TO THE EDITOR:

Acquired aplastic anemia (AA) is caused by immune-mediated destruction of hematopoietic stem and progenitor cells, resulting in bone marrow hypoplasia and pancytopenia in the peripheral blood.1,2  HLA class I allele–lacking (HLA) leukocytes are detected in ∼30% of patients with AA, and are thought to represent the involvement of a cytotoxic T lymphocyte attack against hematopoietic stem cells (HSCs) in the development of AA, based on the high response rate to immunosuppressive therapy (IST).3-6  Similarly to glycosylphosphatidylinositol-anchored protein–deficient (GPI) leukocytes in patients with paroxysmal nocturnal hemoglobinuria, HLA leukocytes in AA patients are often clonal or oligoclonal and expand to account for >50% of the total leukocytes.7  Despite such overwhelming proliferation, somatic mutations in driver genes, as well as telomere shortening that portends clonal evolution, are rarely detected in HLA granulocytes, suggesting the genetic stability of HLA HSCs.7 

Recent studies have reported an association between specific HLA alleles and clonal evolution to myelodysplastic syndrome (MDS)/acute myeloid leukemia (AML) in patients with AA.8,9  However, the association between HLA loss and clonal evolution to MDS/AML has not been extensively analyzed in larger AA cohorts. We therefore addressed this issue by studying the prognosis of a large number of patients with AA with or without HLA leukocytes who had been followed for a long period. We also studied the clonal composition of granulocytes in patients with AA with HLA cells, wherein aberrant clones other than HLA cells might be responsible for clonal evolution to MDS/AML.

We retrospectively analyzed the clinical characteristics of 633 patients with AA and examined peripheral blood samples for the presence of HLA leukocytes using a high-sensitivity flow cytometry assay, droplet digital polymerase chain reaction, single-nucleotide polymorphism arrays, or next-generation sequencing between 2010 and 2020.3-6  GPI cells were detected using a high-sensitivity flow cytometry assay, as previously described.10,11  Further experimental procedures are described in supplemental Methods (available on the Blood Web site). All patients provided their informed consent in accordance with the Declaration of Helsinki. This retrospective study was approved by the Ethics Committee of Kanazawa University School of Medical Science.

Table 1 shows the patient characteristics. The median age at diagnosis was 60 years (range, 2-91 years). The diagnoses of the patients were as follows: nonsevere AA (NSAA) (n = 385; 60.8%); severe AA (SAA) (n = 191; 30.2%), and very severe AA (n = 57; 9.0%). The median follow-up period was 55 months (range, 1-492 months). HLA granulocytes were detected in 127 of the 633 patients (20.0%) with a median clone size of 19.7% (range, 0.04% to 100%); the aberrant granulocytes accounted for >50% of the total granulocytes in 31 of the 127 patients (24.4%) (Figure 1A; supplemental Table 1). Four hundred eighteen of the 633 patients (66.0%) possessed GPI granulocytes with a median size of 0.20% (range, 0.003% to 100%), which was in line with previous studies (Figure 1B).10,12  The prevalence of GPI cells in patients with and without HLA cells did not differ to a statistically significant extent (66.9% vs 65.8%). On the other hand, 91 of 104 patients (87.5%) with HLA cells responded to IST, whereas 249 of 331 (75.2%) without HLA cells responded (P < .05; Figure 1C; supplemental Table 2). Ten patients with HLA cells did not receive any therapy, and only 2 patients (20%) showed spontaneous recovery without IST.

Table 1.

Patient characteristics

CharacteristicsNo. of patients
Median age (range), y 60 (2-91) 
Sex, M/F 295/338 
Diagnosis  
 AA 633 
Onset, n (%)  
 Adult 616 (97.3) 
 Pediatric 17 (2.7) 
Severity at diagnosis, n (%)  
 NSAA 385 (60.8) 
 SAA 191 (30.2) 
 VSAA 57 (9.0) 
Treatment  
 ATG + CsA ± TPO-RA 230 
 CsA ± TPO-RA 205 
 AS 22 
 PSL 
 HSCT 
 Others 
 None 69 
 Unknown 94 
Follow-up, median (range), mo 55 (1-492) 
Patients with HLA cells, n (%) 127 (20.0) 
Patients with GPI cells, n (%) 418 (66.0) 
CharacteristicsNo. of patients
Median age (range), y 60 (2-91) 
Sex, M/F 295/338 
Diagnosis  
 AA 633 
Onset, n (%)  
 Adult 616 (97.3) 
 Pediatric 17 (2.7) 
Severity at diagnosis, n (%)  
 NSAA 385 (60.8) 
 SAA 191 (30.2) 
 VSAA 57 (9.0) 
Treatment  
 ATG + CsA ± TPO-RA 230 
 CsA ± TPO-RA 205 
 AS 22 
 PSL 
 HSCT 
 Others 
 None 69 
 Unknown 94 
Follow-up, median (range), mo 55 (1-492) 
Patients with HLA cells, n (%) 127 (20.0) 
Patients with GPI cells, n (%) 418 (66.0) 

ATG, antithymocyte globulin; AS, anabolic steroids; CsA, cyclosporine; F, female; HSCT, hematopoietic stem cell transplantation; M, male; NSAA, nonsevere aplastic anemia; PSL, prednisolone; SAA, severe aplastic anemia; TPO-RA, thrombopoietin receptor agonist; VSAA, very severe aplastic anemia.

Figure 1.

Response to IST and clonal evolution to MDS/AML in patients with AA with or without HLA cells. (A) The distribution of the HLA granulocyte percentages in AA patients with HLA cells. Red bars indicate the median clone size. (B) The distribution of the GPI granulocyte percentages in AA patients with GPI cells. (C) The response to IST in AA patients with or without HLA cells. *A significant difference P < .05. (D) The cumulative incidence of clonal evolution to MDS/AML in the overall population of patiens with AA. (E) The cumulative incidence of clonal evolution to MDS/AML in AA patients with (red line) or without (black line) HLA cells. (F) The pie diagram indicates the proportions of concomitant aberrant clones detected in 127 patients with AA with HLA cells. The bar graphs depict the number of patients with AA with HLA cells and concomitant aberrant clones. (G) The pie diagrams indicates the proportions of HLA cells and concomitant aberrant clones in 2 representative cases (unique patient number [UPN] 313 and UPN 631).

Figure 1.

Response to IST and clonal evolution to MDS/AML in patients with AA with or without HLA cells. (A) The distribution of the HLA granulocyte percentages in AA patients with HLA cells. Red bars indicate the median clone size. (B) The distribution of the GPI granulocyte percentages in AA patients with GPI cells. (C) The response to IST in AA patients with or without HLA cells. *A significant difference P < .05. (D) The cumulative incidence of clonal evolution to MDS/AML in the overall population of patiens with AA. (E) The cumulative incidence of clonal evolution to MDS/AML in AA patients with (red line) or without (black line) HLA cells. (F) The pie diagram indicates the proportions of concomitant aberrant clones detected in 127 patients with AA with HLA cells. The bar graphs depict the number of patients with AA with HLA cells and concomitant aberrant clones. (G) The pie diagrams indicates the proportions of HLA cells and concomitant aberrant clones in 2 representative cases (unique patient number [UPN] 313 and UPN 631).

Close modal

The prognosis survey revealed that clonal evolution to MDS/AML did not occur in any of the 127 patients with HLA cells who were trackable, after a median follow-up period of 51 months. In contrast, 17 AA patients without HLA cells eventually evolved to MDS/AML (supplemental Table 3). The median time to progression was 24 months (range, 1-309 months). The cumulative incidence of clonal evolution to MDS/AML at 10 years was 4.5% (95% confidence interval, 2.4% to 7.6%) in the overall population of AA patients (Figure 1D), 0% in patients with HLA cells, and 5.8% (95% confidence interval, 3.1% to 9.8%) in patients without HLA cells (P < .05; Figure 1E). When the age at diagnosis, sex, severity, disease duration, GPI cells, and HLA cells were included in the multivariate analysis, the presence of HLA cells represented independent negative predictors of clonal evolution to MDS/AML (supplemental Table 4).

Interestingly, among the 127 patients with HLA cells, 88 patients (69.3%) possessed aberrant clones other than HLA clones, which included 0.005% to 91.6% GPI cells (n = 85), del(13q) cells (n = 3), trisomy 8 cells (n = 1), t(1;10) cells (n = 1), t(9;13) cells (n = 1), and inv12 cells (n = 1) (Figure 1F). Figure 1G shows 2 representative cases in which hematopoiesis was completely replaced by HLA cells and concomitant aberrant clones.

Finally, we identified 13 patients who had >90% HLA cells and who had been in hematological remission for >7 years (supplemental Table 5). Even though HLA cells and other concomitant aberrant cells accounted for >90% of granulocytes, these patients showed no signs of disease progression to MDS/AML. These data suggest that the escape clones were healthy enough to sustain the hematopoietic function of the patients.7,13 

The cumulative incidence of MDS/AML increases after IST, ranging from 5% to 15% within the observation period of 5 to 11.3 years.14-18  The relatively lower cumulative incidence of clonal evolution to MDS/AML at 10 years (4.5%) in comparison with previous studies from western countries may be attributed to the shorter follow-up period (median, 55 months) and the race of our study subjects (Japanese). Moreover, 60.8% our cohort was composed of NSAA cases, which may have also affected the incidence of clonal evolution. However, a recent study from the United States showed that the cumulative incidence of MDS/AML in moderate AA and SAA was not significantly different,19  a finding consistent with our analysis.

Strikingly, our study showed that the presence of HLA cells was an independent negative predictor of clonal evolution to MDS/AML. Our findings are consistent with the results of a previous study in that there was a lower risk of clonal evolution to MDS/AML but a higher rate of clonal hematopoiesis in patients with HLA cells than in those without HLA cells.8  In line with previous reports,20,21  7 (5.5%) had cytogenetic abnormalities in patients with 127 HLA cells, such as del(13q) and trisomy 8. The coexistence of HLA cells and these cytogenetically abnormal clones supports the idea that cells with some cytogenetic abnormalities may arise as a result of immune escape.13,22 

Retrospectives analyses using next-generation sequencing identified risk factors for post-AA MDS, including the age at the onset of AA, short leukocyte telomere length, and HLA class I risk alleles.8,23-27  The age at diagnosis of AA was not associated with an increased risk of clonal evolution to MDS/AML in our analysis. This seems reasonable given that the cumulative incidence of MDS/AML has been reported to be similar between adults and children.14-18  We did not have a chance to examine the telomere lengths of the patients or perform exon sequencing for all patients. However, our recent study showed no telomere attrition in HLA granulocytes; it also showed that somatic mutations related to myeloid malignancies were rarely detected in HLA granulocytes.7  The present study was associated with some limitations, including the heterogeneous patient population and retrospective setting.

In summary, the presence of HLA leukocytes and concomitant aberrant clones was not associated with clonal evolution to MDS/AML in Japanese patients with AA, even in those possessing a large (>90% of the total granulocyte) HLA cell population. Our data suggest that HLA HSC clones that escape immune attack are healthy enough to support hematopoiesis over the long-term in patients with AA.

Detailed data tables of clinical findings are available in supplemental Tables 1-5. For more information, please contact the corresponding author, Shinji Nakao, at snakao8205@staff.kanazawa-u.ac.jp.

The online version of this article contains a data supplement.

The authors thank Rie Ohmi (Kanazawa University) for excellent technical assistance. The authors thank all patients and their physicians of participating institutions who contributed to this study.

This work was supported by the following Ministry of Education, Culture, Sports, Science and Technology (MEXT) Grants-in-Aid for Scientific Research (KAKENHI): Grant-in-Aid for Scientific Research (B), numbers 16H05335 and 19H03686 (S.N.); Grant-in-Aid for Young Scientists (B), numbers 17K16184 and 19K17823 (K. Hosokawa); Grant-in-Aid for Scientific Research (C), number 17K09007 (T.K.); and Grant-in-Aid for Scientific Research on Innovative Areas, numbers 16H06502 and 19H05344 (K. Hosomichi). This work was also supported by a Hokuriku Bank Research Grant for Young Scientists (T.K.) and the Hokkoku Foundation for Cancer Research (T.K. and K. Hosokawa).

Contribution: K. Hosokawa and S.N. designed the research, analyzed the data, and wrote the manuscript; K. Hosokawa, H. Mizumaki, T.Y., H. Maruyama, T.I., N.T., R.U., M.T., Y.Z., K.I., H.Y., T.K., and S.N. recruited patients to participate in the study; H. Mizumaki, T.Y., H. Maruyama, T.I., and N.T. performed flow cytometry; H.T. generated antibodies for flow cytometry; F.A. performed HLA genotyping; S.O. conducted the single-nucleotide polymorphism array analyses; H. Mizumaki, T.I., Y.Z., K. Hosomichi, and A.T. performed deep sequencing; H. Mizumaki, Y.Z., M.A.T.N., and D.C.T. performed the droplet digital polymerase chain reaction; and all authors critically reviewed the manuscript and approved the submission of the final manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Shinji Nakao, Department of Hematology, Faculty of Medicine, Institute of Medical Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa 920-8640 Japan; e-mail: snakao8205@staff.kanazawa-u.ac.jp.

1.
Young
NS
,
Calado
RT
,
Scheinberg
P
.
Current concepts in the pathophysiology and treatment of aplastic anemia
.
Blood
.
2006
;
108
(
8
):
2509
-
2519
.
2.
Young
NS
.
Aplastic anemia
.
N Engl J Med
.
2018
;
379
(
17
):
1643
-
1656
.
3.
Maruyama
H
,
Katagiri
T
,
Kashiwase
K
, et al
.
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
.
4.
Katagiri
T
,
Sato-Otsubo
A
,
Kashiwase
K
, et al;
Japan Marrow Donor Program
.
Frequent loss of HLA alleles associated with copy number-neutral 6pLOH in acquired aplastic anemia
.
Blood
.
2011
;
118
(
25
):
6601
-
6609
.
5.
Mizumaki
H
,
Hosomichi
K
,
Hosokawa
K
, et al
.
A frequent nonsense mutation in exon 1 across certain HLA-A and -B alleles in leukocytes of patients with acquired aplastic anemia [published online ahead of print 21 May 2020]
.
Haematologica
.
doi:10.3324/haematol.2020.247809
.
6.
Zaimoku
Y
,
Takamatsu
H
,
Hosomichi
K
, et al
.
Identification of an HLA class I allele closely involved in the autoantigen presentation in acquired aplastic anemia [published correction appears in Blood. 2017;130(8):1072]
.
Blood
.
2017
;
129
(
21
):
2908
-
2916
.
7.
Imi
T
,
Katagiri
T
,
Hosomichi
K
, et al
.
Sustained clonal hematopoiesis by HLA-lacking hematopoietic stem cells without driver mutations in aplastic anemia
.
Blood Adv
.
2018
;
2
(
9
):
1000
-
1012
.
8.
Babushok
DV
,
Duke
JL
,
Xie
HM
, et al
.
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
.
9.
Zaimoku
Y
,
Adams
SD
,
Patel
BA
, et al
.
Spectrum and clinical significance of HLA class I alleles and their somatic mutations in immune aplastic anemia [abstract]
.
Blood
.
2019
;
134
(
suppl 1
).
Abstract 3738
.
10.
Hosokawa
K
,
Sugimori
C
,
Ishiyama
K
, et al
.
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
.
11.
Hosokawa
K
,
Ishiyama
K
,
Ikemoto
T
, et al
.
The clinical significance of PNH-phenotype cells accounting for < 0.01% of total granulocytes detected by the Clinical and Laboratory Standards Institute methods in patients with bone marrow failure [published online ahead of print 23 October 2020]
.
Ann Hematol
.
doi:10.1007/s00277-020-04314-w
.
12.
Sugimori
C
,
Mochizuki
K
,
Qi
Z
, et al
.
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
.
13.
Hosokawa
K
,
Katagiri
T
,
Sugimori
N
, et al
.
Favorable outcome of patients who have 13q deletion: a suggestion for revision of the WHO “MDS-U” designation
.
Haematologica
.
2012
;
97
(
12
):
1845
-
1849
.
14.
Tichelli
A
,
Schrezenmeier
H
,
Socié
G
, et al
.
A randomized controlled study in patients with newly diagnosed severe aplastic anemia receiving antithymocyte globulin (ATG), cyclosporine, with or without G-CSF: a study of the SAA Working Party of the European Group for Blood and Marrow Transplantation
.
Blood
.
2011
;
117
(
17
):
4434
-
4441
.
15.
Li
Y
,
Li
X
,
Ge
M
, et al
.
Long-term follow-up of clonal evolutions in 802 aplastic anemia patients: a single-center experience
.
Ann Hematol
.
2011
;
90
(
5
):
529
-
537
.
16.
Socié
G
,
Henry-Amar
M
,
Bacigalupo
A
, et al;
European Bone Marrow Transplantation-Severe Aplastic Anaemia Working Party
.
Malignant tumors occurring after treatment of aplastic anemia
.
N Engl J Med
.
1993
;
329
(
16
):
1152
-
1157
.
17.
Kojima
S
,
Ohara
A
,
Tsuchida
M
, et al;
Japan Childhood Aplastic Anemia Study Group
.
Risk factors for evolution of acquired aplastic anemia into myelodysplastic syndrome and acute myeloid leukemia after immunosuppressive therapy in children
.
Blood
.
2002
;
100
(
3
):
786
-
790
.
18.
Frickhofen
N
,
Heimpel
H
,
Kaltwasser
JP
,
Schrezenmeier
H
;
German Aplastic Anemia Study Group
.
Antithymocyte globulin with or without cyclosporin A: 11-year follow-up of a randomized trial comparing treatments of aplastic anemia
.
Blood
.
2003
;
101
(
4
):
1236
-
1242
.
19.
Patel
BJ
,
Barot
SV
,
Kuzmanovic
T
, et al
.
Distinctive and common features of moderate aplastic anaemia
.
Br J Haematol
.
2020
;
189
(
5
):
967
-
975
.
20.
Gupta
V
,
Brooker
C
,
Tooze
JA
, et al
.
Clinical relevance of cytogenetic abnormalities at diagnosis of acquired aplastic anaemia in adults
.
Br J Haematol
.
2006
;
134
(
1
):
95
-
99
.
21.
Ohga
S
,
Ohara
A
,
Hibi
S
, et al;
Aplastic Anaemia Committee of the Japanese Society of Paediatric Haematology
.
Treatment responses of childhood aplastic anaemia with chromosomal aberrations at diagnosis
.
Br J Haematol
.
2002
;
118
(
1
):
313
-
319
.
22.
Hosokawa
K
,
Sugimori
N
,
Katagiri
T
, et al
.
Increased glycosylphosphatidylinositol-anchored protein-deficient granulocytes define a benign subset of bone marrow failures in patients with trisomy 8
.
Eur J Haematol
.
2015
;
95
(
3
):
230
-
238
.
23.
Yoshizato
T
,
Dumitriu
B
,
Hosokawa
K
, et al
.
Somatic mutations and clonal hematopoiesis in aplastic anemia
.
N Engl J Med
.
2015
;
373
(
1
):
35
-
47
.
24.
Negoro
E
,
Nagata
Y
,
Clemente
MJ
, et al
.
Origins of myelodysplastic syndromes after aplastic anemia
.
Blood
.
2017
;
130
(
17
):
1953
-
1957
.
25.
Kulasekararaj
AG
,
Jiang
J
,
Smith
AE
, et al
.
Somatic mutations identify a subgroup of aplastic anemia patients who progress to myelodysplastic syndrome
.
Blood
.
2014
;
124
(
17
):
2698
-
2704
.
26.
Dumitriu
B
,
Feng
X
,
Townsley
DM
, et al
.
Telomere attrition and candidate gene mutations preceding monosomy 7 in aplastic anemia
.
Blood
.
2015
;
125
(
4
):
706
-
709
.
27.
Babushok
DV
.
A brief, but comprehensive, guide to clonal evolution in aplastic anemia
.
Hematology Am Soc Hematol Educ Program
.
2018
;
2018
:
457
-
466
.

Supplemental data

Sign in via your Institution