Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 152
Filter
1.
Blood Adv ; 7(11): 2538-2550, 2023 06 13.
Article in English | MEDLINE | ID: mdl-36689724

ABSTRACT

Cytarabine arabinoside (Ara-C) has been the cornerstone of acute myeloid leukemia (AML) chemotherapy for decades. After cellular uptake, it is phosphorylated into its active triphosphate form (Ara-CTP), which primarily exerts its cytotoxic effects by inhibiting DNA synthesis in proliferating cells. Interpatient variation in the enzymes involved in the Ara-C metabolic pathway has been shown to affect intracellular abundance of Ara-CTP and, thus, its therapeutic benefit. Recently, SAMHD1 (SAM and HD domain-containing deoxynucleoside triphosphate triphosphohydrolase 1) has emerged to play a role in Ara-CTP inactivation, development of drug resistance, and, consequently, clinical response in AML. Despite this, the impact of genetic variations in SAMHD1 on outcome in AML has not been investigated in depth. In this study, we evaluated 25 single nucleotide polymorphisms (SNPs) within the SAMHD1 gene for association with clinical outcome in 400 pediatric patients with newly diagnosed AML from 2 clinical trials, AML02 and AML08. Three SNPs, rs1291128, rs1291141, and rs7265241 located in the 3' region of SAMHD1 were significantly associated with at least 1 clinical outcome: minimal residual disease after induction I, event-free survival (EFS), or overall survival (OS) in the 2 cohorts. In an independent cohort of patients from the COG-AAML1031 trial (n = 854), rs7265241 A>G remained significantly associated with EFS and OS. In multivariable analysis, all the SNPs remained independent predictors of clinical outcome. These results highlight the relevance of the SAMHD1 pharmacogenomics in context of response to Ara-C in AML and warrants the need for further validation in expanded patient cohorts.


Subject(s)
Leukemia, Myeloid, Acute , SAM Domain and HD Domain-Containing Protein 1 , Child , Humans , Arabinofuranosylcytosine Triphosphate/metabolism , Arabinofuranosylcytosine Triphosphate/therapeutic use , Cytarabine/therapeutic use , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Polymorphism, Single Nucleotide , SAM Domain and HD Domain-Containing Protein 1/genetics
2.
J Pharm Biomed Anal ; 211: 114582, 2022 Mar 20.
Article in English | MEDLINE | ID: mdl-35101802

ABSTRACT

In this study, a sensitive and rapid ultra-high-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) method was developed for the simultaneous analysis of cytarabine (ara-C), cytarabine monophosphate (ara-CMP), cytarabine diphosphate (ara-CDP) and cytarabine triphosphate (ara-CTP) in the cytosol and nucleus. The separation of analytes and endogenous interferents was achieved in 8 min on a hypercarb column (2.1 mm × 100 mm, 3 µm) by using a gradient elution with 95% acetonitrile and aqueous 5 mM hexylamine with 0.4% (v/v) diethylamine adjusted to pH 10. The analytes were detected with both negative and positive electrospray ionization in multiple reaction monitoring (MRM) mode. The calibration curve demonstrated good linearity ranging from 5 to 750 nM for ara-C, 50-7500 nM for ara-CMP, 20-3000 nM for ara-CDP and 1-150 nM for ara-CTP in the cytosol. In the nucleus, good linearity was achieved over a concentration range of 1-100 nM for ara-C, 5-500 nM for ara-CMP, 2.5-250 nM for ara-CDP and 0.5-50 nM for ara-CTP. Intra- and interbatch accuracies and precisions met the standards of validation. The matrix effect, recovery and stability were also within acceptable ranges. After incubation with 10 µM ara-C for 3 h, the levels of ara-C, ara-CMP, ara-CDP and ara-CTP in the cytosol and nucleus of HL-60 cells and HL-60/ara-C cells were determined. Most of the metabolites were found within the quantitation range. The results showed that the nuclear ara-CTP level was significantly different than the intracellular ara-CTP level between HL-60 and HL-60/ara-C cells.


Subject(s)
Arabinofuranosylcytosine Triphosphate , Cytarabine , Arabinofuranosylcytosine Triphosphate/analysis , Arabinofuranosylcytosine Triphosphate/metabolism , Chromatography, High Pressure Liquid/methods , Cytosol/metabolism , Diphosphates , Humans , Tandem Mass Spectrometry
3.
EMBO Mol Med ; 12(3): e10419, 2020 03 06.
Article in English | MEDLINE | ID: mdl-31950591

ABSTRACT

The deoxycytidine analogue cytarabine (ara-C) remains the backbone treatment of acute myeloid leukaemia (AML) as well as other haematological and lymphoid malignancies, but must be combined with other chemotherapeutics to achieve cure. Yet, the underlying mechanism dictating synergistic efficacy of combination chemotherapy remains largely unknown. The dNTPase SAMHD1, which regulates dNTP homoeostasis antagonistically to ribonucleotide reductase (RNR), limits ara-C efficacy by hydrolysing the active triphosphate metabolite ara-CTP. Here, we report that clinically used inhibitors of RNR, such as gemcitabine and hydroxyurea, overcome the SAMHD1-mediated barrier to ara-C efficacy in primary blasts and mouse models of AML, displaying SAMHD1-dependent synergy with ara-C. We present evidence that this is mediated by dNTP pool imbalances leading to allosteric reduction of SAMHD1 ara-CTPase activity. Thus, SAMHD1 constitutes a novel biomarker for combination therapies of ara-C and RNR inhibitors with immediate consequences for clinical practice to improve treatment of AML.


Subject(s)
Cytarabine/pharmacology , Leukemia, Myeloid, Acute , Pyrophosphatases/metabolism , Ribonucleotide Reductases/antagonists & inhibitors , SAM Domain and HD Domain-Containing Protein 1/metabolism , Animals , Arabinofuranosylcytosine Triphosphate/metabolism , Mice
4.
Nat Med ; 23(2): 256-263, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28067901

ABSTRACT

The cytostatic deoxycytidine analog cytarabine (ara-C) is the most active agent available against acute myelogenous leukemia (AML). Together with anthracyclines, ara-C forms the backbone of AML treatment for children and adults. In AML, both the cytotoxicity of ara-C in vitro and the clinical response to ara-C therapy are correlated with the ability of AML blasts to accumulate the active metabolite ara-C triphosphate (ara-CTP), which causes DNA damage through perturbation of DNA synthesis. Differences in expression levels of known transporters or metabolic enzymes relevant to ara-C only partially account for patient-specific differential ara-CTP accumulation in AML blasts and response to ara-C treatment. Here we demonstrate that the deoxynucleoside triphosphate (dNTP) triphosphohydrolase SAM domain and HD domain 1 (SAMHD1) promotes the detoxification of intracellular ara-CTP pools. Recombinant SAMHD1 exhibited ara-CTPase activity in vitro, and cells in which SAMHD1 expression was transiently reduced by treatment with the simian immunodeficiency virus (SIV) protein Vpx were dramatically more sensitive to ara-C-induced cytotoxicity. CRISPR-Cas9-mediated disruption of the gene encoding SAMHD1 sensitized cells to ara-C, and this sensitivity could be abrogated by ectopic expression of wild-type (WT), but not dNTPase-deficient, SAMHD1. Mouse models of AML lacking SAMHD1 were hypersensitive to ara-C, and treatment ex vivo with Vpx sensitized primary patient-derived AML blasts to ara-C. Finally, we identified SAMHD1 as a risk factor in cohorts of both pediatric and adult patients with de novo AML who received ara-C treatment. Thus, SAMHD1 expression levels dictate patient sensitivity to ara-C, providing proof-of-concept that the targeting of SAMHD1 by Vpx could be an attractive therapeutic strategy for potentiating ara-C efficacy in hematological malignancies.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Cytarabine/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Monomeric GTP-Binding Proteins/drug effects , Viral Regulatory and Accessory Proteins/pharmacology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Antimetabolites, Antineoplastic/therapeutic use , Arabinofuranosylcytosine Triphosphate/metabolism , Child , Child, Preschool , Cytarabine/therapeutic use , Disease Models, Animal , Female , Humans , In Vitro Techniques , Infant , Leukemia, Myeloid, Acute/metabolism , Male , Mice , Molecular Targeted Therapy , Monomeric GTP-Binding Proteins/metabolism , Prognosis , SAM Domain and HD Domain-Containing Protein 1
5.
DNA Repair (Amst) ; 49: 51-59, 2017 01.
Article in English | MEDLINE | ID: mdl-27989484

ABSTRACT

Human PrimPol is a recently discovered bifunctional enzyme that displays DNA template-directed primase and polymerase activities. PrimPol has been implicated in nuclear and mitochondrial DNA replication fork progression and restart as well as DNA lesion bypass. Published evidence suggests that PrimPol is a Mn2+-dependent enzyme as it shows significantly improved primase and polymerase activities when binding Mn2+, rather than Mg2+, as a divalent metal ion cofactor. Consistently, our fluorescence anisotropy assays determined that PrimPol binds to a primer/template DNA substrate with affinities of 29 and 979nM in the presence of Mn2+ and Mg2+, respectively. Our pre-steady-state kinetic analysis revealed that PrimPol incorporates correct dNTPs with 100-fold higher efficiency with Mn2+ than with Mg2+. Notably, the substitution fidelity of PrimPol in the presence of Mn2+ was determined to be in the range of 3.4×10-2 to 3.8×10-1, indicating that PrimPol is an error-prone polymerase. Furthermore, we kinetically determined the sugar selectivity of PrimPol to be 57-1800 with Mn2+ and 150-4500 with Mg2+, and found that PrimPol was able to incorporate the triphosphates of two anticancer drugs (cytarabine and gemcitabine), but not two antiviral drugs (emtricitabine and lamivudine).


Subject(s)
Coenzymes/metabolism , DNA Primase/metabolism , DNA Replication , DNA-Directed DNA Polymerase/metabolism , DNA/metabolism , Magnesium/metabolism , Manganese/metabolism , Multifunctional Enzymes/metabolism , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Antiviral Agents/metabolism , Antiviral Agents/therapeutic use , Arabinofuranosylcytosine Triphosphate/metabolism , Arabinofuranosylcytosine Triphosphate/therapeutic use , Cations, Divalent/metabolism , Cytidine Triphosphate/analogs & derivatives , Cytidine Triphosphate/metabolism , Cytidine Triphosphate/therapeutic use , Deoxyribonucleotides/metabolism , Dideoxynucleotides/metabolism , Dideoxynucleotides/therapeutic use , Emtricitabine/analogs & derivatives , Emtricitabine/metabolism , Emtricitabine/therapeutic use , Humans , Kinetics , Lamivudine/analogs & derivatives , Lamivudine/metabolism , Lamivudine/therapeutic use
6.
Adv Biochem Eng Biotechnol ; 145: 189-214, 2014.
Article in English | MEDLINE | ID: mdl-25216956

ABSTRACT

Bioluminescent bacterial biosensors can be used in a rapid in vitro assay to predict sensitivity to commonly used chemotherapy drugs in acute myeloid leukemia (AML). The nucleoside analog cytarabine (ara-C) is the key agent for treating AML; however, up to 30 % of patients fail to respond to treatment. Screening of patient blood samples to determine drug response before commencement of treatment is needed. To achieve this aim, a self-bioluminescent reporter strain of Escherichia coli has been constructed and evaluated for use as an ara-C biosensor and an in vitro assay has been designed to predict ara-C response in clinical samples. Transposition mutagenesis was used to create a cytidine deaminase (cdd)-deficient mutant of E. coli MG1655 that responded to ara-C. The strain was transformed with the luxCDABE operon and used as a whole-cell biosensor for development an 8-h assay to determine ara-C uptake and phosphorylation by leukemic cells. Intracellular concentrations of 0.025 µmol/L phosphorylated ara-C were detected by significantly increased light output (P < 0.05) from the bacterial biosensor. Results using AML cell lines with known response to ara-C showed close correlation between the 8-h assay and a 3-day cytotoxicity test for ara-C cell killing. In retrospective tests with 24 clinical samples of bone marrow or peripheral blood, the biosensor-based assay predicted leukemic cell response to ara-C within 8 h. The biosensor-based assay may offer a predictor for evaluating the sensitivity of leukemic cells to ara-C before patients undergo chemotherapy and allow customized treatment of drug-sensitive patients with reduced ara-C dose levels. The 8-h assay monitors intracellular ara-CTP (cytosine arabinoside triphosphate) levels and, if fully validated, may be suitable for use in clinical settings.


Subject(s)
Antimetabolites, Antineoplastic/metabolism , Arabinofuranosylcytosine Triphosphate/analysis , Biological Assay , Cytarabine/metabolism , Drug Screening Assays, Antitumor , Antimetabolites, Antineoplastic/pharmacology , Arabinofuranosylcytosine Triphosphate/metabolism , Biosensing Techniques , Cell Line, Tumor , Cytarabine/pharmacology , Escherichia coli/genetics , Escherichia coli/metabolism , Genes, Reporter , Genetic Engineering , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Limit of Detection , Luciferases, Bacterial/genetics , Luciferases, Bacterial/metabolism , Luminescent Measurements , Operon , Phosphorylation
7.
Front Biosci (Landmark Ed) ; 17(2): 569-74, 2012 01 01.
Article in English | MEDLINE | ID: mdl-22201761

ABSTRACT

Cytarabine (araC) is a highly active antimetabolite against hematological malignancy while the agent shows limited activity for some patients despite maintenance or continued therapy with ara-C-containing regiments. In this study, we focused to elucidate the mechanism of intrinsic resistance to araC. The concentration of intracellular ara-CTP and incorporated ara-CTP were monitored in human leukemia cell line-HL-60 for different passages in parental with its variant HL-60R. The expression of mRNA for deoxycytidine kinase (dCK), cytidine deaminas (CDA), human equilibrative nucleoside transporter 1 (hENT1), and cytosolic 50-nucleotidase II (cN-II) were examined by Real-time PCR in HL-60 and HL-60R for different passages. And activities of two metabolizing enzymes for araC, dCK and CDA were further examined. The results showed that the concentration of intracellular ara-CTP was significantly reduced and the ara-U increased in HL-60 cells for 50 passages compared with the 5 passages, and associated with higher CDA activity. All the factors in HL-60R cells did not change by the incubation of ara-C. In conclusion, the long term cultured cells are intrinsically resistant to ara-C through high CDA activity, but not low DCK activity.


Subject(s)
Cytarabine/pharmacology , Cytidine Deaminase/metabolism , Drug Resistance, Neoplasm/physiology , Arabinofuranosylcytosine Triphosphate/metabolism , Arabinofuranosyluracil/metabolism , Cytidine Deaminase/genetics , HL-60 Cells , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
8.
Pharmacogenomics ; 10(10): 1657-74, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19842938

ABSTRACT

The mainstay of acute myeloid leukemia chemotherapy is the nucleoside analog cytarabine (ara-C). Numerous studies suggest that the intracellular concentrations of the ara-C active metabolite, ara-CTP, vary widely among patients and, in turn, are associated with variability in clinical response to acute myeloid leukemia treatment. Thus, genetic variation in key genes in the ara-C metabolic pathway--specifically, deoxycytidine kinase (a rate-limiting activating enzyme), 5 nucleotidase, cytidine deaminase and deoxycytidylate deaminase (all three are inactivating enzymes), human equilibrative nucleoside transporter (ara-C uptake transporter) and ribonucleotide reductase (RRM1 and RRM2--enzymes regulating intracellular deoxycytidine triphosphate pools)--form the molecular basis of the interpatient variability observed in intracellular ara-CTP concentrations and response to ara-C. Understanding genetic variants in the key candidate genes involved in the metabolic activation of ara-C, as well as the pharmacodynamic targets of ara-C, will provide an opportunity to identify patients at an increased risk of adverse reactions or decreased likelihood of response, based upon their genetic profile, which in future could help in dose optimization to reduce drug toxicity without compromising efficacy. The pharmacogenetic studies on ara-C would also be equally applicable to other nucleoside analogs, such as gemcitabine, decitabine, clofarabine and so on, which are metabolized by the same pathway.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Cytarabine/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , 5'-Nucleotidase/genetics , Arabinofuranosylcytosine Triphosphate/metabolism , Cytidine Deaminase/genetics , Deoxycytidine Kinase/genetics , Equilibrative Nucleoside Transporter 1/genetics , Forecasting , Genetic Variation , Humans , Leukemia, Myeloid, Acute/genetics , Nucleoside Deaminases/genetics , Pharmacogenetics/methods , Ribonucleotide Reductases/genetics
9.
Biochem Pharmacol ; 77(12): 1780-6, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19428333

ABSTRACT

Cytarabine (ara-C) is the key agent for treating acute myeloid leukemia (AML). After being transported into leukemic cells by human equilibrative nucleoside transporter 1 (hENT1), ara-C is phosphorylated to ara-C triphosphate (ara-CTP), an active metabolite, and then incorporated into DNA, thereby inhibiting DNA synthesis. Deoxycytidine kinase (dCK) and cytosolic 5'-nucleotidase II (cN-II) are associated with the production of ara-CTP. Because ara-C's cytotoxicity depends on ara-CTP production, parameters that are most related to ara-CTP formation would predict ara-C sensitivity and the clinical outcome of ara-C therapy. The present study focused on finding any correlation between the capacity to produce ara-CTP and ara-C-metabolizing factors. In vitro ara-CTP production, mRNA levels of hENT1, dCK, and cN-II, and ara-C sensitivity were evaluated in 34 blast samples from 33 leukemic patients including 26 with AML. A large degree of heterogeneity was seen in the capacity to produce ara-CTP and in mRNA levels of hENT1, dCK, and cN-II. Despite the lack of any association between each of the transcript levels and ara-CTP production, the ratio of dCK/cN-II transcript levels correlated significantly with the amount of ara-CTP among AML samples. The HL-60 cultured leukemia cell line and its three ara-C-resistant variants (HL-60/R1, HL-60/R2, HL-60/R3), which were 8-, 10-, and 500-fold more resistant than HL-60, respectively, were evaluated similarly. The dCK/cN-II ratio was again proportional to ara-CTP production and to ara-C sensitivity. The dCK/cN-II ratio may thus predict the capacity for ara-CTP production and ultimately, ara-C sensitivity in AML.


Subject(s)
5'-Nucleotidase/genetics , Arabinofuranosylcytosine Triphosphate/metabolism , Deoxycytidine Kinase/genetics , Equilibrative Nucleoside Transporter 1/genetics , Leukemia, Myeloid, Acute/metabolism , Predictive Value of Tests , Cytarabine/metabolism , Cytarabine/therapeutic use , Drug Resistance, Neoplasm , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/drug therapy , RNA, Messenger/analysis
10.
Biochem Pharmacol ; 73(10): 1548-57, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17324380

ABSTRACT

Gemcitabine and ara-C have multiple mechanisms of action: DNA incorporation and for gemcitabine also ribonucleotide reductase (RNR) inhibition. Since dCTP competes with their incorporation into DNA, dCTP depletion can potentiate their cytotoxicity. We investigated whether additional RNR inhibition by Triapine (3-AP), a new potent RNR inhibitor, enhanced cytotoxicity of gemcitabine and ara-C in four non-small-cell-lung-cancer (NSCLC) cell lines, using the multiple-drug-effect analysis. Simultaneous and sequential exposure (preexposure to 3-AP for 24h) in a constant molar ratio of 3-AP and gemcitabine was antagonistic/additive in all cell lines. Preexposure to 3-AP at an IC(25) concentration for 24h before variable concentrations of gemcitabine was synergistic. RNR inhibition by 3-AP resulted in a more synergistic interaction in combination with ara-C, which does not inhibit RNR. Two cell lines with pronounced synergism (SW1573) or antagonism (H460) for gemcitabine/3-AP, were evaluated for accumulation of the active metabolites, dFdCTP and ara-CTP. Simultaneous exposure induced no or a small increase, but ara-CTP increased after pretreatment with 3-AP, 4-fold in SW1573 cells, but not in H460 (<1.5 fold). Ara-C and gemcitabine incorporation into DNA were more pronounced (about 2-fold increased) for sequential treatment in SW1573 compared to H460 cells (<1.5 fold). This was not related to the activity and expression of deoxycytidine kinase and the M2 subunit of RNR. In conclusion, RNR inhibition by 3-AP prior to gemcitabine or ara-C exposure stimulates accumulation of the active metabolites and incorporation into DNA. The combination 3-AP/Ara-C is more synergistic than 3-AP/gemcitabine possibly because gemcitabine already inhibits RNR, but ara-C does not.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Arabinofuranosylcytosine Triphosphate/metabolism , Cytarabine/pharmacology , DNA/drug effects , Deoxycytidine/analogs & derivatives , Pyridines/pharmacology , Ribonucleotide Reductases/metabolism , Thiosemicarbazones/pharmacology , Animals , DNA/metabolism , Deoxycytidine/metabolism , Deoxycytidine/pharmacology , Drug Synergism , Humans , Pyridines/metabolism , Ribonucleotide Reductases/drug effects , Thiosemicarbazones/metabolism , Tumor Cells, Cultured , Gemcitabine
11.
Int J Hematol ; 85(2): 108-15, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17321987

ABSTRACT

The combination of cytarabine (ara-C) with fludarabine is a common approach to treating resistant acute myeloid leukemia. Success depends on a fludarabine triphosphate (F-ara-ATP)-mediated increase in the active intracellular metabolite of ara-C, ara-C 5'-triphosphate (ara-CTP). Therapy-resistant leukemia may exhibit ara-C resistance, the mechanisms of which might induce cross-resistance to fludarabine with reduced F-ara-ATP formation. The present study evaluated the effect of combining ara-C and fludarabine on ara-C-resistant leukemic cells in vitro. Two variant cell lines (R1 and R2) were 8-fold and 10-fold more ara-C resistant, respectively, than the parental HL-60 cells. Reduced deoxycytidine kinase activity was demonstrated in R1 and R2 cells, and R2 cells also showed an increase in cytosolic 5'-nucleotidase II activity. Compared with HL-60 cells, R1 and R2 cells produced smaller amounts of ara-CTP. Both variants accumulated less F-ara-ATP than HL-60 cells and showed cross-resistance to fludarabine nucleoside (F-ara-A). R2 cells, however, accumulated much smaller amounts of F-ara-ATP and were more F-ara-A resistant than R1 cells. In HL-60 and R1 cells, F-ara-A pretreatment followed by ara-C incubation produced F-ara-ATP concentrations sufficient for augmenting ara-CTP production, thereby enhancing ara-C cytotoxicity. No potentiation was observed in R2 cells. Nucleotidase might preferentially degrade F-ara-A monophosphate over ara-C monophosphate, leading to reduced F-ara-ATP production and thereby compromising the F-ara-A-mediated potentiation of ara-C cytotoxicity in R2 cells. Thus, F-ara-A-mediated enhancement of ara-C cytotoxicity depended on F-ara-ATP accumulation in ara-C-resistant leukemic cells but ultimately was associated with the mechanism of ara-C resistance.


Subject(s)
Arabinofuranosylcytosine Triphosphate/pharmacology , Cytarabine/pharmacology , Drug Resistance, Neoplasm/drug effects , Leukemia, Myeloid, Acute/drug therapy , Vidarabine/analogs & derivatives , Arabinofuranosylcytosine Triphosphate/agonists , Arabinofuranosylcytosine Triphosphate/metabolism , Drug Synergism , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/metabolism , Vidarabine/agonists , Vidarabine/pharmacology
12.
Cancer Chemother Pharmacol ; 60(4): 467-77, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17171362

ABSTRACT

PURPOSE: The prodrug cytosinearabinoside (ara-C) is widely used in the treatment of acute leukemias. The active drug is the intracellular metabolite cytosine-arabinoside-5'-triphosphate (ara-CTP). The purpose of the present study was to investigate the relation between sensitivity and pharmacokinetic parameters Cmax, t1/2 and AUC of ara-CTP. The obtained results were compared to previous studies. EXPERIMENTAL DESIGN: Cmax, t1/2 and AUC of ara-CTP were assessed in leukemic cells of 17 pediatric patients with acute lymphoblastic leukemia (ALL) and in 6 lymphoblastic cell lines and compared with normal lymphocytes of 9 healthy donors by high pressure liquid chromatography (HPLC). The sensitivity of the cells against ara-C was determined by the MTT assay. RESULTS: The intracellular accumulation of ara-CTP was significantly lower in normal lymphocytes (Cmax 47.7-60.9 pmol/10(6) cells) compared to leukemic cell lines (Cmax 11-1128 pmol/10(6) cells) and leukemic cells of our patients (Cmax 85.9-631 pmol/10(6) cells). Similar results were found for the AUC. There was no significant difference between initial and relapsed leukemias in our small cohort. A correlation between sensitivity in terms of IC50 values and the intracellular ara-CTP accumulation was observed in cell lines, but not in leukemic cells and normal lymphocytes from healthy donors. CONCLUSIONS: Pharmacokinetic parameters varied tremendously in leukemic cells in contrast to normal lymphocytes without a difference in sensitivity. It is worthwhile to compare literature data to assess an optimal dosage of ara-C in pediatric patients.


Subject(s)
Arabinofuranosylcytosine Triphosphate/pharmacology , Cytarabine/pharmacokinetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Adolescent , Arabinofuranosylcytosine Triphosphate/metabolism , Cell Line , Cell Line, Tumor , Child , Child, Preschool , Chromatography, High Pressure Liquid , Cytarabine/pharmacology , Half-Life , Humans , Infant , Inhibitory Concentration 50 , Lymphocytes/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Recurrence
13.
Mol Pharmacol ; 70(5): 1832-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16936229

ABSTRACT

Methotrexate (MTX) is the anticancer and antirheumatoid drug that is believed to block nucleotide synthesis and cell cycle by inhibiting dihydrofolate reductase activity. We have developed novel affinity matrices, termed SG beads, that are easy to manipulate and are compatible with surface functionalization. Using the matrices, here we present evidence that deoxycytidine kinase (dCK), an enzyme that acts in the salvage pathway of nucleotide biosynthesis, is another target of MTX. MTX modulates dCK activity differentially depending on substrate concentrations. 1-beta-D-Arabinofuranosylcytosine (ara-C), a chemotherapy agent often used in combination with MTX, is a nucleoside analog whose incorporation into chromosome requires prior phosphorylation by dCK. We show that, remarkably, MTX enhances incorporation and cytotoxicity of ara-C through regulation of dCK activity in Burkitt's lymphoma cells. Thus, this study provides new insight into the mechanisms underlying MTX actions and demonstrates the usefulness of the SG beads.


Subject(s)
Chromatography, Affinity/methods , Drug Screening Assays, Antitumor/instrumentation , Drug Screening Assays, Antitumor/methods , Methotrexate/pharmacology , Microspheres , Amino Acid Sequence , Arabinofuranosylcytosine Triphosphate/metabolism , Cell Death/drug effects , Chromosomes, Human/genetics , Deoxycytidine Kinase/chemistry , Deoxycytidine Kinase/isolation & purification , Humans , Jurkat Cells , Molecular Sequence Data , Molecular Weight , Tetrahydrofolate Dehydrogenase/metabolism
15.
Haematologica ; 91(7): 912-9, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16818278

ABSTRACT

BACKGROUND AND OBJECTIVES: Cytarabine (Ara-C) is commonly used for the treatment of acute leukemia. Incorporation of Ara-C into DNA is a key event in the mechanism of killing of proliferating leukemic cells. Previously, we demonstrated that Ara-C was cytotoxic to proliferating but not to resting (G(O)) malignant cells from patients with acute leukemia. In contrast, here we show unexpected apoptosis of G(O) B-chronic lymphocytic leukemia (CLL) cells by Ara-C in a dose-dependent manner. In this study we analyzed which cellular processes were involved in Ara-C-mediated killing of G(O)-B-CLL cells. DESIGN AND METHODS: Using primary B-CLL cells (>98% in G(O)), we examined the mechanisms of Ara-C-mediated apoptosis in resting G(O) cells. CFSE-based cytotoxicity assays combined with cell cycle analysis were used to perform a long-term analysis of Ara-C-mediated killing of B-CLL cells. The effects of Ara-C on DNA and RNA synthesis were studied using various 3H-incorporation experiments. RESULTS: Ara-C-mediated cell death of B-CLL cells showed the characteristics of normal apoptosis, such as phosphatidyl serine exposure and caspase activation. The mechanism of killing of quiescent B-CLL cells by Ara-C was shown not to be dependent on DNA replication. In contrast, CD40L-activated B-CLL cells showed S-phase-specific depletion of proliferating CLL cells. We demonstrated that Ara-C was converted into its active triphosphate by G(O)-B-CLL cells, coinciding with a 30% inhibition of RNA synthesis. INTERPRETATION AND CONCLUSIONS: In conclusion, our data indicate that Ara-C can induce apoptosis in resting G(O)-B-CLL cells using a mechanism independent of cell proliferation and DNA replication but associated with inhibition of RNA synthesis and downregulation of Mcl-1.


Subject(s)
Apoptosis/drug effects , Cytarabine/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Arabinofuranosylcytosine Triphosphate/metabolism , Cell Proliferation/drug effects , Cytarabine/metabolism , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Male , RNA/antagonists & inhibitors , RNA/biosynthesis
16.
J Natl Cancer Inst ; 97(3): 226-31, 2005 Feb 02.
Article in English | MEDLINE | ID: mdl-15687366

ABSTRACT

Down syndrome children with acute megakaryocytic leukemia (AMkL) have higher cure rates than non-Down syndrome acute myeloid leukemia (AML) patients treated with cytosine arabinoside (ara-C). Megakaryoblasts from Down syndrome AML patients are more sensitive in vitro to ara-C than cells from non-Down syndrome AML patients. Somatic mutations in the GATA1 transcription factor have been detected exclusively and almost uniformly in Down syndrome AMkL patients, suggesting a potential linkage to the chemotherapy sensitivity of Down syndrome megakaryoblasts. Stable transfection of wild-type GATA1 cDNA into the Down syndrome AMkL cell line CMK resulted in decreased (8- to 17-fold) ara-C sensitivity and a threefold-lower generation of the active ara-C metabolite ara-CTP compared with that for mock-transfected CMK cells. High intracellular levels of uridine arabinoside (ara-U) (an inactive ara-C catabolite generated by cytidine deaminase) and cytidine deaminase transcripts were detected in GATA1-transfected CMK sublines, whereas no ara-U was detected in mock-transfected cells. Cytidine deaminase transcripts were a median 5.1-fold (P = .002) lower in Down syndrome megakaryoblasts (n = 16) than in blast cells from non-Down syndrome patients (n = 56). These results suggest that GATA1 transcriptionally upregulates cytidine deaminase and that the presence or absence of GATA1 mutations in AML blasts likely confers differences in ara-C sensitivities due to effects on cytidine deaminase gene expression, which, in turn, contributes to the high cure rate of Down syndrome AMkL patients.


Subject(s)
Cytidine Deaminase/metabolism , DNA-Binding Proteins/metabolism , Down Syndrome/complications , Down Syndrome/metabolism , Leukemia, Megakaryoblastic, Acute/metabolism , Transcription Factors/metabolism , Antimetabolites, Antineoplastic/metabolism , Arabinofuranosylcytosine Triphosphate/metabolism , Arabinofuranosyluracil/metabolism , Blotting, Western , Child , Cytarabine/metabolism , Cytidine Deaminase/genetics , DNA-Binding Proteins/genetics , Down Syndrome/genetics , Erythroid-Specific DNA-Binding Factors , GATA1 Transcription Factor , Humans , Leukemia, Megakaryoblastic, Acute/complications , Leukemia, Megakaryoblastic, Acute/enzymology , Leukemia, Megakaryoblastic, Acute/genetics , Polymerase Chain Reaction , Time Factors , Transcription Factors/genetics , Transcription, Genetic , Transcriptional Activation , Up-Regulation
17.
Blood ; 103(8): 2908-13, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15070662

ABSTRACT

Fludarabine in addition to cytosine-arabinoside (ARA-C) increases the accumulation of ARA-C-5'-triphosphate (ARA-CTP), which is responsible for the cytotoxic effect in leukemic blasts. In a randomized phase 3 trial, patients with high-risk myelodysplastic syndrome (MDS) (n = 91) or elderly patients with acute myeloid leukemia (AML) (n = 43) were randomized to receive 2 induction courses consisting of ARA-C (2 g/m2 days 1 through 5) and granulocyte colony-stimulating factor (G-CSF) (filgrastim, 5 microg/kg) during and after chemotherapy with or without fludarabine (25 mg/m2, days 1 through 5) (FLAG versus AG). Consolidation consisted of daunorubicin (45 mg/m2, days 1 through 3) and ARA-C (200 mg/m2, days 1 through 7). Complete remission (CR) rate following AG was 65% versus 71% with FLAG (P =.49). Overall survival (OS) at 24 months was 24% for AG treatment and 39% for FLAG (P =.32). Event-free survival (EFS) at 2 years was 10% and 19% (P =.31) for the AG and FLAG treatments, respectively. Platelet and granulocyte recovery times after the second cycle were prolonged in the FLAG treatment group. Grades 3 to 4 neurotoxicities were more often reported in the FLAG arm (14% versus 3%, P =.03), whereas no significant differences in other toxicities were observed. In a cohort of patients, the in vivo accumulation of ARA-CTP in leukemic cells was determined. Although ARA-CTP accumulation in leukemic cells after FLAG was enhanced, clinical outcome in terms of CR rate, OS, EFS, and disease-free survival (DFS) was not significantly improved by combining fludarabine with ARA-C.


Subject(s)
Cytarabine/administration & dosage , Granulocyte Colony-Stimulating Factor/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Vidarabine/analogs & derivatives , Vidarabine/administration & dosage , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Arabinofuranosylcytosine Triphosphate/metabolism , Cytarabine/adverse effects , Cytarabine/pharmacokinetics , Disease-Free Survival , Female , Filgrastim , Granulocyte Colony-Stimulating Factor/adverse effects , Hematopoiesis/drug effects , Humans , In Vitro Techniques , Leukemia, Myeloid, Acute/metabolism , Male , Middle Aged , Myelodysplastic Syndromes/metabolism , Prognosis , Recombinant Proteins , Risk Factors , Survival Rate , Vidarabine/adverse effects
18.
Life Sci ; 74(9): 1071-80, 2004 Jan 16.
Article in English | MEDLINE | ID: mdl-14687648

ABSTRACT

Amidox, a new polyhydroxy-substituted benzoic acid derivative, is a potent inhibitor of the enzyme ribonucleotide reductase (RR), which catalyses the de novo synthesis of DNA. RR is considered to be an excellent target for anti cancer chemotherapy. We investigated the biochemical and antineoplastic effects of amidox as a single agent and in combination with Ara-C in human HL-60 promyelocytic leukemia cells. Amidox inhibited the growth of HL-60 cells in a growth inhibition assay with an IC50 of 25 microM. In a soft agar colony forming assay, amidox yielded a 50% inhibition of colony formation at 13 microM. We also investigated the effects of amidox treatment on the formation of deoxynucleosidetriphosphates. Amidox (50 and 75 microM for 24 hours) could significantly decrease intracellular concentrations of dCTP, dATP and dGTP pools, whereas dTTP levels increased. We then tested the combination effects of amidox with Ara-C; this combination yielded additive cytotoxic effects both in growth inhibition and in soft agar colony formation assays. This effect was due to the increased formation of Ara-CTP, the active metabolite of Ara-C, after preincubation with amidox. Preincubation of HL-60 cells with 75 and 100 microM amidox for 24 hours caused an increase in the intracellular Ara-CTP concentrations by 576% and 1143%, respectively. Therefore amidox might offer an additional option for the treatment of leukemia and thus be further investigated in in vivo studies as a single agent and in combination with Ara-C.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Cytarabine/pharmacology , Oximes/pharmacology , Ribonucleotide Reductases/antagonists & inhibitors , Arabinofuranosylcytosine Triphosphate/metabolism , Cell Division/drug effects , Colony-Forming Units Assay , Deoxyribonucleotides/metabolism , Growth Inhibitors/metabolism , HL-60 Cells , Humans
19.
Biochem Pharmacol ; 65(12): 1997-2008, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12787880

ABSTRACT

The effects of enforced expression of p53 on the sensitivity of p53(-/-) human monocytic leukemia cells (U937) to apoptosis following exposure to the S-phase-specific antimetabolite 1-[beta-D-arabinofuranosyl]cytosine (ara-C) were examined. Cells were stably transfected with a plasmid containing a chimeric DNA construct encoding a temperature-sensitive p53 variant (135(ala-->val)), which transactivates at 32 degrees but is non-functional at 37 degrees. A significant reduction in the S-phase population was observed in ptsp53 mutants incubated at 32 degrees. Nevertheless, while vector controls did not exhibit differential sensitivity to ara-C at 32 degrees versus 37 degrees, temperature-sensitive p53 mutants displayed a significant increase in apoptosis at the permissive temperature. This was not accompanied by increased ara-CTP formation, DNA incorporation of [3H]ara-C, or altered expression of Bcl-2 or Bax. Enhanced sensitivity was associated with increased mitochondrial injury (e.g. cytochrome c release), caspase activation, and loss of clonogenic survival. Significantly, ptsp53 cells synchronized in S phase were markedly more sensitive to ara-C-mediated mitochondrial injury and apoptosis at 32 degrees, indicating that wild-type p53 specifically enhances the susceptibility of this subpopulation to ara-C lethality. Consistent with these results, transient transfection of human wild-type p53 cDNA rendered parental U937 cells more sensitive to ara-C-mediated cell death. Collectively, these findings indicate that p53 expression renders S-phase U937 cells more susceptible to ara-C-mediated mitochondrial dysfunction, cytochrome c release, apoptosis, and loss of clonogenic survival without enhancing ara-C metabolism. Such findings raise the possibility that loss of functional p53 activity allows leukemia cells to circumvent ara-C lethality.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Cytarabine/pharmacology , Genes, Tumor Suppressor/physiology , Genes, p53/physiology , Antimetabolites, Antineoplastic/metabolism , Arabinofuranosylcytosine Triphosphate/metabolism , Cytarabine/metabolism , Humans , Leukemia/pathology , U937 Cells
20.
Blood ; 101(4): 1270-6, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12406912

ABSTRACT

Infant acute lymphoblastic leukemia (ALL) is characterized by a high incidence of mixed lineage leukemia (MLL) gene rearrangements, a poor outcome, and resistance to chemotherapeutic drugs. One exception is cytosine arabinoside (Ara-C), to which infant ALL cells are highly sensitive. To investigate the mechanism underlying Ara-C sensitivity in infants with ALL, mRNA levels of Ara-C-metabolizing enzymes were measured in infants (n = 18) and older children (noninfants) with ALL (n = 24). In the present study, infant ALL cells were 3.3-fold more sensitive to Ara-C (P =.007) and accumulated 2.3-fold more Ara-CTP (P =.011) upon exposure to Ara-C, compared with older children with ALL. Real-time quantitative reverse trancriptase-polymerase chain reaction (RT-PCR) (TaqMan) revealed that infants express 2-fold less of the Ara-C phosphorylating enzyme deoxycytidine kinase (dCK) mRNA (P =.026) but 2.5-fold more mRNA of the equilibrative nucleoside transporter 1 (hENT1), responsible for Ara-C membrane transport (P =.001). The mRNA expression of pyrimidine nucleotidase I (PN-I), cytidine deaminase (CDA), and deoxycytidylate deaminase (dCMPD) did not differ significantly between both groups. hENT1 mRNA expression inversely correlated with in vitro resistance to Ara-C (r(s) = -0.58, P =.006). The same differences concerning dCK and hENT1 mRNA expression were observed between MLL gene-rearranged (n = 14) and germ line MLL cases (n = 25). An oligonucleotide microarray screen (Affymetrix) comparing patients with MLL gene-rearranged ALL with those with nonrearranged ALL also showed a 1.9-fold lower dCK (P =.001) and a 2.7-fold higher hENT1 (P =.046) mRNA expression in patients with MLL gene-rearranged ALL. We conclude that an elevated expression of hENT1, which transports Ara-C across the cell membrane, contributes to Ara-C sensitivity in MLL gene-rearranged infant ALL.


Subject(s)
Cytarabine/metabolism , Cytarabine/therapeutic use , DNA-Binding Proteins/genetics , Gene Expression , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Proto-Oncogenes , Transcription Factors , Arabinofuranosylcytosine Triphosphate/metabolism , Cell Survival , Child , Child, Preschool , DCMP Deaminase/genetics , Deoxycytidine Kinase/genetics , Drug Resistance, Neoplasm/genetics , Equilibrative Nucleoside Transporter 1/genetics , Female , Gene Rearrangement , Histone-Lysine N-Methyltransferase , Humans , Infant , Male , Myeloid-Lymphoid Leukemia Protein , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Pyrimidine Nucleotides/genetics , RNA, Messenger/analysis , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...