To the editor:

DNA methyltransferase-inhibiting azanucleosides have become a mainstay of treatment of myeloid neoplasms in adult patients,1  with 5-azacytidine (azacitidine) being the agent in broadest clinical use. Although not curative, treatment with azacitidine achieves hematologic improvement and transfusion independency in many patients and prolongs survival.2,3  Even though most children with juvenile myelomonocytic leukemia (JMML) qualify for allogeneic hematopoietic stem cell transplantation (HSCT), the acceptable toxicity of low-dose azacitidine and its cytoreductive potential make it an attractive option as a bridging therapy before HSCT4  or as palliation after 1 or more transplants have failed. We previously published the first case report of a boy with JMML who achieved a complete clinical and genetic remission after 8 cycles of azacitidine.5  Here, we present a retrospective compilation of 12 children with JMML who received individual off-label treatment with azacitidine before HSCT (N = 9) or for relapsed disease (N = 3).

The children were treated at 11 centers November 2007-April 2012. Ten children were enrolled in the studies “98” or “2006” of the European Working Group of Myelodysplastic Syndromes in Childhood (EWOG-MDS; registered at www.clinicaltrials.gov as #NCT00047268 and #NCT00662090). Approval was obtained from the institutional review board of each institution, and parental informed consent was provided according to the Declaration of Helsinki. Two children were treated at centers not participating in EWOG-MDS studies. One case (D644) was published previously.5  The diagnosis of all children was centrally reviewed, and response was evaluated according to international consensus criteria.6 

The median age of the 12 patients was 4.8 years (range 0.4-9.1) (Table 1). A total of 64 azacitidine cycles were administered (median 5.5 cycles, range 1-11). Seven of 12 treatments consisted of 100 mg/m2 azacitidine per intravenous infusion on 5 consecutive days every 28 days. In the other 5 patients, the substance was administered over 5 to 7 days at a single dose of 50 to 100 mg/m2 per intravenous or subcutaneous route every 28 to 42 days.

Table 1

Response to azacitidine in children with JMML

Disease statusPatient identifierAge (y) and genderCytogeneticsMutational group*Azacitidine cyclesConcomitant treatmentResponse to azacitidineHSCT (total number)StatusFollow-up (mo)
Prior to first HSCT A062 1.1, male Del(5)(q13q33) NRAS None PR Yes (2) Alive with leukemia 38 
2-3 None PD 
Prior to first HSCT CH058 2.8, male Normal PTPN11 1-3 None SD Yes (2) Alive with leukemia 16 
4-7 None PR 
Prior to first HSCT D644§ 1.4, male −7 KRAS None PD Yes (1) Alive in remission 62 
2-4 None SD 
5-8 None CR|| 
Prior to first HSCT D706 5.9, male −7 PTPN11 None PD Yes (3) Alive in remission 66 
Prior to first HSCT D712 6.3, female Normal NRAS None PD Yes (1) Dead, TRM 
Prior to first HSCT D827 0.4, male Normal PTPN11 1-2 6MP Not evaluable Yes (1) Alive in remission 38 
3-7 None CR 
Prior to first HSCT I255 9.1, male Inv(2)(p23q13), −7 No mutation 1-4 AraC, 6MP Not evaluable Yes (1) Alive in remission 49 
Prior to first HSCT NS001 5.4, male −7 NRAS 1-3 None PR No Dead, progressive disease No data 
Prior to first HSCT NS002 0.8, male −7 PTPN11 None SD Yes (1) Dead, TRM 13 
2-3 None PR 
4-11 None CR# 
Relapse after HSCT** NL121 4.6, male Not done PTPN11 2-4 None SD Yes (3) Dead, third relapse 
1, 5-8 None PD 
Relapse after HSCT** SC108 5.0, male Normal†† NF1 1-2 None SD Yes (2) Dead, progressive disease 
3-4 None PR 
None PD 
Relapse after HSCT** SC156 5.3, male Normal†† PTPN11 2-4 None PR Yes (3) Dead, third relapse 
1, 5-6 None PD 
Disease statusPatient identifierAge (y) and genderCytogeneticsMutational group*Azacitidine cyclesConcomitant treatmentResponse to azacitidineHSCT (total number)StatusFollow-up (mo)
Prior to first HSCT A062 1.1, male Del(5)(q13q33) NRAS None PR Yes (2) Alive with leukemia 38 
2-3 None PD 
Prior to first HSCT CH058 2.8, male Normal PTPN11 1-3 None SD Yes (2) Alive with leukemia 16 
4-7 None PR 
Prior to first HSCT D644§ 1.4, male −7 KRAS None PD Yes (1) Alive in remission 62 
2-4 None SD 
5-8 None CR|| 
Prior to first HSCT D706 5.9, male −7 PTPN11 None PD Yes (3) Alive in remission 66 
Prior to first HSCT D712 6.3, female Normal NRAS None PD Yes (1) Dead, TRM 
Prior to first HSCT D827 0.4, male Normal PTPN11 1-2 6MP Not evaluable Yes (1) Alive in remission 38 
3-7 None CR 
Prior to first HSCT I255 9.1, male Inv(2)(p23q13), −7 No mutation 1-4 AraC, 6MP Not evaluable Yes (1) Alive in remission 49 
Prior to first HSCT NS001 5.4, male −7 NRAS 1-3 None PR No Dead, progressive disease No data 
Prior to first HSCT NS002 0.8, male −7 PTPN11 None SD Yes (1) Dead, TRM 13 
2-3 None PR 
4-11 None CR# 
Relapse after HSCT** NL121 4.6, male Not done PTPN11 2-4 None SD Yes (3) Dead, third relapse 
1, 5-8 None PD 
Relapse after HSCT** SC108 5.0, male Normal†† NF1 1-2 None SD Yes (2) Dead, progressive disease 
3-4 None PR 
None PD 
Relapse after HSCT** SC156 5.3, male Normal†† PTPN11 2-4 None PR Yes (3) Dead, third relapse 
1, 5-6 None PD 

AraC, cytarabine; CR, complete response; PD, progressive disease; PR, partial response; SD, stable disease; 6MP, 6-mercaptopurine; TRM, transplantation-related mortality.

*

All mutations were confirmed to be somatic. Neurofibromatosis type 1 was diagnosed clinically.

Cycles of azacitidine with concomitant antineoplastic medication were considered not evaluable.

From the end of azacitidine treatment to death or last follow-up.

§

This case was published previously by Furlan et al.5 

||

The patient reached genetic CR with disappearance of monosomy 7 and KRAS mutation after 5 and 7 cycles, respectively. Monosomy 7 was tested by fluorescence in situ hybridization, and KRAS mutation was assessed by Sanger sequencing.

Material for mutational studies was not available at the time of clinical CR.

#

The patient reached genetic CR with disappearance of monosomy 7 and PTPN11 mutation after 6 cycles. Monosomy 7 was tested by fluorescence in situ hybridization, and PTPN11 mutation was assessed by Sanger sequencing.

**

Relapse after second HSCT.

††

Last cytogenetic analysis: SC108, at start of azacitidine treatment; SC156, prior to second HSCT.

Severe neutropenia (≤500/µL) was observed in 4 children. Cytopenias led to dose reduction in 2 children, both treated for relapse after second HSCT. Other adverse events were gastrointestinal problems including nausea and vomiting in 2 children, skin rash in 2 children, and fatigue or slight creatinine elevation in 1 patient each. Seven episodes of infection were reported for a total of 64 azacitidine cycles (10.9%).

Of 9 children treated prior to HSCT, 3 normalized blood counts and spleen size (scored as clinical CR) (Table 1). In 2 of these patients, monosomy 7 was present in leukemic cells but disappeared after cycles 5 and 6, respectively. The leukemic karyotype was normal in the other child with clinical CR, precluding the assessment of cytogenetic response. Two of the 3 CR patients featured a somatic PTPN11 gene mutation. The mutation became undetectable after cycle 6 in 1 child (NS002); material for mutational analysis under azacitidine was unavailable from the other child (D827). The third leukemia carried a somatic KRAS mutation, which was no longer detectable after 7 cycles of azacitidine (D644).5  One child (CH058) experienced considerable regression of spleen size and became transfusion independent (scored as clinical PR). All 4 children underwent HSCT after 7 to 11 cycles. A fifth child (NS001) responded unusually early, as indicated by reduction of splenomegaly after the first cycle and hematologic improvement after cycle 3 (scored as clinical PR). Azacitidine was then discontinued because of parental choice. Three children progressed rapidly under azacitidine and underwent expedited HSCT. One child (I255) was not evaluable for response because of concomitant treatment. Three children with JMML received azacitidine for leukemia recurrence after the second HSCT. They achieved clinical PR or could be maintained in stable disease for 4 cycles before progressing.

In summary, this retrospective series indicates that low-dose azacitidine is effective and tolerable in JMML and documents 3 cases of JMML where azacitidine induced complete clinical, cytogenetic, and/or molecular genetic remissions before allogeneic HSCT. Importantly, complete remissions without HSCT have not been documented for JMML thus far, regardless of whether conventional cytostatic chemotherapy or newer experimental agents were applied.7-10  Prospective clinical investigation of azacitidine, such as the ongoing collaborative Innovative Therapies in Childhood Cancer/EWOG-MDS phase 1/2 trial (Eudra-CT 2010-022235-10), is needed to clarify these questions.

Acknowledgments: This work was supported by Deutsche Forschungsgemeinschaft grants FL 345/4-1 (C.M.N., C.F.) and CRC 992-C05 (C.F.).

Contribution: Annamaria C., C.M.N., and C.F. designed research, analyzed data, and wrote the manuscript; and A.Y., M.D., H.H., M.M.v.d.H.-E., F.L., R.M., M.S., U.G.-W., Andrea C., A.E.K., L.O., M.S., I.F., and B.S. performed research.

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

Correspondence: C. Flotho, Division of Pediatric Hematology-Oncology, Department of Pediatrics and Adolescent Medicine, University Medical Center, Mathildenstrasse 1, 79106 Freiburg, Germany; e-mail: christian.flotho@uniklinik-freiburg.de.

1
Santini
 
V
Novel therapeutic strategies: hypomethylating agents and beyond.
Hematology Am Soc Hematol Educ Program
2012
, vol. 
2012
 (pg. 
65
-
73
)
2
Silverman
 
LR
Demakos
 
EP
Peterson
 
BL
, et al. 
Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: a study of the cancer and leukemia group B.
J Clin Oncol
2002
, vol. 
20
 
10
(pg. 
2429
-
2440
)
3
Fenaux
 
P
Mufti
 
GJ
Hellstrom-Lindberg
 
E
, et al. 
International Vidaza High-Risk MDS Survival Study Group
Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study.
Lancet Oncol
2009
, vol. 
10
 
3
(pg. 
223
-
232
)
4
Gerds
 
AT
Gooley
 
TA
Estey
 
EH
Appelbaum
 
FR
Deeg
 
HJ
Scott
 
BL
Pretransplantation therapy with azacitidine vs induction chemotherapy and posttransplantation outcome in patients with MDS.
Biol Blood Marrow Transplant
2012
, vol. 
18
 
8
(pg. 
1211
-
1218
)
5
Furlan
 
I
Batz
 
C
Flotho
 
C
, et al. 
Intriguing response to azacitidine in a patient with juvenile myelomonocytic leukemia and monosomy 7.
Blood
2009
, vol. 
113
 
12
(pg. 
2867
-
2868
)
6
Niemeyer
 
CM
Loh
 
ML
Cseh
 
A
, et al. 
Criteria for evaluating response and outcome in clinical trials for children with juvenile myelomonocytic leukemia.
Haematologica
2015
, vol. 
100
 
1
(pg. 
17
-
22
)
7
Castleberry
 
RP
Emanuel
 
PD
Zuckerman
 
KS
, et al. 
A pilot study of isotretinoin in the treatment of juvenile chronic myelogenous leukemia.
N Engl J Med
1994
, vol. 
331
 
25
(pg. 
1680
-
1684
)
8
Niemeyer
 
CM
Arico
 
M
Basso
 
G
, et al. 
European Working Group on Myelodysplastic Syndromes in Childhood (EWOG-MDS)
Chronic myelomonocytic leukemia in childhood: a retrospective analysis of 110 cases.
Blood
1997
, vol. 
89
 
10
(pg. 
3534
-
3543
)
9
Castleberry
 
RP
Loh
 
ML
Jayaprakash
 
N
, et al. 
Phase II window study of the farnesyltransferase inhibitor R115777 (Zarnestra ®) in untreated juvenile myelomonocytic leukemia (JMML): a children's oncology group study [abstract].
Blood
2005
, vol. 
106
 
11
 
Abstract 2587
10
Bergstraesser
 
E
Hasle
 
H
Rogge
 
T
, et al. 
Non-hematopoietic stem cell transplantation treatment of juvenile myelomonocytic leukemia: a retrospective analysis and definition of response criteria.
Pediatr Blood Cancer
2007
, vol. 
49
 
5
(pg. 
629
-
633
)
Sign in via your Institution