Abstract
Leukocytes that lack HLA allelic expression are frequently detected in patients with acquired aplastic anemia (AA) who respond to immunosuppressive therapy (IST), although the exact mechanisms underlying the HLA loss and HLA allele repertoire likely to acquire loss-of-function mutations are unknown. We identified a common nonsense mutation at position 19 (c.19C>T, p.R7X) in exon 1 (Exon1mut) of different HLA-A and -B alleles in HLA-lacking granulocytes from AA patients. A droplet digital PCR (ddPCR) assay capable of detecting as few as 0.07% Exon1mut HLA alleles in total DNA revealed the mutation was present in 29% (101/353) of AA patients, with a median allele frequency of 0.42% (range, 0.071% to 21.3%). Exon1mut occurred in only 12 different HLA-A (n=4) and HLA-B (n=8) alleles, including B*40:02 (n=31) and A*02:06 (n=15), which correspond to 4 HLA supertypes (A02, A03, B07, and B44). The percentages of patients who possessed at least one of these 12 HLA alleles were significantly higher in the 353 AA patients (92%, P<0.001) and in 83 AA patients with copy number neutral loss of heterozygosity in chromosome 6p (6pLOH) (100%, P<0.001) than that (81%) in 18,604 Japanese healthy individuals. 82% (37/45) of AA patients with Exon1mut responded to IST. Small populations of leukocytes that lack particular HLA-A or B alleles due to Exon1mut are common in AA patients. The detection of Exon1mut using the ddPCR assay without the need for HLA typing may serve as a powerful tool for diagnosing the immune pathophysiology of patients with bone marrow failure.Introduction
Acquired aplastic anemia (AA) is a rare condition characterized by pancytopenia and bone marrow hypoplasia resulting from immune-mediated suppression of hematopoietic stem progenitor cells (HSPC).1 Among several different immune mechanisms, cytotoxic T lymphocytes that recognize auto-antigens presented by HSPC are thought to play a critical role in the development of AA,2-7 based on the finding that leukocytes that lack particular HLA-A or HLA-B alleles (HLA-allelelacking leukocytes: HLA-LL) are often detected in the peripheral blood of AA patients.8,9 The presence of HLALL represents compelling evidence to support the involvement of cytotoxic T lymphocytes specific to HSPC in the development of bone marrow failure, and the detection of these leukocytes would be useful for diagnosing immune pathophysiology in patients with AA and other types of bone marrow failure, including low-risk myelodysplastic syndrome. However, assays for detecting HLA-LL, such as flow cytometry using monoclonal antibodies specific to HLA-A or HLA-B alleles and single nucleotide polymorphism (SNP) arrays, have not been popularized because of the need for HLA typing and low frequencies of copy number neutral loss of heterozygosity of the short arm of chromosome 6 (6pLOH).10,11
6pLOH was considered the most common way for HSPC to lose HLA alleles.10-15 We recently reported that, using targeted deep sequencing with a next-generation sequencer, somatic loss-of-function mutations of HLAB *40:02 were frequently detected in granulocytes of AA patients possessing HLA-B*40:02. These results strongly suggested that antigen presentation by HSPC via HLAB4002 plays a critical role in the pathogenesis of AA.9 Loss-of-function mutations in HLA class I alleles other than HLA-B*40:02 were also detected in patients with AA. Babushok et al. identified mutations in several HLA class I alleles in leukocytes of AA patients, including HLAA *33:03, A*68:01 and HLA-B*14:02.14 We recently analyzed leukocytes of AA patients with 6pLOH and detected somatic loss-of-function mutations in HLA-A*02:06 and B*54:01.16,17 However, HLA class I alleles responsible for autoantigen presentation in AA patients without HLAB *40:02, who account for approximately 80% of all AA patients, are largely unknown due to the limited number of AA patients who have been studied for loss-of-function mutations in HLA class I alleles.
To identify HLA class I alleles other than HLA-B*40:02 that are involved in the autoantigen presentation of AA, we performed targeted next-generation sequencing in AA patients with HLA-LL who had HLA class I alleles other than HLA-B*40:02. During the course of the mutation analysis, we identified a nonsense mutation at codon 19 (c.19C>T, p.R7X) in exon 1 (Exon1mut) of some HLA-A or HLA-B alleles. Surprisingly, Exon1mut was shared by different HLA-A or HLA-B alleles and was prevalent in AA patients although their variant allelic frequencies (VAF) were very low. A sensitive assay that can detect Exon1mut could help to identify HLA-A or HLA-B alleles that are responsible for autoantigen presentation and provide insight into the immune pathophysiology of bone marrow failure.
Against this backdrop, we developed a highly sensitive droplet digital PCR (ddPCR) assay for detecting Exon1mut, and determined the prevalence of Exon1mut and HLA alleles likely to acquire this mutation in AA patients.
Methods
Detailed information on the materials and methods are provided in the Online Supplementary Data.
Patients
Twenty Japanese AA patients with HLA-LL who did not have an HLA-B*40:02 allele were analyzed for the presence of loss-offunction mutations in HLA alleles. We studied a total of 353 Japanese AA patients, including the 20 patients who were further analyzed for the prevalence and clinical significance of Exon1mut in AA between 2010 and 2018 (Table 1). A schematic of the experiments is provided in Online Supplementary Figure S1. All patients were genotyped for HLA-A, HLA-B, HLA-C, and HLA-DRB1 alleles using the PCR sequence-specific oligonucleotide method. All patients provided consent to participation in this study, which was conducted in accordance with the Declaration of Helsinki and approved by the ethics committee of the Kanazawa University Institute of Medical, Pharmaceutical, and Health Sciences.
Detection of glycosylphosphatidylinositol-deficient cells and cells with 6p loss of heterozygosity
Glycosylphosphatidylinositol-anchored protein-deficient (GPI–) cells were detected using high sensitivity flow cytometry, as previously described.18 6pLOH was assessed using a SNP array-based method with GeneChip 500K arrays (Affymetrix, Japan) or a ddPCR assay with a QX200 AutoDG Droplet Digital PCR System (Bio-Rad, CA, USA), as previously described.9,10
Deep sequencing of HLA class I genes
From peripheral blood samples of the 20 patients with HLA-LL, which were stained with anti-HLA-allele-specific and lineage-specific monoclonal antibodies, paired fractions of granulocytes and CD3+ T cells were sorted and were subjected to DNA extraction (Online Supplementary Figure S2). The monoclonal antibodies used in this study are summarized in Online Supplementary Table S1. Nucleotide sequences of HLA-A and HLA-B genes in sorted granulocytes of patients with HLA-LL were determined using a nextgeneration sequencer (Miseq; Illumina, CA, USA).
Digital droplet polymerase chain reaction assay for detecting Exon1mut
We developed a sensitive ddPCR assay for precise detection of Exon1mut in the peripheral blood of AA patients using the QX200 ddPCR system. Briefly, we designed two different sets of primer pairs complementary to the consensus sequences of HLA-A and HLA-B alleles, and locked nucleic acid-based probes complementary to wild-type and mutant-specific sequences (Online Supplementary Table S2). Detailed protocols for ddPCR are provided in the Online Supplementary Methods.
Determination of HLA alleles that acquired Exon1mut
HLA alleles that acquired Exon1mut were determined by deep sequencing with the next-generation sequencer, or deduced from alleles contained in the lost haplotype due to 6pLOH that was accompanied by Exon1mut (Online Supplementary Figure S3). The low VAF of Exon1mut (VAF <1%) was confirmed by deep sequencing with unique molecular identifiers (xGen® Dual Index UMI Adapters: Integrated DNA Technologies, IA, USA).19 The correlation between Exon1mut’ VAF determined by deep sequencing with unique molecular identifiers and those determined by the ddPCR assay was examined using 24 different samples (Online Supplementary Figure S4). HLA class I alleles acquiring Exon1mutwere determined using the nearest allele-specific SNP. Details on deep sequencing with unique molecular identifiers are provided in the Online Supplementary Methods.
Statistical analysis
Comparisons were performed using the Fisher exact test for categorical variables and Mann-Whitney U test for continuous variables with a two-tailed significance level of 0.05. Statistical analyses were performed using the EZR software program.20 Graphs were generated using GraphPad PRISM7.0 (GraphPad Software Inc, CA, USA).
Results
Identification of Exon1mut in different HLA-A and HLA-B alleles in HLA-lacking leukocytes from patients with aplastic anemia
To identify HLA class I alleles other than HLA-B*40:02 that are critically involved in autoantigen presentation in AA, we sequenced HLA-A and HLA-B alleles of sorted granulocytes from 20 patients with HLA-LL not possessing HLA-B*40:02. The clinical characteristics of these 20 patients are shown in Online Supplementary Table S3. HLAA– granulocytes or HLA-A+B– granulocytes, accounting for 2.4-99.8% of all granulocytes, were detected in these patients (Online Supplementary Figure S5, Online Supplementary Table S3). Median read depths of the HLAA and HLA-B alleles were 925 and 1,012 for targeted deep sequencing and 43,013 and 35,267 for amplicon sequencing, respectively. Of the 20 AA patients assessed, six had 6pLOH alone, ten had various loss-of-function mutations in addition to 6pLOH, and four had only somatic loss-offunction mutations in HLA-A. Three (UPN 210, 335, and 348) of the 14 patients with loss-of-function mutations had the mutations in HLA-B of sorted HLA-A+ granulocytes. Of note, 12 of 14 patients with loss-of-function mutations had Exon1mut in HLA-A (A*02:06, n=7; A*31:01, n=1) and HLA-B (B*13:01, n=1; B*40:01, n=2; and B*54:01, n=1). The other two patients (UPN 335 and UPN 210) had different loss-of-function mutations from Exon1mut in HLAB *40:03 and HLA-B*54:01, respectively. Interestingly, a frameshift mutation of HLA-B*54:01 also occurred at codon 19 (c.19delC, p.R7Efs) in exon 1 (Figure 1A and B, Online Supplementary Table S4).
Exon1mut detection using a sensitive droplet digital polymerase chain reaction assay
To detect Exon1mut with high sensitivity and specificity, we established a ddPCR assay that allows for precise measurement of mutant allele frequency without the need for HLA typing. Tested samples containing a fixed amount of wild-type DNA and serial dilutions of Exon1mut template DNA revealed a detection limit of 0.07% for both HLA-A and HLA-B (Online Supplementary Figure S6). The ddPCR assay yielded 0% to 0.042% (median, 0.009%) positive dots in peripheral blood of 24 healthy individuals, validating the cut-off value of 0.07%. The ddPCR assay was able to detect Exon1mut, which had an allelic frequency of <1.0% (Figure 2A), clearly showing the presence of HLA-LL in patients for whom flow cytometry analysis of monocytes using anti-HLA-A2 antibodies produced unequivocal results regarding the presence of HLA-A2-lacking monocytes (Figure 2B).
Prevalence of Exon1mut in patients with aplastic anemia
Using two different ddPCR mixtures for HLA-A and HLA-B, the presence of Exon1mut was evaluated in all of the 353 patients. Exon1mut was detected in 101 (28.6%) of the 353 patients, with the median frequency of 0.42% (range, 0.071% to 21.3%). The prevalence of Exon1mut was similar in HLA-A and HLA-B alleles in both treatment-naïve patients (n=291) and treated patients (n=62). Among the AA patients with Exon1mut, those who had been treated had a higher median frequency of Exon1mut than those who were untreated (0.96% vs. 0.33%, P=0.0079) (Figure 3A). Figure 3B shows the relationships between the presence of cells with Exon1mut, 6pLOH and a paroxysmal nocturnal hemoglobinuria (PNH) phenotype in the patients. Fifty-six (55.4%) and 67 (66.3%) of the 101 patients with Exon1mut had 6pLOH and PNH phenotype cells, respectively. Ten (2.8%) of the 353 patients had Exon1mut alone. The frequency of Exon1mut was much lower than that of 6pLOH in 36 patients possessing both clones (P<0.001) (Figure 3C).
Long-term persistence of Exon1mut-positive cells
Serial blood samples were available for longitudinal analyses of Exon1mut in 13 patients who responded to immunosuppressive therapy (cyclosporine [CsA] alone in 6 and rabbit antithymocyte globulin [rATG] + CsA in 5) or anabolic steroids (n=2). Exon1mut was persistently detected for 14-86 months in nine patients, including one patient (UPN 299) who had been off treatment for 7 years, suggesting that Exon1mut-positive leukocytes are derived from long-lasting HSPC (Figure 4A). The VAF of Exon1mut increased in two (15%, UPN 333 and UPN 339), remained stable in four (31%), and decreased in three (23%) patients. Exon1mut became undetectable at 7-33 months after the first detection of Exon1mut in the other four patients, all of whom were being treated with CsA. Figure 4B shows a gradual decline of the Exon1mut frequency over 3 years in one patient (UPN 213).
HLA-A and HLA-B alleles that acquire Exon1mut
Among the 101 patients with Exon1mut, HLA alleles that acquired Exon1mut could be determined by targeted deep sequencing with (n=21) or without (n=37) unique molecular identifiers, or deduced from alleles contained in the lost haplotype due to 6pLOH that was accompanied by Exon1mut (n=10) (Online Supplementary Figure S2). In the other 33 patients with Exon1mut, HLA-A or HLA-B alleles that acquired Exon1mut could not be determined or deduced due to very low VAF (<0.2%), the absence of allele-specific SNP near Exon1mut in HLA-A or HLA-B alleles that are useful for identify missing HLA alleles, or the absence of coexisting 6pLOH. For the 68 patients in whom HLA alleles that acquired Exon1mut could be determined, the following 12 alleles were identified: A*02:01 (n=2), A*02:06 (n=15), A*02:07 (n=1), A*31:01 (n=3), B*13:01 (n=2), B*40:01 (n=3), B*40:02 (n=31), B*40:03 (n=1), B*44:03 (n=1), B*54:01 (n=6), B*55:02 (n=2), and B*56:01 (n=1) (Figure 5A). HLA class I supertypes of these alleles, which are defined by similarities in the antigen-presenting amino-acid motif of HLA alleles, were confined to only four supertypes: A02, A03, B07, and B44, except for HLAB *13:01 that does not belong to any of the 14 supertypes.21
When comparing the frequency of these 12 alleles between a healthy control population and our study cohort, 81% of 18,604 healthy Japanese individuals possessed at least one of the 12 alleles, while the prevalence was 92% in the 353 patients with AA (P<0.001) and 100% in the 83 patients with 6pLOH (P<0.001) (Figure 5B), suggesting the involvement of these alleles in the development of AA.
Loss of HLA-B expression from Exon1mut-positive leukocytes
Although Exon1mut in leukocytes is expected to result in lack of the corresponding HLA allele, the phenotype of these leukocytes is difficult to examine since the VAF of Exon1mut is very low. We previously established six induced pluripotent stem (iPS) cell clones from peripheral blood monocytes of an AA patient (UPN 333) whose monocytes included approximately 60% HLA-A24+Bw6– cells (Figure 6A).17 Deep sequencing revealed the presence of Exon1mut in sorted HLA-A24+Bw6– cells and also in one (clone C1) of the six iPS cell clones. When a wild-type iPS clone (clone E1) and clone C1 were induced to differentiate into CD34+ cells, all clone E1-derived CD34+ cells expressed HLA-Bw6 (B5401), while all clone C1-derived CD34+ cells lacked HLA-Bw6 (Figure 6B). The ddPCR assay using DNA from wild-type and Exon1mut-positive iPS cell-derived CD34+ cells revealed that the VAF of Exon1mut were 0.041% and 49%, respectively, as expected (Figure 6C).
Clinical characteristics of aplastic anemia patients with Exon1mut
Of the 291 patients whose peripheral blood samples were examined for Exon1mut and GPI– cells before treatment, 151 were evaluable for response to immunosuppressive therapy (CsA alone, n=68; CsA+rATG, n=83). The other 140 patients were excluded from the analysis of the relationship between the response to immunosuppressive therapy and the presence of Exon1mut or GPI– cells because no data on the response to immunosuppressive therapy were available in 84, and the remaining 56 received no treatment (n=25) or treatments other than immunosuppressive therapy (n=31), such as anabolic steroids and thrombopoietin receptor agonists, and allogeneic stem cell transplantation. An increase in GPI– cells was noted in 76% (34/45) of patients with Exon1mut and in 76% (81/106) without Exon1mut (P=1.0). In terms of response to immunosuppressive therapy, 82% (37/45) of patients with Exon1mut responded to CsA (n=14) or CsA+rATG (n=23), while 75% (79/106) of those without Exon1mut responded to CsA (n=35) or CsA+rATG (n=44) (P=0.40). The response rate to immunosuppressive therapy in patients with Exon1mut was significantly higher than that (54%, 13/24) in patients who were negative for all of Exon1mut, GPI– cells, and 6pLOH (P=0.023).
Discussion
Targeted deep sequencing of HLA genes of leukocytes obtained from AA patients with HLA-LL revealed a unique nonsense mutation at codon 19 (c.19C>T, p.R7X) in exon 1 (Exon1mut) of different HLA-A and HLA-B alleles. This mutation has been previously reported in Japanese and American AA patients, but did not draw attention because the mutation was detectable in only a limited number of patients.9,14 Our highly sensitive ddPCR assay enabled the detection of minor Exon1mut clones and detected the mutant DNA in nearly one third of Japanese AA patients regardless of the presence of 6pLOH. Exon1mut was also detected in two of eight Finnish AA patients we studied (unpublished observation). Interestingly, a frameshift mutation (c.19delC, p.R7Efs) was also identified at codon 19 of HLA-B*54:01 in a patient (UPN 210) without Exon1mut, suggesting that the codon 19 in exon 1 of HLA-A and HLA-B may be a specific position at which somatic mutations are likely to occur.
The loss of HLA from CD34+ cells due to Exon1mutwas verified by phenotypic analysis of Exon1mut-positive iPS cell- HSPC that were derived from monocytes of an AA patient who had approximately 14% Exon1mut-positive cells among the granulocyte population.17 Exon1mut has also been detected in several squamous cell carcinomas, such as head and neck tumors, oral cancers, and anal cancers, in previous studies.22-25 The solid tumors that lost HLA class I expression due to Exon1mut were thought to have escaped T-cell attack and acquired a proliferative advantage. Taken together, these findings suggest that Exon1mut is a common mechanism by which HSPC lose HLA, allowing them to escape from the effects of cytotoxic T lymphocytes in AA patients.
We previously used targeted deep sequencing to identify frequent loss-of-function mutations in three HLA class I alleles, B*40:02, A*02:06, and B*54:01.9,16,17 The highly sensitive ddPCR assay described herein that was capable of detecting Exon1mut newly identified three HLA-A (A*02:01, A*02:07, A*31:01) and six HLA-B alleles (B*13:01, B*40:01, B*40:03, B*44:03, B*55:02, B*56:01) as HLA alleles that are susceptible to allelic loss. Compared with their frequency in the general Japanese population, these HLA alleles were found to be highly enriched in AA patients. Among the 14 HLA class I supertypes that are defined based on similarities in the antigen-presenting peptide motif, the 12 alleles mentioned above belong to only four of the supertypes.21 These findings suggest that autoantigens of AA may be presented to T cells by these specific HLA alleles on HSPC.
Like HSPC positive for 6pLOH, those positive for Exon1mut are thought to escape the attack of cytotoxic T lymphocytes specific to autoantigens presented by the missing HLA-A or HLA-B allele and contribute to hematopoiesis over the long-term. However, it is unclear why Exon1mut occurs more frequently in HSPC than loss-offunction mutations in other positions of HLA class I alleles. Shukla et al. reported different hotspots of mutations in class I HLA genes according to cancer type, and identified Exon1mut only in head and neck squamous cell cancers. 24 HSPC may thus share a common property in that Exon1mut is likely to occur in class I HLA genes in head and neck squamous cell cancers.
The median VAF of Exon1mut in patients with Exon1mut patients was only 0.42%, a level that cannot be detected by targeted deep sequencing. This low VAF was in sharp contrast to the high proportion of concomitant 6pLOH in individual patients (Figure 3C). We previously reported that 6pLOH+ leukocytes were often polyclonal, consisting of leukocytes having different breakpoints of uniparental disomy in the short arm of chromosome 6.10 This polyclonality may account for the high proportion of 6pLOH. Although the leukocytes with Exon1mut represent a minor leukocyte population, the long-term (1-7 years) persistence of these mutated leukocytes indicates that they are derived from HSPC with self-renewal capacity. Arends et al. showed that clone size of cells with somatic mutations of epigenetic regulation genes expanded from most immature hematopoietic stem cells to mature peripheral blood cells in patients with clonal hematopoiesis of indeterminate potential.26 Leukocyte positive for Exon1mut may also be derived from most immature hematopoietic stem cells. The persistence of similarly minor clones in peripheral blood has been reported for GPI– granulocytes in AA, the median frequency of which was 0.25%.18,27 In contrast to PIGA-mutated or 6pLOH+ leukocytes, which can be oligoclonal and dysfunctional due to the lack of all GPIanchored proteins or a large segment of 6p, Exon1mut-positive leukocytes are derived from a single HSPC that is phenotypically normal except for the lack of one HLA allele. According to Dingli’s hypothesis, approximately 400 HSPC are actively involved in human hematopoiesis.28 Thus, the small proportion of Exon1mut-positive leukocytes among the entire leukocyte population may reflect an average clone size of individual HSPC in the bone marrow.
HLA-LL are useful markers that indicate the presence of an immune pathophysiology in patients with bone marrow failure. Here we showed a high response rate to immunosuppressive therapy in patients with Exon1mut, although patients without Exon1mut also had a high response rate likely due to the high prevalence of GPI– cells.29-31 Several methods can be used to detect HLA-LL, including flow cytometry assays with monoclonal antibodies specific to HLA-A or HLA-B alleles, ddPCR or SNP arrays for detecting 6pLOH, and targeted deep sequencing. 8-10,12 However, these methods require HLA typing of patients, take a long time to produce results, and are unable to detect HLA-LL that account for less than 1% of total leukocytes. The ddPCR assay used in the present study to detect Exon1mut enables the detection of HLA-LL accounting for as few as 0.07% of the total leukocyte population within 6 h of blood collection, highlighting the powerful nature of this assay for diagnosing immune pathophysiology in patients with bone marrow failure.
Footnotes
- Received February 16, 2020
- Accepted May 14, 2020
Correspondence
Disclosures No conflicts of interest to disclose.
Contributions
HM, TI, KT, YZ and SN collected clinical data and blood samples. FA performed HLA genotyping. YF and SO conducted the SNP array analyses. YZ, HT, TO, HK and AM generated an original monoclonal antibody specific to HLA-B13, B60 and B61. HM and TY performed cell sorting. HM, KHosomichi, TI, YZ and AT performed deep sequencing. HM, YZ, NMA and TCD performed the droplet digital polymerase chain reaction. KC and YY generated the induced pluripotent stem cells. MIE performed the in vitro experiments. HM, KHosokawa and SN designed the research and wrote the manuscript. All authors critically reviewed the manuscript and checked the final version.
References
- Young NS. Aplastic anemia. N Engl J Med. 2018; 379(17):1643-1656. https://doi.org/10.1056/NEJMra1413485PubMedPubMed CentralGoogle Scholar
- Zeng W, Nakao S, Takamatsu H. Characterization of T-cell repertoire of the bone marrow in immune-mediated aplastic anemia: evidence for the involvement of antigen-driven T-cell response in cyclosporine-dependent aplastic anemia. Blood. 1999; 93(9):3008-3016. https://doi.org/10.1182/blood.V93.9.3008.409a10_3008_3016PubMedGoogle Scholar
- Nakao S, Takami A, Takamatsu H. Isolation of a T-cell clone showing HLADRB1 *0405-restricted cytotoxicity for hematopoietic cells in a patient with aplastic anemia. Blood. 1997; 89(10):3691-3699. https://doi.org/10.1182/blood.V89.10.3691PubMedGoogle Scholar
- Risitano AM, Maciejewski JP, Green S. In-vivo dominant immune responses in aplastic anaemia: molecular tracking of putatively pathogenetic T-cell clones by TCR beta-CDR3 sequencing. Lancet. 2004; 364(9431):355-364. https://doi.org/10.1016/S0140-6736(04)16724-XPubMedGoogle Scholar
- Wlodarski MW, Gondek LP, Nearman ZP. Molecular strategies for detection and quantitation of clonal cytotoxic T-cell responses in aplastic anemia and myelodysplastic syndrome. Blood. 2006; 108(8):2632-2641. https://doi.org/10.1182/blood-2005-09-3902PubMedPubMed CentralGoogle Scholar
- Inaguma Y, Akatsuka Y, Hosokawa K. Induction of HLA-B*40:02-restricted T cells possessing cytotoxic and suppressive functions against haematopoietic progenitor cells from a patient with severe aplastic anaemia. Br J Haematol. 2016; 172(1):131-134. https://doi.org/10.1111/bjh.13464PubMedGoogle Scholar
- Espinoza JL, Elbadry MI, Chonabayashi K. Hematopoiesis by iPSC-derived hematopoietic stem cells of aplastic anemia that escape cytotoxic T-cell attack. Blood Adv. 2018; 2(4):390-400. https://doi.org/10.1182/bloodadvances.2017013342PubMedPubMed CentralGoogle Scholar
- Maruyama H, Katagiri T, Kashiwase K. Clinical significance and origin of leukocytes that lack HLA-A allele expression in patients with acquired aplastic anemia. Exp Hematol. 2016; 44(10):931-939.e3. https://doi.org/10.1016/j.exphem.2016.05.013PubMedGoogle Scholar
- Zaimoku Y, Takamatsu H, Hosomichi K. Identification of an HLA class I allele closely involved in the autoantigen presentation in acquired aplastic anemia. Blood. 2017; 129(21):2908-2916. https://doi.org/10.1182/blood-2016-11-752378PubMedGoogle Scholar
- Katagiri T, Sato-Otsubo A, Kashiwase K. Frequent loss of HLA alleles associated with copy number-neutral 6pLOH in acquired aplastic anemia. Blood. 2011; 118(25):6601-6609. https://doi.org/10.1182/blood-2011-07-365189PubMedGoogle Scholar
- Yoshizato T, Dumitriu B, Hosokawa K. Somatic mutations and clonal hematopoiesis in aplastic anemia. N Engl J Med. 2015; 373(1):35-47. https://doi.org/10.1056/NEJMoa1414799PubMedPubMed CentralGoogle Scholar
- Afable MG, Wlodarski M, Makishima H. SNP array-based karyotyping: differences and similarities between aplastic anemia and hypocellular myelodysplastic syndromes. Blood. 2011; 117(25):6876-6884. https://doi.org/10.1182/blood-2010-11-314393PubMedPubMed CentralGoogle Scholar
- Betensky M, Babushok D, Roth JJ. Clonal evolution and clinical significance of copy number neutral loss of heterozygosity of chromosome arm 6p in acquired aplastic anemia. Cancer Genet. 2016; 209(1-2):1-10. https://doi.org/10.1016/j.cancergen.2015.10.002PubMedPubMed CentralGoogle Scholar
- Babushok DV, Duke JL, Xie HM. Somatic HLA mutations expose the role of class I-mediated autoimmunity in aplastic anemia and its clonal complications. Blood Adv. 2017; 1(22):1900-1910. https://doi.org/10.1182/bloodadvances.2017010918PubMedPubMed CentralGoogle Scholar
- Montes P, Kerick M, Bernal M. Genomic loss of HLA alleles may affect the clinical outcome in low-risk myelodysplastic syndrome patients. Oncotarget. 2018; 9(97):36929-36944. https://doi.org/10.18632/oncotarget.26405PubMedPubMed CentralGoogle Scholar
- Imi T, Katagiri T, Hosomichi K. Sustained clonal hematopoiesis by HLAlacking hematopoietic stem cells without driver mutations in aplastic anemia. Blood Adv. 2018; 2(9):1000-1012. https://doi.org/10.1182/bloodadvances.2017013953PubMedPubMed CentralGoogle Scholar
- Elbadry MI, Mizumaki H, Hosokawa K. Escape hematopoiesis by HLA-B5401- lacking hematopoietic stem progenitor cells in men with acquired aplastic anemia. Haematologica. 2019; 104(10):e447-e450. https://doi.org/10.3324/haematol.2018.210856PubMedPubMed CentralGoogle Scholar
- Hosokawa K, Sugimori C, Ishiyama K. Establishment of a flow cytometry assay for detecting paroxysmal nocturnal hemoglobinuria- type cells specific to patients with bone marrow failure. Ann Hematol. 2018; 97(12):2289-2297. https://doi.org/10.1007/s00277-018-3443-1PubMedGoogle Scholar
- MacConaill LE, Burns RT, Nag A. Unique, dual-indexed sequencing adapters with UMIs effectively eliminate index crosstalk and significantly improve sensitivity of massively parallel sequencing. BMC Genomics. 2018; 19(1):30. https://doi.org/10.1186/s12864-017-4428-5PubMedPubMed CentralGoogle Scholar
- Kanda Y. Investigation of the freely available easy-to-use software 'EZR' for medical statistics. Bone Marrow Transplant. 2013; 48(3):452-458. https://doi.org/10.1038/bmt.2012.244PubMedPubMed CentralGoogle Scholar
- Sidney J, Peters B, Frahm N. HLA class I supertypes: a revised and updated classification. BMC Immunol. 2008; 9:1. https://doi.org/10.1186/1471-2172-9-1PubMedPubMed CentralGoogle Scholar
- Pickering CR, Zhang J, Yoo SY. Integrative genomic characterization of oral squamous cell carcinoma identifies frequent somatic drivers. Cancer Discov. 2013; 3(7):770-781. https://doi.org/10.1158/2159-8290.CD-12-0537PubMedPubMed CentralGoogle Scholar
- Mouw KW, Cleary JM, Reardon B. Genomic evolution after chemoradiotherapy in anal squamous cell carcinoma. Clin Cancer Res. 2017; 23(12):3214-3222. https://doi.org/10.1158/1078-0432.CCR-16-2017PubMedPubMed CentralGoogle Scholar
- Shukla SA, Rooney MS, Rajasagi M. Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat Biotechnol. 2015; 33(11):1152-1158. https://doi.org/10.1038/nbt.3344PubMedPubMed CentralGoogle Scholar
- Li YY, Chung GT, Lui VW. Exome and genome sequencing of nasopharynx cancer identifies NF-kappaB pathway activating mutations. Nat Commun. 2017; 8:14121. https://doi.org/10.1038/ncomms14121PubMedPubMed CentralGoogle Scholar
- Arends CM, Galan-Sousa J, Hoyer K. Hematopoietic lineage distribution and evolutionary dynamics of clonal hematopoiesis. Leukemia. 2018; 32(9):1908-1919. https://doi.org/10.1038/s41375-018-0047-7PubMedGoogle Scholar
- Sugimori C, Mochizuki K, Qi Z. Origin and fate of blood cells deficient in glycosylphosphatidylinositol- anchored protein among patients with bone marrow failure. Br J Haematol. 2009; 147(1):102-112. https://doi.org/10.1111/j.1365-2141.2009.07822.xPubMedGoogle Scholar
- Dingli D, Traulsen A, Pacheco JM. Compartmental architecture and dynamics of hematopoiesis. PLoS One. 2007; 2(4):e345. https://doi.org/10.1371/journal.pone.0000345PubMedPubMed CentralGoogle Scholar
- Sugimori C, Chuhjo T, Feng X. Minor population of CD55-CD59- blood cells predicts response to immunosuppressive therapy and prognosis in patients with aplastic anemia. Blood. 2006; 107(4):1308-1314. https://doi.org/10.1182/blood-2005-06-2485PubMedGoogle Scholar
- Kulagin A, Lisukov I, Ivanova M. Prognostic value of paroxysmal nocturnal haemoglobinuria clone presence in aplastic anaemia patients treated with combined immunosuppression: results of two-centre prospective study. Br J Haematol. 2014; 164(4):546-554. https://doi.org/10.1111/bjh.12661PubMedGoogle Scholar
- Narita A, Muramatsu H, Sekiya Y. Paroxysmal nocturnal hemoglobinuria and telomere length predicts response to immunosuppressive therapy in pediatric aplastic anemia. Haematologica. 2015; 100(12):1546-1552. https://doi.org/10.3324/haematol.2015.132530PubMedPubMed CentralGoogle Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.