Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Anticancer Res ; 44(3): 981-991, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38423659

ABSTRACT

BACKGROUND/AIM: Methionine metabolism contributes to supplying sulfur-containing amino acids, controlling the methyl group transfer reaction, and producing polyamines in cancer cells. Polyamines play important roles in various cellular functions. Methylthioadenosine phosphorylase (MTAP), the key enzyme of the methionine salvage pathway, is reported to be deficient in 15-62% of cases of hematological malignancies. MTAP-deficient cancer cells accumulate polyamines, resulting in enhanced cell proliferation. The aim of this study was to investigate the combined effects of the polyamine synthesis inhibitor SAM486A and the anticancer antimetabolite cytarabine in MTAP-deficient leukemic cells in vitro. MATERIALS AND METHODS: The leukemia cell line U937 and the subline, U937/MTAP(-), in which MTAP was knocked down by shRNA, were used. The experiments were performed in media supplemented with 20% methionine (low methionine), which was the minimum concentration for maintaining cellular viability. RESULTS: The knockdown efficiency test confirmed a 70% suppression of the expression of the MTAP gene in U937/MTAP(-) cells. Even in the media with low methionine, the intracellular methionine concentration was not reduced in U937/MTAP(-) cells, suggesting that the minimum supply of methionine was sufficient to maintain intracellular levels of methionine. Both U937/MTAP(+) and U937/MTAP(-) cells were comparably sensitive to anticancer drugs (cytarabine, methotrexate, clofarabine and 6-thioguanine). The combination of SAM486A and cytarabine was demonstrated to have synergistic cytotoxicity in U937/MTAP(-) cells with regard to cell growth inhibition and apoptosis induction, but not in U937/MTAP(+) cells. Mechanistically, SAM486A altered the intracellular polyamine concentrations and reduced the antiapoptotic proteins. CONCLUSION: Methionine metabolism and polyamine synthesis can be attractive therapeutic targets in leukemia.


Subject(s)
Amidines , Antineoplastic Agents , Indans , Leukemia , Humans , Cytarabine/pharmacology , Purine-Nucleoside Phosphorylase/genetics , Purine-Nucleoside Phosphorylase/metabolism , Polyamines , Methionine/pharmacology , Methionine/metabolism , Leukemia/drug therapy
2.
BMC Cancer ; 21(1): 879, 2021 Jul 31.
Article in English | MEDLINE | ID: mdl-34332580

ABSTRACT

BACKGROUND: Pralatrexate (PDX) is a novel antifolate approved for the treatment of patients with relapsed/refractory peripheral T-cell lymphoma, but some patients exhibit intrinsic resistance or develop acquired resistance. Here, we evaluated the mechanisms underlying acquired resistance to PDX and explored potential therapeutic strategies to overcome PDX resistance. METHODS: To investigate PDX resistance, we established two PDX-resistant T-lymphoblastic leukemia cell lines (CEM and MOLT4) through continuous exposure to increasing doses of PDX. The resistance mechanisms were evaluated by measuring PDX uptake, apoptosis induction and folate metabolism-related protein expression. We also applied gene expression analysis and methylation profiling to identify the mechanisms of resistance. We then explored rational drug combinations using a spheroid (3D)-culture assay. RESULTS: Compared with their parental cells, PDX-resistant cells exhibited a 30-fold increase in half-maximal inhibitory concentration values. Induction of apoptosis by PDX was significantly decreased in both PDX-resistant cell lines. Intracellular uptake of [14C]-PDX decreased in PDX-resistant CEM cells but not in PDX-resistant MOLT4 cells. There was no significant change in expression of dihydrofolate reductase (DHFR) or folylpolyglutamate synthetase (FPGS). Gene expression array analysis revealed that DNA-methyltransferase 3ß (DNMT3B) expression was significantly elevated in both cell lines. Gene set enrichment analysis revealed that adipogenesis and mTORC1 signaling pathways were commonly upregulated in both resistant cell lines. Moreover, CpG island hypermethylation was observed in both PDX resistant cells lines. In the 3D-culture assay, decitabine (DAC) plus PDX showed synergistic effects in PDX-resistant cell lines compared with parental lines. CONCLUSIONS: The resistance mechanisms of PDX were associated with reduced cellular uptake of PDX and/or overexpression of DNMT3B. Epigenetic alterations were also considered to play a role in the resistance mechanism. The combination of DAC and PDX exhibited synergistic activity, and thus, this approach might improve the clinical efficacy of PDX.


Subject(s)
Aminopterin/analogs & derivatives , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm , Folic Acid Antagonists/pharmacology , Aminopterin/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers , DNA Methylation , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Humans , Tumor Cells, Cultured
3.
BMC Cancer ; 20(1): 984, 2020 Oct 12.
Article in English | MEDLINE | ID: mdl-33046037

ABSTRACT

BACKGROUND: Cytarabine (ara-C) is the major drug for the treatment of acute myeloid leukemia (AML), but cellular resistance to ara-C is a major obstacle to therapeutic success. The present study examined enhanced anti-apoptosis identified in 3 newly established nucleoside analogue-resistant leukemic cell line variants and approaches to overcoming this resistance. METHODS: HL-60 human AML cells were used to develop the ara-C- or clofarabine (CAFdA)-resistant variants. The Bcl-2 inhibitor venetoclax and the Mcl-1 inhibitor alvocidib were tested to determine whether they could reverse these cells' resistance. RESULTS: A 10-fold ara-C-resistant HL-60 variant, a 4-fold CAFdA-resistant HL-60 variant, and a 30-fold CAFdA-resistant HL-60 variant were newly established. The variants demonstrated reduced deoxycytidine kinase and deoxyguanosine kinase expression, but intact expression of surface transporters (hENT1, hENT2, hCNT3). The variants exhibited lower expression of intracellular nucleoside analogue triphosphates compared with non-variant HL-60 cells. The variants also overexpressed Bcl-2 and Mcl-1. Venetoclax as a single agent was not cytotoxic to the resistant variants. Nevertheless, venetoclax with nucleoside analogs demonstrated synergistic cytotoxicity against the variants. Alvocidib as a single agent was cytotoxic to the cells. However, alvocidib induced G1 arrest and suppressed the cytotoxicity of the co-administered nucleoside analogs. CONCLUSIONS: Three new nucleoside analogue-resistant HL-60 cell variants exhibited reduced production of intracellular analogue triphosphates and enhanced Bcl-2 and Mcl-1 expressions. Venetoclax combined with nucleoside analogs showed synergistic anti-leukemic effects and overcame the drug resistance.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Clofarabine/therapeutic use , Cytarabine/therapeutic use , Flavonoids/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Piperidines/therapeutic use , Sulfonamides/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Proliferation , Clofarabine/pharmacology , Cytarabine/pharmacology , Flavonoids/pharmacology , Humans , Piperidines/pharmacology , Sulfonamides/pharmacology
5.
Cancer Sci ; 107(7): 1029-38, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27166836

ABSTRACT

The major mechanism of imatinib (IM) resistance of CML is the reactivation of ABL kinase either through BCR-ABL gene amplification or mutation. We investigated the cytotoxicity of a pan-ABL tyrosine kinase inhibitor, ponatinib, and a pan-histone deacetylase inhibitor, panobinostat, against IM-resistant CML cells in vitro. Two different IM-resistant cell lines, K562/IM-R1 and Ba/F3/T315I were evaluated in comparison with their respective, parental cell lines, K562 and Ba/F3. K562/IM-R1 overexpressed BCR-ABL due to gene amplification. Ba/F3/T315I was transfected with a BCR-ABL gene encoding T315I-mutated BCR-ABL. Ponatinib inhibited the growth of both K562/IM-R1 and Ba/F3/T315I as potently as it inhibited their parental cells with an IC50 of 2-30 nM. Panobinostat also similarly inhibited the growth of all of the cell lines with an IC50 of 40-51 nM. This was accompanied by reduced histone deacetylase activity, induced histone H3 acetylation, and an increased protein level of heat shock protein 70, which suggested disruption of heat shock protein 90 chaperone function for BCR-ABL and its degradation. Importantly, the combination of ponatinib with panobinostat showed synergistic growth inhibition and induced a higher level of apoptosis than the sum of the apoptosis induced by each agent alone in all of the cell lines. Ponatinib inhibited phosphorylation not only of BCR-ABL but also of downstream signal transducer and activator of transcription 5, protein kinase B, and ERK1/2 in both K562/IM-R1 and Ba/F3/T315I, and the addition of panobinostat to ponatinib further inhibited these phosphorylations. In conclusion, panobinostat enhanced the cytotoxicity of ponatinib towards IM-resistant CML cells including those with T315I-mutated BCR-ABL.


Subject(s)
Drug Resistance, Neoplasm , Hydroxamic Acids/pharmacology , Imatinib Mesylate/pharmacology , Imidazoles/pharmacology , Indoles/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Pyridazines/pharmacology , Acetylation/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , HSP70 Heat-Shock Proteins/metabolism , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Panobinostat , Phosphorylation/drug effects , Signal Transduction/drug effects
6.
Anticancer Res ; 34(10): 5487-94, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25275045

ABSTRACT

BACKGROUND/AIM: Gemtuzumab ozogamicin (GO) consists of the cluster of differentiation 33 (CD33) antibody linked to calicheamicin. The binding of GO to the CD33 antigen on leukemic cells results in internalization and subsequent release of calicheamicin, thereby inducing DNA strand breaks. We hypothesized that the poly (ADP-ribose) polymerase inhibitor olaparib might inhibit DNA repair initiated by GO-induced DNA strand breaks, thereby increasing cytotoxicity. MATERIALS AND METHODS: The human myeloid leukemia cell line HL-60 and a GO-resistant variant (HL/GO20) were used. RESULTS: The 50% growth-inhibitory concentrations (IC50) were 24 ng/ml for HL-60 cells and 550 ng/ml for GO-resistant variant HL/GO20 cells. HL/GO20 cells were also refractory to GO-induced apoptosis. CD33 positivity was reduced in HL/GO20 cells. Olaparib-alone did not inhibit the cell growth and did not induce apoptosis in either HL-60 cells or HL/GO20 cells at concentrations of up to 10 µM. When cells were treated with different concentrations of GO in the presence of 10 µM olaparib, the IC50 of GO for HL-60 cells was 13 ng/ml. The combination index was 0.86, indicating synergistic cytotoxicity of GO and olaparib in combination. Such a combination was ineffective for HL/GO20 cells. CONCLUSION: GO and olaparib exerted synergistic cytotoxicity in CD33-positive myeloid leukemia cells in vitro.


Subject(s)
Aminoglycosides/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Leukemia, Myeloid/metabolism , Phthalazines/pharmacology , Piperazines/pharmacology , Sialic Acid Binding Ig-like Lectin 3/metabolism , Aminoglycosides/toxicity , Antibodies, Monoclonal, Humanized/toxicity , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Drug Synergism , Gemtuzumab , HL-60 Cells , Humans , Phthalazines/toxicity , Piperazines/toxicity , Poly(ADP-ribose) Polymerase Inhibitors
7.
Anticancer Res ; 34(9): 4885-92, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25202070

ABSTRACT

BACKGROUND/AIM: Forodesine inhibits purine nucleoside phosphorylase, resulting in an accumulation of intracellular dGTP and consequently cell death. 9-ß-D-Arabinofuranosylguanine (ara-G) is an active compound of nelarabine that is intracellularly phosphorylated to a triphosphate form, which inhibits DNA synthesis. Both agents show cytotoxicity toward T-cell malignancies. In the present study, we investigated the cytotoxicity of forodesine in vitro using ara-G-resistant leukemia cells. MATERIALS AND METHODS: T-Lymphoblastic leukemia cell line CCRF-CEM and ara-G-resistant CEM variant cell line CEM/ara-G that we had previously established were used. RESULTS: A growth-inhibition assay demonstrated that CEM cells were insensitive to single-agent forodesine treatment. The cells were also insensitive to deoxyguanosine at a maximal concentration of 10 µM. CEM/ara-G cells were 80-fold more resistant to ara-G than were CEM cells, and the mode of sensitivity to forodesine and deoxyguanosine was similar to that of CEM cells. In the presence of 10 µM deoxyguanosine, forodesine effectively inhibited the growth of CEM cells but not that of CEM/ara-G cells. Flow cytometric analyses showed that combination of forodesine and deoxyguanosine induced apoptosis of CEM cells but not of CEM/ara-G cells. The addition of ara-G did not augment the cytotoxicity of the forodesine/deoxyguanosine combination towards CEM cells or CEM/ara-G cells. The combination index revealed antagonism between forodesine and ara-G. The intracellular production of ara-G triphosphate was reduced in the presence of forodesine. CONCLUSION: Nelarabine-resistant CEM/ara-G cells are insensitive to forodesine.


Subject(s)
Arabinonucleosides/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Purine Nucleosides/pharmacology , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Pyrimidinones/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Arabinonucleosides/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , HL-60 Cells , Humans , Lymphoma, B-Cell/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Purine Nucleosides/toxicity , Pyrimidinones/toxicity
8.
BMC Cancer ; 14: 547, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25070259

ABSTRACT

BACKGROUND: Nine-beta-D-arabinofuranosylguanine (ara-G), an active metabolite of nelarabine, enters leukemic cells through human Equilibrative Nucleoside Transporter 1, and is then phosphorylated to an intracellular active metabolite ara-G triphosphate (ara-GTP) by both cytosolic deoxycytidine kinase and mitochondrial deoxyguanosine kinase. Ara-GTP is subsequently incorporated into DNA, thereby inhibiting DNA synthesis. METHODS: In the present study, we developed a novel ara-G-resistant variant (CEM/ara-G) of human T-lymphoblastic leukemia cell line CCRF-CEM, and elucidated its mechanism of ara-G resistance. The cytotoxicity was measured by using the growth inhibition assay and the induction of apoptosis. Intracellular triphosphate concentrations were quantitated by using HPLC. DNA synthesis was evaluated by the incorporation of tritiated thymidine into DNA. Protein expression levels were determined by using Western blotting. RESULTS: CEM/ara-G cells were 70-fold more ara-G-resistant than were CEM cells. CEM/ara-G cells were also refractory to ara-G-mediated apoptosis. The transcript level of human Equilibrative Nucleoside Transporter 1 was lowered, and the protein levels of deoxycytidine kinase and deoxyguanosine kinase were decreased in CEM/ara-G cells. The subsequent production of intracellular ara-GTP (21.3 pmol/107 cells) was one-fourth that of CEM cells (83.9 pmol/107 cells) after incubation for 6 h with 10 µM ara-G. Upon ara-G treatment, ara-G incorporation into nuclear and mitochondrial DNA resulted in the inhibition of DNA synthesis of both fractions in CEM cells. However, DNA synthesis was not inhibited in CEM/ara-G cells due to reduced ara-G incorporation into DNA. Mitochondrial DNA-depleted CEM cells, which were generated by treating CEM cells with ethidium bromide, were as sensitive to ara-G as CEM cells. Anti-apoptotic Bcl-xL was increased and pro-apoptotic Bax and Bad were reduced in CEM/ara-G cells. CONCLUSIONS: An ara-G-resistant CEM variant was successfully established. The mechanisms of resistance included reduced drug incorporation into nuclear DNA and antiapoptosis.


Subject(s)
Antineoplastic Agents/metabolism , Arabinonucleosides/metabolism , DNA/metabolism , Drug Resistance, Neoplasm/drug effects , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Arabinonucleosides/pharmacology , Cell Line, Tumor , DNA Replication/drug effects , Humans , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology
9.
Anticancer Res ; 34(4): 1657-62, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24692694

ABSTRACT

BACKGROUND/AIM: Clofarabine is transported into leukemic cells via the equilibrative nucleoside transporters (hENT) 1 and 2 and the concentrative nucleoside transporter (hCNT) 3, then phosphorylated by deoxycytidine kinase (dCK) and deoxyguanosine kinase (dGK) to an active triphosphate metabolite. Cytarabine uses hENT1 and dCK for its activation. We hypothesized that cytarabine-resistant leukemia cells retain sensitivity to clofarabine. MATERIALS AND METHODS: Human myeloid leukemia HL-60 cells and cytarabine-resistant variant HL/ara-C20 cells were used in the present study. RESULTS: Despite 20-fold cytarabine resistance, the HL/ara-C20 cells exhibited only a 6-fold resistance to clofarabine compared to HL-60 cells. The intracellular concentration of the triphosphate metabolite of cytarabine was reduced to 1/10, and that of clofarabine was halved in the HL/ara-C20 cells. hENT1 and dCK were reduced, but hCNT3 and dGK were not altered in the HL/ara-C20 cells, which might contribute to their retained capability to produce intracellular triphosphate metabolite of clofarabine. CONCLUSION: Clofarabine was cytotoxic to leukemia cells that were resistant to cytarabine.


Subject(s)
Adenine Nucleotides/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Arabinonucleosides/pharmacology , Cytarabine/pharmacology , Drug Resistance, Neoplasm , Adenine Nucleotides/metabolism , Adenine Nucleotides/toxicity , Antimetabolites, Antineoplastic/metabolism , Antimetabolites, Antineoplastic/toxicity , Apoptosis/drug effects , Arabinonucleosides/metabolism , Arabinonucleosides/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Clofarabine , Cytarabine/toxicity , Dose-Response Relationship, Drug , Equilibrative Nucleoside Transporter 1/metabolism , HL-60 Cells , Humans , Intracellular Space/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Membrane Transport Proteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism
10.
Nephron Extra ; 3(1): 73-85, 2013.
Article in English | MEDLINE | ID: mdl-24052801

ABSTRACT

BACKGROUND: Hyperuricemia cases (HU) can be classified into four subgroups by combining the two main causes of hyperuricemia, i.e. urate underexcretion and overproduction. These subgroups are as follows: underexcretion-type cases (UE); overproduction-type cases (OP); combined-type cases, and normal-type cases. Since urinary urate excretion (Uua) and urate clearance differ significantly between UE and OP, urate transport in the nephrons and the intratubular urate contents might also differ. Such differences might help clarify the pathophysiology of urate underexcretion in subgroups of hyperuricemia, and thus reveal its underlying mechanisms. METHODS: Urate transport coefficients in each subtype of HU were determined employing the previously reported benzbromarone-loading urate clearance tests. The subtype cases of HU were plotted on a graph of urate transport coefficients versus Uua as coordinates. The characteristic features in the distribution of subtype cases on graphs were analyzed in relation to Uua. RESULTS: The mean (±standard error) tubular secretion rate (TSR) in the UE (48.7 ± 1.7 ml/min) was significantly lower and the postsecretory urate reabsorption rate (R2) in the UE (0.904 ± 0.004) was significantly higher than those in the normal controls (78.0 ± 2.1 ml/min and 0.877 ± 0.003) or the OP (61.1 ± 3.2 ml/min and 0.861 ± 0.009). Decrements of TSR and increments of R2 in the UE were largest in the subtypes of the HU, in terms of case numbers and the deviation rate of the group. Conversely, decrements of TSR and increments of R2 were smallest in the OP. A significant correlation was identified between TSR and Uua (r = 0.345, p < 0.0001), and a significant negative correlation was also found between R2 and Uua (r = -0.393, p < 0.0001). CONCLUSION: IN THE UE, HYPERURICEMIA IS INDUCED MAINLY BY URATE UNDEREXCRETION, WHICH RESULTS FROM THE COMBINATION OF TWO MAIN CAUSES IN URATE TRANSPORTERS OF THE NEPHRON: significantly lower TSR and significantly higher R2. Neither of these was observed in OP. Differences in urate transporters in subtypes of the HU might be important not only for understanding the pathophysiology and mechanisms of urate underexcretion and hyperuricemia, but also for providing a strategic therapy for hyperuricemia.

11.
Cancer Sci ; 104(4): 502-7, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23320492

ABSTRACT

Cytarabine (ara-C) is the key agent for treating acute myeloid leukemia. After being transported into leukemic cells, ara-C is phosphorylated, by several enzymes including deoxycytidine kinase (dCK), to ara-C triphosphate (ara-CTP), an active metabolite, and then incorporated into DNA, thereby inhibiting DNA synthesis. Therefore, the cytotoxicity of ara-C depends on the production of ara-CTP and the induction of apoptosis. Here, we established a new ara-C-resistant acute myeloid leukemia cell line (HL-60/ara-C60) with dual resistance characteristics of the anti-antimetabolic character of decreased ara-CTP production and an increase in the antiapoptotic factors Bcl-2 and Bcl-XL. We further attempted to overcome resistance by augmenting ara-CTP production and stimulating apoptosis. A relatively new nucleoside analog, 9-ß-d-arabinofuranosylguanine (ara-G), and the small molecule Bcl-2 antagonist YC137 were used for this purpose. HL-60/ara-C60 was 60-fold more ara-C-resistant than the parental HL-60 cells. HL-60/ara-C60 cells exhibited low dCK protein expression, which resulted in decreased ara-CTP production. HL-60/ara-C60 cells were also refractory to ara-C-induced apoptosis due to overexpression of Bcl-2 and Bcl-XL. Combination treatment of ara-C with ara-G augmented the dCK protein level, thereby increasing ara-CTP production and subsequent cytotoxicity. Moreover, the combination of ara-C with YC137 produced a greater amount of apoptosis than ara-C alone. Importantly, the three-drug combination of ara-C, ara-G and YC137 provided greater cytotoxicity than ara-C+ara-G or ara-C+YC137. These findings suggest possible combination strategies for overcoming ara-C resistance by augmenting ara-CTP production and reversing refractoriness against the induction of apoptosis in ara-C resistant leukemic cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Arabinonucleosides/pharmacology , Drug Resistance, Neoplasm/drug effects , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Thiazoles/pharmacology , Apoptosis , Arabinonucleosides/administration & dosage , Cytarabine/pharmacology , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/drug therapy
12.
Anticancer Res ; 31(9): 2863-7, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21868530

ABSTRACT

BACKGROUND/AIM: An active metabolite of the anti-leukemia agent clofarabine (Cl-F-ara-A) is an intracellular triphosphate form, Cl-F-ara-ATP. Monitoring this active form could provide crucial information for optimizing treatment regimens based on Cl-F-ara-A. A simple, isocratic HPLC method was developed. MATERIALS AND METHODS: Samples (500 µl) from leukemic cells were loaded onto an anion-exchange column and eluted with a phosphate-acetonitrile buffer (flow rate: 0.7 ml/min) at ambient temperature. The Cl-F-ara-ATP concentration was determined by measuring absorbance at 254 nm. RESULTS: The standard curve was linear, with minimal within-day and inter-day variability. Recovery was excellent; low and high quantitation limits were 10 pmol and 5,000 pmol, respectively. Cl-F-ara-ATP eluted independently of ATP, GTP, UTP, and CTP. Production of Cl-F-ara-ATP was successfully measured in cultured leukemia HL-60 cells treated in vitro with Cl-F-ara-A. CONCLUSION: This method is simple, sensitive and applicable for determination of the Cl-F-ara-ATP content of biological materials.


Subject(s)
Adenine Nucleotides/analysis , Antineoplastic Agents/analysis , Arabinonucleosides/analysis , Chromatography, High Pressure Liquid/methods , Leukemia/pathology , Clofarabine , HL-60 Cells , Humans , Reproducibility of Results
13.
Int J Oncol ; 38(4): 911-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21290089

ABSTRACT

Cytarabine (ara-C) is the key drug for treatment of acute myeloid leukemia. Since intracellular cytarabine triphosphate (ara-CTP) is an active metabolite of ara-C, factors that reduce the amount of ara-CTP are known to induce drug resistance. However, these factors do not fully explain the development of resistance to ara-C. The present study was conducted to search for new candidate ara-C resistance factors, including those that are unrelated to ara-CTP production. For this purpose, we newly established five ara-C-resistant leukemic clones from different blood cell lineage leukemic cell lines (HL-60, K562, CEM, THP1 and U937). The resistant subclones were 5-58-fold more ara-C-resistant than their parental counterparts. All of the ara-C-resistant subclones, except for ara-C-resistant CEM cells, displayed alteration of ara-CTP-related factors such as ara-C membrane transport capacity, deoxycytidine kinase activity or cytosolic nucleotidase II activity. To identify new candidate factors, we used two comprehensive approaches: DNA microarray and proteome analyses. The DNA microarray analysis revealed eight genes (C19orf2, HSPA8, LGALS1, POU4F3, PSAP, AKT1, MBC2 and CACNA2D3) that were altered in all five ara-C-resistant lines compared to parental cells. Both proteome and DNA microarray analyses further detected a reduced protein level of stathmin1 in the ara-C-resistant CEM subclone compared to its parental line. Thus, the present findings suggested the involvement of novel multiple mechanisms in mediating the ara-C resistance of leukemic cells. The role of some of these molecules in resistance is still unclear.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Cell Line, Tumor/drug effects , Cytarabine/pharmacology , Drug Resistance, Neoplasm , Gene Expression Profiling , Leukemia/pathology , Proteome/metabolism , 5'-Nucleotidase/metabolism , Cell Line, Tumor/metabolism , Cell Lineage , Cell Survival , Deoxycytidine Kinase/metabolism , Electrophoresis, Gel, Two-Dimensional , Enzyme Assays , Genes, Neoplasm , Humans , Oligonucleotide Array Sequence Analysis
14.
Nephron Extra ; 1(1): 55-68, 2011 Jan.
Article in English | MEDLINE | ID: mdl-22470379

ABSTRACT

BACKGROUND: A four-component system for urate transport in nephrons has been proposed and widely investigated by various investigators studying the mechanisms underlying urinary urate excretion. However, quantitative determinations of urate transport have not been clearly elucidated yet. METHODS: The equation C(ua) = {C(cr)(1 - R(1)) + TSR}(1 - R(2)) was designed to approximate mathematically urate transport in nephrons, where R(1) = urate reabsorption ratio; R(2) = urate postsecretory reabsorption ratio; TSR = tubular secretion rate; C(ua) = urate clearance, and C(cr) = creatinine clearance. To investigate relationships between the three unknown variables (R(1), R(2), and TSR), this equation was expressed as contour lines of one unknown on a graph of the other two unknowns. Points at regular intervals on each contour line for the equation were projected onto a coordinate axis and the high-density regions corresponding to high-density intervals of a coordinate were investigated for three graph types. For benzbromarone (BBR)-loading C(ua) tests, C(ua) was determined before and after oral administration of 100 mg of BBR and C(ua)BBR(∞) was calculated from the ratio of C(ua)BBR(100)/C(ua). RESULTS: Before BBR administration, points satisfying the equation on the contour line for R(1) = 0.99 were highly dense in the region R(2) = 0.87-0.92 on all three graphs, corresponding to a TSR of 40-60 ml/min in hyperuricemia cases (HU). After BBR administration, the dense region was shifted in the direction of reductions in both R(1) and R(2), but TSR was unchanged. Under the condition that R(1) = 1 and R(2) = 0, urate tubular secretion (UTS) was considered equivalent to calculated urinary urate excretion (U(ex)) in a model of intratubular urate flow with excess BBR; C(ua)BBR(∞) = TSR was deduced from the equation at R(1) = 1 and R(2) = 0. In addition, TSR of the point under the condition that R(1) = 1 and R(2) = 0 on the graph agreed with TSR for the dense region at excess BBR. TSR was thus considered approximately equivalent to C(ua)BBR(∞), which could be determined from a BBR-loading C(ua) test. Approximate values for urate glomerular filtration, urate reabsorption, UTS, urate postsecretory reabsorption (UR(2)), and U(ex) were calculated as 9,610; 9,510; 4,490; 4,150, and 440 µg/min for HU and 6,890; 6,820; 4,060; 3,610, and 520 µg/min for normal controls (NC), respectively. The most marked change in HU was the decrease in TSR (32.0%) compared to that in NC, but UTS did not decrease. Calculated intratubular urate contents were reduced more by higher UR(2) in HU than in NC. This enhanced difference resulted in a 15.4% decrease in U(ex) for HU. CONCLUSION: Increased UR(2) may represent the main cause of urate underexcretion in HU.

15.
Oncol Rep ; 23(2): 499-504, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20043113

ABSTRACT

The 9-beta-D-arabinofuranosylguanine (ara-G), an active compound of nelarabine, demonstrates potent cytotoxicity specifically on T-cell malignancies. In cells, ara-G is phosphorylated to ara-G triphosphate (ara-GTP), which is subsequently incorporated into DNA, thereby inhibiting DNA synthesis. Because ara-GTP is crucial to ara-G's cytotoxicity, the determination of ara-GTP production in cancer cells is informative for optimizing nelarabine administration. Here, we developed a new, sensitive isocratic-elution HPLC method for quantifying ara-GTP. Samples were eluted isocratically by using phosphate buffer at a constant flow rate. Ara-GTP was clearly separated from other nucleotides by using an anion-exchange column and it was quantitated by its peak area at 254 nm. The standard curve was linear with low variability and a sensitive detection limit (10 pmol). Furthermore, due to ara-G's specificity to T-cells we hypothesized that nelarabine might be effective against adult T-cell leukemia (ATL). The ara-GTP production was compared between T-lymphoblastic leukemia CCRF-CEM and ATL cell lines in vitro. When CEM cells were incubated with ara-G, the ara-GTP production increased in a concentration- and time-dependent manner. In contrast, 5 ATL cell lines accumulated lower ara-GTP in the same condition. While ara-G inhibited the growth of CEM cells with a 50% growth inhibition concentration of 2 microM, the inhibitory-concentration values were >1 mM in 8 of the 12 ATL cell lines. This ineffectiveness appeared to correspond with the low ara-GTP production. The present study is the first to evaluate the potential of ara-G against ATL cells; our results suggest that nelarabine would not be effective against ATL.


Subject(s)
Arabinonucleosides/pharmacokinetics , Arabinonucleotides/analysis , Guanosine Triphosphate/analogs & derivatives , Leukemia-Lymphoma, Adult T-Cell/metabolism , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Arabinonucleosides/metabolism , Arabinonucleosides/pharmacology , Arabinonucleosides/therapeutic use , Arabinonucleotides/metabolism , Biomarkers/analysis , Biomarkers/metabolism , Cell Proliferation/drug effects , Chemical Fractionation/methods , Chromatography, High Pressure Liquid/economics , Chromatography, High Pressure Liquid/methods , Chromatography, High Pressure Liquid/standards , Guanosine Triphosphate/analysis , Guanosine Triphosphate/metabolism , Humans , Leukemia-Lymphoma, Adult T-Cell/diagnosis , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Prodrugs/metabolism , Prodrugs/pharmacokinetics , Reference Standards , Sensitivity and Specificity , Treatment Outcome , Tumor Cells, Cultured
16.
Cancer Sci ; 97(11): 1274-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17034368

ABSTRACT

A deoxycytidine analog, gemcitabine (dFdC), is effective for treating solid tumors and hematological malignancies. After being transported into cancer cells, dFdC is phosphorylated to dFdC triphosphate (dFdCTP), which is subsequently incorporated into the DNA strand, thereby inhibiting DNA synthesis. Intracellular dFdCTP is the critical determinant for dFdC cytotoxicity, so therapeutic drug monitoring or in vitro testing of the capability of cancer cells to accumulate dFdCTP may be informative for optimizing dFdC administration. We have developed a new isocratic-elution high-performance liquid chromatography method for quantifying dFdCTP in cancer cells. Samples (500 microL) were eluted isocratically using 0.06 M Na(2)HPO(4) (pH 6.9) containing 20% acetonitrile, at a constant flow rate of 0.7 mL/min and at ambient temperature. Separation was carried out using an anion-exchange column (TSK gel DEAE-2SW; 250 mm x 4.6 mm inside diameter, particle size 5 microL) and monitored at 254 nm. The standard curve was linear with low within-day and interday variability. The lower detection limit (20 pmol) was as sensitive as that of the previous gradient-elution method. dFdCTP was well separated from other nucleoside triphosphates. The method could measure dFdCTP in cultured or primary leukemic cells treated in vitro with dFdC. The method was also applicable to simultaneous determination of dFdCTP and cytarabine triphosphate, the results of which demonstrated ara-CTP production augmented by dFdC pretreatment. Thus, our isocratic high-performance liquid chromatography assay method will be of great use because of its sensitivity and simplicity as well as its applicability to biological materials.


Subject(s)
Antimetabolites, Antineoplastic/analysis , Chromatography, High Pressure Liquid , Deoxycytidine/analogs & derivatives , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Arabinofuranosylcytosine Triphosphate/analysis , Cytarabine/analysis , Deoxycytidine/analysis , HL-60 Cells , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Sensitivity and Specificity , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...