You are on page 1of 9

Annals of Hematology

https://doi.org/10.1007/s00277-020-04296-9

ORIGINAL ARTICLE

Long-term outcomes of 172 children with severe aplastic anemia


treated with rabbit antithymocyte globulin and cyclosporine
Yang Lan 1 & Lixian Chang 1 & Meihui Yi 1 & Yuli Cai 1 & Jing Feng 1 & Yuanyuan Ren 1 & Chao Liu 1 & Xiaoyan Chen 1 &
Shuchun Wang 1 & Ye Guo 1 & Aoli Zhang 1 & Lipeng Liu 1 & Jingliao Zhang 1 & Xiaofan Zhu 1

Received: 27 May 2020 / Accepted: 3 October 2020


# Springer-Verlag GmbH Germany, part of Springer Nature 2020

Abstract
This study retrospectively analyzed the clinical outcome of 172 children with newly diagnosed severe aplastic anemia (SAA) between
January 2008 and April 2018, who received rabbit antithymocyte globulin (ATG) and cyclosporine (CsA) as first-line treatment. The
median age at diagnosis was 5 years (range, 1–14). The overall response rates were 22.7%, 45.3%, and 61% at 40 days, 3 months, and
6 months, respectively, after rabbit ATG. In multivariate analysis, mild disease severity was the only predictor of favorable response at
6 months (P = 0.006). In the present study, median follow-up period was 63 months (range, 1–135). The 5-year overall survival (OS)
and failure-free survival (FFS) rates were 90.5% and 70.4%. Multivariate analysis showed that erythroid burst-forming units (BFU-E)
> 2/105 bone marrow mononuclear cell (BMMNC) (P = 0.037) and time interval before IST ≤ 30 days (P = 0.017) were independent
positive predictors for OS, meanwhile BFU-E > 2/105BMMNC (P = 0.029) was the only favorable prognostic factor for FFS.

Keywords Antithymocyte globulin . Child . Immunosuppressive therapy . Predictor . Severe aplastic anemia

Abbreviations aAA Acquired aplastic anemia


IST Immunosuppressive therapy HSCs Hematopoietic stem cells
ATG Antithymocyte globulin HSCT Hematopoietic stem cell transplantation
CsA Cyclosporine ALG Antilymphocyte globulin
SAA Severe aplastic anemia AA Aplastic anemia
HLA Human leukocyte antigen ANC Absolute neutrophil count
vSAA Very severe aplastic anemia PLT Platelet
OS Overall survival ARC Absolute reticulocyte count
FFS Failure-free survival CR Complete response
BFU-E Erythroid burst-forming units HGB Hemoglobin
BMMNC Bone marrow mononuclear cell NR Nonresponse
PB Peripheral blood PR Partial response
BM Bone marrow IMDM Iscove’s Modified Dulbecco’s Medium
FBS Fetal bovine serum
PNH Paroxysmal nocturnal hemoglobinuria
Electronic supplementary material The online version of this article CFU-GM Granulocyte-macrophage
(https://doi.org/10.1007/s00277-020-04296-9) contains supplementary
material, which is available to authorized users. colony-forming units
AML Acute myelogenous leukemia
* Xiaofan Zhu MDS Myelodysplastic syndrome
xfzhu@ihcams.ac.cn CFU-E Erythroid colony-forming units
NIH National Institutes of Health
1
Division of Pediatric Blood Diseases Center, State Key Laboratory of WBC White blood cell
Experimental Hematology, National Clinical Research Center for
ALC Absolute lymphocyte count
Blood Diseases, Institute of Hematology & Blood Diseases Hospital,
Chinese Academy of Medical Sciences & Peking Union Medical CFU-GEMM Granulocyte erythrocyte macrophage
College, 288 Nanjing Road, Heping District, Tianjin 300020, China megakaryocyte colony-forming units
Ann Hematol

Introduction for SAA, and ANC was < 0.2 × 109/L. Complete history and
physical examination, routine cytogenetic analysis, mitomy-
Severe aplastic anemia (SAA) is a rare but heterogeneous cin C testing, comet assay, and if possible, next-generation
disorder. It is characterized by peripheral blood (PB) pancy- sequencing methods were performed to exclude inherited
topenia and bone marrow (BM) hypoplasia [1]. The underly- bone marrow failure syndrome. The patients were diagnosed
ing pathogenesis of acquired aplastic anemia (aAA) is be- as SAA when they finished BM investigation and met the
lieved to be immune-mediated damage of hematopoietic stem criteria. So, the time point for diagnosis and assessment of
cells (HSCs), with activated type 1 cytotoxic T cells implicat- severity mainly based on BM investigation. None of the 172
ed. For patients with SAA who lack a human leukocyte anti- patients had a HLA-matched sibling donor for HSCT at
gen (HLA)-compatible sibling donor for hematopoietic stem diagnosis.
cell transplantation (HSCT), the mainstay of treatment is im- The study protocol conformed to the ethical guidelines of
munosuppressive therapy (IST) [2]. the Declaration of Helsinki and was approved by the Ethics
The IST regimen usually involves antithymocyte globulin Committee and Institutional Review Board of Chinese
(ATG)/antilymphocyte globulin (ALG) and cyclosporine Academy of Medical Sciences and Peking Union Medical
(CsA). In many countries, horse ATG along with CsA has College. All legal guardians of pediatric patients provided
been regarded as the standard IST for SAA patients, with a informed consent before participation.
60–75% hematologic response and 85% 5-year overall surviv-
al (OS) rate [3–5]. However, horse ATG is unavailable in Treatment protocol
some countries including China. In China, both rabbit ATG
and porcine ALG are currently used as first-line treatment. IST consisted of rabbit ATG (Thymoglobuline®, Genzyme)
The outcome of 70 newly diagnosed SAA children treated intravenously 2.5–3.5 mg/kg/day for 5 consecutive days and
with porcine ALG and CsA at our institution was previously oral CsA 5 mg/kg/day for at least 2 years. For the prevention
reported [6]. Multiple studies indicated that the application of of serum sickness, intravenous methylprednisolone (1 mg/kg/
horse ATG yields better outcomes in comparison with rabbit day) was administered on days 1–5 followed by oral prednis-
ATG [7–9]. Hence, we retrospectively analyzed the clinical olone (1 mg/kg/day) for 2 weeks, and then tapered until with-
features, hematologic response, and long-term outcome of pe- drawal on day 28. The dosage of CsA was adjusted to achieve
diatric patients with newly diagnosed SAA who received rab- a trough whole-blood level at 100–200 ng/mL. Blood counts
bit ATG and CsA in our center last decade. We also explored and serum biochemistry were supervised accordingly.
the predictive factors for response and survival.
Response criteria and follow-up

Materials and methods Hematologic responses were assessed at 40 days, 3 months,


and 6 months after the initiation of IST. Complete response
Study population (CR) was defined as ANC ≥ 1.5 × 109/L, PLT ≥ 100 × 109/L,
and hemoglobin (HGB) normal for age and gender; nonre-
We conducted a single-center study at the Division of sponse (NR) was defined as persistence of severe disease;
Pediatric Blood Diseases Center, Institute of Hematology and partial response (PR) was defined as transfusion indepen-
and Blood Diseases Hospital (Tianjin, China). A total of 243 dent and not meeting the criteria for SAA [12, 13]. Relapse
patients younger than 18 years with newly diagnosed SAA was considered if the peripheral blood counts of patients de-
were admitted from January 2008 to April 2018. Among clined to a level requiring blood products transfusion, further
them, 70 patients were treated with porcine ALG and CsA, intensive immunotherapy, or HSCT [14, 15]. OS referred to
and one did not have sufficient data. Therefore, 172 cases with the time from initiation of IST until death from any cause or
complete clinical information, receiving rabbit ATG plus CsA last follow-up. Failure-free survival (FFS) was defined as the
as first-line treatment, and follow-up period over a minimum time from IST until the date of treatment failure, relapse, clon-
of 6 months were included in the retrospective study. The al evolution, or death, whichever came first. For non-
severity of aplastic anemia (AA) was classified by the pro- responders who did not receive HSCT and finally died or
posed criteria of Camitta et al. [10] and Bacigalupo et al. maintained alive in NR, 6 months were considered as FFS.
[11]. SAA was defined as BM cellularity < 25% (or 25–50%
with < 30% residual hematopoietic cells) and at least two pe- Culture assay of hematopoietic progenitor cells
ripheral cytopenias: (1) absolute neutrophil count (ANC) <
0.5 × 109/L, (2) platelet (PLT) count < 20 × 109/L, (3) absolute BM samples were treated with ammonium chloride solution
reticulocyte count (ARC) < 20 × 109/L. Very severe aplastic and separated using Ficoll-Paque® PLUS gradient centrifuga-
anemia (vSAA) was considered if patients satisfied the criteria tion. Then, we utilized CD34 + cell sorting method to enrich
Ann Hematol

precursor cells from BM samples. The prepared cells were Table 1 Baseline characteristics of 172 pediatric patients with SAA
diluted 1:1 with Iscove’s Modified Dulbecco’s Medium Characteristics Value
(IMDM) and 2% fetal bovine serum (FBS). Next, cell samples
were added to the MethoCult® medium H4434, shaken, and Median age at diagnosis, years (range) 5 (1–14)
inoculated into a petri dish. These cells were cultured for Gender, male (%) 74 (43%)
14 days in a 37 °C and 5% CO2 incubator. Finally, an inverted Median duration from diagnosis to IST, days (range) 31 (3–187)
microscope and a grid counting dish were used to count and PNH clone detected
judge the colony type. Minor, n (%) 58 (33.7%)
Major (> 10%), n (%) 0
Statistical analysis Disease severity
SAA, n (%) 82 (47.7%)
The statistical analysis was performed using the SPSS 18.0 vSAA, n (%) 90 (52.3%)
(SPSS, Chicago, IL, USA) and GraphPad Prism 7.04 software Peripheral blood count
(GraphPad, La Jolla, CA, USA). The chi-square test was used Median ANC, × 109/L (range) 0.18 (0–1.03)
for categorical variables. Multivariate logistic regression mod- Median HGB, g/L (range) 64 (32–117)
el was applied to evaluate which factors could predict re- Median PLT, × 109/L (range) 8 (0–41)
sponse. The OS and FFS rates were analyzed utilizing the Median ARC, × 109/L (range) 13.1 (1.5–62.5)
Kaplan-Meier method. Log-rank test was used for the com- Median MCV, fl (range) 83 (67.5–111.1)
parison between subgroups. Cox proportional hazard models Bone marrow morphology
were applied to assess risk factors for survival. P value < 0.05 Median marrow granulocyte, % (range) 8 (0–68)
was considered statistically significant. Median marrow erythrocyte, % (range) 8 (0–64)
Median marrow lymphocyte, % (range) 77 (19–99.5)
Hematopoietic progenitor cells culture of BM
Results Median CFU-E, /105BMMNC (range) 10 (0–88)
Median BFU-E, /105BMMNC (range) 2 (0–25)
Patient characteristics Median CFU-GM, /105BMMNC (range) 2 (0–31)

A total of 172 pediatric patients (74 males, 98 females) were SAA, severe aplastic anemia; IST, immunosuppressive therapy; PNH,
included in the study. The median age at diagnosis was 5 years paroxysmal nocturnal hemoglobinuria; vSAA, very severe aplastic ane-
mia; ANC, absolute neutrophil count; HGB, hemoglobin; PLT, platelet;
(range, 1–14). Ninety (52.3%) patients were diagnosed as
ARC, absolute reticulocyte count; MCV, mean corpuscular volume; BM,
vSAA. The median interval from diagnosis to initiation of bone marrow; CFU-E, erythroid colony-forming units; BMMNC, bone
IST was 31 days (range, 3–187). Flow cytometry of PB for marrow mononuclear cell; BFU-E, erythroid burst-forming units; CFU-
paroxysmal nocturnal hemoglobinuria (PNH) antigens GM, granulocyte-macrophage colony-forming units
(CD55, CD59) was performed for all patients at diagnosis
and a minor PNH clone (less than 10%) was found in 58
(33.7%) patients. The demographic and clinical features at and antibiotics. Additional 11 patients responded within 6–
baseline are shown in Table 1. 12 months after IST. At the time point of last follow-up, 4
new patients achieved delayed response, and 11 patients were
Hematologic response and response predictors maintained alive in NR. Therefore, the overall response rate
was 69.8% (120 responders) during the observation time.
Table 2 shows the hematologic response to IST at different To identify predictive factors for response to IST at
time points. Two (1.2%) children died within 3 months after 6 months, we compared the baseline variables between re-
treatment. Both of them were vSAA patients and died of seri- sponders and non-responders. Univariate analysis showed that
ous infection. At 40 days, 3 months, 6 months, and 12 months disease severity, baseline ARC, marrow granulocyte %, and
after IST, the overall response rates of all patients were 22.7%, marrow lymphocyte % were correlated with the response rate
45.3%, 61%, and 67.4%, respectively. The SAA group had a (Table 3). We further performed multivariate logistic regres-
significantly better response rate than the vSAA group at sion analysis which included the following baseline variables:
40 days (31.7 versus 14.4%, P = 0.007) and 6 months (72 gender, disease severity, ARC, marrow granulocyte %, mar-
versus 51.1%, P = 0.005). At 6 months post-IST, a total of row lymphocyte %, erythroid burst-forming units (BFU-E),
67 patients showed no response. Owing to financial and per- and granulocyte-macrophage colony-forming units (CFU-
sonal reasons, only 26 non-responders underwent HSCT, and GM). We confirmed that mild disease severity (P = 0.006)
the remaining non-responders received high-dose CsA and was a significant predictor of favorable response at 6 months
supportive care which mainly in the form of blood transfusion (Supplementary Table S1).
Ann Hematol

Table 2 Hematologic response to


IST at different time points Total (n = 172) SAA (n = 82) vSAA (n = 90) P value

Response at 40 days after IST


CR 1 (0.6%) 0 (0%) 1 (1.1%)
PR 38 (22.1%) 26 (31.7%) 12 (13.3%)
NR 133 (77.3%) 56 (68.3%) 77 (85.6%)
ORR (CR/PR) 39 (22.7%) 26 (31.7%) 13 (14.4%) 0.007*
Response at 3 months after IST
CR 13 (7.5%) 8 (9.7%) 5 (5.6%)
PR 65 (37.8%) 35 (42.7%) 30 (33.3%)
NR 94 (54.7%) 39 (47.6%) 55 (61.1%)
ORR (CR/PR) 78 (45.3%) 43 (52.4%) 35 (38.9%) 0.075
Response at 6 months after IST
CR 47 (27.3%) 29 (35.4%) 18 (20%)
PR 58 (33.7%) 30 (36.6%) 28 (31.1%)
NR 67 (39%) 23 (28%) 44 (48.9%)
ORR (CR/PR) 105 (61%) 59 (72%) 46 (51.1%) 0.005*
Response at 12 months after IST
CR 57 (33.1%) 38 (46.3%) 19 (21.1%)
PR 59 (34.3%) 23 (28.1%) 36 (40%)
NR 56 (32.6%) 21 (25.6%) 35 (38.9%)
ORR (CR/PR) 116 (67.4%) 61 (74.4%) 55 (61.1%) 0.063

IST, immunosuppressive therapy; SAA, severe aplastic anemia; vSAA, very severe aplastic anemia; CR, complete
response; PR, partial response; NR, nonresponse; ORR, overall response rate
*
Represents P value < 0.05

Long-term outcome and survival analysis The 5-year overall survival (OS) rate was 90.5% and the 5-
year FFS rate was 70.4% for all patients (Fig. 1a, b). Prognostic
For all patients, the follow-up endpoint was July 2019. The me- factors correlated with OS and FFS were evaluated in univariate
dian follow-up was 63 months (range, 1–135). A total of 16 analysis (Supplementary Table S2). Log-rank test indicated that
(9.3%) patients died, including 2 (1.2%) early deaths and 14 BFU-E (P = 0.042) and time interval before IST (P = 0.022)
(8.1%) late deaths. Among the 14 late deaths, 6 died of serious were associated with OS, while BFU-E (P = 0.025) and ery-
bacterial and fungal infection, 6 died of hemorrhage, 1 died of throid colony-forming units (CFU-E) (P = 0.045) showed influ-
secondary acute myelogenous leukemia (AML), and 1 died of ence on FFS. Patients with BFU-E > 2/105 bone marrow mono-
pulmonary fibrosis after HSCT. The median time of late deaths nuclear cell (BMMNC) or time interval before IST ≤ 30 days
was 10 months (range, 5–110). Relapse was observed in 7 of the had a significantly higher OS rate (Fig. 2a, b). Meanwhile, pa-
120 responders at a median of 49 months (range, 40–98) follow- tients with BFU-E > 2/105 BMMNC or CFU-E > 10/105
ing IST, and the cumulative incidence of relapse was 5.8%. Five BMMNC had a significantly higher FFS rate (Fig. 2c, d). Cox
of the relapsed children were treated with increasing doses of proportional hazard models were also used to evaluate risk fac-
CsA and the other two received HSCT. At the time point of last tors for OS and FFS. BFU-E > 2/105 BMMNC (P = 0.037) and
follow-up, 51 (42.5%) of the 120 patients with response contin- time interval before IST ≤ 30 days (P = 0.017) were independent
ued to receive CsA treatment. Two (1.2%) patients who had positive predictors for OS in multivariate analysis
normal chromosomes before IST suffered from clonal evolution (Supplementary Table S3). We also confirmed that BFU-E >
at the follow-up endpoint. Between them, the one who 2/105 BMMNC (P = 0.029) was the only favorable prognostic
progressed to myelodysplastic syndrome (MDS) underwent factor for FFS (Supplementary Table S4).
HSCT and was evaluated as CR at last follow-up while another
one who developed AML received chemotherapy and died dur-
ing the treatment. A total of 29 (16.9%) patients underwent Discussion
HSCT as salvage settings due to lack of response to IST (n =
26), relapse (n = 2), or clonal evolution (n = 1). Except one pa- IST with ATG/ALG and CsA is often regarded as the initial
tient died of pulmonary fibrosis at 1 year after HSCT, the remain- therapy for SAA, since most patients lack an available HLA-
ing 28 patients were all alive at the follow-up endpoint. matched sibling donor or are not suitable for HSCT. In China,
Ann Hematol

Table 3 Univariate analysis for


response to IST at 6 months after Factors Responders (n = 105) Non-responders (n = 67) χ2 value P value
treatment
Gender 3.385 0.066
Male 51 23
Female 54 44
Age 1.727 0.189
> 5 years 45 22
≤ 5 years 60 45
Time interval before IST 0.024 0.876
> 30 days 53 33
≤ 30 days 52 34
PNH clone 0.038 0.845
Positive 36 22
Negative 69 45
Disease severity 7.836 0.005*
SAA 59 23
vSAA 46 44
Baseline HGB 0.007 0.932
> 60 g/L 62 40
≤ 60 g/L 43 27
Baseline PLT 2.376 0.123
> 10 × 109/L 35 15
≤ 10 × 109/L 70 52
Baseline ARC 4.058 0.044*
> 10 × 109/L 68 33
≤ 10 × 109/L 37 34
Baseline MCV, fl 1.018 0.313
> 80 fl 78 45
≤ 80 fl 27 22
Baseline marrow granulocyte % 5.733 0.017*
> 10% 54 22
≤ 10% 51 45
Baseline marrow erythrocyte % 3.114 0.078
> 10% 52 24
≤ 10% 53 43
Baseline marrow lymphocyte % 6.598 0.010*
> 80% 39 38
≤ 80% 66 29
Baseline CFU-E 1.828 0.176
> 10/105BMMNC 42 20
≤ 10/105BMMNC 63 47
Baseline BFU-E 3.680 0.055
> 2/105BMMNC 45 19
≤ 2/105BMMNC 60 48
Baseline CFU-GM 3.281 0.070
> 2/105BMMNC 54 25
≤ 2/105BMMNC 51 42

IST, immunosuppressive therapy; PNH, paroxysmal nocturnal hemoglobinuria; SAA, severe aplastic anemia;
vSAA, very severe aplastic anemia; HGB, hemoglobin; PLT, platelet; ARC, absolute reticulocyte count; MCV,
mean corpuscular volume; CFU-E, erythroid colony-forming units; BMMNC, bone marrow mononuclear cell;
BFU-E, erythroid burst-forming units; CFU-GM, granulocyte and macrophage colony-forming units
*
Represents P value < 0.05

rabbit ATG was the only ATG formulation available and was Our study retrospectively analyzed a large cohort of 172
approved in 2003 as a first-line agent to treat SAA. A previous SAA children treated with rabbit ATG and CsA at a single
study showed an obvious difference in 6-year OS rate among center. In our work, the overall response rates at 3 months and
patients of different age receiving ATG and CsA: < 20 years (n = 6 months after IST were 45.3% and 61%, respectively. It was
31, 100%), 20–40 years (n = 51, 92%), 40–60 years (n = 51, comparable with the study of Jeong et al. [9] and Chen et al.
71%), and > 60 years (n = 59, 56%) [16]. This finding indicates [17] in which rabbit ATG along with CsA was used as first-
that the survival rates of children and adults who receive the same line therapy for childhood SAA. However, a recent study by
treatment differ significantly. So, the results of IST for adult and North American Pediatric Aplastic Anemia Consortium
pediatric patients with SAA should be analyzed separately. showed a higher response rate of 71.2% in subjects treated
Ann Hematol

(a)
with horse ATG and CsA [18]. Considering baseline clinical
and laboratory data may put a salient impact on the response to
IST, we further performed univariate and multivariate analy-
ses on the baseline factors between responders and non-re-
sponders. The pre-treatment variables involved in our study
were comprehensive, which not only covered basic informa-
tion, blood counts, and PNH clone detection, but also included
the findings of BM aspiration such as BM morphology and
hematopoietic progenitor cells culture.
Several studies showed a favorable outcome in patients (b)

with AA who were PNH clone positive [19–21]. In the work


of Tutelman et al. [19], 73% of pediatric AA patients had a
PNH clone positivity and a high proportion (94%) of them
achieved either CR or PR to IST. Meanwhile, the patients
who were PNH negative had a lower response rate of 33%.
In our pediatric cohort, 58 (33.7%) patients had a PNH clone
positivity at diagnosis. However, the PNH clone detected at

(c)
(a)

(d)

(b)

Fig. 2 a Overall survival (OS) of 172 children with severe aplastic anemia
(SAA) who received rabbit antithymocyte globulin (ATG) and cyclospor-
ine (CsA) categorized by erythroid burst-forming units (BFU-E). Five-year
OS rate was significantly higher in patients with BFU-E > 2/105 bone
marrow mononuclear cell (BMMNC), 96.6 versus 87.0% (P = 0.042). b
Overall survival (OS) of 172 children with severe aplastic anemia (SAA)
who received rabbit antithymocyte globulin (ATG) and cyclosporine
(CsA) categorized by time interval between diagnosis and IST. Five-year
OS rate was significantly higher in patients with time interval ≤ 30 days,
96.5 versus 83.8% (P = 0.022). c Failure-free survival (FFS) of 172 chil-
dren with severe aplastic anemia (SAA) who received rabbit antithymocyte
globulin (ATG) and cyclosporine (CsA) categorized by erythroid burst-
forming units (BFU-E). Five-year FFS rate was significantly higher in
patients with BFU-E > 2/105 bone marrow mononuclear cell (BMMNC),
Fig. 1 a Overall survival (OS) of 172 children with severe aplastic ane- 83.2 versus 63.8% (P = 0.025). d Failure-free survival (FFS) of 172 chil-
mia (SAA) who received rabbit antithymocyte globulin (ATG) and cy- dren with severe aplastic anemia (SAA) who received rabbit antithymocyte
closporine (CsA). Five-year OS rate was 90.5% for all patients. b Failure- globulin (ATG) and cyclosporine (CsA) categorized by erythroid colony-
free survival (FFS) of 172 children with severe aplastic anemia (SAA) forming units (CFU-E). Five-year FFS rate was significantly higher in
who received rabbit antithymocyte globulin (ATG) and cyclosporine patients with CFU-E > 10/10 5 bone marrow mononuclear cell
(CsA). Five-year FFS rate was 70.4% for all patients (BMMNC), 78.8 versus 65.6% (P = 0.045)
Ann Hematol

baseline was not correlated with an increased response rate at year OS rate was comparable with horse ATG cohort in the
6 months following IST. This outcome was consistent with study of North American Pediatric Aplastic Anemia
the work of Yoshida et al. [22] and the National Institutes of Consortium [18]. Regarding the association between prognos-
Health (NIH) group [23] which demonstrated the absence of tic factors and survival, we further performed univariate and
prognostic value with PNH clone. multivariate analyses. Higher absolute lymphocyte count
A previous multicenter study in Japan reported that gender, (ALC) and ARC were previously reported to have a signifi-
duration from diagnosis to treatment, and white blood cell cant connection to a higher survival rate [25] while in another
(WBC) count were confirmed as predictive factors for hema- study, only higher ARC was of prognostic value [27]. A mul-
tologic response [24]. Another Asian study also indicated that ticenter study in Europe and Asia reported that patients age,
male gender and shorter time interval before IST were inde- disease severity, and time interval between diagnosis and
pendent predictors of better response in childhood SAA [9]. In treatment were predictors of survival [29]. In our study, uni-
our study, male group displayed better response than female variate analysis showed that BFU-E and time interval before
group, although the difference was not of statistical signifi- IST were correlated with OS, while BFU-E and CFU-E
cance (P = 0.066). Besides, the response rates were not signif- showed influence on FFS. Our multivariate analysis showed
icantly different grouped by time interval before IST. that BFU-E > 2/105 BMMNC and time interval before IST ≤
In univariate analysis, we found that disease severity, base- 30 days were independent positive predictors for OS, mean-
line ARC, marrow granulocyte %, and marrow lymphocyte % while BFU-E > 2/105 BMMNC was the only favorable prog-
were significantly associated with the response rate. nostic factor for FFS. Hematopoietic progenitor cells are ex-
Moreover, we identified that disease severity was the only istent in the blood, as well as in the BM. Changes in the
predictive factor of response in multivariate analysis. As for numbers of hematopoietic progenitor cells reveal alteration
pediatric patients with SAA, baseline ARC and marrow gran- of pluripotent stem cells. In AA, the most consistent finding
ulocyte % can reflect the status of hematopoiesis. Thus, a is the very low numbers of hematopoietic progenitor cells
higher pre-treatment ARC and marrow granulocyte % may including CFU-GM, BFU-E, CFU-E, and granulocyte eryth-
show better residual hematopoiesis function and the ability rocyte macrophage megakaryocyte colony-forming units
to support blood cell production following IST. The NIH (CFU-GEMM) [34]. In the present study, the median numbers
group [25] showed that baseline ARC was the predominant of CFU-E, BFU-E, and CFU-GM were much decreased in the
factor for predicting the response at 6 months. When it comes BM of children with SAA. Besides, BFU-E > 2/105 BMMNC
to the correlation between response and disease severity, con- was verified as a good predictor for both OS and FFS. The
troversial results have been reported. A prospective multicen- study of Rosenfeld et al. [35] had described in adults levels of
ter trial in Europe reported favorable response rates in pediat- residual hematopoiesis as a predictor of response to immuno-
ric patients with vSAA compared with SAA [26], but several suppressive therapy, which was in line with our work. The 5-
studies indicated the opposite results [11, 27–29]. Our data year OS rates of children with intervals more than 30 and of
were consistent with those published studies which demon- 30 days or less were 83.8% and 96.5%, respectively. Yoshida
strated that SAA patients had a better response than their et al. [24] reported that AA children with long-standing dis-
vSAA counterparts. In our work, the overall response rates ease may experience irreversible destruction to hematopoietic
of SAA children and vSAA children were 72% and 51.1% progenitor cells or stromal components as time passed by.
at 6 months post-IST, respectively. Therefore, we suggested the administration of IST as soon
The long-term survival of responders is mainly influenced as possible in all pediatric patients with SAA who lack a
by disease relapse and clonal evolution to MDS or AML. matched sibling donor.
Besides, the occurrence of relapse and clonal evolution are Although our study is limited by its retrospective design
associated with CsA tapering strategy [30] and residual hema- such as potential confounding factors and data availability, it
topoietic cell biology [31]. In our work, the median follow-up supplies a contemporary analysis of the diagnostic assessment
period was 63 months, and the actuarial risk of relapse was and treatment outcome for childhood SAA. Considering that
only 5.8%, which was much lower than previous studies [32, horse ATG is unavailable in China, we cannot directly com-
33]. The implementation of slow CsA tapering schedule and pare the efficacy of rabbit ATG and horse ATG.
continuous CsA treatment in nearly half of the responders at With the application of rabbit ATG, the result of our study
last follow-up may account for the low relapse rate. The pres- is comparable with those reported results using horse ATG as
ent study reported that 2 (1.2%) patients experienced clonal first-line treatment.
evolution, and the incidence of evolution was similar to rabbit In conclusion, our study has demonstrated that rabbit ATG
ATG cohort in the study of Jeong et al. [9]. along with CsA is an effective first-line therapy for pediatric
In our pediatric cohort treated with rabbit ATG and CsA, patients with SAA, with a hematologic response rate of 61%
the 5-year OS and FFS rates were 90.5% and 70.4%, which at 6 months following IST, a 5-year OS rate of 90.5%, and a 5-
were obviously higher than previous report [32]. And the 5- year FFS rate of 72.1%. Mild disease severity was the only
Ann Hematol

predictor of favorable response to IST. Independent prognos- 6. Liu LP, Chen XJ, Yang WY, Yi MH, Zhou K, Ruan M, Liu F,
Chen X, Chang LX, Liu TF, Zhang L, Zou Y, Chen YM, Zhang
tic factors for OS were BFU-E and time interval before IST,
FK, Zhu XF, Guo Y (2019) Predicting response to porcine
while the only predictor for FFS was BFU-E. antilymphocyte globulin plus cyclosporine A in children with ac-
quired severe aplastic anemia. Pediatr Res 86(3):360–364. https://
Acknowledgments The authors would like to sincerely thank the patients doi.org/10.1038/s41390-019-0437-1
that participated in the follow-up and the support of AiYou Foundation. 7. Scheinberg P, Nunez O, Weinstein B, Scheinberg P, Biancotto A,
Wu CO, Young NS (2011) Horse versus rabbit antithymocyte glob-
Authors’ contributions Yang Lan designed the study, collected and ana- ulin in acquired aplastic anemia. N Engl J Med 365(5):430–438.
lyzed the data, and wrote the article; Lixian Chang designed the study, https://doi.org/10.1056/NEJMoa1103975
collected the data, and reviewed the article; Meihui Yi, Yuli Cai, and Jing 8. Atta EH, Dias DS, Marra VL, de Azevedo AM (2010) Comparison
Feng collected and analyzed the data; Chao Liu, Xiaoyan Chen, Aoli between horse and rabbit antithymocyte globulin as first-line treat-
Zhang, and Lipeng Liu analyzed the data; Yuanyuan Ren, Shuchun ment for patients with severe aplastic anemia: a single-center retro-
Wang, Ye Guo, and Jingliao Zhang collected the data; Xiaofan Zhu spective study. Ann Hematol 89(9):851–859. https://doi.org/10.
designed the study and reviewed the article. All authors read and ap- 1007/s00277-010-0944-y
proved the final manuscript. 9. Jeong DC, Chung NG, Cho B, Zou Y, Ruan M, Takahashi Y,
Muramatsu H, Ohara A, Kosaka Y, Yang W, Kim HK, Zhu X,
Kojima S (2014) Long-term outcome after immunosuppressive
Funding This study was funded by the National Key Research and
therapy with horse or rabbit antithymocyte globulin and cyclospor-
Development Program of China (grant No.2016YFC0901503).
ine for severe aplastic anemia in children. Haematologica 99(4):
664–671. https://doi.org/10.3324/haematol.2013.089268
Data availability The datasets generated during and/or analyzed during 10. Camitta BM, Rappeport JM, Parkman R, Nathan DG (1975)
the current study are available from the corresponding author on reason- Selection of patients for bone marrow transplantation in severe
able request. aplastic anemia. Blood 45(3):355–363
11. Bacigalupo A, Hows J, Gluckman E, Nissen C, Marsh J, Van Lint
Compliance with ethical standards MT, Congiu M, De Planque MM, Ernst P, McCann S et al (1988)
Bone marrow transplantation (BMT) versus immunosuppression
for the treatment of severe aplastic anaemia (SAA): a report of the
Conflict of interest The authors declare that they have no conflict of EBMT SAA working party. Br J Haematol 70(2):177–182. https://
interest. doi.org/10.1111/j.1365-2141.1988.tb02460.x
12. Subspecialty Group of H, Society of P, Chinese Medical
Ethics approval This study was performed in line with the principles of Association The Editorial B, Chinese Journal of P (2014)
the Declaration of Helsinki. Approval was obtained from the Ethics Recommendations for diagnosis and treatment of acquired aplastic
Committee and Institutional Review Board of Chinese Academy of anemia in children. Zhonghua Er Ke Za Zhi 52(2):103–106
Medical Sciences and Peking Union Medical College. 13. Camitta BM (2000) What is the definition of cure for aplastic ane-
mia? Acta Haematol 103(1):16–18. https://doi.org/10.1159/
Consent to participate Informed consent was obtained from legal 000040999
guardians. 14. Scheinberg P, Wu CO, Nunez O, Scheinberg P, Boss C, Sloand
EM, Young NS (2009) Treatment of severe aplastic anemia with a
Consent for publication Patients signed informed consent regarding combination of horse antithymocyte globulin and cyclosporine,
publishing their data. with or without sirolimus: a prospective randomized study.
Haematologica 94(3):348–354. https://doi.org/10.3324/haematol.
13829
Code availability Not applicable.
15. Nishikawa E, Yagasaki H, Hama A, Yabe H, Ohara A, Kosaka Y,
Kudo K, Kobayashi R, Ohga S, Morimoto A, Watanabe KI,
Yoshida N, Muramatsu H, Takahashi Y, Kojima S (2017) Long-
References term outcomes of 95 children with moderate aplastic anemia treated
with horse antithymocyte globulin and cyclosporine. Pediatr Blood
Cancer 64(5). https://doi.org/10.1002/pbc.26305
1. Scheinberg P, Young NS (2012) How I treat acquired aplastic ane- 16. Tichelli A, Schrezenmeier H, Socie G, Marsh J, Bacigalupo A,
mia. Blood 120(6):1185–1196. https://doi.org/10.1182/blood- Duhrsen U, Franzke A, Hallek M, Thiel E, Wilhelm M,
2011-12-274019 Hochsmann B, Barrois A, Champion K, Passweg JR (2011) A
2. Young NS, Calado RT, Scheinberg P (2006) Current concepts in randomized controlled study in patients with newly diagnosed se-
the pathophysiology and treatment of aplastic anemia. Blood vere aplastic anemia receiving antithymocyte globulin (ATG), cy-
108(8):2509–2519. https://doi.org/10.1182/blood-2006-03-010777 closporine, with or without G-CSF: a study of the SAA Working
3. Frickhofen N, Heimpel H, Kaltwasser JP, Schrezenmeier H (2003) Party of the European Group for Blood and Marrow
Antithymocyte globulin with or without cyclosporin A: 11-year Transplantation. Blood 117(17):4434–4441. https://doi.org/10.
follow-up of a randomized trial comparing treatments of aplastic 1182/blood-2010-08-304071
anemia. Blood 101(4):1236–1242. https://doi.org/10.1182/blood- 17. Chen C, Xue HM, Xu HG, Li Y, Huang K, Zhou DH, Guo HX,
2002-04-1134 Fang JP, Huang SL (2012) Rabbit-antithymocyte globulin com-
4. Peslak SA, Olson T, Babushok DV (2017) Diagnosis and treatment bined with cyclosporin A as a first-line therapy: improved, effec-
of aplastic anemia. Curr Treat Options in Oncol 18(12):70. https:// tive, and safe for children with acquired severe aplastic anemia. J
doi.org/10.1007/s11864-017-0511-z Cancer Res Clin Oncol 138(7):1105–1111. https://doi.org/10.1007/
5. Luzzatto L, Risitano AM (2018) Advances in understanding the s00432-012-1184-4
pathogenesis of acquired aplastic anaemia. Br J Haematol 182(6): 18. Rogers ZR, Nakano TA, Olson TS, Bertuch AA, Wang W, Gillio
758–776. https://doi.org/10.1111/bjh.15443 A, Coates TD, Chawla A, Castillo P, Kurre P, Gamper C, Bennett
Ann Hematol

CM, Joshi S, Geddis AE, Boklan J, Nalepa G, Rothman JA, Huang 27. Chang MH, Kim KH, Kim HS, Jun HJ, Kim DH, Jang JH, Kim K,
JN, Kupfer GM, Cada M, Glader B, Walkovich KJ, Thompson AA, Jung CW (2010) Predictors of response to immunosuppressive ther-
Hanna R, Vlachos A, Malsch M, Weller EA, Williams DA, apy with antithymocyte globulin and cyclosporine and prognostic
Shimamura A (2019) Immunosuppressive therapy for pediatric factors for survival in patients with severe aplastic anemia. Eur J
aplastic anemia: a North American Pediatric Aplastic Anemia Haematol 84(2):154–159. https://doi.org/10.1111/j.1600-0609.
Consortium study. Haematologica 104(10):1974–1983. https:// 2009.01378.x
doi.org/10.3324/haematol.2018.206540 28. Marsh JC, Hows JM, Bryett KA, Al-Hashimi S, Fairhead SM,
19. Tutelman PR, Aubert G, Milner RA, Dalal BI, Schultz KR, Deyell Gordon-Smith EC (1987) Survival after antilymphocyte globulin
RJ (2014) Paroxysmal nocturnal haemoglobinuria phenotype cells therapy for aplastic anemia depends on disease severity. Blood
and leucocyte subset telomere length in childhood acquired aplastic 70(4):1046–1052
anaemia. Br J Haematol 164(5):717–721. https://doi.org/10.1111/ 29. Bacigalupo A, Oneto R, Schrezenmeier H, Hochsmann B, Dufour
bjh.12656 C, Kojima S, Zhu X, Chen X, Issaragrisil S, Chuncharunee S, Jeong
20. Kulagin A, Lisukov I, Ivanova M, Golubovskaya I, Kruchkova I, DC, Giammarco S, Van Lint MT, Zheng Y, Vallejo C (2018) First
Bondarenko S, Vavilov V, Stancheva N, Babenko E, Sipol A, line treatment of aplastic anemia with thymoglobuline in Europe
Pronkina N, Kozlov V, Afanasyev B (2014) Prognostic value of and Asia: outcome of 955 patients treated 2001-2012. Am J
paroxysmal nocturnal haemoglobinuria clone presence in aplastic Hematol 93(5):643–648. https://doi.org/10.1002/ajh.25081
anaemia patients treated with combined immunosuppression: re- 30. Saracco P, Quarello P, Iori AP, Zecca M, Longoni D, Svahn J,
sults of two-Centre prospective study. Br J Haematol 164(4):546– Varotto S, Del Vecchio GC, Dufour C, Ramenghi U, Bacigalupo
554. https://doi.org/10.1111/bjh.12661 A, Locasciulli A (2008) Cyclosporin A response and dependence in
21. Scheinberg P, Marte M, Nunez O, Young NS (2010) Paroxysmal children with acquired aplastic anaemia: a multicentre retrospective
nocturnal hemoglobinuria clones in severe aplastic anemia patients study with long-term observation follow-up. Br J Haematol 140(2):
treated with horse anti-thymocyte globulin plus cyclosporine. 197–205. https://doi.org/10.1111/j.1365-2141.2007.06903.x
Haematologica 95(7):1075–1080. https://doi.org/10.3324/ 31. Calado RT, Cooper JN, Padilla-Nash HM, Sloand EM, Wu CO,
haematol.2009.017889 Scheinberg P, Ried T, Young NS (2012) Short telomeres result in
22. Yoshida N, Yagasaki H, Takahashi Y, Yamamoto T, Liang J, chromosomal instability in hematopoietic cells and precede malig-
Wang Y, Tanaka M, Hama A, Nishio N, Kobayashi R, Hotta N, nant evolution in human aplastic anemia. Leukemia 26(4):700–707.
Asami K, Kikuta A, Fukushima T, Hirano N, Kojima S (2008) https://doi.org/10.1038/leu.2011.272
Clinical impact of HLA-DR15, a minor population of paroxysmal 32. Jeong D-C, Chung NG, Lee JW, P-s J, Cho B, Kim H-K (2011)
nocturnal haemoglobinuria-type cells, and an aplastic anaemia- Long-term outcome of immunosuppressive therapy with rabbit
associated autoantibody in children with acquired aplastic anaemia. antithymocyte globulin (rATG) for childhood severe aplastic ane-
Br J Haematol 142(3):427–435. https://doi.org/10.1111/j.1365- mia for 15 years. Blood 118(21):1346–1346. https://doi.org/10.
2141.2008.07182.x 1182/blood.V118.21.1346.1346
23. Bacigalupo A (2017) How I treat acquired aplastic anemia. Blood 33. Jain R, Trehan A, Bansal D, Varma N (2019) Aplastic anemia in
129(11):1428–1436. https://doi.org/10.1182/blood-2016-08- children: how good is immunosuppressive therapy? Pediatr
693481 Hematol Oncol 36(4):211–221. https://doi.org/10.1080/08880018.
24. Yoshida N, Yagasaki H, Hama A, Takahashi Y, Kosaka Y, 2019.1621970
Kobayashi R, Yabe H, Kaneko T, Tsuchida M, Ohara A, 34. Issaragrisil S, Up Y, Yimyam M, Pakdeesuwan K, Khuhapinant A,
Nakahata T, Kojima S (2011) Predicting response to immunosup- Muangsup W, Pattanapanyasat K (1998) Hematopoietic progenitor
pressive therapy in childhood aplastic anemia. Haematologica cells in the blood and bone marrow in various hematologic disor-
96(5):771–774. https://doi.org/10.3324/haematol.2010.032805 ders. Stem Cells 16(Suppl 1):123–128. https://doi.org/10.1002/
25. Scheinberg P, Wu CO, Nunez O, Young NS (2009) Predicting stem.5530160815
response to immunosuppressive therapy and survival in severe 35. Rosenfeld S, Follmann D, Nunez O, Young NS (2003)
aplastic anaemia. Br J Haematol 144(2):206–216. https://doi.org/ Antithymocyte globulin and cyclosporine for severe aplastic ane-
10.1111/j.1365-2141.2008.07450.x mia: association between hematologic response and long-term out-
26. Fuhrer M, Rampf U, Baumann I, Faldum A, Niemeyer C, Janka- come. Jama 289(9):1130–1135. https://doi.org/10.1001/jama.289.
Schaub G, Friedrich W, Ebell W, Borkhardt A, Bender-Goetze C 9.1130
(2005) Immunosuppressive therapy for aplastic anemia in children:
a more severe disease predicts better survival. Blood 106(6):2102– Publisher’s note Springer Nature remains neutral with regard to jurisdic-
2104. https://doi.org/10.1182/blood-2005-03-0874 tional claims in published maps and institutional affiliations.

You might also like