You are on page 1of 9

Hematology

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/yhem20

Hyperleukocytosis predicts inferior clinical


outcome in pediatric acute myeloid leukemia

Lu-Hong Xu , Jing-Wen Wang , Yin Wang & Feng-Ying Yang

To cite this article: Lu-Hong Xu , Jing-Wen Wang , Yin Wang & Feng-Ying Yang (2020)
Hyperleukocytosis predicts inferior clinical outcome in pediatric acute myeloid leukemia,
Hematology, 25:1, 507-514, DOI: 10.1080/16078454.2020.1859169

To link to this article: https://doi.org/10.1080/16078454.2020.1859169

© 2020 The Author(s). Published by Informa


UK Limited, trading as Taylor & Francis
Group

View supplementary material

Published online: 15 Dec 2020.

Submit your article to this journal

Article views: 264

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=yhem20
HEMATOLOGY
2020, VOL. 25, NO. 1, 507–514
https://doi.org/10.1080/16078454.2020.1859169

Hyperleukocytosis predicts inferior clinical outcome in pediatric acute myeloid


leukemia
Lu-Hong Xu , Jing-Wen Wang, Yin Wang and Feng-Ying Yang
Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Pediatrics, Sun Yat-Sen
Memorial Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China

ABSTRACT KEYWORDS
Objectives: Hyperleukocytosis (HL) is a laboratory abnormality commonly presented in Acute myeloid leukemia;
patients with acute myeloid leukemia (AML). However, large cohort studies on the clinical hyperleukocytosis; pediatric
significance of HL in pediatric AML are paucity. Moreover, the effect of stem cell patients; clinical outcome
transplantation in HL patients remains unknown.
Methods: The clinical profiles of 885 pediatric patients with AML were downloaded from the
TARGET dataset. HL was defined as an initial peripheral WBC count of ≥ 100 ×109/L. We
analyzed the prevalence, clinical profile and prognosis of HL in these patients.
Results: The frequency of HL among all the pediatric AML was 22.6%. FMS-like tyrosine kinase
3/internal tandem duplication (FLT3/ITD) mutation and gene fusion of NUP98/NSD1 occurred
with higher incidence in HL patients. Overall, HL was associated with a low induction complete
remission rate, and high risk of induction death. Moreover, HL predicted a significantly inferior
5-year event-free survival (EFS) (P < 0.001) and a trend of inferior 5-year overall survival (OS) (P
= 0.059). However, compared with chemotherapy, stem cell transplantation had no significant
effect on the survival of HL patients in terms of 5-year leukemia-free survival (P = 0.449) or OS
(P = 0.447). Multivariate analysis revealed that HL was an independent prognosis factor for EFS
(Hazard ratio:1.352, P = 0.013) but not for OS (Hazard ratio:1.225, P = 0.170) in pediatric AML.
Conclusion: HL might predict inferior clinical outcome in pediatric AML. SCT is an effective
therapy for AML, but it may have no better effect on the survival of patients with HL,
compared to chemotherapy.

1. Introduction in the 1980s. Recently, routine clinical care have been


incorporated with more advanced supportive care,
Acute myeloid leukemia (AML) is a heterogeneous
such as prompt transfusion of platelets, leukapheresis,
disease that accounts for about 20% of all leukemias
hydroxyurea, urate oxidase and hemodialysis [12, 13].
in children [1, 2]. Hyperleukocytosis (HL) is a laboratory
Thus, it is necessary to investigate the clinical signifi-
abnormality, commonly defined by a white blood cell
cance of HL in pediatric AML. Furthermore, the role
count of ≥100 ×109/L. HL is presented in 5% to 20%
of stem cell transplantation (SCT) in pediatric AML
of patients with AML [3]. Two main pathogenetic
patients with HL remains unclear. In this study, we
factors were reported to be responsible for the devel-
evaluated the prevalence, clinical profile, and progno-
opment of HL: first, a rapid blast proliferation leading
sis of HL in a cohort of 885 pediatric patients with AML.
to a high leukemic tumor burden; second, disruption
Our results show that HL predicts inferior clinical
in normal hematopoietic cell adhesion leading to a
outcome in pediatric AML. However, SCT does not sig-
reduced affinity to the bone marrow [3, 4]. Thus, HL
nificantly benefit HL patients.
may result in 3 main complications: leukostasis,
tumor lysis syndrome and disseminated intravascular
coagulation. Pulmonary and central nervous system 2. Methods
injuries are the major issues associated with HL
2.1. Data Sources
patients [5, 6]. Many adult studies showed that HL pre-
dicted poor prognosis [7–10]. However, studies on the Data on patients with complete clinical data from the
clinical significance of HL in pediatric AML in a large therapeutically applicable research to generate
cohort are paucity. effective treatment (TARGET) dataset were downloaded
Bunin et al. [11] found that pediatric AML with HL (https://ocg.cancer.gov/programs/target/data-matrix).
was associated with higher frequency of early death In total, 885 pediatric patients younger than 18 years

CONTACT Lu-Hong Xu xulvhong@126.com


Supplemental data for this article can be accessed https://doi.org/10.1080/16078454.2020.1859169.
© 2020 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits unrest-
ricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
508 L.-H. XU ET AL.

old with AML were included in our study. Year of diag- higher than that in non-HL group (22.0% vs 12.0%, P
nosis ranged from 1996 to 2010. Year of last follow up = 0.001). Notably, the FMS-like tyrosine kinase 3/
ranged from 1997 to 2015. The diagnosis of AML as internal tandem duplication (FLT3/ITD) mutation was
well as risk group stratification were done according substantially higher in HL group (26.5%) compared
to the Children’s Oncology Group (COG) guidelines with non-HL group (13.9%) (P < 0.001). However, no
[14]. Subtype classifications of AML were assigned significant difference was found in the median of
according to the French–American–British (FAB) FLT3/ITD allelic ratio and nucleophosmin (NPM1)
classifications. Treatment protocols for AML included mutations between HL group and non-HL group. In
AAML03P1, AAML0531 and CCG-2961. All these treat- addition, SCT and the treatment protocols for pediatric
ment protocols consisted of a remission induction AML were equally distributed between the two groups.
phase followed by an intensification phase. Stem cell Though there was no significant association
transplantation (SCT) was considered for patients in between HL and cytogenetic status, we found distinc-
the first complete remission. HL was defined as an tively different distribution of abnormal cytogenetics
initial peripheral WBC count of 100 ×109/L or over. in the two groups. As shown in Table S1, HL occurred
more frequently in patients with inv(16) (P < 0.001) and
MLL (P = 0.046), but less frequently in patients with t
2.2. Statistical analysis (8;21) (P = 0.001) and del 9q (P = 0.001). We further ana-
The clinical and biological characteristics of patients lyzed gene fusion data available in 429 cases. As shown
were summarized by descriptive statistics. To in Table S2, HL patients had significantly higher inci-
compare categorical variables, we used χ2 analysis dences of CBFB-MYH11 (P = 0.005) and NUP98-NSD1
and Fisher’s exact test in case of small numbers. The (P < 0.001), but lower incidence of RUNX1-RUNX1T1
nonparametric Mann–Whitney U-test was applied for (P < 0.001) than those without HL.
continuous variables. To assess outcome, the following
parameters were used: complete remission (CR, defined 3.2. Clinical outcomes
as a normocellular BM containing fewer than 5% blasts)
rate, event-free survival (EFS, defined as time between We examined the CR rate in the pediatric patients with
diagnosis and first event, including induction failure, AML. At the end of the first course of therapy, 142
relapse or death of any cause), overall survival (OS, (71.4%) of the 199 patients with HL achieved a CR com-
defined as time between diagnosis and death from pared with 528 (78.1%) of 794 patients without HL (P =
any cause), leukemia-free survival (LFS, defined as survi- 0.048). Moreover, the death rate at the end of first
val without leukemia relapse or progression). EFS, OS course of therapy was significantly correlated with HL
and LFS were estimated by the Kaplan–Meier method, (4.0% vs 1.2%, P = 0.015). At the end of the second
and compared using the log-rank test. Prognostic course of therapy, low CR rate (P < 0.001) and high
factors were examined by multivariate Cox regression death rate (P = 0.032) were found in HL group.
analysis. Hazard ratios (HR) were presented with 95% Next, we analyzed survival data in all the 885 pedi-
confidence intervals. A two-sided P-value < 0.05 was atric patients. The median follow-up time for the survi-
considered statistically significant for all statistical ana- vors was 5.62 years (5.90 years for HL patients and 5.58
lyses. The data were analyzed with the Statistical years for non-HL patients, respectively, P = 0.873). As
Package for the Social Sciences (SPSS®) version, 24.0 shown in Figure 1(A), HL patients had a markedly
(IBM Corporation, Armonk, NY, U.S.A.). worse 5-year EFS (37.4 ± 3.5%) compared with non-
HL patients (49.7 ± 1.9%; P < 0.001). HL patients also
had a trend of worse 5-year OS (57.4 ± 3.6%) compared
3. Results with non-HL patients (64.3 ± 1.9%) (P = 0.059) (Figure
1B). In the subgroup of normal cytogenetics, HL
3.1. Patient characteristics
patients showed a significantly lower 5-year EFS
Overall, among 885 pediatric AML patients, 200 (36.6 ± 7.5% vs 50.5 ± 4.1%, P = 0.010) and a trend
(22.6%) had HL at diagnosis. As shown in Table 1, no towards reduced 5-year OS (54.2 ± 8.0% vs 64.6 ±
correlation was found in median age between HL 3.9%, P = 0.076) (Figure 1C, D). In the subgroup of
group and non-HL group. However, the age under abnormal cytogenetics, HL was associated with poor
one year was associated with an increased risk of HL. clinical outcome in term of EFS (P = 0.007), but not
The median of WBC count was 182.1×109/L in HL OS (P = 0.403) (Figure 1E, F).
group and 19.6 ×109/L in non-HL group. Moreover,
patients with HL had higher percentage of blasts in
3.3. Stratification analysis of FLT3/ITD and
peripheral blood (P < 0.001) and in bone marrow (P <
NUP98-NSD1
0.001) compared with those in patients without HL.
The FAB subtypes in HL group were mainly M1, M4 We further investigated the prognostic impact of FLT3/
and M5. The percentage of high risk in HL group was ITD and NUP98-NSD1 in AML patients by stratification
HEMATOLOGY 509

Table 1. Characteristics of study population according to HL status.


All patients Hyperleukocytosis (WBC ≥ 100 ×109/L) No hyperleukocytosis (WBC < 100 ×109/L) P-value
Number (%) 885 200 (22.6%) 685 (77.4%)
Age, median (year) 9.6 8.9 9.7 0.481
Age<1 year, n (%) 81 (9.2%) 34 (17%) 47 (6.9%) <0.001
1≤Age<10 years, n (%) 375 (42.4%) 72 (36%) 303 (44.2%) 0.038
10≤Age<18 years, n (%) 429 (48.5%) 94 (47%) 335 (48.9%) 0.635
Gender (% female) 47.80% 47.50% 47.90% 0.924
WBC, ×109/L, Median (range) 31.7 (0.2-610) 182.1 (100.7-610) 19.6 (0.2-99.3) <0.001
% PB blast, Median (range) 47 (0-99) 80 (0-99) 36 (0-99) <0.001
% BM blast, Median (range) 70 (0-100) 82 (0-100) 64.7 (0.4-100) <0.001
FAB classification: N (%) 0.869
M0 21 (2.9%) 6 (3.6%) 15 (2.7%) 0.596
M1 98 (13.4%) 31 (18.7%) 67 (11.9%) 0.025
M2 196 (26.9%) 20 (12.0%) 176 (31.3%) <0.001
M3 2 (0.3%) 0 (0%) 2 (0.4%) >0.999
M4 200 (27.4%) 61 (36.7%) 139 (24.7%) 0.002
M5 160 (21.9%) 46 (27.7%) 114 (20.2%) 0.041
M6 12 (1.6%) 0 (0%) 12 (2.1%) 0.078
M7 40 (5.5%) 2 (1.2%) 38 (6.7%) 0.006
Risk group: N (%) 0.017
Low risk 333 (38.9%) 67 (35.1%) 266 (40.1%) 0.213
Intermediate risk 400 (46.8%) 82 (42.9%) 318 (47.9%) 0.226
High risk 122 (14.3%) 42 (22.0%) 80 (12.0%) 0.001
FLT3/ITD <0.001
Positive, n (%) 148 (16.8%) 53 (26.5%) 95 (13.9%)
Negative, n (%) 734 (83.2%) 147 (73.5%) 587 (86.1%)
FLT3/ITD allelic ratio,
Median (range) 0.55 (0.03-9.50) 0.63 (0.05-9.50) 0.53 (0.03-5.19) 0.732
NPM1 mutation 0.226
Positive, n (%) 66 (7.6%) 11 (5.6%) 55 (8.2%)
Negative, n (%) 803 (92.4%) 186 (94.4%) 617 (91.8%)
Cytogenetic status 0.575
Normal (n, %) 198 (23.5%) 41 (21.9%) 157 (23.9%)
Abnormal (n, %) 646 (76.5%) 146 (78.1%) 500 (76.1%)
SCT in 1st CR 0.204
No (n, %) 676 (83.9%) 138 (80.7%) 538 (84.7%)
Yes (n, %) 130 (16.1%) 33 (19.3%) 97 (15.3%)
Protocol 0.202
AAML03P1 (n, %) 93 (10.5%) 26 (13%) 67 (9.8%) 0.192
AAML0531 (n, %) 737 (83.3%) 163 (81.5%) 574 (83.8%) 0.444
CCG-2961 (n, %) 55 (6.2%) 11 (5.5%) 44 (6.4%) 0.634
CR status at end of course 1 0.033
CR 670 (76.6%) 142 (71.4%) 528 (78.1%) 0.048
Not CR 189 (21.6%) 49 (24.6%) 140 (20.7%) 0.238
Death 16 (1.8%) 8 (4.0%) 8 (1.2%) 0.015
CR status at end of course 2 <0.001
CR 749 (87.2%) 148 (77.1%) 601 (90.1%) <0.001
Not CR 89 (10.4%) 35 (18.2%) 54 (8.1%) <0.001
Death 21 (2.4%) 9 (4.7%) 12 (1.8%) 0.032
Abbreviations: BM, bone marrow; CR, complete remission; FAB, French-American-British morphology classification; FLT3/ITD, FMS-like tyrosine kinase 3/
internal tandem duplication; NPM1, nucleophosmin; PB, peripheral blood; SCT, stem cell transplantation; WBC: white blood cell.

analysis. As shown in Figure S1, FLT3/ITD conferred in Figure 3. When restricted to no NUP98-NSD1
poor clinical outcomes in AML patients with decreased group, HL was correlated with worse 5-year EFS (40.6
5-year EFS (P < 0.001) and OS (P = 0.004). As shown by ± 4.6% HL vs 51.8 ± 2.6% non-HL, P = 0.018) but not
the survival curves of pediatric AML patients grouped 5-year OS (60.3 ± 4.7% HL vs 68.2 ± 2.4% non-HL, P =
based on HL and FLT3/ITD status (Figure 2), HL in 0.142). However, when restricted to NUP98-NSD1
FLT3/ITD negative patients was associated with poor group, HL showed both significantly worse 5-year
prognosis in 5-year EFS (40.7 ± 4.1% HL vs 51.7 ± EFS (11.8 ± 7.8% HL vs 40.3 ± 12.1% non-HL, P =
2.1% non-HL; P = 0.009). However, HL showed no 0.004) and OS (22.1 ± 10.9% HL vs 75.5 ± 10.7% non-
impact on 5-year OS (61.7 ± 4.1% HL vs 65.7 ± 2.0% HL, P = 0.002).
non-HL; P = 0.308) in FLT3/ITD negative patients. Simi-
larly, HL in FLT3/ITD positive patients also had negative
3.4. The association between HL and SCT
impact on 5-year EFS (P = 0.036) but not on 5-year OS
(P = 0.218). To evaluate the association of HL and outcome of SCT,
As shown in Figure S2, NUP98-NSD1 gene fusion patients with induction failure or death without remis-
conferred poor clinical outcomes in AML patients in sion were excluded from our analysis. As shown in
term of 5-year EFS (P < 0.001) and OS (P = 0.029). The Figure 4(A, B), when restricted to SCT, HL had no sig-
survival curves of pediatric AML patients according to nificant impact on SCT outcome in term of 5-year LFS
the combined HL and NUP98-NSD1 status are shown (51.5 ± 8.7% HL vs. 63.0 ± 5.0% non-HL, P = 0.255) or
510 L.-H. XU ET AL.

Figure 1. Survival curves of pediatric AML patients according to hyperleukocytosis (HL) status. (A) Probability of EFS for all
patients. (B) Probability of OS for all patients. (C) Probability of EFS for patients with normal cytogenetics. (D) Probability of
OS for patients with normal cytogenetics. (E) Probability of EFS for patients with abnormal cytogenetics. (F) Probability of OS
for patients with abnormal cytogenetics.

OS (59.4 ± 8.7% HL vs. 66.9 ± 4.9% non-HL, P = 0.582). to HL patients, compared with chemotherapy (no
In addition, we evaluated the impact of SCT on HL SCT), SCT had no significant impact on the clinical
patients. As shown in Figure 4(C, D), when restricted outcome in term of 5-year LFS (51.5 ± 8.7% SCT vs.
HEMATOLOGY 511

Figure 2. Survival curves of pediatric AML patients according to the combined hyperleukocytosis (HL) and FLT3/ITD status. (A)
Probability of EFS for AML patients. (B) Probability of OS for AML patients.

46.1 ± 4.5% non-SCT, P = 0.449) or OS (59.4 ± 8.7% SCT 4. Discussion


vs. 68.5 ± 4.3% non-SCT, P = 0.447).
The frequency of HL among the 885 pediatric AML
patients was 22.6%. Our study showed that pediatric
AML with HL was associated to the age ( < 1 year),
3.5. Multivariate analysis
FAB subtypes of M1, M4 and M5, FLT3/ITD, inv (16),
In the Cox model (Table 2), we analyzed HL status with MLL, gene fusion of CBFB-MYH11 and NUP98-NSD1.
other co-variables including age (utilizing 1 year as the However, no association was found between HL and
cutoff value), high risk, standard risk and SCT. High risk NPM1 mutations. These results are consistent with an
and standard risk were significantly associated with earlier pediatric study [15]. In another large cohort of
worse clinical outcomes. In contrast, SCT was significantly adult study, the frequency of HL among 3510 newly
associated with better clinical outcomes. Age affect diagnosed AML patients was only 10%. Moreover,
neither EFS nor OS. Importantly, we identified that HL adult AML with HL was associated with higher frequen-
was an independent factor for EFS but not OS in pediatric cies of monocytic subtype of AML, FLT3/ITD and NPM1
patients with AML. HL was a highly significant predictor mutations [3]. It has been reported that monocytic leu-
for inferior EFS (HR:1.352, 95% CI:1.066-1.715, P = 0.013), kemic cells have a larger volume and markedly
but not for OS (HR:1.225, 95% CI: 0.917-1.636, P = 0.170). increased lysozyme activities. There was a greater

Figure 3. Survival curves of pediatric AML patients according to the combined hyperleukocytosis (HL) and NUP98-NSD1 status. (A)
Probability of EFS for AML patients. (B) Probability of OS for AML patients.
512 L.-H. XU ET AL.

Figure 4. Survival curves of pediatric AML patients according to the combined hyperleukocytosis (HL) and stem cell transplan-
tation (SCT) status. (A) Probability of LFS for patients with SCT according to HL status. (B) Probability of OS for patients with
SCT according to HL status. (C) Probability of LFS for HL patients according to SCT status. (D) Probability of OS for HL patients
according to SCT status.

degree of cytoreduction in M4 and M5 than in other Studies have reported various significance of HL in
subtypes of AML after chemotherapy began, and treatment response and clinical outcome. In 1987,
massive tumor lysis caused coagulopathy and meta- Dutcher et al. [10] found that HL predicted high risk
bolic disturbance with electrolyte abnormalities [16]. of early death, low CR rate and survival rate. In the
database of the Alliance Leukemia study group, early
death in HL group was higher after 1 week and 30
Table 2. Multivariate analysis for EFS and OS in pediatric
days of chemotherapy compared with non-HL group,
patients with AML.
Outcome Variable Hazard ratio (95% CI) P-value
the 5-year OS and relapse-free survival rates were
EFS HL 1.352 (1.066-1.715) 0.013
lower in HL group [3]. However, other adult studies
Age > 1 year 0.959 (0.677-1.359) 0.815 showed that HL did not impact the clinical outcomes
High risk 3.247 (2.326-4.534) <0.001 in term of OS and disease-free survival rates [17, 18].
Standard risk 2.270 (1.789-2.879) <0.001
SCT 0.440 (0.321-0.604) <0.001 Furthermore, the clinical significance of HL on clinical
OS HL 1.225 (0.917-1.636) 0.17 outcome in pediatric patients with AML was shown
Age > 1 year 1.187 (0.776-1.816) 0.429
High risk 4.164 (2.795-6.204) <0.001 by Inaba et al. that pediatric patients with or without
Standard risk 2.971 (2.189-4.031) <0.001 HL had similar CR rates and OS rate, but those with
SCT 0.600 (0.424-0.849) 0.004
HL had a lower EFS rate [19].
Abbreviations: CI, confidence interval; EFS, event-free survival; FLT3/ITD,
FMS-like tyrosine kinase 3/internal tandem duplication; HL, hyperleuko- Our study showed that HL in pediatric patients was
cytosis; OS, overall survival; SCT, stem cell transplantation. associated with high risk of induction death,
HEMATOLOGY 513

significantly inferior 5-year EFS and a trend of inferior Data Availability


OS. Moreover, the inferior impact on EFS caused by
The datasets generated and/or analyzed during the
HL was found irrelevant of cytogenetics status. FLT3/
current study are available from the TARGET website:
ITD mutation and gene fusion of NUP98/NSD1 were
https://ocg.cancer.gov/programs/target/data-matrix
characterized as adverse prognostic factors in AML
[20, 21]. We found that HL was related to higher fre-
quencies of FLT3/ITD mutation and NUP98/NSD1. Acknowledgements
After stratifications studies, we found that HL had sig-
Thanks are due to TARGET researchers as well as all study
nificantly adverse impacts on 5-year EFS in spite of the
participants for public data. This work was supported by
status of FLT3/ITD and NUP98/NSD1. Multivariate the Natural Science Foundation of Guangdong Province,
analysis confirmed that HL was an independent prog- China (2018A030313680 to L.H. Xu) and Guangdong Basic
nosis factor for EFS in pediatric AML. Compared with and Applied Basic Research Foundation (2020A1515010312
the previous reports in 1980s [10, 11, 22], the rates of to L.H. Xu).
induction death and OS in HL patients were improved.
The difference may be due to improvements in thera-
Disclosure statement
peutic protocols and dedicated intensive supportive
care. No potential conflict of interest was reported by the author(s).
Recently, the management of HL involves suppor-
tive care and reducing the number of circulating leuke-
mic blast cells by induction chemotherapy, Funding
hydroxyurea, low-dose chemotherapy and leukapher- Thanks are due to TARGET researchers as well as all study
esis. However, it is worth noted that neither hydro- participants for public data. This work was supported by
the Natural Science Foundation of Guangdong Province,
xyurea nor leukapheresis showed benefit in clinical
China (2018A030313680 to L.H. Xu) and Guangdong Basic
outcomes in patients with symptomatic HL [23, 24]. and Applied Basic Research Foundation (2020A1515010312
The impact of HL on SCT remains controversial in to L.H. Xu).
patients with AML. In the registry of the acute leukemia
working party of the European Society for Blood and
Marrow Transplantation (EBMT), HL in 1275 adult trans- ORCID
planted AML patients was independently associated Lu-Hong Xu http://orcid.org/0000-0001-7898-7720
with increased relapse rate, decreased LFS, and inferior
OS after SCT. HL retained a significant prognostic role
for adult AML undergoing SCT [25]. However, Tien References
et al. found that HL patients had similar relapse rate, [1] Ribeiro RC. Advances in treatment of de-novo pediatric
OS and disease-free survival after SCT as non-HL acute myeloid leukemia. Curr Opin Oncol. 2014;26
patients [26]. Our results showed that HL had no sig- (6):656–662.
nificant impact on LFS and OS after SCT in pediatric [2] Creutzig U, van den Heuvel-Eibrink MM, Gibson B, et al.
AML. On the other hand, the effect of SCT in HL Diagnosis and management of acute myeloid leukemia
in children and adolescents: recommendations from an
patients remains unknown. Tien et al. [26]. found that international expert panel. Blood. 2012;120(16):3187–
SCT ameliorated the poor survival impact of HL, and 3205.
suggested that SCT might be beneficial for HL patients [3] Rollig C, Ehninger G. How I treat hyperleukocytosis in
in first CR. In our study, we found SCT was significantly acute myeloid leukemia. Blood. 2015;125(21):3246–
associated with better clinical outcomes for AML by 3252.
[4] Reuss-Borst MA, Klein G, Waller HD, et al. Differential
multivariate analysis. However, compared with che-
expression of adhesion molecules in acute leukemia.
motherapy, SCT had no better effect on the survival Leukemia. 1995;9(5):869–874.
of pediatric AML patients with HL in term of LFS or [5] Abla O, Angelini P, Di Giuseppe G, et al. Early compli-
OS. Thus, the role of SCT in AML patients with HL cations of hyperleukocytosis and leukapheresis in
requires further investigation. Childhood acute leukemias. J Pediatr Hematol Oncol.
Taken together, we analyzed the impact of HL in a 2016;38(2):111–117.
[6] Ali AM, Mirrakhimov AE, Abboud CN, et al. Leukostasis
large cohort of pediatric patients with AML. Our
in adult acute hyperleukocytic leukemia: a clinician’s
findings showed that pediatric AML with HL was digest. Hematol Oncol. 2016;34(2):69–78.
associated with the age (< 1 year), FAB M1, M4 and [7] Daver N, Kantarjian H, Marcucci G, et al. Clinical charac-
M5, as well as FLT3/ITD mutation and gene fusion of teristics and outcomes in patients with acute promyelo-
NUP98/NSD1. HL predicted a low induction CR rate cytic leukaemia and hyperleucocytosis. Br J Haematol.
2015;168(5):646–653.
and a significantly inferior EFS rate. SCT is an
[8] Greenwood MJ, Seftel MD, Richardson C, et al.
effective therapy for AML, but it may have no better Leukocyte count as a predictor of death during remis-
effect on the survival of patients with HL, compared sion induction in acute myeloid leukemia. Leuk
to chemotherapy. Lymphoma. 2006;47(7):1245–1252.
514 L.-H. XU ET AL.

[9] Oliveira LC, Romano LG, Prado-Junior BP, et al. Outcome cytosine arabinoside for de novo acute myeloid leuke-
of acute myeloid leukemia patients with hyperleukocyto- mia. Ann Hematol. 2000;79(10):533–542.
sis in Brazil. Med Oncol. 2010;27(4):1254–1259. [19] Inaba H, Fan Y, Pounds S, et al. Clinical and biologic fea-
[10] Dutcher JP, Schiffer CA, Wiernik PH. Hyperleukocytosis tures and treatment outcome of children with newly
in adult acute nonlymphocytic leukemia: impact on diagnosed acute myeloid leukemia and hyperleukocy-
remission rate and duration, and survival. J Clin Oncol. tosis. Cancer. 2008;113(3):522–529.
1987;5(9):1364–1372. [20] Hollink IH, van den Heuvel-Eibrink MM, Arentsen-Peters
[11] Bunin NJ, Pui CH. Differing complications of hyperleu- ST, et al. NUP98/NSD1 characterizes a novel poor prog-
kocytosis in children with acute lymphoblastic or nostic group in acute myeloid leukemia with a distinct
acute nonlymphoblastic leukemia. J Clin Oncol. 1985;3 HOX gene expression pattern. Blood. 2011;118
(12):1590–1595. (13):3645–3656.
[12] Villgran V, Agha M, Raptis A, et al. Leukapheresis in [21] Ostronoff F, Othus M, Gerbing RB, et al. NUP98/NSD1 and
patients newly diagnosed with acute myeloid leukemia. FLT3/ITD coexpression is more prevalent in younger
Transfus Apher Sci. 2016;55(2):216–220. AML patients and leads to induction failure: a COG and
[13] Badawy SM. The use of hydroxyurea and leukapheresis SWOG report. Blood. 2014;124(15):2400–2407.
in childhood acute leukemia with hyperleukocytosis. J [22] Ventura GJ, Hester JP, Smith TL, et al. Acute myeloblas-
Pediatr Hematol Oncol. 2016;38(5):407. tic leukemia with hyperleukocytosis: risk factors for
[14] Gamis AS, Alonzo TA, Meshinchi S, et al. Gemtuzumab early mortality in induction. Am J Hematol. 1988;27
ozogamicin in children and adolescents with de novo (1):34–37.
acute myeloid leukemia improves event-free survival [23] Oberoi S, Lehrnbecher T, Phillips B, et al. Leukapheresis
by reducing relapse risk: results from the randomized and low-dose chemotherapy do not reduce early mor-
phase III children’s Oncology group trial AAML0531. J tality in acute myeloid leukemia hyperleukocytosis: a
Clin Oncol. 2014;32(27):3021–3032. systematic review and meta-analysis. Leuk Res.
[15] Sung L, Aplenc R, Alonzo TA, et al. Predictors and short- 2014;38(4):460–468.
term outcomes of hyperleukocytosis in children with [24] Korkmaz S. The management of hyperleukocytosis in
acute myeloid leukemia: a report from the children’s 2017: do we still need leukapheresis? Transfus Apher
Oncology group. Haematologica. 2012;97(11):1770–1773. Sci. 2018;57(1):4–7.
[16] Hijiya N, Metzger ML, Pounds S, et al. Severe cardiopul- [25] Canaani J, Labopin M, Socie G, et al. Long term impact
monary complications consistent with systemic inflam- of hyperleukocytosis in newly diagnosed acute myeloid
matory response syndrome caused by leukemia cell leukemia patients undergoing allogeneic stem cell
lysis in childhood acute myelomonocytic or monocytic transplantation: an analysis from the acute leukemia
leukemia. Pediatr Blood Cancer. 2005;44(1):63–69. working party of the EBMT. Am J Hematol. 2017;92
[17] Marbello L, Ricci F, Nosari AM, et al. Outcome of hyper- (7):653–659.
leukocytic adult acute myeloid leukaemia: a single- [26] Tien FM, Hou HA, Tsai CH, et al. Hyperleukocytosis is
center retrospective study and review of literature. associated with distinct genetic alterations and is an
Leuk Res. 2008;32(8):1221–1227. independent poor-risk factor in de novo acute
[18] Flasshove M, Meusers P, Schutte J, et al. Long-term sur- myeloid leukemia patients. Eur J Haematol. 2018;101
vival after induction therapy with idarubicin and (1):86–94.

You might also like