Chapter 2
The HLA System in Hematopoietic Stem Cell Transplantation Effie Petersdorf1 and Ge´rard Socie´2 1
Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, United States; 2Department of Hematology, Paris, France
Chapter Outline Introduction Classical HLA Genes Organization Classical Class I Genes Nonclassical MHC Genes: HLA-E, HLA-F, HLA-G, and MIC HLA-E Human Leukocyte Antigen G MIC Genes Class II Genes Class III Genes Nomenclature LD and Haplotypes The Role of Classical HLA in Unrelated Donor Hematopoietic Cell Transplantation
15 16 16 16 16 16 17 18 18 19 19 19 19
Alleles and Antigens Additive Effects of HLA Disparity Beneficial Effects of HLA Mismatching: Graft-VersusLeukemia New Approaches for Defining Permissive HLA Mismatches Toward a Haplotypic View of Allogenecity The Clinical Significance of Nonclassical HLA Genes: HLA-E, HLA-F, HLA-G, and MIC Genes HLA-E in HCT: GVH HLA-G in HCT-GVH MIC Genes in HCT-GVH Conclusions Acknowledgments References
20 20 21 21 22 23 23 24 25 25 26 26
The human major histocompatibility complex (MHC) is residence to the human leukocyte antigen (HLA) genes that play a fundamental role in the acceptance of transplanted tissues. The ubiquitous expression of HLA antigens on nucleated cells is a key feature of their function in both health and disease. In this chapter, the content, organization, and diversity of genes within the MHC are discussed, and the clinical implications of genetic polymorphism on risks of graft-versus-host disease (GVHD) in hematopoietic stem cell transplantation (HSCT) are summarized.
INTRODUCTION Since the discovery of the HLA system [1,2], there has been unprecedented discovery of the gene number, structure and sequences, polymorphism, haplotype composition, and linkage disequilibrium (LD) within the major histocompatibility complex (MHC) [3]. More than 300 genes reside within the MHC, and of these, approximately 15%e20% have immunerelated function including antigen processing and presentation, immune regulation, inflammation, complement, maternalfetal immunology, stress response, leukocyte maturation, and the immunoglobulin superfamily [3]. New information on the regulatory polymorphisms is emerging from efforts of the Human Epigenome Project and provides insight into methylation and histone acetylation profiles within the MHC [4,5].
Immune Biology of Allogeneic Hematopoietic Stem Cell Transplantation. https://doi.org/10.1016/B978-0-12-812630-1.00002-5 Copyright © 2019 Elsevier Inc. All rights reserved.
15
16
Immune Biology of Allogeneic Hematopoietic Stem Cell Transplantation
CLASSICAL HLA GENES Organization The MHC is composed of three regions termed class I, class III, and class II (Fig. 2.1). The HLA system includes the classical loci HLA-A, HLA-C, and HLA-B and the nonclassical genes HLA-E, HLA-F, HLA-G, MICA, and MICB that reside within the class I region. HLA-DR, HLA-DQ, and HLA-DP reside within the class II region. The class III region comprises genes of importance to the stress response (TNF, HSP, LTA) and the complement cascade.
Classical Class I Genes Class I HLA-A, -B, and -C genes are each composed of a series of eight exons delineated by intervening introns. Each class I exon has a unique function: exon 1 encodes the leader sequence; exons 2, 3, and 4 encode the a1, 2, and 3 domains, respectively; exon 5 encodes the transmembrane portion, and exons 6, 7, and 8 encode the cytoplasmic tail. These products give rise to the “heavy” a chain of the mature HLA class I molecule and define the HLA phenotype (e.g., HLA-A1 or HLA-A2). The heavy chain is noncovalently bound to a b2-microglobulin “light” chain whose gene resides on chromosome 15. The delineation of the crystallographic structure of HLA-A2 in 1987 was pivotal to understanding the structureefunction relationship of MHC molecules. Those landmark studies revealed that class I molecules are composed of two a-helical regions that overlay an eight-stranded antiparallel b-pleated sheet; together, these form the functional groove of class I molecules for peptide binding [6]. Sequence variation in exons 2, 3, and 4 define the allospecificity of HLA-A, HLA-B, and HLA-C antigens, respectively. As of June 2017, over 3913 HLA-A, 4765 HLA-B, and 3510 HLA-C alleles are recognized by the World Health Organization Nomenclature Committee for Factors of the HLA System [7,8]. The nucleotide substitutions in exons 2, 3, and 4 are commonly observed at hypervariable positions that define the peptide-binding groove and T-cell receptor (TCR) contact residues of the a1 and a2 domains [9]. In addition to interactions with the TCR, HLA-C and some HLA-B and HLA-A molecules serve as ligands for natural killer inhibitory receptors (KIRs). The specificity of ligandereceptor interactions is defined by residues 77e80 for HLA-C and by residues 77e83 (the Bw4 epitope) for HLA-B.
Nonclassical MHC Genes: HLA-E, HLA-F, HLA-G, and MIC In addition to the classical class I HLA loci, the class I region of the MHC encodes a series of genes that are termed the “nonclassical” class I genes, HLA-E, HLA-F, HLA-G, and the MHC class Ierelated chain A and B genes (MICA and MICB, respectively). Known as the class 1b family [10], these genes share homology but differ with respect to their specific regulation, expression patterns, and epigenetic profiles. A role of MICA ligands in transplantation outcome has recently been elucidated (Table 2.1).
HLA-E HLA-E is an extensively studied MHC class Ib antigen. In contrast to the exceptional diversity of classical MHC class I HLA-A, HLA-B, and HLA-C genes, HLA-E encodes 18 recognized proteins, with HLA-E*01:01 and HLA-E*01:03 the most frequently observed alleles worldwide [8]. HLA-E*01:03 differs from HLA-E*01:01 by a single amino acid substitution (gly to arg) at position 107 of the a2 heavy chain domain [8]. Although the two alleles appear to be evenly distributed in the human population (approximately 50% each), they differ with respect to their quantitative cell surface expression. HLA-E*01:03 is expressed at higher concentration on transfected cells compared to HLA-E*01:01 [11]. The HLA-E molecules preferentially bind nonameric self-peptides derived from the leader sequences of the various HLA class I
FIGURE 2.1 Map of the human major histocompatibility complex (MHC). The MHC is encoded on chromosome 6p21 and is organized into three major regions: class I, class III, and class II. The figure shows the location of each of the classical and nonclassical HLA genes that have been described as playing a role in hematopoietic cell transplantation. HLA, human leukocyte antigen.
The HLA System in Hematopoietic Stem Cell Transplantation Chapter | 2
17
TABLE 2.1 The Role of MHC Genes in Hematopoietic Cell Transplantation Mismatched Locus
Survival
GVHD
HLA-A
Y
[
[68,69,71e73,80,81,83,86]
HLA-B
Y
[
[68,69,71e73,80,81,83,85,86,107,108,112]
HLA-C
Y
[
[68,69,71,73,80e83,86,87,108,112,129]
HLA-DRB1
Y
[ [
HLA-DPB1
Relapse
References
[68,69,71e73,80,83,86,88,90,114] Y
[69,74e78,114e126,130]
HLA-E*
Y
[
[152e158]
HLA-G*
Y
[
[160e162,174e177]
[
[178,179,181,182]
MICA*
GVHD, graft-versus-host disease; HLA, human leukocyte antigen; MHC, major histocompatibility complex.
molecules. HLA-E also presents noncanonical peptides derived from pathogens or stress-related proteins. Overall, the HLA-E/peptide complexes differ in thermal stability [12]. As a ligand for the CD94/NKG2 receptors on NK cells [13] and for the TCR on NKT cells, HLA-E molecules are involved in both innate and adaptive immunity. Recent evidence supports a role for the involvement of HLA-E in presentation of peptides to the ab TCR expressed on CD8þ T cells. The HLA-E CD94/NKG2 A/C system modulates either inhibition or activation of the NK cellemediated cytotoxicity and cytokine production; at the same time, HLA-E can present microbial-derived peptides from human viruses or bacteria, thereby inducing T-cell responses. These diverse roles highlight the importance of HLA-E molecules as restriction elements for the specific T-cell responses against pathogens such as CMV, EpsteineBarr virus (EBV), or mycobacteria (Mycobacterium tuberculosis) [14e16]. In the same way, the Qa-1 molecule, a murine counterpart of the human HLA-E, has been shown to bind and present an immunodominant peptide recognized by salmonella-specific CD8þ T lymphocytes (CTL) and also participates in the host response against Listeria monocytogenes.
Human Leukocyte Antigen G The human leukocyte antigen G (HLA-G) is a nonclassical HLA class Ib locus whose products have distinct immunomodulatory, anti-inflammatory, and tolerogenic properties. HLA-G maps telomeric to HLA-A (Fig. 2.1). Although HLA-G molecules are structurally similar to their classical counterparts, they are distinguished by their limited tissue distribution in physiological conditions, the diversity of isoforms generated by alternative splicing (four membrane-bound [HLA-G1 to G4] and three soluble [HA-G5 to G7] isoforms [sHLA-G]) [17e19], and their unique pattern of polymorphisms in the noncoding regions, especially within the promoter and the 30 -untranslated region (30 UTR; [20]). To date, HLA-G alleles encode a limited number of exonic mutations; these polymorphisms are often silent mutations that result in 53 unique alleles that give rise to 18 protein variants [8]. HLA-G allele frequencies vary extensively between Caucasian, Asian, and African populations. HLA-G mediates immune responses of NK and T lymphocytes by interacting directly with a series of inhibitory receptors: KIR 2DL4 (CD158d) on NK and a subpopulation of T cells; ILT2 (CD85j) on T, B, NK, dendritic cell (DC) and monocytes, and ILT4 (CD85d) expressed exclusively on antigen presenting cells. HLA-G expression is regulated by variation within its 50 and 30 untranslated regions [21e23]. Furthermore, each HLA-G allele bears either a 14-base pair (bp) insertion (ins) or deletion (del) polymorphism in the 30 UTR which influences HLA-G expression through the shortening of exon 8 [24,25]. Such indels have functional consequences. The insertion allele (þ14 bp), albeit initially described to increase HLA-G mRNA stability, was subsequently correlated with lower levels of HLA-G mRNA and serum sHLA-G isoforms [25,26] with likely functional consequences on the properties of HLA-G molecules. Most recently, the rs1063320 single nucleotide polymorphism (SNP) has been shown to regulate HLA-G expression and correlate with disease risk [23,27e29]. Cell surface HLA-G was first found on cytotrophoblasts and shown to maintain fetalematernal tolerance, and it has been the maternalefetal tolerance model in which HLA-G has been studied most extensively [30,31]. HLA-G is also physiologically expressed on CD14þ monocytes and thymic epithelium. Overall, HLA-G molecules are involved in the
18
Immune Biology of Allogeneic Hematopoietic Stem Cell Transplantation
inhibition of NK cell activity, CD4þ T lymphocyte and DC maturation, apoptosis of CD8þ cytotoxic T cells (CTL), and development of regulatory T cells (Tregs). A role of HLA-G in both T and NK pathways has been studied in models of cancer, transplantation, and autoimmunity [19,20,32e35].
MIC Genes The MIC gene family comprises two expressed genes (MICA and MICB) and five pseudo genes (MICC, MICD, MICE, MICF, and MICG) [36]. Located at the centromeric end of HLA classical class I region, MICA maps approximately 46 kb from HLA-B. MICA is the most polymorphic nonclassical class I gene with over 106 alleles and 82 recognized proteins reported so far [8]. A distinguishing feature of MICA sequence variation is the presence of polymorphism in both the a2 and the a3 domains. In fact, none of the polymorphic residues of MICA correlate with those of the a1 and a2 domains of classical class I molecules, the latter of which contact the antigenic peptide and/or TCR. Overall, the significance of most MICA/B polymorphisms remains to be elucidated in terms of their ancestral origins and evolution. It is suggested that, based on the a3 domain polymorphisms, MICA alleles can be divided into two large families that might have evolved from two different ancestral lineages. From a functional point of view, a methionine (met) to valine (val) change at position 129 of the a2-heavy domain categorizes MICA alleles into strong (MICA-129 met) and weak (MICA-129 val) binders of the NKG2D receptor involved in activation and costimulation of NK and T cells [37]. Another level of diversity has been identified in the transmembrane (TM) region of MICA with the insertion of short tandem repeats (STRs) that result in a series of alleles (A4eA10). In addition, a nucleotide insertion (GCT / GGCT) in the TM region of the A5.1STR allele results in a premature stop codon. This sequence is present in MICA*008, the most frequent MICA allele described in various populations. Although similar in structure to an HLA class I heavy chain, MICA does not bind b2 microglobulin or any specific peptide. Therefore, MICA molecules are not involved in TCR-mediated immunity but rather engage NKG2D, a C-type lectin expressed on effector cells, including NK and ab- and gd-T cells [38e44]. Such engagement triggers NK cells and costimulates T lymphocytes to mount subsequent immune responses. Furthermore, a soluble isoform of MICA (sMICA), resulting from the proteolytic shedding of the membrane-bound molecules, was shown to result in NKG2D receptor downregulation. The ensuing immune modulation highlights the functional duality of the membrane-bound and soluble MICA isoforms. MICA expression is induced by cellular stress [45]. Preferentially expressed on epithelial and endothelial cells, MICA and MICB appear more ubiquitous at the mRNA level. Expression has also been reported in activated immune cells including DCs and T lymphocytes. The specific patterns of expression of MICA/B are related to the regulatory sequences that are devoid of interferon response elements, while a stress response element has been identified in the promoter region [46].
Class II Genes Historically the class II region has been known as the “HLA-D” region following the characterization of HLA-A, HLA-B, and HLA-C. The class II region has since been extensively sequenced, and its organization has been well defined in many populations [3]. Not only does the class II region encode genes that define the classical HLA-DR, HLA-DQ, and HLA-DP alloantigens, but also it contains genes that play a critical role in antigen loading and presentation (TAP and LMP); the concentration of genes with related function make class II a truly unique region of the human genome. The specific genes that define class II phenotypes are DRA1 and DRB1 (HLA-DR), DRB3 (HLA-DR52), DRB4 (HLA-DR53), DRB5 (HLA-DR54), DQA1 and DQB1 (HLA-DQ), and DPA1 and DPB1 (HLA-DP). The A genes (DRA1, DQA1, and DPA1) each encode the a chain of the mature class II molecule and have limited sequence diversity. The B genes (DRB1, DQB1, and DPB1) are each defined by a highly polymorphic exon 2 that gives rise to the b chain of the molecule. The a chain is noncovalently bound to the b chain [47]. The a-b heterodimer defines the phenotype of the antigen (e.g., DR1 or DQ2), with the b chain contributing the majority of the polymorphism that distinguishes unique allelic variants. The “supratypic” loci, DRB3, DRB4, and DRB5, are highly organized on DRB1 haplotypes, such that certain DRB1 genes have DRB3, DRB4, or DRB5 genes linked to their haplotype, whereas other DRB1 genes have no additional supratypic loci. In summary, DR1 and DR10 phenotypes have no additional supratypic gene; DR2 phenotypes have DRB5 that defines DR52; DR3, DR5, and DR6 have the DRB3 gene that defines the DR53 specificity, and DR4, DR7, and DR9 have the DRB4 gene that defines the DR54 specificity. Since a variable number of DRB genes are linked on haplotypes, different haplotypes have different lengths within the class II region. Yet another source of potential variation arising from
The HLA System in Hematopoietic Stem Cell Transplantation Chapter | 2
19
class II is contributed by DQA1 and DQB1. Trans pairing of DQ a encoded by one parental chromosome with a DQ b encoded by the other parental chromosome can give rise to up to four unique HLA-DQ antigens [48]. Like class I genes, HLA-DRB1, HLA-DQB1, and HLA-DPB1 genes are highly polymorphic, with more than 2311 HLA-DRB1, 1079 HLA-DQB1, and 828 DPB1 alleles recognized as of June 2017 [7,8]. Also similar to class I, the polymorphic sites within class II molecules are localized to discrete regions of the a1 and b1 domains of the a and b chains, respectively, to promote a large array of peptides that can be presented [9,49].
Class III Genes The Class III region resides in between class I and II and is now known to be the most gene-dense region of the entire human genome [50,51]. Importantly, the class III region displays strong positive LD with HLA class I loci to its telomere and with class II loci to its centromere; this LD is what characterizes the highly conserved “ancestral” haplotypes that have been defined in many human populations [52]. The class III region harbors genes that participate in the stress response, several of which have been found to influence risk of GVHD after allogeneic transplantation [53e55].
NOMENCLATURE The application of DNA-based methods for typing HLA genes has resulted in the discovery of novel alleles at an extraordinary rate [7,8]. To accommodate new sequence information, HLA nomenclature was recently modified using a naming system that offers unlimited numerical digits. The nomenclature captures four properties of sequences, in the following order: the serological equivalent of the allele, the unique sequence that gives rise to the unique protein, synonymous (silent) substitutions, and level of expression of the molecule. Each of these four characteristics is delimited by a colon (:). Using HLA-A*02:101:01:02N as an example, this name provides information on the serological specificity (02), the unique sequence (101), the synonymous substitution (01), and the null allele (02N). Additional letter suffixes denote proteins whose expression is low (L), soluble (S), aberrant (A), or whose product is cytoplasmic (C), or of questionable expression (Q).
LD and Haplotypes A hallmark of the MHC is its long-range positive LD, a mathematical measurement of two or more markers that have a higher observed frequency than would be predicted by their individual allele frequencies [56,57]. LD across the MHC demonstrates that occurrence of HLA tissue types is not random [52]. Linked HLA genes are inherited from each parent as a haplotype in classical Mendelian fashion. “Ancestral” haplotypes represent highly conserved HLA-A, HLA-B, HLA-DR haplotypes that display conservation for “blocks” or stretches of sequences in the class I, III, and II regions [52]. In disease mapping, haplotypes serve as proxies for ungenotyped markers because haplotypes define not one but many physically linked markers. In hematopoietic cell transplantation from unrelated donors, LD can both help and hinder the identification of suitable donors. In general, the probability of identifying an HLA-matched donor is higher when the patient and donor share a similar ethnic background [58e62]. When high LD exists, such as with the HLA-A1, -B8, -DR3 haplotype, the probability of finding donors with the same genotype is very high; when a patient has inherited a maternal or paternal recombination event, however, LD is disrupted, and the likelihood of identifying matched donors will then depend on the frequency of those alleles and antigens in the donor pool [63,64]. This is the case even for patients of European Caucasian background. In a recent analysis by the National Marrow Donor Program (NMDP), the overall rate of donorerecipient matching for HLA-A, HLA-B, HLA-C, HLA-DRB1, and HLA-DQB1 alleles was higher for Caucasian-Americans than for patients of all other races and ethnicities [65]. The likelihood of a patient identifying a donor with one HLA mismatch at either HLAA, HLA-B, HLA-C, or HLA-DRB1 was also higher for Caucasian-American patients but significantly increased the chances that a patient of non-Caucasian background could find an acceptable donor for transplantation [66]. These data suggest that understanding the properties of HLA mismatch combinations that are not associated with higher transplantrelated risks remains an important research effort so that more patients can benefit from transplantation.
THE ROLE OF CLASSICAL HLA IN UNRELATED DONOR HEMATOPOIETIC CELL TRANSPLANTATION The degree of HLA matching at the DNA or allele level is one of the strongest factors for transplant success [67e71]. Based on extensive worldwide data, HLA matching of the patient and the unrelated donor for HLA-A, -C, -B, -DRB1,
20
Immune Biology of Allogeneic Hematopoietic Stem Cell Transplantation
and -DQB1 alleles is associated with lower posttransplant risks and higher survival compared to mismatching [68,71e73]. Recent data support the prospective typing and matching of HLA-DPB1 to lower risks of GVHD [74e78]. Inclusion of HLA-DPB1 in standard HLA typing gives rise to 6 HLA loci, 12 determinants, and “12/12” matching with the patient and unrelated donor are HLA-A, HLA-B, HLA-C, HLA-DRB1, HLA-DQB1, HLA-DPB1 matched.
Alleles and Antigens Clinical practice in unrelated donor transplantation has mirrored the advances in HLA typing methodology over the past 4 decades. Antigens are defined by serological reagents in microcytotoxicity assays [79]. Alleles are defined by DNA-based methods that permit ascertainment of selected nucleotide positions of exons or full-length sequences. Since the definition of alleles and antigens is, in part, methodological, intense efforts have been made to determine whether there are biological differences associated with HLA mismatches that are detectable using serology (“antigen-level” or “low-resolution” mismatches) apart from mismatches that can only be detected using DNA-based methods (“allele-level” or “highresolution” mismatches). With the availability of molecular typing assays, it was appreciated that among phenotypically matched patients and unrelated donors, DNA-based typing methods can detect allelic differences that are clinically relevant [68,72,80e83]. One of the earliest demonstrations that alleles can be highly immunogenic came from observations in the setting of graft rejection after transplantation of a B*44:03 donor for a B*44:02 patient [84]. Donor-derived cytotoxic T lymphocytes could selectively recognize the patient’s HLA-B*44:02 allele. Following this report, the application of molecular methods was used to identify HLA-C allele mismatching and its role in graft failure [85]. These early studies firmly placed DNAbased typing methods at the forefront of clinical testing. Subsequently, large retrospective analyses have extended those findings to other HLA loci [68,71]. In a study from the NMDP-CIBMTR [68], survival was decreased by 10% for each mismatched HLA-A, HLA-B, HLA-C, or HLA-DRB1 locus. Allele mismatches were as detrimental as antigen mismatches with the exception being HLA-C, where antigen mismatches were more detrimental than allele mismatches. In peripheral blood stem cell transplantation [86], pronounced differences in mortality among patients mismatched with their donors for alleles versus antigens were found for HLA-C, but not for other loci. HLA-C has served as an important model for understanding differential risks conferred by allele and antigen mismatches [68,72,81,85]. Since its discovery as a classical transplantation antigen [87], donor mismatching for HLA-C has consistently been shown to be a risk factor after myeloablative, nonmyeloablative, unrelated donor, cord blood, marrow, and peripheral blood stem cell transplantation. In the era of growth factoremobilized peripheral blood stem cell transplantation, the potential effects of HLA disparity on outcome have been reassessed [86]. In a large retrospective analysis of HLA-A, -C, -B, -DRB1 alleleematched transplants, “8/8” donor matching was associated with improved 1-year survival compared to any single mismatch (“7/8”). Clinical differences in risks associated with HLA-C antigen versus HLA-C allele mismatches were observed. Specifically, donor HLA-C antigen mismatching was associated with the worst outcomes compared to HLA-A, -B, or -DRB1 mismatching. Whereas HLA-C antigen mismatching was associated with increased mortality, lowered disease-free survival, and increased grades IIIeIV acute GVHD, HLA-C allele mismatches did not increase risks. HLA-B allele or antigen mismatches were associated with GVHD; there were no statistically significant associations of HLA-A, -DRB1, or -DQB1 mismatches with transplant outcomes. These data suggest that for peripheral blood stem cell recipients, criteria for the selection of mismatched donors may not necessarily be the same as that for marrow sources. Future studies are warranted to fully evaluate the clinical significance of the stem cell source on the risks conferred by specific loci and resolution of donor matching.
Additive Effects of HLA Disparity A step-up of risks associated with increasing numbers of HLA mismatches is in keeping with a biological model for alloreactivity. Such multilocus mismatch effects have been observed in graft failure, GVHD, and mortality [68,72,81,83,85,88e91]. Clinical evidence demonstrating the deleterious nature of multilocus mismatching has led to the current practice of limiting the total number of HLA mismatches to one [67,69,70,74,78,85,92]. The impact of mismatching at a single HLA locus also depends on nongenetic factors that influence transplant outcome. Of these nongenetic factors, the stage of disease at the time of transplantation remains a strong factor for risk of disease recurrence and survival. Among good (low)-risk patients, the risks associated with donor HLA mismatching appear to be higher than those in patients with high-risk disease [68,88]. These observations are likely due to the cumulative effects of genetic and
The HLA System in Hematopoietic Stem Cell Transplantation Chapter | 2
21
nongenetic factors. Given the importance of an efficient search [93,94], careful consideration should be taken for extending a search when no HLA-matched donors are available. Not only is the total number of HLA mismatches an important determinant of risks, but the specific combination of mismatched loci can also define relative risks [68]. The criteria for prioritizing the selection of mismatched donors indicate that of all the classical HLA loci, an isolated HLA-DQB1 mismatch is better tolerated than mismatching at HLA-A, -C, -B, or -DRB1 [68,71]. Recently, the JMDP analyzed the effect of single DRB1, single DQB1, and two-locus DRB1/DQB1 mismatches; whereas single-locus mismatches did not increase risk, two-locus mismatches were associated with significantly inferior overall outcomes [69]. HLA-C disparity in the presence of mismatching at any other HLA locus (class I and/or class II) is associated with significantly increased incidence of grades IIeIV acute GVHD [72]. In the Japanese experience, HLA-C allele mismatching was an independent risk factor for severe acute GVHD. Lower rates of relapse among patients with GVHD were observed (the “graft-versus-leukemia effect” or GVLE as described below); when HLA-C mismatching occurred together with HLA-A, -B, -DR, or -DQ mismatching, survival was significantly lower.
Beneficial Effects of HLA Mismatching: Graft-Versus-Leukemia The lower risk of relapse observed in patients with GVHD is known as the graft-versus-leukemia effect [95e98]. Data demonstrate that of all the HLA loci, HLA-C and HLA-DPB1 mismatches are most strongly associated with graft-versusleukemia (GVL) [69], suggesting that not all allotypes can impart equivalent GVLE. In vitro data support the powerful GVLE of HLA-DPB1 allotypes [99] and the potential to harness this effect clinically to lower posttransplant disease recurrence.
NEW APPROACHES FOR DEFINING PERMISSIVE HLA MISMATCHES The feasibility of identifying HLA-matched donors depends on the HLA alleles, antigens and haplotypes of the patient, along with the size and composition of donor registries [58,61,100e106]. However, since every patient has a mismatched donor, intense efforts have been made to determine the characteristics of HLA mismatches that do not increase posttransplant risks (“permissive” or “tolerable” HLA mismatches). There are two models for permissive HLA mismatching that have substantial independent validation to support their use clinically: mismatching at key residues or epitopes of class I and II allotypes, and the level of HLA expression. The basis for the residue approach is the potential that certain HLA mismatches may be matched at key residues that define the peptide-binding grove of HLA molecules and that matching for such residues may be associated with lower risks than HLA mismatches that involve mismatching at key residues. Among the earliest studies, donorerecipient mismatching for residue 116 of HLA-B was significantly associated with increased risk of acute GVHD and transplant-related mortality [107]. In more recent analyses, donorerecipient mismatching for Tyr9ePhe9 of HLA-A and for Tyr9eSer9, Asn77e Ser77, Lys80eAsn80, Tyr99ePhe99, Leu116eSer116, and Arg156eLeu156 of HLA-C were identified in Japanese patients to be associated with increased risk of acute GVHD [108]. Since HLA-C serves as a ligand for the natural KIR (refer to section below), HLA-C mismatched pairs were further evaluated for the presence of residues that could be associated with GVHD risk beyond the two positions that define KIR receptor binding (positions 77 and 80); donore recipient mismatching at positions 9, 99, 156, and 163 strongly correlated with GVHD risk. In a subsequent analysis, certain HLA-C and HLA-DPB1 mismatch combinations were associated with both relapse and GVHD [109]. This study demonstrates that risks are not necessarily the same for every HLA mismatch; future studies are needed to separate mismatch combinations involved in GVL from those influencing GVHD. To this end, the use of statistical and structural modeling has been explored to identify HLA alleles with diverse peptide-binding repertoires [110,111]. Most recently, donorerecipient mismatching at residue 116 of class I was validated to be a risk factor for mortality [112]. HLA-DP has served as a model locus for understanding the immunogenicity of residue mismatching by way of in vitro cytotoxicity assays to identify the most immunogenetic epitopes [74,113]. Population studies have shown that HLA-DP is unique among other HLA genes because of very weak LD between HLA-DP and HLA-A, HLA-B, HLA-C, HLA-DR, and HLA-DQ. As a result, less than 20% of HLA-A, HLA-B, HLA-C, HLA-DRB1, and HLA-DQB1-matched unrelated donor pairs are also matched for HLA-DP. Retrospective examination of HLA-DP has required very large transplant populations so that sufficient numbers of HLA-DP-matched pairs can be compared with mismatched pairs [114e119]. Furthermore, the measured effects attributed to single loci in early studies likely measured additive effects of HLA-DP with HLA-A, HLA-B, and HLA-DR. HLA-DP does function as a classical transplantation antigen with respect to GVHD [117e119].
22
Immune Biology of Allogeneic Hematopoietic Stem Cell Transplantation
Mismatching for two DPB1 alleles increases the risk of acute GVHD compared to one or no HLA-DP mismatch [74]. More recent genome-wide association studies have validated HLA-DP as a classical transplantation determinant [75,77]. Given that 80% of HLA 10/10 allele matched donors are DPB1-mismatched, the frequencies suggest that for patients with common HLA haplotypes who can identify several HLA-A, HLA-C, HLA-B, DRB1, DQB1-matched donors, one of five donors could be DPB1-matched. Hence, the patients who are likely to benefit from prospective evaluation of DPB1 are patients who have common HLA haplotypes and for whom transplantation is not urgent. Considering that every patient has a DPB1-mismatched donor, studies have addressed whether all DPB1 mismatches are equally detrimental. These analyses have focused on the identification of donorerecipient mismatching at specific T-cell epitopes (TCEs) defined by exon 2 sequence variation, that are most closely associated with GVHD risk [120e123]. Using T-cell clones to identify DPB1 alleles that generate high cytotoxic potential, a schema for predicting high immunogenetic combinations of DPB1 alleles is possible. The TCE model has been independently validated in a large cohort of HLA-matched and -mismatched unrelated donor transplants from the International Histocompatibility Working Group in HCT as a clinically practical approach for selecting DPB1-mismatched donors whose mismatch may not increase risks [123]. The study furthermore demonstrates that the presence of permissive DPB1 mismatches in HLA-A-, HLA-C-, HLA-B-, DRB1-, or DQB1-mismatched transplants, may afford comparable outcomes with those observed after HLA 10/10-matched nonpermissive DPB1 mismatches. These data collectively suggest that when HLA 10/10-matched donors are not available, DPB1 typing and selection of TCE permissible donors may afford these patients similar favorable outcomes as a traditional HLA 10/10-matched transplant [120e126]. The second model for identifying permissive HLA mismatches focuses on the level of HLA expression of the patient’s mismatched HLA allotype that is recognized by the donor as foreign. Allotype-specific HLA-C expression has been elegantly demonstrated in cohorts of patients with HIV-AIDS and hepatitis [127,128]. In both scenarios, higher HLA-C expression is associated with lower progression to HIV-AIDS, and higher HLA-DP expression is associated with lower progression of hepatitis viremia. Recently, the impact of level of HLA-C and HLA-DP expression on GVH recognition was explored in two independent cohorts. Among unrelated donor transplants mismatched for a single HLA-C allotype, GVHD risk increased as the mean fluorescence intensity of the patient’s mismatched HLA-C increased [129]. Among HLA-A-, HLA-B-, HLA-C-, HLA-DRB1-, HLA-DQB1-matched unrelated donor transplants with a single HLA-DPB1 mismatch, mismatching against a high-expression HLA-DPB1 recipient allotype was associated with significantly increased GVHD [130]. Both the HLA-C and the HLA-DP models demonstrate that quantitative as well as qualitative differences between HLA-mismatched allotypes contribute to graft-versus-host allorecognition.
TOWARD A HAPLOTYPIC VIEW OF ALLOGENECITY HLA haplotypes represent a series of HLA genes and other loci that are physically linked on the same chromosomal strand (Fig. 2.1). The HLA region has served as a model for understanding patterns of LD [5,52,131e139]. HLA haplotypes are characterized by conserved “blocks” of genes and sequences of variable lengths [52]. This block-like structure distinguishes “ancestral” haplotypes that carry HLA-A, HLA-C, HLA-B, HLA-DRB1, and HLA-DQB1 alleles in strong positive LD with one another, and with key markers that reside in between the classical HLA loci [52]. Since the five classical HLA loci represent a fraction of the total gene content of the MHC [3], undetectable haplotypelinked variation could be responsible for increased risks after HLA-matched unrelated donor transplantation. Early observations pointed to donor-recipient disparity for variation that resides outside of the classical HLA loci [140,141]. In the DNA typing era, even with HLA 10/10 allele matching of unrelated donors, patients are at increased risk of GVHD and mortality. Since HLA-matched patients and unrelated donors are not identical by descent, it is possible that patient and donor HLA-A, HLA-B, and HLA-DR haplotypes are not the same. Using a patient and donor who are both HLA-A1,2/ B7,8/DR2,3 matched as an example, the HLA-A antigen could be linked to different HLA-B/DR antigens: A1/B8/DR3, A2/B7/DR2, A2/B8/DR3, and A1/B7/DR2. To test the hypothesis that HLA-A, HLA-B, HLA-DR-defined haplotypes can serve as markers for undetected linked genes that confer clinical effects, a novel method for phasing HLA alleles was developed to physically separate the two HLA haplotypes [142]. Application of the phasing method to HLA 10/10 allelematched unrelated patients and donors has revealed that a subset of identical pairs encode different HLA-A, HLA-B, HLADR haplotypes [143]. Patients transplanted from HLA-matched but haplotype-mismatched donors had significantly increased risk of severe acute GVHD. These observations suggest that there are undetected differences contributed by different haplotypes. What are the candidates within the MHC that could be responsible for GVHD risk? Microsatellite markers have been instrumental in probing the gene-dense MHC region for potential areas that harbor new transplantation determinants [144]. One of the earliest studies to define susceptibility genes was conducted in a Japanese population using microsatellite
The HLA System in Hematopoietic Stem Cell Transplantation Chapter | 2
23
markers to query the MHC [145]. Among patients who developed clinical acute GVHD, variation within the TNF complex was associated with lower survival. The importance of TNF polymorphisms has recently been confirmed in a study of Caucasian patients and transplant donors [54]. Microsatellites were used to define TNF alleles in HLA 10/10-matched transplant pairs. The pairs were further defined by their HLA-DPB1 match status. The presence of DPB1 mismatching concurrent with TNFd alleles was associated with increased GVHD risk and poorer overall outcome compared to their absence. This study suggests that variation within the class III TNF-defined region has clinical relevance. Furthermore, negative additive effects can be measured at both the TNF and the DP loci. The application of microsatellite markers paved the way for the use of state-of-the-art SNP platforms to understand haplotype content [146]. In a recent study by the Japanese Marrow Donor Program [147], HLA 10/10-matched patientedonor pairs were characterized using an SNP-based genotyping platform. Three high-frequency Japanese haplotypes were fully characterized for SNP content and LD. Interestingly, the Japanese patients and donors displayed a very high degree of SNP conservation across the MHC and precluded an analysis of the effect of SNP mismatching. However, a high frequency of HLA homozygosity permitted the presence of certain haplotypes to be analyzed for associated transplant risks. Three major haplotypes were identified, each having a different association to posttransplant complications. This study highlights the fundamental role of haplotypes and their associated variants in transplant outcome. In North American transplants, SNPs have been used to identify candidate regions for fine-mapping genes that influence clinical outcome after unrelated donor HCT. SNPs have been identified as markers for susceptibility genes in the class I and II regions in HLA-matched unrelated donor transplants [148]. In HLA-mismatched transplantation, candidate SNP markers provide a means to interrogate the gene-dense MHC for an efficient approach to identifying novel genes encoded on HLA haplotypes responsible for GVHD and mortality [149]. Future efforts to identify clinically relevant MHC region variants will be facilitated by the availability of nextgeneration sequencing methods [150]. Next-generation sequencing technology may permit phase to be established between markers and will significantly advance understanding of HLA haplotype content. The diversity of haplotypes in certain populations may permit donorerecipient mismatching of non-HLA variants to be more fully defined. The characterization of both common and rare haplotypes for the entire MHC region will greatly facilitate mapping efforts to localize novel transplantation determinants.
THE CLINICAL SIGNIFICANCE OF NONCLASSICAL HLA GENES: HLA-E, HLA-F, HLA-G, AND MIC GENES The measurement of transplant-associated risks conferred by the nonclassical HLA genes has been successful in selected sibling cohorts that limit the confounding effects of the classical HLA genes; however, due to the inherent long-range LD of the MHC, the number of sibling transplant pairs required to analyze the differential effect of the nonclassical HLA genes is necessarily high to reach sufficient statistical power [55,151].
HLA-E IN HCT: GVH As described previously, HLA-E can participate in the innate and adaptive immune pathways and has been studied as a transplantation determinant in related and unrelated donor transplantation [152e156]. The effect of HLA-E*01:03 homozygosity was first assessed in a cohort of 187 HLA-matched genotypically identical siblings from a single institution and revealed a protective effect for acute GVHD and transplant-related mortality (TRM) [152]. Patients with HLAE*01:01,01:01 or 01:01,01:03 genotypes showed a higher incidence of acute GVHD compared with patients with the HLA-E*01:03,01:03 genotype. This protective role was subsequently confirmed [154]. Similarly, a significant reduction of TRM was associated with HLA-E*01:03 homozygosity in a cohort of 83 related and unrelated HLA-matched transplants and translated into better overall survival [157]. The role of the HLA-E was confirmed in a study on 124 patients receiving an unrelated donor HCT, showing a decreased risk of acute GVHD, but no effect on survival [153], while in another study of 116 HLA-matched unrelated donor transplants, HLA-E polymorphism did not influence acute GVHD, TRM, or diseasefree survival [156]. The number of confounding factors is likely to be higher in unrelated donor than in sibling HCT and may very well explain the heterogeneous results in the two most recent studies. Interestingly, combined data from siblings and unrelated donor marrow transplants suggest a potential paradigm of an HLA-E*01:03 molecule that may present minor histocompatibility antigen peptides inefficiently without inducing T-cell recognition, while HLA-E*01:01 displays a lower capacity to present nominal antigen from pathogens [158]. More functionally integrated data are needed to elucidate and quantify the impact of HLA-E on GVH and overall in HCT outcomes. In addition, identification in renal transplantation of
24
Immune Biology of Allogeneic Hematopoietic Stem Cell Transplantation
anti-HLA-E antibodies that account for nonedonor-specific antibodies and correlate with lower graft survival may well have to be taken into account in the near future in HCT [159].
HLA-G IN HCT-GVH The pleiomorphic role in modulating immune responses justifies the interest of studying the HLA-G system in HCT both at genetic and functional pathophysiological levels [160e175]. The first study of HLA-G in transplantation was performed in 53 HLA-matched unrelated donor transplants for b thalassemia and found that the HLA-G 14-bp deletion polymorphism correlated with acute GVHD. While the acute GVHD risk associated with the HLA-G 14-bp del/del genotype [160] appears to be in contradiction with the established fact that the 14-bp del allele is associated with higher levels of sHLA-G than the 14-bp ins allele, the mechanism leading to clinical acute GVHD could however be different in the unrelated donor transplant situation. Because the HLA-matched unrelated donorerecipient pairs are not identical by descent, it could be possible that the HLA-G 14-bp del/del genotype may reflect the MHC non-HLA haplotype disparity. More recently, an analysis of 47 transplant patients with a variety of hematological malignancies failed to observe any significant association between the HLA-G 14-bp ins/del polymorphism and acute GVHD [161] but found that patients homozygous for the 14-bp ins allele were characterized by lower survival rate and disease-free survival. The authors related their findings to the possible relationship between HLA-G 14-bp dimorphism, methotrexate (MTX)-based acute GVHD prophylaxis and HLA-G expression. By contrast, in a recent series of 157 sibling pairs from a single institution, the HLA-G low expression genotype (ins/ins) was associated with severe acute GVHD [162]. In this study, the donorerecipient sibling pairs were fully matched for HLA-G genotypes with frequencies comparable to those previously reported. Univariate analysis using competing risk showed that the homozygous state of the HLA-G 14-bp ins/ins genotype was more prevalent among patients who experienced acute GVHD (grade 0, vs. II, III, IV) but failed to reach statistical significance (P ¼ .06). Nevertheless, additional univariate analyses after patient stratification based on acute GVHD severity (grade 0, I, II, vs. III, IV) revealed a significant association between the HLA-G 14bp ins/ins genotype and severe acute GVHD (22% in HLA-G 14-bp ins/ins vs. 6% in other; P ¼ .008). These data were further confirmed using two different multivariate analyses adjusted for confounding variables (gender, CMV status, age of recipient, and disease status). The only factor that remained statistically significant was the HLA-G 14-bp ins/ins genotype. Given the potential effect of MTX on HCT outcome in the context of HLA-G polymorphism [161], the subgroup who received this drug for acute GVHD prophylaxis (n ¼ 144) was reanalyzed. No change was found in the aforementioned association, which was strengthened despite a reduction in the sample size. These findings are in line with the data published in different clinical settings including gestational complications, auto-immunity, infections, cancers, as well as solid organ transplantation [163], although recent analysis has not observed correlations between regulatory region, 14 bp indel, and clinical outcomes [174,175]. Taken together, these results suggest that further investigation in larger transplant populations is warranted. The ins/del polymorphic variation in HLA-G seems to influence the RNA splicing stability by mechanisms that are yet to be understood. The presence of the 14-bp insertion introduces an additional splice site which results in the removal of the first 92 bp of exon 8 thereby generating more stable HLA-G mRNAs species than the complete mRNA [21,24,164]. In fact, the 30 UTR 14-bp insertion has been consistently associated with low expression of HLA-G mRNA and low serum sHLA-G [25,26]. Such a discrepancy between stability and output, termed “the 14-bp polymorphism paradox”, predicts complex mechanisms of regulation of HLA-G expression. Recent studies implicate the potential role of micro-RNAs which, by interacting with the HLA-G 30 mRNA region, may regulate its phenotypic expression [23,165]. Indeed, the observed effect of the insertion allele on acute GVHD could be either due to a haplotypic effect or due to another yet to be identified linked functional variant. The 14-bp ins/þ3142G/þ3187A haplotype has been studied with respect to risk of preeclampsia [163]. Data on the effect of the insertion allele in the incidence of acute GVHD are internally consistent with the concept that downregulated expression of HLA-G molecules could decrease immunosuppressive/tolerogenic properties that consequently result in the development of acute GVHD. The data are also in concordance with those showing that high pretransplantation and posttransplantation levels of sHLA-G molecules correlate not only with a decreased incidence of acute GVHD but also with a high frequency of circulating Tregs. Similar correlation between sHLA-G and Treg cells was also observed in in vitro mixed leukocyte reaction assay [166], in the context of liver transplantation [167] and in the transgenic murine model system [168]. These findings are also in agreement with most expression studies of HLA-G in solid organ transplantation that show beneficial effects of HLA-G molecules and lowered acute rejection/chronic dysfunction of the transplanted heart and kidney [169,170] and also with those establishing correlations between the presence of the HLA-G 14-bp ins allele and organ rejection [171e173]. HCT is a sensitive in vivo setting capable of revealing fine immune
The HLA System in Hematopoietic Stem Cell Transplantation Chapter | 2
25
response traits, undetectable in physiological situations. This may be a plausible reason why the effect of the ins/ins HLAG genotype on conferring low immune tolerance has been uncovered. More recently, studies have shown an increased level of HLA-G expression in GVHD target organs in patients receiving allogeneic hematopoietic stem cell transplants [176]. Furthermore, higher levels of soluble HLA-G can be found in the weeks after transplantation and correlate with protection against GVHD [177]. These studies collectively provide evidence for soluble HLA-G as an important factor in GVH responses.
MIC GENES IN HCT-GVH The investigation of MICA and MICB genes in transplantation has been hampered by the strong positive LD between these two loci and HLA-B; the LD favors MICA matching among HLA-matched transplants, thus necessitating very large transplant populations to isolate potential effects contributed by MICA. Nonetheless, the functional implications of the MICA 129 dimorphism have been investigated in many clinical settings including transplantation. In a cohort of 211 consecutive patients who underwent noneT-celledepleted allogenic HCT in a single institution from HLA identical siblings [178], MICA 129 genotyping (val/met) revealed that a recipient MICA 129 val/val genotype is a risk factor for chronic GVHD (63% vs. 45% at 3 years, P ¼ .03). The data were confirmed in multivariate analysis adjusted for confounding variables. Because acute GVHD is a major risk factor for subsequent chronic GVHD, acute GVHD was introduced as a time-dependent covariate in the multivariate analysis model. This analysis confirmed that the risk conferred by the MICA 129 val/val genotype is independent from acute GVHD. In an exploratory study of 236 URD transplants of which 73% of pairs were matched for HLA 10/10 alleles (HLA-A, -B, -C, -DRB1, -DQB1), MICA polymorphism was assessed by sequence-based typing methods and MICA mismatching was observed in 8.4%. A higher rate of grade IIeIV acute GVHD was observed in MICA disparate pairs (80% vs. 40%, P ¼ .003) irrespective of the degree of HLA matching. Furthermore, the rate of gastrointestinal acute GVHD was higher in MICA-mismatched patients (35% vs. 17%, P ¼ .5) [179] although the association of MICA mismatching with gastrointestinal GVHD was not observed in an independent study [180]. These results are reminiscent of the data obtained using the MHC block matching technique, a DNA-based MHC matching approach that uses non-HLA DNA polymorphisms in the MHC as markers of blocks of ancestral haplotypes. Matching for the MHC beta block inclusive of MIC genes was found to be beneficial over the classical HLA-B, HLA-C matching for survival in HCT [141]. In addition to MICA genetic polymorphism effects, additional MICA-related phenotypic features could influence the outcome of HCT. An elevated serum level of MICA was found to be associated with the incidence of chronic GVHD while the presence of MICA antibodies before transplantation was shown to confer protection against chronic GVHD [178]. The inverse relationship between MICA antibodies and sMICA suggests an antibody-based neutralization of deleterious effects of sMICA. Therefore, both genotypes and phenotypes of MICA represent important integrated biomarkers in HCT monitoring. One can speculate that the lower engagement of NKG2D receptors by the weak binder MICA 129val allele could impair the NK/cytotoxic T lymphocyte cell activation costimulation and possibly skew Th1 toward Th2 and subsequent B-cell activation and antibody production, two hallmarks of chronic GVHD pathogenesis. This is corroborated by the recent observation that sMICA-NKG2D engagement upregulates INF-g expression by CD56 bright NK cells and therefore contributes to systemic inflammation, a feature of chronic GVHD [162]. Two recent studies provide the most convincing evidence of MICA as a transplantation determinant [181,182]. In a large multicenter analysis coordinated in France [181], 922 HLA-10/10-matched unrelated donor transplants were evaluated and MICA mismatching was significantly associated with increased risk of clinical severe grade IIIeIV acute GVHD. In an independent study conducted by the German Registry for Stem Cell Transplantation [182], 2172 matched or mismatched patients were evaluated, and HLA 12/12,MICA-matched transplants had superior outcomes. By contrast, an analysis of North American patients by the CIBMTR did not observe an association between MICA mismatching and risks after transplantation [183]. Differing patient populations and transplant procedures may have contributed to these heterogeneous results. Future validation studies are needed to establish the rationale for prospective MICA typing and matching in unrelated donor transplantation.
CONCLUSIONS The assessment of the genetic risks associated with each locus of the MHC provides critical information for implementing comprehensive pretransplant genetic assessment and matching to lower risks for patients and for developing algorithms for targeted GVHD prevention strategies in HCT. A systems biology approach based on data from experimental and modeling outcomes represents a forward-looking strategy in this field. Analysis of the sequence, polymorphism, transcription, and
26
Immune Biology of Allogeneic Hematopoietic Stem Cell Transplantation
protein expression profiles of MHC and of the three-dimensional structure of the histocompatibility antigens has enabled the elucidation of the structureefunction relationship using innovative experimental approaches and computational tools. Gene expression profiling applied to an in situ skin explant model of GVHD was recently used to identify new candidate genes for controlling the risk of GVHD [184]. Hematopoietic cell transplantation is a unique model with which to understand the complex and systemic effects of introducing a healthy new genome into a patient to cure hematologic disorders. The drastic biological perturbations caused depend on both the genetic make-up of the donor and that of the recipient. Donor/recipient histocompatibility can be assessed by correlating biological and physiological effects, thus identifying the genetic contribution of MHC alleles, haplotypes, and their combined effects. This opens up, for example, the possibility of neutralizing MHC genetic differences by performing hematopoietic cell transplantation between HLA identical siblings. Such a clinical setting makes it possible to highlight the effects of other immunogenetic systems, such as minor histocompatibility loci, cytokines, receptors, which currently may have a more limited yet significant influence on the outcome of transplantation. Among the major bottlenecks in translating systems biology into individualized systems medicine is the limited number of clinical cases that can be included in randomized trials and the number of genetic and environmental variables that cannot be easily accounted for. With the increasing number of genetic systems and alleles that have to be taken into account, the number of transplants available for investigation is a major limiting factor. Are virtual patient models that mimic the patients’ main characteristics, from which testable hypotheses can be generated and validated on the small number of actual patients available, one solution? In the future, a systems biology approach and integrative methodologies will undoubtedly be needed to unravel the role of immunogenetics in transplantation to bring tailored and personalized treatment to the individual patient [185].
ACKNOWLEDGMENTS EWP is supported by grants CA18029, CA100019, CA162194, and AI069197 from the National Institutes of Health.
REFERENCES [1] [2] [3] [4] [5] [6] [7] [8] [9] [10] [11] [12] [13] [14] [15] [16] [17] [18]
Dausset J. Leuco-agglutinins IV: leuco-agglutinins and blood transfusion. Vox Sang 1954;4:190e8. van Rood JJ, van Leeuwen A. Leukocyte grouping. A method and its application. J Clin Invest 1963;42:1382e90. Trowsdale J. HLA genomics in the third millennium (Review). Curr Opin Immunol 2005;17:498e504. Charron D. Immunogenetics today: HLA, MHC and much more. Curr Opin Immunol 2005;17:493e7. Traherne JA. Human MHC architecture and evolution: implications for disease association studies. Int J Immunogenet 2008;35:179e92. Bjorkman PJ, Saper MA, Samraoui B, Bennett WS, Strominger JL, Wiley DC. Structure of the human class I histocompatibility antigen, HLA-A2. Nature 1987;329:506e12. Marsh SG, Albert ED, Bodmer WF, Bontrop RE, Dupont B, Erlich HA, et al. Nomenclature for factors of the HLA system 2004. Tissue Antigens 2005;65:301e69. European Bioinformatics Institute. IPD-IMGT/HLA database. Hinxton: European Bioinformatics Institute; 2016 [cited 2016 Dec 07]. Available from: http://www.ebi.ac.uk/ipd/imgt/hla/. Rammensee HG. Chemistry of peptides associated with MHC class I and class II molecules (Review). Curr Opin Immunol 1995;7:85e96. Rodgers JR, Cook RG. MHC class Ib molecules bridge innate and acquired immunity. Nat Rev Immunol 2005;5:459e71. Ulbrecht M, Couturier A, Martinozzi S, Pla M, Srivastava R, Peterson PA, et al. Cell surface expression of HLA-E: interaction with human beta2microglobulin and allelic differences. Eur J Immunol 1999;29:537e47. Strong RK, Holmes MA, Li P, Braun L, Lee N, Geraghty DE. HLA-E allelic variants. Correlating differential expression, peptide affinities, crystal structures, and thermal stabilities. J Biol Chem 2003;278:5082e90. Braud VM, Allan DS, O’Callaghan CA, Söderström K, D’Andrea A, Ogg GS, et al. HLA-E binds to natural killer cell receptors CD94/NKG2A. B and C. Nature 1998;391:795e9. Tomasec P, Braud VM, Rickards C, Powell MB, McSharry BP, Gadola S, et al. Surface expression of HLA-E, an inhibitor of natural killer cells, enhanced by human cytomegalovirus gpUL40. Science 2000;287:1031. Ulbrecht M, Modrow S, Srivastava R, Peterson PA, Weiss EH. Interaction of HLA-E with peptides and the peptide transporter in vitro: implications for its function in antigen presentation. J Immunol 1998;160:4375e85. Heinzel AS, Grotzke JE, Lines RA, Lewinsohn DA, McNabb AL, Streblow DN, et al. HLA-E-dependent presentation of Mtb-derived antigen to human CD8þ T cells. J Exp Med 2002;196:1473e81. Paul P, Cabestre FA, Ibrahim EC, Lefebvre S, Khalil-Daher I, Vazeux G, et al. Identification of HLA-G7 as a new splice variant of the HLA-G mRNA and expression of soluble HLA-G5, -G6, and -G7 transcripts in human transfected cells. Hum Immunol 2000;61:1138e49. Carosella ED, Moreau P, Le Maoult J, Le Discorde M, Dausset J, Rouas-Freiss N. HLA-G molecules: from maternal-fetal tolerance to tissue acceptance. Adv Immunol 2003;81:199e252.
The HLA System in Hematopoietic Stem Cell Transplantation Chapter | 2
27
[19] Carosella ED, Rouas-Freiss N, Tronik-Le Roux D, Moreau P, LeMaoult J. HLA-G: an immune checkpoint molecule. Adv Immunol 2015;127:33e144. [20] Larsen MH, Hviid TV. Human leukocyte antigen-G polymorphism in relation to expression, function, and disease. Hum Immunol 2009;70:1026e34. [21] Hviid TV, Hylenius S, Rorbye C, Nielsen LG. HLA-G allelic variants are associated with differences in the HLA-G mRNA isoform profile and HLA-G mRNA levels. Immunogenetics 2003;55:63e79. [22] Tan Z, Shon AM, Ober C. Evidence of balancing selection at the HLA-G promoter region. Hum Mol Genet 2005;14:3619e28. [23] Castelli EC, Mendes-Junior CT, Deghaide NH, et al. The genetic structure of 30 untranslated region of the HLA-G gene: polymorphisms and haplotypes. Genes Immun 2010;11:134e41. [24] Rousseau P, Le Discorde M, Mouillot G, Marcou C, Carosella ED, Moreau P. The 14 bp deletion-insertion polymorphism in the 3’ UT region of the HLA-G gene influences HLA-G mRNA stability. Hum Immunol 2003;64:1005e10. [25] Chen XY, Yan WH, Lin A, Xu HH, Zhang JG, Wang XX. The 14 bp deletion polymorphisms in HLA-G gene play an important role in the expression of soluble HLA-G in plasma. Tissue Antigens 2008;72:335e41. [26] Rizzo R, Rubini M, Govoni M, Padovan M, Melchiorri L, Stignani M, et al. HLA-G 14-bp polymorphism regulates the methotrexate response in rheumatoid arthritis. Pharmacogenet Genomics 2006;16:615e23. [27] Rizzo R, Hviid TV, Govoni M, et al. HLA-G genotype and HLA-G expression in systemic lupus erythematosus: HLA-G as a putative susceptibility gene in systemic lupus erythematosus. Tissue Antigens 2008;71:520e9. [28] Veit TD, Vianna P, Scheibel I, et al. Association of the HLA-G 14-bp insertion/deletion polymorphism with juvenile idiopathic arthritis and rheumatoid arthritis. Tissue Antigens 2008;71:440e6. [29] Veit TD, Cordero EA, Mucenic T, et al. Association of the HLA-G 14 bp polymorphism with systemic lupus erythematosus. Lupus 2009;18:424e30. [30] Geraghty DE, Pei J, Lipsky B, et al. Cloning and physical mapping of the HLA class I region spanning the HLA-E-to-HLA-F interval by using yeast artificial chromosomes. Proc Natl Acad Sci U S A 1992;89:2669e73. [31] Carosella ED, Gregori S, Rouas-Freiss N, LeMaoult J, Menier C, Favier B. The role of HLA-G in immunity and hematopoiesis. Cell Mol Life Sci 2011;68:353e68. [32] Rouas-Freiss N, Marchal RE, Kirszenbaum M, Dausset J, Carosella ED. The alpha1 domain of HLA-G1 and HLA-G2 inhibits cytotoxicity induced by natural killer cells: is HLA-G the public ligand for natural killer cell inhibitory receptors? Proc Natl Acad Sci U S A 1997;94:5249e54. [33] LeMaoult J, Krawice-Radanne I, Dausset J, Carosella ED. HLA-G1-expressing antigen-presenting cells induce immunosuppressive CD4þ T cells. Proc Natl Acad Sci U S A 2004;101:7064e9. [34] McIntire RH, Hunt JS. Antigen presenting cells and HLA-Gea review. Placenta 2005;26(Suppl A):S104e9. [35] Tripathi P, Abbas A, Naik S, Agrawal S. Role of 14-bp deletion in the HLA-G gene in the maintenance of pregnancy. Tissue Antigens 2004;64:706e10. [36] Bahram S. MIC genes: from genetics to biology. Adv Immunol 2000;76:1e60. [37] Steinle A, Li P, Morris DL, Groh V, Lanier LL, Strong RK, et al. Interactions of human NKG2D with its ligands MICA, MICB, and homologs of the mouse RAE-1 protein family. Immunogenetics 2001;53:279e87. [38] Tieng V, Le Bouguenec C, du Merle L, et al. Binding of Escherichia coli adhesin AfaE to CD55 triggers cell-surface expression of the MHC class I-related molecule MICA. Proc Natl Acad Sci U S A 2002;99:2977e82. [39] Bahram S, Bresnahan M, Geraghty DE, Spies T. A second lineage of mammalian major histocompatibility complex class I genes. Proc Natl Acad Sci U S A 1994;91:6259e63. [40] Bauer S, Groh V, Wu J, et al. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science 1999;285:727e9. [41] Groh V, Rhinehart R, Randolph-Habecker J, Topp MS, Riddell SR, Spies T. Costimulation of CD8alphabeta T cells by NKG2D via engagement by MIC induced on virus-infected cells. Nat Immunol 2001;2:255e60. [42] Billadeau DD, Upshaw JL, Schoon RA, Dick CJ, Leibson PJ. NKG2D-DAP10 triggers human NK cell-mediated killing via a Syk-independent regulatory pathway. Nat Immunol 2003;4:557e64. [43] Isernhagen A, Malzahn D, Viktorova E, et al. The MICA-129 dimorphism affects NKG2D signaling and outcome of hematopoietic stem cell transplantation. EMBO Mol Med 2015;7:1480e502. [44] Wesselkamper SC, Eppert BL, Motz GT, Lau GW, Hassett DJ, Borchers MT. NKG2D is critical for NK cell activation in host defense against Pseudomonas aeruginosa respiratory infection. J Immunol 2008;181:5481e9. [45] Groh V, Bahram S, Bauer S, Herman A, Beauchamp M, Spies T. Cell stress-regulated human major histocompatibility complex class I gene expressed in gastrointestinal epithelium. Proc Natl Acad Sci U S A 1996;93:12445e50. [46] Bahram S, Inoko H, Shiina T, Radosavljevic M. MIC and other NKG2D ligands: from none to too many. Curr Opin Immunol 2005;17:505e9. [47] Brown JH, Jardetzky TS, Gorga JC, Stern LJ, Urban RG, Strominger JL, et al. Three-dimensional structure of the human class II histocompatibility antigen HLA-DR1. Nature 1993;364:33e9. [48] Charron DJ, Lotteau V, Turmel P. Hybrid HLA-DC antigens provide molecular evidence for gene trans-complementation. Nature 1984;312:157e9. [49] Marshall KW, Liu AF, Canales J, Perahia B, Jorgensen B, Gantzos RD, et al. Role of the polymorphic residues in HLA-DR molecules in allelespecific binding of peptide ligands. J Immunol 1994;152:4946e57.
28
Immune Biology of Allogeneic Hematopoietic Stem Cell Transplantation
[50] Traherne JA, Horton R, Roberts AN, Miretti MM, Hurles ME, Stewart CA, et al. Genetic analysis of completely sequenced disease-associated MHC haplotypes identifies shuffling of segments in recent human history. PLoS Genet 2006;2:e9. [51] Horton R, Gibson R, Coggill P, Miretti M, Allcock RJ, Almeida J, et al. Variation analysis and gene annotation of eight MHC haplotypes: the MHC Haplotype Project. Immunogenetics 2008;60:1e18. [52] Yunis EJ, Larsen CE, Fernandez-Viña M, Awdeh ZL, Romero T, Hansen JA, et al. Inheritable variable sizes of DNA stretches in the human MHC: conserved extended haplotypes and their fragments or blocks. Tissue Antigens 2003;62:1e20. [53] Socie G, Loiseau P, Tamouza R, Janin A, Busson M, Gluckman E, et al. Both genetic and clinical factors predict the development of graft-versushost disease after allogeneic hematopoietic stem cell transplantation. Transplantation 2001;72:699e706. [54] Bettens F, Passweg J, Schanz U, Chalandon Y, Heim D, Gungor T, et al. Impact of HLA-DPB1 haplotypes on outcome of 10/10 matched unrelated hematopoietic stem cell donor transplants depends on MHC-linked microsatellite polymorphisms. Biol Blood Marrow Transpl 2012;18:608e16. [55] Dickinson AM, Charron D. Non-HLA immunogenetics in hematopoietic stem cell transplantation (Review). Curr Opin Immunol 2005;17:517e25. [56] Piazza A. Haplotype and linkage disequilibrium from the three-locus phenotypes. In: Kissmeyer-Nielsen F, editor. Histocompatibility testing. Copenhagen, Denmark: Munksgaard; 1975. p. 923e7. [57] Bodmer WF. HLA: a super supergene (Review). Harvey Lect 1978;72:91e138. [58] Beatty PG, Boucher KM, Mori M, Milford EL. Probability of finding HLA-mismatched related or unrelated marrow or cord blood donors. Hum Immunol 2000;61:834e40. [59] Lonjou C, Clayton J, Cambon-Thomsen A, Raffoux C. HLA -A, -B, -DR haplotype frequencies in France e implications for recruitment of potential bone marrow donors. Transplantation 1995;60:375e83. [60] Schipper RF, D’Amaro J, Oudshoorn M. The probability of finding a suitable related donor for bone marrow transplantation in extended families. Blood 1996;87:800. [61] Tiercy JM, Bujan-Lose M, Chapuis B, Gratwohl A, Gmur J, Seger R, et al. Bone marrow transplantation with unrelated donors: what is the probability of identifying an HLA-A/B/Cw/DRB1/B3/B5/DQB1-matched donor? Bone Marrow Transplant 2000;26:437e41. [62] Gragert L, Eapen M, Williams E, et al. HLA match likelihoods for hematopoietic stem-cell grafts in the U.S. registry. N Engl J Med 2014;371:339e48. [63] The Allele Frequency Net Database. Allele, haplotype and genotype frequencies in Worldwide Populations. 2016. Allelefrequencies.net. [cited 2016 Dec 013]. Available from: http://www.allelefrequencies.net. [64] National Marrow Donor Program. NMDP frequencies. Frequency.nmdp.org; 2016 [cited 2016 Dec 013]. Available from: http://frequency.nmdp. org/NMDPFrequencies2011/. [65] Dehn J, Buck K, Maiers M, et al. 8/8 and 10/10 high-resolution match rate for the be the match unrelated donor registry. Biol Blood Marrow Transplant 2015;21:137e41. [66] Buck K, Wadsworth K, Setterholm M, et al. High-resolution match rate of 7/8 and 9/10 or better for the Be the match unrelated donor registry. Biol Blood Marrow Transplant 2016;22:759e63. [67] Kollman C, Spellman SR, Zhang MJ, et al. The effect of donor characteristics on survival after unrelated donor transplantation for hematologic malignancy. Blood 2016;127:260e7. [68] Lee SJ, Klein J, Haagenson M, et al. High-resolution donor-recipient HLA matching contributes to the success of unrelated donor marrow transplantation. Blood 2007;110:4576e83. [69] Morishima Y, Kashiwase K, Matsuo K, et al. Biological significance of HLA locus matching in unrelated donor bone marrow transplantation. Blood 2015;125:1189e97. [70] Spellman S, Setterholm M, Maiers M, et al. Advances in the selection of HLA-compatible donors: refinements in HLA typing and matching over the first 20 years of the National Marrow Donor Program Registry. Biol Blood Marrow Transplant 2008;14:37e44. [71] Petersdorf EW, Anasetti C, Martin PJ, et al. Limits of HLA mismatching in unrelated hematopoietic cell transplantation. Blood 2004;104:2976e80. [72] Morishima Y, Sasazuki T, Inoko H, Juji T, Akaza T, Yamamoto K, et al. The clinical significance of human leukocyte antigen (HLA) allele compatibility in patients receiving a marrow transplant from serologically HLA-A, HLA-B, and HLA-DR matched unrelated donors. Blood 2002;99:4200e6. [73] Chalandon Y, Tiercy JM, Schanz U, Gungor T, Seger R, Halter J, Nicoloso de Faveri G, Roosnek E, Passweg JR, et al. Wiss Transplant Working Group for Blood, Marrow Transplantation (STABMT). Swiss National Donor Registry. Impact of high-resolution matching in allogeneic unrelated donor stem cell transplantation in Switzerland. Bone Marrow Transplant 2006;37:909. [74] Petersdorf EW, Gooley T, Malkki M, et al. The biological significance of HLA-DP gene variation in haematopoietic cell transplantation. Br J Haematol 2001;112:988e94. [75] Ogawa S, Matsubara A, Onizuka M, et al. Exploration of the genetic basis of GVHD by genetic association studies. Biol Blood Marrow Transplant 2009;15:39e41. [76] Shaw BE, Mayor NP, Russell NH, et al. Diverging effects of HLA-DPB1 matching status on outcome following unrelated donor transplantation depending on disease stage and the degree of matching for other HLA alleles. Leukemia 2010;24:58e65. [77] Martin PJ, Levine DM, Storer BE, et al. Genome-wide minor histocompatibility matching as related to the risk of graft-versus-host disease. Blood 2016. [78] Fernandez-Vina MA, Klein JP, Haagenson M, et al. Multiple mismatches at the low expression HLA loci DP, DQ, and DRB3/4/5 associate with adverse outcomes in hematopoietic stem cell transplantation. Blood 2013;121:4603e10.
The HLA System in Hematopoietic Stem Cell Transplantation Chapter | 2
29
[79] National Institutes of Health. NIH lymphocyte microcytotoxicity technique. In: Anonymous NIAID manual of tissue typing techniques. Atlanta, GA: Department of Health, Education, Welfare; 1979. NIH publication 80. [80] Petersdorf EW, Gooley TA, Anasetti C, Martin PJ, Smith AG, Mickelson EM, et al. Optimizing outcome after unrelated marrow transplantation by comprehensive matching of HLA class I and II alleles in the donor and recipient. Blood 1998;92:3515e20. [81] Sasazuki T, Juji T, Morishima Y, Kinukawa N, Kashiwabara H, Inoko H, et al. Effect of matching of class I HLA alleles on clinical outcome after transplantation of hematopoietic stem cells from an unrelated donor. N Engl J Med 1998;339:1177e85. [82] Ho VT, Kim HT, Liney D, Milford E, Gribben J, Culter C, et al. HLA-C mismatch is associated with inferior survival after unrelated donor nonmyeloablative hematopoietic stem cell transplantation. Bone Marrow Transplant 2006;37:845. [83] Flomenberg N, Baxter-Lowe LA, Confer D, Fernandez-Vina M, Filipovich A, Horowitz M, et al. Impact of HLA class I and class II high resolution matching on outcomes of unrelated donor bone marrow transplantation: HLA-C mismatching is associated with a strong adverse effect on transplant outcome. Blood 2004;104:1923e30. [84] Keever CA, Leong N, Cunningham I, Copelan EA, Avalos BR, Klein J, et al. HLA-B44-directed cytotoxic T cells associated with acute graftversus-host disease following unrelated bone marrow transplantation. Bone Marrow Transplant 1994;14:137e45. [85] Petersdorf EW, Hansen JA, Martin PJ, Woolfrey A, Malkki M, Gooley T, et al. Major-histocompatibility-complex class I alleles and antigens in hematopoietic-cell transplantation. N Engl J Med 2001;345:1794e800. [86] Woolfrey A, Klein JP, Haagenson M, Spellman S, Petersdorf E, Oudshoorn M, et al. HLA-C antigen mismatch is associated with worse outcome in unrelated donor peripheral blood stem cell transplantation. Biol Blood Marrow Transplant 2011;17:885e92. [87] Nagler A, Brautbar C, Slavin S, Bishara A. Bone marrow transplantation using unrelated and family related donors: the impact of HLA-C disparity. Bone Marrow Transplant 1996;18:891e7. [88] Petersdorf EW, Kollman C, Hurley CK, Dupont B, Nademanee A, Begovich AB, et al. Effect of HLA class II gene disparity on clinical outcome in unrelated donor hematopoietic cell transplantation for chronic myeloid leukemia: the US National Marrow Donor Program experience. Blood 2001;98:2922. [89] Petersdorf EW, Longton GM, Anasetti C, Mickelson EM, Smith AG, Martin PJ, et al. Definition of HLA-DQ as a transplantation antigen. Pro Natl Acad Sci USA 1996;93:15358e63. [90] Gajewski J, Gjertson D, Cecka M, Tonai R, Przepirka D, Hunt L, et al. The impact of T-cell depletion on the effects of HLA DR beta 1 and DQ beta allele matching in HLA serologically identical unrelated donor bone marrow transplantation. Biol Blood Marrow Transplant 1997;3:76e82. [91] Keever-Taylor CA, Bredeson C, Loberiza FR, Casper JT, Lawton C, Rizzo D, et al. Analysis of risk factors for the development of GVHD after Tcell-depleted allogeneic BMT: effect of HLA disparity, ABO incompatibility, and method of T-cell depletion. Biol Blood Marrow Transplant 2001;7:620e30. [92] Anasetti C, Hansen JA. Effect of HLA incompatibility in marrow transplantation from unrelated and HLA-mismatched related donors. Transfus Sci 1994;15:221e30. [93] Oudshoorn M, Cornelissen JJ, Fibbe WE, de Graeff-Meeder ER, Lie JL, Schreuder GM, et al. Problems and possible solutions in finding an unrelated bone marrow donor. Results of consecutive searches for 240 Dutch patients. Bone Marrow Transplant 1997;20:1011e7. [94] Heemskerk MB, van Walraven SM, Cornelissen JJ, Barge RM, Bredius RG, Egeler R, et al. How to improve the search for an unrelated haematopoietic stem cell donor. Faster is better than more! Bone Marrow Transplant 2005;35:645e52. [95] Weiden PL, Flournoy N, Thomas ED, et al. Antileukemic effect of graft-versus-host disease in human recipients of allogeneic-marrow grafts. N Engl J Med 1979;300:1068e73. [96] Horowitz MM, Gale RP, Sondel PM, et al. Graft-versus-leukemia reactions after bone marrow transplantation. Blood 1990;75:555e62. [97] Kolb HJ, Socie G, Duell T, et al. Malignant neoplasms in long-term survivors of bone marrow transplantation. Late effects working party of the European Cooperative Group for Blood and Marrow Transplantation and the European Late Effect Project Group. Ann Intern Med 1999;131:738e44. [98] Gratwohl A, Brand R, Apperley J, et al. Graft-versus-host disease and outcome in HLA-identical sibling transplantations for chronic myeloid leukemia. Blood 2002;100:3877e86. [99] Herr W, Eichinger Y, Beshay J, et al. HLA-DPB1 mismatch alleles represent powerful leukemia rejection antigens in CD4 T-cell immunotherapy after allogeneic stem-cell transplantation. Leukemia 2017;31:434e45. [100] Foeken LM, Green A, Hurley CK, Marry E, Wiegand T, Oudshoorn M. Donor Registries Working Group of the World Donor Association (WMDA). Monitoring the international use of unrelated donors for transplantation: the WMDA annual reports. Bone Marrow Transplant 2010;45:811e8. [101] Mori M, Beatty PG, Graves M, Boucher KM, Milford EL. HLA gene and haplotype frequencies in the North American population: the national marrow donor program donor registry. Transplantation 1997;64:1017e27. [102] Schipper RF, D’Amaro J, Bakker JT, Bakker J, van Rood JJ, Oudshoorn M. HLA gene haplotype frequencies in bone marrow donors worldwide registries. Hum Immunol 1997;52:54e71. [103] Oh HB, Kim SI, Park MH, Akaza T, Juji T. Probability of finding HLA-matched unrelated marrow donors for Koreans and Japanese from the Korean and Japan Marrow Donor Programs. Tissue Antigens 1999;53:347e9. [104] Velickovic ZM, Carter JM. Feasibility of finding an unrelated bone marrow donor on international registries for New Zealand patients. Bone Marrow Transplant 1999;23:291e4. [105] O’Shea J, Cleaver S, Little A-M, Madrigal A. Searching for an unrelated haemopoietic stem cell donor: a United Kingdom perspective. In: Cecka JM, Terasaki PI, editors. Clinical transplants 1999. Los Angeles, CA: UCLA Immunogenetics Center; 2000. p. 129e37.
30
Immune Biology of Allogeneic Hematopoietic Stem Cell Transplantation
[106] Takahashi K, Juji T, Miyazaki H. Determination of an appropriate size of unrelated donor pool to be registered for HLA-matched bone marrow transplantation. Transfusion 1989;29:311e6. [107] Ferrara GB, Bacigalupo A, Lamparelli T, Lanino E, Delfino L, Morabito A, et al. Bone marrow transplantation from unrelated donors: the impact of mismatches with substitutions at position 116 of the human leukocyte antigen class I heavy chain. Blood 2001;98:3150e5. [108] Kawase T, Morishima Y, Matsuo K, Kashiwase K, Inoko H, Saji H, et al. Japan Marrow Donor Program. High-risk HLA allele mismatch combinations responsible for severe acute graft versus host disease and implication for its molecular mechanism. Blood 2007;110:2235e41. [109] Kawase T, Matsuo K, Kashiwase K, Inoko H, Saji H, Ogawa S, et al. Japan Marrow Donor Program. HLA mismatch combinations associated with decreased risk of relapse: implications for the molecular mechanism. Blood 2009;113:2851e8. [110] DeLuca DS, Khattab B, Blasczyk R. A modular concept of HLA for comprehensive peptide binding prediction. Immunogenetics 2007;59:25e35. [111] Yanover C, Petersdorf EW, Malkki M, Gooley T, Spellman S, Velardi A, et al. HLA mismatches and hematopoietic cell transplantation: structural simulations assess the impact of changes in peptide binding specificity on transplant outcome. Immunome Res 2011;7:4. [112] Pidala J, Wang T, Haagenson M, et al. Amino acid substitution at peptide-binding pockets of HLA class I molecules increases risk of severe acute GVHD and mortality. Blood 2013;122:3651e8. [113] Fleischhauer K, Locatelli F, Zecca M, Orofino MG, Giardini C, De Stefano P, et al. Graft rejection after unrelated donor hematopoietic stem cell transplantation for thalassemia is associated with nonpermissive HLA-DPB1 disparity in host-versus-graft direction. Blood 2006;107:2984e92. [114] al Daccak R, Loiseau P, Rabian C, Devergie A, Bourdeau H, Raffoux C, et al. HLA-DR, DQ, and/or DP genotypic mismatches between recipientdonor pairs in unrelated bone marrow transplantation and transplant clinical outcome. Transplantation 1990;50:960e4. [115] Pawelec G, Ehninger G, Schmidt H, Schmidt H, Wernet P. HLA-DP matching and graft-versus-host disease in allogeneic bone marrow transplantation [published erratum appears in Transplantation 43:608, 1987] Transplantation 1986;42:558. [116] Kato Y, Mitsuishi Y, Cecka M, Hopfield J, Hunt L, Champlin R, et al. HLA-DP incompatibilities and severe graft-versus-host disease in unrelated bone marrow transplants. Transplantation 1991;52:374e6. [117] Varney MD, Lester S, McCluskey J, Gao X, Tait BD. Matching for HLA DPA1 and DPB1 alleles in unrelated bone marrow transplantation. Hum Immunol 1999;60:532e8. [118] Loiseau P, Espérou H, Busson M, Sghiri R, Tamouza R, Hilarious M, et al. DPB1 disparities contribute to severe GVHD and reduced patient survival after unrelated donor bone marrow transplantation (abstr). Blood 2001;98(Pt 1):660a. [119] Shaw BE, Marsh SG, Mayor NP, Russell NH, Madrigal JA. HLA-DPB1 matching status has significant implications for recipients of unrelated donor stem cell transplants. Blood 2006;107:1220e6. [120] Fleischhauer K, Zino E, Mazzi B, et al. Peripheral blood stem cell allograft rejection mediated by CD4(þ) T lymphocytes recognizing a single mismatch at HLA-DP beta 1*0901. Blood 2001;98:1122e6. [121] Zino E, Frumento G, Marktel S, et al. A T-cell epitope encoded by a subset of HLA-DPB1 alleles determines nonpermissive mismatches for hematologic stem cell transplantation. Blood 2004;103:1417e24. [122] Crocchiolo R, Zino E, Vago L, et al. Nonpermissive HLA-DPB1 disparity is a significant independent risk factor for mortality after unrelated hematopoietic stem cell transplantation. Blood 2009;114:1437e44. [123] Fleischhauer K, Shaw BE, Gooley T, Malkki M, Bardy P, Bignon JD, et al. On behalf of the International Histocompatibility Working Group in Hematopoietic Cell Transplantation. Effect of T-cell-epitope matching at HLA-DPB1 in recipients of unrelated-donor haemopoietic-cell transplantation: a retrospective study. Lancet Oncol 2012;13:366e74. [124] Pidala J, Lee SJ, Ahn KW, et al. Nonpermissive HLA-DPB1 mismatch increases mortality after myeloablative unrelated allogeneic hematopoietic cell transplantation. Blood 2014;124:2596e606. [125] Crivello P, Heinold A, Rebmann V, et al. Functional distance between recipient and donor HLA-DPB1 determines nonpermissive mismatches in unrelated HCT. Blood 2016;128:120e9. [126] Inamoto Y, Martin PJ, Flowers ME, et al. Genetic risk factors for sclerotic graft-versus-host disease. Blood 2016;128:1516e24. [127] Apps R, Qi Y, Carlson JM, et al. Influence of HLA-C expression level on HIV control. Science 2013;340:87e91. [128] Thomas R, Thio CL, Apps R, et al. A novel variant marking HLA-DP expression levels predicts recovery from hepatitis B virus infection. J Virol 2012;86:6979e85. [129] Petersdorf EW, Gooley TA, Malkki M, et al. HLA-C expression levels define permissible mismatches in hematopoietic cell transplantation. Blood 2014;124:3996e4003. [130] Petersdorf EW, Malkki M, O’HUigin C, et al. High HLA-DP expression and graft-versus-host disease. N Engl J Med 2015;373:599e609. [131] Miretti MM, Walsh EC, Ke X, Delgado M, Griffths M, Hunt S, et al. A high-resolution linkage-disequilibrium map of the human major histocompatibility complex and first generation of tag single-nucleotide polymorphisms. Am J Hum Genet 2005;76:634e6. [132] Dawkins R, Leelayuwat C, Gaudieri S, Tay G, Hui J, Cattley S, et al. Genomics of the major histocompatibility complex: haplotypes, duplication, retroviruses and disease (Review). Immunol Rev 1999;167:275e304. [133] De Bakker PI, McVean G, Sabeti PC, Miretti MM, Green T, Marchini J, et al. A high-resolution HLA and SNP haplotype map for disease association studies in the extended human MHC. Nat Genet 2006;38:1166e72. [134] Ahmad T, Neville M, Marshall SE, Armuzzi A, Mulcahy-Hawes K, Crawshaw J, et al. Haplotype-specific linkage disequilibrium patterns define the genetic topography of the human MHC. Hum Mol Genet 2003;12:647e56. [135] Allcock RJ, Atrazhev AM, Beck S, de Jong PJ, Elliott JF, Forbes S, et al. The MHC haplotype project: a resource for HLA-linked association studies. Tissue Antigens 2002;59:520e1.
The HLA System in Hematopoietic Stem Cell Transplantation Chapter | 2
31
[136] Baschal EE, Aly TA, Jasinski JM, Steck AK, Noble JA, Erlich HA, et al. Type 1 Diabetes Genetics Consortium. Defining multiple common “completely” conserved major histocompatibility complex SNP haplotypes. Clin Immunol 2009;132:203e14. [137] Smith WP, Vu Q, Li SS, Hansen JA, Zhao LP, Geraghty DE. Toward understanding MHC disease associations: partial resequencing of 46 distinct HLA haplotypes. Genomics 2006;87:561e71. [138] Walsh EC, Mather KA, Schaffner SF, Farwell L, Daly MJ, Patterson N, et al. An integrated haplotype map of the human major histocompatibility complex. Am J Hum Genet 2003;73:580e90. [139] Jeffreys AJ, Kauppi L, Neumann R. Intensely punctate meiotic recombination in the class II region of the major histocompatibility complex. Nat Genet 2001;29:217e22. [140] Ketheesan N, Gaudieri S, Witt CS, Tay GK, Townend DC, Christiansen FT, et al. Reconstruction of the block matching profiles. Hum Immunol 1999;60:171e6. [141] Tay GK, Witt CS, Christiansen FT, Charron D, Baker D, Herrmann R, et al. Matching for MHC haplotypes results in improved survival following unrelated bone marrow transplantation. Bone Marrow Transplant 1995;15:381e5. [142] Guo Z, Hood L, Malkki M, Petersdorf EW. Long-range multilocus haplotype phasing of the MHC [erratum appears in Proc Natl Acad Sci USA. 2006 Jun 13;103(24):9374] Proc Natl Acad Sci USA 2006;103:6964e9. [143] Petersdorf EW, Malkki M, Gooley TA, Martin PJ, Guo Z. MHC haplotype matching for unrelated hematopoietic cell transplantation. PLoS Med 2007;4:e8. [144] Malkki M, Gooley TA, Horowitz MM, Absi L, Christiansen FT, Cornelisseen JJ, et al. International histocompatibility Working Group in transplantation. Mapping MHC-resident transplantation determinants. Biol Blood Marrow Transplant 2007;13:986e95. [145] Li S, Kawata H, Katsuyama Y, Ota M, Morishima Y, Mano S, et al. Association of polymorphic MHC microsatellites with GVHD, survival, and leukemia relapse in unrelated hematopoietic stem cell transplant donor/recipient pairs matched at five HLA loci. Tissue Antigens 2004;63:362e8. [146] The Human Genome Project e Welcome Trust. Sanger Institute. http://www.sanger.ac.uk/HGP/Chr6?MHC. [147] Morishima S, Ogawa S, Matsubura A, Kawase T, Nannya Y, Kashiwase K, et al. Japan Marrow Donor Program. Impact of highly conserved HLA haplotype on acute graft-versus-host disease. Blood 2010;115:4664e70. [148] Petersdorf EW, Malkki M, Gooley TA, et al. MHC-resident variation affects risks after unrelated donor hematopoietic cell transplantation. Sci Transl Med 2012;4:144ra01. [149] Petersdorf EW, Malkki M, Horowitz MM, Spellman SR, Haagenson MD, Wang T. Mapping MHC haplotype effects in unrelated donor hematopoietic cell transplantation. Blood 2013;121:1896e905. [150] Norman PJ, Norberg SJ, Nemat-Gorgani N, et al. Very long haplotype tracts characterized at high resolution from HLA homozygous cell lines. Immunogenetics 2015;67:479e85. [151] Charron DJ. HLA matching in unrelated donor bone marrow transplantation. Curr Opin Hematol 1996;3:416e22. [152] Tamouza R, Busson M, Rocha V, et al. Homozygous status for HLA-E*0103 confers protection from acute graft-versus-host disease and transplant-related mortality in HLA-matched sibling hematopoietic stem cell transplantation. Transplantation 2006;82:1436e40. [153] Ludajic K, Rosenmayr A, Fae I, et al. Association of HLA-E polymorphism with the outcome of hematopoietic stem-cell transplantation with unrelated donors. Transplantation 2009;88:1227e8. [154] Hosseini E, Schwarer AP, Ghasemzadeh M. The impact of HLA-E polymorphisms in graft-versus-host disease following HLA-E matched allogeneic hematopoietic stem cell transplantation. Iran J Allergy, Asthma Immunol 2012;11:15e21. [155] Harkensee C, Oka A, Onizuka M, et al. Single nucleotide polymorphisms and outcome risk in unrelated mismatched hematopoietic stem cell transplantation: an exploration study. Blood 2012;119:6365e72. [156] Furst D, Bindja J, Arnold R, et al. HLA-E polymorphisms in hematopoietic stem cell transplantation. Tissue Antigens 2012;79:287e90. [157] Danzer M, Polin H, Pröll J, Haunschmid R, Hofer K, Stabentheiner S, et al. Clinical significance of HLA-E*0103 homozygosity on survival after allogeneic hematopoietic stem-cell transplantation. Transplantation 2009;27(88):528e32. [158] Tamouza R, Rocha V, Busson M, Fortier C, El Sherbini SM, Esperou H, et al. Association of HLA-E polymorphism with severe bacterial infection and early transplant-related mortality in matched unrelated bone marrow transplantation. Transplantation 2005;80:140e4. [159] Ravindranath MH, Pham T, Ozawa M, Terasaki PI. Antibodies to HLA-E may account for the non-donor-specific anti-HLA class-Ia antibodies in renal and liver transplant recipients. Int Immunol 2012;24:43e57. [160] La Nasa G, Littera R, Locatelli F, Lai S, Alba F, Caocci G, et al. The human leucocyte antigen-G 14-basepair polymorphism correlates with graftversus-host disease in unrelated bone marrow transplantation for thalassaemia. Br J Haematol 2007;139:284e8. [161] Chiusolo P, Bellesi S, Piccirillo N, Giammarco S, Marietti S, De Ritis D, et al. The role of HLA-G 14-bp polymorphism in allo-HSCT after shortterm course MTX for GvHD prophylaxis. Bone Marrow Transplant 2012;47:120e4. [162] Boukouaci W, Busson M, Fortier C, Amokrane K, Peffault de Latour R, Robin M, et al. Association of HLA-G low expressor genotype with severe acute graft-versus-host disease after sibling bone marrow transplantation. Front Immun 2011;2:1e6. [163] Larsen MH, Hylenius S, Andersen AM, Hviid TV. The 30 -untranslated region of the HLA-G gene in relation to pre-eclampsia: revisited. Tissue Antigens 2010;75:253e61. [164] O’Brien M, McCarthy T, Jenkins D, Paul P, Dausset J, Carosella ED, et al. Altered HLA-G transcription in pre-eclampsia is associated with allele specific inheritance: possible role of the HLA-G gene in susceptibility to the disease. Cell Mol Life Sci 2001;58:1943e9. [165] Veit TD, Chies JA. Tolerance versus immune response - microRNAs as important elements in the regulation of the HLA-G gene expression. Transpl Immunol 2009;20:229e31.
32
Immune Biology of Allogeneic Hematopoietic Stem Cell Transplantation
[166] Le Maux A, Noël G, Birebent B, Grosset JM, Vu N, De Guibert S, et al. Soluble human leucocyte antigen-G molecules in peripheral blood haematopoietic stem cell transplantation: a specific role to prevent acute graft-versus-host disease and a link with regulatory T cells. Clin Exp Immunol 2008;152:50e6. [167] Le Rond S, Le Maoult J, Créput C, Menier C, Deschamps M, Le Friec G, et al. Alloreactive CD4þ and CD8þ T cells express the immunotolerant HLA-G molecule in mixed lymphocyte reactions: in vivo implications in transplanted patients. Eur J Immunol 2004;34:649e60. [168] Ristich V, Zhang W, Liang S, Horuzsko A. Mechanisms of prolongation of allograft survival by HLA-G/ILT4-modified dendritic cells. Hum Immunol 2007;68:264e71. [169] Lila N, Carpentier A, Amrein C, Khalil-Daher I, Dausset J, Carosella ED. Implication of HLA-G molecule in heart-graft acceptance. Lancet 2000;355:2138. [170] Crispim JC, Duarte RA, Soares CP, Costa R, Silva JS, Mendes-Júnior CT, et al. Human leukocyte antigen-G expression after kidney transplantation is associated with a reduced incidence of rejection. Transpl Immunol 2008;18:361e7. [171] Crispim JC, Mendes-Junior CT, Wastowski IJ, Costa R, Castelli EC, Saber LT, et al. Frequency of insertion/deletion polymorphism in exon 8 of HLA-G and kidney allograft outcome. Tissue Antigens 2008;71:35e41. [172] Piancatelli D, Maccarone D, Liberatore G, Parzanese I, Clemente K, Azzarone R, et al. HLA-G 14-bp insertion/deletion polymorphism in kidney transplant patients with metabolic complications. Transplant Proc 2009;41:1187e8. [173] Twito T, Joseph J, Mociornita A, Rao V, Ross H, Delgado DH. The 14-bp deletion in the HLA-G gene indicates a low risk for acute cellular rejection in heart transplant recipients. J Heart Lung Transplant 2011;30:778e82. [174] Sizzano F, Testi M, Zito L, et al. Genotypes and haplotypes in the 30 untranslated region of the HLA-G gene and their association with clinical outcome of hematopoietic stem cell transplantation for beta-thalassemia. Tissue Antigens 2012;79:326e32. [175] Waterhouse M, Duque-Afonso J, Wasch R, Bertz H, Finke J. Soluble HLA-G molecules and HLA-G 14-base pair polymorphism after allogeneic hematopoietic cell transplantation. Transplant Proc 2013;45:397e401. [176] Lazana I, Zoudiari A, Kokkinou D, et al. Identification of a novel HLA-Gþ regulatory population in blood: expansion after allogeneic transplantation and de novo HLA-G expression at graft-versus-host disease sites. Haematologica 2012;97:1338e47. [177] Liu H, Chen Y, Xuan L, et al. Soluble human leukocyte antigen G molecule expression in allogeneic hematopoietic stem cell transplantation: good predictor of acute graft-versus-host disease. Acta Haematol 2013;130:160e8. [178] Boukouaci W, Busson M, Peffault de Latour R, Rocha V, Suberbielle C, Bengoufa D, et al. MICA-129 genotype, soluble MICA, and anti-MICA antibodies as biomarkers of chronic graft-versus-host disease. Blood 2009;114:5216e24. [179] Parmar S, Del Lima M, Zou Y, Patah PA, Liu P, Cano P, et al. Donor-recipient mismatches in MHC class I chain-related gene A in unrelated donor transplantation lead to increased incidence of acute graft-versus-host disease. Blood 2009;114:2884e7. [180] Anderson E, Grzywacz B, Wang H, et al. Limited role of MHC class I chain-related gene A (MICA) typing in assessing graft-versus-host disease risk after fully human leukocyte antigen-matched unrelated donor transplantation. Blood 2009;114:4753e4. author reply 4e5. [181] Carapito R, Jung N, Kwemou M, et al. Matching for the nonconventional MHC-I MICA gene significantly reduces the incidence of acute and chronic GVHD. Blood 2016;128:1979e86. [182] Fuerst D, Neuchel C, Niederwieser D, et al. Matching for the MICA-129 polymorphism is beneficial in unrelated hematopoietic stem cell transplantation. Blood 2016;128:3169e76. [183] Askar M, Sobecks R, Wang T, et al. MHC class I chain-related gene A (MICA) donor-recipient mismatches and MICA-129 Polymorphism in unrelated donor hematopoietic cell transplantations has no impact on outcomes in acute lymphoblastic leukemia, acute myeloid leukemia, or myelodysplastic syndrome: a Center for International Blood and Marrow Transplant Research study. Biol Blood Marrow Transplant 2017;23:436e44. [184] Novota P, Zinëcker S, Norden J, Wang XN, Sviland L, Opitz L, et al. Expression profiling of major histocompatibility and natural killer complex genes reveals candidates for controlling risk of graft versus host disease. PLoS One 2011;6:e16582. [185] Charron D. HLA, immunogenetics, pharmacogenetics and personalized medicine. Vox Sang 2011;100:163e6.