Startseite Review of pharmacogenetics studies of L-asparaginase hypersensitivity in acute lymphoblastic leukemia points to variants in the GRIA1 gene
Artikel Öffentlich zugänglich

Review of pharmacogenetics studies of L-asparaginase hypersensitivity in acute lymphoblastic leukemia points to variants in the GRIA1 gene

  • Maria Lopez-Santillan , Leire Iparraguirre , Idoia Martin-Guerrero , Angela Gutierrez-Camino und Africa Garcia-Orad EMAIL logo
Veröffentlicht/Copyright: 6. März 2017
Veröffentlichen auch Sie bei De Gruyter Brill

Abstract

Acute lymphoblastic leukemia (ALL) is a major pediatric cancer in developed countries. Although treatment outcome has improved owing to advances in chemotherapy, there is still a group of patients who experience severe adverse events. L-Asparaginase is an effective antineoplastic agent used in chemotherapy of ALL. Despite its indisputable indication, hypersensitivity reactions are common. In those cases, discontinuation of treatment is usually needed and anti-asparaginase antibody production may also attenuate asparaginase activity, compromising its antileukemic effect. Till now, six pharmacogenetic studies have been performed in order to elucidate possible genetic predisposition for inter-individual differences in asparaginase hypersensitivity. In this review we have summarized the results of those studies which describe the involvement of four different genes, being polymorphisms in the glutamate receptor, ionotropic, AMPA 1 (GRIA1) the most frequently associated with asparaginase hypersensitivity. We also point to new approaches focusing on epigenetics that could be interesting for consideration in the near future.

Introduction

Acute lymphoblastic leukemia (ALL) is a major pediatric cancer in developed countries, accounting for 30% of all malignancies in children [1]. During the last few years, treatment outcome has improved substantially owing to advances in chemotherapy, with cure rates now exceeding 80% [2], [3], [4]. However, there are groups of patients who still remain refractory to therapy or experience severe toxicity affecting their quality of life during and after treatment. Consequently, there is a current interest in detecting markers which recognize, in advance, patients who will be resistant to treatment or suffer from the adverse effects of therapy, in order to adjust the treatment from the beginning.

In this context, pharmacogenetic studies can be useful in childhood ALL for several reasons, as it was mentioned in a review by Lopez-Lopez et al. [5]:

  1. Chemotherapeutic drugs used in ALL treatment have a very narrow therapeutic range. This means there is a small difference between the effective dose and the dose that causes toxicity [6], [7]. Consequently, toxicity prevention is challenging because it is difficult to predict, and dose reduction is dangerous as under-dosage is usually associated with decreased survival.

  2. Genes involved in the metabolic pathways of the drugs used in ALL treatment are highly variable [8], [9]. In fact, several genetic variants have been described that lead to inter-individual variability in protein levels or activity in those pathways. These variations can lead to changes in drug exposure or activity and impact treatment response.

  3. Treatment protocols for ALL are standardized and well established, which allows the inclusion of large and homogeneously treated groups of patients in the studies. As a result, larger and homogeneous samples increase the statistical power of the studies and strengthen the reliability of the results.

Till date, several studies have been performed in order to search for pharmacogenetic markers of toxicity and outcome in pediatric ALL [5], [10], [11], [12], [13], [14], [15], [16]. However, the results are controversial for most of the analyzed polymorphisms. The lack of replication could be due to the characteristics of the samples included (small or mixed patients populations), to the toxicity criteria studied (different among studies), or to the differences among treatment protocols.

In Spain, the treatment of ALL was recently unified with LAL/SEHOP-PETHEMA 2013 protocol. According to this protocol, the treatment lasts an average of 2 years including induction, consolidation, re-induction and maintenance phases, in which different drugs are combined. One of the main drugs used is L-asparaginase (L-ASP), which is administered in the induction and re-induction phases, as well as in consolidation or intensification phase in high risk patients (Figure 1). Asparaginase is an enzyme that metabolizes extracellular asparagine into aspartic acid. Its antileukemic effect is based on the relative inability of leukemic cells to synthesize asparagine, as opposed to normal cells. The depletion of asparagine diminishes protein synthesis, leading to leukemic cell death [17], [18].

Figure 1: LAL/SEHOP-PETHEMA 2013 treatment protocol.The phases where asparaginase is administered are pointed out.
Figure 1:

LAL/SEHOP-PETHEMA 2013 treatment protocol.

The phases where asparaginase is administered are pointed out.

Several studies have demonstrated that increased dose of L-ASP improves response in patients with ALL [19], [20]. Despite this widespread success, hypersensitivity to L-ASP remains a commonly reported adverse event and often requires termination of asparaginase treatment. Therefore, the detection of markers which recognize, in advance, patients who will develop hypersensitivity to L-ASP will help to optimize the treatment.

Asparaginase

There are currently three forms of asparaginase that are used in clinical practice: native and pegylated form derived from Escherichia coli (E. coli-ASP and PEG-ASP, respectively), and an enzyme isolated from Erwiniachrysanthemi, known as Erwiniaasparaginase (Erwinia-ASP), antigenically distinct from E. coli-derived asparaginase forms [21]. All commercially available asparaginase preparations have been reported to have the potential to elicit an immune response characterized by the development of anti-asparaginase antibodies. The presence of these L-ASP antibodies is often associated with clinical or symptomatic hypersensitivity, which is the adverse effect more frequently described. Nevertheless, L-ASP preparations do not always lead to clinical hypersensitivity, but may instead cause rapid inactivation of the asparaginase, resulting in suboptimal asparagine depletion [21]. This is commonly referred to as ‘silent hypersensitivity’ or ‘silent inactivation’ and may occur in approximately 30% of the patients and it is characterized by circulating antibodies without clinical manifestations [22].

The incidence of hypersensitivity is variable depending on three different formulations of L-ASP used in clinical practice, with rates that vary from 20% to 31.5% in native E. coli-treated patients [23], [24], [25], from 1.8% to 9.4% in PEG-treated patients [26], [27], [28], and from 10.9% to 33% [29], [30], [31] in Erwinia-treated patients. Typically, patients exhibiting clinical allergy symptoms to one formulation of asparaginase are switched to another product to ensure they receive the most efficacious treatment regimen possible [22], [32]. In comparison with E. coli-derived asparaginase forms, Erwinia-ASP has a significantly reduced duration of action that permits tighter control of drug exposure should adverse effects occur. In fact, currently, hypersensitivity is the only recommended reason, as well as the only Food and Drug Administration-approved indication, for switching to Erwinia-ASP [33]. Nevertheless, the risk for allergic asparaginase-related adverse effects does still exist with the use of this drug. Additionally, other factors such as the number of asparaginase applications, route of administration, readministration after hiatus, and concomitant therapy can influence [34]. For example, Kutszegi et al. noticed that the highest occurrence of E. coli L-ASP hypersensitivity happened in the first dose of L-ASP in the reinduction, after a 3-month long break (consolidation phase) on medium risk (MR) arm [34]; in accordance with the fact that re-exposure to L-ASP after hiatus increases risk to hypersensitivity.

Clinical hypersensitivity is characterized by an allergic reaction, and can include the following sympthoms: rash, pain around the injection site, flushing, fever, chills, dyspnea, bronchospasm and anaphylaxis (which can be life-threatening situation requiring urgent interventions [35]). Hypersensitivity reactions are classified according to gravity in grades from 1 to 5 according to Common Terminology Criteria for Adverse Events, being the most frequent grade 1 and 2 ones. Accordingly, developing clinical hypersensitivity negatively impacts outcomes in ALL due to the fact that the discontinuation of L-ASP therapy is usually needed in these cases [36], and that the presence of L-ASP antibodies can substantially reduce L-ASP activity [22], [37].

Other important adverse effects, often enhanced by concomitant use of corticoids in the induction and re-induction phases, are as following: deep vein thrombosis (5.2%, mostly in the induction), pancreatitis (5%–10% usually after first doses of L-ASP) [33], [38], hepatotoxicity (~75% in the first 2 weeks), hyperglycemia (20%–35% [39]) and severe hypertriglyceridemia (>1000 mg/dL) (7%, [40]).

Remarkably, these adverse events exhibit large inter-individual differences. Therefore, predicting which patients will suffer from L-ASP toxicity and which type of toxicity will be developed is one of the challenges of modern medicine.

Pharmacogenetics

To date, only 10 pharmacogenetics studies analyzing L-ASP have been performed in pediatric ALL [34], [41], [42], [43], [44], [45], [46], [47], [48], [49]. Among them, six specifically focused on the analysis of the risk to L-ASP hypersensitivity, finding the most remarkable results with 15 polymorphisms located at GRIA1, HLA-DRB1, NFATC2 and ASNS (Table 1) [34], [41], [42], [43], [44], [49]. Interestingly, out of these 15 significant polymorphisms, 11 were located in the GRIA1 gene (glutamate receptor, ionotropic, AMPA 1). In 2010, Chen et al. performed a genome wide association study (GWAS) interrogating 364,033 single nucleotide polymorphisms (SNPs) in 485 children with ALL [assigned into discovery (n=322) and validation (n=163) cohorts, as well as the combined cohort (n=485)]. From the top 100 SNPs associated with allergy in the discovery cohort, they found that chromosome 5 was overrepresented with 10 significant SNPs. Among them, rs4958351 located at GRIA1 gene was remarkably significant in the discovery, validation and combined cohorts. Focused on this gene, they found 9 SNPs (rs4958351, rs10070447, rs6890057, rs4958676, rs6889909, rs11167640, rs10072570, rs13354399 and rs17356099) significantly associated with native E-coli-ASP hypersensitivity in the discovery and combined cohorts. Notably, five of them (rs4958351, rs10070447, rs6890057, rs4958676 and rs6889909) also remained significant in the validation cohort [41], although no correction for multiple comparisons was applied. In 2015, Rajić et al. replicated the association of these last 5 SNPs with native E. coli L-ASP hypersensitivity in a cohort of 146 Caucasian children with ALL [42]. However, the association between rs4958351 and the risk to native E. coli L-ASP hypersensitivity was only replicated by Kutszegi et al. specifically in T-ALL subgroup, but with opposite effect. They found that A allele reduced the risk to hypersensitivity in this subgroup (n=69); whereas A allele was associated with a slightly but not significantly increased risk to hypersensitivity in [pre-B-ALL (n=402)] [34]. Kutszegi et al. explained this difference among studies because Chen et al. and Rajić et al. did not analyze specifically T-ALL due to the low number of this type of samples in their cohorts. Additionally, Kutszegi et al. found other two SNPs in GRIA1 (rs2055083 and rs707176) associated with hypersensitivity but only in the medium risk group [34]. GRIA1 encodes a subunit (GluR1) of the ionotropic α-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) receptor, a tetrameric ligand-gated ion channel that transmits glutamatergic signals in the brain. Glutamate has been recently shown to have a role not only as a neurotransmitter, but also as an immunomodulator [50], [51]. In 2003, AMPA-activated ionotropicGluRs were detected in human lymphocytes [52]. Moreover, glutamate was shown to be able to activate and modulate T cell activity by itself [50], [53], [54], [55]. In addition, GRIA1 gene is located in chromosome 5q31-33, which has been considered as a susceptibility locus for several inflammatory or autoimmune diseases, including asthma. These results suggest that drug allergy and asthma share a range of candidate genes [56], [57], [58], [59], [60], [61].

Table 1:

Polymorphisms associated with L-ASP hypersensitivity in B-/T-ALL pediatric patients.

GenePolymorphismAlleleaStudy SNPsSamplesbEthnicityp-ValueRisk (95% CI)Ref.
GRIA1 (Chr.5)rs4958351G/AGWAS322W/B/H/A/Oc1.8 E–5HZ=1.72 (1.34–2.20)[41]
GWAS163W/B/H/A/Od0.0029HZ=1.93 (1.25–2.98)[41]
5146Caucasian0.003OR=1.74 (1.18–2.57)[42]
20464HungarianNSNS[34]
rs10070447C/TGWAS322W/B/H/A/Oc1.3E–4HZ=1.59 (1.25–2.02)[41]
GWAS163W/B/H/A/Od0.0023HZ=1.96 (1.27–3.01)[41]
5146Caucasian0.006OR=1.65 (1.11–2.45)[42]
rs6890057C/TGWAS322W/B/H/A/Oc4.3 E–4HZ=1.64 (1.24–2.15)[41]
GWAS163W/B/H/A/Od0.0035HZ=1.80 (1.21–2.68)[41]
5146Caucasian0.005OR=1.59 (1.17–2.17)[42]
rs4958676G/AGWAS322W/B/H/A/Oc0.0014HZ=1.57 (1.19–2.08)[41]
GWAS163W/B/H/A/Od0.0066HZ=1.66 (1.15–2.40)[41]
5146Caucasian0.005OR=1.59 (1.17–2.17)[42]
rs6889909C/TGWAS322W/B/H/A/Oc0.0020HZ=1.53 (1.17–2.00)[41]
GWAS163W/B/H/A/Od0.016HZ=1.58 (1.09–2.29)[41]
5146Caucasian0.005OR=1.59 (1.17–2.17)[42]
rs11167640C/TGWAS322W/B/H/A/Oc0.0025HZ=1.72 (1.21–2.45)[41]
GWAS163W/B/H/A/OdNSNS[41]
20490HungarianNSNS[34]
rs10072570G/AGWAS322W/B/H/A/Oc0.0038HZ=1.47 (1.13–1.91)[41]
GWAS163W/B/H/A/OdNSNS[41]
rs13354399G/AGWAS322W/B/H/A/Oc0.0069HZ=1.41 (1.10–1.80)[41]
GWAS163W/B/H/A/OdNSNS[41]
rs17356099T/CGWAS322W/B/H/A/Oc0.017HZ=1.85 (1.12–3.07)[41]
GWAS163W/B/H/A/OdNSNS[41]
rs2055083G/A20490Hungarian0.104OR=0.66 (0.39–1.09)[34]
rs707176T/C20477Hungarian0.041OR=1.90 (1.02–3.48)[34]
HLA-DRB1 (Chr.6)HLA-DRB1*07 :01541870European0.002OR=1.64 (1.28–2.09)[43]
rs17885382C/TGWAS3308E/Af/H/A/Oe3.5E-5OR=1.66 (1.3–2.1)[44]
NFATC2 (Chr.20)rs6021191A/TGWAS3308E/Af/H/A/Oe5.4E-6OR=3.07 (1.87–4.94)[44]
ASNS (Chr.7)Haplotype*114285Caucasian0.01OR=0.4 (0.2–0.8)[49]
14248Caucasian0.002OR=0.3 (0.1–0.6)[49]

HZ, hazard ratio; OR, odds ratio; CI, confidence interval; NS, no significant. aAlleles are listed with the risk allele as the second allele. bNumber of individuals with genotyping data for the SNP of interest. cWhite 65.5%/Black 12.5%/Hispanic 14.3%/Asian 1.2%/Other 6.5%. dWhite 60.7%/Black 20.3%/Hispanic 14.7%/Asian 1.2%/Other 3.1%. eEuropean 59.5%/Hispanic 22.0%/African 7.7%/Asian 1.6%/Other 9.2%.

Regarding the HLA-DRB1 (major histocompatibility complex, class II, DR β 1) gene, Fernandez et al. identified the polymorphism HLA-DRB1*07:01 allele associated with L-ASP hypersensitivity. These authors analyzed two cohorts of 541 and 1329 Caucasian patients, and suggested that risk allele conferred high-affinity binding to asparaginase epitopes leading to a higher frequency of hypersensitivity reactions [43]. In line with these results, other studies linked variants in HLA genes on chromosome 6 with autoimmune diseases [62], [63]. Additionally, several HLA-B alleles were associated with small molecule drug allergy and adverse reactions [64], [65], [66], [67], [68], [69]. Specifically, HLA-DRB1*07:01 allele was associated with elevated levels of serum alanine transferase during ximelagatrn and lapatinib treatment through a drug-induced adaptative immune response [70], [71]. In 2015, the same group performed a genome-wide approach and found significant the nonsynonymous polymorphism rs17885382 in the exon 2 of HLA-DRB1 (Arg >Gln, 54), which is in complete linkage disequilibrium with the HLA-DRB1*07:01 allele previously mentioned. In addition, they pointed out another gene, nuclear factor of activated T cells 2 (NFATC2), as a possible candidate gene implicated in the risk of hypersensitivity. They found that the intronic rs6021191 polymorphism was associated with higher risk of L-ASP hypersensitivity increasing NFATC2 expression [44]. NFATC2 gene encodes a cytoplasmatic component of the NFAT transcription factor family. Upon T-cell receptor stimulation, cytoplasmatic NFATC2 is dephosphorylated and traslocated to the nucleus where it participates in gene regulation. So far, diverse studies have shown that NFATC2 can influence the development and function of regulatory T cells and can have either negative or positive regulatory influence on the immune response, depending on the antigen [72], [73]. Nevertheless, other studies have demonstrated that inhibition of the NFAT pathway can attenuate an immune response [74], [75], [76], [77], [78], [79].

Finally, in 2015 Ben Tanfous et al. studying 14 SNPs in ATF5 (activating transcription factor 5), ASNS (asparagine synthetase) and ASS1 (argininosuccinate synthase) genes in 285 Caucasian pediatric ALL, discovered that homozygosity for haplotype*2 (3R allele) in ASNS, conferred higher risk of pancreatitis and hypersensitivity in the discovery cohort, while haplotype*1 (defined by C-181 of rs3757676 and 2R alleles) was associated with protective effect for hypersensitivity in the discovery (n=285) and replication (n=248) cohorts [49].

Noncoding RNAs

To date, most of pharmacogenetic research of L-ASP hypersensitivity in ALL has focused on the analysis of variants in coding genes. Nevertheless, these regions correspond only to approximately 1.5% of the entire genome [80]. Nowadays, it is known that regions that do not codify proteins (98.5%), such as microRNAs (miRNAs), may have an important regulatory function. MiRNAs can regulate the expression of 50% of the genes, including those involved in the administration, distribution, metabolism and excretion (ADME) of drugs (ADME genes). Therefore, deregulation or polymorphisms of miRNAs, by regulating the expression of drug-related genes, can play a pivotal role in drug response or side effects. In fact, a new field in pharmacogenetics has been proposed: the mir-pharmacogenetics [81].

Regarding childhood ALL therapy, several studies have already reported variations in miRNA expression or miRNA-related SNPs associated with the response to treatment as well as with drug-related adverse effects [10], [11], [12], [82], [83], [84], [85]. However, to date, there are no studies analyzing miRNAs involved in L-ASP hypersensitivity. Nevertheless, three studies have shown deregulated miRNAs expression associated with L-ASP response in childhood B-ALL [86], [87], [88]. In 2011, Schotte et al. found that miR-454 was expressed at a 1.9-fold lower level in L-ASP-resistant cases [86]. In another study, Mei et al. in 2014 observed that low levels of miR-210 in B-ALL patients were associated with a lower response to L-ASP treatment and higher risk of relapse. Moreover, in vitro analysis also showed that low levels of mir-210 in cell lines increased L-ASP resistance [87]. Finally, MesrianTanha et al., in 2016 found miR-491-5p and miR-93-5p deregulated in patients who presented L-ASP resistance [88].

Therefore, taking into account that all the four genes associated with L-ASP hypersensitivity have both predicted and validated miRNA binding sites in miRWalk (http://zmf.umm.uni-heidelberg.de/apps/zmf/mirwalk/), it is tempting to speculate that changes in miRNAs could be also associated with L-ASP hypersensitivity. (Supplemental table 1 gives characteristics of the 6 pharmacogenetic studies analyzing the L-ASP hypersensitivity in pediatric ALL patients).

Conclusions and future perspective

During the last few years, several studies have been performed in the context of ALL pharmacogenetics. Some of them have focused on L-ASP induced hypersensitivity pharmacogenetics, pointing out the involvement of four genes GRIA1, HLA-DRB1, NFATC2 and ASNS. However, the low number of studies performed and the lack of replication have failed to produce consistent results until now.

In this review, considering all the published data together, we show up that there is growing evidence of the involvement of GRIA1 in L-ASP hypersensitivity, for which SNPs located in this gene were significantly associated in three different studies [34], [41], [42]. Thus, those SNPs could be new potential predictors of L-ASP hypersensitivity, but further studies are required. Indeed, the majority of the pharmacogenetic studies analyzing L-ASP hypersensitivity have been conducted in ALL patients treated with native E. coli L-ASP (removed from the market in some countries [89], although it continues being used in the first line of ALL therapy in many others), so it would be interesting to analyze the effects of other formulations in the future. In this context, we think that measurement of antibody assessment and asparaginase levels could become useful tools to predict future allergic reaction or to alert physicians to the possibility of silent hypersensitivity, so further pharmacogenetic studies analyzing L-ASP induced hypersensitivity, including clinical and also silent forms, could be also of interest. Moreover, as ALL is a complex and heterogeneous disease, the subtype of ALL should be also taken into account in these studies. Similarly, information about when hypersensitivity occurs, number of asparaginase injections and co-administrations should be available in the different studies with the goal of optimizing the therapy schedule and limiting this toxicity.

Additionally, data about treatment outcomes and resistance to asparaginase therapy are also interesting. In this line, recently, it has been demonstrated that KMT2E-ASNS fusion transcript contributes to acquired chemotherapy resistance and relapse in ETP-ALL through ASNS over-expression [90].

On the other hand, considering that polymorphisms in miRNA genes have been already associated with other childhood B-ALL treatment-related toxicities, but there are no studies in L-ASP toxicity, it would be interesting to analyze polymorphisms in miRNAs regulating GRIA1, HLA-DR1, NFATC2 and ASNS genes. In fact, these genes contain miRNA binding sites, so miRNAs regulating them could also be L-ASP hypersensitivity markers. This field of miRNA pharmacogenomics is very promising; especially if we consider that miRNA expression could be exogenously controlled by blocking the expression of upregulated miRNAs or by restoring the expression of downregulated miRNAs. Finally, other epigenetic regulators, such as DNA methylation, histone modifications and long noncoding RNAs could also have a role in the development of L-ASP hypersensitivity. In this line, it has been already demonstrated that ALL ETV6-RUNX1 subtype displays hypermethylation of ASNS [91], which was associated with a higher sensitivity of leukemic cells to L-ASP [92] in ETV6-RUNX1 patients [93]. This developing field of pharmacoepigenetics has started to produce promising results and epigenetic variants have great potential to be used as biomarkers for personalized therapy. Therefore, future studies in this area could be a breakthrough in L-ASP pharmacogenetics.

  1. Author contributions: All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

  2. Research funding: None declared.

  3. Employment or leadership: None declared.

  4. Honorarium: None declared.

  5. Competing interests: The funding organization(s) played no role in the study design; in the collection, analysis, and interpretation of data; in the writing of the report; or in the decision to submit the report for publication.

References

1. Pui CH, Evans WE. Treatment of acute lymphoblastic leukemia. N Engl J Med 2006;354:166–78.10.1056/NEJMra052603Suche in Google Scholar PubMed

2. Hunger SP, Lu X, Devidas M, Camitta BM, Gaynon PS, Winick NJ, et al. Improved survival for children and adolescents with acute lymphoblastic leukemia between 1990 and 2005: a report from the children’s oncology group. J Clin Oncol 2012;30:1663–9.10.1200/JCO.2011.37.8018Suche in Google Scholar PubMed PubMed Central

3. Möricke A, Zimmermann M, Reiter A, Henze G, Schrauder A, Gadner H, et al. Long-term results of five consecutive trials in childhood acute lymphoblastic leukemia performed by the ALL-BFM study group from 1981 to 2000. Leukemia 2010;24:265–84.10.1038/leu.2009.257Suche in Google Scholar PubMed

4. Pui CH, Campana D, Pei D, Bowman WP, Sandlund JT, Kaste SC, et al. Treating childhood acute lymphoblastic leukemia without cranial irradiation. N Engl J Med 2009;360:2730–41.10.1056/NEJMoa0900386Suche in Google Scholar PubMed PubMed Central

5. Lopez-Lopez E, Gutierrez-Camino A, Bilbao-Aldaiturriaga N, Pombar-Gomez M, Martin-Guerrero I, Garcia-Orad A. Pharmacogenetics of childhood acute lymphoblastic leukemia. Pharmacogenomics 2014;15:1383–98.10.2217/pgs.14.106Suche in Google Scholar PubMed

6. Cheok MH, Evans WE. Acute lymphoblastic leukaemia: a model for the pharmacogenomics of cancer therapy. Nat Rev Cancer 2006;6:117–29.10.1038/nrc1800Suche in Google Scholar PubMed

7. Paugh SW, Stocco G, McCorkle JR, Diouf B, Crews KR, Evans WE. Cancer pharmacogenomics. Clin Pharmacol Ther 2011;90:461–6.10.1038/clpt.2011.126Suche in Google Scholar PubMed PubMed Central

8. Wall AM, Rubnitz JE. Pharmacogenomic effects on therapy for acute lymphoblastic leukemia in children. Pharmacogenomics J 2003;3:128–35.10.1038/sj.tpj.6500174Suche in Google Scholar PubMed

9. Cheok MH, Pottier N, Kager L, Evans WE. Pharmacogenetics in acute lymphoblastic leukemia. Semin Hematol 2009;46:39–51.10.1053/j.seminhematol.2008.09.002Suche in Google Scholar PubMed PubMed Central

10. López-López E, Gutiérrez-Camino Á, Piñán M, Sánchez-Toledo J, Uriz JJ, Ballesteros J, et al. Pharmacogenetics of microRNAs and microRNAs biogenesis machinery in pediatric acute lymphoblastic leukemia. PLoS One 2014;9:e91261.10.1371/journal.pone.0091261Suche in Google Scholar PubMed PubMed Central

11. Lopez-Lopez E, Gutierrez-Camino A, Astigarraga I, Navajas A, Echebarria-Barona A, Garcia-Miguel P, et al. Vincristine pharmacokinetics pathway and neurotoxicity during early phases of treatment in pediatric acute lymphoblastic leukemia. Pharmacogenomics 2016;17:731–41.10.2217/pgs-2016-0001Suche in Google Scholar PubMed

12. Iparraguirre L, Gutierrez-Camino A, Umerez M, Martin-Guerrero I, Astigarraga I, Navajas A, et al. MiR-pharmacogenetics of methotrexate in childhood B-cell acute lymphoblastic leukemia. Pharmacogenet Genomics 2016;26:517–25.10.1097/FPC.0000000000000245Suche in Google Scholar PubMed

13. Mei L, Ontiveros EP, Griffiths EA, Thompson JE, Wang ES, Wetzler M. Pharmacogenetics predictive of response and toxicity in acute lymphoblastic leukemia therapy. Blood Rev 2015;29:243–9.10.1016/j.blre.2015.01.001Suche in Google Scholar PubMed PubMed Central

14. Pui CH. Genomic and pharmacogenetic studies of childhood acute lymphoblastic leukemia. Front Med 2015;9:1–9.10.1007/s11684-015-0381-3Suche in Google Scholar PubMed

15. Dulucq S, Laverdière C, Sinnett D, Krajinovic M. Pharmacogenetic considerations for acute lymphoblastic leukemia therapies. Expert Opin Drug Metab Toxicol 2014;10:699–719.10.1517/17425255.2014.893294Suche in Google Scholar PubMed

16. Relling MV, Ramsey LB. Pharmacogenomics of acute lymphoid leukemia: new insights into treatment toxicity and efficacy. Hematol Am Soc Hematol Educ Program 2013;2013:126–30.10.1182/asheducation-2013.1.126Suche in Google Scholar PubMed

17. Ho DH, Whitecar JP, Luce JK, Frei E. L-asparagine requirement and the effect of L-asparaginase on the normal and leukemic human bone marrow. Cancer Res 1970;30:466–72.Suche in Google Scholar

18. Onuma T, Waligunda J, Holland JF. Amino acid requirements in vitro of human leukemic cells. Cancer Res 1971;31:1640–4.Suche in Google Scholar

19. Avramis VI, Sencer S, Periclou AP, Sather H, Bostrom BC, Cohen LJ, et al. A randomized comparison of native Escherichia coli asparaginase and polyethylene glycol conjugated asparaginase for treatment of children with newly diagnosed standard-risk acute lymphoblastic leukemia: a Children’s Cancer Group study. Blood 2002;99:1986–94.10.1182/blood.V99.6.1986Suche in Google Scholar

20. Silverman LB, Declerck L, Gelber RD, Dalton VK, Asselin BL, Barr RD, et al. Results of Dana-Farber cancer institute consortium protocols for children with newly diagnosed acute lymphoblastic leukemia (1981–1995). Leukemia 2000;14:2247–56.10.1038/sj.leu.2401980Suche in Google Scholar PubMed

21. Pieters R, Hunger SP, Boos J, Rizzari C, Silverman L, Baruchel A, et al. L-asparaginase treatment in acute lymphoblastic leukemia: a focus on Erwinia asparaginase. Cancer 2011;117:238–49.10.1002/cncr.25489Suche in Google Scholar PubMed PubMed Central

22. Panosyan EH, Seibel NL, Martin-Aragon S, Gaynon PS, Avramis IA, Sather H, et al. Asparaginase antibody and asparaginase activity in children with higher-risk acute lymphoblastic leukemia: Children’s Cancer Group Study CCG-1961. J Pediatr Hematol Oncol 2004;26:217–26.10.1097/00043426-200404000-00002Suche in Google Scholar PubMed

23. Müller HJ, Beier R, Löning L, Blütters-Sawatzki R, Dörffel W, Maass E, et al. Pharmacokinetics of native Escherichia coli asparaginase (Asparaginasemedac) and hypersensitivity reactions in ALL-BFM 95 reinduction treatment. Br J Haematol 2001;114:794–9.10.1046/j.1365-2141.2001.03009.xSuche in Google Scholar

24. Woo MH, Hak LJ, Storm MC, Sandlund JT, Ribeiro RC, Rivera GK, et al. Hypersensitivity or development of antibodies to asparaginase does not impact treatment outcome of childhood acute lymphoblastic leukemia. J Clin Oncol 2000;18:1525–32.10.1200/JCO.2000.18.7.1525Suche in Google Scholar

25. Vrooman LM, Supko JG, Neuberg DS, Asselin BL, Athale UH, Clavell L, et al. Erwiniaasparaginase after allergy to E. coli asparaginase in children with acute lymphoblastic leukemia. Pediatr Blood Cancer 2010;54:199–205.10.1002/pbc.22225Suche in Google Scholar

26. Douer D, Aldoss I, Lunning MA, Burke PW, Ramezani L, Mark L, et al. Pharmacokinetics-based integration of multiple doses of intravenous pegaspargase in a pediatric regimen for adults with newly diagnosed acute lymphoblastic leukemia. J Clin Oncol 2014;32:905–11.10.1200/JCO.2013.50.2708Suche in Google Scholar

27. DeAngelo DJ, Stevenson K, Neuberg DS. A multicenter phase II study using a dose intensified pegylated-asparaginase pediatric regimen in adults with untreated acute lymphoblastic leukemia: a DFCI ALL consortium trial. Blood 2015;126:80.10.1182/blood.V126.23.80.80Suche in Google Scholar

28. Advani AS, Sanford B, Luger S, Devidas M, Larsen EC, Liedtke M, et al. Frontline-treatment of acute lymphoblastic leukemia (ALL) In older adolescents and young adults (AYA) using a pediatric regimen is feasible: toxicity results of the prospective US intergroup trial C10403 (Alliance). Blood 2013;122:3903.10.1182/blood.V122.21.3903.3903Suche in Google Scholar

29. Salzer WL, Asselin B, Supko JG, Devidas M, Kaiser NA, Plourde P, et al. Erwinia asparaginase achieves therapeutic activity after pegaspargase allergy: a report from the Children’s Oncology Group. Blood 2013;122:507–14.10.1182/blood-2013-01-480822Suche in Google Scholar

30. Plourde PV, Jeha S, Hijiya N, Keller FG, Silverman LB, Rheingold SR, et al. Safety profile of asparaginase Erwiniachrysanthemi in a large compassionate-use trial. Pediatr Blood Cancer 2014;61:1232–8.10.1002/pbc.24938Suche in Google Scholar

31. Billett AL, Carls A, Gelber RD, Sallan SE. Allergic reactions to Erwiniaasparaginase in children with acute lymphoblastic leukemia who had previous allergic reactions to Escherichia coli asparaginase. Cancer 1992;70:201–6.10.1002/1097-0142(19920701)70:1<201::AID-CNCR2820700131>3.0.CO;2-MSuche in Google Scholar

32. Wenner KA, Vieira Pinheiro JP, Escherich G, Wessalowski R, Jorch N, Wolff J, et al. Asparagine concentration in plasma after 2,500 IU/m(2) PEG-asparaginasei.v. in children with acute lymphoblastic leukemia. Klin Paediatr 2005;217:321–6.10.1055/s-2005-872516Suche in Google Scholar

33. Stock W, Douer D, DeAngelo DJ, Arellano M, Advani A, Damon L, et al. Prevention and management of asparaginase/pegasparaginase-associated toxicities in adults and older adolescents: recommendations of an expert panel. Leuk Lymphoma 2011;52:2237–53.10.3109/10428194.2011.596963Suche in Google Scholar PubMed

34. Kutszegi N, Semsei Á, Gézsi A, Sági JC, Nagy V, Csordás K, et al. Subgroups of paediatric acute lymphoblastic leukaemia might differ significantly in genetic predisposition to asparaginase hypersensitivity. PLoS One 2015;10:e0140136.10.1371/journal.pone.0140136Suche in Google Scholar PubMed PubMed Central

35. Kawedia JD, Rytting ME. Asparaginase in acute lymphoblastic leukemia. Clin Lymphoma Myeloma Leuk 2014;14:S14–17.10.1016/j.clml.2014.06.017Suche in Google Scholar PubMed

36. Silverman LB, Gelber RD, Dalton VK, Asselin BL, Barr RD, Clavell LA, et al. Improved outcome for children with acute lymphoblastic leukemia: results of Dana-Farber Consortium Protocol 91-01. Blood 2001;97:1211–18.10.1182/blood.V97.5.1211Suche in Google Scholar

37. Zalewska-Szewczyk B, Andrzejewski W, Młynarski W, Jedrychowska-Dańska K, Witas H, Bodalski J. The anti-asparagines antibodies correlate with L-asparagines activity and may affect clinical outcome of childhood acute lymphoblastic leukemia. Leuk Lymphoma 2007;48:931–6.10.1080/10428190701292049Suche in Google Scholar PubMed

38. Kearney SL, Dahlberg SE, Levy DE, Voss SD, Sallan SE, Silverman LB. Clinical course and outcome in children with acute lymphoblastic leukemia and asparaginase-associated pancreatitis. Pediatr Blood Cancer 2009;53:162–7.10.1002/pbc.22076Suche in Google Scholar PubMed PubMed Central

39. Earl M. Incidence and management of asparaginase-associated adverse events in patients with acute lymphoblastic leukemia. Clin Adv Hematol Oncol 2009;7:600–6.Suche in Google Scholar

40. Bhojwani D, Darbandi R, Pei D, Ramsey LB, Chemaitilly W, Sandlund JT, et al. Severe hypertriglyceridaemia during therapy for childhood acute lymphoblastic leukaemia. Eur J Cancer 2014;50:2685–94.10.1016/j.ejca.2014.06.023Suche in Google Scholar PubMed PubMed Central

41. Chen SH, Pei D, Yang W, Cheng C, Jeha S, Cox NJ, et al. Genetic variations in GRIA1 on chromosome 5q33 related to asparaginase hypersensitivity. Clin Pharmacol Ther 2010;88:191–6.10.1038/clpt.2010.94Suche in Google Scholar PubMed PubMed Central

42. Rajić V, Debeljak M, Goričar K, Jazbec J. Polymorphisms in GRIA1 gene are a risk factor for asparaginase hypersensitivity during the treatment of childhood acute lymphoblastic leukemia. Leuk Lymphoma 2015;56:3103–8.10.3109/10428194.2015.1020802Suche in Google Scholar PubMed

43. Fernandez CA, Smith C, Yang W, Daté M, Bashford D, Larsen E, et al. HLA-DRB1*07:01 is associated with a higher risk of asparaginase allergies. Blood 2014;124:1266–76.10.1182/blood-2014-03-563742Suche in Google Scholar PubMed PubMed Central

44. Fernandez CA, Smith C, Yang W, Mullighan CG, Qu C, Larsen E, et al. Genome-wide analysis links NFATC2 with asparaginase hypersensitivity. Blood 2015;126:69–75.10.1182/blood-2015-02-628800Suche in Google Scholar PubMed PubMed Central

45. Chen SH, Yang W, Fan Y, Stocco G, Crews KR, Yang JJ, et al. A genome-wide approach identifies that the aspartate metabolism pathway contributes to asparaginase sensitivity. Leukemia 2011;25:66–74.10.1038/leu.2010.256Suche in Google Scholar PubMed PubMed Central

46. Rousseau J, Gagné V, Labuda M, Beaubois C, Sinnett D, Laverdière C, et al. ATF5 polymorphisms influence ATF function and response to treatment in children with childhood acute lymphoblastic leukemia. Blood 2011;118:5883–90.10.1182/blood-2011-05-355560Suche in Google Scholar PubMed PubMed Central

47. Pastorczak A, Fendler W, Zalewska-Szewczyk B, Górniak P, Lejman M, Trelińska J, et al. Asparagine synthetase (ASNS) gene polymorphism is associated with the outcome of childhood acute lymphoblastic leukemia by affecting early response to treatment. Leuk Res 2014;38:180–3.10.1016/j.leukres.2013.10.027Suche in Google Scholar PubMed

48. Liu C, Yang W, Devidas M, Cheng C, Pei D, Smith C, et al. Clinical and genetic risk factors for acute pancreatitis in patients with acute lymphoblastic leukemia. J Clin Oncol 2016;34:2133–40.10.1200/JCO.2015.64.5812Suche in Google Scholar PubMed PubMed Central

49. Ben Tanfous M, Sharif-Askari B, Ceppi F, Laaribi H, Gagné V, Rousseau J, et al. Polymorphisms of asparaginase pathway and asparaginase-related complications in children with acute lymphoblastic leukemia. Clin Cancer Res 2015;21:329–34.10.1158/1078-0432.CCR-14-0508Suche in Google Scholar PubMed PubMed Central

50. Franco R, Pacheco R, Lluis C, Ahern GP, O’Connell PJ. The emergence of neurotransmitters as immune modulators. Trends Immunol 2007;28:400–7.10.1016/j.it.2007.07.005Suche in Google Scholar PubMed

51. Pacheco R, Gallart T, Lluis C, Franco R. Role of glutamate on T-cell mediated immunity. J Neuroimmunol 2007;185:9–19.10.1016/j.jneuroim.2007.01.003Suche in Google Scholar PubMed

52. Ganor Y, Besser M, Ben-Zakay N, Unger T, Levite M. Human T cells express a functional ionotropic glutamate receptor GluR3, and glutamate by itself triggers integrin-mediated adhesion to laminin and fibronectin and chemotactic migration. J Immunol 2003;170:4362–72.10.4049/jimmunol.170.8.4362Suche in Google Scholar PubMed

53. Boldyrev AA, Kazey VI, Leinsoo TA, Mashkina AP, Tyulina OV, Johnson P, et al. Rodent lymphocytes express functionally active glutamate receptors. Biochem Biophys Res Commun 2004;324:133–9.10.1016/j.bbrc.2004.09.019Suche in Google Scholar PubMed

54. Boldyrev AA, Carpenter DO, Johnson P. Emerging evidence for a similar role of glutamate receptors in the nervous and immune systems. J Neurochem 2005;95:913–18.10.1111/j.1471-4159.2005.03456.xSuche in Google Scholar

55. Levite M. Neurotransmitters activate T-cells and elicit crucial functions via neurotransmitter receptors. Curr Opin Pharmacol 2008;8:460–71.10.1016/j.coph.2008.05.001Suche in Google Scholar

56. Beyer K, Nickel R, Freidhoff L, Björkstén B, Huang SK, Barnes KC, et al. Association and linkage of atopic dermatitis with chromosome 13q12-14 and 5q31-33 markers. J Invest Dermatol 2000;115:906–8.10.1046/j.1523-1747.2000.00096.xSuche in Google Scholar

57. Blumenthal MN, Langefeld CD, Beaty TH, Bleecker ER, Ober C, Lester L, et al. A genome-wide search for allergic response (atopy) genes in three ethnic groups: collaborative study on the genetics of asthma. Hum Genet 2004;114:157–64.10.1007/s00439-003-1030-5Suche in Google Scholar

58. Xu J, Meyers DA, Ober C, Blumenthal MN, Mellen B, Barnes KC, et al. Genomewide screen and identification of gene-gene interactions for asthma-susceptibility loci in three U.S. populations: collaborative study on the genetics of asthma. Am J Hum Genet 2001;68:1437–46.10.1086/320589Suche in Google Scholar

59. Hizawa N, Freidhoff LR, Ehrlich E, Chiu YF, Duffy DL, Schou C, et al. Genetic influences of chromosomes 5q31-q33 and 11q13 on specific IgE responsiveness to common inhaled allergens among African American families. Collaborative Study on the Genetics of Asthma (CSGA). J Allergy Clin Immunol 1998;102:449–53.10.1016/S0091-6749(98)70134-4Suche in Google Scholar

60. Koppelman GH, Stine OC, Xu J, Howard TD, Zheng SL, Kauffman HF, et al. Genome-wide search for atopy susceptibility genes in Dutch families with asthma. J Allergy Clin Immunol 2002;109:498–506.10.1067/mai.2002.122235Suche in Google Scholar PubMed

61. Noguchi E, Shibasaki M, Arinami T, Takeda K, Maki T, Miyamoto T, et al. Evidence for linkage between asthma/atopy in childhood and chromosome 5q31-q33 in a Japanese population. Am J Respir Crit Care Med 1997;156:1390–3.10.1164/ajrccm.156.5.9702084Suche in Google Scholar PubMed

62. Price P, Witt C, Allcock R, Sayer D, Garlepp M, Kok CC, et al. The genetic basis for the association of the 8.1 ancestral haplotype (A1, B8, DR3) with multiple immunopathological diseases. Immunol Rev 1999;167:257–74.10.1111/j.1600-065X.1999.tb01398.xSuche in Google Scholar

63. Trowsdale J. The MHC, disease and selection. Immunol Lett 2011;137:1–8.10.1016/j.imlet.2011.01.002Suche in Google Scholar PubMed

64. Mallal S, Nolan D, Witt C, Masel G, Martin AM, Moore C, et al. Association between presence of HLA-B*5701, HLA-DR7, and HLA-DQ3 and hypersensitivity to HIV-1 reverse-transcriptase inhibitor abacavir. Lancet 2002;359:727–32.10.1016/S0140-6736(02)07873-XSuche in Google Scholar

65. Hetherington S, Hughes AR, Mosteller M, Shortino D, Baker KL, Spreen W, et al. Genetic variations in HLA-B region and hypersensitivity reactions to abacavir. Lancet 2002;359:1121–2.10.1016/S0140-6736(02)08158-8Suche in Google Scholar

66. Chung WH, Hung SI, Hong HS, Hsih MS, Yang LC, Ho HC, et al. Medical genetics: a marker for Stevens-Johnson syndrome. Nature 2004;428:486.10.1038/428486aSuche in Google Scholar PubMed

67. Hung SI, Chung WH, Liou LB, Chu CC, Lin M, Huang HP, et al. HLA-B*5801 allele as a genetic marker for severe cutaneous adverse reactions caused by allopurinol. Proc Natl Acad Sci USA 2005;102:4134–9.10.1073/pnas.0409500102Suche in Google Scholar PubMed PubMed Central

68. Tassaneeyakul W, Jantararoungtong T, Chen P, Lin PY, Tiamkao S, Khunarkornsiri U, et al. Strong association between HLA-B*5801 and allopurinol-induced Stevens-Johnson syndrome and toxic epidermal necrolysis in a Thai population. Pharmacogenet Genomics 2009;19:704–9.10.1097/FPC.0b013e328330a3b8Suche in Google Scholar PubMed

69. Kaniwa N, Saito Y, Aihara M, Matsunaga K, Tohkin M, Kurose K, et al. HLA-B locus in Japanese patients with anti-epileptics and allopurinol-related Stevens-Johnson syndrome and toxic epidermal necrolysis. Pharmacogenomics 2008;9:1617–22.10.2217/14622416.9.11.1617Suche in Google Scholar PubMed

70. Kindmark A, Jawaid A, Harbron CG, Barratt BJ, Bengtsson OF, Andersson TB, et al. Genome-wide pharmacogenetic investigation of a hepatic adverse event without clinical signs of immunopathology suggests an underlying immune pathogenesis. Pharmacogenomics J 2008;8:186–95.10.1038/sj.tpj.6500458Suche in Google Scholar PubMed

71. Spraggs CF, Budde LR, Briley LP, Bing N, Cox CJ, King KS, et al. HLA-DQA1*02:01 is a major risk factor for lapatinib-induced hepatotoxicity in women with advanced breast cancer. J ClinOncol 2011;29:667–73.10.1200/JCO.2010.31.3197Suche in Google Scholar PubMed

72. Xanthoudakis S, Viola JP, Shaw KT, Luo C, Wallace JD, Bozza PT, et al. An enhanced immune response in mice lacking the transcription factor NFAT1. Science 1996;272:892–5.10.1126/science.272.5263.892Suche in Google Scholar PubMed

73. Karwot R, Übel C, Bopp T, Schmitt E, Finotto S. Increased immunosuppressive function of CD4(+)CD25(+)Foxp3(+)GITR+ T regulatory cells from NFATc2((-/-)) mice controls allergen-induced experimental asthma. Immunobiology 2012;217:905–11.10.1016/j.imbio.2012.01.004Suche in Google Scholar PubMed

74. Nagamoto-Combs K, Combs CK. Microglial phenotype is regulated by activity of the transcription factor, NFAT (nuclear factor of activated T cells). J Neurosci 2010;30:9641–6.10.1523/JNEUROSCI.0828-10.2010Suche in Google Scholar PubMed PubMed Central

75. Choi JM, Sohn JH, Park TY, Park JW, Lee SK. Cell permeable NFAT inhibitory peptide Sim-2-VIVIT inhibits T-cell activation and alleviates allergic airway inflammation and hyper-responsiveness. Immunol Lett 2012;143:170–6.10.1016/j.imlet.2012.01.016Suche in Google Scholar PubMed

76. Elloumi HZ, Maharshak N, Rao KN, Kobayashi T, Ryu HS, Mühlbauer M, et al. A cell permeable peptide inhibitor of NFAT inhibits macrophage cytokine expression and ameliorates experimental colitis. PLoS One 2012;7:e34172.10.1371/journal.pone.0034172Suche in Google Scholar PubMed PubMed Central

77. Noguchi H, Matsushita M, Okitsu T, Moriwaki A, Tomizawa K, Kang S, et al. A new cell-permeable peptide allows successful allogeneic islet transplantation in mice. Nat Med 2004;10:305–9.10.1038/nm994Suche in Google Scholar PubMed

78. Kuriyama M, Matsushita M, Tateishi A, Moriwaki A, Tomizawa K, Ishino K, et al. A cell-permeable NFAT inhibitor peptide prevents pressure-overload cardiac hypertrophy. ChemBiol Drug Des 2006;67:238–43.10.1111/j.1747-0285.2006.00360.xSuche in Google Scholar PubMed

79. Zhang S, Luo L, Wang Y, Gomez MF, Thorlacius H. Nuclear factor of activated T cells regulates neutrophil recruitment, systemic inflammation, and T-cell dysfunction in abdominal sepsis. Infect Immun 2014;82:3275–88.10.1128/IAI.01569-14Suche in Google Scholar PubMed PubMed Central

80. McPherson JD, Marra M, Hillier L, Waterston RH, Chinwalla A, Wallis J, et al. A physical map of the human genome. Nature 2001;409:934–41.10.1038/35057157Suche in Google Scholar PubMed

81. Rukov JL, Shomron N. MicroRNA pharmacogenomics: post-transcriptional regulation of drug response. Trends Mol Med 2011;17:412–23.10.1016/j.molmed.2011.04.003Suche in Google Scholar PubMed

82. Mishra PJ, Humeniuk R, Longo-Sorbello GS, Banerjee D, Bertino JR. A miR-24 microRNA binding-site polymorphism in dihydrofolate reductase gene leads to methotrexate resistance. Proc Natl Acad Sci USA 2007;104:13513–8.10.1073/pnas.0706217104Suche in Google Scholar PubMed PubMed Central

83. Han BW, Feng DD, Li ZG, Luo XQ, Zhang H, Li XJ, et al. A set of miRNAs that involve in the pathways of drug resistance and leukemic stem-cell differentiation is associated with the risk of relapse and glucocorticoid response in childhood ALL. Hum Mol Genet 2011;20:4903–15.10.1093/hmg/ddr428Suche in Google Scholar PubMed PubMed Central

84. Wang SM, Sun LL, Zeng WX, Wu WS, Zhang GL. Effects of a microRNA binding site polymorphism in SLC19A1 on methotrexate concentrations in Chinese children with acute lymphoblastic leukemia. Med Oncol 2014;31:62.10.1007/s12032-014-0062-0Suche in Google Scholar PubMed

85. AkbariMoqadam F, Lange-Turenhout EA, Ariës IM, Pieters R, den Boer ML. MiR-125b, miR-100 and miR-99a co-regulate vincristine resistance in childhood acute lymphoblastic leukemia. Leuk Res 2013;37:1315–21.10.1016/j.leukres.2013.06.027Suche in Google Scholar PubMed

86. Schotte D, De Menezes RX, Akbari Moqadam F, Khankahdani LM, Lange-Turenhout E, Chen C, et al. MicroRNA characterize genetic diversity and drug resistance in pediatric acute lymphoblastic leukemia. Haematologica 2011;96:703–11.10.3324/haematol.2010.026138Suche in Google Scholar PubMed PubMed Central

87. Mei Y, Gao C, Wang K, Cui L, Li W, Zhao X, et al. Effect of microRNA-210 on prognosis and response to chemotherapeutic drugs in pediatric acute lymphoblastic leukemia. Cancer Sci 2014;105:463–72.10.1111/cas.12370Suche in Google Scholar PubMed PubMed Central

88. MesrianTanha H, MojtabaviNaeini M, Rahgozar S, Moafi A, Honardoost MA. Integrative computational in-depth analysis of dysregulated miRNA-mRNA interactions in drug-resistant pediatric acute lymphoblastic leukemia cells: an attempt to obtain new potential gene-miRNA pathways involved in response to treatment. Tumour Biol 2016;37:7861–72.10.1007/s13277-015-4553-1Suche in Google Scholar PubMed

89. RE: Discontinuation of Elspar, (asparaginase for injection) 10,000 IU. 2012. (accessed 03.03.16) at http://www.fda.gov/downloads/drugs/drugsafety/drugshortages/ucm321556.pdf.Suche in Google Scholar

90. Khater F, Lajoie M, Langlois S, Healy J, Cellot S, Richer C, et al. KMT2E-ASNS: a novel relapse-specific fusion gene in early T-cell precursor acute lymphoblastic leukemia. Blood 2017;blood-2016-10-744219 [Epub ahead of print].10.1182/blood-2016-10-744219Suche in Google Scholar PubMed PubMed Central

91. Chatterton Z, Morenos L, Mechinaud F, Ashley DM, Craig JM, Sexton-Oates A, et al. Epigenetic deregulation in pediatric acute lymphoblastic leukemia. Epigenetics 2014;9:459–67.10.4161/epi.27585Suche in Google Scholar PubMed PubMed Central

92. Li BS, Gu LJ, Luo CY, Li WS, Jiang LM, Shen SH, et al. The downregulation of asparagine synthetase expression can increase the sensitivity of cells resistant to l-asparaginase. Leukemia 2006;20:2199–201.10.1038/sj.leu.2404423Suche in Google Scholar PubMed

93. Ramakers-van Woerden NL, Pieters R, Loonen AH, Hubeek I, van Drunen E, Beverloo HB, et al. TEL/AML1 gene fusion is related to in vitro drug sensitivity for L-asparaginase in childhood acute lymphoblastic leukemia. Blood 2000;96:1094–9.Suche in Google Scholar


Supplemental Material:

The online version of this article (DOI: 10.1515/dmpt-2016-0033) offers supplementary material, available to authorized users.


Received: 2016-10-5
Accepted: 2017-2-7
Published Online: 2017-3-6
Published in Print: 2017-3-1

©2017 Walter de Gruyter GmbH, Berlin/Boston

Heruntergeladen am 27.9.2025 von https://www.degruyterbrill.com/document/doi/10.1515/dmpt-2016-0033/html
Button zum nach oben scrollen