You are on page 1of 10

Received: 16 November 2020 | Revised: 12 December 2020 | Accepted: 20 December 2020

DOI: 10.1111/iji.12525

INVITED REVIEW

Matchmaker, matchmaker make me a match: Opportunities


and challenges in optimizing compatibility of HLA eplets in
transplantation

William Lemieux1 | Hossein Mohammadhassanzadeh1 | William Klement1,2 |


Claude Daniel3 | Ruth Sapir-Pichhadze1,4,5

1
Centre for Outcomes Research and
Evaluation (CORE), Research Institute of Abstract
McGill University Health Centre, Montréal, The development of donor-specific antibodies (DSAs) is a major complication in
QC, Canada
2 transplantation, which is associated with inferior graft survival, impaired quality of
Canadian Blood Services, Ottawa, Ontario,
Canada life, and increased healthcare costs. DSA develop upon recognition of nonself HLA
3
Division of Hematology, McGill University by the recipient's immune system. HLA molecules contain epitopes, which are the
Health Centre, Montréal, QC, Canada
4
surface regions of HLA molecules recognized by antibodies. HLAMatchmaker is an
Division of Nephrology and the Multi-
Organ Transplant Program, Royal Victoria algorithm for assessing donor:recipient HLA compatibility at the level of structurally
Hospital, McGill University Health Centre, defined HLA targets called eplets. The consideration of eplets, rather than the whole
Montréal, QC, Canada
5 HLA molecule, could offer some advantages when classifying the immune risk associ-
Department of Epidemiology, Biostatistics
and Occupational Health, McGill University, ated with particular donor:recipient pairs. Assessing compatibility at the level of HLA
Montréal, QC, Canada
eplets could decrease misclassification of post-transplant immune risk by improving
Correspondence specificity, when antibodies are confirmed to be directed against donor eplets miss-
Ruth Sapir-Pichhadze, Centre for Outcomes
ing from the recipient's repertoire of eplets. Consideration of eplets may also increase
Research and Evaluation (CORE), McGill
University Health Centre, Montréal, QC, the sensitivity of immune risk assessment, when identifying mismatched eplets that
Canada.
could give rise to new, not previously detected, donor-specific antibodies post-trans-
Email: ruth.sapir-pichhadze@mcgill.ca
plant. Eplet matching can serve as a rational strategy for immune risk mitigation.
Funding information
Herein, we review the evolution of HLA (in) compatibility assessment for organ al-
Fonds de Recherche du Québec - Santé,
Grant/Award Number: chercheur boursier location. We outline challenges in the implementation of eplet-based donor:recipient
clinician award (254386); Génome Québec,
matching, including unavailability of allele-level donor genotypes for 11 HLA loci at
Grant/Award Number: Precision Medicine
CanPREVENT AMR the time of organ allocation and difficulty in assessing the hierarchy of immune risk
associated with particular HLA eplet mismatches. Opportunities to address some of
the current shortcomings of donor genotyping and HLAMatchmaker are also dis-
cussed. While there is a demonstrated benefit in the application of HLAMatchmaker
for donor: recipient HLA (in)compatibility assessment, evolving long-read genotyping
methods, compilation of large data sets with allele-level genotypes, and standardiza-
tion of methods to verify eplets as determinants of immune-mediated injuries are
required before HLA eplet matching is implemented in organ allocation to improve
upon transplant outcomes.

Int J Immunogenet. 2021;48:135–144. wileyonlinelibrary.com/journal/iji© 2021 John Wiley & Sons Ltd | 135
136 | LEMIEUX et al.

KEYWORDS

allograft, histocompatibility, immune response, immunology, matching, transplantation, typing

1 | I NTRO D U C TI O N from the pool. Acknowledging this, transplant centres may use the
cumulative cPRA, which considers anti-HLA antibodies that have
Currently, the assessment of donors and candidates’ compatibil- been detected over the entire course of pre-transplant screening
ity relies on acceptable mismatch strategies, with unacceptable (Beausang et al., 2017; Tinckam et al., 2015). The cumulative cPRA
mismatches defined by the presence of preformed donor-specific is also dependent on the detection of antibodies, which is itself de-
antibodies (DSAs) targeting human leucocyte antigens (HLAs). Anti- pendent on laboratory practices. This dependency makes standard-
HLA antibodies are produced in response to previous exposure to ization of SAB assay analysis challenging (Reed et al., 2013). Factors,
HLA through blood transfusions, pregnancy and transplantation. including serum handling, choice of mean fluorescence intensity
While the verification of preformed antibodies relied historically on (MFI) threshold to determine the presence of anti-HLA antibody,
cell-dependent crossmatches, nowadays donor:recipient compatibil- and the specific test kit used, are critical to the final interpretation
ity is assessed by virtual crossmatch or flow cytometry crossmatch of the assay (Konvalinka & Tinckam, 2015). Further complicating the
(Tait, 2016; Tinckam, 2009). Complement-dependent cytotoxicity interpretation of SAB assays, is their sensitivity to issues intrinsic to
(CDC) assays inform the highest risk of immune-mediated injuries. the serum sample, such as inhibitory substances (referred to as the
On the other hand, flow cytometry and virtual crossmatches are ‘prozone’ effect) and bead saturation (Konvalinka & Tinckam, 2015;
considered more sensitive assays, allowing detection of lower titre Tambur et al., 2020; Tambur & Wiebe, 2018). Thus, the virtual cross-
and noncomplement-binding DSAs that pose a lower immune risk match is vulnerable to misclassification of candidates’ risk of recall
than that identified by positive CDC. In contrast to flow cytome- responses due to immune memory, which may yield diverse reper-
try and CDC, virtual crossmatches rely on knowledge of donor HLA toires of unacceptable mismatches.
genotype and are deemed positive when single antigen bead (SAB) While cPRA is informed by existing (and/or historic) anti-HLA
assays confirm presence of DSA (Tait, 2016; Tambur et al., 2020; antibodies, it fails to capture the risk of developing new, previously
Tinckam, 2009). unobserved, antibodies against donor HLA. When detected for the
first-time post-transplant, these antibodies are referred to as de
novo DSAs. Epitope analysis could overcome some of the limitations
2 | PA N E L R E AC TI V E A NTI B O D I E S A N D of cPRA as a determinant of historic, current and potentially de novo
O PP O RT U N ITI E S FO R CO N S I D E R ATI O N O F DSA (Duquesnoy, 2001; Terasaki et al., 1989). The focus of epitope
H L A E PITO PE S analysis is on specific polymorphic regions on the HLA that can bind
to antibodies. The availability of a panel of monoclonal anti-HLA
The implementation of virtual crossmatch is based on the verifica- antibodies has helped grow the understanding of the concept of
tion of panel reactive antibodies (PRAs). The PRA offers a score from epitopes (Tambur & Claas, 2015). Some epitopes may be restricted
zero to 100% to represent the proportion of the donor pool a given to target a specific HLA protein, but others are shared between
transplant candidate may have antibodies against. To determine the different proteins, with each allele coding for HLA protein and the
PRA score, transplant candidates undergo routine screening by SAB unique combination of epitopes associated with it. Epitopes that are
assays. This is often referred to as a calculated PRA (cPRA), which common to different HLA molecules explain how certain anti-HLA
considers the entire donor population rather than a select panel of antibodies may demonstrate cross-reactivity to proteins coded by
representative donors as in the case of cell-based PRA (Tinckam different alleles. Immunogenetics allow the recognition of epitope
et al., 2015). The introduction of cPRA served to limit the variabil- repertoire from the HLA genotype.
ity observed when relying on different cell-based panels (Kransdorf Structural analysis of crystalized HLA complexes allowed the
et al., 2017). Moreover, anti-HLA antibody screening in candidates identification of accessible amino acids on HLA molecules that
and the calculation of PRA have improved the capability to predict can be bound by antibodies (Duquesnoy, 2001; Gu et al., 2019).
cellular crossmatch results and, consequently, have resulted in a de- Based on this principle, Duquesnoy and colleagues developed
creased likelihood of transplantation in the presence of unaccepta- HLAMatchmaker, a structurally based matching program. Each HLA
ble HLA mismatches, rejection and graft failure (Cherikh et al., 2006; antigen was viewed as a string of eplets or short sequences involving
Morris et al., 2019; Roll et al., 2020; Tinckam, 2009). polymorphic amino acid residues in antibody-accessible positions.
Despite the advantages of cPRA and the virtual crossmatch, Initially, HLAMatchmaker determined which triplets, linear se-
some limitations must be noted. First, cPRA may be highly depen- quences of three amino acid residues, were different between donor
dent on the timing and context of the testing performed. Anti-HLA and recipient (Duquesnoy, 2001). In subsequent iterations of the al-
antibodies may wax and wane over time, resulting in a fluctuating gorithm, a revised concept of eplets was defined (Duquesnoy, 2006).
repertoire of unacceptable mismatches and, consequently, also re- This concept arose from the observation that, in addition to a func-
sulting in different proportions of potentially compatible donors tional epitope, which includes about 2–5 residues that dominate the
LEMIEUX et al. | 137

strength and specificity of binding with antibodies, HLA antigens reasonable chance of finding a suitable donor (Keith & Vranic, 2016).
also have structural epitopes consisting of 15–22 residues that pro- This can translate to extended wait times for transplantation. To in-
vide supplementary interactions that increase the stability of the crease access to transplantation for the above disadvantaged popu-
antigen–antibody complex. Different from triplets, eplets did not as- lation, Eurotransplant developed the Acceptable Mismatch (AM)
sume sequentially contiguous amino acids but rather structurally de- program (Claas et al., 1988) that allowed mismatches in HLA-A or
fined amino acid polymorphisms within proximity in the context of HLA-B when DSAs were not present against the donor HLA pro-
each protein's 3D configuration. As a result, many (but not all) eplets teins as determined by cell-based crossmatching (Heidt et al., 2015).
were also triplets. This updated definition (in the form of eplets) is Noninherited maternal antigens were also considered acceptable
considered a more accurate representation of the constituents of in the AM program, as tolerance of these HLA is supposed to be
functional epitopes. acquired during foetal development (Claas et al., 1988). To further
A comparison between the donor's repertoire of eplets with expand the donor pool, mismatches were also allowed at other HLA
that of the recipient's allows for the estimation of the cumula- loci, while simultaneously ensuring a negative crossmatch result
tive mismatch load, or the sum of eplet mismatches, by HLA locus (Claas & Doxiadis, 2009). Advances in immunogenetics have allowed
and/or class. To document the evolving appreciation of eplets and an increased understanding of amino acid variations and their role
their validation as epitopes through antibody verification, the HLA in DSA development. This was followed by an evolution of the AM
Epitope Registry (https://www.epreg​istry.com.br/) was established program to consider transplantation in the presence of 0–2 triplet
(Duquesnoy et al., 2018). The HLA Epitope Registry captures lists of mismatches (Heidt et al., 2015).
alleles associated with each eplet, provides data on whether a puta- The consideration of epitope compatibility in highly sensitized
tive eplet has associated antibody reactivity (indicating that it may patients has the potential to increase the specificity with which
be an immunologically recognized epitope) and outlines the valida- donors are classified as incompatible (Figure 1, Highly Sensitized).
tion method used for antibody testing. Practically, in addition to transplantation occurring on the rare occa-
As the biologically rational measure for donor:recipient HLA sions when HLA allele-compatible donor is identified, this strategy
compatibility assessment, consideration of HLA eplets could min- also enables organ allocation to highly sensitized patients despite
imize the misclassification of transplant candidate's immune risk. presence of mismatched HLA alleles, provided that the associated
Further, eplet analysis offers a comprehensive and ‘static’ assessment repertoire of eplets is compatible: as more allele combinations are
of HLA (in)compatibility, independent of time and MFI threshold for deemed compatible, this inevitably helps expand the donor pool.
detecting HLA antibodies. To date, however, the assessment of eplet Indeed, it has been shown that matching at the eplet level (rather
compatibility by the HLAMatchmaker algorithm has relied on the than the antigen level) allows for a higher probability of identify-
cumulative eplet mismatch load (Lim et al., 2018; Sapir-Pichhadze ing compatible donors for highly sensitized patients (Duquesnoy
et al., 2015; Wiebe et al., 2013). While eplet mismatch load has been et al., 2003). By drawing parallels from Eurotransplant's AM program
shown to be associated with inferior transplant outcomes, optimiz- and comparing to the current allocation strategy used in Australia,
ing epitope compatibility at the time of organ allocation requires Nguyen et al. were also able to determine that eplet matching was
consideration of risk informed by the immunogenicity and antigenic- beneficial for the highly sensitized patients, without being a bur-
ity of individual eplet mismatches. den and/or compromising access to transplantation on the part of
nonhighly sensitized transplant candidates (Nguyen et al., 2014).
Importantly, the AM program was shown to decrease rejection rates
3 | A S S E S S I N G I N CO M PATI B I LIT Y AT TH E in AM patients and unsensitized individuals (Heidt et al., 2019).
LE V E L O F H L A E PLE T S

3.1 | Applications in highly sensitized patients 3.2 | Applications across transplant candidate
populations
A consideration of HLA eplet-based donor and recipient compat-
ibility has been implemented by the Eurotransplant program to In addition to increased access to transplantation in highly sensitized
identify compatible donors for highly sensitized patients. In this patients, the assessment of eplet compatibility may offer advantages
context, patients with a PRA > 6% are deemed sensitized and across transplant candidates whether sensitized or not. The advan-
patients with PRA > 85% are deemed highly sensitized as they tage of this strategy can be attributed to its ability to explain (and help
express anti-HLA antibodies against more than 85% of the popu- avoid) not only preformed antibodies but also de novo DSA (Figure 1,
lation (Heidt et al., 2015). Highly sensitized patients are less likely PRA 0% and PRA > 0%). A perfect donor:recipient HLA allele match-
to access transplantation and be matched with a compatible donor ing may be clinically impractical in the presence of over 27,000 HLA
(Sebastiaan Heidt & Claas, 2018; Smits et al., 1998). Many groups alleles across 11 HLA genes. In contrast, by virtue of a smaller number
recognized the need to address the scarcity of donors available for of potential eplets which may be common to different HLA alleles of
highly sensitized patients and especially patients with a PRA nearing the same locus or across loci, accomplishing compatibility at the level
100% who would require exceedingly large donor pools to have a of the eplet is expected to be more feasible. When perfect matching
138 | LEMIEUX et al.

F I G U R E 1 Qualitative representation of the impact of transitioning from calculated panel reactive antigens (cPRA) to eplet-based cPRA
on assessment of immune risk. Assessing compatibility at the level of HLA epitopes could decrease misclassification of post-transplant
immune risk by improving specificity (when antibodies detected are linked to eplets missing from the recipient's repertoire) and sensitivity
(when identifying mismatched eplets that could give rise to immune-mediated injuries even in the absence of preformed antibodies). Specific
eplet mismatches may inform variable degrees of risk and this may be further modified given the role of T cells in the alloimmune response
as well as the recipient's HLA type [Colour figure can be viewed at wileyonlinelibrary.com]

is not possible to achieve, assessing (in)compatibility at the level of at all 11 loci of HLA genes (Class I (A, B, C) and Class II (DRB1,
HLA eplets can be used for risk stratification. Indeed, eplet mismatch DRB3/4/5, DQA1/B1, DPA1/B1)) is necessary to determine the
load has been linked to the development of de novo DSA, T-cell medi- candidate's and donor's eplet repertoire. HLA genotyping methods
ated rejection, transplant glomerulopathy and graft failure (Hamada can be classified into two broad categories: probe-based meth-
et al., 2020; Sapir-Pichhadze et al., 2015; Sapir-Pichhadze et al., 2020; ods and sequence-based methods. Probe-based methods include
Senev, Coemans, et al., 2020; Wiebe et al., 2019). While many studies sequence-specific primer (SSP) typing and sequence-specific oli-
focus on kidney transplantation, the link between eplet (in)compat- gonucleotide probe typing (SSO). These two techniques offer low-
ibility and patient outcomes remains consistent in the context of other cost rapid turnaround typing (Smith et al., 2019). Yet, both methods
solid organ transplants (Hamada et al., 2020; McCaughan et al., 2018; have the disadvantage of providing low or, at best, intermediate
Walton et al., 2016). Importantly, a better understanding of immune resolution genotyping, with residual ambiguity of allele assign-
risk associated with eplet incompatibility can inform surveillance ment. Of these two, SSO typing is more common in clinical prac-
schedules and personalized immunosuppression regimens in both tice as it can increase efficiency through batching multiple samples
adult and paediatric patients (Sharma et al., 2020; Wiebe et al., 2015; (Smith et al., 2019).
Wiebe & Nickerson, 2020; Wiebe et al., 2017). In contrast, in sequence-based genotyping, the specific allele
is determined by resolving the exact genomic sequence of alleles.
Sanger sequencing is often referred to as sequence-based typing
4 | ROA D M A P TO I M PLE M E NTATI O N (SBT) that enables high-resolution typing of HLA alleles. Next-
O F E PLE T M ATC H I N G — C H A LLE N G E S A N D generation sequencing (NGS) has reduced the infrequent inaccura-
O PP O RT U N ITI E S cies observed in SBT but is associated with higher implementation
costs, which can be a barrier to its integration in HLA typing labo-
4.1 | Measured and imputed HLA Genotypes ratories. Moreover, NGS is not immune to residual ambiguity with
occasional allele dropouts (Klasberg et al., 2019). The processing
The first step towards implementing eplet matching relies on the time required by SBT and NGS methods makes them less useful for
availability of allele-level HLA genotyping (Figure 2). Genotyping deceased donor genotyping. Therefore, imputation of allele-level
LEMIEUX et al. | 139

F I G U R E 2 Challenges and opportunities on path to incorporating HLA eplet compatibility in immune risk assessment and organ
allocation. Abbreviations: SAB, single antigen bead assay [Colour figure can be viewed at wileyonlinelibrary.com]

genotypes is often used for clinical decision making and for analysis would allow the rapid generation of high-resolution typing in a mat-
of large legacy data sets. ter of hours as required in the deceased donor transplant context
Imputation of allele-level genotypes from low or intermediate (Liu, 2020). Indeed recently, Mosbruger et al. reported on the appli-
resolution typing has been utilized to overcome HLA allele ambigu- cation of nanopore sequencing technology for the rapid (<6 hr) and
ity. The imputation process relies on probabilistic inference, which comprehensive characterization of 11 HLA loci increasing the likeli-
considers population-level allele and haplotype frequencies. Recent hood that this technology will be implemented in clinical practice to
studies have shown that imputations may result in inaccuracies in improve donor:recipient compatibility (Mosbruger et al., 2020).
allele and, consequently, in eplet assignments (D'Souza et al., 2018;
Engen et al., 2020; Senev, Emonds, et al., 2020). The inaccuracies
were more pronounced when applied to HLA class II and among pa- 4.2 | The issue of rare and null HLA alleles
tients of non-Caucasian self-reported ancestry. As with any tool that
is being used for research or clinical purposes, it is of paramount Apart from allele ambiguity, when relying on probe-based (and
importance to be familiar with its advantages and limitations in order even sequence-based) methods to assign allele-level genotypes, the
to apply the tool responsibly. Imputation accuracy is expected to im- eplet repertoire assignment may be limited in the case of rare al-
prove when 2-field allele and haplotype frequencies are available leles. An algorithm can be trained to automate and accelerate the
for the population considered and by providing the imputation tool incorporation of rare alleles, which are currently not represented in
the most detailed genotyping information possible (e.g. intermediate the HLAMatchmaker algorithm or the HLA Epitope Registry. This
resolution typing with NMDP ambiguity codes for 11 HLA loci). algorithm will identify nucleotide sequences that encode the same
With advances in genotyping technologies, it is expected that protein sequences for the peptide binding domains (exon 2 and exon
sequencing-based typing could be implemented with shorter turn- 3 for HLA class I and exon 2 for HLA class II alleles) as the more com-
around times or that an epitope-based typing strategy could be de- mon and well-documented alleles. Over time, the algorithm would
veloped. This could allow for the use of eplet matching for organ also be able to identify new polymorphic DNA sequences that can
allocation without imputation. Importantly, a novel approach has code for antibody targets with physiochemical properties within 3D
been designed using long-read nanopore sequencing (LNS). While HLA molecules that are likely to represent epitopes.
the long reads minimize inaccuracies introduced when assembling Another limitation of the current HLAMatchmaker algorithm is
short sequencing reads, this technique still suffers from some short- the way it handles null alleles (Elsner & Blasczyk, 2004). Null alleles
comings in read analysis and interpretation (Matern et al., 2020). are characterized by a lack of a serologically detectable product.
Should these deficiencies be resolved, the turnaround time of LNS While they should not represent a major issue, it is important to
140 | LEMIEUX et al.

acknowledge the existence of alleles with various degrees of ex- Fusion v4.4 and HLAMatchmaker v3.1 were also available). It is im-
pression in transplantation and how they may contribute to over- portant to ensure that as our knowledge on clinically relevant eplet
estimation (e.g. when donors have HLA proteins with null (N) or low repertoires evolves, changes are applied across all available software
(L) expression) or underestimation (e.g. when recipients have HLA packages consistently.
proteins with null (N) or low (L) expression) of donor:recipient HLA
eplet incompatibility (Elsner & Blasczyk, 2004). Currently, users of
the Excel version of HLAMatchmaker are instructed to maintain 4.5 | Steps towards verification of eplets
an ‘x’ (indicating no allele present) in the appropriate field for a null as epitopes
allele. This ensures that null alleles will not be counted as present.
Future updates of HLAMatchmaker should allow direct entry of null Although the analysis of eplet (in)compatibility enables risk strati-
alleles in their standard nomenclature format. fication, it is essential to define the repertoire of eplets that must
be prioritized for matching. One must acknowledge that on the one
hand, incompatibility of high-risk eplets, even in the absence of pre-
4.3 | Re-exposure to eplets and memory response formed HLA antibodies, can give rise to immune-mediated injuries.
On the other hand, efforts to secure donor:recipient compatibility
Eplet incompatibility predicts the potential for immune-mediated in- on clinically irrelevant or low-risk eplets may unjustifiably delay ac-
juries, and efforts are underway to identify the most immunogenic cess to transplantation in certain individuals. Promoting eplet-based
eplets (Hönger et al., 2020). Risk of immune-mediated injuries may organ allocation requires the verification of individual eplet mis-
be accentuated with re-exposure to similar HLA eplets. The cur- matches as biomarkers predictive of immune-mediated injury.
rent HLAMatchmaker algorithm does not consider prior sensitiz- To record clinically relevant eplets, Duquesnoy and collaborators
ing events and shared eplets between prior and current allografts. have initiated a process to document and track antibody-verified
Patient sensitization after transfusions and prior pregnancies are epitopes on the HLA Epitope Registry. However, the experimental
even harder to account for when considering HLA eplet compatibil- validation of each eplet in the registry varies; it includes verification
ity. Candidates for re-transplantation or those with prior exposure by human monoclonal antibodies, elution/absorption, mouse mono-
to eplets consistent with those of the new donor (e.g. because of clonal antibodies, and sera from multiparous women. Additionally,
pregnancy) may experience a memory response and immune injury. the opportunity for antibody verification may be limited because
Even in the absence of preformed DSA, avoiding repeated exposure of additional factors such as the collection of serum, choice of sol-
to mismatched eplets encountered with a prior donor could help re- id-phase assay kit for antibody detection, the repertoire and integrity
duce risk. Similarly, recognizing nonimmunogenic eplets, which are of epitopes represented on the assay, MFI threshold used to assign
unlikely to induce an antibody response and, thus, do not present a positive/negative antibody status, strategies for serum handling and
problem upon re-exposure, is also important. While applying a grow- antibody verification. There is a need for clarity and standardization
ing number of restrictions at the time of organ allocation may affect of experimental procedures required for epitope validation within
access to transplantation, mitigating the risk of immune-mediated laboratories and across transplant centres. Moreover, considering
injury may facilitate the expansion of the existing donor pool by the dynamic nature of some anti-HLA antibodies, it is also important
virtue of decreasing the number of candidates likely to require re- to study the impact of eplet mismatches on other clinical outcomes.
transplantation after a premature graft failure. In addition to popula- Such outcomes may include graft dysfunction, rejection and failure.
tion-based policies, analysis of risk versus benefit must consider the Further insights are also needed on how to prioritize compatibil-
context and preferences of the individual waitlisted patient. ity on specific eplets. For this purpose, one must estimate the weight
each eplet may carry (and in reference to other eplets) in predicting
immune risk when mismatched. Establishing whether some eplets,
4.4 | Software for assessing HLA eplet compatibility by virtue of their inherent properties or due to their combination
with other specific eplets, could be associated with an increased risk
The growing popularity of the concept of HLA eplet compatibility of immune injuries is an important step towards considering eplet
gave rise to the development of several software packages. Tassone compatibility in organ allocation.
et al conducted eplet analysis of the same donor:recipient pairs by Eplet mismatches may vary in their tendency to trigger an im-
the HLAMatchmaker program (v2.1) and OLI Fusion MatchMaker mune response. Varied immunogenicity may be determined by the
(v4.2) software. The different types of software used yielded dif- physiochemical properties of polymorphic amino acid sequences,
ferent total eplet mismatch loads, inconsistent eplet mismatch recipient HLA, and the presence or absence of accompanying
repertoires, and variable antibody verification status (Tassone T-helper cell epitopes. Kosmoliapsis et al outlined a strategy to de-
et al., 2020). At times, certain software also incorrectly assigned termine immunogenicity of HLA by comparing the physiochemical
eplets to HLA alleles. As our understanding of eplets improves, the properties of HLA proteins expressed in donors versus recipients
HLA Epitope Registry as well as Fusion and HLAMatchmaker un- (Kosmoliaptsis et al., 2009, 2011). The immunogenicity algorithm
dergo intermittent updates (at the time this manuscript was written, developed by Kosmoliapsis and colleagues informs the likelihood
LEMIEUX et al. | 141

of observing an alloantibody response by considering, in addition Traditionally, health-related information presents challenges
to the number of amino acid mismatches, the hydrophobicity and attributed to the collection, incompleteness, sparsity and limited
electrostatic mismatch scores. This topic has also been reviewed in sample size of medical data. While statistical methods struggle with
Ref. Tambur (2018). data representativeness, skewness of distributions and low signal-
It is also important to acknowledge the role of T-helper cell epi- to-noise ratio, machine learning methods spare no effort in counter-
+
topes in immunogenicity. CD4 T-helper cells are important for the ing the detrimental effect of such limitations. The advent of genomic
development of a productive antibody response. After recognition data, including NGS data, intensifies the complexity of data analysis
by the B-cell receptor, HLA can be internalized and degraded into due to substantial increases in dimensionality, greater variations be-
peptides. These peptides may then be presented on self HLA class tween patients and greater variations in patients’ response to vari-
II on B cells. Presentation of self or nonself allo-HLA-derived pep- ous treatments (Lee & Yoon, 2017). In the context of transplantation,
tides by recipient's class II HLA may determine whether a specific these variations become more crucial because they are multiplied
eplet mismatch will lead to an antibody response and whether a DSA and amplified by the number of permutations of possible HLA loci,
isotype switch would occur from IgM to IgG (Dankers et al., 2004; alleles and eplets observed in donors and recipients, notwithstand-
Kramer et al., 2017, 2019). Algorithms have been developed to con- ing the variations in the response to immunosuppressant therapies.
sider Predicted Indirectly Recognizable HLA Epitopes (PIRCHE-II) Methods of optimizing matching, allocation or management of trans-
binding to HLA class II (Otten et al., 2013). These represent antigens plants need to address all these challenges systematically to ensure
from donor HLA class I that can bind to the donor's class II HLA and that the outcome of an individual patient is optimized while at the
can lead to T-helper cell responses. PIRCHE-II is associated with an same time benefiting the entire candidate population given the avail-
increased risk of DSA development, rejection and graft loss (Hamada able donor pool.
et al., 2020; Meszaros et al., 2020; Zheng et al., 2019). There is a need to balance the risks of immune-mediated injuries
As all mismatches do not seem to have equivalent impact on risk with the probability of finding a suitable donor to avoid unnecessary
of immune-mediated injury, it is critical to elucidate the respective protraction of waiting time to transplantation. While decision-mak-
importance of particular eplets to prioritize matching accordingly. ers should be aware of the trade-offs involved, in parallel, the direc-
Efforts to establish relative immunogenicity of eplets have been put tives and procedures for organ allocation should concur with views,
forth by establishing the relative frequency of DSA against HLA-DQ values and preferences of the public. Strategies to ensure equity in
in the context of pregnancy, where mothers have up to 1 HLA-DQ access to transplantation when considering patients’ ancestry and
allele difference in reference to babies (Schawalder et al., 2020). immune risk require careful deliberation. After the implementation
With a similar objective, Tambur et al. conceived the 2MM1DSA par- of policies, an additional discussion may be needed with patients
adigm. Planning to study a cohort of patients transplanted across themselves as to their degree of risk they are willing to accept in
2 HLA-DQ mismatches and formed de novo DSA to one mismatch general given current organ allocation schemes and at a time spe-
(referred to as foe) but not the other (friend), the team plans to focus cific organ offers are put forth. Inevitably, such decisions will include
on mismatches exhibited by the allele informing the de novo DSA but will not be limited to eplet compatibility as additional donor and
development (Tambur et al., 2019). In addition to immunogenicity transplant characteristics may inform long-term outcomes in partic-
represented by DSA development, there is a need to study how ular patient subgroups.
specific class I and class II eplet mismatch may affect hard clinical
endpoints like graft survival at the population level. Distinguishing
between eplets associating with an increased risk versus those that 5 | CO N C LU S I O N
do not can determine lower risk eplets representing acceptable
mismatches. This knowledge is bound to increase the feasibility of In conclusion, eplet matching represents an important refinement
matching at the eplet level. of HLA compatibility assessment in organ transplantation with a
potential to mitigate immune-mediated injuries and prolong graft
survival. This review brought to notice some of the challenges on
4.6 | Informing feasibility of eplet matching path to ensuring eplet compatibility at the time of organ allocation,
including the current impracticality of timely high-resolution donor
The feasibility of matching at the level of the eplet can be estimated 11-loci HLA typing, the fact that the repertoire of antibody-verified
as the sum of the products of the probabilities of observing com- eplets in HLAMatchmaker is in flux, lack of data on the differential
binations of HLA haplotypes giving rise to the recipient's complete risk associated with particular eplet mismatches (informed by their
eplet repertoire. To avoid unnecessary stringency in access to trans- immunogenicity and antigenicity), and a sufficiently diverse donor
plantation, efforts may also focus on a smaller subset of higher risk pool to facilitate the optimization of HLA compatibility. This review
eplets. To establish the risk of transplant outcomes as a function of also highlighted opportunities to overcome some of the challenges
particular eplet mismatches, analyses should disentangle simultane- towards the consideration of eplet compatibility, including evolv-
ously observed eplet mismatch profiles informed by the donor's and ing rapid high-resolution genotyping techniques, standardization
recipient's repertoires of eplets. of antibody-verification efforts, and automation of eplet repertoire
142 | LEMIEUX et al.

and mismatch determination from genotyping data. Large-scale col- epitope database. Human Immunology, 80(2), 103–106. https://doi.
org/10.1016/j.humimm.2018.11.007
laborations are needed to ensure an adequate power to detect risks
Elsner, H. A., & Blasczyk, R. (2004). Immunogenetics of
associated with particular eplet mismatches and inform priorities for HLA null alleles: Implications for blood stem cell trans-
donor:recipient matching. plantation. Tissue Antigens, 64(6), 687–695. https://doi.
org/10.1111/j.1399-0039.2004.00322.x
Engen Rachel M., Jedraszko Aneta M., Conciatori Michael A., Tambur
AC K N OW L E D G E M E N T S
Anat R. (2020). Substituting imputation of HLA antigens for
This work was supported by Genome Canada Large Scale Applied high-resolution HLA typing: Evaluation of a multiethnic popu-
Research Program Award ‘Precision Medicine CanPREVENT AMR' lation and implications for clinical decision making in transplan-
funded by Genome Quebec, Genome British Columbia, Genome tation. American Journal of Transplantation, 1–9. http://dx.doi.
Alberta and Canadian Institutes of Health Research. R.S.-P. is also org/10.1111/ajt.16070
Gu, Y., Wong, Y. H., Liew, C. W., Chan, C. E. Z., Murali, T. M., Yap, J.,
supported by a Fonds de recherche du Quebec—Santé chercheur
Too, C. T., Purushotorman, K., Hamidinia, M., El Sahili, A., Goh, A.
boursier clinician award (grant no. 254386). T. H., Teo, R. Z. C., Wood, K. J., Hanson, B. J., Gascoigne, N. R. J.,
Lescar, J., Vathsala, A., & MacAry, P. A. (2019). Defining the structural
ORCID basis for human alloantibody binding to human leukocyte antigen
allele HLA-A*11:01. Nature Communications, 10(1), 893. https://doi.
William Lemieux https://orcid.org/0000-0001-5684-9773
org/10.1038/s4146​7-019-08790​-1
Ruth Sapir-Pichhadze https://orcid.org/0000-0003-0745-004X Hamada, S., Dumortier, J., Thévenin, C., Pageaux, G.-P., Faure, S.,
Guillaud, O., Boillot, O., Lachaux, A., Luscalov, D.-A., Dubois, V.,
REFERENCES & Meszaros, M. (2020). Predictive value of HLAMatchmaker
and PIRCHE-II scores for de novo donor-specific antibody
Beausang, J. F., Fan, H. C., Sit, R., Hutchins, M. U., Jirage, K., Curtis, R.,
formation after adult and pediatric liver transplantation.
Hutchins, E., Quake, S. R., & Yabu, J. M. (2017). B cell repertoires in
Transplant Immunology, 61, 101306. https://doi.org/10.1016/j.
HLA-sensitized kidney transplant candidates undergoing desensiti-
trim.2020.101306
zation therapy. Journal of Translational Medicine, 15(1), 9. https://doi.
Heidt, S., & Claas, F. H. J. (2018). Transplantation in highly sensi-
org/10.1186/s1296​7-017-1118-7
tized patients: Challenges and recommendations. Expert Review of
Cherikh, W., Baker, T., Nelson, K., Land, G., Leffell, M., & Nikaein, A.
Clinical Immunology, 14(8), 673–679. https://doi.org/10.1080/17446​
(2006). Broader geographic sharing through accurate prediction of
66X.2018.1498335
crossmatch results. Transplantation, 82(1), 360.
Heidt, S., Haasnoot, G. W., Witvliet, M. D., Linden-van Oevelen, M. J. H.,
Claas, F. H., & Doxiadis, I. I. (2009). Management of the highly sensitized
Kamburova, E. G., Wisse, B. W., Joosten, I., Allebes, W. A., Meer, A.,
patient. Current Opinion in Immunology, 21(5), 569–572. https://doi.
Hilbrands, L. B., Baas, M. C., Spierings, E., Hack, C. E., Reekum, F. E.,
org/10.1016/j.coi.2009.07.010
Zuilen, A. D., Verhaar, M. C., Bots, M. L., Drop, A. C. A. D., Plaisier, L.,
Claas, F., Gijbels, Y., D'Amaro, J., Hendriks, G., & Persijn, G. (1988). A
… Claas, F. H. J. (2019). Allocation to highly sensitized patients based
special strategy to increase the chance of finding cross-match neg-
on acceptable mismatches results in low rejection rates comparable
ative kidneys for highly sensitized patients. Paper presented at the
to nonsensitized patients. American Journal of Transplantation, 19(10),
Transplantation Proceedings
2926–2933. https://doi.org/10.1111/ajt.15486
Claas, F. H., Gijbels, Y., van der Velden-de Munck, J., & van Rood,
Heidt, S., Witvliet, M. D., Haasnoot, G. W., & Claas, F. H. (2015). The 25th
J. J. (1988). Induction of B cell unresponsiveness to noninher-
anniversary of the Eurotransplant acceptable mismatch program
ited maternal HLA antigens during fetal life. Science, 241(4874),
for highly sensitized patients. Transplant Immunology, 33(2), 51–57.
1815–1817.
https://doi.org/10.1016/j.trim.2015.08.006
Dankers, M. K., Roelen, D. L., Nagelkerke, N. J., de Lange, P., Persijn, G.
Hönger, G., Niemann, M., Schawalder, L., Jones, J., van Heck, M. R., van
G., Doxiadis, I. I., & Claas, F. H. (2004). The HLA-DR phenotype of the
de Pasch, L. A. L.,Vendelbosch, S., Rozemuller, E. H., Hösli, I., Blümel,
responder is predictive of humoral response against HLA class I an-
S., & Schaub, S. (2020). Toward defining the immunogenicity of HLA
tigens. Human Immunology, 65(1), 13–19. https://doi.org/10.1016/j.
epitopes: Impact of HLA class I eplets on antibody formation during
humimm.2003.09.017
pregnancy. HLA, 96(5), 589–600.
D'Souza, Y., Ferradji, A., Saw, C., Oualkacha, K., Richard, L., Popradi, G., &
Keith, D. S., & Vranic, G. M. (2016). Approach to the highly sensi-
Sapir-Pichhadze, R. (2018). Inaccuracies in epitope repertoire estima-
tized kidney transplant candidate. Clinical Journal of the American
tions when using Multi-Locus Allele-Level hla genotype imputation
Society of Nephrology, 11(4), 684–693. https://doi.org/10.2215/
tools. HLA, 92(1), 33–39. https://doi.org/10.1111/tan.13307
CJN.05930615
Duquesnoy, R. J. (2001). HLAMMATCHMAKER: A molecularly
Klasberg, S., Surendranath, V., Lange, V., & Schofl, G. (2019).
based donor selection algorithm for highly alloimmunized pa-
Bioinformatics strategies, challenges, and opportunities for next
tients. Transplantation Proceedings, 33(1–2), 493–497. https://doi.
generation sequencing-based HLA genotyping. Transfusion Medicine
org/10.1016/s0041​-1345(00)02108​- 4
and Hemotherapy, 46(5), 312–325. https://doi.org/10.1159/00050​
Duquesnoy, R. J. (2006). A structurally based approach to determine
2487
HLA compatibility at the humoral immune level. Human Immunology,
Konvalinka, A., & Tinckam, K. (2015). Utility of HLA antibody test-
67(11), 847–862. https://doi.org/10.1016/j.humimm.2006.08.001
ing in kidney transplantation. Journal of the American Society
Duquesnoy, R. J., Howe, J., & Takemoto, S. (2003). HLAmatchmaker: A
of Nephrology, 26(7), 1489–1502. https://doi.org/10.1681/
molecularly based algorithm for histocompatibility determination.
ASN.20140​8 0837
IV. An alternative strategy to increase the number of compatible do-
Kosmoliaptsis, V., Chaudhry, A. N., Sharples, L. D., Halsall, D. J., Dafforn,
nors for highly sensitized patients. Transplantation, 75(6), 889–897.
T. R., Bradley, J. A., & Taylor, C. J. (2009). Predicting HLA class I allo-
https://doi.org/10.1097/01.TP.00000​55097.58209.83
antigen immunogenicity from the number and physiochemical prop-
Duquesnoy, R. J., Marrari, M., Marroquim, M. S., Borges, A. G., da
erties of amino acid polymorphisms. Transplantation, 88(6), 791–798.
Mata Sousa, L., Socorro, A., & do Monte, S. (2019). Second update
https://doi.org/10.1097/TP.0b013​e3181​b 4a9ff
of the international registry of HLA epitopes. I. The HLA-ABC
LEMIEUX et al. | 143

Kosmoliaptsis, V., Sharples, L. D., Chaudhry, A. N., Halsall, D. J., Bradley, HLA IgG antibodies after kidney transplantation. Human Immunology,
J. A., & Taylor, C. J. (2011). Predicting HLA class II alloantigen immu- 74(3), 290–296. https://doi.org/10.1016/j.humimm.2012.12.004
nogenicity from the number and physiochemical properties of amino Reed, E. F., Rao, P., Zhang, Z., Gebel, H., Bray, R. A., Guleria, I., Lunz,
acid polymorphisms. Transplantation, 91(2), 183–190. https://doi. J., Mohanakumar, T., Nickerson, P., Tambur, A. R., Zeevi, A., Heeger,
org/10.1097/TP.0b013​e3181​f fff99 P. S., & Gjertson, D. (2013). Comprehensive assessment and stan-
Kramer, C. S. M., Israeli, M., Mulder, A., Doxiadis, I. I. N., Haasnoot, G. W., dardization of solid phase multiplex-bead arrays for the detection of
Heidt, S., & Claas, F. H. J. (2019). The long and winding road towards antibodies to HLA. American Journal of Transplantation, 13(7), 1859–
epitope matching in clinical transplantation. Transplant International, 1870. https://doi.org/10.1111/ajt.12287
32(1), 16–24. https://doi.org/10.1111/tri.13362 Roll, G. R., Webber, A. B., Gae, D. H., Laszik, Z., Tavakol, M., Mayen, L.,
Kramer, C. S. M., Roelen, D. L., Heidt, S., & Claas, F. H. J. (2017). Defining Cunniffe, K., Syed, S., Hirose, R., Freise, C., Feng, S., Roberts, J. P.,
the immunogenicity and antigenicity of HLA epitopes is crucial for Ascher, N. L., Stock, P. G., & Rajalingam, R. (2020). A virtual cross-
optimal epitope matching in clinical renal transplantation. HLA, 90(1), match-based strategy facilitates sharing of deceased donor kidneys
5–16. https://doi.org/10.1111/tan.13038 for highly sensitized recipients. Transplantation, 104(6), 1239–1245.
Kransdorf, E. P., Pando, M. J., Gragert, L., & Kaplan, B. (2017). HLA https://doi.org/10.1097/TP.00000​0 0000​0 02924
population genetics in solid organ transplantation. Transplantation, Sapir-Pichhadze, R., Tinckam, K., Quach, K., Logan, A. G., Laupacis, A.,
101(9), 1971–1976. https://doi.org/10.1097/TP.00000​0 0000​ John, R., Beyene, J., & Kim, S. J. (2015). HLA-DR and -DQ eplet
001830 mismatches and transplant glomerulopathy: A nested case-control
Lee, C. H., & Yoon, H.-J. (2017). Medical big data: Promise and chal- study. American Journal of Transplantation, 15(1), 137–148. https://
lenges. Kidney Research and Clinical Practice, 36(1), 3–11. https://doi. doi.org/10.1111/ajt.12968
org/10.23876/​j.krcp.2017.36.1.3 Sapir-Pichhadze, R., Zhang, X., Ferradji, A., Madbouly, A., Tinckam, K. J.,
Lim, W. H., Wong, G., Heidt, S., & Claas, F. H. J. (2018). Novel aspects Gebel, H. M., Blum, D., Marrari, M., Kim, S. J., Fingerson, S., Bashyal,
of epitope matching and practical application in kidney transplanta- P., Cardinal, H., & Foster, B. J. (2020). Epitopes as characterized by an-
tion. Kidney International, 93(2), 314–324. https://doi.org/10.1016/j. tibody-verified eplet mismatches determine risk of kidney transplant
kint.2017.08.008 loss. Kidney International, 97(4), 778–785. https://doi.org/10.1016/j.
Liu, C. (2020). A long road/read to rapid high-resolution HLA typing: kint.2019.10.028
The nanopore perspective. Human Immunology, 1–8. https://doi. Schawalder, L., Honger, G., Kleiser, M., van Heck, M. R., van de Pasch,
org/10.1016/j.humimm.2020.04.00900 L. A. L., Vendelbosch, S., Rozemuller, E. H., & Schaub, S. (2020).
Matern, B. M., Olieslagers, T. I., Groeneweg, M., Duygu, B., Wieten, L., Development of an immunogenicity score for HLA-DQ eplets: A con-
Tilanus, M. G. J., & Voorter, C. E. M. (2020). Long-read nanopore se- ceptual study. HLA, 97, 30-43. https://doi.org/10.1111/tan.14110
quencing validated for human leukocyte antigen class I typing in rou- Senev, A., Coemans, M., Lerut, E., Van Sandt, V., Kerkhofs, J., Daniels,
tine diagnostics. The Journal of Molecular Diagnostics, 22(7), 912–919. L., Driessche, M. V., Compernolle, V., Sprangers, B., Van Loon, E.,
https://doi.org/10.1016/j.jmoldx.2020.04.001 Callemeyn, J., Claas, F., Tambur, A. R., Verbeke, G., Kuypers, D.,
McCaughan, J. A., Battle, R. K., Singh, S. K. S., Tikkanen, J. M., Moayedi, Emonds, M.-P., & Naesens, M. (2020). Eplet mismatch load and de
Y., Ross, H. J., Singer, L. G., Keshavjee, S., & Tinckam, K. J. (2018). novo occurrence of donor-specific anti-HLA antibodies, rejection,
Identification of risk epitope mismatches associated with de novo and graft failure after kidney transplantation: An observational co-
donor-specific HLA antibody development in cardiothoracic trans- hort study. Journal of the American Society of Nephrology, 31, 2193–
plantation. American Journal of Transplantation, 18(12), 2924–2933. 2204. https://doi.org/10.1681/ASN.20200​10019
https://doi.org/10.1111/ajt.14951 Senev, A., Emonds, M. P., Van Sandt, V., Lerut, E., Coemans, M.,
Meszaros, M., Niemann, M., Ursic-Bedoya, J., Faure, S., Meunier, L., Rivière, Sprangers, B., Kuypers, D., & Naesens, M. (2020). The clinical im-
B., Costes-Martineau, V., Thevenin, C., & Pageaux, G.-P. (2020). portance of extended 2nd field high-resolution HLA genotyping for
Exploring predicted indirectly recognizable HLA epitopes (PIRCHE-II) kidney transplantation. American Journal of Transplantation, 20, 3367-
in liver transplant recipients on calcineurin inhibitor-free maintenance 3378. https://doi.org/10.1111/ajt.15938
immunosuppression. A retrospective single center study. Transplant Sharma, A., Taverniti, A., Graf, N., Teixeira-Pinto, A., Lewis, J. R., Lim, W. H.,
Immunology, 59, 101272. https://doi.org/10.1016/j.trim.2020.101272 Alexander, S. I., Durkan, A., Craig, J. C., & Wong, G. (2020). The asso-
Morris, A. B., Sullivan, H. C., Krummey, S. M., Gebel, H. M., & Bray, R. ciation between human leukocyte antigen eplet mismatches, de novo
A. (2019). Out with the old, in with the new: Virtual versus physical donor-specific antibodies, and the risk of acute rejection in pediatric
crossmatching in the modern era. HLA, 94(6), 471–481. https://doi. kidney transplant recipients. Pediatric Nephrology(Berlin, Germany),
org/10.1111/tan.13693 35(6), 1061–1068. https://doi.org/10.1007/s0046​7-020-04474​-x
Mosbruger, T. L., Dinou, A., Duke, J. L., Ferriola, D., Mehler, H., Pagkrati, Smith, A. G., Pereira, S., Jaramillo, A., Stoll, S. T., Khan, F. M., Berka, N.,
I., Damianos, G., Mbunwe, E., Sarmady, M., Lyratzakis, I., Tishkoff, Mostafa, A. A., Pando, M. J., Usenko, C. Y., Bettinotti, M. P., Pyo,
S. A., Dinh, A., & Monos, D. S. (2020). Utilizing nanopore sequenc- C.-W., Nelson, W. C., Willis, A., Askar, M., & Geraghty, D. E. (2019).
ing technology for the rapid and comprehensive characterization Comparison of sequence-specific oligonucleotide probe vs next gen-
of eleven HLA loci; addressing the need for deceased donor expe- eration sequencing for HLA-A, B, C, DRB1, DRB3/B4/B5, DQA1,
dited HLA typing. Human Immunology, 81(8), 413–422. https://doi. DQB1, DPA1, and DPB1 typing: Toward single-pass high-resolution
org/10.1016/j.humimm.2020.06.004 HLA typing in support of solid organ and hematopoietic cell trans-
Nguyen, H. D., Wong, G., Howard, K., Claas, F. H. J., Craig, J. C., Fidler, S., plant programs. HLA, 94(3), 296–306. https://doi.org/10.1111/
D’Orsogna, L., Chapman, J. R., Irish, A., Ferrari, P., Christiansen, F. T., tan.13619
& Lim, W. H. (2014). Modeling the benefits and costs of integrating an Smits, J., van Houwelingen, H. C., De Meester, J., Persijn, G. G., & Claas, F.
acceptable HLA mismatch allocation model for highly sensitized pa- H. J. (1998). Analysis of the renal transplant waiting list: Application
tients. Transplantation, 97(7), 769–774. https://doi.org/10.1097/01. of a parametric competing risk method. Transplantation, 66(9), 1146–
TP.00004​38639.36838.ac 1153. https://doi.org/10.1097/00007​890-19981​1150-00006
Otten, H. G., Calis, J. J., Kesmir, C., van Zuilen, A. D., & Spierings, E. Tait, B. D. (2016). Detection of HLA antibodies in organ transplant recipi-
(2013). Predicted indirectly recognizable HLA epitopes presented by ents - triumphs and challenges of the solid phase bead assay. Frontiers
HLA-DR correlate with the de novo development of donor-specific in Immunology, 7, 570. https://doi.org/10.3389/fimmu.2016.00570
144 | LEMIEUX et al.

Tambur, A. R. (2018). HLA-epitope matching or eplet risk stratification: for primary alloimmunity. American Journal of Transplantation, 19(6),
The devil is in the details. Frontiers in Immunology, 9, 2010. https:// 1708–1719. https://doi.org/10.1111/ajt.15177
doi.org/10.3389/fimmu.2018.02010 Wiebe, C., Nevins, T. E., Robiner, W. N., Thomas, W., Matas, A. J., &
Tambur, A. R., Campbell, P., Chong, A. S., Feng, S., Ford, M. L., Gebel, H., Nickerson, P. W. (2015). The synergistic effect of class II HLA epi-
Gill, R. G., Kelsoe, G., Kosmoliaptsis, V., Mannon, R. B., Mengel, M., tope-mismatch and nonadherence on acute rejection and graft sur-
Reed, E. F., Valenzuela, N. M., Wiebe, C., Dijke, I. E., Sullivan, H. C., & vival. American Journal of Transplantation, 15(8), 2197–2202. https://
Nickerson, P. (2020). Sensitization in transplantation: Assessment of doi.org/10.1111/ajt.13341
risk (STAR) 2019 Working Group Meeting Report. American Journal Wiebe, C., & Nickerson, P. W. (2020). Human leukocyte antigen mo-
of Transplantation, 20(10), 2652–2668. https://doi.org/10.1111/ lecular mismatch to risk stratify kidney transplant recipients.
ajt.15937 Current Opinion in Organ Transplantation, 25(1), 8–14. https://doi.
Tambur, A. R., & Claas, F. H. (2015). HLA epitopes as viewed by antibod- org/10.1097/MOT.00000​0 0000​0 00714
ies: What is it all about? American Journal of Transplantation, 15(5), Wiebe, C., Pochinco, D., Blydt-Hansen, T. D., Ho, J., Birk, P. E., Karpinski,
1148–1154. https://doi.org/10.1111/ajt.13192 M., Goldberg, A., Storsley, L. J., Gibson, I. W., Rush, D. N. ,. &
Tambur, A. R., McDowell, H., Hod-Dvorai, R., Casillas Abundis, M. A., Nickerson, P. W. (2013). Class II HLA epitope matching-a strategy to
& Pinelli, D. F. (2019). The quest to decipher HLA immunogenicity: minimize de novo donor-specific antibody development and improve
Telling Friend from Foe. American Journal of Transplantation, 19(10), outcomes. American Journal of Transplantation, 13(12), 3114–3122.
2910–2925. https://doi.org/10.1111/ajt.15489 https://doi.org/10.1111/ajt.12478
Tambur, A. R., & Wiebe, C. (2018). HLA diagnostics: Evaluating DSA Wiebe, C., Rush, D. N., Nevins, T. E., Birk, P. E., Blydt-Hansen, T., Gibson,
strength by titration. Transplantation, 102(1S), S23–S30. https://doi. I. W., Goldberg, A., Ho, J., Karpinski, M., Pochinco, D., Sharma, A.,
org/10.1097/TP.00000​0 0000​0 01817 Storsley, L., Matas, A. J., & Nickerson, P. W. (2017). Class II eplet
Tassone, G., De Santis, D., Vukovic, I., Downing, J., Martinez, O. P., & mismatch modulates tacrolimus trough levels required to pre-
D'Orsogna, L. J. (2020). Different eplet software programs give dis- vent donor-specific antibody development. Journal of the American
cordant and incorrect results: An analysis of HLAMatchmaker vs Society of Nephrology, 28(11), 3353–3362. https://doi.org/10.1681/
Fusion Matchmaker Eplet calling software. HLA, 96(1), 52–63. ASN.20170​3 0287
Terasaki, P. I., Park, M. S., Takemoto, S., Cecka, J. M., Clark, B., Corcoran, Zheng, J., Kuang, P. D., Zhang, Y., Zhao, Q., He, X. L., Ding, X. M., & Xue, W.
S.,Cicciarelli, J., Barbetti, A., Yuge, J., & Carnahan, E. (1989). Overview J. (2019). Relationship of distribution frequency of HLA antigen/anti-
and epitope matching. Clinical Transplants, 3, 499–516. body and PIRCHE score with DSA production and AMR occurrence.
Tinckam, K. (2009). Histocompatibility methods. Transplantation Reviews, Zhonghua Yi Xue Za Zhi, 99(12), 901–906. https://doi.org/10.3760/cm
23(2), 80–93. https://doi.org/10.1016/j.trre.2009.01.001 a.j.issn.0376-2491.2019.12.005
Tinckam, K. J., Liwski, R., Pochinco, D., Mousseau, M., Grattan, A.,
Nickerson, P., & Campbell, P. (2015). cPRA increases with DQA, DPA,
and DPB unacceptable antigens in the Canadian cPRA calculator.
How to cite this article: Lemieux W, Mohammadhassanzadeh
American Journal of Transplantation, 15(12), 3194–3201. https://doi.
H, Klement W, Daniel C, Sapir-Pichhadze R. Matchmaker,
org/10.1111/ajt.13355
Walton, D. C., Hiho, S. J., Cantwell, L. S., Diviney, M. B., Wright, S. T., matchmaker make me a match: Opportunities and challenges
Snell, G. I., Paraskeva, M. A., & Westall, G. P. (2016). HLA matching in optimizing compatibility of HLA eplets in transplantation. Int
at the eplet level protects against chronic lung allograft dysfunction. J Immunogenet. 2021;48:135–144. https://doi.org/10.1111/
American Journal of Transplantation, 16(9), 2695–2703. https://doi.
iji.12525
org/10.1111/ajt.13798
Wiebe, C., Kosmoliaptsis, V., Pochinco, D., Gibson, I. W., Ho, J., Birk, P. E.,
Goldberg, A., Karpinski, M., Shaw, J., Rush, D. N., & Nickerson, P. W.
(2019). HLA-DR/DQ molecular mismatch: A prognostic biomarker

You might also like