Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Review

Interpreting Anti-HLA Antibody Testing Data:


A Practical Guide for Physicians
Carrie A. Schinstock, MD,1 Manish J. Gandhi, MD,2 and Mark D. Stegall, MD1

Abstract: The development of sensitive methods for alloantibody detection has been a significant advance in clinical trans-
plantation. However, the complexity of the data from solid phase and crossmatch assays has led to potential confusion about
how to use the results for clinical decision making. The goal of this review is to provide a practical guide for transplant phy-
sicians for the interpretation of antibody data to supplement consultation with local tissue typing experts. Sources of variability
in both the solid phase and crossmatch assay are discussed as are recent data regarding C1q binding antibodies and IgG sub-
class testing. Although definitive approaches to alloantibody testing are not possible with our current knowledge, we outline a
pragmatic approach that we hope will enhance clinical management in this area.

(Transplantation 2016;100: 1619–1628)

D espite improvements in patient selection and manage-


ment, every transplant carries some risk of graft loss.
Donor-specific alloantibody (DSA) either present at the time
the basic principles apply posttransplantation and to other
solid organ transplants—particularly heart, lung, and pancreas.

of transplantation or arising de novo posttransplant is a risk General Principles: The Initial Evaluation
factor for antibody mediated rejection (AMR) and potentially The initial assessment of a potential renal transplant candi-
allograft loss in almost all types of organ transplants.1-6 date involves HLA typing of the donor and recipient, screening
Ideally, all DSAwould be avoided, but this is often impractical for alloantibody against HLA, and obtaining a history of sen-
in the setting of organ scarcity and recipient sensitization. sitizing events (previous transplant, blood transfusion, and/or
Instead, the clinician must estimate the risk of AMR in each pregnancy). HLA typing ideally includes A; B; C; DRB1;
situation, while considering the consequences of remaining DRB3,4,5; DQB1; DQA; DPB1; and DPA; but this is not
on dialysis. Understanding the complexities and limitations always performed.7 The single antigen bead (SAB) solid-phase
of DSA detection techniques is key for making an accurate [LAB screen (One Lambda) or LifeScreen (LifeCodes-Immucor)]
risk assessment while improving access to transplantation. assay is most commonly used as the first line of screening for
The goal of this overview is to provide a practical guide for alloantibody, but multi-antigen screening beads can be used.
using solid phase assays and crossmatch (XM) testing. We also Fortunately, many renal transplant candidates—especially
provide possible explanations for ambiguous test results and rec- in the absence of a sensitizing event—have little or no alloanti-
ommendations for further investigation. This guide is not a sub- body detected on the solid phase assay. These patients usually
stitute for meaningful interaction with your local tissue typing require no further investigation. However, if the solid phase
laboratory experts because their guidance remains crucial for assay is positive suggesting the presence of alloantibody, fur-
proper decision making. Our major emphasis is on pretransplant ther evaluation is necessary. A basic understanding of the tech-
alloantibody assessment in kidney transplant candidates, but nical aspects of the solid phase and XM assays is essential for
optimal interpretation.
Received 17 November 2015. Revision received 5 January 2016.
The Solid Phase Single Antigen Bead (SAB) Assays
Accepted 20 January 2016.
1
William J. von Liebig Transplant Center, Mayo Clinic, Rochester, MN.
The current fluorescent-based solid phase assay is able to
2 concurrently distinguish up to 100 different microparticles
Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN.
or beads. Phenotypic or multi-antigen beads contain class I
This publication was made possible by CTSA Grant Number UL1 TR000135 from
the National Center for Advancing Translational Sciences (NCATS), a component
and class II antigens from a single cell line, whereas each
of the National Institutes of Health (NIH). Its contents are solely the responsibility of SAB contains a single HLA antigen.8 In both cases, the beads
the authors and do not necessarily represent the official view of NIH. contain a unique ratio of different fluorochromes that pro-
The authors declare no conflicts of interest. vide a distinctive signal that is recognized by Luminex technol-
C.A.S. designed and drafted manuscript. M.J.G. helped with the design of the work ogy.8 The patient’s serum is incubated with the solid phase
and critical revision. M.D.S. helped with the design of the work and critical revision. platform (beads) and a fluorescent-conjugated anti-human
All authors reviewed and approved the final version of the manuscript. IgG, which binds to alloantibody bound to its antigenic target.
Correspondence: Carrie A Schinstock, MD, Division of Nephrology and Hypertension, If the multi-antigen beads used for screening are positive, we
Mayo Clinic, 200 First Street SW, Rochester, MN 55905. (Schinstock.carrie@mayo.edu). suggest doing SABs to precisely identify the HLA antigen to
Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved. which the patient has alloantibody. For the purposes of this
ISSN: 0041-1337/16/10008-1619 overview, we will mostly discuss SAB testing. The SAB output
DOI: 10.1097/TP.0000000000001203 is a semiquantitative measure of the amount of antibody (total

Transplantation ■ August 2016 ■ Volume 100 ■ Number 8 www.transplantjournal.com 1619

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.


1620 Transplantation ■ August 2016 ■ Volume 100 ■ Number 8 www.transplantjournal.com

IgG—all subclasses combined) referred to as mean fluorescence antibodies bind both T and B cells, whereas antibodies
intensity (MFI). However, the assay is only FDA approved as against class II antigens usually bind only B cells. Activated
a qualitative assay (negative or positive). Determining a negative donor T cells express class II antigen in the presence of certain
SAB result is often the single most important aspect of alloanti- cytokines, which is a consideration in some cases.12
body testing because it is used for donor selection and calculated
panel reactive antibody (cPRA) assignment, yet this can be
difficult because results are not dichotomous and depend on Identifying a Transplant that is Low Risk for AMR
laboratory conditions. The MFI threshold to distinguish nega- Every transplant comes with some risk. When the SABs are
tive and positive also depends on a particular center’s risk tol- completely negative, especially if the patient has not had a
erance for AMR and clinical context. This MFI cutoff also sensitizing event, the risk for AMR is exceedingly low. How-
differs depending on the organ to be transplanted. An MFI ever, it is important to recognize that not uncommonly either
of less than 2000 is usually considered negative at our kidney the SAB or XM are falsely positive for anti-HLA alloantibody.
transplant center. We recognize that DSA may be present using In these situations, the risk of AMR is also low (Table 1).
this cutoff, but it also increases our patients’ access to trans-
plantation. Regardless, an MFI <1000 in a patient who has
not had a sensitizing event is considered negative at most kid- Alloantibody to Denatured HLA
ney transplant centers. The SAB assay does not measure antibody directly, but
rather measures the binding of antibody towards an antigen,
which is made up of multiple epitopes. This is essential to
Crossmatch Testing conceptualize when interpreting SAB results. When the HLA
The XM9-11 is considered the final test to determine the antigen is attached to the bead, a conformational change can
safety of moving forward with transplantation, but now sen- expose cryptic binding sites that are otherwise not accessible
sitive SAB assays have made the virtual crossmatch possible. on donor cells.13-17 Alloantibody binding to these denatured
When SAB testing is completely negative in a patient who has rather than intact HLA antigens leads to SAB positivity, yet
not had a sensitizing event, XM testing is unlikely to add to the flow XM will be negative. Recognizing this phenomenon
the AMR risk assessment. The XM is most valuable when- is valuable because antibodies toward denatured antigen alone
ever there is SAB positivity. Different XM techniques exist are not clinically relevant and by making the corresponding
and each has its own set of limitations and varied sensitivity. HLA antigen acceptable for organ offers increases a patient’s
The complement-dependent cytotoxicity crossmatch (CDC-XM) access to transplantation. One way of detecting these antibod-
was historically performed routinely. For this test, donor lym- ies is to treat the SABs with acid to denature all HLA antigens.
phocytes and recipient serum are mixed with complement. If the antibody only binds denatured antigen, the SAB result
The membrane attack complex forms when alloantibody is will remain positive despite acid treatment.16 Few studies have
cell bound leading to cell lysis. This testing is not sensitive for been done on this subject, but 21% to 39% of patients are
noncomplement binding DSA of low affinity/avidity and there- thought to have at least one antibody towards a denatured an-
fore the modified T cell anti-human globulin-enhanced cytotox- tigen.14,17 These antibodies have only been reported to be
icity XM (T cell AHG CDC) was developed. The addition of directed towards class I HLA and are not associated with
AHG to the cytotoxic assay improved its sensitivity, yet its previous sensitization.14 Transplant recipients with alloanti-
sensitivity is variable. It also does not detect DSA against body exclusively towards denatured antigen appear to have
class II (T cells do not express class II HLA). The CDC-XM similar long-term allograft survival as compared to transplant
with enriched B cells can be used to detect class II DSA, but recipients with no DSA.13,17
this technique can be technically difficult and AHG treatment
is not recommended.
The flow cytometric crossmatch (flow XM) has improved Laboratory Factors
sensitivity for the detection of both complement and non- Occasionally, the SAB test is mildly positive to multiple
complement binding class I and class II DSA as compared antigen specificities, yet the patient has not had a sensitizing
to the gold-standard CDC-XM.10 This method also reduces event. Likewise, the MFI may be 800 on one occasion and
the subjectivity and inter-observer variability associated with 1200 the next. These results do not necessary indicate that
the CDC-XM because it is reported as mean channel shift the patient has alloantibody or even that there has been an in-
rather than a positive or negative result based on visual inter- crease in alloantibody. Inter- and intra-laboratory variability
pretation. Therefore, to best estimate a patient’s risk for and background SAB fluorescence can sometimes lead to these
AMR, we suggest using the flow XM as adjunct to SAB, as op- results. The SAB assay is only semiquantitative, and the varia-
posed to other methods. This is our personal recommenda- tion in MFI has been reported as high as 62%, especially when
tion. The international Antibody Consensus Group supported the MFI is relatively low (1000-3000).18
by the Transplantation Society has not made recommenda- Laboratory variability exists because of differences in the
tions for or against the use of CDC versus flow XM.11 SAB product itself depending on the bead manufacturer,
The flow XM is performed by incubating donor lympho- batch, or lot.19-21 Manufacturers have different antigen sources,
cytes, recipient serum, and a fluorescent labeled anti-human distinctive specificity representation, and varied antigen density
immunoglobulin together and running them through a flow on the beads.18 Even with an identical product, intra-laboratory
cytometer.9 The level of fluorescence is proportional to the variability can occur depending on laboratory personnel, re-
amount of antibody bound to the cell. Markers with different agents, equipment, and conditions.19,20,22 As with any test using
fluorochromes are also added to separate T cell and B cell flow fluorescent technology, there is also background fluorescence.
reactivity. This is important because class I anti-HLA Laboratories validate their assays with clearly defined positive and

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.


© 2016 Wolters Kluwer Schinstock et al 1621

TABLE 1.
Risk stratification of antibody mediated rejection based on the combination of single antigen beads and flow
cytometric crossmatch

Risk for AMR Considerations SAB results Flow XM results Further evaluation
Low No DSA present Negative Negative T/B cell None
Alloantibody towards Positive Negative T/B cell Acid treatment of SAB. If still positive—the
denatured HLA alloantibody is towards denatured HLA
Laboratory factors Positive Negative T/B Cell Consult with HLA laboratory
Recent anti-CD20 treatment Negative Positive T and/or B cell Pronase treatment of donor lymphocytes
Auto-antibodies Negative Positive T and/or B cell Flow auto-crossmatch
High DSA present Positive Positive B cell Consider testing for further risk stratification*
Prozone effect Weakly positive Strongly positive B cell Repeat SAB with 1:8 serum dilution to identify
possible prozone
Incomplete donor/recipient Negative Positive B cell Be sure that the donor and recipient have been
HLA typing typed for DQ alpha and DP
SAB does not include donor Negative Positive B cell If SAB does not include donor antigen, use
HLA antigen supplemental SAB kit or another SAB vendor
Alloantibody to shared epitope Weakly positive Positive B cell Review SAB epitope binding patterns with
HLA expert
Uncertain Low HLA expression on donor Positive Negative B cell Consult with HLA laboratory
cells (ex. Cw)
DSA with low affinity/avidity Positive Negative B cell None
Non-HLA antibody Weakly positive or negative Positive T and/or B cell Consider checking for non-HLA antibody
The risk of AMR depends on several factors, but mostly depends on SAB and XM testing in clinical context. Ancillary testing, including acid treatment of SAB, auto-flow XM, and serum dilution, among others, can
provide additional information for risk stratification.
AMR, antibody mediated rejection; SAB, single antigen beads; DSA, donor-specific antibody; Flow XM, flow cytometric crossmatch; HLA, human leukocyte antigen.
*Further risk stratification includes DSA quantification, C1q binding ability, and IgG subclass identification if indicated.

negative controls to compensate for this, yet it is important to at obvious that the patient is at higher risk for AMR. But there
least consider. Whenever it is suspected that laboratory factors are also high-risk situations that are not as apparent when
are contributing to positive SAB results, it is important to con- looking at SAB tests alone (Table 1). Whenever a patient is
sult with your local tissue typing expert for advice. sensitized (especially highly sensitized, eg, cPRA >80%),
you need to have a heightened concern for AMR, and it is im-
Nonspecific Binding on Donor Lymphocytes: Recent portant to consider situations in which the virtual crossmatch
Anti-CD20 Treatment or “Auto-antibodies” is falsely negative to avoid an unanticipated positive final
A false-positive flow XM can occur with nonspecific flow XM. For this reason, we suggest consulting with the
binding of IgG to Fc receptors by factors in recipient serum. HLA laboratory whenever your patient is highly sensitized.
Anti-CD20 monoclonal antibody (rituximab) has most
commonly been implicated. Pretreatment of donor lym- Prozone Effect
phocytes with pronase can be helpful in removing the effect High levels of anti-HLA antibody with strong affinity and
of CD 20 binding.23 Treatment of donor cells with pronase avidity can saturate the solid phase platform or interfere
is only recommended when needed because it can interfere with alloantibody binding to the beads leading to SAB re-
with the donor HLA expression, which can lead to unreliable sults that are lower than expected for a particular antigenic
results.24 Other unidentified substances or “auto-antibodies” specificity. This is referred as the prozone or hook effect.
in recipient serum can also occasionally lead to nonspecific External substances (IgM antibody,26 intravenous immu-
binding to Fc receptors, and an auto-flow XM (recipient lym- noglobulin, antithymocyte globulin,27 immune complexes,
phocytes and serum) can provide clues to this phenomenon.25 complement,28,29 or nonspecific binding of external substances)
The long-term significance of an isolated auto-flow XM has can also interfere with antibody binding to the bead or the
not been thoroughly studied in the era of sensitive testing for secondary detection agent and also lead to this phenome-
anti-HLA antibody. Our unpublished single-center experience non.30,31 This effect does not necessarily affect all antigen
has been that nonsensitized patients with negative SAB tests specificities for a particular patient at one time and is mainly
and positive donor/recipient flow XM thought secondary to a consideration for highly sensitized patients (cPRA >80%).
auto-antibody (positive auto-flow XM) have short-term out- In one highly sensitized cohort studied, 71% had at least
comes comparable to recipients who have a pretransplant one anti-HLA antibody specificity demonstrating a prozone.31
negative B flow XM. However, this amounted to only 0.5% of the total SAB tested.
Serum treatment with EDTA/dithiothreitol, heat, or hypo-
tonic dialysis treatment29,30 can mitigate the prozone effect
Identifying a Transplant that is High Risk for AMR somewhat. These treatments are not part of the SAB manu-
When a patient has a history of a sensitizing event, SAB facturer’s standard operating procedure and are therefore
testing is positive for DSA, and the flow XM is positive; it is not routinely recommended. These treatments can also

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.


1622 Transplantation ■ August 2016 ■ Volume 100 ■ Number 8 www.transplantjournal.com

non-uniformly dilute the sample, making the MFI results less specifically donor typing from a previous transplant or paternal
reproducible. HLA typing in the setting of pregnancy. Furthermore, many
The most reliable way to recognize this effect is to do a serum sensitized patients have historically positive SAB results. Nota-
dilution. If the MFI does not decrease linearly as expected, the bly, the peak MFI of a particular DSA is most predictive of
prozone effect should be suspected. For example, a patient with AMR and graft loss, even if it is a historical measurement.35
a cPRA of 95% has an MFI of 800 to B44 that increases to The risk of AMR in the setting of historically positive DSA
3000 after a 1:8 serum dilution. We routinely do a 1:8 se- and negative crossmatch is not known. To minimize the risk
rum dilution in all patients with cPRA of greater than of AMR many transplant centers avoid HLA antigens to
80% who are eligible for a deceased donor transplant. If which the patient has been previously exposed or has had his-
prozone is detected, the corresponding antigen is considered torical alloantibody.
unacceptable for transplant. We recognize that further serum
dilution is needed to obtain the antibody titer, which is opti-
Low HLA Expression on Donor Cells
mal to determine the level of antibody. However, in most in-
stances, the detection of prozone alone (not antibody titer) is Regardless of the strength of alloantibody, without an an-
sufficient to consider the transplant high risk. tigenic target, the risk of AMR is generally low. Occasionally,
the HLA expression on donor cells is low or at least different
Incomplete Donor/Recipient HLA Typing than that represented on the beads. Low expression of HLA
Cw and DP is very common, but occasionally other HLA an-
An unexpected final positive crossmatch can occur if the tigens have low expression.20 Although the risk to undergo
donor and/or recipient are not completely typed for HLA. transplantation is generally low, it is unclear whether the HLA
This is especially a consideration for highly sensitized trans- expression could change during an inflammatory event, for
plant candidates. The United Network of Organ Sharing example. Low HLA expression should be considered when
(UNOS) does not currently require the reporting of DQA the SABs are positive, but flow XM and testing for alloanti-
or DP typing, yet 1.6% to 21.6% of transplant candidates body towards denatured antigen is negative. Although there
have alloantibody towards DQA and 6.6% to 33.7% have are no readily available tests to determine HLA expression
alloantibody towards DP antigens7 according to a survey of on the intended donor cells, occasionally a XM can be done
the UNOS Histocompatibility Committee members. The in- with a surrogate donor (a different donor with same HLA),
ternational Antibody Consensus Group supported by the which would be positive.
Transplantation Society recommends that donor typing for
HLA-A,B,C, DRB1, DRB3, DRB4, DRB5, DQA, DQB,
DPA, and DPB be performed for all potential recipients en- DSA With Low Affinity/Avidity
rolled in paired exchange registries.11 This group also recom- Even if donor HLA is expressed, sometimes DSA is of low
mends that for a renal transplant to be performed without a enough affinity and avidity that it does not produce a positive
prospective pretransplant XM, consistent absence of DSA flow XM. Although ideally all DSA would be avoided, it is
against the above stated antigens must be confirmed.11 not always possible. Although the presence of DSA in the set-
ting of positive CDC or T cell AHG is associated with AMR
SABs Do Not Include Donor HLA Antigen and allograft loss, the significance of DSA with low affinity/
Although the beads in the SAB assay include most poten- avidity with a corresponding negative flow XM is not as clear.
tial donor HLA antigens, it is not entirely comprehensive. If transplantation is performed in this setting, frequent early
Just as it is important to ensure that donor typing is accurate, posttransplant DSA monitoring may be helpful.
it is important that the donor antigen is represented on the
SABs. If the donor antigen is not represented in the particular Non-HLA Antibodies
kit used, then it may be worthwhile using a different vendor
Sensitized transplant recipients of kidney transplants from
for this particular patient. Supplemental kits are also avail-
HLA identical siblings have inferior 10-year allograft sur-
able if needed.
vival as compared to nonsensitized recipients,36 and specific
cases of AMR after donation from an HLA identical sibling
Shared Epitopes have been reported.37 Although these reports suggest a role
Alloantibody can bind a shared epitope on multiple anti- of non-HLA antibodies in AMR, the significance of these an-
gens resulting in the positivity of more than one bead.19,32-34 tibodies completely independent of alloantibody towards
The strength of this antibody would be diluted and under- HLA remains unclear.
represent its clinical importance if the donor cell only has Testing for non-HLA antibodies has historically been la-
one of these antigens. This is most relevant for alloantibody bor intensive and not validated. Most non-HLA antibodies
towards public epitopes present such as those on HLA Bw4 are expressed on endothelial cells and not lymphocytes.
or Bw6.11,20 The HLA laboratory can identify whether it ap- Some of the antigenic targets on the endothelial cell that have
pears that an antibody is present towards a shared epitope. been studied include anti-vimentin, anti-major histocompati-
The flow XM would also be positive in this scenario. bility I related chain A, anti-major histocompatibility I re-
lated chain B, perlecan, Kα-tubulin, protein kinase C, and
anti-angiotensin A1 receptor, among others.38-47 An endo-
Uncertain Risk for AMR
thelial XM has also been developed recently (XM-ONE;
Historical Alloantibody AbSorber AB, Stockholm, Sweden).48,49 Antigen targets other
Careful attention to the patient’s sensitization history than those present on the endothelial cell are less well studied
can provide clues about previous HLA antigen exposure, and include anti-myosin, anti-nuclear, anti-smooth muscle,

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.


© 2016 Wolters Kluwer Schinstock et al 1623

anti-nucleoprotein, anti-DNA, anti-cytoplasmic, and anti- detection, it has been used clinically as a semiquantitative
mitochondrial among others.38,50 assay to estimate the amount (strength and avidity) of anti-
The study of non-HLA antibody was previously limited HLA antibody52,53 and attempt to assess posttransplant risk.
by the lack of concurrent sensitive testing for anti-HLA anti- Numerous groups have shown that a higher DSA MFI and/or
bodies and small numbers of patients. The true prevalence of XM has been associated with increased AMR and allograft
non-HLA antibodies, their relationship to allograft pathol- loss,35,52,53but the correlation between MFI and these out-
ogy in the absence of anti-HLA antibody, and their mecha- comes is far from precise. Another way of determining the
nism of action remains unclear; thus, routine testing for strength and avidity of alloantibody is by obtaining the an-
them cannot be recommended currently. That being said, if tibody titer. This is especially important in patients with pro-
a sensitized patient has AMR without detectable anti-HLA zone as described above, but may be helpful in any patient
DSA, testing for non-HLA antibodies using the few commer- with alloantibody. The antibody titer is determined by doing
cially available assays is a consideration. serial dilutions. The dilution at which antibody is no longer de-
tected is considered its titer.31 Figure 1 shows a comparison
Not All DSA are the Same: Risk Stratification in the
between neat MFI, serum dilutions, C1q MFI, and the MFI
Setting of Known DSA
of EDTA treated samples in a cohort of sensitized patients.
Although it is advantageous to completely avoid DSA, it Obtaining the antibody titer is clearly a more reliable means
is not always possible given a patient’s sensitization. Patient of quantifying antibody strength and avidity as compared to
survival has been shown to be improved with positive XM MFI alone in highly sensitized transplant candidates because
transplantation and desensitization as compared to waiting of the limitations of the SAB assay,31 yet it remains unclear
on dialysis for a negative XM transplant.51 For this reason, whether a higher antibody titer is more strongly correlated
some programs accept the potential risk for AMR and proceed with AMR and allograft loss than MFI alone.
to transplantation in the setting of known DSA. Not all DSA The disadvantage of determining antibody titer is the in-
is the same and its pathogenicity remains difficult to predict. creased cost and labor involved with doing multiple tests,
Understanding DSA characteristics beyond positive and neg- but this is not always needed. Our personal recommendation
ative can help further stratify the risk of AMR. is to perform only one serum dilution in highly sensitized
patients at baseline and as needed to detect prozone. The in-
Alloantibody Quantification formation can be used when assigning unacceptable antigens
Although the current SAB assay is FDA approved as a and for future reference because the mere presence of pro-
qualitative assay for alloantibody (positive versus negative) zone indicates high risk for transplantation. Serial serum

FIGURE 1. Comparison of the strength of DSA using serial dilutions, C1q binding, and EDTA treatment.

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.


1624 Transplantation ■ August 2016 ■ Volume 100 ■ Number 8 www.transplantjournal.com

dilutions and antibody titer determination can often be re- available modified SAB tests have been developed to specifi-
served to assess the response to antibody depletion therapy cally detect C1q and C3d binding. C1q or C3d binding pos-
in clinical practice or research. itivity represents the presence of DSA that is a complement
fixing subtype of IgG at a high enough level to bind comple-
Single Versus Multiple DSA ment.59,60 DSA with C1q and/or C3d binding positivity is
It remains unclear whether there is a difference in long-term associated with the development of AMR and allograft
outcomes depending on the number of DSA the patient has at loss,57,60,61 and some groups routinely use these tests to dis-
the time of transplant, independent of the DSA MFI or cross- tinguish the most deleterious DSA However, the presence of
match results. Several groups have reported outcomes based C1q binding antibodies is also associated with DSA MFI
on the immunodominant (highest MFI) at the time of trans- and antibody titer.62,63 It remains unknown whether C1q
plantation,54,55 but this approach is complicated because a positivity is any more predictive of AMR or graft loss than
DSA that appears immunodominant pretransplant may not MFI or antibody titer31,62,64 and thus testing for C1q or
be immunodominant posttransplant. Other groups have C3d binding is not yet universally performed.
correlated the MFI sum with long-term outcomes.52,56 That
approach is also problematic because the SAB is only semi- Immunoglobulin Subclasses
quantitative. Clearly, further study is needed in this area. SABs provide a semiquantitative measure of total immu-
For now, we suggest using the B flow XM to guide decision noglobulin G (IgG) rather than IgG subclasses. This is relevant
making as it provides a summation of the effect of the DSA because each subclass has a differing role in the immune re-
on the donor cell. sponse and varied ability to bind the Fc receptor or activate
complement.65 Specific subclass profiles may have a stronger
Complement Binding DSA association with the development of chronic AMR or allograft
Recent studies have suggested that assessing antibodies for loss than total IgG. IgG subclass characterization is now per-
the ability to bind C1q can help differentiate pathogenic from formed in the research setting using a modified SAB assay in
nonpathogenic DSA.57 Complement activation is an impor- which the conventional phycoerythrin-conjugated anti-human
tant mechanism by which DSA leads to allograft damage, IgG is replaced with an IgG subclass-specific anti-human IgG.66
and this activation begins by antibody binding of C1q.58 Of the 4 subclasses, IgG1/IgG3 has the strongest ability
Downstream, C3 is cleaved leaving C3d. Commercially to bind complement. Emerging evidence suggests that DSA

FIGURE 2. Algorithm for DSA testing for candidate on deceased donor waiting list. † Refer to Table 1.

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.


© 2016 Wolters Kluwer Schinstock et al 1625

FIGURE 3. Algorithm for DSA testing for candidates with potential living donor. † Refer to Figure 4. ‡ Refer to Figure 2.

of the IgG 3 subclass is particularly deleterious to the renal and apply to renal transplant recipients only. These algo-
and liver allografts.54,66-72 However, studies of IgG subclasses rithms may differ depending on the organ transplanted.
in transplantation are limited because few groups have ex- For transplant candidates awaiting a deceased donor, the
amined complete IgG subclass characterization [IgG1- goal is to optimize access to transplantation while avoiding
IgG4] serially. The complexity of studying IgG subclasses an unexpected final XM. Time constraints limit the ability
is overwhelming but provides clues about the pathogenicity to do further testing after an unexpected positive final XM,
of certain DSA and the immune response, and thus it will likely and thus we recommend as much testing as possible before
become an important aspect in donor selection in highly sensi- or at the time of the final XM.
tized patients. More study of the significance of DSA subclass If the patient has completely negative solid phase testing
characterization and testing methods are needed before rou- (single antigen bead or multi-antigen beads), their risk for
tine clinical use. AMR is low. Crossmatch testing does not add considerably
to risk stratification, and in that case, one could argue that
it is not needed. However, for patients with SAB positivity
Practical Approach to Alloantibody Testing or history of sensitizing event, unacceptable antigens (or ac-
In an ideal world, comprehensive DSA testing would be ceptable mismatch) must be assigned and a final XM is valu-
performed for each patient including antibody titer, non- able. Because highly sensitized transplant candidates (cPRA
HLA antibody, auto-flow XM etc., but this is not practical >80%) have the least access to transplantation and are the
given time constraints and is not cost-effective. We suggest most likely to have an unexpected positive XM, we recom-
the evaluation be tailored based on type of donor (deceased mend testing for prozone and alloantibody towards denatured
versus living) and the patient’s sensitization (Figures 2 and 3). HLA before unacceptable antigen assignment. The assignment
Additionally, we always recommend consultation with local of unacceptable antigens is variable among centers and is de-
tissue typing experts and consideration of clinical context. pendent on center preferences and underlying patient character-
These algorithms are based on our personal recommendations istics (age, sensitization, historical alloantibody, and waiting

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.


1626 Transplantation ■ August 2016 ■ Volume 100 ■ Number 8 www.transplantjournal.com

FIGURE 4. Evaluation when the single antigen beads and flow crossmatch are inconsistent.

time). Inter- and intra-laboratory variability and background alloantibody towards denatured antigen. The risk of trans-
positivity of the single antigen bead assay should also be plantation in the setting of a positive XM and negative
taken into consideration. For the most part, if alloantibody DSA is less certain. Although the XM is often falsely positive
is present with MFI of >1000 or prozone is present, the cor- from nonspecific binding of donor lymphocytes (auto-flow
responding antigen is considered unacceptable. Conversely, if XM positivity), prozone effect, incomplete donor typing,
the SAB is only positive because alloantibody is toward dena- SAB noninclusive of donor HLA, alloantibody towards a
tured HLA, it can be considered acceptable. shared epitope, and non-HLA alloantibody should be consid-
When an organ is offered, we also recommend obtaining ered especially if the patient has a cPRA of greater than 80%.
an auto-flow XM at the time of final crossmatch and treating Although we provide a framework for DSA testing, it does
donor lymphocytes with pronase if the potential recipient has not substitute clinical judgment. Consultation with the HLA
had recent exposure to anti-CD20. Usually all XM testing is laboratory is crucial to accurately estimate your patient’s risk
negative, and transplantation can proceed. However, occasionally, for AMR. Consideration of the underlying clinical context is
the final crossmatch is positive in which case the auto-flow also critical. We optimally avoid DSA in the setting of a
XM is helpful. If the auto-flow XM is positive, and you have young nonsensitized patient who is not yet on dialysis, yet
confirmed that the patient has not had any recent sensitiza- consider positive XM transplantation with known DSA in
tion and the donor typing is complete, you can proceed to a highly sensitized candidate with prolonged waiting time.
transplant. If the final XM is unexpectedly positive, but the Although beyond the scope of this article, it is also always im-
auto-flow XM is negative, the risk of transplantation is un- portant to have a protocol to manage AMR because AMR
certain. The potential considerations include recent sensitiza- can unexpectedly occur even in presumably low-risk situations.
tion (previous SAB testing is not current), incomplete donor/
recipient HLA typing, SAB noninclusive of donor HLA, allo-
antibody toward a public epitope, or alloantibody towards CONCLUSION
non-HLA (Figure 2). Our understanding of DSA has advanced overtime, and
When a potential living donor has been identified, the sensitive techniques for DSA detection are now available, but
timing of DSA testing is somewhat modified as shown in the interpretation remains difficult. Optimal patient manage-
Figure 3. Again, if the solid phase assay is completely nega- ment involves experts from the HLA laboratory and careful
tive, the risk for AMR is low, and the XM does not add con- interpretation of alloantibody testing while considering the
siderably to the evaluation. However, in other cases, we clinical context. Recent modifications of the SAB assay to
recommend a flow XM at the time of donor identification. determine particular DSA characteristics including com-
Further testing is only done if the flow XM result is not con- plement binding and IgG subclass will complicate alloanti-
sistent with the SAB—whether it is a positive SAB suggestive body interpretation, but likely provide clarity about the
of DSA and negative XM or negative SAB and positive XM prognostic significance of particular DSA. An efficient
(Figure 4). The risk of AMR in the setting of positive and cost-effective approach to alloantibody testing can
SAB and a negative XM is generally low, but we recommend minimize the risk of AMR for patients, yet optimize access
consulting with the HLA laboratory and sometimes testing for to transplantation.

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.


© 2016 Wolters Kluwer Schinstock et al 1627

ACKNOWLEDGMENTS 22. Gandhi MJ, Degoey S, Falbo D, et al. Inter and intra laboratory concor-
The authors thank Adriana Zeevi PhD, ABHI for her critical dance of HLA antibody results obtained by single antigen bead based
assay. Hum Immunol. 2013;74:310.
review of the manuscript and suggestions for improvement. 23. Vaidya S, Cooper TY, Avandsalehi J, et al. Improved flow cyto-
metric detection of HLA alloantibodies using pronase: potential-
implications in renal transplantation. Transplantation. 2001;71:
REFERENCES 422–428.
1. Gloor JM, Sethi S, Stegall MD, et al. Transplant glomerulopathy: sub- 24. Hetrick SJ, Schillinger KP, Zachary AA, et al. Impact of pronase on flow cy-
clinical incidence and association with alloantibody. Am J Transplant. tometric crossmatch outcome. Hum Immunol. 2011;72:330.
2007;7:2124. 25. Le Bas-Bernardet S, Hourmant M, Valentin N, et al. Identification of
2. Issa N, Cosio FG, Gloor JM, et al. Transplant glomerulopathy: risk and the antibodies involved in B-cell crossmatch positivity in renal trans-
prognosis related to anti-human leukocyte antigen class II antibody levels. plantation. Transplantation. 2003;75:477.
Transplantation. 2008;86:681. 26. Kosmoliaptsis V, Bradley JA, Peacock S, et al. Detection of immunoglob-
3. Hidalgo LG, Campbell PM, Sis B, et al. De novo donor-specific anti- ulin G human leukocyte antigen-specific alloantibodies in renal transplant
body at the time of kidney transplant biopsy associates with microvas- patients using single-antigen-beads is compromised by the presence
cular pathology and late graft failure. Am J Transplant. 2009;9:2532. of immunoglobulin M human leukocyte antigen-specific alloantibodies.
4. Hodges AM, Lyster H, McDermott A, et al. Late antibody-mediated re- Transplantation. 2009;87:813.
jection after heart transplantation following the development of de 27. Gloor JM, Moore SB, Schneider BA, et al. The effect of antithymocyte
novo donor-specific human leukocyte antigen antibody. Transplanta- globulin on anti-human leukocyte antigen antibody detection assays.
tion. 2012;93:650. Transplantation. 2007;84:258–264.
5. Taner T, Stegall MD, Heimbach JK. Antibody-mediated rejection in liver 28. Visentin J, Vigata M, Daburon S, et al. Deciphering complement interfer-
transplantation: current controversies and future directions. Liver Transpl. ence in anti-human leukocyte antigen antibody detection with flow beads
2014;20:514–527. assays. Transplantation. 2014;98:625.
6. Niederhaus SV, Leverson GE, Lorentzen DF, et al. Acute cellular and 29. Schnaidt M, Weinstock C, Jurisic M, et al. HLA antibody specification
antibody-mediated rejection of the pancreas allograft: incidence, risk factors using single-antigen beads—a technical solution for the prozone effect.
and outcomes. Am J Transplant. 2013;13:2945. Transplantation. 2011;92:510–515.
7. OPTN/UNOS. Greater consistency in candidate and deceased donor 30. Zachary AA, Lucas DP, Detrick B, et al. Naturally occurring interference
HLA typing requirements across organ types. U.S. Department of Health in Luminex assays for HLA-specific antibodies: characteristics and reso-
& Human Services. https://optn.transplant.hrsa.gov/PublicComment/ lution. Hum Immunol. 2009;70:496.
pubcommentPropSub_335.pdf. Published 2014. Accessed January 22, 2016. 31. Tambur AR, Herrera ND, Haarberg KM, et al. Assessing antibody strength:
8. Lachmann N, Todorova K, Schulze H, et al. Luminex® and its applications comparison of MFI, C1q, and titer information. Am J Transplant. 2015;15:
for solid organ transplantation, hematopoietic stem cell transplantation, 2421–2430.
and transfusion. Transfus Med Hemother. 2013;40:182–189. 32. Ellis TM. Interpretation of HLA single antigen bead assays. Transplant Rev
9. Bray R. Transplantation immunology: methods and protocols. In: Zachary (Orlando). 2013;27:108–111.
AL M, editor. Lymphocyte Crossmatching by Flow Cytometry. vol. 103. 33. Tait BD, Hudson F, Brewin G, et al. Solid phase HLA antibody de-
New York City, New York: Springer; 2013. Pages 285–296. tection technology—challenges in interpretation. Tissue Antigens.
10. Taylor CJ, Kosmoliaptsis V, Summers DM, et al. Back to the future: appli- 2010;76:87.
cation of contemporary technology to long-standing questions about 34. Duquesnoy RJ, Marrari M. Correlations between Terasaki’s HLA class I
the clinical relevance of human leukocyte antigen-specific alloanti- epitopes and HLAMatchmaker-defined eplets on HLA-A, -B and -C antigens.
bodies in renal transplantation. Hum Immunol. 2009;70:563. Tissue Antigens. 2009;74:117.
11. Tait BD, Süsal C, Gebel HM, et al. Consensus guidelines on the testing 35. Lefaucheur C, Loupy A, Hill GS, et al. Preexisting donor-specific HLA
and clinical management issues associated with HLA and non-HLA antibodies predict outcome in kidney transplantation. J Am Soc Nephrol.
antibodies in transplantation. Transplantation. 2013;95:19. 2010;21:1398.
12. Heo WB, Kim CD, Won DI. HLA Class II-specific antibodies can react 36. Opelz G. Non-HLA transplantation immunity revealed by lymphocytotoxic
with T cells in flow cytometry crossmatch: a case report. Transplant antibodies. Lancet. 2005;365:1570.
Proc. 2007;39:3485. 37. Grafft CA, Cornell LD, Gloor JM, et al. Antibody-mediated rejection
13. Otten HG, Verhaar MC, Borst HP, et al. The significance of pretrans- following transplantation from an HLA-identical sibling. Nephrol Dial
plant donor-specific antibodies reactive with intact or denatured human Transplant. 2010;25:307–310.
leucocyte antigen in kidney transplantation. Clin Exp Immunol. 2013; 38. Sigdel TK, Sarwal MM. Moving beyond HLA: a review of nHLA antibodies
173:536–543. in organ transplantation. Hum Immunol. 2013;74:1486.
14. Visentin J, Guidicelli G, Bachelet T, et al. Denatured class I human 39. Dinavahi R, George A, Tretin A, et al. Antibodies reactive to non-
leukocyte antigen antibodies in sensitized kidney recipients: preva- HLA antigens in transplant glomerulopathy. J Am Soc Nephrol. 2011;
lence, relevance, and impact on organ allocation. Transplantation. 2014; 22:1168.
98:738. 40. Cerilli J, Clarke J, Doolin T, et al. The significance of a donor-specific vessel
15. Pereira S, Perkins S, Lee JH, et al. Donor-specific antibody against crossmatch in renal transplantation. Transplantation. 1988;46:359.
denatured HLA-A1: clinically nonsignificant? Hum Immunol. 2011; 41. Paul LC, Claas FH, van Es LA, et al. Accelerated rejection of a renal
72:492. allograft associated with pretransplantation antibodies directed
16. Jacob EK, De Goey SR, Gandhi MJ. Positive virtual crossmatch with against donor antigens on endothelium and monocytes. N Engl J Med.
negative flow crossmatch results in two cases. Transpl Immunol. 1979;300:1258.
2011;25:77–81. 42. Fredrich R, Toyoda M, Czer LS, et al. The clinical significance of anti-
17. Oaks M, Michel K, Sulemanjee NZ, et al. Practical value of identifying bodies to human vascular endothelial cells after cardiac transplanta-
antibodies to cryptic HLA epitopes in cardiac transplantation. J Heart tion. Transplantation. 1999;67:385–391.
Lung Transplant. 2014;33:713. 43. Glotz D, Lucchiari N, Pegaz-Fiornet B, et al. Endothelial cells as targets of
18. Reed EF, Rao P, Zhang Z, et al. Comprehensive assessment and allograft rejection. Transplantation. 2006;82(1 Suppl):S19.
standardization of solid phase multiplex-bead arrays for the detection 44. Mizutani K, Terasaki PI, Shih RN, et al. Frequency of MIC antibody in
of antibodies to HLA-drilling down on key sources of variation. Am J rejected renal transplant patients without HLA antibody. Hum Immunol.
Transplant. 2013;13:3050. 2006;67:223.
19. Konvalinka A, Tinckam K. Utility of HLA antibody testing in kidney trans- 45. Mizutani K, Terasaki P, Rosen A, et al. Serial ten-year follow-up of HLA and
plantation. J Am Soc Nephrol. 2015;26:1489. MICA antibody production prior to kidney graft failure. Am J Transplant.
20. Gebel HM, Bray RA. HLA antibody detection with solid phase assays: 2005;5:2265.
great expectations or expectations too great? Am J Transplant. 2014; 46. Terasaki PI, Ozawa M, Castro R. Four-year follow-up of a prospective trial
14:1964–1975. of HLA and MICA antibodies on kidney graft survival. Am J Transplant.
21. Gandhi MJ, Carrick DM, Jenkins S, et al. Lot-to-lot variability in HLA anti- 2007;7:408.
body screening using a multiplexed bead-based assay. Transfusion. 47. Zou Y, Stastny P, Susal C, et al. Antibodies against MICA antigens and
2013;53:1940. kidney-transplant rejection. N Engl J Med. 2007;357:1293.

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.


1628 Transplantation ■ August 2016 ■ Volume 100 ■ Number 8 www.transplantjournal.com

48. Jackson AM, Sigdel TK, Delville M, et al. Endothelial cell antibodies asso- 61. Baid-Agrawal S, Lachmann N, Budde K. Complement-binding anti-HLA
ciated with novel targets and increased rejection. J Am Soc Nephrol. antibodies and kidney transplantation. N Engl J Med. 2014;370:84.
2015;26:1161. 62. Zeevi A, Lunz J, Feingold B, et al. Persistent strong anti-HLA antibody at
49. Jackson AM, Lucas DP, Melancon JK, et al. Clinical relevance and IgG high titer is complement binding and associated with increased risk of
subclass determination of non-HLA antibodies identified using endothelial antibody-mediated rejection in heart transplant recipients. J Heart Lung
cell precursors isolated from donor blood. Transplantation. 2011;92:54–60. Transplant. 2013;32:98–105.
50. Valenzuela NM, Reed EF. Antibodies in transplantation: the effects 63. Schaub S, Hönger G, Koller MT, et al. Determinants of C1q binding in the
of HLA and non-HLA antibody binding and mechanisms of injury. single antigen bead assay. Transplantation. 2014;98:387–393.
Methods Mol Biol. 2013;1034:41–70. 64. Gandhi M, Degoey S, Henderson N, et al. C1q single antigen bead assay
51. Montgomery RA, Lonze BE, King KE, et al. Desensitization in HLA- only detects high titer/avidity class-I anti HLA antibodies detected by sin-
incompatible kidney recipients and survival. N Engl J Med. 2011;365:318. gle antigen beads. Hum Immunol. 2014;75:73.
52. Gloor JM, Winters JL, Cornell LD, et al. Baseline donor-specific antibody 65. Nimmerjahn F, Ravetch JV. Divergent immunoglobulin g subclass activity
levels and outcomes in positive crossmatch kidney transplantation. Am through selective Fc receptor binding. Science. 2005;310:1510.
J Transplant. 2010;10:582. 66. Kaneku H, O'Leary JG, Taniguchi M, et al. Donor-specific human leuko-
53. Burns JM, Cornell LD, Perry DK, et al. Alloantibody levels and acute hu- cyte antigen antibodies of the immunoglobulin G3 subclass are associ-
moral rejection early after positive crossmatch kidney transplantation. ated with chronic rejection and graft loss after liver transplantation. Liver
Am J Transplant. 2008;8:2684. Transpl. 2012;18:984.
54. Lefaucheur C, Viglietti D, Bentlejewski C, et al. IgG donor-specific anti- 67. Taner T, Gandhi MJ, Sanderson SO, et al. Prevalence, course and impact
human HLA antibody subclasses and kidney allograft antibody- of HLA donor-specific antibodies in liver transplantation in the first year. Am
mediated injury. J Am Soc Nephrol. 2015. J Transplant. 2012;12:1504.
55. Woodle ES, Shields AR, Ejaz NS, et al. Prospective iterative trial of 68. O'Leary JG, Klintmalm GB. Impact of donor-specific antibodies
proteasome inhibitor-based desensitization. Am J Transplant. 2015;15: on results of liver transplantation. Curr Opin Organ Transplant.
101–118. 2013;18(3):279.
56. Kannabhiran D, Lee J, Schwartz JE, et al. Characteristics of circulating 69. O'Leary JG, Demetris AJ, Friedman LS, et al. The role of donor-
donor human leukocyte antigen-specific immunoglobulin G antibodies specific HLA alloantibodies in liver transplantation. Am J Transplant.
predictive of acute antibody-mediated rejection and kidney allograft fail- 2014;14:779.
ure. Transplantation. 2015;99:1156. 70. Everly MJ, Rebellato LM, Haisch CE, et al. Impact of IgM and IgG3 anti-
57. Loupy A, Lefaucheur C, Vernerey D, et al. Complement-binding anti- HLA alloantibodies in primary renal allograft recipients. Transplantation.
HLA antibodies and kidney-allograft survival. N Engl J Med. 2013; 2014;97:494.
369:1215. 71. Schinstock C, Cosio F, Cornell L, et al. Donor-Specific Alloantibody After Positive
58. Stegall MD, Chedid MF, Cornell LD. The role of complement in antibody- Crossmatch Kidney Transplantation With Eculizumab (EC): Correlation With
mediated rejection in kidney transplantation. Nat Rev Nephrol. 2012;8:670–678. Transplant Glomerulopathy and Graft Failure [abstract]. Am J Transplant.
59. Chen G, Sequeira F, Tyan DB. Novel C1q assay reveals a clinically 2015;15(suppl 3). Available at: http://www.atcmeetingabstracts.com/
relevant subset of human leukocyte antigen antibodies independent abstract/donor-specific-alloantibody-after-positive-crossmatch-kidney-
of immunoglobulin G strength on single antigen beads. Hum Immunol. transplantation-with-eculizumab-ec-correlation-with-transplant-glomerulopathy-
2011;72:849. and-graft-failure/. Accessed March 31, 2016.
60. Sicard A, Ducreux S, Rabeyrin M, et al. Detection of C3d-binding donor- 72. Freitas MC, Rebellato LM, Ozawa M, et al. The role of immunoglobulin-G
specific anti-HLA antibodies at diagnosis of humoral rejection predicts subclasses and C1q in de novo HLA-DQ donor-specific antibody kidney
renal graft loss. J Am Soc Nephrol. 2015;26:457–467. transplantation outcomes. Transplantation. 2013;95:1113–1119.

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.

You might also like