Search

Recent Articles

IJMPO—A Journey of a Thousand Miles

Author : Padmaj S. Kulkarni

Coronavirus Disease 2019 Treatment—T-Cells Hold the Key in Severe Cases

Author : Kunal Das, Nitika Agrawal, Mansi Kala, Rakhee Khanduri

Why Is China Importing COVID-19 Vaccine Now?

Author : Purvish M. Parikh

Treating acute myeloid leukemia among children with down syndrome

CC BY-NC-ND 4.0 · Indian J Med Paediatr Oncol 2020; 41(06): 841-845

DOI: DOI: 10.4103/ijmpo.ijmpo_175_20

Abstract

Background: Down Syndrome (DS) children with acute myeloid leukemia (AML) have unique differences in clinical features, epidemiologic nature, and biologic patterns of disease compared with AML in children without DS. Aims and Objective: AML in DS children should be considered distinct disorder from AML in Non DS population and treatment needs to be customized for this population. In this retrospective study spanning from 2014 to 2019 we present our experience of managing leukemia in children with DS. Materials and Methods: From 2014 and 2019, 72 children aged below 18 years were managed at our institute with acute myeloid leukemia (AML). Out of these 72 children with AML, 7 children were with DS which was confirmed by karyotyping. Majority of these children had M7 while M2 and M4 subtypes were seen in one child each. On conventional karyotyping in addition to trisomy 21 additional cytogenetic abnormalities were seen in 4 patients. Two children had trisomy 8. One child had deletion of 11 chromosomes and one had translocation between 8 and 21 chromosomes. Results: All 7 children were administered intensive chemotherapy with curative intent after informed parental consent. All 7 children achieved complete remission. Four out of 7 children had complications related to severe neutropenia. Conclusion: All patients of DS with AML should be offered chemotherapy with curative intent. Endeavour should be to give less aggressive chemotherapy protocol to bring down treatment related mortality.



Publication History

Received: 20 April 2020

Accepted: 19 August 2020

Article published online:
14 May 2021

© 2020. Indian Society of Medical and Paediatric Oncology. This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial-License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/.)

Thieme Medical and Scientific Publishers Pvt. Ltd.
A-12, 2nd Floor, Sector 2, Noida-201301 UP, India

Abstract

Background: Down Syndrome (DS) children with acute myeloid leukemia (AML) have unique differences in clinical features, epidemiologic nature, and biologic patterns of disease compared with AML in children without DS. Aims and Objective: AML in DS children should be considered distinct disorder from AML in Non DS population and treatment needs to be customized for this population. In this retrospective study spanning from 2014 to 2019 we present our experience of managing leukemia in children with DS. Materials and Methods: From 2014 and 2019, 72 children aged below 18 years were managed at our institute with acute myeloid leukemia (AML). Out of these 72 children with AML, 7 children were with DS which was confirmed by karyotyping. Majority of these children had M7 while M2 and M4 subtypes were seen in one child each. On conventional karyotyping in addition to trisomy 21 additional cytogenetic abnormalities were seen in 4 patients. Two children had trisomy 8. One child had deletion of 11 chromosomes and one had translocation between 8 and 21 chromosomes. Results: All 7 children were administered intensive chemotherapy with curative intent after informed parental consent. All 7 children achieved complete remission. Four out of 7 children had complications related to severe neutropenia. Conclusion: All patients of DS with AML should be offered chemotherapy with curative intent. Endeavour should be to give less aggressive chemotherapy protocol to bring down treatment related mortality.

Introduction

Down syndrome (DS) is the most common genetic anomaly with incidence ranging from 1 in 600 to 1 in 1000 live births.[1] This syndrome is recognized by characteristic dysmorphic features and developmental abnormalities. Children affected with DS have trisomy of 21 chromosome, and besides characteristic features, extra genetic material of 21st chromosome also renders them uniquely susceptible to myeloid leukemia.[2] DS children with myeloid leukemia exhibit unique differences in clinical features, epidemiologic nature, and biologic patterns of disease compared with myeloid leukemia among children who do not have DS.[3] Children with DS have 500 fold increased incidence acute megakaryoblastic leukemia (AMkL) which is a subtype of myeloid leukemia[4] AMkL is rare among children without DS and is associated with poor survival.[5] Paradoxically outcomes of AMkL in children with DS are better than general population.[6],[7] Nearly all DS AMkL cases are positive for somatic mutations of the GATA1 gene which is not present in non DS population with AMkL.[8] This finding suggest that DS AMkL should be considered distinct disorder from AMkL in Non DS population and treatment needs to be customized for this population. In this retrospective study spanning from 2014 to 2019 we present our experience of managing leukemia in children with DS.

Objectives

Objective of presenting this case series is to highlight the distinctive features of acute myeloid leukemia (AML) in children with DS and to increase awareness among referring pediatricians about better prognosis for AMl in these children as compared to general population.

Methods

It is a retrospective study spanning 5 years from 2014 to 2019. Descriptive statistics design has been used to describe 6 cases of AML in children with DS managed during these 5 years.

Case series

From 2014 and 2019, 72 children aged below 18 years were managed at our institute with AML. Out of these 72 children with AML, 7 children were with DS which was confirmed by karyotyping. Clinical features of these 7 children are summarized in [Table 1]. Fever, hepatomegaly and splenomegaly were the common presenting features. Only one child (Patient number 4) had extramedullary disease and he had significant delay of 120 days between onset of symptoms and presentation. All but one child had blasts cells present on peripheral smear. All patients were subjected to bone marrow aspiration and biopsy. AML was further sub classified on morphology as per French American British classification [Table 2]. Majority (5/7) of these children had M7 while M2 and M4 subtypes were seen in one child each (Patient number 5 and 1 respectively). Bone marrow samples were sent for conventional karyotyping as well as polymerase chain reaction for established translocations and mutations. On conventional karyotyping in addition to trisomy 21 additional cytogenetic abnormalities were seen in 4 patients. Two children had trisomy 8 (Patient number 1 and 6). One child had deletion of 11 chromosomes (Patient number 3) and one had translocation between 8 and 21 chromosomes (Patient number 5). None of these 7 children had history of preceding transient myeloproliferative disorder. All 7 children were administered intensive chemotherapy after informed parental consent [Table 3]. Briefly chemotherapy consisted of induction, consolidation (high dose cytosine arabinoside [AraC]) and maintenance (monthly cycles of 6thioguanine and low dose AraC) for 18 months. Etoposide was administered during induction phase [Table 3]. CNS directed therapy was administered by intrathecal AraC, during each cycle of induction and consolidation followed by 3 monthly during maintenance chemotherapy. All patients were treated in single room with reverse barrier nursing. As per our institutional protocol we do not use antifungal prophylaxis in children with AML during induction chemotherapy. Septran prophylaxis was started during maintenance chemotherapy.{Table 1}{Table 2}{Table 3}

Table 1

Base line characteristics at the time of diagnosis

Patient number

Age (months)

Sex

Duration of symptoms (days)

EMD (yes/no)

Fever

Hepatomegaly (yes/no)

Splenomegaly (yes/no)

Presentation Hb (gm/dL)

Presentation TLC (×109/L)

Peripheral blood blasts%

Presentation platelets (×109/L)

LDH, above ULN (yes/no)

EMD –Extramedullary disease; Hb – Hemoglobin; TLC – Total leukocyte counts; LDH – Lactate dehydrogenase; ULN – Upper limit of normal

1

48

Male

60

No

No

Yes

Yes

9

9.4

40

40

Yes

2

14

Male

100

No

Yes

Yes

Yes

9.4

6.1

11

11

Yes

3

25

Female

30

No

Yes

Yes

No

7.2

61

60

53

Yes

4

26

Male

120

Yes

Yes

Yes

Yes

7.4

32

62

131

No

5

24

Male

45

No

Yes

Yes

Yes

5.9

2.5

12

120

Yes

6

32

Male

60

No

Yes

Yes

No

4.9

1.9

0

30

No

7

18

Female

90

No

Yes

Yes

Yes

6.5

52

71

15

No

Table 2

French American British classification and cytogentic abnormalities of all 7 patients

Patient number

FAB classification

Additional cytogenetic abnormalities

PCR/FISH for translocations

FAB – French American British classification; PCR – Polymerase chain reaction; FISH – Fluorescent in situ hybridization

1

M4

Trisomy 8

Negative

2

M7

None

Negative

3

M7

Del 11

Negative

4

M7

None

Negative

5

M2

t(8;21)

t(8;21)

6

M7

Trisomy 8

Negative

7

M7

Add (5) (p15), del 7 (p13)

Negative

Table 3

Treatment and outcome details

Results

Outcomes of treatment have been summarized in [Table 3]. Early response to treatment was evaluated by peripheral blood blast clearance after end of induction chemotherapy at D + 7. Granulocyte colony stimulating factor (G-CSF) was used in all patients in dose of 5 mcg/kg subcutaneously. G-CSF was started on D + 8 of chemotherapy. G-CSF was administered till two consecutive reports of ANC > 500/mm3. Median days of use were 10 days (Range 4–14 days) during induction cycles. No patients were given prophylactic antibiotics or antifungals. All 7 patients are currently under follow-up, patient number 1 and 2 have been followed up for more than 60 months. Remaining 5 children have been under follow-up for median duration of 24 months (9–48 months) at the time of writing. Assessment for minimal residual disease was not done for any of these children during follow-up because as it is not part of our institutional protocol.

Discussion

Incidence of AML among pediatric population is estimated to be 7.7 cases per million,[9] accounting for 18% of all childhood leukemia.[10] Unlike Acute lymphoblastic leukemia, treatment of AML is associated with poor prognosis and significant relapse rates. Almost 50%–80% patients in India suffer severe toxicity, relapse or refractory disease.[11] Radhakrishnan et al. evaluated and found twodrug induction regimen to be more suitable then the conventional three drug induction regimen in order to reduce treatment related toxicity among sick patients.[12] In India cure rates have been inferior for pediatric malignancies when compared to developed countries.[13] With improvement is supportive care and standardization of treatment protocols, improved survival has also been reported in pediatric AML management in India.[14] Abandonment of therapy is common in the Indian/low middle income country scenario. Socioeconomic factors and long-term morbidity associated with DS also play part in failure to complete therapy in AML.[15] Historically patients with DS with AML were considered to have high risk disease and patients with DS who developed AML were not administered protocol based therapy largely owing to DS per se In a large multi centre study from India 54% patients of AML were not given any form of therapy due to various reasons.[16] Though there is no study from India, one can safely assume that higher percentage of DS with AML do not get any form of treatment due to reasons discussed above. In last three decades outcomes have improved dramatically once patients with DS were treated according to tailor made protocols for them.[17] Initial study of 12 patients with DS published in 1992 was first one to show excellent outcomes with chemotherapy protocol with AraC as backbone. In this study 3-year event free survival (EFS) was 100% for patients of DS compared to only 28% for non DS patients.[18] Since then more than 10 studies have been published studying patients of DS with AML and all have reported significantly better outcome in these patients in comparison to non DS AML.[17] One of recently published results is from COG-A2971 study which included 132 patients of DS with AML. In this study induction consisted of oral thio guanine (T), continuous infusion of AraC (A) and daunorubicin (D) also known as TAD protocol. Consolidation was as per Capizzi regimen with high dose AraC (3 g/m2) and L asparginase. There was no maintenance chemotherapy. Five-year overall survival and EFS rates were impressive 84% and 79%.[19] Therapy reduction and its outcome were studied in 170 patients treated with International Ml-DS 2006 trial. In this study, lower cumulative dose of etoposide was used along with AraC and Idarubicin (AIE) during induction. There was no maintenance chemotherapy. Despite being less intensive, there were comparable results as far as disease outcomes were concerned.[20] This proved the concept that altered drug metabolism in myeloid leukemic-DS cells and the altered capacity for DNA repair in normal DS cells both permit and necessitate reduced exposures to cytotoxic therapies for optimal survival benefit. Two groups have published findings after studying large number of DS with AML. Based on these findings we have modified our induction protocol to less intensive 3 + 7 or ADE from 2017 onward. Anthracycline cardiotoxicity is more pronounced in children with DS. Cardiac assessment in all patients was done by baseline echocardiography and repeated at 06 monthly intervals till the follow-up period. No patient exceeded cumulative threshold dose of anthracycline and we did not find any evidence of cardiac toxicity in any of the patient. In present study all patients developed febrile neutropenia during induction, out of which 4 patients who received 3 drug induction developed severe complications necessitating intensive care. All patients successfully recovered with good supportive care. Though these are small numbers, they do indicate that addition of third drug during induction increases severe neutropenic complications. In our study, we used prophylactic G-CSF in all patients with no adverse outcomes even though some studies have documented higher risk of relapse with G-CSF.[21],[22] Our policy of use of G-CSF in children with AML post induction chemotherapy is guided by various publications in literature where G-CSF has not been associated with increased risk of relapse.[23] In a Cochrane review where 19 studies with more than 5000 patients were analyzed, relapse rates did not increase due to use of CSFs.[24] Shortening the duration of neutropenia in children with AML is very important in our setting but authors would like to caution that long-term follow-up is required to assess the risk of relapse in these children due to use of G-CSF. With advances in cytogenetics it is clear that besides trisomy 21, DS children with AML may show additional chromosomal abnormalities.[25] Recent study has suggested that normal karyotype, besides trisomy 21 may indicate poor prognosis among this specific group of children.[26] In our study only 2 children (Patient number 2 and 4) did not have additional cytogenetic anomaly while other 5 had additional trisomy (Patient number 1 and 6), deletion (Patient number 3 and 7), or translocation (Patient number 5). However, this is too small a series to comment on significance of these anomalies in terms of management or prognosis. Additional cytogenetic abnormalities are important especially in long-term outcome. We intend to follow-up these children to better understand effects of additional cytogenetic abnormalities.

Conclusion

DS children are particularly susceptible to AML, which is relatively uncommon disorder among children without DS. However, prognosis among DS children is much better than non-DS children for AML. During this time period, 65 non DS AML children were also treated. At 2 years of follow-up, 38 children were in remission giving an OS of 58.4% in this cohort. To conclude all patients of DS with AML should be offered chemotherapy, as they show excellent response to AraC, which was remains keystone in any chemotherapy protocol. Endeavour should be to give less aggressive chemotherapy protocol to bring down treatment related mortality. G-CSF should be judiciously used from D + 8 during induction to bring down duration and complications of febrile neutropenia in these patients.

Conflict of Interest

There are no conflicts of interest.

References

  1. Amayreh A, Al QaqaK, Ali AH, Issa K. Clinical and cytogenetic profile of down syndrome at king hussein medical centre. JRMS 2012; 19: 14-8
  2. Mitelman F, Heim S, Mandahl N. Trisomy 21 in neoplastic cells. Am J Med Genet Suppl 1990; 7: 262-6
  3. Falini B, Tiacci E, Martelli MP, Ascani S, Pileri SA. New classification of acute myeloid leukemia and precursor-related neoplasms: Changes and unsolved issues. Discov Med 2010; 10: 281-92
  4. Lange B. The management of neoplastic disorders of haematopoiesis in children with Down'ssyndrome. Br J Haematol 2000; 110: 512-24
  5. Hama A, Yagasaki H, Takahashi Y, Nishio N, Muramatsu H, Yoshida N. et alAcute megakaryoblasticleukaemia (AMkL) in children: Acomparison of AMkL with and without Down syndrome. Br J Haematol 2008; 140: 552-61
  6. Abildgaard L, Ellebaek K, Gustafsson G, Abildgaard L, Ellebaek E, Gustafsson G. et al Optimal treatment intensity in children with downsyndrome and myeloid leukaemia: Data from 56 children treated on NOPHO-AML protocols and areview of the literature. Ann Hematol 2006; 85: 275-80
  7. Taga T, Shimomura Y, Horikoshi Y, Ogawa A, Itoh M, Okada M. et alContinuous and high-dose cytarabine combinedchemotherapy in children with down syndrome and acute myeloid leukemia: Report from theJapanese children's cancer and leukemia study group (JCCLSG) AML 9805 down study. Pediatr Blood Cancer 2011; 57: 36-40
  8. Wechsler J, Greene M, McDevitt MA, Anastasi J, Karp JE, Le BeauMM. et alAcquired mutations in GATA1 in the megakaryoblastic leukemia of Down syndrome. Nat Genet 2002; 32: 148-52
  9. Linabery AM, Ross JA. Trends in childhood cancer incidence in the U.S. (1992-2004). Cancer 2008; 112: 416-32
  10. Howlader N, Howlader A, Krapcho MS, Noone AM, Neyman N, Aminou R. et al. SEER cancer statistics review, 1975-2009 (vintage 2009 populations).
  11. Arora RS, Arora B. Acute leukemia in children: A review of the current Indian data. South Asian J Cancer 2016; 5: 155-60
  12. Radhakrishnan V, Thampy C, Ganesan P, Rajendranath R, Ganesan TS, Rajalekshmy KR. et alAcute myeloid leukemia in children: Experience from tertiary cancer centre in India. Indian J Hematol Blood Transfus 2016; 32: 257-61
  13. Kulkarni KP, Marwaha RK. Childhood acute myeloid leukemia: An Indian perspective. Pediatr Hematol Oncol 2011; 28: 257-68
  14. Seth R, Pathak N, Singh A, Chopra A, Kumar R, Kalaivani M. Pediatric acute myeloid leukemia: Improved survival rates in India. Indian J Pediatr 2017; 84: 166-7
  15. Arora RS, Pizer B, Eden T. Understanding refusal and abandonment in the treatment of childhood cancer. Indian Pediatr 2010; 47: 1005-10
  16. Kapoor R, Mathews V, Sengar M, Bhurani D, Radhakrishnan V, Philip C. et alHematological cancer consortium: Multi-center acute myeloid leukemia registry data from India. Blood 2018; 132 (Suppl 1): 4006
  17. Caldwell JT, Ge Y, Taub JW. Prognosis and management of acute myeloid leukemia in patients with Down syndrome. Expert Rev Hematol 2014; 7: 831-40
  18. Ravindranath Y, Abella E, Krischer JP, Wiley J, Inoue S, Harris M. et alAcute myeloid leukemia (AML) in down's syndrome is highly responsive to chemotherapy: Experience on pediatric oncology group AML Study 8498. Blood 1992; 80: 2210-4
  19. Sorrell AD, Alonzo TA, Hilden JM, Gerbing RB, Loew TW, Hathaway L. et alFavorable survival maintained in children who have myeloid leukemia associated with down syndrome using reduced-dose chemotherapy on children's oncology group trial A2971: A report from the children's oncology group. Cancer 2012; 118: 4806-14
  20. Uffmann M, Rasche M, Zimmermann M, Neuhoff CV, Creutzig U, Dworzak M. et alTherapy reduction in patients with Down syndrome and myeloid leukemia: The international ML-DS 2006 trial. Blood 2017; 129: 3314-21
  21. Lehrnbecher T, Zimmermann M, Reinhardt T, Dworzak M, Stary J, Creutzig U. et alProphylactic human granulocyte colony-stimulating factor after induction therapy in Pediatric acute myeloid leukemia. Blood 2007; 109: 936-43
  22. Lohmann DJ, Asdahl PH, Abrahamsson J, Ha SY, Jónsson ÓG, Kaspers GJL. et al Use of granulocyte colony-stimulating factor and risk of relapse in pediatric patients treated for acute myeloid leukemia according to NOPHO-AML 2004 and DB AML-01. Pediatr Blood Cancer 2019; 66: e27701
  23. Feng X, Lan H, Ruan Y, Li C, Sarin YK. Impact on acute myeloid leukemia relapse in granulocyte colony-stimulating factor application: A meta-analysis. Hematology 2018; 23: 581-9
  24. Gurion R, Belnik-Plitman Y, Gafter-Gvili A, Paul M, Vidal L, Ben-Bassat I. et alColony-stimulating factors for prevention and treatment of infectious complications in patients with acute myelogenous leukemia. Cochrane Database Syst Rev 2012; 6: CD008238
  25. Von Neuhoff C, Reinhardt D, Sander A, Bradtke J, Betts DR, Zemanova Z. et alPrognostic impact of specific chromosomal aberrations in a large group of pediatric patients with acute myeloid leukemia treated uniformly according to trial AML-BFM 98. J Clin Oncol 2010; 28: 2682-9
  26. Blink M, Zimmermann M, von Neuhoff C, Reinhardt D, Haas V, Hasle H. et alNormal karyotype is a poor prognostic factor in myeloid leukemia of Down syndrome: A retrospective, international study. Haematologica 2014; 99: 299-307

Address for correspondence

Dr. A K Simalti
Army Hospital (Research and Referral),
New Delhi - 110 010
India   

Publication History

Received: 20 April 2020

Accepted: 19 August 2020

Article published online:
14 May 2021

© 2020. Indian Society of Medical and Paediatric Oncology. This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial-License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/.)

Thieme Medical and Scientific Publishers Pvt. Ltd.
A-12, 2nd Floor, Sector 2, Noida-201301 UP, India

Patient number

Induction-1

Number of induction cycles

Absolute day 7 blast count

Severe neutropenic complications (yes/no)

Remission status

Type of consolidation

Follow-up duration (months)

TAD-6 – Thioguanine (100 mg/m2 × 4 days), Cytosine arabinoside (200 mg/m2 daily as continuous infusion × 4 days), Daunorubicin (20 mg/m2 daily over 1 h infusion × 4 days); AIE – Cytosine arabinoside (100 mg/m2 continuous infusion × 7 days), Idarubicin (12 mg/m2 as 4 h infusion × 3 days), Etoposide (150 mg/m2 as 1 h infusion × 3 days), 3+7 (Idarubicin 12 mg/m2 × 3 days + cytosine arabinoside 100 mg/m2 daily as continuous infusion × 7 days); HaM – High dose cytosine arabinoside, mitoxantrone (cytosinearabinoside 1 g/m2 as a 4 h infusion every 12 h × 3 days, Mitoxantrone 10 mg/m2 as 30 min infusion × 2 days; HiDAC – High dose cytosine arabinoside (3 g/m2 as a 3 h infusion × 3 days); CR – Complete remission

1

TAD

4

100

Yes

CR

HaM

>60

2

TAD

4

0

Yes

CR

HaM

>60

3

3+7

2

100

No

CR

HiDAC

48

4

3+7

1

0

No

CR

HiDAC

36

5

AIE

2

200

Yes

CR

HiDAC

24

6

AIE

2

0

Yes

CR

HiDAC

18

7

3+7

1

0

No

CR

HiDAC

09

References

  1. Amayreh A, Al QaqaK, Ali AH, Issa K. Clinical and cytogenetic profile of down syndrome at king hussein medical centre. JRMS 2012; 19: 14-8
  2. Mitelman F, Heim S, Mandahl N. Trisomy 21 in neoplastic cells. Am J Med Genet Suppl 1990; 7: 262-6
  3. Falini B, Tiacci E, Martelli MP, Ascani S, Pileri SA. New classification of acute myeloid leukemia and precursor-related neoplasms: Changes and unsolved issues. Discov Med 2010; 10: 281-92
  4. Lange B. The management of neoplastic disorders of haematopoiesis in children with Down'ssyndrome. Br J Haematol 2000; 110: 512-24
  5. Hama A, Yagasaki H, Takahashi Y, Nishio N, Muramatsu H, Yoshida N. et alAcute megakaryoblasticleukaemia (AMkL) in children: Acomparison of AMkL with and without Down syndrome. Br J Haematol 2008; 140: 552-61
  6. Abildgaard L, Ellebaek K, Gustafsson G, Abildgaard L, Ellebaek E, Gustafsson G. et al Optimal treatment intensity in children with downsyndrome and myeloid leukaemia: Data from 56 children treated on NOPHO-AML protocols and areview of the literature. Ann Hematol 2006; 85: 275-80
  7. Taga T, Shimomura Y, Horikoshi Y, Ogawa A, Itoh M, Okada M. et alContinuous and high-dose cytarabine combinedchemotherapy in children with down syndrome and acute myeloid leukemia: Report from theJapanese children's cancer and leukemia study group (JCCLSG) AML 9805 down study. Pediatr Blood Cancer 2011; 57: 36-40
  8. Wechsler J, Greene M, McDevitt MA, Anastasi J, Karp JE, Le BeauMM. et alAcquired mutations in GATA1 in the megakaryoblastic leukemia of Down syndrome. Nat Genet 2002; 32: 148-52
  9. Linabery AM, Ross JA. Trends in childhood cancer incidence in the U.S. (1992-2004). Cancer 2008; 112: 416-32
  10. Howlader N, Howlader A, Krapcho MS, Noone AM, Neyman N, Aminou R. et al. SEER cancer statistics review, 1975-2009 (vintage 2009 populations).
  11. Arora RS, Arora B. Acute leukemia in children: A review of the current Indian data. South Asian J Cancer 2016; 5: 155-60
  12. Radhakrishnan V, Thampy C, Ganesan P, Rajendranath R, Ganesan TS, Rajalekshmy KR. et alAcute myeloid leukemia in children: Experience from tertiary cancer centre in India. Indian J Hematol Blood Transfus 2016; 32: 257-61
  13. Kulkarni KP, Marwaha RK. Childhood acute myeloid leukemia: An Indian perspective. Pediatr Hematol Oncol 2011; 28: 257-68
  14. Seth R, Pathak N, Singh A, Chopra A, Kumar R, Kalaivani M. Pediatric acute myeloid leukemia: Improved survival rates in India. Indian J Pediatr 2017; 84: 166-7
  15. Arora RS, Pizer B, Eden T. Understanding refusal and abandonment in the treatment of childhood cancer. Indian Pediatr 2010; 47: 1005-10
  16. Kapoor R, Mathews V, Sengar M, Bhurani D, Radhakrishnan V, Philip C. et alHematological cancer consortium: Multi-center acute myeloid leukemia registry data from India. Blood 2018; 132 (Suppl 1): 4006
  17. Caldwell JT, Ge Y, Taub JW. Prognosis and management of acute myeloid leukemia in patients with Down syndrome. Expert Rev Hematol 2014; 7: 831-40
  18. Ravindranath Y, Abella E, Krischer JP, Wiley J, Inoue S, Harris M. et alAcute myeloid leukemia (AML) in down's syndrome is highly responsive to chemotherapy: Experience on pediatric oncology group AML Study 8498. Blood 1992; 80: 2210-4
  19. Sorrell AD, Alonzo TA, Hilden JM, Gerbing RB, Loew TW, Hathaway L. et alFavorable survival maintained in children who have myeloid leukemia associated with down syndrome using reduced-dose chemotherapy on children's oncology group trial A2971: A report from the children's oncology group. Cancer 2012; 118: 4806-14
  20. Uffmann M, Rasche M, Zimmermann M, Neuhoff CV, Creutzig U, Dworzak M. et alTherapy reduction in patients with Down syndrome and myeloid leukemia: The international ML-DS 2006 trial. Blood 2017; 129: 3314-21
  21. Lehrnbecher T, Zimmermann M, Reinhardt T, Dworzak M, Stary J, Creutzig U. et alProphylactic human granulocyte colony-stimulating factor after induction therapy in Pediatric acute myeloid leukemia. Blood 2007; 109: 936-43
  22. Lohmann DJ, Asdahl PH, Abrahamsson J, Ha SY, Jónsson ÓG, Kaspers GJL. et al Use of granulocyte colony-stimulating factor and risk of relapse in pediatric patients treated for acute myeloid leukemia according to NOPHO-AML 2004 and DB AML-01. Pediatr Blood Cancer 2019; 66: e27701
  23. Feng X, Lan H, Ruan Y, Li C, Sarin YK. Impact on acute myeloid leukemia relapse in granulocyte colony-stimulating factor application: A meta-analysis. Hematology 2018; 23: 581-9
  24. Gurion R, Belnik-Plitman Y, Gafter-Gvili A, Paul M, Vidal L, Ben-Bassat I. et alColony-stimulating factors for prevention and treatment of infectious complications in patients with acute myelogenous leukemia. Cochrane Database Syst Rev 2012; 6: CD008238
  25. Von Neuhoff C, Reinhardt D, Sander A, Bradtke J, Betts DR, Zemanova Z. et alPrognostic impact of specific chromosomal aberrations in a large group of pediatric patients with acute myeloid leukemia treated uniformly according to trial AML-BFM 98. J Clin Oncol 2010; 28: 2682-9
  26. Blink M, Zimmermann M, von Neuhoff C, Reinhardt D, Haas V, Hasle H. et alNormal karyotype is a poor prognostic factor in myeloid leukemia of Down syndrome: A retrospective, international study. Haematologica 2014; 99: 299-307