Skip to main content
Erschienen in: Journal of Translational Medicine 1/2018

Open Access 01.12.2018 | Research

Association of genetic polymorphisms in genes involved in Ara-C and dNTP metabolism pathway with chemosensitivity and prognosis of adult acute myeloid leukemia (AML)

verfasst von: Ke-Wei Zhu, Peng Chen, Dao-Yu Zhang, Han Yan, Han Liu, Li-Na Cen, Yan-Ling Liu, Shan Cao, Gan Zhou, Hui Zeng, Shu-Ping Chen, Xie-Lan Zhao, Xiao-Ping Chen

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2018

Abstract

Background

Cytarabine arabinoside (Ara-C) has been the core of chemotherapy for adult acute myeloid leukemia (AML). Ara-C undergoes a three-step phosphorylation into the active metabolite Ara-C triphosphosphate (ara-CTP). Several enzymes are involved directly or indirectly in either the formation or detoxification of ara-CTP.

Methods

A total of 12 eQTL (expression Quantitative Trait Loci) single nucleotide polymorphisms (SNPs) or tag SNPs in 7 genes including CMPK1, NME1, NME2, RRM1, RRM2, SAMHD1 and E2F1 were genotyped in 361 Chinese non-M3 AML patients by using the Sequenom Massarray system. Association of the SNPs with complete remission (CR) rate after Ara-C based induction therapy, relapse-free survival (RFS) and overall survival (OS) were analyzed.

Results

Three SNPs were observed to be associated increased risk of chemoresistance indicated by CR rate (NME2 rs3744660, E2F1 rs3213150, and RRM2 rs1130609), among which two (rs3744660 and rs1130609) were eQTL. Combined genotypes based on E2F1 rs3213150 and RRM2 rs1130609 polymorphisms further increased the risk of non-CR. The SAMHD1 eQTL polymorphism rs6102991 showed decreased risk of non-CR marginally (P = 0.055). Three SNPs (NME1 rs3760468 and rs2302254, and NME2 rs3744660) were associated with worse RFS, and the RRM2 rs1130609 polymorphism was marginally associated with worse RFS (P = 0.085) and OS (P = 0.080). Three SNPs (NME1 rs3760468, NME2 rs3744660, and RRM1 rs183484) were associated with worse OS in AML patients.

Conclusion

Data from our study demonstrated that SNPs in Ara-C and dNTP metabolic pathway predict chemosensitivity and prognosis of AML patients in China.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12967-018-1463-1) contains supplementary material, which is available to authorized users.
Abkürzungen
Ara-C
cytarabine arabinoside
dNTP
deoxy-ribonucleoside triphosphate
AML
acute myeloid leukemia
ara-CTP
cytarabine arabinoside triphosphate
CR
complete remission
RFS
relapse-free survival
OS
overall survival
eQTL
expression Quantitative Trait Loci
SNPs
single nucleotide polymorphisms
SLC29A1
solute carrier family 29 member 1
DCK
deoxycytidine kinase
NDPKs
nucleoside diphosphate kinases
dCTP
deoxycytidine triphosphate
SAMHD1
SAM domain and HD domain 1
dATP
deoxyadenosine triphosphate
dTTP
deoxythymidine triphosphate
dGTP
deoxyguanosine triphosphate
RR
ribonucleotide reductase
CTP
cytidine triphosphate
RRM1
ribonucleotide reductase catalytic subunit M1
RRM2
ribonucleotide reductase catalytic subunit M2
WT1
Wilms tumor 1
DNMT3A
DNA methyltransferase 3 alpha
E2F1
transcription factor 1
FLT3
FMS-like Tyrosine Kinase3
NMP1
nucleophosmin
NME1
nucleoside diphosphate kinase 1
NME2
nucleoside diphosphate kinase 2
EFS
event free survival
CMPK1
cytidine/uridine monophosphate kinase 1
EMR
electronic medical record
HSCT
haematopoietic stem cell transplantation
HWE
Hardy–Weinberg Equilibrium
FAB
French–Britain–American
WBC
white blood cell
LDH
lactate dehydrogenase
BM
bone marrow
RBC
red blood cell
Allo-SCT
allogeneic hematopoietic stem cell transplantation
CR
complete remission
THP
pirarubicin
MA
Ara-C + mitoxantrone
IA
Ara-C + idarubicin
TA
Ara-C + pirarubicin
DA
Ara-C + daunorubicin
CAG
Ara-C + aclarubicin + G-CSF
G-CSF
granulocyte colony-stimulating factor
OR
odds ratio
HR
hazard ratio
CI
confidence interval
LD
linkage disequilibrium
HIV
human immunodeficiency virus

Background

Acute myeloid leukemia (AML) is a hematological malignancy characterized by malignant proliferation of the hemopoietic system. AML is a heterogeneous collection of diseases characterized by distinct morphological, chromosomal and cytogenetic abnormalities. Subtype specific therapy for AML is not available despite the FAB M3 subtype. Cytarabine arabinoside (Ara-C) remains the first-line chemotherapeutic agent for the treatment of AML for decades [13]. Clinical studies have demonstrated that the complete remission (CR) rate ranges in 50–70% and 5 year survival rate ranges in 27–40% in AML patients receiving Ara-C based chemotherapy [46].
Ara-C is a nucleotide analog that is transmembrane transported into leukemia cells by the nucleoside transporters include the solute carrier family 29 member 1 (SLC29A1) [7]. Intracellular Ara-C undergoes a three-step phosphorylation into the activate metabolite Ara-C triphosphate (ara-CTP) by deoxycytidine kinase (DCK) [8], cytidine monophosphate kinase 1 (CMPK1) [9], and nucleoside diphosphate kinases (NDPKs) in turn [10]. Ara-CTP competes with deoxycytidine triphosphate (dCTP) for incorporation into DNA, and thus results in blockade of DNA synthesis and cell death [11]. More recently, Schneider C and colleagues found that ara-CTP is hydrolyzed by the deoxynucleoside triphosphate (dNTP) triphosphohydrolase SAM domain and HD domain 1 (SAMHD1), which promotes the detoxification of intracellular ara-CTP pools [12]. Disruption of SAMHD1 increases Ara-C sensitivity in both cultured leukemia cells and mouse model of AML [13].
Intracellular level of dNTPs, which are consisted of dCTP, dATP, dTTP and dGTP, is another potential mechanism that affect Ara-C sensitivity. High intracellular dNTPs can inhibit Ara-C phosphorylation and decrease the accumulation of ara-CTP through inhibition of DCK activity by a feedback mechanism. Exhaustion of the intracellular CTP/dCTP pools facilitates Ara-C phosphorylation and increases the incorporation of ara-CTP into DNA by reducing the feedback inhibition on DCK [14]. Ribonucleotide reductase (RR) is a key enzyme responsible for the reduction of ribonucleotides to deoxyribonucleotides and plays important roles in the regulation of intracellular CTP/dCTP pools [15, 16] (Fig. 1). RR is mainly composed of RRM1 and RRM2 subunits, and the expression of RRM2 is regulated by the transcription factor E2F1 [17]. An inverse correlation between RRM1/RRM2 mRNA expression and intracellular ara-CTP in leukemia blasts after Ara-C treatment is observed in clinic. Ex vivo ara-C sensitivity study with primary AML samples has also shown correlationship between RRM1 mRNA expression and Ara-C sensitivity [18]. In addition, higher RRM1 expression in leukemia blast cells predicts better relapse-free survival (RFS) in AML patients [19].
Influence of genetic factors on either Ara-C chemosensitivity and/or AML prognosis has raised much interest in recent years [20, 21]. Our previous studies have shown associations of single nucleotide polymorphisms (SNPs) in DCK, Wilms tumor 1 (WT1) and DNA methyltransferase 3 alpha (DNMT3A) with drug response to Ara-C based induction therapy in AML [2224]. In addition, somatic mutations in genes such as DNMT3A, FMS-like Tyrosine Kinase3 (FLT3) and nucleophosmin (NPM1) can also affect AML prognosis [2426]. For the Ara-C metabolic pathway genes, a promoter polymorphism rs2302254 in NME1 (encoding NDPK-A) is associated increased risk of Ara-C associated neurotoxicity [27]. The RRM1 3′-UTR SNP rs1042919 and promoter SNP rs1561876 are reported to be associated with decreased intracellular ara-CTP levels during Ara-C therapy, and increased risk of relapse and/or worse event free survival (EFS) in Caucasian AML patients treated with cytarabine and cladribine [18]. More recently, association of RRM1 rs9937 variant with induction therapy related death and survival rates in AML patients is also reported [20]. In the study by Cao et al. the RRM2 nonsynonymous SNP rs1130609 (S59A) is associated with decreased Ara-C cytotoxicity in HapMap lymphoblast cell lines from the CEU population and poorer EFS for the St Jude AML97 cohort, but the clinical association was not replicated in St Jude AML02 cohort [18]. There is no studies focused on the association of CMPK1, NME2, SAMHD1 and E2F1 polymorphisms with Ara-C response in AML patients presently.
In this study, we sought to identify genetics polymorphisms in Ara-C and dNTP metabolic pathway including CMPK1, NME1/NME2 (encoding NDPK), SAMHD1, RRM1, RRM2 and E2F1 with drug response to Ara-C based chemotherapy and AML prognosis in Chinese AML patients.

Methods

Patients

Three hundred and sixty-one newly diagnosed AML patients were recruited from Xiangya hospital from May 2009 to Nov 2017. Peripheral venous blood or bone marrow samples were obtained before chemotherapy. Patients with AML FAB M3 subtype, or combined critical illness and cancer, less than 16 years old, and secondary leukemia were excluded. M3 subtype was excluded because this subtype of patients had unique chemotherapy regimens.
All patients received an “7 + 3” induction regimen consisting of Ara-C (100–200 mg/m2 for day 1–7) and anthracyclines (mitoxantrone 8–16 mg/m2, or daunorubicin 45–60 mg/m2, or aclarubicin 20 mg/m2, or pirarubicin 30 mg/m2, or idarubicin 10–20 mg/m2, for day 1–3). One or two courses of induction chemotherapy were given to obtain complete remission (CR). Achievement of CR was defined according to the international recommendations as we described previously [23], which include: < 5% blasts in the bone marrow; absence of extramedullary disease; absolute neutrophil count > 1.0 × 109/L; platelet count > 100 × 109/L. Once CR was achieved, the patients received sequential consolidation therapy consisting of Ara-C and anthracyclines or haematopoietic stem cell transplantation (HSCT). Regular follow-up was carried out by outpatient review or telephone.
The clinical and pathological information of the AML patients were obtained by chart review of electronic medical record (EMR) from the hospital. Overall survival (OS) and relapse free survival (RFS) were used to indicate disease outcomes. Relapse was defined as the presence of > 5% of blast cells in the bone marrow or reappearance of blast cells in the peripheral blood or development of extramedullary disease. RFS was calculated from the date of achievement of CR until the date of relapse or death from any cause. OS was calculated from the date of AML diagnosis to the date of death from any cause. For those patients without relapse or death event by the end of the study follow-up, survival end-points were censored at the date of last follow-up. The risk stratification criteria based on cytogenetics and molecular characteristics was described elsewhere [23].

SNPs selection and genotyping

Based on literature searching on Ara-C metabolism related genes in the PubMed database, 7 genes involved in Ara-C metabolic pathway (CMPK1, NEM1, NEM2, SAMHD1, RRM1, RRM2, E2F1) were selected. Candidate tag SNPs in these genes were initially selected based on the database from NCBI (https://​www.​ncbi.​nlm.​nih.​gov/​snp/​) and the 1000 Genomes Project (https://​www.​ncbi.​nlm.​nih.​gov/​variation/​tools/​1000 genomes/) using the Haploview 4.2 (Cambridge, MA, USA). Then, potential influence of the SNPs on expression the corresponding genes was analyzed by the expression quantitative trait locus (eQTL) database (https://​gtexportal.​org/​home/​). A total of 12 SNPs (CMPK1 rs7543016, NME1 rs3760468 and rs2302254, NME2 rs3744660, SMAD1 rs6102991, rs28372906 and rs6029941, E2F1 rs3213150 and rs3213180, RRM1 rs2412344 and rs183484, and RRM2 rs1130609) were finally selected (Table 1, Fig. 2).
Table 1
Candidate SNPs selected in the study and influence on gene expression according to eQTL database
SNP
Gene
Position
MAF
Alleles
Region
eQTL P
rs7543016
CMPK1
1:47333967
0.467
C/G
Gly8Arg
0.3
rs3760468
NME1
17:51153130
0.376
A/T
Promoter
2.3e−21
rs2302254
NME1
17:51153539
0.087
C/T
Promoter
2.7e−13
rs3744660
NME2
17:51168540
0.162
A/G
Intron
2.5e−12
rs6102991
SAMHD1
20:36956238
0.386
A/G
Promoter
2.0e−6
rs28372906
SAMHD1
20:36951753
0.063
C/T
5′-UTR
NA
rs6029941
SAMHD1
20:36891027
0.447
A/G
3′-UTR
2.1e−8
rs3213150
E2F1
20:33687284
0.357
C/T
Intron
NA
rs3213180
E2F1
20:33675818
0.286
C/G
3′-UTR
NA
rs2412344
RRM1
11:4086354
0.430
C/T
Promoter
1.1e−6
rs183484
RRM1
11:4119902
0.381
G/T
Arg284Arg
9.4e−6
rs1130609
RRM2
2:10122793
0.343
G/T
5′-UTR/Ser59Ala
1.2e−7
NA expression data is not available in the eQTL database
Genomic DNA was extracted from blood samples using Blood DNA Kit II (Omega Bio-Tek, USA) according to the manufacturer’s protocol. The 12 candidate SNPs were genotyped by ionization-time-of-flight mass spectrometry (Sequenom, SanDiego, CA). The genotyping success rates were higher than 96.4% except for the rs7543016 polymorphism (89.8%) by the Massarray system. Genotyping results for the SNPs were confirmed in 10% randomly selected samples by PCR-based sequencing.

Statistical analysis

Statistical analyses were carried out with the IBM SPSS software (version 22.0; IBM SPSS Inc., Chicago, IL, USA). Comparisons of continuous variables between genotype groups were performed by nonparametric test (Mann–Whitney U test). Fitness of the genotype distribution to Hardy–Weinberg Equilibrium (HWE) was analyzed by using the χ2 test. Associations of genotypes with CR or non-CR was analyzed by unconditional logistic regression and indicated by odds ratio (OR) and the 95% confidence interval (CI). Survival probabilities were estimated by the Kaplan–Meier method, and differences between genotypes were evaluated using the log-rank test. Cox proportional hazard models were constructed for RFS and OS, adjusting for potential confounding covariates including gender, age at diagnosis, risk stratification, allo-SCT, WBC count, serum lactate dehydrogenase (LDH) level and bone marrow blast counts at diagnosis as necessary. A stepwise selection method was used to determine the potential confounding covariates. Hazard ratio (HR) was used to estimate association of risk factors with RFS and OS. Three genetic models (dominant, recessive and additive model) were tested for the genotype association analysis. P < 0.05 was considered to be significant.

Results

Baseline characteristics and the overall CR status of the AML patients

The baseline clinical characteristics of the 361 AML patients were summarized in Table 2. The median age of the patients was 43 [range 16–76] years old. Among the AML patients, 196 were male and 165 were female. According to FAB classification, the patients were classified into 7 subtypes. M2 was the most common subtype (53.5%), followed by M5 (20.8%), and no patient with the M7 or M0 subtype was recruited. The median number of WBC was 14.7 × 109/L, the mean serum level of LDH was 363 U/L. Other clinical information was also shown in Table 2. When stratified by cytogenetics and molecular abnormalities, 71, 187, and 78 patients were classified as low risk, intermediate risk, and high risk, respectively. Information of karyotype and somatic mutations was not available for 25 patients. For the 361 patients received Ara-C based induction therapy, 211 patients (58.4%) received Ara-C + mitoxantrone (MA) or Ara-C + idarubicin (IA) regimens, 47 patients (13.0%) received Ara-C + THP (TA), 52 patients (14.4%) received Ara-C + aclarubicin + G-CSF (CAG) and 31 patients (8.6%) received Ara-C + daunorubicin (DA). A total of 205 patients (56.8%) achieved CR after one or two courses of chemotherapy, and CR could not be evaluated accurately for 36 (10.0%) patients due to insufficiencies in clinical evidence. Sixty-four (17.7%) patients received hematopoietic stem cell transplantation, 178 patients died during the follow-up time, and 7 (1.9%) patients were out of touch after several follow-up. The median and mean follow-up period for OS analysis were 926 and 1235 days, respectively.
Table 2
The baseline characteristics of 316 AML patients
Characteristics
Totality, n (%)
Median (range)
Gender
361
 
 Male
196 (54.3%)
 
 Female
165 (45.7%)
 
Age
361
43 (16–76) years
FAB classification
 M1
19 (5.3%)
 
 M2
193 (53.5%)
 
 M4
63 (17.5%)
 
 M5
75 (20.8%)
 
 M6
6 (1.7%)
 
 Undefined
5 (1.4%)
 
WBC
356 (98.6%)
14.7 (0.4–426.0), × 109/L
LDH
341 (94.5%)
363 (17–5853), U/L
BM blast %
348 (96.4%)
71% (17–99%)
RBC
356 (98.6%)
2.19 (0.67–4.98), × 1012/L
Hemoglobin
356 (98.6%)
72 (27–149), g/L
Platelets
356 (98.6%)
34 (2–546), × 109/L
Neutrophil
356 (98.6%)
2.2 (0.0–340.3), × 109/L
Risk stratifications
 Low risk
71 (19.7%)
 
 Intermediate risk
187 (51.8%)
 
 High risk
78 (21.6%)
 
 Undefined
25 (6.9%)
 
FLT3-ITD mutation
 Positive
42 (11.6%)
 
 Negative
294 (81.4%)
 
 Unknown
25 (6.9%)
 
NPM1 mutation
 Positive
67 (18.6%)
 
 Negative
269 (74.5%)
 
 Unknown
25 (6.9%)
 
CEBPA mutation
 Positive
50 (13.9%)
 
 Negative
286 (79.2%)
 
 Unknown
25 (6.9%)
 
Karyotype
 Normal
255 (70.6%)
 
 Abnormal
84 (23.3%)
 
 Unknown
22 (6.1%)
 
Allo-HCT
 Yes
64 (17.7%)
 
 No
297 (82.3%)
 
CR after two courses of induction therapy
 Yes
205 (56.8%)
 
 No
120 (33.2%)
 
 Not evaluated
36 (10.0%)
 
Chemotherapy regimens
 MA
125 (34.6%)
 
 IA
86 (23.8%)
 
 TA
47 (13.0%)
 
 DA
31 (8.6%)
 
 CAG
52 (14.4%)
 
 Other regimens
20 (5.5%)
 
FAB French–Britain–American, WBC white blood cell, LDH lactate dehydrogenase, BM bone marrow, RBC red blood cell, Allo-SCT allogeneic hematopoietic stem cell transplantation, CR complete remission, THP pirarubicin, MA Ara-C + mitoxantrone, IA Ara-C + idarubicin, TA Ara-C + THP, DA Ara-C + daunorubicin, CAG Ara-C + aclarubicin + G-CSF

Comparison of CR rate among genotypes after Ara-C based induction therapy and results of unconditional logistic regression analysis

Significant differences in CR rates among genotypes for the NME2 rs3744660 (P = 0.047), E2F1 rs3213150 (P = 0.043), and RRM2 rs1130609 (P = 0.042) polymorphisms were observed (p values from Chi square test by three genotype groups). For the NME2 rs3744660 polymorphism, the CR rate was 68.5, 53.9, and 57.1%, respectively, for the GG, GA and AA genotypes, and carriers of the rs3744660 A allele (dominant model) showed significantly increased non-CR (indicative of chemoresistance) rate (GA+AA vs GG, 45.6% vs 31.5%, crude OR = 1.829, 95% CI 1.127–2.967, P = 0.014) (Table 3). For the E2F1 rs3213150 polymorphism, the CR rate was 60.4, 69.2, and 50.0%, respectively, for the CC, CT, and TT genotypes, and rs3213150 TT homozygous showed significantly increased non-CR rate as compared with carriers of the rs3213150 C allele (recessive model) (50.0% vs 34.4%, crude OR = 1.903, 95% CI 1.014–3.574, P = 0.043). For the RRM2 rs1130609 polymorphism, the CR rate was 64.1, 67.1, and 44.1%, respectively, for the TT, TG and GG genotypes, and patients with the rare GG genotype showed significantly increased non-CR rate as compared with carriers of the T allele (GG vs TT+TG, 55.9% vs 34.3%, crude OR = 2.428, 95% CI 1.181–4.990, P = 0.014). Carriers of the SAMHD1 rs6102991 G allele (GA+GG) also showed marginally decreased non-CR rate as compared with the wild-type AA homozygotes (crude OR = 0.611, 95% CI 0.369–1.013, P = 0.055). No differences in CR rates among genotypes for other SNPs were observed (Table 3).
Table 3
Comparison of CR rate among genotypes after 2 courses of Ara-C based chemotherapy
Genotype
Total (n)
CR, n (%)
Non-CR, n (%)
P
OR (95% CI)
P a
OR (95% CI)a
CMPK1 rs7543016
 CC
93
57 (61.3%)
36 (38.7%)
 
1.00 (reference)
 
1.00 (reference)
 GC
167
106 (63.5%)
61 (36.5%)
0.727
0.911 (0.540–1.537)
0.921
0.970 (0.531–1.772)
 GG
30
23 (76.7%)
7 (23.3%)
0.125
0.482 (0.188–1.238)
0.076
0.365 (0.120–1.111)
NME1 rs3760468
 AA
115
77 (67.0%)
38 (33.0%)
 
1.00 (reference)
 
1.00 (reference)
 AT
154
96 (62.3%)
58 (37.7%)
0.434
1.224 (0.737–2.033)
0.256
1.420 (0.775–2.597)
 TT
44
26 (59.1%)
18 (40.9%)
0.353
1.403 (0.686–2.869)
0.465
1.359 (0.597–3.096)
NME1 rs2302254
 CC
186
123 (66.1%)
63 (33.9%)
 
1.00 (reference)
 
1.00 (reference)
 CT
114
65 (57.0%)
49 (43.0%)
0.114
1.473 (0.912–2.375)
0.288
1.355 (0.775–2.370)
 TT
19
13 (68.4%)
6 (31.6%)
0.840
0.901 (0.327–2.481)
0.534
0.688 (0.212–2.232)
NME2 rs3744660
 GG
213
146 (68.5%)
67 (31.5%)
 
1.00 (reference)
 
1.00 (reference)
 AG
89
48 (53.9%)
41 (46.1%)
0.016
1.861 (1.121–3.091)
0.024
1.953 (1.092–3.484)
 AA
14
8 (57.1%)
6 (42.9%)
0.376
1.634 (0.545–4.897)
0.770
1.299 (0.361–4.673)
 AA+AG
103
56 (54.4%)
47 (45.6%)
0.014
1.829 (1.127–2.967)
0.030
1.852 (1.062–3.236)
SAMHD1 rs6102991
 AA
88
49 (55.7%)
39 (44.3%)
 
1.00 (reference)
 
1.00 (reference)
 GA
161
107 (66.5%)
54 (33.5%)
0.093
0.634 (0.372–1.080)
0.183
0.669 (0.370–1.209)
 GG
62
43 (69.4%)
19 (30.6%)
0.090
0.555 (0.280–1.101)
0.063
0.483 (0.224–1.038)
 AG+GG
223
150 (67.3%)
73 (32.7%)
0.055
0.611 (0.369–1.013)
0.086
0.611 (0.349–1.071)
SAMHD1 rs28372906
 TT
284
180 (63.4%)
104 (36.6%)
 
1.00 (reference)
 
1.00 (reference)
 CT
32
21 (65.6%)
11 (34.4%)
0.802
0.907 (0.420–1.955)
0.878
0.935 (0.394–2.217)
 CC
4
3 (75.0%)
1 (25.0%)
0.632
0.577 (0.059–5.618)
0.953
0.933 (0.091–9.524)
SAMHD1 rs6029941
 GG
88
50 (56.8%)
38 (43.2%)
 
1.00 (reference)
 
1.00 (reference)
 GA
161
107 (66.5%)
54 (33.5%)
0.132
0.664 (0.389–1.132)
0.339
0.739 (0.398–1.374)
 AA
65
42 (64.6%)
23 (35.4%)
0.330
0.721 (0.372–1.395)
0.370
0.706 (0.330–1.512)
E2F1 rs3213180
 GG
149
95 (63.8%)
54 (36.2%)
 
1.00 (reference)
 
1.00 (reference)
 GC
141
90 (63.8%)
51 (36.2%)
0.990
0.997 (0.617–1.610)
0.915
0.970 (0.552–1.704)
 CC
29
16 (55.2%)
13 (44.8%)
0.383
1.429 (0.639–3.195)
0.429
1.449 (0.578–3.636)
E2F1 rs3213150
 CC
111
67 (60.4%)
44 (39.6%)
 
1.00 (reference)
 
1.00 (reference)
 CT
159
110 (69.2%)
49 (30.8%)
0.133
0.678 (0.408–1.127)
0.105
0.616 (0.343–1.106)
 TT
46
23 (50.0%)
23 (50.0%)
0.232
1.523 (0.762–3.042)
0.128
1.835 (0.839–4.016)
 CC+CT
270
177 (65.6%)
93 (34.4%)
 
1.00 (reference)
 
1.00 (reference)
 TT
46
23 (50.0%)
23 (50.0%)
0.043
1.903 (1.014–3.574)
0.033
2.208 (1.065–4.567)
RRM1 rs183484
 GG
100
62 (62.0%)
38 (38.0%)
 
1.00 (reference)
 
1.00 (reference)
 GT
177
118 (66.7%)
59 (33.3%)
0.434
0.816 (0.490–1.359)
0.112
0.617 (0.340–1.119)
 TT
42
22 (52.4%)
20 (47.6%)
0.287
1.483 (0.716–3.071)
0.263
1.616 (0.697–3.731)
RRM1 rs2412344
 TT
89
56 (62.9%)
33 (37.1%)
 
1.00 (reference)
 
1.00 (reference)
 CT
183
119 (65.0%)
64 (35.0%)
0.734
0.913 (0.539–1.545)
0.510
0.817 (0.449–1.488)
 CC
48
28 (58.3%)
20 (41.7%)
0.599
1.212 (0.592–2.483)
0.967
1.018 (0.429–2.415)
RRM2 rs1130609
 TT
131
84 (64.1%)
47 (35.9%)
 
1.00 (reference)
 
1.00 (reference)
 GT
149
100 (67.1%)
49 (32.9%)
0.599
0.876 (0.534–1.436)
0.920
0.972 (0.557–1.697)
 GG
34
15 (44.1%)
19 (55.9%)
0.034
2.264 (1.053–4.867)
0.069
2.347 (0.935–5.882)
 TT+GT
280
184 (65.7%)
96 (34.3%)
 
1.00 (reference)
 
1.00 (reference)
 GG
34
15 (44.1%)
19 (55.9%)
0.014
2.428 (1.181–4.990)
0.044
2.398 (1.025–5.618)
aAdjusted by age, risk stratification, WBC count, and serum LDH level
Logistic regression analysis showed significant associations of risk stratification, age, pretreatment WBC counts and LDH levels with non-CR risk (Additional file 1: Table S1). When adjusted by these risk factors, NME2 rs3744660, E2F1 rs3213150 and RRM2 rs1130609 were still significantly associated with risk of non-CR (Table 3). NME2 rs3744660 AG genotype or carriers of the rs3744660 A allele showed increased risk for non-CR (AG vs GG: adjusted OR = 1.953, 95% CI 1.092–3.484, P = 0.024; AG+AA vs GG: adjusted OR = 1.852, 95% CI 1.062–3.236, P = 0.030). For the E2F1 rs3213150 polymorphism, as compared with patients carrying the E2F1 rs3213150 C allele, those with the TT genotype showed increased risk of non-CR (adjusted OR = 2.208, 95% CI 1.065–4.567, P = 0.033). While for the RRM2 rs1130609 polymorphism, the rare GG homozygotes showed significantly increased risk of non-CR in comparison with carriers of the T allele (adjusted OR = 2.398, 95% CI 1.025–5.618, P = 0.044). Marginally decreased risk of non-CR was also observed for the SAMHD1 rs6102991 polymorphism in a dominant model (GA+GG vs AA, adjusted OR = 0.611, 95% CI 0.349–1.071, P = 0.086).

Combined influence of E2F1 and RRM2 polymorphisms on chemosensitivity to Ara-C based induction therapy in AML patients

As E2F1 is a transcription factor involved in the regulation of RRM2 expression, a combined genotype analysis of the E2F1 and RRM2 polymorphisms on CR rate was carried out. As both E2F1 rs3213150 and RRM2 rs1130609 polymorphisms were associated with increased risk for non-CR in negative models, patients were grouped based on genotypes of the two SNPs (Table 5): rs1130609 TT+GT/rs3213150 CC+CT (both favorable for CR, n = 237), rs1130609 TT+GT/rs3213150 TT, n = 37), rs1130609 GG/rs3213150 CC+CT (n = 27), and rs1130609 GG/rs3213150 TT (both unfavorable for CR, n = 6). Significant difference in CR rate was observed among the four groups (χ2 = 10.637, P = 0.014), which was 66.7, 54.1, 48.1, and 16.7%, respectively (Table 5). As compared with the favorable genotypes combination group (rs1130609 TT+GT/rs3213150 CC+CT), carriers of two unfavorable genotypes (rs1130609 GG/rs3213150 TT) and those carrying at least one unfavorable genotype showed significantly increased risk of non-CR (OR = 10.00, 95% CI 1.149–87.06, P = 0.004; OR = 2.118, 95 CI 1.233–3.637, P = 0.006, respectively, Table 4). After adjustment for age, risk stratification, and WBC counts, results of logistic regression analysis indicated that patients carrying both or at least one unfavorable genotype showed significantly increased risk of non-CR (OR = 9.780, 95% CI 1.099–87.069, P = 0.041; OR = 2.257, 95 CI 1.273–4.002, P = 0.005, respectively, Table 4).
Table 4
Combined genotypes of RRM2 and E2F1 polymorphisms and chemotherapy sensitivity in AML patients
rs1130609
rs3213150
CR, n (%)
Non-CR, n (%)
P
OR (95% CI)
P b
OR (95% CI)b
TT+GT
CC+CT
158 (66.7%)
79 (33.3%)
 
1.00 (reference)
 
1.00 (reference)
TT+GT
TT
20 (54.1%)
17 (45.9%)
0.134
1.700 (0.844–3.426)
0.048
2.115 (1.006–4.450)
GG
CC+CT
13 (48.1%)
14 (51.9%)
0.056
2.154 (0.966–4.802)
0.159
1.840 (0.787–4.301)
GG
TT
1 (16.7%)
5 (83.3%)
0.004
10.00 (1.149–87.06)
0.041
9.780 (1.099–87.069)
Unfavorable combinationsa
34 (48.6%)
36 (51.4%)
0.006
2.118 (1.233–3.637)
0.005
2.257 (1.273–4.002)
aUnfavorable combination genotypes included rs1130609 GG genotype or rs3213150 TT genotype
bAdjusted by age, risk stratification, WBC count, and serum LDH level

Influence of candidate SNPs on RFS and OS for AML patients

Univariate analysis showed significant differences in RFS among genotypes of NME1 rs3760468, NME1 rs2302254, and NME2 rs3744660 (Table 5, Fig. 3). All the three SNPs were associated with worse RFS in a recessive model (rs3760468 TT vs AA+AT: HR = 1.752, 95% CI 1.142–2.686, P = 0.009; rs2302254 TT vs CC+CT: HR = 1.912, 95% CI 1.117–3.272, P = 0.016; rs3744660 AA vs GG+AG: HR = 2.087, 95% CI 1.051–4.145, P = 0.036). The RRM2 rs1130609 GG genotype also showed marginally worse RFS (GG vs TT+GT: HR = 1.577, 95% CI 0.935–2.660, P = 0.085).
Table 5
Univariate and multivariate Cox regression analysis of SNPs associated with AML relapse-free survival (RFS)
Genotype
n
Mean ± SE (day)
Median (range, day)
HR (95% CI)
P
HR (95% CI)a
P a
NME1 rs3760468
 AA
113
1058 ± 202
751 (294–1208)
1.00 (reference)
 
1.00 (reference)
 
 AT
121
1163 ± 212
894 (378–1410)
0.946 (0.657–1.362)
0.764
1.027 (0.709–1.488)
0.887
 TT
39
655 ± 207
357 (285–429)
1.703 (1.070–2.709)
0.025
1.744 (1.085–2.801)
0.022
 AA+AT
234
1110 ± 149
761 (379–1143)
1.00 (reference)
 
1.00 (reference)
 
 TT
39
655 ± 207
357 (285–429)
1.752 (1.142–2.686)
0.009
1.854 (1.197–2.871)
0.006
NME1 rs2302254
 CC
169
941 ± 129
724 (4484–964)
1.00 (reference)
 
1.00 (reference)
 
 CT
87
1324 ± 266
1282 (516–2048)
0.770 (0.526–1.126)
0.177
0.728 (0.494–1.071)
0.107
 TT
22
581 ± 253
373 (286–460)
1.756 (1.014–3.042)
0.044
1.688 (0.973–2.928)
0.062
 CC+CT
256
1111 ± 156
760 (398–1122)
1.00 (reference)
 
1.00 (reference)
 
 TT
22
581 ± 253
373 (286–460)
1.912 (1.117–3.272)
0.016
1.878 (1.096–3.217)
0.022
NME2 rs3744660
 GG
191
987 ± 124
751 (397–1105)
1.00 (reference)
 
1.00 (reference)
 
 AG
71
1239 ± 283
976 (186–1766)
0.929 (0.627–1.377)
0.714
0.958 (0.644–1.425)
0.832
 AA
12
382 ± 116
373 (95–651)
2.087 (1.051–4.145)
0.036
2.224 (1.114–4.444)
0.024
 GG+AG
262
1136 ± 157
760 (399–1121)
1.00 (reference)
 
1.00 (reference)
 
 AA
12
382 ± 116
373 (95–651)
2.127 (1.078–4.194)
0.026
2.250 (1.135–4.460)
0.020
RRM2 rs1130609
 TT
123
1126 ± 208
751 (368–1134)
1.00 (reference)
 
1.00 (reference)
 
 GT
125
1129 ± 216
724 (209–1239)
1.561 (0.692–1.386)
0.907
0.926 (0.651–1.317)
0.668
 GG
29
679 ± 272
371 (200–542)
1.561 (0.900–2.707)
0.113
1.437 (0.825–2.501)
0.200
 TT+GT
248
1127 ± 159
729 (381–1077)
1.00 (reference)
 
1.00 (reference)
 
 GG
29
679 ± 272
371 (200–542)
1.577 (0.935–2.660)
0.085
1.493 (0.882–2.528)
0.136
aAdjusted for risk stratification, WBC count, and allo-SCT
Multivariate analysis indicated that risk stratification, age, allo-SCT, WBC counts and serum LDH levels were associated with RFS of AML patients (Additional file 2: Table S2). When adjusted by these risk factors, NME1 rs3760468 TT, NME1 rs2302254 TT, and NME2 rs3744660 GG genotypes were still associated with worse RFS (rs3760468 TT vs AA+AT: adjusted HR = 1.854, 95% CI 1.197–2.871, P = 0.006; rs2302254 TT vs CC+CT: adjusted HR = 1.878, 95% CI 1.096–3.217, P = 0.022; rs3744660 AA vs GG+AG: adjusted HR = 2.250, 95% CI 1.135–4.460, P = 0.020).
Results of univariate analysis showed that the NME1 rs3760468, NME2 rs3744660, and RRM1 rs183484 polymorphisms were associated with OS of AML patients significantly (Table 6, Fig 4). As compared with rs3760468 AA homozygotes, rs3760468 TT homozygotes showed significantly worse OS (TT vs AA, HR = 1.652, 95% CI 1.067–2.557, P = 0.024) and carriers of the rs3760468 T allele showed a trend of worse OS (TT+AT vs AA: HR = 1.328, 95% CI 0.973–1.812, P = 0.073). For the rs3744660 polymorphisms, the rare AA homozygotes showed significantly worse OS (HR = 2.214, 95% CI 1.154–4.247, P = 0.017). For the RRM1 rs183484 polymorphism, the GT heterozygotes showed significantly better OS than the wild-type GG homozygotes (HR = 0.692, 95% CI 0.496–0.964, P = 0.030). Marginally significant difference in OS between the RRM2 rs1130609 GG and GT+TT genotypes was also observed (HR = 1.516, 95% CI 0.948–2.422, P = 0.080, Table 6).
Table 6
Univariate and multivariate Cox regression analysis of SNPs associated with AML overall survival (OS)
Genotype
n
Mean ± SE (day)
Median (range) (day)
HR (95% CI)
P
HR (95% CI)a
P a
NME1 rs3760468
 AA
135
1326 ± 209
1534 (947–2121)
1.00 (reference)
 
1.00 (reference)
 
 AT
160
1232 ± 218
768 (388–1148)
1.238 (0.890–1.724)
0.205
1.455 (1.015–2.087)
0.041
 TT
47
801 ± 231
513 (398–638)
1.652 (1.067–2.557)
0.024
1.702 (1.072–2.704)
0.024
 AT+TT
207
1159 ± 185
663 (442–884)
1.328 (0.973–1.812)
0.073
1.518 (1.082–2.129)
0.016
NME2 rs3744660
 GG
230
1283 ± 168
1159 (615–1703)
1.00 (reference)
 
1.00 (reference)
 
 AG
98
1175 ± 252
817 (410–1224)
1.283 (0.924–1.780)
0.137
1.181 (0.831–1.679)
0.353
 AA
15
442 ± 149
359 (167–551)
2.214 (1.154–4.247)
0.017
2.284 (1.145–4.559)
0.019
 GG+AG
328
1261 ± 143
988 (682–1294)
1.00 (reference)
 
1.00 (reference)
 
 AA
15
442 ± 149
359 (167–511)
2.054 (1.080–3.905)
0.025
2.172 (1.098–4.294)
0.026
RRM1 rs183484
 GG
109
940 ± 167
672 (454–890)
1.00 (reference)
 
1.00 (reference)
 
 GT
194
1506 ± 202
1749 (799–2699)
0.692 (0.496–0.964)
0.030
0.599 (0.413–0.868)
0.007
 TT
46
993 ± 305
414 (162–666)
1.112 (0.717–1.726)
0.635
1.063 (0.665–1.698)
0.799
RRM2 rs1130609
 TT
148
1243 \± 201
1025 (426–1624)
1.00 (reference)
 
1.00 (reference)
 
 GT
161
1380 ± 209
843 (520–1166)
0.988 (0.719–1.357)
0.941
1.025 (0.746–1.408)
0.881
 GG
36
753 ± 248
473 (137–809)
1.506 (0.917–2.475)
0.106
1.489 (0.906–2.448)
0.116
 TT+GT
309
1297 ± 153
935 (645–1225)
1.00 (reference)
 
1.00 (reference)
 
 GG
36
753 ± 248
473 (137–809)
1.516 (0.948–2.422)
0.080
1.471 (0.920–2.352)
0.107
aAdjusted for risk stratification, WBC count, allo-SCT, age, and serum LDH level
Results from multivariate analysis indicated that risk stratification, age, allo-SCT, WBC counts and serum LDH levels were associated with OS of AML patients (Additional file 2: Table S2). When adjusted by these OS risk factors, rs3760468, rs3744660, and rs183484 polymorphisms were still associated with OS risk. As compared with rs3760468 AA homozygotes, rs3760468 TT homozygotes or carriers of the rs3760468 T allele showed significantly worse OS (TT+AT vs AA: adjusted HR = 1.518, 95% CI 1.082–2.129, P = 0.016). For the rs3744660 polymorphisms, the rare AA homozygotes showed significantly worse OS (adjusted HR = 2.284, 95% CI 1.145–4.559, P = 0.019). For the RRM1 rs183484 polymorphism, the GT heterozygotes showed significantly longer OS than the wild-type GG homozygotes (HR = 0.599, 95% CI 0.413–0.868, P = 0.007).

Discussion

In this study, we observed the associations of 12 SNPs in 7 genes involved in Ara-C and dNTP metabolic pathway with chemosensitivity to Ara-C based therapy as well as disease prognosis in Chinese AML patients. We observed that 3 SNPs including NME2 rs3744660 (dominant model), E2F1 rs3213150 (recessive model) and RRM2 rs1130609 (recessive model) were associated with increased risk for non-CR after Ara-C based therapy, while the SAMHD1 rs6102991 polymorphism showed a tendency with decreased risk of non-CR. We also observed a combined effect of E2F1 and RRM2 polymorphisms on CR rate. In addition, we observed that the NME1 rs3760468 and rs2302254, and NME2 rs3744660 polymorphisms predicted worse RFS, while NME1 rs3760468, NME2 rs3744660, and RRM1 rs183484 predicted worse OS in the AML patients.
For the 7 genes involved in our study, 3 genes (CMPK1, NME1 and NME2) were involved in the metabolic activation of Ara-C, while 4 genes (SAMHD1, E2F1, RRM1 and RRM2) were involved in the Ara-C inactivation or DCK feedback inhibition (dNTP/dCTP synthesis) pathways. Four genes (CMPK1, NME2, SAMHD1 and E2F1) were studied for the first time in our study. The majority of the SNPs included in our study were eQTL variants. For the 4 SNPs associated with CR rate observed in our study, difference in mRNA expression among genotypes of NME2 rs3744660 (P = 2.5e−12), RRM2 rs1130609 (P = 1.2e−7), and SAMHD1 rs6102991 (P = 2.0e−6) were indicated by eQTL analysis, and 2 of these SNPs (NME2 rs3744660, SAMHD1 rs6102991) were reported for the first time in our study.
NME1 (alias DNPK-A) and NME2 (alias DNPK-B) are two most important members of the NDPK family of proteins and mediate phosphorylation of ara-CDP into ara-CTP. The coding genes NME1 and NME2 are closely neighbored on chromosome 17. We observed that carriers of the NME2 intronic variant rs3744660 was associated with decreased NME2 mRNA expression shown by eQTL analysis, which indicated decreased DNPK activity and ara-CTP formation, this is in agreement with the observation of decreased CR rate in patients carrying the rs3744660 A allele or worse prognosis in AA genotyped patients. Few studies have focused on NME1NME2 locus presently. NME1 is initially identified for its metastatic suppressive potential for cancer cells [28]. Qu et al. reported that the NME1 promoter SNPs rs2302254 and rs3760468 can alter nuclear proteins binding capacity and reduce NME1 promoter activity by about 20%, which may account for increased breast cancer mortality for these SNPs in Chinese [29]. In a study by Braunagel et al. the rs2302254 TT genotype (mutant homozygotes) was an independent risk factor for Ara-C associated neurotoxicity but not for OS or RFS in AML patients [26]. However, neither rs2302254 nor rs3760468 was associated with CR rate in our study, though both polymorphisms can results in decreased NME1 mRNA expression and predicted worse prognosis indicated by RFS and/or OS in our study. We assume that the positive findings in AML prognosis could also be explained, at least partially, by the direct functions of NME1/NME2, both can stimulate the growth and survival of AML cells through activation of the MAPK and transducers and activators of transcription signaling pathways [30, 31]. Of note, the rs3744660 is located in intron 4 of NME2 (NM_001018137.2) and intron 7 of the NME1NME2 transcript (NM_001018136.2), the exact function of this SNP remains to be explored.
The RRM2 rs1130609 polymorphism is a non-synonymous SNP that leads to amino acid change from Ser59 to Ala59. We found that the rare GG genotype showed increased RRM2 mRNA expression as indicated by eQTL analysis, which may suggest increased dNTP synthesis and increased feedback inhibition on DCK activity in these patients. This is in accordance with the findings of increased non-CR risk and marginally worse prognosis in AML patients with the GG genotype in our study. In consistent with our study, higher RRM1/RRM2 mRNA expression was observed to be associated with decreased Ara-C sensitivity in the HapMap YRI samples (30 trios). Our findings are also in agreement with a previous study by Cao et al. who reported that the rs1130609 polymorphism was associated with decreased Ara-C cytotoxicity in the HapMap CEU samples and worse EFS in Caucasian AML patient cohorts [18]. The exact function of the SNP remains unknown despite its influence on RRM2 mRNA expression. Search of the dbSNP database shows that the rs1130609 polymorphism is also a 5′-UTR SNP for the RRM2 transcript NM_001034.3. Further functional study of this SNP is required. Cao et al. reported significant associations of several RRM1 SNPs in the regulatory regions (e.g. rs2412344, rs11030907, rs7929397, rs2898950, rs1042919, and rs2412344) with ex vivo Ara-C cytotoxicity and/or in vivo Ara-C response [18]. A recent study in Chinese AML patients also observed association of the 3′-UTR potential miRNA binding site SNP rs1042919 with decreased CR rate but not disease prognosis in AML patients [32]. However, we failed to observed associations between the two RRM1 SNPs rs2412344 (in promoter) and rs183484 (Arg284Arg) and risk of non-CR, though significant differences in mRNA expression among the genotypes of both SNPs were indicated by eQTL analysis. We observed that the rs183484 GT genotype showed better OS (HR = 0.599, 95% CI 0.413–0.868, P = 0.007). The discrepancies among the studies should be further validated in future studies.
E2F1 is a newly identified risk gene for Ara-C response in our study. We selected this gene based on a recent report of ERK/E2F1 signaling in regulating RRM2 expression, dCTP pool and gemcitabine sensitivity [17]. The E2F1 rs3213150 polymorphism is an intronic and tag SNP selected in our study. Interesting, we observed significant association of this SNP with CR rate after Ara-C based induction therapy. In addition, a combined effect of the E2F1 rs3213150 and RRM2 rs1130609 polymorphisms on CR rate is observed. The CR rate decreased with the number of unfavorable genotypes carried at both loci (66.7% in carriers of both favorable genotypes, 51.6% in carriers of one unfavorable genotype, and 16.6% in carriers of unfavorable genotypes for both SNPs), and unfavorable genotypes at both loci results in about ninefold increase in non-CR risk after Ara-C based chemotherapy (OR = 9.780, 95% CI 1.099–87.069, P = 0.041). Our study suggest the importance of concomitant consideration of genetic variants of both E2F1 and RRM2 in evaluation of Ara-C response in AML patients.
SAMHD1 is a recently identified ara-CTPase that can hydrolyze and detoxify intracellular ara-CTP [12]. Inhibition of the enzyme or CRISPR–Cas9-mediated disruption of SAMHD1 can sensitize AML cells to Ara-C [13]. In our study, we analyzed associations of three SAMHD1 SNPs (rs6102991 in the promoter, rs28372906 in 5′-UTR, and rs6029941 in 3′-UTR) with drug response and AML prognosis. We observed that carriers of the rs6102991 G allele showed a tendency with decreased risk of non-CR after Ara-C based therapy in a dominant model (OR = 0.611, 95% CI 0.369–1.013, P = 0.055). The rs6102991 polymorphism is located at − 4395 upstream transcription start site (T-4395C). eQTL analysis indicates that the rs6102991 polymorphism results in decreased SAMHD1 mRNA expression, which supports our clinic findings. As the SNPs is far from the transcription start site, our findings indicate potential influence of rs6102991 or other variants in high LD with it on Ara-C response through affecting SAMHD1 mRNA expression. Few studies have focused on functions of rare SAMHD1 coding variations on HIV infection and replication [33, 34], however, there is still a gap between SAMHD1 variations and the clinical relevance. Because SNPs information for the SAMHD1 promoter is very limited in the SNP database available, further study is required to explore the potentially causative variant(s).
There were some limitations in our study. For instance, the outcomes of our study failed to undergo multiple test adjusting, some P values lost statistical significance when Bonferroni correction was performed possibly due to limited sample size included in our study. In addition, Ara-C response is affected by various genetic factors, the contribution of any unique gene in the drug response might be limited. And due to the limitations of the Massarray genotyping system, we failed to genotype some of the important SNPs such as RRM1rs1561876 [18]. For the AML patients involved in our study, genotype distribution for 4 SNPs (CMPK1 rs7543016, SAMHD1 rs28372906, RRM1 rs183484, and RRM2 rs1130609) were not in Hardy–Weinberg equilibrium, these may partially be explained by the internal function of the genes in etiology of AML rather than in Ara-c response. We suppose that these polymorphisms may also act as susceptibility genes for AML, which may account for the abnormal distribution of the genotype for these SNPs in the AML patients. Of course, this should be confirmed in further study.

Conclusions

Our current study demonstrated that SNPs in genes involved in Ara-C metabolic pathway are associated with drug response to Ara-C based induction therapy and prognosis of AML in Chinese patients, though further confirmatory studies and functional evaluations of the newly identified SNPs are needed to further support our findings. In addition, we suggest consideration of combined genotypes in the Ara-C metabolic pathway in evaluation of the associations between genetic variations and drug response in AML. Our findings provide insightful information for the understanding of individual difference in drug response and potential biomarkers for identification of patients with increased risk of chemoresistance or poor prognosis.

Authors’ contributions

ZKW made main contributions to read document and design research, specimen collection, sorting date, date analysis and drafting manuscript; CP, ZDY, YH and LYL were assisting in specimen collection and acquisition of clinical date; LH and CLN were mainly responsible for follow-up; and CXP provided substantial guidance and revised manuscript; CS and ZG given some useful advice; ZH, CSP, ZXL helped to collect specimen smoothly. All authors read and approved the final manuscript.

Acknowledgements

The authors would like to thank Prof. Hui Zeng, Shu-ping Chen, Xie-lan Zhao for unselfish assistance with colleting specimen and acquiring clinical date.

Competing interests

The authors declare that they have no competing interests.

Availability of date and materials

The datasets supporting the conclusions of this article are included within the article (and its additional files).
Not applicable.
This study was approved by the Ethics Committee of Institute of Clinical Pharmacology, Central South University (Registration Number: CTXY-120025-2). Clinical study admission (Registration Number: ChiCTR-PPC-14005297) was approved by the Chinese Clinical Trial Register and informed consent was obtained from each patient or their family members.

Funding

This project was supported by Chinese National Science Foundation (Nos. 81673518, 81422052), the National key R&D program (No. 2017YFC0909302).

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
2.
Zurück zum Zitat Dohner H, Estey EH, Amadori S, Appelbaum FR, Buchner T, Burnett AK, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115:453–74.CrossRefPubMed Dohner H, Estey EH, Amadori S, Appelbaum FR, Buchner T, Burnett AK, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115:453–74.CrossRefPubMed
3.
Zurück zum Zitat Wang JJ, Selawry OS, Vietti TJ, Bodey GP Sr. Prolonged infusion of arabinosyl cytosine in childhood leukemia. Cancer. 1970;25:1–6.CrossRefPubMed Wang JJ, Selawry OS, Vietti TJ, Bodey GP Sr. Prolonged infusion of arabinosyl cytosine in childhood leukemia. Cancer. 1970;25:1–6.CrossRefPubMed
5.
Zurück zum Zitat Boffo S, Damato A, Alfano L, Giordano A. CDK9 inhibitors in acute myeloid leukemia. J Exp Clin Canc Res. 2018;37:36.CrossRef Boffo S, Damato A, Alfano L, Giordano A. CDK9 inhibitors in acute myeloid leukemia. J Exp Clin Canc Res. 2018;37:36.CrossRef
6.
Zurück zum Zitat Miyawaki S, Ohtake S, Fujisawa S, Kiyoi H, Shinagawa K, Usui N, et al. A randomized comparison of 4 courses of standard-dose multiagent chemotherapy versus 3 courses of high-dose cytarabine alone in postremission therapy for acute myeloid leukemia in adults: the JALSG AML201 Study. Blood. 2011;117:2366–72.CrossRefPubMed Miyawaki S, Ohtake S, Fujisawa S, Kiyoi H, Shinagawa K, Usui N, et al. A randomized comparison of 4 courses of standard-dose multiagent chemotherapy versus 3 courses of high-dose cytarabine alone in postremission therapy for acute myeloid leukemia in adults: the JALSG AML201 Study. Blood. 2011;117:2366–72.CrossRefPubMed
7.
Zurück zum Zitat Wiley JS, Jones SP, Sawyer WH, Paterson AR. Cytosine arabinoside influx and nucleoside transport sites in acute leukemia. J Clin Investig. 1982;69:479–89.CrossRefPubMedPubMedCentral Wiley JS, Jones SP, Sawyer WH, Paterson AR. Cytosine arabinoside influx and nucleoside transport sites in acute leukemia. J Clin Investig. 1982;69:479–89.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Yamauchi T, Negoro E, Kishi S, Takagi K, Yoshida A, Urasaki Y, et al. Intracellular cytarabine triphosphate production correlates to deoxycytidine kinase/cytosolic 5′-nucleotidase II expression ratio in primary acute myeloid leukemia cells. Biochem Pharmacol. 2009;77:1780–6.CrossRefPubMed Yamauchi T, Negoro E, Kishi S, Takagi K, Yoshida A, Urasaki Y, et al. Intracellular cytarabine triphosphate production correlates to deoxycytidine kinase/cytosolic 5′-nucleotidase II expression ratio in primary acute myeloid leukemia cells. Biochem Pharmacol. 2009;77:1780–6.CrossRefPubMed
9.
Zurück zum Zitat Van Rompay AR, Johansson M, Karlsson A. Phosphorylation of nucleosides and nucleoside analogs by mammalian nucleoside monophosphate kinases. Pharmacol Ther. 2000;87:189–98.CrossRefPubMed Van Rompay AR, Johansson M, Karlsson A. Phosphorylation of nucleosides and nucleoside analogs by mammalian nucleoside monophosphate kinases. Pharmacol Ther. 2000;87:189–98.CrossRefPubMed
10.
Zurück zum Zitat Attwood PV, Muimo R. The actions of NME1/NDPK-A and NME2/NDPK-B as protein kinases. Lab Invest. 2018;98:283–90.CrossRefPubMed Attwood PV, Muimo R. The actions of NME1/NDPK-A and NME2/NDPK-B as protein kinases. Lab Invest. 2018;98:283–90.CrossRefPubMed
11.
Zurück zum Zitat Crisp LB, Smith SM, Mathers MA, Young GA, Lyons SD, Christopherson RI. Effects of cytosine arabinoside on human leukemia cells. Int J Biochem Cell Biol. 1996;28:1061–9.CrossRefPubMed Crisp LB, Smith SM, Mathers MA, Young GA, Lyons SD, Christopherson RI. Effects of cytosine arabinoside on human leukemia cells. Int J Biochem Cell Biol. 1996;28:1061–9.CrossRefPubMed
12.
Zurück zum Zitat Schneider C, Oellerich T, Baldauf H, Schwarz SM, Thomas D, Flick R, et al. SAMHD1 is a biomarker for cytarabine response and a therapeutic target in acute myeloid leukemia. Nat Med. 2017;23:250–5.CrossRefPubMed Schneider C, Oellerich T, Baldauf H, Schwarz SM, Thomas D, Flick R, et al. SAMHD1 is a biomarker for cytarabine response and a therapeutic target in acute myeloid leukemia. Nat Med. 2017;23:250–5.CrossRefPubMed
13.
Zurück zum Zitat Herold N, Rudd SG, Ljungblad L, Sanjiv K, Myrberg IH, Paulin CB, et al. Targeting SAMHD1 with the Vpx protein to improve cytarabine therapy for hematological malignancies. Nat Med. 2017;23:256–63.CrossRefPubMed Herold N, Rudd SG, Ljungblad L, Sanjiv K, Myrberg IH, Paulin CB, et al. Targeting SAMHD1 with the Vpx protein to improve cytarabine therapy for hematological malignancies. Nat Med. 2017;23:256–63.CrossRefPubMed
14.
Zurück zum Zitat Whelan J, Smith T, Phear G, Rohatiner A, Lister A, Meuth M. Resistance to cytosine arabinoside in acute leukemia: the significance of mutations in CTP synthetase. Leukemia. 1994;8:264–5.PubMed Whelan J, Smith T, Phear G, Rohatiner A, Lister A, Meuth M. Resistance to cytosine arabinoside in acute leukemia: the significance of mutations in CTP synthetase. Leukemia. 1994;8:264–5.PubMed
15.
Zurück zum Zitat Chimploy K, Tassotto ML, Mathews CK. Ribonucleotide reductase, a possible agent in deoxyribonucleotide pool asymmetries induced by hypoxia. J Biol Chem. 2000;275:39267–71.CrossRefPubMed Chimploy K, Tassotto ML, Mathews CK. Ribonucleotide reductase, a possible agent in deoxyribonucleotide pool asymmetries induced by hypoxia. J Biol Chem. 2000;275:39267–71.CrossRefPubMed
17.
Zurück zum Zitat Xia G, Wang H, Song Z, Meng Q, Huang X, Huang X. Gambogic acid sensitizes gemcitabine efficacy in pancreatic cancer by reducing the expression of ribonucleotide reductase subunit-M2 (RRM2). J Exp Clin Cancer Res. 2017;36:107.CrossRefPubMedPubMedCentral Xia G, Wang H, Song Z, Meng Q, Huang X, Huang X. Gambogic acid sensitizes gemcitabine efficacy in pancreatic cancer by reducing the expression of ribonucleotide reductase subunit-M2 (RRM2). J Exp Clin Cancer Res. 2017;36:107.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Cao X, Mitra AK, Pounds S, Crews KR, Gandhi V, Plunkett W, et al. RRM1 and RRM2 pharmacogenetics: association with phenotypes in HapMap cell lines and acute myeloid leukemia patients. Pharmacogenomics. 2013;14:1449–66.CrossRefPubMed Cao X, Mitra AK, Pounds S, Crews KR, Gandhi V, Plunkett W, et al. RRM1 and RRM2 pharmacogenetics: association with phenotypes in HapMap cell lines and acute myeloid leukemia patients. Pharmacogenomics. 2013;14:1449–66.CrossRefPubMed
19.
Zurück zum Zitat Minkovsky A, Sahakyan A, Bonora G, Damoiseaux R, Dimitrova E, Rubbi L, et al. A high-throughput screen of inactive X chromosome reactivation identifies the enhancement of DNA demethylation by 5-aza-2′-dC upon inhibition of ribonucleotide reductase. Epigenetics Chromatin. 2015;8:42.CrossRefPubMedPubMedCentral Minkovsky A, Sahakyan A, Bonora G, Damoiseaux R, Dimitrova E, Rubbi L, et al. A high-throughput screen of inactive X chromosome reactivation identifies the enhancement of DNA demethylation by 5-aza-2′-dC upon inhibition of ribonucleotide reductase. Epigenetics Chromatin. 2015;8:42.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Megías-Vericat JE, Montesinos P, Herrero MJ, Moscardó F, Bosó V, Martínez-Cuadrón D, et al. Influence of cytarabine metabolic pathway polymorphisms in acute myeloid leukemia induction treatment. Leuk Lymphoma. 2017;58:2880–94.CrossRefPubMed Megías-Vericat JE, Montesinos P, Herrero MJ, Moscardó F, Bosó V, Martínez-Cuadrón D, et al. Influence of cytarabine metabolic pathway polymorphisms in acute myeloid leukemia induction treatment. Leuk Lymphoma. 2017;58:2880–94.CrossRefPubMed
21.
Zurück zum Zitat Yan H, Zhang DY, Li X, Yuan XQ, Yang YL, Zhu KW, et al. Long non-coding RNA GAS5 polymorphism predicts a poor prognosis of acute myeloid leukemia in Chinese patients via affecting hematopoietic reconstitution. Leuk Lymphoma. 2017;58:1948–57.CrossRefPubMed Yan H, Zhang DY, Li X, Yuan XQ, Yang YL, Zhu KW, et al. Long non-coding RNA GAS5 polymorphism predicts a poor prognosis of acute myeloid leukemia in Chinese patients via affecting hematopoietic reconstitution. Leuk Lymphoma. 2017;58:1948–57.CrossRefPubMed
22.
Zurück zum Zitat Zhang DY, Yan H, Cao S, Zhang W, Li XL, Zeng H, et al. Wilms tumor 1 rs16754 predicts favorable clinical outcomes for acute myeloid leukemia patients in South Chinese population. Leuk Res. 2015;39:568–74.CrossRefPubMed Zhang DY, Yan H, Cao S, Zhang W, Li XL, Zeng H, et al. Wilms tumor 1 rs16754 predicts favorable clinical outcomes for acute myeloid leukemia patients in South Chinese population. Leuk Res. 2015;39:568–74.CrossRefPubMed
23.
Zurück zum Zitat Zhang DY, Yuan XQ, Yan H, Cao S, Zhang W, Li XL, et al. Association between DCK 35708 T>C variation and clinical outcomes of acute myeloid leukemia in South Chinese patients. Pharmacogenomics. 2016;17:1519–31.CrossRefPubMed Zhang DY, Yuan XQ, Yan H, Cao S, Zhang W, Li XL, et al. Association between DCK 35708 T>C variation and clinical outcomes of acute myeloid leukemia in South Chinese patients. Pharmacogenomics. 2016;17:1519–31.CrossRefPubMed
24.
Zurück zum Zitat Yuan XQ, Zhang DY, Yan H, Yang YL, Zhu KW, Chen YH, et al. Evaluation of DNMT3A genetic polymorphisms as outcome predictors in AML patients. Oncotarget. 2016;7:60555–74.PubMedPubMedCentral Yuan XQ, Zhang DY, Yan H, Yang YL, Zhu KW, Chen YH, et al. Evaluation of DNMT3A genetic polymorphisms as outcome predictors in AML patients. Oncotarget. 2016;7:60555–74.PubMedPubMedCentral
25.
Zurück zum Zitat Zhu GZ, Yang YL, Zhang YJ, Liu W, Li MP, Zeng WJ, et al. High expression of AHSP, EPB42, GYPC and HEMGN predicts favorable prognosis in FLT3-ITD-negative acute myeloid leukemia. Cell Physiol Biochem. 2017;42:1973–84.CrossRefPubMed Zhu GZ, Yang YL, Zhang YJ, Liu W, Li MP, Zeng WJ, et al. High expression of AHSP, EPB42, GYPC and HEMGN predicts favorable prognosis in FLT3-ITD-negative acute myeloid leukemia. Cell Physiol Biochem. 2017;42:1973–84.CrossRefPubMed
26.
Zurück zum Zitat Yuan XQ, Peng L, Zeng WJ, Jiang BY, Li GC, Chen XP. DNMT3A R882 mutations predict a poor prognosis in AML: a meta-analysis from 4474 patients. Medicine (Baltimore). 2016;95:e3519.CrossRefPubMedPubMedCentral Yuan XQ, Peng L, Zeng WJ, Jiang BY, Li GC, Chen XP. DNMT3A R882 mutations predict a poor prognosis in AML: a meta-analysis from 4474 patients. Medicine (Baltimore). 2016;95:e3519.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Braunagel D, Schaich M, Kramer M, Dransfeld C, Ehninger G, Mahlknecht U. The T_T genotype within the NME1 promoter single nucleotide polymorphism − 835 C/T is associated with an increased risk of cytarabine induced neurotoxicity in patients with acute myeloid leukemia. Leuk Lymphoma. 2012;53:952–7.CrossRefPubMed Braunagel D, Schaich M, Kramer M, Dransfeld C, Ehninger G, Mahlknecht U. The T_T genotype within the NME1 promoter single nucleotide polymorphism − 835 C/T is associated with an increased risk of cytarabine induced neurotoxicity in patients with acute myeloid leukemia. Leuk Lymphoma. 2012;53:952–7.CrossRefPubMed
28.
Zurück zum Zitat Horak CE, Lee JH, Elkahloun AG, Boissan M, Dumont S, Maga TK, et al. Nm23-H1 suppresses tumor cell motility by down-regulating the lysophosphatidic acid receptor EDG2. Cancer Res. 2007;67:7238–46.CrossRefPubMed Horak CE, Lee JH, Elkahloun AG, Boissan M, Dumont S, Maga TK, et al. Nm23-H1 suppresses tumor cell motility by down-regulating the lysophosphatidic acid receptor EDG2. Cancer Res. 2007;67:7238–46.CrossRefPubMed
29.
Zurück zum Zitat Qu S, Long J, Cai Q, Shu XO, Cai H, Gao YT, et al. Genetic polymorphisms of metastasis suppressor gene NME1 and breast cancer survival. Clin Cancer Res. 2008;14:4787–93.CrossRefPubMedPubMedCentral Qu S, Long J, Cai Q, Shu XO, Cai H, Gao YT, et al. Genetic polymorphisms of metastasis suppressor gene NME1 and breast cancer survival. Clin Cancer Res. 2008;14:4787–93.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Yokoyama A, Okabe-Kado J, Sakashita A, Maseki N, Kaneko Y, Hino K, et al. Differentiation inhibitory factor nm23 as a new prognostic factor in acute monocytic leukemia. Blood. 1996;88:3555–61.PubMed Yokoyama A, Okabe-Kado J, Sakashita A, Maseki N, Kaneko Y, Hino K, et al. Differentiation inhibitory factor nm23 as a new prognostic factor in acute monocytic leukemia. Blood. 1996;88:3555–61.PubMed
31.
Zurück zum Zitat Okabe-Kado J, Kasukabe T, Honma Y, Kobayashi H, Maseki N, Kaneko Y. Extracellular NM23 protein promotes the growth and survival of primary cultured human acute myelogenous leukemia cells. Cancer Sci. 2009;100:1885–94.CrossRefPubMed Okabe-Kado J, Kasukabe T, Honma Y, Kobayashi H, Maseki N, Kaneko Y. Extracellular NM23 protein promotes the growth and survival of primary cultured human acute myelogenous leukemia cells. Cancer Sci. 2009;100:1885–94.CrossRefPubMed
32.
Zurück zum Zitat Cao HX, Miao CF, Yan L, Tang P, Zhang LR, Sun L. Polymorphisms at microRNA binding sites of Ara-C and anthracyclines-metabolic pathway genes are associated with outcome of acute myeloid leukemia patients. J Transl Med. 2017;15:235.CrossRefPubMedPubMedCentral Cao HX, Miao CF, Yan L, Tang P, Zhang LR, Sun L. Polymorphisms at microRNA binding sites of Ara-C and anthracyclines-metabolic pathway genes are associated with outcome of acute myeloid leukemia patients. J Transl Med. 2017;15:235.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Rentoft M, Lindell K, Tran P, Chabes AL, Buckland RJ, Watt DL, et al. Heterozygous colon cancer-associated mutations of SAMHD1 have functional significance. Proc Natl Acad Sci USA. 2016;113:4723–8.CrossRefPubMedPubMedCentral Rentoft M, Lindell K, Tran P, Chabes AL, Buckland RJ, Watt DL, et al. Heterozygous colon cancer-associated mutations of SAMHD1 have functional significance. Proc Natl Acad Sci USA. 2016;113:4723–8.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat He DD, Mao SL, Wang W, Jin YB, Zhang HL, Ling F. SAMHD1 polymorphisms were significantly correlated with HIV/SIV virus load in PBMC from Chinese rhesus macaques and cynomolgus macaques. Curr HIV Res. 2017;15:15–22.CrossRefPubMed He DD, Mao SL, Wang W, Jin YB, Zhang HL, Ling F. SAMHD1 polymorphisms were significantly correlated with HIV/SIV virus load in PBMC from Chinese rhesus macaques and cynomolgus macaques. Curr HIV Res. 2017;15:15–22.CrossRefPubMed
Metadaten
Titel
Association of genetic polymorphisms in genes involved in Ara-C and dNTP metabolism pathway with chemosensitivity and prognosis of adult acute myeloid leukemia (AML)
verfasst von
Ke-Wei Zhu
Peng Chen
Dao-Yu Zhang
Han Yan
Han Liu
Li-Na Cen
Yan-Ling Liu
Shan Cao
Gan Zhou
Hui Zeng
Shu-Ping Chen
Xie-Lan Zhao
Xiao-Ping Chen
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2018
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-018-1463-1

Weitere Artikel der Ausgabe 1/2018

Journal of Translational Medicine 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.