Concentrative nucleoside transporter 3 as a prognostic indicator for favorable outcome of t(8;21)-positive acute myeloid leukemia patients after cytarabine-based chemotherapy

  • Authors:
    • Ju Han Song
    • Kyung-Min Cho
    • Hyeoung-Joon Kim
    • Yeo-Kyeoung Kim
    • Nan Young Kim
    • Hee-Je Kim
    • Tae-Hyang Lee
    • Seung Yong Hwang
    • Tae Sung Kim
  • View Affiliations

  • Published online on: May 7, 2015     https://doi.org/10.3892/or.2015.3959
  • Pages: 488-494
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Although acute myeloid leukemia (AML) exhibits diverse responses to chemotherapy, patients harboring the t(8;21) translocation are part of a favorable risk group. However, the reason why this subgroup is more responsive to cytarabine-based therapy has not been elucidated. In the present study, we analyzed expression levels of cytarabine metabolism-related genes in patients diagnosed with AML with or without t(8;21) and investigated their correlation with clinical outcomes after cytarabine-based therapy. Among the 8 genes studied, expression of the concentrative nucleoside transporter 3 (CNT3) gene was significantly higher in t(8;21)-positive patients compared to the others in the test population and the validation cohort (P<0.001 in Mann-Whitney U test; P<0.002 in Pearson's correlation analysis). Additionally, in both multivariate and univariate analyses, t(8;21)-positive patients categorized in a higher CNT3 expression tertile had longer disease-free survival [hazard ratio (HR), 0.117; 95% confidence interval (CI), 0.025-0.557; P=0.008] and overall survival (HR, 0.062; 95% CI, 0.007-0.521; P=0.010) compared to t(8;21)-positive patients in a lower CNT3 expression tertile. Notably, these trends did not occur in t(8;21)-negative patients. Our results demonstrate that CNT3 expression is associated with overall favorable outcomes and is predictive of clinical outcomes in AML patients with t(8;21). This suggests that CNT3 expression can be used to optimize treatment strategies for AML patients.

Introduction

Acute myeloid leukemia (AML) is a highly heterogeneous hematologic malignancy that displays diverse responses to chemotherapy. Although a number of clinical factors affect treatment outcomes, the cytogenetic features of AML are generally accepted as strong predictors of therapeutic response (1). Pediatric and adult patients carrying the t(8;21) chromosomal translocation, which is one of the most frequent AML subtypes, are part of a favorable risk group (2). Although these patients are initially highly responsive to treatment, a number of patients relapse and fail to achieve long-term disease-free survival (DFS) (35). However, little is currently known concerning the functional mechanism by which t(8;21) is associated with both a higher remission rate in some patients, and occasionally with poor outcomes in others, except in cases that are characterized by other factors such as distinctive immunophenotypical changes and multidrug resistance genes (68).

Cytarabine is a nucleoside analog that is intensively used for the treatment of AML (911). Because of its hydrophilic nature, cytarabine can only enter cells via nucleoside transporters such as concentrative nucleoside transporters (CNT1/2/3) and equilibrative nucleoside transporters (ENT1/2) (12,13). Cytarabine must also be converted to its active form by specific enzymes such as deoxycytidine kinase (dCK) (14). Several groups have reported that low gene expression and protein activity of these factors confers resistance to nucleoside drugs of leukemia cells in in vivo and in vitro studies (1518). Alternatively, activation of certain components of the metabolic machinery such as cytidine deaminase (CDA), ecto-5′-nucleotidase (CD73) and 5′,3′-nucleotidase (NT5C), may cause resistance to cytarabine therapy (1922). Microarray analysis of an in vitro gemcitabine-resistant model revealed upregulation of ribonucleotide reductase M subunits (RRM1/2) as part of the intracellular detoxification process (23). Nevertheless, the prognostic implications of changes in the expression of these genes have not been elucidated in specific subpopulations, such as in AML patients with t(8;21).

In the present study, we hypothesized that specific genes that are closely involved in the antitumor action of cytarabine may be associated with the clinical response to treatment of AML patients carrying t(8;21). Therefore, we attempted to identify a novel marker that was predictive of outcomes of patients carrying the t(8;21) abnormality and evaluated the prognostic impact of the candidate genes.

Materials and methods

Patient samples

Bone marrow (BM) mononuclear cells (MNCs) from adults newly diagnosed with AML and 4 healthy donors were collected at Chonnam National University Hwasun Hospital and Catholic University Seoul St. Mary’s Hospital. Written informed consent for the cryopreservation and use of the samples for further research were obtained from the patients. The Institutional Review Board approved all research on the human subjects participating in the present study. The 54 individuals in the test study and the 44 individuals in the validation study had M2 subtype disease according to the French-American-British (FAB) classification criteria; their clinical information is summarized in Table I. In the test cohort, 16 patients had normal karyotypes, 25 patients had the cytogenetically t(8;21) abnormality, and 13 patients had diverse chromosomal abnormalities. In the validation cohort, 15 patients had normal karyotypes, 19 patients had t(8;21), and 10 patients had other abnormalities. Intensive remission induction therapy was executed by administration of the combination of 12 mg/m2/day idarubicin for 3 days with 100 mg/m2/day cytarabine for 7 days or N4-behenoyl-1-D-arabinofuranosyl cytosine (BH-AC) at either 300 mg/m2/day for patients younger than 40 years or 200 mg/m2/day for patients older than 40 years. Four patients received cytarabine alone at a dose of 100 mg/m2/day; one patient, daunorubicin plus cytarabine; one patient, cytarabine plus etoposide. Patients who failed to achieve complete remission (CR) after the first round of induction chemotherapy received re-induction chemotherapy with the same regimen. Patients achieving a CR received 3 courses of high-dose Ara-C (3 g/m2 every 12 h/day on days 1, 3 and 5) or the combination of idarubicin and BH-AC for consolidation therapy. Thirty-three patients in the test cohort and 13 patients in the validation cohort received hematopoietic stem cell transplantation (HSCT).

Table I

Patient samples.

Table I

Patient samples.

CategoryTestValidation
Patients, n5444
Age, median years (range) Gender, n (%)41 (20–72)49 (17–73)
 Male28 (51.9)26 (59.1)
 Female26 (48.1)18 (40.9)
Blasts, mean % (range)50 (2–92)60 (20–91)
M2 (FAB), n5444
Cytogenetics, n
 t(8;21)2519
 Normal1615
 Others1310
RNA preparation and real-time polymerase chain reaction (real-time PCR)

Total RNA was extracted from the BM-MNCs of patients using the Qiagen RNA isolation kit (Qiagen, Venlo, The Netherlands) and converted to cDNA with a reverse transcription kit (Invitrogen, Carlsbad, CA, USA). The cDNA was mixed with SYBR-Green PCR Master Mix (PE Applied Biosystems, Foster City, CA, USA) and specific primers for 8 genes including CD73, CDA, NT5C, dCK, CNT3, ENT1, RRM1 and RRM2. Amplification reactions were performed in triplicate using the ABI Prism 7900 Sequence Detection system (PE Applied Biosystems). GAPDH served as an experimental control. Primer sequences, PCR conditions, and data processing were previously described (24). In the validation study, the expression levels of the membrane transporters, CNT1, CNT2, CNT3, ENT1 and ENT2, were evaluated by the method described above. However, we excluded the results for CNT1 and CNT2 from further analysis because they had low or undetectable signals that were not as reliable.

Survival estimate and correlation analyses

The study population was separated based on the cytogenetic abnormalities of the patients. For the purpose of statistical analysis of the target gene, the patients were also subdivided into 3 groups (low, intermediate and high) according to candidate gene expression tertiles. DFS was defined as the time from first remission to relapse or death. Overall survival (OS) was defined as the time from diagnosis to the date of death or last followup. Kaplan-Meier estimates were used to construct survival curves. Statistical differences in the treatment outcomes were determined by the univariate analyses with log-rank test and Cox proportional hazards univariate model. In multivariate analysis, a backward stepwise method of Cox proportional hazards model was used to investigate the association of clinical variables and gene expression pattern with DFS and OS and to assess hazard ratio (HR) with 95% confidence interval (CI). Correlations between clinical and molecular features were obtained by Pearson’s correlation statistics and represented as a correlation coefficient (r). All statistical analyses were performed using SPSS version 12.0 (SPSS, Inc., Chicago, IL, USA).

Results

Prolonged survival in patients carrying t(8;21)

We collected BM-MNCs from 54 newly diagnosed patients with M2 subtype AML according to the FAB classification who received cytarabine-based chemotherapy. Although it is well established that AML patients with the t(8;21) abnormality respond better to chemotherapy than patients with normal cytogenetics or other cytogenetic abnormalities (1,2,25), we confirmed this in our study population. In a univariate analysis, Kaplan-Meier estimates showed longer survival rates in terms of DFS (P=0.028 in log-rank; P=0.037 in Cox model) and OS (P=0.029 in log-rank; P=0.034 in Cox model) in the t(8;21)-positive AML cohort compared to the t(8;21)-negative group (Fig. 1).

Elevated CNT3 expression in AML patients with t(8;21)

To explore possible pathological reasons for the better response to cytarabine-based therapy in t(8;21)-positive patients, we examined the mRNA expression levels of 8 candidate genes in a total of 54 AML patients. These genes included CD73, CDA, NT5C, dCK, CNT3, ENT1, RRM1 and RRM2, which are genes that are closely involved in metabolic processing of nucleoside analogs. Of the 8 genes, only CNT3 mRNA levels were significantly different between the t(8;21)-positive and -negative patients in non-parametric statistical analysis with Mann-Whitney U test (P<0.001); no significant differences were observed for the remaining genes (Fig. 2A). Using the level of CNT3 in the healthy donors as a reference, the median expression values were 1.89 (range, 0.36–7.67) for t(8;21)-positive patients and 0.63 (range, 0.05–2.16) for t(8;21)-negative patients. For the study of correlation between expression patterns of the candidate genes and cytogenetic characteristics of the patients, we conducted Pearson’s correlation analysis. This analysis revealed that only the CNT3 level had a definite correlation with the presence of cytogenetic abnormalities (r=0.520; P<0.001) (at bottom in Fig. 2A).

To further clarify the distinctive expression patterns of CNT3 in patients, we divided the patients into 3 groups: t(8;21) (n=25), normal cytogenetics (n=16), and other cytogenetic abnormalities (n=13). The patients carrying t(8;21) still had higher CNT3 expression compared to either of the other patient groups, even though the t(8;21)-negative population was divided into 2 subpopulations (Fig. 2B).

The t(8;21) abnormality-specific increase in CNT3 expression was confirmed in a validation cohort of 44 patients diagnosed with M2 FAB subtype AML. To further examine the association between nucleoside transporter genes and cytogenetic abnormalities, we analyzed expression levels of solute carrier (SLC) family genes, including SLC28 (CNT1, CNT2 and CNT3) and SLC29 (ENT1 and ENT2). The levels of CNT1 and CNT2 mRNA were low or undetectable in most of the validation cohort and, therefore, results for these genes were excluded from the following analysis. As shown in Fig. 3A, we observed that CNT3 expression was significantly elevated in patient samples that were t(8;21)-positive, compared to those that were t(8;21)-negative (P<0.001 in U test and P=0.002 in Pearson’s correlation statistics). The median expression values were 2.23 (range, 0.39–15.76) for t(8;21)-positive patients and 0.47 (0.04–5.12) for t(8;21)-negative patients. Levels of ENT1 and ENT2 were not statistically different between the two groups. We also compared DFS and OS between t(8;21)-positive and -negative patient groups. Prolonged survival was observed in patients harboring t(8;21), although the results did not reach statistical significance for survival indices (P=0.342 for DFS and P=0.250 for OS in log-rank tests; Fig. 3B). The t(8;21)-specific higher expression of CNT3 was consistent with the observation in Fig. 2 and implies a particular contribution of CNT3 to favorable outcomes of these patients.

Prognostic impact of CNT3 expression on treatment outcomes

We hypothesized that expression of CNT3 in patients with t(8;21) could enable more precise prediction of treatment outcomes. To examine the potential role of CNT3 in predicting outcomes, 45 t(8;21)-positive patients from the test and validation cohorts were clustered into 3 groups (tertiles) based on their levels of CNT3 expression (high, intermediate or low). First, the prognostic impact of CNT3 was evaluated by univariate analysis. As shown in Fig. 4, patients with high CNT3 expression had significantly longer survival compared to the other two patient groups (DFS, P=0.008; OS, P=0.019 in log-rank tests). In a Cox proportional hazards univariate model, HRs for the patients in the high CNT3 tertile compared to the patients in the low tertile were 0.18 (95% CI, 0.05–0.63; P=0.008) for DFS and 0.14 (95% CI, 0.03–0.66; P=0.013) for OS. However, when the prognostic value of CNT3 was investigated in a separate, restricted group of patients lacking the t(8;21), there were no significant differences in survival among the subpopulations with different CNT3 expression levels (data not shown).

We conducted a multivariate analysis using the Cox proportional hazards model, which adjusted for the influence of the remission induction (RI) regimen, hematopoietic stem cell transplantation (HSCT), blast percentage, age, gender and CNT3 mRNA; this model confirmed that high CNT3 expression was an independent risk factor associated with DFS (HR, 0.12; P=0.007) and OS (HR, 0.06; P=0.010) (Table II). Alternatively, clinical outcomes of patients with t(8;21)-negative AML were affected by the RI regimen and HSCT, but not by the CNT3 expression status (data not shown). These findings suggest that CNT3 expression levels can be used to stratify overall clinical outcomes after cytarabine-based standard chemotherapy in AML patients with t(8;21).

Table II

Multivariate analysis of clinical outcomes in the t(8;21)-positive patients.

Table II

Multivariate analysis of clinical outcomes in the t(8;21)-positive patients.

VariableDisease-free survival
Overall survival
HR (95% CI)P-valueHR (95% CI)P-value
CNT3 expression
CNT3 <1.34 (n=15)1 (reference)1 (reference)
CNT3 ≥1.34 and <3.37 (n=14)0.44 (0.14–1.38)0.1580.80 (0.24–2.64)0.717
CNT3 ≥3.37 (n=15)0.12 (0.03–0.56)0.0070.06 (0.01–0.52)0.010
RI regimen
 Ida/AraC (n=24)1 (reference)1 (reference)
 Ida/BHAC (n=20)1.11 (0.36–3.45)0.8602.20 (0.67–7.23)0.196
HSCT
 Yes (n=24)1 (reference)1 (reference)
 No (n=20)1.27 (0.45–3.59)0.6581.66 (0.51–5.39)0.401
Blast (%)
 <50 (n=16)1 (reference)1 (reference)
 ≥50 (n=27)0.47 (0.14–1.56)0.2140.46 (0.12–1.72)0.251
Age (years)
 ≤60 (n=39)1 (reference)1 (reference)
 >60 (n=5)0.54 (0.11–2.53)0.4310.69 (0.14–3.51)0.654
Gender
 Male (n=26)1 (reference)1 (reference)
 Female (n=18)1.37 (0.46–4.14)0.5731.80 (0.52–6.27)0.355

[i] HR, hazard ratio; CI, confidence interval; RI, remission induction; HSCT, hematopoietic stem cell transplantation.

Discussion

We showed that cytogenetic abnormality-specific expression of CNT3 in leukemic blasts from AML patients correlated with favorable responses to cytarabine-based chemotherapy and prolonged survival of patients with t(8;21). We also found that CNT3 expression level was an independent predictor of the response to cytarabine-based chemotherapy in the t(8;21)-positive AML patients.

The t(8;21) translocation is one of the most frequent cytogenetic abnormalities, occurring in a large proportion (up to 15%) of AML patients. This type of AML has a high remission rate to chemotherapy, particularly when high-dose cytarabine is administered (2,9,26). Nevertheless, some patients with t(8;21) experience relapse and fail to maintain long-term survival (35). For this reason, most studies concerning the treatment response in AML with t(8;21) have been devoted to identification of useful markers that can be measured at diagnosis to predict poor outcomes. Representative clinically significant examples include the association of natural killer cell antigen CD56 and P-glycoprotein with inferior outcomes in t(8;21)-positive AML patients (68,27). However, neither of these factors is linked with favorable outcomes in patients carrying this aberration. Although a limited number of patients were examined in the present study, we were able to provide the first example of a gene related to cytarabine metabolism that can explain the varied responses of these patients to cytarabine therapy.

Nucleoside transporters including both influx and efflux pumps and drug metabolism-associated enzymes have been steadily identified as predictive markers of clinical outcomes and as potential therapeutic targets (24,2831). In this study, we examined two types of specialized influx pumps of nucleoside analogs, ENTs and CNTs, as potential candidates for predicting patient outcomes. We chose these transporters because of previous reports indicating that they have a broad spectrum of substrate flux and are predominantly expressed in BM cells (3234). CNT3 expression significantly correlated with the presence of the t(8;21) aberration in both the test and validation data sets. Previous reports indicated that ENT1 was clinically significant for cytarabine-mediated therapy; however, the present study found a slight but insignificant correlation between ENT1 and the t(8;21) translocation in correlation analysis and survival analysis in AML patients with t(8;21) (33,35,36). These results indicate that CNT3, but not ENT1 or other functional genes, is a specific marker of cytogenetics in AML and is also a critical factor in the responsiveness of t(8;21)-positive patients to cytarabine.

Nucleoside transporters are required not only for nucleotide synthesis, but also for import of a variety of nucleoside-derived anticancer drugs. Specifically, CNT3 plays a role in the uptake of a broad range of nucleosides and their analogs, while CNT1 and CNT2 are responsible for specialized substrates (pyrimidine and purine nucleosides for CNT1 and CNT2, respectively) (34). Most studies that have only analyzed CNT3 have used in vitro assays to identify a critical role of CNT3 in the antitumor effect of nucleoside drugs and the resistance to these drugs (34,37,38). Alternatively, a study by Mackey et al (30) showed a negative correlation between CNT3 and clinical outcomes after fludarabine therapy in patients with chronic lymphocytic leukemia. Furthermore, distinct expression of CNT3 has not been reported in AML with specific cytogenetics, even though there were several genome-wide approaches to dissect gene profiles in large numbers of patients (3941). The present study is the first to demonstrate that CNT3 levels can be used to stratify patients based on survival outcomes in, at least in part, t(8;21)-positive but not t(8;21)-negative AML patients. We questioned why the prognostic potential of CNT3 preferentially applied to t(8;21)-positive AML patients. For the survival analysis, we divided t(8;21)-positive or -negative patients into subpopulations based on their CNT3 expression level. The median values for CNT3 expression were 2.07 (range, 0.36–15.76) for the t(8;21)-positive cohort and 0.56 (range, 0.04–5.12) for the t(8;21)-negative cohort. One potential explanation for this observation is that, at least in the t(8;21)-negative population, the small range of CNT3 expression might be insufficient to allow stratification for outcome prediction. Although we did not determine whether CNT3 transcript levels were consistent with protein expression levels in individual patients, and we did not ensure that the protein encoded by the CNT3 gene was functional, it is believed that low mRNA levels are indicative of low CNT3 protein levels, which are insufficient for CNT3 to carry out its novel function.

In the CNT nucleoside transporter family, expression levels of CNT1 and CNT2 are known to be upregulated by extracellular stimuli including cell proliferation and activation (4244). However, the mechanism of transcriptional regulation of CNT3 remains largely unknown. Based on our novel finding that CNT3 was upregulated in AML patients with t(8;21) but not in patients with other cytogenetic changes, we hypothesize that there may be a physiological interaction between the AML1 or AML1-ETO chimera proteins, which are derived from the t(8;21) translocation, and the promoter region of CNT3. Indeed, a virtual analysis for promoter prediction performed using transcription factor binding site search software identified 3 putative binding sites for AML1 at −3438 to −3433, −888 to −883, and +526 to +531 relative to the transcription start site of CNT3. Therefore, additional study is necessary to identify a role of these putative binding sites.

In summary, our results imply that higher CNT3 expression in AML patients with t(8;21) contributes to the prolonged survival observed in this population. Our results also suggest that CNT3 can be used to predict clinical responses to cytarabine-based therapies in AML patients with t(8;21).

Acknowledgments

The present study was supported by grant A111218-GM06 of the National Project for Personalized Genomic Medicine, Ministry for Health and Welfare, the National Research Foundation of Korea (NRF) grant 2011-0001388 funded by the Korean government and a Korea University grant.

References

1 

Grimwade D, Walker H, Oliver F, Wheatley K, Harrison C, Harrison G, Rees J, Hann I, Stevens R, Burnett A, et al The Medical Research Council Adult and Children’s Leukaemia Working Parties: The importance of diagnostic cytogenetics on outcome in AML: Analysis of 1,612 patients entered into the MRC AML 10 trial. Blood. 92:2322–2333. 1998.PubMed/NCBI

2 

Estey EH: Acute myeloid leukemia: 2012 update on diagnosis, risk stratification, and management. Am J Hematol. 87:89–99. 2012. View Article : Google Scholar

3 

Bloomfield CD, Herzig GP, Peterson BA and Wolff SN: Long-term survival of patients with acute myeloid leukemia: Updated results from two trials evaluating postinduction chemotherapy. Cancer. 80(Suppl): 2186–2190. 1997. View Article : Google Scholar : PubMed/NCBI

4 

O’Brien S, Kantarjian HM, Keating M, Gagnon G, Cork A, Trujillo J and McCredie KB: Association of granulocytosis with poor prognosis in patients with acute myelogenous leukemia and translocation of chromosomes 8 and 21. J Clin Oncol. 7:1081–1086. 1989.

5 

Prébet T, Boissel N, Reutenauer S, Thomas X, Delaunay J, Cahn JY, Pigneux A, Quesnel B, Witz F, Thépot S, et al Acute Leukemia French Association; Groupe Ouest-Est des leucémies et autres maladies du sang (GOELAMS); Core Binding Factor Acute Myeloid Leukemia (CBF AML) intergroup: Acute myeloid leukemia with translocation (8;21) or inversion (16) in elderly patients treated with conventional chemotherapy: A collaborative study of the French CBF-AML intergroup. J Clin Oncol. 27:4747–4753. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Baer MR, Stewart CC, Lawrence D, Arthur DC, Byrd JC, Davey FR, Schiffer CA and Bloomfield CD: Expression of the neural cell adhesion molecule CD56 is associated with short remission duration and survival in acute myeloid leukemia with t(8;21)(q22;q22). Blood. 90:1643–1648. 1997.PubMed/NCBI

7 

Schaich M, Koch R, Soucek S, Repp R, Ehninger G and Illmer T: A sensitive model for prediction of relapse in adult acute myeloid leukaemia with t(8;21) using white blood cell count, CD56 and MDR1 gene expression at diagnosis. Br J Haematol. 125:477–479. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Yang DH, Lee JJ, Mun YC, Shin HJ, Kim YK, Cho SH, Chung IJ, Seong CM and Kim HJ: Predictable prognostic factor of CD56 expression in patients with acute myeloid leukemia with t(8:21) after high dose cytarabine or allogeneic hematopoietic stem cell transplantation. Am J Hematol. 82:1–5. 2007. View Article : Google Scholar

9 

Bloomfield CD, Lawrence D, Byrd JC, Carroll A, Pettenati MJ, Tantravahi R, Patil SR, Davey FR, Berg DT, Schiffer CA, et al: Frequency of prolonged remission duration after high-dose cytarabine intensification in acute myeloid leukemia varies by cytogenetic subtype. Cancer Res. 58:4173–4179. 1998.PubMed/NCBI

10 

Weick JK, Kopecky KJ, Appelbaum FR, Head DR, Kingsbury LL, Balcerzak SP, Bickers JN, Hynes HE, Welborn JL, Simon SR, et al: A randomized investigation of high-dose versus standard-dose cytosine arabinoside with daunorubicin in patients with previously untreated acute myeloid leukemia: A Southwest Oncology Group study. Blood. 88:2841–2851. 1996.PubMed/NCBI

11 

Schnetzke U, Fix P, Spies-Weisshart B, Schrenk K, Glaser A, Fricke HJ, La Rosée P, Hochhaus A and Scholl S: Efficacy and feasibility of cyclophosphamide combined with intermediate- dose or high-dose cytarabine for relapsed and refractory acute myeloid leukemia (AML). J Cancer Res Clin Oncol. 140:1391–1397. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Damaraju VL, Damaraju S, Young JD, Baldwin SA, Mackey J, Sawyer MB and Cass CE: Nucleoside anticancer drugs: The role of nucleoside transporters in resistance to cancer chemotherapy. Oncogene. 22:7524–7536. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Zhang J, Visser F, King KM, Baldwin SA, Young JD and Cass CE: The role of nucleoside transporters in cancer chemotherapy with nucleoside drugs. Cancer Metastasis Rev. 26:85–110. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Momparler RL and Fischer GA: Mammalian deoxynucleoside kinase. I. Deoxycytidine kinase: Purification, properties, and kinetic studies with cytosine arabinoside. J Biol Chem. 243:4298–4304. 1968.PubMed/NCBI

15 

Galmarini CM, Thomas X, Graham K, El Jafaari A, Cros E, Jordheim L, Mackey JR and Dumontet C: Deoxycytidine kinase and cN-II nucleotidase expression in blast cells predict survival in acute myeloid leukaemia patients treated with cytarabine. Br J Haematol. 122:53–60. 2003. View Article : Google Scholar : PubMed/NCBI

16 

Molina-Arcas M, Bellosillo B, Casado FJ, Montserrat E, Gil J, Colomer D and Pastor-Anglada M: Fludarabine uptake mechanisms in B-cell chronic lymphocytic leukemia. Blood. 101:2328–2334. 2003. View Article : Google Scholar

17 

Song JH, Kim SH, Kweon SH, Lee TH, Kim HJ, Kim HJ and Kim TS: Defective expression of deoxycytidine kinase in cytarabine-resistant acute myeloid leukemia cells. Int J Oncol. 34:1165–1171. 2009.PubMed/NCBI

18 

Takagaki K, Katsuma S, Kaminishi Y, Horio T, Nakagawa S, Tanaka T, Ohgi T and Yano J: Gene-expression profiling reveals down-regulation of equilibrative nucleoside transporter 1 (ENT1) in Ara-C-resistant CCRF-CEM-derived cells. J Biochem. 136:733–740. 2004. View Article : Google Scholar

19 

Abraham A, Varatharajan S, Abbas S, Zhang W, Shaji RV, Ahmed R, Abraham A, George B, Srivastava A, Chandy M, et al: Cytidine deaminase genetic variants influence RNA expression and cytarabine cytotoxicity in acute myeloid leukemia. Pharmacogenomics. 13:269–282. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Galmarini CM, Cros E, Graham K, Thomas X, Mackey JR and Dumontet C: 5′-(3′)-nucleotidase mRNA levels in blast cells are a prognostic factor in acute myeloid leukemia patients treated with cytarabine. Haematologica. 89:617–619. 2004.PubMed/NCBI

21 

Momparler RL, Eliopoulos N, Bovenzi V, Létourneau S, Greenbaum M and Cournoyer D: Resistance to cytosine arabi-noside by retrovirally mediated gene transfer of human cytidine deaminase into murine fibroblast and hematopoietic cells. Cancer Gene Ther. 3:331–338. 1996.PubMed/NCBI

22 

Ujházy P, Berleth ES, Pietkiewicz JM, Kitano H, Skaar JR, Ehrke MJ and Mihich E: Evidence for the involvement of ecto-5′-nucleotidase (CD73) in drug resistance. Int J Cancer. 68:493–500. 1996. View Article : Google Scholar

23 

Smid K, Bergman AM, Eijk PP, Veerman G, van Haperen VW, van den Ijssel P, Ylstra B and Peters GJ: Micro-array analysis of resistance for gemcitabine results in increased expression of ribonucleotide reductase subunits. Nucleosides Nucleotides Nucleic Acids. 25:1001–1007. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Song JH, Kweon SH, Kim HJ, Lee TH, Min WS, Kim HJ, Kim YK, Hwang SY and Kim TS: High TOP2B/TOP2A expression ratio at diagnosis correlates with favourable outcome for standard chemotherapy in acute myeloid leukaemia. Br J Cancer. 107:108–115. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Cho EK, Bang SM, Ahn JY, Yoo SM, Park PW, Seo YH, Shin DB and Lee JH: Prognostic value of AML 1/ETO fusion transcripts in patients with acute myelogenous leukemia. Korean J Intern Med. 18:13–20. 2003.PubMed/NCBI

26 

Downing JR: The AML1-ETO chimaeric transcription factor in acute myeloid leukaemia: Biology and clinical significance. Br J Haematol. 106:296–308. 1999. View Article : Google Scholar : PubMed/NCBI

27 

Schaich M, Harbich-Brutscher E, Pascheberg U, Mohr B, Soucek S, Ehninger G and Illmer T: Association of specific cytogenetic aberrations with mdr1 gene expression in adult myeloid leukemia and its implication in treatment outcome. Haematologica. 87:455–464. 2002.PubMed/NCBI

28 

Jin G, Matsushita H, Asai S, Tsukamoto H, Ono R, Nosaka T, Yahata T, Takahashi S and Miyachi H: FLT3-ITD induces ara-C resistance in myeloid leukemic cells through the repression of the ENT1 expression. Biochem Biophys Res Commun. 390:1001–1006. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Jordheim LP and Dumontet C: Review of recent studies on resistance to cytotoxic deoxynucleoside analogues. Biochim Biophys Acta. 1776:138–159. 2007.PubMed/NCBI

30 

Mackey JR, Galmarini CM, Graham KA, Joy AA, Delmer A, Dabbagh L, Glubrecht D, Jewell LD, Lai R, Lang T, et al: Quantitative analysis of nucleoside transporter and metabolism gene expression in chronic lymphocytic leukemia (CLL): Identification of fludarabine-sensitive and -insensitive populations. Blood. 105:767–774. 2005. View Article : Google Scholar

31 

Guo Y, Köck K, Ritter CA, Chen ZS, Grube M, Jedlitschky G, Illmer T, Ayres M, Beck JF, Siegmund W, et al: Expression of ABCC-type nucleotide exporters in blasts of adult acute myeloid leukemia: Relation to long-term survival. Clin Cancer Res. 15:1762–1769. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Griffiths M, Beaumont N, Yao SY, Sundaram M, Boumah CE, Davies A, Kwong FY, Coe I, Cass CE, Young JD, et al: Cloning of a human nucleoside transporter implicated in the cellular uptake of adenosine and chemotherapeutic drugs. Nat Med. 3:89–93. 1997. View Article : Google Scholar : PubMed/NCBI

33 

Hubeek I, Stam RW, Peters GJ, Broekhuizen R, Meijerink JP, van Wering ER, Gibson BE, Creutzig U, Zwaan CM, Cloos J, et al: The human equilibrative nucleoside transporter 1 mediates in vitro cytarabine sensitivity in childhood acute myeloid leukaemia. Br J Cancer. 93:1388–1394. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Ritzel MW, Ng AM, Yao SY, Graham K, Loewen SK, Smith KM, Ritzel RG, Mowles DA, Carpenter P, Chen XZ, et al: Molecular identification and characterization of novel human and mouse concentrative Na+-nucleoside cotransporter proteins (hCNT3 and mCNT3) broadly selective for purine and pyrimidine nucleosides (system cib). J Biol Chem. 276:2914–2927. 2001. View Article : Google Scholar

35 

Galmarini CM, Thomas X, Calvo F, Rousselot P, El Jafaari A, Cros E and Dumontet C: Potential mechanisms of resistance to cytarabine in AML patients. Leuk Res. 26:621–629. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Stam RW, den Boer ML, Meijerink JP, Ebus ME, Peters GJ, Noordhuis P, Janka-Schaub GE, Armstrong SA, Korsmeyer SJ and Pieters R: Differential mRNA expression of Ara-C-metabolizing enzymes explains Ara-C sensitivity in MLL gene-rearranged infant acute lymphoblastic leukemia. Blood. 101:1270–1276. 2003. View Article : Google Scholar

37 

Fotoohi AK, Lindqvist M, Peterson C and Albertioni F: Involvement of the concentrative nucleoside transporter 3 and equilibrative nucleoside transporter 2 in the resistance of T-lymphoblastic cell lines to thiopurines. Biochem Biophys Res Commun. 343:208–215. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Karim H, Hashemi J, Larsson C, Moshfegh A, Fotoohi AK and Albertioni F: The pattern of gene expression and gene dose profiles of 6-Mercaptopurine- and 6-Thioguanine-resistant human leukemia cells. Biochem Biophys Res Commun. 411:156–161. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Verhaak RG, Wouters BJ, Erpelinck CA, Abbas S, Beverloo HB, Lugthart S, Löwenberg B, Delwel R and Valk PJ: Prediction of molecular subtypes in acute myeloid leukemia based on gene expression profiling. Haematologica. 94:131–134. 2009. View Article : Google Scholar :

40 

Balgobind BV, Van den Heuvel-Eibrink MM, De Menezes RX, Reinhardt D, Hollink IH, Arentsen-Peters ST, van Wering ER, Kaspers GJ, Cloos J, de Bont ES, et al: Evaluation of gene expression signatures predictive of cytogenetic and molecular subtypes of pediatric acute myeloid leukemia. Haematologica. 96:221–230. 2011. View Article : Google Scholar :

41 

Ichikawa H, Tanabe K, Mizushima H, Hayashi Y, Mizutani S, Ishii E, Hongo T, Kikuchi A and Satake M: Common gene expression signatures in t(8;21)- and inv(16)-acute myeloid leukaemia. Br J Haematol. 135:336–347. 2006. View Article : Google Scholar : PubMed/NCBI

42 

Duflot S, Riera B, Fernández-Veledo S, Casadó V, Norman RI, Casado FJ, Lluís C, Franco R and Pastor-Anglada M: ATP-sensitive K+ channels regulate the concentrative adenosine transporter CNT2 following activation by A1 adenosine receptors. Mol Cell Biol. 24:2710–2719. 2004. View Article : Google Scholar : PubMed/NCBI

43 

Fernández-Veledo S, Valdés R, Wallenius V, Casado FJ and Pastor-Anglada M: Up-regulation of the high-affinity pyrimidine-preferring nucleoside transporter concentrative nucleoside transporter 1 by tumor necrosis factor-alpha and interleukin-6 in liver parenchymal cells. J Hepatol. 41:538–544. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Soler C, Felipe A, García-Manteiga J, Serra M, Guillén-Gómez E, Casado FJ, MacLeod C, Modolell M, Pastor-Anglada M and Celada A: Interferon-gamma regulates nucleoside transport systems in macrophages through signal transduction and activator of transduction factor 1 (STAT1)-dependent and -independent signalling pathways. Biochem J. 375:777–783. 2003. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2015
Volume 34 Issue 1

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Song JH, Cho K, Kim H, Kim Y, Kim NY, Kim H, Lee T, Hwang SY and Kim TS: Concentrative nucleoside transporter 3 as a prognostic indicator for favorable outcome of t(8;21)-positive acute myeloid leukemia patients after cytarabine-based chemotherapy. Oncol Rep 34: 488-494, 2015
APA
Song, J.H., Cho, K., Kim, H., Kim, Y., Kim, N.Y., Kim, H. ... Kim, T.S. (2015). Concentrative nucleoside transporter 3 as a prognostic indicator for favorable outcome of t(8;21)-positive acute myeloid leukemia patients after cytarabine-based chemotherapy. Oncology Reports, 34, 488-494. https://doi.org/10.3892/or.2015.3959
MLA
Song, J. H., Cho, K., Kim, H., Kim, Y., Kim, N. Y., Kim, H., Lee, T., Hwang, S. Y., Kim, T. S."Concentrative nucleoside transporter 3 as a prognostic indicator for favorable outcome of t(8;21)-positive acute myeloid leukemia patients after cytarabine-based chemotherapy". Oncology Reports 34.1 (2015): 488-494.
Chicago
Song, J. H., Cho, K., Kim, H., Kim, Y., Kim, N. Y., Kim, H., Lee, T., Hwang, S. Y., Kim, T. S."Concentrative nucleoside transporter 3 as a prognostic indicator for favorable outcome of t(8;21)-positive acute myeloid leukemia patients after cytarabine-based chemotherapy". Oncology Reports 34, no. 1 (2015): 488-494. https://doi.org/10.3892/or.2015.3959