You are on page 1of 7

| |

Received: 26 July 2018    Revised: 10 September 2018    Accepted: 20 September 2018

DOI: 10.1111/ijlh.12939

ORIGINAL ARTICLE

Apoptosis: A biomarker of high-­risk phenotype in pediatric


acute myeloid leukemia?

Anudishi Tyagi1 | Raja Pramanik1 | Radhika Bakhshi2 | Sreenivas Vishnubhatla3 | 


Sameer Bakhshi1

1
Department of Medical Oncology, Dr. B. R.
A. Institute Rotary Cancer Hospital, All India Abstract
Institute of Medical Sciences, New Delhi, Introduction: Dysregulation of apoptosis has been explored in acute myeloid leuke-
India
2
mia (AML); yet, its correlation with clinical outcomes in pediatric AML is unknown.
Shaheed Rajguru College of Applied
Sciences, University of Delhi, New Delhi, This study was aimed to analyze percentage of apoptosis and apoptosis mediated
India
through the intrinsic pathway with clinical outcomes in patients with pediatric
3
Department of Biostatistics, All India
AML.
Institute of Medical Sciences, New Delhi,
India Methods: This prospective study included pediatric AML patients enrolled from July
2013 to August 2016. Annexin-­V (marker of total apoptosis) and caspase-­9 expres-
Correspondence
Dr. Sameer Bakhshi, Department of Medical sion (marker of intrinsic pathway) was determined in baseline bone marrow (BM)
Oncology, Dr. B. R.A. Institute Rotary
samples by flow cytometery and compared with controls (unaffected BM of solid
Cancer Hospital, All India Institute of
Medical Sciences, New Delhi, India. tumors and peripheral blood [PB] of unaffected siblings). Overall survival (OS) and
Email: sambakh@hotmail.com
event-­free survival (EFS) were compared using log-­rank test.
Funding information Results: A total of 151 AML patients were enrolled, median age 10 (range: 0.7-­
Department of Biotechnology, Ministry
18 years). Annexin-­V expression in blast cells was significantly high in AML patients
of Science and Technology, Grant/Award
Number: BT/PR7197/MED/30/899/2012 as compared to BM of subjects with solid tumors (P = 0.01) and PB of healthy sub-
jects (P = 0.04). Caspase-­9 expression in blast cells was not significantly different.
Median annexin-­V expression was significantly higher in patients with WBC count
≥11 000/mm3 (P = 0.02), poor-­risk cytogenetics (P = 0.02), the absence of RUNX1-­
RUNX1T1 translocation (P = 0.004), and the absence of NPM1 mutation (P = 0.05).
Patients with high annexin-­V expression had significantly inferior OS (P = 0.05) in
univariate analysis but not in multivariate analysis (P = 0.32).
Conclusion: Apoptosis as a whole was found to be activated in baseline BM samples
of AML patients. High apoptosis may be associated with high-­risk phenotype in this
disease.

KEYWORDS
acute myeloid leukemia, annexin-V, apoptosis, caspase-9

1 |  I NTRO D U C TI O N hematopoiesis. AML develops through a multistep process of ge-


nomic and epigenetic alterations which results in disease progres-
Acute myeloid leukemia (AML) is a genetic heterogeneous dis- sion.1 Dismal survival and poor outcomes in this disease result in a
ease characterized by the rapid proliferation of immature myeloid large unmet need to discover newer biomarkers and potential ther-
progenitor cells, ultimately resulting in suppression of normal apeutic targets. 2,3

Int J Lab Hematol. 2018;1–7. © 2018 John Wiley & Sons Ltd |  1
wileyonlinelibrary.com/journal/ijlh  
|
2       TYAGI et al.

Abnormalities in apoptotic pathways lead to the development are no published reports correlating total apoptosis and intrinsic
of hematological malignancies and reduce the efficacy of chemo- apoptosis with clinical outcome in pediatric AML patients. The
therapy.4 There are two interconnected pathways of apoptotic cell present study was designed to analyze total apoptosis and apop-
death; the intrinsic pathway involves the mitochondria and cyto- tosis induced due to intrinsic pathway, and correlate these data
chrome C, activation of the apaf-­1 complex finally culminating into with clinic-­p athological features and outcome in pediatric AML
caspase-­9, and the extrinsic pathway, on the other hand, comprises subjects.
the death receptor (DR) DR4, DR5, FAS leading to the proteolytic ac-
tivation of caspase-­8, 3 and 7 finally leading to cell death. Decreased
susceptibility to apoptosis is an important biological feature of leu- 2 | M ATE R I A L S A N D M E TH O DS
kemic cells and reflects their survival potential and resistance to
chemotherapeutic drugs. As a genuine hallmark of cancer, “evasion
2.1 | Patient’s sample
of apoptosis” has been the focus of research for many tumors in
recent years. 5 Several preclinical studies have evaluated the role of This was a single-­centre, prospective cohort study conducted on de
6
extrinsic apoptotic pathway in the pathogenesis of AML. However, novo pediatric (≤18 year) AML subjects at the tertiary cancer centre
the role of intrinsic apoptotic pathway in AML appears less clear. at our institute, who were registered between July 2013 and June
Annexin-­V is widely used as a marker for total apoptosis in 2016. Study was approved by the institute ethics committee.Assent
scientific research.7 Some preclinical studies have used caspase-­9 form from parents was taken from subject’s ≤8 years of age and
as a marker of the intrinsic apoptotic pathway. 8-10 To date, there informed consent was taken from rest. We excluded patients with

(A) (B) (C) (D)

Necrotic Late apoptosis

Caspase-9
PI

Viable cell Early apoptosis

CD45 Annexin-V Annexin-V

(E) ** (F) **
80

60

P* =0.76
* P* =0.01 * P**=0.78
P**=0.04
60

Caspase -9 Expression
40
Annexin-V (%)
40

20
20
0

BM Patients BM Controls PB Controls BM Patients BM Controls PB Controls


(n = 151) (n = 30) (n = 50) (n = 151) (n = 30) (n = 50)

FIGURE 1 Analysis of annexin-­V (total apoptosis) and caspase-­9 expression (intrinsic pathway of apoptosis) by flow cytometery: (A) Side
scattered vs Forward scattered dot plot showing normal distribution of different cells population, (B) Blasts were gated based on low
side scatter and dim CD45 (C) Gated blasts showing percentage of annexin-­V expression (total apoptosis) and (D) Gated blasts showing
percentage of caspase-­9 expression (intrinsic pathway of apoptosis) (E) Comparison of Annexin-­V expression and (F) Caspase-­9 expression
between AML patients, PB controls and BM controls. Y axis of each graph shows the percentage of annexin-­V expression and caspase-­9
expression. P* represents the comparison of patients with PB controls while P** represents their comparison with BM controls. BM, bone
marrow; PB, peripheral blood [Colour figure can be viewed at wileyonlinelibrary.com]
TYAGI et al. |
      3

TA B L E   1   Association of baseline
Variable Annexin-­V Caspase-­9
parameter with caspase-­9 and annexin-­V
(N = 151) (Median IQ Range) P (Median IQ Range) P

Age (y)
<10 (77) 46.7 (34.6-­56.4) 0.97 20.4 (10.5-­32.1) 0.65
≥10 (74) 47.1 (35.4-­55.6) 19.3 (9.9-­33.6)
Sex
Male (108) 46.8 (35.5-­56.7) 0.70 18.6 (10.1-­3 0.3) 0.20
Female (43) 46.7 (32.6-­55.1) 27.3 (11.2-­3 4.5)
Hb (g/dL)
<8 (82) 46.2 (34.6-­53.3) 0.73 19.9 (9.0-­3 0.2) 0.49
≥8 (69) 47.5 (35.1-­57.3) 20.3 (12.4-­36.3)
Platelet (/mm3)
<50 000(48) 47.1 (35.1-­56.7) 0.94 18.9 (12.4-­31.6) 0.90
≥50 000(103) 46.7 (34.6-­55.6) 20.4 (9.9-­33.6)
WBC (/mm3)
<11 000 (55) 42.6 (30.7-­51.5) 0.02 18.8 (12-­27.7) 0.63
≥11 000 (96) 48.3 (42.3-­57.4) 20.6 (6.6-­33.8)
LDH (n = 93)
<280 (12) 42.6 (30.6-­49.2) 0.56 17.5 (10.1-­38.3) 0.82
≥280 (81) 45.5 (32-­52.9) 18.2 (5.9-­31.6)
Karyotype (n = 125)
Good (61) 42.5 (29.8-­51.3) 0.02 22.4 (12.4-­36.3) 0.09
Intermediate (49) 46.7 (38.5-­51.5) 18.1 (7.2-­27.6)
Poor (15) 52.5 (42.6-­62.7) 20.3 (13.7-­39.4)
NPM1 (n = 122)
Negative (113) 48.3 (42.5-­56.7) 0.05 20.1 (9-­3 0.6) 0.17
Positive (9) 31.7 (23.7-­52.9) 35.2 (17.8-­36.4)
*RUNX1-­RUNX1T1 (n = 125)
Negative (76) 45.7 (42.3-­55.6) 0.004 15.2 (9.4-­3 0.1) 0.43
Positive (49) 38.0 (28.2-­51.3) 15.0 (12-­36.3)
FlT3ITD (n = 127)
Negative (117) 47.3 (36.5-­55.7) 0.16 20.1 (9-­32.1) 0.53
Positive (10) 55.9 (47.5-­57.4) 23.9 (14.5-­37.7)

FLT-3ITD, FMS like tyrosine kinase 3 internal tandem duplication; Hb, hemoglobin; IQ, inter-­quartile;
LDH, lactate dehydrogenase; NPM1, nucleophosmin; WBC, white blood cells.
*
Done by cytogenetics.

AML-­M3 and MDS-­related AML. Peripheral blood (PB) samples from and FLT3-­ITD was done using PCR.12,13 Based on this information,
50 age and sex-­matched normal individuals (siblings accompanying the European leukemia network (ELN) classification was used to
the patients) were taken along with bone marrow (BM) samples from categorize the patients into three different prognostic risk groups;
30 age-­and sex-­matched patients with solid tumors without any mi- good, intermediate, and poor risk.14
croscopic bone marrow involvement.

2.3 | Treatment
2.2 | Baseline cytogenetic and molecular analysis
All patients were treated on a uniform protocol. They were induced
Pretreatment samples from all patients underwent cytogenetic with 3 + 7 regimen (daunorubicin 60 mg/m2 for 3 days and cyto-
analysis at NABL-­accredited laboratory, and chromosomal abnor- sine arabinoside 100 mg/m2 as a 24-­hour continuous infusion for
malities were described according to the International System for 7 days). Patients, who did not achieve CR after 1st induction, were
Human Cytogenetic Nomenclature.11 Molecular analysis for NPM1 given ADE as a 2nd induction.15 After achieving remission, the
4       | TYAGI et al.

(A) Apoptosis (Overall survival) (B) Apoptosis (Event-free survival)


1.00

1.00
Log rank; P = 0.07
Log rank; P = 0.05
Cumulative survival

Cumulative survival
0.75

0.75
Apoptosis Low (n = 74)
0.50

0.50
Apoptosis High (n = 77) Apoptosis Low (n = 74)

Apoptosis High (n = 77)


0.25

0.25
0.00

0.00
0 10 20 30 40 50 0 10 20 30 40 50
Time (Mo) Time (Mo)

F I G U R E   2   Kaplan-­Meier curves showing (A) Overall survival and (B) Event-­free survival for apoptosis [Colour figure can be viewed at
wileyonlinelibrary.com]

patients received three cycles of high-­dose cytosine arabinoside at used to estimate OS and EFS and log-­rank test was used to compare
18 g/m2. Twenty-­eight patients received consolidation at 12 g/m2 differences between survival curves. Comparison of Annexin-­V and
as a part of a randomized controlled trial, which was underway dur- caspase-­9 expression between controls and AML patients was done
ing this period at our institute. Patients who relapsed received al- using Mann-­Whitney test. Comparison between Annexin-­V and cas-
logeneic transplantation in CR2, if a matched sibling was available. pase-­9 expression with baseline patient’s characteristics was done
using Dunn test or Ranksum test. For multivariate analysis, a Cox
proportional hazards model was constructed for outcome. Stepwise
2.4 | Measurement of apoptosis using
multivariate Cox regression method was employed to evaluate the
Annexin-­V and caspase-­9
independent prognostic factors. P value ≤0.05 was considered sig-
Five milliliters BM samples was collected in EDTA vials and pro- nificant. All statistical analysis was done using STATA 11.2 (Lakeway,
6
cessed within 5 hour after collection. The whole blood (10 cells) TX, USA).
were mixed with the BD FACS lysing solution and kept in dark for
10 minutes. Followed centrifugation, supernatant was discarded
3 | R E S U LT S
and white blood cell (WBC) pellets were washed with phosphate-­
buffered saline. One microliter of Caspase-­9 (FITC) was added
3.1 | Baseline parameters
into the tube and incubated for 30-­6 0 minutes in 37°C incubator
with 5% CO2. Cells were again washed with phosphate-­buffered A total of 200 patients were registered at the centre during the
saline wash buffer twice and suspended in 200 μL Annexin-­V bind- study period. Out of these, 49 were excluded from the study (five
ing buffer. Cells were then incubated with 5 μL Annexin-­V (APC) patients were acute promyelocytic leukemia, 15 patients had only
and 5 μL CD45 (Percp-­c y5.5) (e-­bioscience, Thermo fisher, San one visit and 29 patients had technical failure during processing).
Diego, CA, USA) for 15 minutes in dark at room temperature. The Therefore, 151 patients were eligible for this study. Median age
cells were then washed and resuspended in 200 μL binding buffer. was 10 years (0.7-­18 years); male: female ratio was 2.5:1. Median
Finally, the cells were mixed with 5 μL propidium iodide (PE) (e-­ blast percentage of AML patients and BM controls was 75 (range:
bioscience, Thermo fisher). Cells were immediately acquired into 10-­95) and 0.8 (range: 0.2-­2 .3), respectively. Cytogenetics was
FACS Calibur (BD Bioscience, Becton Dickinson, San Jose, CA, evaluable in 125 patients (82.8%), 48.8% of the patients had
USA) and analyzed using the Cell Quest Pro software program (BD). good-­r isk cytogenetics while intermediate and poor-­r isk cytoge-
netics were present in 39.2% and 12.0% subjects, respectively.
French American British (FAB) subclassification of AML was avail-
2.5 | Statistical analysis
able in 59 cases. Out of these, 23/59 (38.9%) cases were of M2,
Baseline characteristics for all the patients were summarized using 11/59 (18.6%) cases of M5, 9/59 (15.2%) cases of M4, 6/59 (10.2%)
median and range. Primary overall survival (OS) was measured as the cases of M0 and M1 while 2/59 (3.4%) cases were of M6 and M7.
duration from the date of enrollment to death from any cause. Event-­ Recurrent RUNX1-­RUNX1T1 translocation was seen in 49/125
free survival (EFS) was measured as the duration from date of enroll- (39.2%) cases. FLT3-­I TD and NPM1 mutations were analyzed
ment to date of relapse or death due to any cause. These data were in 127 and 122 cases, respectively. Out of these, 10/127 (7.9%)
censored at the date of last follow-­up (22-­December-­2017) for alive had mutated FLT3-­I TD while 9/122 (7.4%) had mutated NPM1.
patients. Annexin-­V and caspase-­9 expression were categorized into Complete remission (CR) was achieved in 123 patients with induc-
high or low based on their mean value. Kaplan-­Meier curves were tion chemotherapy. Median follow-­u p was 33.8 months.
TYAGI et al.       5|
TA B L E   2   Association of baseline
Variable Event-­free survival Overall survival
parameters and apoptosis with clinical
(n = 151) Hazard Ratio (95%CI) P Hazard Ratio (95%CI) P
outcomes
Age (y)
≤10 (n = 77) 1.00 0.46 1.00 0.62
≥10 (n = 74) 0.8 (0.6-­1.3) 0.9 (0.6-­1.4)
Sex
Male (n = 108) 1.00 0.16 1.00 0.55
Female (n = 43) 1.3 (0.9-­2.1) 1.2 (0.7-­1.8)
Hemoglobin (g/dL)
≤8 (n = 82) 1.00 0.14 1.00 0.18
≥8 (n = 69) 1.4 (0.9-­2.1) (1.3 (0.8-­2.1)
WBC (/mm3)
≤11 000 (=55) 1.00 0.46 1.00 0.57
≥11 000 (=96) 1.2 (0.8-­1.8) 1.1 (0.7-­1.8)
Platelets (/mm3)
≤50 000 (n = 48) 1.00 0.87 1.00 0.84
≥50 000 (n = 103) 1.0 (0.7-­1.6) 1.0 (0.7-­1.7)
LDH (n = 93)
≤280 (n = 12) 1.00 0.51 1.00 0.29
≥280 (n = 81) 0.8 (0.4-­1.6) 0.6 (0.3-­1.4)
CSF (n = 105)
Negative (n = 92) 1.00 0.20 1.00 0.20
Positive (n = 13) 1.5 (0.8-­2.9) 1.5 (0.7-­2.9)
Karyotype (n = 125)
Good (n = 61) 1.00 0.05 1.00 0.002
Intermediate (n = 49) 1.2 (0.7-­1.9) 1.3 (0.8-­2.3)
Poor (n = 15) 2.2 (1.2-­4.3) 3.1 (1.6-­6.2)
FLT3-­ITD (n = 127)
Negative (n = 117) 1.00 0.39 1.00 0.16
Positive (n = 10) 1.4 (0.6-­3.0) 1.8 (0.8-­3.9)
NPM1 (n = 122)
Negative (n = 113) 1.00 0.41 1.00 0.43
Positive (n = 9) 1.4 (0.6-­3.0) 1.4 (0.6-­3.3)
*
RUNX1-­RUNX1T1 (n = 125)
Negative (n = 76) 1.00 0.68 1.00 0.48
Positive (n = 49) 0.9 (0.6-­1.4) 0.8 (0.5-­1.4)
Total apoptosis
Apoptosis (Median=46.7)
≤Median (n = 74) 1.00 0.07 1.00 0.05
≥Median (n = 77) 1.4 (0.9-­2.1) 1.5 (0.9-­2.4)
Caspase-­9 (Median=20.1)
≤Median (n = 74) 1.00 0.28 1.00 0.15
≥Median (n = 77) 1.2 (0.8-­1.8) 1.4 (0.9-­2.1)

CI, confidence interval; CSF, cerebrospinal fluid; FLT-3ITD, FMS like tyrosine kinase 3 internal tan-
dem duplication; LDH, lactate dehydrogenase; NPM1, nucleophosmin; WBC, white blood cells.
*
Done by cytogenetics.
|
6       TYAGI et al.

TA B L E   3   Factors associated with


Event-­free survival Overall survival
–event-­free survival and -­overall survival,
based on multivariate analysis Variables HR (95% CI) P value HR (95% CI) P value

Apoptosis 1.2 (0.8-­1.8) 0.43 1.3 (0.8-­2.1) 0.32


*
Karyotype (others vs 1.9 (1.0-­3.6) 0.04 2.6 (1.3-­4.9) 0.004
poor)

CI, confidence interval; HR, hazard ratio.


*
Others include good and intermediate risk group of cytogenetics.

on the genomic landscape of AML, several questions remain unan-


3.2 | Comparison of apoptosis between AML
swered; which apoptotic circuitry is activated (intrinsic or extrinsic)?
patients and controls
Can there be some predictive biomarker for targeting apoptosis in
Annexin-­V expression in blast cells (Figure 1A-­C) of AML patients AML? Can some reliable prognostic biomarker be identified related
was significantly higher than the lymphocytes of PB (P = 0.04) and to these cascades?
BM (P = 0.01) controls at diagnosis (Figure 1E). However, intrinsic We envisaged to explore these points and got some key find-
pathway of apoptosis as determined by caspase-­
9 expression in ings. Firstly, Annexin-­V (a marker of total apoptosis) was signifi-
blast cells (Figure 1D) was not significantly different between AML cantly overexpressed in baseline BM of AML patients as compared
patients, PB and BM controls (Figure 1F). to BM or PB of the matched controls. Increased apoptosis defi-
nitely gives a survival advantage to leukemic cells; it generates
vacant niches that potentially become repopulated by more ag-
3.3 | Association of baseline patient’s
gressive subclones. Thus, apoptosis increases proliferative pres-
characteristics with annexin-­V and caspase-­9
sure, promotes clonal selection, and drives tumor evolution.18
Annexin-­V was found to be significantly higher in patients with high However, caspase-­9, the key component of intrinsic pathway, was
WBC count (P = 0.02), poor-­risk cytogenetics (P = 0.02), the absence not overexpressed in the blast of AML patients. This indirect clue
of NPM1 mutation (P = 0.05), and the absence of RUNX1-­RUNX1T1 suggests that the extrinsic pathway might be more active in AML,
translocation (P < 0.01). However, no association was observed a less clear fact that needs validation in large studies. Secondly,
between caspase-­9 and any of the baseline patient characteristics Annexin-­V was overexpressed significantly in patients with poor
(Table 1). prognostic factors such as poor cytogenetics, high WBC count,
lower expression of RUNX1-­RUNX1T1 and NPM1, which appears
quite logical. Thirdly, Annexin-­V over expression was significantly
3.4 | Association of baseline patient’s
associated with inferior OS in univariate analysis although not in
characteristics, annexin-­V and caspase-­9 with
multivariate analysis.
survival outcomes
Hess et al6 in their study using reverse transcriptase multiplex
In univariate analysis, poor-­r isk cytogenetics adversely affected ligation-­dependent probe amplification (RT-­MLPA) of several pro
EFS and OS. Higher Annexin-­V expression was significantly as- and antiapoptotic genes, validated a three gene expression signature
sociated with inferior overall survival (P = 0.05) (Figure 2A). (BIRC3, BAX-­(1) and BMF) which predicted OS. In another study,
However, no association was found between Annexin-­V and EFS Kohler et al19 concluded that high levels of Bax and Bad correlate
(Table 2; Figure 2B). No association could be demonstrated be- with poor outcome, particularly in patients who do not enter CR and
tween caspase-­9 with either EFS or OS (Table 2). In multivariate may serve as prognostic markers in AML.19 Another study by our
analysis, karyotype was the only significant factor (P = 0.004) group also suggested that high BAX/BCL2 RMFI ratio was positively
(Table 3). associated with CR rates in AML. 20 It is interesting to note that com-
ponents of the intrinsic cascade upstream of the mitochondria seem
to be upregulated while caspase-­9 (the final components of the in-
4 |  D I S CU S S I O N trinsic cascade) is neither overexpressed nor prognostic for survival.
In a recent study by Singh et al, 21 the apoptotic index (AI) at base-
Recent studies have demonstrated that the true “Achilles heel” of line was reported to be significantly associated with poor prognostic
leukemia may be exploited by targeting both signal transduction features of pediatric ALL. We also observed a similar association of
and apoptotic programs simultaneously (synthetic lethality).16,17 total apoptosis at baseline with features of a high-­risk phenotype for
Indeed, apoptosis has been established as a mechanism of antican- pediatric AML.
cer defense and several components of the apoptotic machinery Our study is unique because for the first time, we have seen the
are being currently targeted in clinical trials of leukemia.18 Although association of apoptosis at baseline directly with survival. Although
alterations in the apoptotic cascade have been revealed by studies apoptosis had a significant association with OS in univariate analysis
TYAGI et al. |
      7

it could not retain its effect in multivariate model. We hypothesized 4. Ossenkoppele G. Minimal residual disease in acute myeloid leuke-
that the interaction of karyotype (a strong prognostic factor in AML) mia is predicted by an apoptosis-­resistant protein profile at diagno-
sis. Clin Cancer Res. 2005;11(7):2540‐2546.
is the reason for this effect. Larger studies would be beneficial in
5. Hanahan D, Weinberg RA. Review hallmarks of cancer : the next
elucidating the exact relationship. generation. Cell. 2011;144(5):646‐674.
As more and more newer molecules targeting different compo- 6. Hess CJ, Berkhof J, Denkers F, Ossenkoppele GJ, Schouten JP,
nents of the apoptotic circuitry are making moves into early phase Oudejans JJ. Activated intrinsic apoptosis pathway is a key re-
lated prognostic parameter in acute myeloid leukemia. J Clin Oncol.
clinical trials of AML, a disease which is molecularly highly heteroge-
2018;25(10):1209‐1215.
neous, it becomes all the more essential to select out patients who 7. Savitskiy VP, Shman TV, Potapnev MP. Comparative measurement
have specific defects in the apoptotic machinery. It is also import- of spontaneous apoptosis in pediatric acute leukemia by different
ant to identify which component (intrinsic or extrinsic) is activated techniques. Cytometery B Clin Cytom. 2003;56(1):16‐22.
8. Lu Y, Chen G. Effector caspases and leukemia. Int J Cell Biol.
in a particular patient, so that we may have appropriately enriched
2011;2011(49):738301.
dose expansion cohorts during these clinical trials. Markers such as 9. Degterev A, Boyce M, Yuan J. A decade of caspases. Oncogene.
Annexin-­V can thus be simple enrichments strategies. 2003;22(53):8543‐8567.
To conclude, apoptosis as a whole was found to be activated in 10. Chowdhury I, Tharakan B, Bhat GK. Comparative biochemistry and
physiology, Part B caspases — an update. Comp Biochem Physiol B
baseline BM samples of AML patients. High apoptosis may be as-
Biochem Mol Biol. 2008;151(1):10‐27.
sociated with high-­risk phenotype in this disease and the extrinsic
11. Nomenclature ISC on HC. ISCN 2013: an International System for
pathway may actually be overexpressed in these clones, which is Human Cytogenetic Nomenclature (2013). Basel, Switzerland: Karger
hypothesis generating. Publishers; 2013:140.
12. Chopra A, Soni S, Pati H, et al. Nucleophosmin mutation analysis in
acute myeloid leukaemia: immunohistochemistry as a surrogate for
AC K N OW L E D G E M E N T S molecular techniques. Indian J Med Res. 2016;143(6):763‐768.
13. Sharawat SK, Raina V, Kumar L, et al. High fms-­like tyrosine kinase-­3
We thank our nursing staff, data entry operator, patients, and their par- (FLT3) receptor surface expression predicts poor outcome in FLT3
ents who participated in the study. We also acknowledge the follow- internal tandem duplication (ITD) negative patients in adult acute
myeloid leukaemia: a prospective pilot study from India. Indian J
ing funding sources: (a) Department of Biotechnology, Government
Med Res. 2016;143(Supplement1):S11‐S16.
of India under grant (Number: BT/PR7197/MED/30/899/2012 14. Döhner H, Estey E, Grimwade D, Amadori S. Diagnosis and manage-
Dated: 26 September 2013) (b) AIIMS, Institutional Junior Research ment of AML in adults: 2017 ELN recommendations from an inter-
Fellowship grant. national expert panel. Blood. 2016;129(4):424‐447.
15. Burnett AK, Russell NH, Hills RK, et  al. A randomized comparison of
daunorubicin 90 mg/m2 vs 60 mg/m2 in AML induction: results from the
C O N FL I C T O F I N T E R E S T UK NCRI AML17 trial in 1206 patients. Blood. 2015;125(25):3878‐3886.
16. Dasgupta A, Nomura M, Shuck R, Yustein J. Cancer’s achil-
All authors declare no conflict of interests. les’ heel: apoptosis and necroptosis to the rescue. Int J Mol Sci.
2017;18(1):1‐20.
17. Samudio I, Konopleva M, Carter B, Andreeff M. Apoptosis in leu-
AU T H O R S C O N T R I B U T I O N kemias: regulation and therapeutic targeting. Cancer Treat Res.
2010;145:197‐217.
AT, RP, RB, and SB: contribution to design the research study, ac- 18. Labi V, Erlacher M. How cell death shapes cancer. Cell Death Dis.
quisition of data, interpretation of data, drafting the manuscript and 2015;6(3):e1675‐11.
19. Kohler T, Schill C, Krahl R, et al. High Bad and Bax mRNA expression
critical review the intellectual content of the manuscript; SV: was the
correlate with negative outcome in acute myeloid leukemia (AML).
statistician. The final draft was approved by all. Leukemia. 2002;16(1):22‐29.
20. Sharawat SK, Bakhshi R, Vishnubhatla S, Gupta R, Bakhshi S. Brief
report BAX/BCL2 RMFI ratio predicts better induction response
ORCID in pediatric patients with acute myeloid leukemia. Pediatr Blood
Cancer. 2013;60(8):E63‐E66.
Sameer Bakhshi  http://orcid.org/0000-0001-9367-4407
21. Singh A, Bhatia P, Trehan A, Bansal D, Singh A, Bhatia A. Low spon-
taneous apoptosis index at diagnosis predicts a high risk pheno-
type in paediatric acute lymphoblastic leukemia. Indian J Med Res.
REFERENCES 2015;147(3):248‐255.

1. Shih AH, Jiang Y, Meydan C, et al. Gain of function in acute myeloid


leukemia. Cancer Cell. 2016;27(4):502‐515.
2. Arora RS, Arora B. Acute leukemia in children : a review of the cur- How to cite this article: Tyagi A, Pramanik R, Bakhshi R,
rent Indian data. South Asian J Cancer. 2016;5(3):155‐160.
Vishnubhatla S, Bakhshi S. Apoptosis: A biomarker of
3. Bonaventure A, Harewood R, Stiller CA, et al. Articles Worldwide
comparison of survival from childhood leukaemia for 1995–2009, high-­risk phenotype in pediatric acute myeloid leukemia? Int
by subtype, age, and sex (CONCORD-­2): a population-­based study J Lab Hematol. 2018;00:1–7. https://doi.org/10.1111/
of individual data for 89 828 children from 198 registries in 53 ijlh.12939
countries. Lancet Hematol. 2017;4(5):202‐217.

You might also like