: A European Society for Blood and Marrow Transplantation registry retrospective analysis. Lancet Haematol. 2019, 6, e573–e584. [Google Scholar] [CrossRef]
  2. Snowden, J.A.; Sánchez-Ortega, I.; Corbacioglu, S.; Basak, G.W.; Chabannon, C.; de la Camara, R.; Dolstra, H.; Duarte, R.F.; Glass, B.; Greco, R.; et al. Indications for haematopoietic cell transplantation for haematological diseases, solid tumours and immune disorders: Current practice in Europe, 2022. Bone Marrow Transplant. 2022, 57, 1217–1239. [Google Scholar] [CrossRef] [PubMed]
  3. Little, A.M.; Akbarzad-Yousefi, A.; Anand, A.; Diaz Burlinson, N.; Dunn, P.P.J.; 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. [Google Scholar] [CrossRef] [PubMed]
  4. WMDA Website. Available online: https://wmda.info/ (accessed on 2 December 2022).
  5. Yakoub-Agha, I.; Mesnil, F.; Kuentz, M.; Boiron, J.M.; Ifrah, N.; Milpied, N.; Chehata, S.; Esperou, H.; Vernant, J.P.; Michallet, M.; et al. Allogeneic marrow stem-cell transplantation from human leukocyte antigen-identical siblings versus human leukocyte antigen-allelic-matched unrelated donors (10/10) in patients with standard-risk hematologic malignancy: A prospective study from the French Society of Bone Marrow Transplantation and Cell Therapy. J. Clin. Oncol. 2006, 24, 5695–5702. [Google Scholar] [CrossRef] [PubMed]
  6. Saber, W.; Opie, S.; Rizzo, J.D.; Zhang, M.J.; Horowitz, M.M.; Schriber, J. Outcomes after matched unrelated donor versus identical sibling hematopoietic cell transplantation in adults with acute myelogenous leukemia. Blood 2012, 119, 3908–3916. [Google Scholar] [CrossRef] [Green Version]
  7. Shimoni, A.; Labopin, M.; Savani, B.; Byrne, M.; Volin, L.; Finke, J.; Niederwieser, D.; Ehninger, G.; Blaise, D.; Beelen, D.; et al. Comparable Long-Term Outcome after Allogeneic Stem Cell Transplantation from Sibling and Matched Unrelated Donors in Patients with Acute Myeloid Leukemia Older Than 50 Years: A Report on Behalf of the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation. Biol. Blood Marrow Transplant. 2019, 25, 2251–2260. [Google Scholar] [CrossRef] [PubMed]
  8. Lee, S.J.; Klein, J.; Haagenson, M.; Baxter-Lowe, L.A.; Confer, D.L.; Eapen, M.; Fernandez-Vina, M.; Flomenberg, N.; Horowitz, M.; Hurley, C.K.; et al. High-resolution donor-recipient HLA matching contributes to the success of unrelated donor marrow transplantation. Blood 2007, 110, 4576–4583. [Google Scholar] [CrossRef] [Green Version]
  9. Tiercy, J.M. How to select the best available related or unrelated donor of hematopoietic stem cells? Haematologica 2016, 101, 680–687. [Google Scholar] [CrossRef] [Green Version]
  10. Shaw, B.E.; Logan, B.R.; Spellman, S.R.; Marsh, S.G.E.; Robinson, J.; Pidala, J.; Hurley, C.; Barker, J.; Maiers, M.; Dehn, J.; et al. Development of an Unrelated Donor Selection Score Predictive of Survival after HCT: Donor Age Matters Most. Biol. Blood Marrow Transplant. 2018, 24, 1049–1056. [Google Scholar] [CrossRef] [Green Version]
  11. Kröger, N.; Zabelina, T.; de Wreede, L.; Berger, J.; Alchalby, H.; van Biezen, A.; Milpied, N.; Volin, L.; Mohty, M.; Leblond, V.; et al. Allogeneic stem cell transplantation for older advanced MDS patients: Improved survival with young unrelated donor in comparison with HLA-identical siblings. Leukemia 2013, 27, 604–609. [Google Scholar] [CrossRef]
  12. Zino, E.; Frumento, G.; Marktel, S.; Sormani, M.P.; Ficara, F.; Di Terlizzi, S.; Parodi, A.M.; Sergeant, R.; Martinetti, M.; Bontadini, A.; et al. A T-cell epitope encoded by a subset of HLA-DPB1 alleles determines nonpermissive mismatches for hematologic stem cell transplantation. Blood 2004, 103, 1417–1424. [Google Scholar] [CrossRef] [Green Version]
  13. Crocchiolo, R.; Zino, E.; Vago, L.; Oneto, R.; Bruno, B.; Pollichieni, S.; Sacchi, N.; Sormani, M.P.; Marcon, J.; Lamparelli, T.; et al. Nonpermissive HLA-DPB1 disparity is a significant independent risk factor for mortality after unrelated hematopoietic stem cell transplantation. Blood 2009, 114, 1437–1444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Fleischhauer, K.; Shaw, B.E.; Gooley, T.; Malkki, M.; Bardy, P.; Bignon, J.D.; Dubois, V.; Horowitz, M.M.; Madrigal, J.A.; Morishima, Y.; et al. Effect of T-cell-epitope matching at HLA-DPB1 in recipients of unrelated-donor haemopoietic-cell transplantation: A retrospective study. Lancet Oncol. 2012, 13, 366–374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Petersdorf, E.W.; Malkki, M.; O’hUigin, C.; Carrington, M.; Gooley, T.; Haagenson, M.D.; Horowitz, M.M.; Spellman, S.R.; Wang, T.; Stevenson, P. High HLA-DP Expression and Graft-versus-Host Disease. N. Engl. J. Med. 2015, 373, 599–609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Petersdorf, E.W.; Bengtsson, M.; De Santis, D.; Dubois, V.; Fleischhauer, K.; Gooley, T.; Horowitz, M.; Madrigal, J.A.; Malkki, M.; McKallor, C.; et al. Role of HLA-DP Expression in Graft-Versus-Host Disease After Unrelated Donor Transplantation. J. Clin. Oncol. 2020, 38, 2712–2718. [Google Scholar] [CrossRef]
  17. Shaw, B.E.; Marsh, S.G.; Mayor, N.P.; Russell, N.H.; Madrigal, J.A. HLA-DPB1 matching status has significant implications for recipients of unrelated donor stem cell transplants. Blood 2006, 107, 1220–1226. [Google Scholar] [CrossRef] [Green Version]
  18. Mayor, N.P.; Hayhurst, J.D.; Turner, T.R.; Szydlo, R.M.; Shaw, B.E.; Bultitude, W.P.; Sayno, J.R.; Tavarozzi, F.; Latham, K.; Anthias, C.; et al. Recipients Receiving Better HLA-Matched Hematopoietic Cell Transplantation Grafts, Uncovered by a Novel HLA Typing Method, Have Superior Survival: A Retrospective Study. Biol. Blood Marrow Transplant. 2019, 25, 443–450. [Google Scholar] [CrossRef] [Green Version]
  19. Dehn, J.; Spellman, S.; Hurley, C.K.; Shaw, B.E.; Barker, J.N.; Burns, L.J.; Confer, D.L.; 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. [Google Scholar] [CrossRef] [Green Version]
  20. Picardi, A.; Sacchi, N.; Miotti, V.; Lorentino, F.; Oldani, E.; Rambaldi, A.; Sessa, M.; Bruno, B.; Cerno, M.; Vago, L.; et al. Allelic HLA Matching and Pair Origin Are Favorable Prognostic Factors for Unrelated Hematopoietic Stem Cell Transplantation in Neoplastic Hematologic Diseases: An Italian Analysis by the Gruppo Italiano Trapianto di Cellule Staminali e Terapie Cellulari, Italian Bone Marrow Donor Registry, and Associazione Italiana di Immunogenetica e Biologia dei Trapianti. Transplant. Cell Ther. 2021, 27, e1–e406. [Google Scholar] [CrossRef]
  21. Fürst, D.; Bindja, J.; Arnold, R.; Herr, W.; Schwerdtfeger, R.; Müller, C.H.; Recker, K.; Schrezenmeier, H.; Mytilineos, J. HLA-E polymorphisms in hematopoietic stem cell transplantation. Tissue Antigens 2012, 79, 287–290. [Google Scholar] [CrossRef]
  22. Neuchel, C.; Gowdavally, S.; Tsamadou, C.; Platzbecker, U.; Sala, E.; Wagner-Drouet, E.; Valerius, T.; Kröger, N.; Wulf, G.; Einsele, H.; et al. Higher risk for chronic graft-versus-host disease (GvHD) in HLA-G mismatched transplants following allogeneic hematopoietic stem cell transplantation: A retrospective study. HLA 2022, 100, 349–360. [Google Scholar] [CrossRef]
  23. Kekre, N.; Mak, K.S.; Stopsack, K.H.; Binder, M.; Ishii, K.; Brånvall, E.; Cutler, C.S. Impact of HLA-Mismatch in Unrelated Donor Hematopoietic Stem Cell Transplantation: A Meta-Analysis. Am. J. Hematol. 2016, 91, 551–555. [Google Scholar] [CrossRef] [Green Version]
  24. Tie, R.; Zhang, T.; Yang, B.; Fu, H.; Han, B.; Yu, J.; Tan, Y.; Huang, H. Clinical implications of HLA locus mismatching in unrelated donor hematopoietic cell transplantation: A meta-analysis. Oncotarget 2017, 8, 27645–27660. [Google Scholar] [CrossRef] [Green Version]
  25. Iacobelli, S.; EBMT Statistical Committee. Suggestions on the use of statistical methodologies in studies of the European Group for Blood and Marrow Transplantation. Bone Marrow Transplant. 2013, 48 (Suppl. 1), S1–S37. [Google Scholar] [CrossRef] [Green Version]
  26. Glucksberg, H.; Storb, R.; Fefer, A.; Buckner, C.D.; Neiman, P.E.; Clift, R.A.; Lerner, K.G.; Thomas, E.D. Clinical manifestations of graft-versus-host disease in human recipients of marrow from HL-A-matched sibling donors. Transplantation 1974, 18, 295–304. [Google Scholar] [CrossRef]
  27. Souchet, L.; Masouridi, S.; Marçais, A.; Ibrahim, A.; Chauvel, C.; Turquet, E.; Derail, M.; Yakoub-Agha, I.; Crocchiolo, R. Diagnosis, prophylaxis and therapeutic management of acute GVH: Recommendations of the Société Francophone de Greffe de Moelle et Thérapie Cellulaire (SFGM-TC). Bull. Cancer 2022, 48, S1–S37. [Google Scholar] [CrossRef]
  28. Ferrara, G.B.; Bacigalupo, A.; Lamparelli, T.; Lanino, E.; Delfino, L.; Morabito, A.; Parodi, A.M.; Pera, C.; Pozzi, S.; Sormani, M.P.; et al. Bone marrow transplantation from unrelated donors: The impact of mismatches with substitutions at position 116 of the human leukocyte antigen class I heavy chain. Blood 2001, 98, 3150–3155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Kawase, T.; Morishima, Y.; Matsuo, K.; Kashiwase, K.; Inoko, H.; Saji, H.; Kato, S.; Juji, T.; Kodera, Y.; Sasazuki, T.; et al. High-risk HLA allele mismatch combinations responsible for severe acute graft-versus-host disease and implication for its molecular mechanism. Blood 2007, 110, 2235–2241. [Google Scholar] [CrossRef] [Green Version]
  30. Pidala, J.; Lee, S.J.; Ahn, K.W.; Spellman, S.; Wang, H.L.; Aljurf, M.; Askar, M.; Dehn, J.; Fernandez Viña, M.; Gratwohl, A.; et al. Nonpermissive HLA-DPB1 mismatch increases mortality after myeloablative unrelated allogeneic hematopoietic cell transplantation. Blood 2014, 124, 2596–2606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Morishima, Y.; Kashiwase, K.; Matsuo, K.; Azuma, F.; Morishima, S.; Onizuka, M.; Yabe, T.; Murata, M.; Doki, N.; Eto, T.; et al. Biological significance of HLA locus matching in unrelated donor bone marrow transplantation. Blood 2015, 125, 1189–1197. [Google Scholar] [CrossRef]
  32. Fürst, D.; Müller, C.; Vucinic, V.; Bunjes, D.; Herr, W.; Gramatzki, M.; Schwerdtfeger, R.; Arnold, R.; Einsele, H.; Wulf, G.; et al. High-resolution HLA matching in hematopoietic stem cell transplantation: A retrospective collaborative analysis. Blood 2013, 122, 3220–3229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Woolfrey, A.; Klein, J.P.; Haagenson, M.; Spellman, S.; Petersdorf, E.; Oudshoorn, M.; Gajewski, J.; Hale, G.A.; Horan, J.; Battiwalla, M.; et al. HLA-C antigen mismatch is associated with worse outcome in unrelated donor peripheral blood stem cell transplantation. Biol. Blood Marrow Transplant. 2011, 17, 885–892. [Google Scholar] [CrossRef] [Green Version]
  34. Gooley, T.A.; Chien, J.W.; Pergam, S.A.; Hingorani, S.; Sorror, M.L.; Boeckh, M.; Martin, P.J.; Sandmaier, B.M.; Marr, K.A.; Appelbaum, F.R.; et al. Reduced mortality after allogeneic hematopoietic-cell transplantation. N. Engl. J. Med. 2010, 363, 2091–2101. [Google Scholar] [CrossRef] [Green Version]
  35. Apps, R.; Meng, Z.; Del Prete, G.Q.; Lifson, J.D.; Zhou, M.; Carrington, M. Relative expression levels of the HLA class-I proteins in normal and HIV-infected cells. J. Immunol. 2015, 194, 3594–3600. [Google Scholar] [CrossRef] [Green Version]
  36. Fernandez-Viña, M.A.; Wang, T.; Lee, S.J.; Haagenson, M.; Aljurf, M.; Askar, M.; Battiwalla, M.; Baxter-Lowe, L.A.; Gajewski, J.; Jakubowski, A.A.; et al. Identification of a permissible HLA mismatch in hematopoietic stem cell transplantation. Blood 2014, 123, 1270–1278. [Google Scholar] [CrossRef]
  37. Petersdorf, E.W.; Gooley, T.A.; Malkki, M.; Bacigalupo, A.P.; Cesbron, A.; Du Toit, E.; Ehninger, G.; Egeland, T.; Fischer, G.F.; Gervais, T.; et al. HLA-C expression levels define permissible mismatches in hematopoietic cell transplantation. Blood 2014, 124, 3996–4003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Ducreux, S.; Dubois, V.; Amokrane, K.; Yakoub-Agha, I.; Labalette, M.; Michallet, M.; Rubio, M.T.; Kennel, A.; Forcade, E.; Lafarge, X.; et al. HLA-DRB3/4/5 mismatches are associated with increased risk of acute GVHD in 10/10 matched unrelated donor hematopoietic cell transplantation. Am. J. Hematol. 2018. online ahead of print. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Tsamadou, C.; Engelhardt, D.; Platzbecker, U.; Sala, E.; Valerius, T.; Wagner-Drouet, E.; Wulf, G.; Kröger, N.; Murawski, N.; Einsele, H.; et al. HLA-DRB3/4/5 Matching Improves Outcome of Unrelated Hematopoietic Stem Cell Transplantation. Front. Immunol. 2021, 12, 771449. [Google Scholar] [CrossRef]
  40. Fernández-Viña, M.A.; Klein, J.P.; Haagenson, M.; Spellman, S.R.; Anasetti, C.; Noreen, H.; Baxter-Lowe, L.A.; Cano, P.; Flomenberg, N.; Confer, D.L.; et al. Multiple mismatches at the low expression HLA loci DP, DQ, and DRB3/4/5 associate with adverse outcomes in hematopoietic stem cell transplantation. Blood 2013, 121, 4603–4610. [Google Scholar] [CrossRef] [Green Version]
  41. Petersdorf, E.W.; Stevenson, P.; Bengtsson, M.; De Santis, D.; Dubois, V.; Gooley, T.; Horowitz, M.; Hsu, K.; Madrigal, J.A.; Malkki, M.; et al. HLA-B leader and survivorship after HLA-mismatched unrelated donor transplantation. Blood 2020, 136, 362–369. [Google Scholar] [CrossRef]
  42. Brooks, C.F.; Moore, M. Differential MHC class II expression on human peripheral blood monocytes and dendritic cells. Immunology 1988, 63, 303–311. [Google Scholar] [PubMed]
  43. Petersdorf, E.W.; Longton, G.M.; Anasetti, C.; Martin, P.J.; Mickelson, E.M.; Smith, A.G.; Hansen, J.A. The significance of HLA-DRB1 matching on clinical outcome after HLA-A, B, DR identical unrelated donor marrow transplantation. Blood 1995, 86, 1606–1613. [Google Scholar] [CrossRef] [Green Version]
  44. Detrait, M.; Morisset, S.; Chalandon, Y.; Yakoub-Agha, I.; Dufossé, F.; Labalette, M.; Top, I.; Elsermans, V.; Barraco, F.; Quintela, A.; et al. Suggestive evidence of a role of HLA-DRB4 mismatches in the outcome of allogeneic hematopoietic stem cell transplantation with HLA-10/10-matched unrelated donors: A French-Swiss retrospective study. Bone Marrow Transplant. 2015, 50, 1316–1320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Crocchiolo, R.; Mele, L.; Testi, M.; Scollo Chiara, M.; Murgia, B.; Rossi, A.; Vecchiato, C.; Grammatico, P.; Mininni, D.; Longhi, E.; et al. Use of DPB1 T-cell epitope algorithm among italian transplant centers: A survey on behalf of Associazione Italiana di Immunogenetica e Biologia dei Trapianti. HLA 2021, 98, 114–121. [Google Scholar] [CrossRef] [PubMed]
  46. Fleischhauer, K. Immunogenetics of HLA-DP—A New View of Permissible Mismatches. N. Engl. J. Med. 2015, 373, 669–672. [Google Scholar] [CrossRef]
  47. Crocchiolo, R.; Girgenti, D.; D’Amico, F.; Cornacchini, G.; Lando, G.; De Marco, B.; Magliano, G.; Grillo, G.; Rossini, S. Potential selection of unrelated donor based on HLA-DPB1 T-cell epitope matching using data from a single-center analysis. Bone Marrow Transplant. 2022, 57, 1626–1628. [Google Scholar] [CrossRef]
  48. Hurley, C.K.; Woolfrey, A.; Wang, T.; Haagenson, M.; Umejiego, J.; Aljurf, M.; Askar, M.; Battiwalla, M.; Dehn, J.; Horan, J.; et al. The impact of HLA unidirectional mismatches on the outcome of myeloablative hematopoietic stem cell transplantation with unrelated donors. Blood 2013, 121, 4800–4806. [Google Scholar] [CrossRef]
  49. Lazaryan, A.; Wang, T.; Spellman, S.R.; Wang, H.L.; Pidala, J.; Nishihori, T.; Askar, M.; Olsson, R.; Oudshoorn, M.; Abdel-Azim, H.; et al. Human leukocyte antigen supertype matching after myeloablative hematopoietic cell transplantation with 7/8 matched unrelated donor allografts: A report from the Center for International Blood and Marrow Transplant Research. Haematologica 2016, 101, 1267–1274. [Google Scholar] [CrossRef]
  50. Geneugelijk, K.; Thus, K.A.; van Deutekom, H.W.M.; Calis, J.J.A.; Borst, E.; Keşmir, C.; Oudshoorn, M.; van der Holt, B.; Meijer, E.; Zeerleder, S.; et al. Exploratory Study of Predicted Indirectly ReCognizable HLA Epitopes in Mismatched Hematopoietic Cell Transplantations. Front. Immunol. 2019, 10, 880. [Google Scholar] [CrossRef]
  51. Spellman, S.; Klein, J.; Haagenson, M.; Askar, M.; Baxter-Lowe, L.A.; He, J.; Hsu, S.; Blasczyk, R.; Hurley, C. Scoring HLA class I mismatches by HistoCheck does not predict clinical outcome in unrelated hematopoietic stem cell transplantation. Biol. Blood Marrow Transplant. 2012, 18, 739–746. [Google Scholar] [CrossRef]
  52. Roerden, M.; Nelde, A.; Heitmann, J.S.; Klein, R.; Rammensee, H.G.; Bethge, W.A.; Walz, J.S. HLA Evolutionary Divergence as a Prognostic Marker for AML Patients Undergoing Allogeneic Stem Cell Transplantation. Cancers 2020, 12, 1835. [Google Scholar] [CrossRef] [PubMed]
  53. Camacho-Bydume, C.; Wang, T.; Sees, J.A.; Fernandez-Viña, M.; Abid, M.B.; Askar, M.; Beitinjaneh, A.; Brown, V.; Castillo, P.; Chhabra, S.; et al. Specific Class I HLA Supertypes but Not HLA Zygosity or Expression Are Associated with Outcomes following HLA-Matched Allogeneic Hematopoietic Cell Transplant: HLA Supertypes Impact Allogeneic HCT Outcomes. Transplant. Cell Ther. 2021, 27, e1–e142. [Google Scholar] [CrossRef] [PubMed]
  54. Petersdorf, E.W.; Malkki, M.; Gooley, T.A.; Martin, P.J.; Guo, Z. MHC haplotype matching for unrelated hematopoietic cell transplantation. PLoS Med. 2007, 4, e8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Merli, P.; Crivello, P.; Strocchio, L.; Pinto, R.M.; Algeri, M.; Del Bufalo, F.; Pagliara, D.; Becilli, M.; Carta, R.; Gaspari, S.; et al. Human leukocyte antigen evolutionary divergence influences outcomes of paediatric patients and young adults affected by malignant disorders given allogeneic haematopoietic stem cell transplantation from unrelated donors. Br. J. Haematol. 2022. online version of record before inclusion in an issue. [Google Scholar] [CrossRef]
  56. Hou, L.; Vierra-Green, C.; Lazaro, A.; Brady, C.; Haagenson, M.; Spellman, S.; Hurley, C.K. Limited HLA sequence variation outside of antigen recognition domain exons of 360 10 of 10 matched unrelated hematopoietic stem cell transplant donor-recipient pairs. HLA 2017, 89, 39–46. [Google Scholar] [CrossRef] [Green Version]
  57. Pasi, A.; Crocchiolo, R.; Bontempelli, M.; Carcassi, C.; Carella, G.; Crespiatico, L.; Garbarino, L.; Mascaretti, L.; Mazzi, B.; Mazzola, G.; et al. The conundrum of HLA-DRB1*14:01/*14:54 and HLA-DRB3*02:01/*02:02 mismatches in unrelated hematopoietic SCT. Bone Marrow Transplant. 2011, 46, 916–922. [Google Scholar] [CrossRef] [Green Version]
  58. Bettens, F.; Schanz, U.; Tiercy, J.M. Lack of recognition of HLA class I mismatches outside α1/α2 domains by CD8+ alloreactive T lymphocytes: The HLA-B44 paradigm. Tissue Antigens 2013, 81, 414–418. [Google Scholar] [CrossRef]
  59. Hurley, C.K.; Spellman, S.; Dehn, J.; Barker, J.N.; Devine, S.; Fernandez-Vina, M.; Gautreaux, M.; Logan, B.; Maiers, M.; Mueller, C.; et al. Regarding “Recipients Receiving Better HLA-Matched Hematopoietic Cell Transplantation Grafts, Uncovered by a Novel HLA Typing Method, Have Superior Survival: A Retrospective Study”. Biol. Blood Marrow Transplant. 2019, 25, e268–e269. [Google Scholar] [CrossRef]
  60. Mayor, N.P.; Wang, T.; Lee, S.J.; Kuxhausen, M.; Vierra-Green, C.; Barker, D.J.; Auletta, J.; Bhatt, V.R.; Gadalla, S.M.; Gragert, L.; et al. Impact of Previously Unrecognized HLA Mismatches Using Ultrahigh Resolution Typing in Unrelated Donor Hematopoietic Cell Transplantation. J. Clin. Oncol. 2021, 39, 2397–2409. [Google Scholar] [CrossRef]
  61. Hurley, C.K.; Ng, J. Continue to focus clinical decision-making on the antigen recognition domain for the present. Hum. Immunol. 2019, 80, 79–84. [Google Scholar] [CrossRef]
  62. Kolb, H.J. Graft-versus-leukemia effects of transplantation and donor lymphocytes. Blood 2008, 112, 4371–4383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Crocchiolo, R.; Ciceri, F.; Fleischhauer, K.; Oneto, R.; Bruno, B.; Pollichieni, S.; Sacchi, N.; Sormani, M.P.; Fanin, R.; Bandini, G.; et al. HLA matching affects clinical outcome of adult patients undergoing haematopoietic SCT from unrelated donors: A study from the Gruppo Italiano Trapianto di Midollo Osseo and Italian Bone Marrow Donor Registry. Bone Marrow Transplant. 2009, 44, 571–577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Cooley, S.; Trachtenberg, E.; Bergemann, T.L.; Saeteurn, K.; Klein, J.; Le, C.T.; Marsh, S.G.; Guethlein, L.A.; Parham, P.; Miller, J.S.; et al. Donors with group B KIR haplotypes improve relapse-free survival after unrelated hematopoietic cell transplantation for acute myelogenous leukemia. Blood 2009, 113, 726–732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Ruggeri, L.; Vago, L.; Eikema, D.J.; de Wreede, L.C.; Ciceri, F.; Diaz, M.A.; Locatelli, F.; Jindra, P.; Milone, G.; Diez-Martin, J.L.; et al. Natural killer cell alloreactivity in HLA-haploidentical hematopoietic transplantation: A study on behalf of the CTIWP of the EBMT. Bone Marrow Transplant. 2021, 56, 1900–1907. [Google Scholar] [CrossRef] [PubMed]
  66. Vago, L.; Perna, S.K.; Zanussi, M.; Mazzi, B.; Barlassina, C.; Stanghellini, M.T.; Perrelli, N.F.; Cosentino, C.; Torri, F.; Angius, A.; et al. Loss of mismatched HLA in leukemia after stem-cell transplantation. N. Engl. J. Med. 2009, 361, 478–488. [Google Scholar] [CrossRef] [Green Version]
  67. Crucitti, L.; Crocchiolo, R.; Toffalori, C.; Mazzi, B.; Greco, R.; Signori, A.; Sizzano, F.; Chiesa, L.; Zino, E.; Lupo Stanghellini, M.T.; et al. Incidence, risk factors and clinical outcome of leukemia relapses with loss of the mismatched HLA after partially incompatible hematopoietic stem cell transplantation. Leukemia 2015, 29, 1143–1152. [Google Scholar] [CrossRef]
  68. Christopher, M.J.; Petti, A.A.; Rettig, M.P.; Miller, C.A.; Chendamarai, E.; Duncavage, E.J.; Klco, J.M.; Helton, N.M.; O’Laughlin, M.; Fronick, C.C.; et al. Immune Escape of Relapsed AML Cells after Allogeneic Transplantation. N. Engl. J. Med. 2018, 379, 2330–2341. [Google Scholar] [CrossRef]
  69. Jan, M.; Leventhal, M.J.; Morgan, E.A.; Wengrod, J.C.; Nag, A.; Drinan, S.D.; Wollison, B.M.; Ducar, M.D.; Thorner, A.R.; Leppanen, S.; et al. Recurrent genetic HLA loss in AML relapsed after matched unrelated allogeneic hematopoietic cell transplantation. Blood Adv. 2019, 3, 2199–2204. [Google Scholar] [CrossRef]
  70. Luznik, L.; O’Donnell, P.V.; Symons, H.J.; Chen, A.R.; Leffell, M.S.; Zahurak, M.; Gooley, T.A.; Piantadosi, S.; Kaup, M.; Ambinder, R.F.; et al. HLA-haploidentical bone marrow transplantation for hematologic malignancies using nonmyeloablative conditioning and high-dose, posttransplantation cyclophosphamide. Biol. Blood Marrow Transplant. 2008, 14, 641–650. [Google Scholar] [CrossRef] [Green Version]
  71. Bacigalupo, A. Post-transplant cyclophosphamide: Overcoming the HLA barrier to hematopoietic stem cell transplants. Haematologica 2022, 107, 1230–1231. [Google Scholar] [CrossRef]
  72. Baron, F.; Labopin, M.; Tischer, J.; Ciceri, F.; Raiola, A.M.; Blaise, D.; Sica, S.; Vydra, J.; Fanin, R.; Diez-Martin, J.L.; et al. Comparison of HLA-mismatched unrelated donor transplantation with post-transplant cyclophosphamide versus HLA-haploidentical transplantation in patients with active acute myeloid leukemia. Bone Marrow Transplant. 2022, 57, 1657–1663. [Google Scholar] [CrossRef] [PubMed]
  73. Shaw, B.E.; Jimenez-Jimenez, A.M.; Burns, L.J.; Logan, B.R.; Khimani, F.; Shaffer, B.C.; Shah, N.N.; Mussetter, A.; Tang, X.Y.; McCarty, J.M.; et al. Three-year outcomes in recipients of mismatched unrelated bone marrow donor transplants using post-transplant cyclophosphamide: Follow-up from a National Marrow Donor Program-sponsored prospective clinical trial. Transplant. Cell Ther. 2022, in press. [Google Scholar] [CrossRef] [PubMed]
Table 1. Clinical impact of HLA matching in the allo-HSCT setting as reported in most recent relevant studies.
Table 1. Clinical impact of HLA matching in the allo-HSCT setting as reported in most recent relevant studies.
HLA LocusMismatchArticlenHazard Ratio (95% CI)
OSGvHD II-IV
Amismatched vs. matchedWoolfrey 2011 [33]19331.17 (0.93–1.47)1.18 (0.94–1.50)
Furst 2013 [32]26461.43 (1.19–1.72)ND
Pidala 2014 [30]80031.3 (1.2–1.5)1.3 (1.2–1.5)
Morishima 2015 [31]78981.29 (1.17–1.42)1.18 (1.06–1.32)
Picardi 2021 [20]1788ND (p = n.s.)1.34 (1.04–1.74)
Kekre 2016 [23]13,4461.48 (1.19–1.86)ND
Bmismatched vs. matchedWoolfrey 2011 [33]19331.22 (0.90–1.67)1.11 (0.81–1.52)
Furst 2013 [32]26461.52 (1.20–1.93)ND
Pidala 2014 [30]80031.2 (1.0–1.4)1.3 (1.1–1.6)
Morishima 2015 [31]78981.27 (1.11–1.45)1.28 (1.11–1.48)
Picardi 2021 [20]1788ND (p = n.s.)2.02 (1.53–2.67)
Kekre 2016 [23]13,4461.45 (1.20–1.75)ND
B-leader mismatched: MT vs. TTPetersdorf 2020 [16]17,100ND1.11 (1.04–1.19)
B-leader mismatched: MM vs. TTPetersdorf 2020 [16]17,100ND1.11 (1.00–1.24)
Cmismatched vs. matchedWoolfrey 2011 [33]19331.41 (1.16–1.70) #1.12 (0.90–1.39)
Furst 2013 [32]26461.35 (1.17–1.56)ND
Pidala 2014 [30]80031.3 (1.2–1.5)1.1 (1.0–1.3)
Morishima 2015 [31]78981.21 (1.13–1.30)1.27 (1.17–1.37)
Picardi 2021 [20]1788ND (p = n.s.)ND (p = n.s.)
Kekre 2016 [23]13,4461.58 (1.23–2.01)ND
C03:03/C03:04 mismatchFernandez-Vina 2014 [36]73490.98 (0.78–1.23)0.97 (0.76–1.25)
Ag vs. All mismatch aT C*03 or C*07Petersdorf 2014 [37]1975ND1.07 (0.75–1.53)
residue 116 mismatch vs. matchPetersdorf 2014 [37]19751.12 (1.00–1.26)1.14 (0.91–1.41)
residue 77/80 mismatch vs. matchPetersdorf 2014 [37]19751.13 (1.01–1.26)1.02 (0.82–1.27)
DRB1 Woolfrey 2011 [33]19331.30 (0.87–1.94)1.60 (1.06–1.80)
Furst 2013 [32]26461.42 (1.10–1.82)ND
Pidala 2014 [30]80031.1 (0.9–1.3)1.2 (0.9–1.5)
Morishima 2015 [31]78981.09 (0.98–1.21)1.24 (1.11–1.39)
Kekre 2016 [23]13,4461.16 (0.84–1.59)ND
DRB3/4/5mismatched vs. matchedDucreux 2018 [38]19751.10 (0.91–1.33)1.43 (1.07–1.90)
DRB3/4/5mismatched vs. matchedTsamadou 2021 [39]34101.25 (1.02–1.54)1.16 (0.89–1.52)
DQB1mismatched vs. matchedFurst 2013 [32]26461.23 (1.00–1.51)ND
Morishima 2015 [31]78981.08 (0.97–1.19)1.09 (0.98–1.22)
Picardi 2021 [20]1788ND (p = n.s.)ND (p = n.s.)
Kekre 2016 [23]13,4460.95 (0.74–1.21)ND
DPB1single-mismatched vs. matchedPidala 2014 [30]8003ND1.4 (1.2–1.6)
double-mismatched vs. matchedPidala 2014 [30]8003ND1.6 (1.3–1.9)
matched vs. TCE permissiveFleischhauer 2012 [14]85390.96 (0.87–1.06)ND
TCE nonpermissive vs. permissiveFleischhauer 2012 [14]85391.15 (1.05–1.25)ND
TCE nonpermissive vs. permissivePidala 2014 [30]80031.2 (1.1–1.4)1.1 (1.0–1.3)
LEL (DRB3/4/5, DQ, DP)≥3 vs. 0 among 7/8Fernandez-Vina 2013 [40]38531.45 (1.06–1.96)ND
≥3 vs. 1 among 7/8Fernandez-Vina 2013 [40]38531.43 (1.09–1.87)ND
# antigen mismatch. List of abbreviations: MT: methionine–threonine genotype; MM: methionine–methionine genotype; TT: threonine–threonine genotype; TCE: T-cell epitope; ND: not done; OS: overall survival; GvHD: graft-versus-host disease. Respective p-value is statistically significant in bold.
Table 2. Suggestions for the selection of unrelated donors according to the HLA matching status.
Table 2. Suggestions for the selection of unrelated donors according to the HLA matching status.
(a) Multiple 10/10 (or 8/8) HLA-matched donors:
     (1) avoid T-cell epitope DPB1 non-permissive donors;
     (2) match at DRB3/4/5 if possible.
(b) Multiple single-locus mismatched (9/10 HLA-matched, or 7/8) donors:
     (1) avoid T-cell epitope DPB1 non-permissive donors;
     (2) prefer DQB1 (not applicable for 8/8) or C*03:03/C*03:04 mismatches if possible, otherwise:
     (3) select DRB1-mismatched donors if possible, otherwise:
     (4) select A-, B- or C-mismatched donors # (all comparable) if possible;
     (5) if 7/8, prefer no more than two low-expression loci mismatches;
     (6) if patient homozygous, select the donor mismatched at the homozygous loci (HvG vector).
# other than C*03:03/C*03:04.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Crocchiolo, R.; Rombolà, G. Human Leucocyte Antigen System and Selection of Unrelated Hematopoietic Stem Cell Donors: Impact of Patient–Donor (Mis)matching and New Challenges with the Current Technologies. J. Clin. Med. 2023, 12, 646. https://doi.org/10.3390/jcm12020646

AMA Style

Crocchiolo R, Rombolà G. Human Leucocyte Antigen System and Selection of Unrelated Hematopoietic Stem Cell Donors: Impact of Patient–Donor (Mis)matching and New Challenges with the Current Technologies. Journal of Clinical Medicine. 2023; 12(2):646. https://doi.org/10.3390/jcm12020646

Chicago/Turabian Style

Crocchiolo, Roberto, and Gianni Rombolà. 2023. "Human Leucocyte Antigen System and Selection of Unrelated Hematopoietic Stem Cell Donors: Impact of Patient–Donor (Mis)matching and New Challenges with the Current Technologies" Journal of Clinical Medicine 12, no. 2: 646. https://doi.org/10.3390/jcm12020646

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop