Cellular methotrexate (MTX) resistance may cause treatment failure in childhood common/preB-acute lymphoblastic leukemia (c/preB-ALL), T-lineage ALL (T-ALL), and acute myeloid leukemia (AML). The ex vivo potency of several antifolates (MTX, trimetrexate [TMQ], GW1843U89, multitargeted antifolate [MTA], Raltitrexed, and ZD9331) was studied via in situ inhibition of thymidylate synthase (TS). After short-term exposure, relapsed c/preB-ALL (rALL, n = 21), T-ALL (n = 22), and AML (n = 22) were 3-fold, 10-fold, and 6-fold less sensitive to MTX (P ≤ .01) compared with initial c/preB-ALL (n = 43). This difference in resistance was not observed for TMQ. Also for GW1843U89 and MTA, no resistance was observed in rALL and AML compared with c/preB-ALL. T-ALL compared with c/preB-ALL tended to be less resistant to GW1843U89 (3-fold) and MTA (6-fold) than to MTX (10-fold) (P= .06). Raltitrexed was more active against c/preB-ALL compared with the other subtypes. After 21 hours continuous incubation, T-ALL and AML samples were equally sensitive as c/preB-ALL to MTX, but rALL was 3-fold resistant to MTX compared with initial c/preB-ALL (P = .003). The resistance of rALL was circumvented by TMQ (1-fold; P = .03) and GW1843U89 (1.4-fold; P= .004). Novel antifolates, except MTA, displayed a more potent TS inhibition than MTX during continuous exposure. These results suggest that MTX resistance in AML and T-ALL can be circumvented by continuous exposure, and that novel antifolates should be explored further for MTX-resistant T-ALL, rALL, and AML cells.

RESISTANCE TO methotrexate (MTX) may contribute to treatment failure as observed in one fifth of children diagnosed with common/preB-acute lymphoblastic leukemia (c/preB-ALL).1 The risk for relapse is partly associated with pharmacokinetic parameters, as individualized treatment to sustain MTX plasma levels at a specific level improved survival of B-lineage ALL patients.2 The correlation of accumulation of MTX and MTX polyglutamates in leukemic blasts with short-term antileukemic effect3 and with event-free survival4 suggested that also cellular MTX resistance is involved in treatment failure.

At relapse, ALL patients have a relatively poor prognosis with an event-free survival of 20% to 40%.5,6 Knowledge of the frequency of occurrence and mechanisms of MTX resistance in relapsed ALL is limited. Diminished MTX membrane transport via the reduced folate carrier (RFC)7 and elevated levels of dihydrofolate reductase (DHFR),8 the main target enzyme of MTX, have been detected in the majority of relapsed ALL samples. The latter might be associated with DHFR gene amplification as observed after MTX treatment in one third of relapsed ALL samples.9 Mutations in DHFR resulting in a lower affinity for MTX do not seem to play an important role in acquired MTX resistance in the clinical situation.10,11 

Compared with B-lineage ALL, T-lineage ALL (T-ALL) requires more intensive therapy to be cured.1 Cellular MTX resistance, as was detected in T-ALL by an in situ enzyme inhibition assay,12 may be involved in this relative chemoresistance. T-ALL cells were reported to accumulate lower levels of MTX compared with B-lineage ALL cells, accompanied by a less efficient polyglutamylation, in vitro13,14 and in vivo.15,16 Additional studies demonstrated that the inefficient polyglutamylation was associated with a lower folylpolyglutamate synthetase (FPGS) activity,14,17 but not with a higher folylpolyglutamate hydrolase (FPGH) activity.14 In addition, DHFR levels were higher in blasts obtained from patients with T-ALL compared with B-lineage ALL,8 resulting in lower levels of unbound MTX available for polyglutamylation.18 No difference in MTX uptake was observed, which could have explained the lower accumulation in T-ALL compared with c/preB-ALL.8 

Children diagnosed with acute myeloid leukemia (AML) are usually not treated with MTX because early clinical trials demonstrated low response rates to MTX in this type of leukemia.19Combination chemotherapy for AML, applying maximum tolerated doses that are sometimes even surpassed, results in an event-free survival of 40% to 50%.20 In addition to the toxicity of the treatment, the high rate of relapse in AML explains the poorer prognosis as compared with ALL. The high rate of relapse is likely to be associated with cellular drug resistance.21 To improve prognosis of these children, novel drugs with activity against AML are required.22,23 The presumed intrinsic MTX resistance in AML cells has been ascribed to an impaired MTX polyglutamylation14,24,25 associated with both a decrease in FPGS activity14,17,26 and an increase in activity of FPGH.14 In addition, MTX uptake may be defective as observed in one third of the AML samples,7 whereas also high levels of (altered) DHFR have been reported for AML blasts.27 

We recently described that cellular resistance to MTX and the potency of new antifolates against blast cells from leukemia patients can be evaluated using an in situ thymidylate synthase (TS) inhibition assay.12,28 In the present study, we investigated the potency of novel antifolates designed to circumvent MTX resistance by (1) bypassing transport deficiency (trimetrexate29[TMQ]), (2) not being dependent on polyglutamylation (TMQ and ZD933130), (3) having an improved affinity towards FPGS and RFC (GW1843U89,31 Raltitrexed,32 multitargeted antifolate33 [MTA]), or by (4) targeting other enzymes than DHFR (GW1843U89,31 Raltitrexed,32ZD9331,30 and MTA33). We show that continuous long-term MTX exposure can circumvent MTX resistance in T-ALL and AML and that novel antifolates show potency to circumvent MTX resistance after short-term exposure in relapsed ALL, T-ALL, and AML.

Patient Specimens

Bone marrow and peripheral blood samples were obtained after informed consent from 49 children with newly diagnosed common/preB-ALL, 22 with B-precursor ALL at relapse (rALL) previously treated with MTX, 24 with newly diagnosed T-lineage ALL, and from 26 AML patients (French-American-British [FAB] classifications: 1 M0; 5 M1; 6 M2; 2 M3; 6 M4; 2 M5; 1 M6; 1 M7; and 2 unknown). None of the patients have been treated with 1 of the novel antifolates. The number of antifolates screened per sample was dependent on the number of cells available. MTX sensitivity data for 75% of the untreated ALL samples, presented as reference values in the present report, have been reported previously.12 Immunophenotype of the ALL patients was determined by immunocytochemistry: c/preB-ALL was defined as HLA-DR+/CD19+ and terminal deoxynucleotidyl transferase (TdT) positive, excluding the proB-ALL phenotype (CD10, cytoplasmic μ-chain negative). Samples were scored T-ALL when positive for TdT, CD3, and CD7. Newly diagnosed ALL patients were classified as high-risk if 1 or more of the following features were present: T-lineage phenotype, white blood cell counts ≥25/nL and younger than 1 or older than 10 years of age. Eighty percent of the newly diagnosed c/preB-ALL samples were derived from high-risk patients.

Mononuclear cells were isolated within 48 hours after sampling by centrifugation (500g, 25 minutes) with Ficoll Isopaque and washed twice in RPMI containing 1% fetal calf serum (FCS) with 10-minute periods of centrifugation at 300g and suspended at 1 × 106 cells/mL in culture medium (RPMI 1640 containing 20% heat inactivated FCS, 100 IU/mL penicillin, 100 μg/mL streptomycin, 0.125 μg/mL fungizone, 200 μg/mL gentamycin, 2 mmol/L L-glutamine, 5 μg/mL insulin, 5 μg/mL transferrin, and 5 ng/mL sodium selenite). If necessary, contaminating normal cells were eliminated using monoclonal antibodies linked to magnetic beads, as previously described.34 At the time of assay, the percentage of blasts was greater than 80% as morphologically determined by May-Grünwald-Giemsa staining of the cytospins. Samples, which had been cryopreserved in 10% dimethyl sulfoxide and 20% FCS, were assayed shortly after cells were thawed, washed, and resuspended in prewarmed culture medium.

Chemicals

MTX was a gift from Pharmachemie (Haarlem, The Netherlands). Trimetrexate35 (5-methyl-6-[(3,4,5-trimethoxyphenyl amino) methyl] 2,4-quinazolinediamide, TMQ), GW1843U8931((S)-2-(5-(((1,2-dihydro-3-methyl-1-oxo-benzo(F)quinazolin-9-yl)methyl)-amino)-1-oxo-2-isoindolinyl)glutaric acid) and MTA33(N-{4-[2-(2-amino-3,4-dihydro-4-oxo-7H-pyrrolo[2,3-d]pyrimidin-5-yl)ethyl]-benzoyl]-L-glutamic acid, LY231514) were provided by Warner-Lambert/ Parke Davis (Ann Arbor, MI), Glaxo Wellcome (Research Triangle Park, NC), and Eli Lilly Research Laboratories (Indianapolis, IN), respectively. Raltitrexed32{N-[5-[N-(3,4-dihydro-2-methyl-4-oxo-quinazolin-6-ylmethyl)-N-methylamino]-2-thenoyl]-L-glutamic acid} (Tomudex, ZD1694) and ZD933130 (gamma tetrazole analogue of 2-desamino-2,7-dimethyl-N10-propargyl-2’fluoro-5,8-dideazafolic acid) were obtained from Zeneca Pharmaceuticals (Macclesfield, UK).

FCS, penicillin, streptomycin, fungizone, gentamycin, and L-glutamine were obtained from Flow Laboratories (Irvine, UK), bovine serum albumin (BSA) from Organon Technika (Oss, The Netherlands), and Ficoll Isopaque (Lymphoprep 1.077 g/mL) was purchased from Nyegaard (Oslo, Norway). Insulin, transferrin, and sodium selenite were purchased from Sigma (Zwijndrecht, The Netherlands) and [5-3H]-2′-deoxycytidine (25 Ci/mmol) from Moravek Biochemicals (Brea, CA). RNAzol was purchased from Campro Scientific (Veenendaal, The Netherlands); Moloney Murine Leukemia Virus Reverse Transcriptase (M-MLV-RT) from Promega (Madison, WI), and Taq Polymerase from Pharmacia Biotech (Roosendaal, The Netherlands).

In Situ TS Inhibition Assay

Because antifolate sensitivity of patient-derived leukemia samples cannot be determined using conventional cytotoxicity assays,12 we determined TS inhibition in intact cells based on a previously described assay,12,28,36 measuring the TS-catalyzed conversion of 3H-deoxyuridylate (3H-dUMP) to deoxythymidylate (dTMP) and3H2O. In cell lines, we previously demonstrated a good correlation between antifolate sensitivity determined by conventional cytotoxicity assays and the in situ TS inhibition assay for MTX and for novel rationally designed antifolates.12,28 In short, 135-μL cell suspensions (1 × 106 cells/mL) were incubated at 37°C with either 15 μL RPMI as controls or with 15 μL drug solution. Blanks in triplicate were included containing 135 μL culture medium and 15 μL RPMI. Two conditions were tested: (1) long-term continuous incubation in which cells were incubated with drugs for 21 hours and (2) short-term exposure in which the drug was washed away after 3 hours followed by an 18-hour drug-free period. Five concentrations of each antifolate were tested in duplicate for each condition as demonstrated in Table 1. Controls without drug were included in triplicate for both conditions. [5-3H]-2′-deoxycytidine (final concentration 1 μmol/L, 2.5 Ci/mmol) was added 4 hours after the start of the experiment as precursor for [5-3H]-dUMP, the substrate for TS.

Table 1.

Concentrations of Antifolates Tested in the In Situ TS Inhibition Assay

Antifolate Short Incubation (μmol/L) Continuous Incubation (μmol/L)
MTX  0.156-40  0.0039-1  
TMQ 0.08-50  0.0016-1  
GW1843U89  0.001-0.63  0.00025-0.064 
MTA  0.039-10  0.039-10  
Raltitrexed  0.0013-0.32 0.00063-0.16  
ZD9331  0.04-25  0.0016-0.4 
Antifolate Short Incubation (μmol/L) Continuous Incubation (μmol/L)
MTX  0.156-40  0.0039-1  
TMQ 0.08-50  0.0016-1  
GW1843U89  0.001-0.63  0.00025-0.064 
MTA  0.039-10  0.039-10  
Raltitrexed  0.0013-0.32 0.00063-0.16  
ZD9331  0.04-25  0.0016-0.4 

After a total incubation time of 21 hours, cells were put on ice and 150 μL 35% ice-cold trichloroacetic acid (TCA) was added together with 750 μL 10% activated charcoal solution (10 g washed charcoal, 0.5 g Dextran, and 2.5 g BSA in 100 mL demineralized water).37 After vortexing, samples were left on ice for 30 minutes and centrifuged (800g, 30 minutes, 4°C); 450 μL of the aqueous phase, containing 3H2O, was transferred to a scintillation vial and counted for radioactivity. After subtraction of the mean blank counts, the data were evaluated by calculation of the concentration of drug needed to inhibit 50% of the control TS activity, assuming a linear dose-response curve between the 2 flanking concentration points. Data were expressed as TSI50,cont referring to the continuous exposure condition and as TSI50,short for the short exposure condition. To evaluate the potency of novel antifolates to circumvent MTX resistance within individual samples, normalized TSI50 ratios were calculated by dividing the TSI50 values for each antifolate with the median TSI50 value obtained for that antifolate for the group of “MTX-sensitive” c/preB-ALL samples.

Competitive Template Reverse Transcriptase-Polymerase Chain Reaction (RT-PCR)

RNA was extracted from five million cells by the RNAzol method and reversed transcribed by Moloney Murine Leukemia Virus reverse transcriptase, as described by the manufacturer with minimal modifications. Competitive templates (CTs) were designed for RFC, DHFR, TS, FPGS, and FPGH and dissolved in standardized mixtures to avoid irreproducibilities as described elsewhere38 (manuscript submitted). PCRs were performed by adding different primer pairs to different aliquots of the same mastermix containing patients’ cDNA and CT mixture. PCR products (cDNA, CT, and heterodimers) were separated on agarose gel and measured by densitometry.

Statistics

The Mann-Whitney U test was applied to evaluate differences in antifolate sensitivity between the group of c/preB-ALL versus the group of T-ALL, relapsed ALL, or AML cells. The Wilcoxon signed ranks test was conducted to analyze the potency of a novel antifolate to circumvent MTX resistance within individual samples by comparing the normalized TSI50 ratios for MTX with those for a novel antifolate. To analyze any correlation between antifolate sensitivity profiles and mRNA expression levels, Spearman rank correlation coefficients were calculated.

Potency of MTX to Inhibit In Situ TS Activity in Childhood Leukemia Subtypes

To determine whether relative clinical chemoresistance of relapsed c/preB-ALL, T-lineage ALL and AML compared with c/preB-ALL was reflected by ex vivo testing of MTX sensitivity, we used the in situ TS inhibition assay as described previously.12Ranges of TSI50 values are presented in Table 2. Compared with c/preB-ALL, the median TSI50,short values for MTX were higher for relapsed c/preB-ALL, T-lineage ALL and AML (Table 3and Fig 1A through D [closed squares]).

Table 2.

Range of In Situ TSI50 Values for Novel Antifolates in Clinical Leukemia Samples and Plasma Levels Obtained in Phase I/II Clinical Trials

TSIcont (μmol/L) TSIshort (μmol/L) Plasma Levels Trial Reference
MTX 0.01-0.78  <0.16 − >40.0  25 μmol/L  Children Evans et al49 
TMQ  <0.0016-0.16 <0.08-14.3  7 μmol/L  Children  Hum et al45 
GW1843  <0.00025-0.047  <0.0010 >0.63  3 μmol/L  Adults  Burris et al50 
MTA  <0.039 − >10.0  <0.039 − >10.0 300 μmol/L  Adults  Rinaldi51 
Raltitrexed 0.0014-0.092  0.0043 − >0.32  3 μmol/L  Children Widemann et al46 
ZD9331  0.0016-0.19 0.9 − >25.0  No data published 
TSIcont (μmol/L) TSIshort (μmol/L) Plasma Levels Trial Reference
MTX 0.01-0.78  <0.16 − >40.0  25 μmol/L  Children Evans et al49 
TMQ  <0.0016-0.16 <0.08-14.3  7 μmol/L  Children  Hum et al45 
GW1843  <0.00025-0.047  <0.0010 >0.63  3 μmol/L  Adults  Burris et al50 
MTA  <0.039 − >10.0  <0.039 − >10.0 300 μmol/L  Adults  Rinaldi51 
Raltitrexed 0.0014-0.092  0.0043 − >0.32  3 μmol/L  Children Widemann et al46 
ZD9331  0.0016-0.19 0.9 − >25.0  No data published 
Table 3.

Median TSI50,short Values (μmol/L) for MTX and Novel Antifolates and Normalized TSI50,short Ratios in Childhood Leukemia Subtypes

Initial c/preB-ALLRelapsed c/preB-ALL (rALL)T-ALL AML
TSI50,short (μmol/L)TSI50,short (μmol/L) Normalized TSI50,short Ratio TSI50,short (μmol/L) Normalized TSI50,short RatioTSI50,short (μmol/L) Normalized TSI50,short Ratio
MTX  0.38  1.13-150 2.9 3.73-151 9.8  2.23-151 5.7  
TMQ  1.5  1.3  0.853-152 1.4  0.893-151 0.333-151 0.223-151 
GW1843U89  0.0067 0.0037  0.553-150 0.0213-150 3.13-153 0.0056  0.843-151 
MTA 0.21  0.33  1.63-153 1.43-153 6.53-153 0.41  1.93-150 
Raltitrexed  0.0096  0.0473-152 4.9  0.0373-150 3.9 0.0423-150 4.4 
Initial c/preB-ALLRelapsed c/preB-ALL (rALL)T-ALL AML
TSI50,short (μmol/L)TSI50,short (μmol/L) Normalized TSI50,short Ratio TSI50,short (μmol/L) Normalized TSI50,short RatioTSI50,short (μmol/L) Normalized TSI50,short Ratio
MTX  0.38  1.13-150 2.9 3.73-151 9.8  2.23-151 5.7  
TMQ  1.5  1.3  0.853-152 1.4  0.893-151 0.333-151 0.223-151 
GW1843U89  0.0067 0.0037  0.553-150 0.0213-150 3.13-153 0.0056  0.843-151 
MTA 0.21  0.33  1.63-153 1.43-153 6.53-153 0.41  1.93-150 
Raltitrexed  0.0096  0.0473-152 4.9  0.0373-150 3.9 0.0423-150 4.4 

TSI50,short values were measured by the in situ TS inhibition assay after a 3-hour drug exposure followed by an 18-hour drug-free period. Mann-Whitney U test was applied to analyze differences between untreated c/preB-ALL and relapsed c/preB-ALL (rALL), T-ALL or AML. Normalized TSI50,short was calculated as (TSI50,short for individual sample)/(median TSI50,short for group of c/preB-ALL). Wilcoxon signed ranks test was applied to analyze differences between normalized TSI50,short ratios for a novel antifolate and for MTX. No results are presented for ZD9331 because the majority of the samples required more than the highest concentration tested in this study to inhibit 50% of the TS activity.

F3-151

P ≤ .001;

F3-150

P ≤ .01;

F3-152

P < .05;

F3-153

P < .1.

Fig. 1.

Ranges of sensitivities to MTX, TMQ, GW1843U89, MTA, Raltitrexed, and ZD9331 as determined by the in situ TS inhibition assay for (A) c/preB-ALL; (B) relapsed c/preB-ALL (rALL); (C) T-ALL; and (D) AML. Number of samples tested are depicted at the bottom. Median values are represented by squares, the 25th and 75th percentiles by horizontal lines. Closed squares represent the short-term exposure, open squares the continuous exposure. For ZD9331, which has not been assayed for T-ALL samples, the closed squares represent a median TSI50,short of >25 μmol/L.

Fig. 1.

Ranges of sensitivities to MTX, TMQ, GW1843U89, MTA, Raltitrexed, and ZD9331 as determined by the in situ TS inhibition assay for (A) c/preB-ALL; (B) relapsed c/preB-ALL (rALL); (C) T-ALL; and (D) AML. Number of samples tested are depicted at the bottom. Median values are represented by squares, the 25th and 75th percentiles by horizontal lines. Closed squares represent the short-term exposure, open squares the continuous exposure. For ZD9331, which has not been assayed for T-ALL samples, the closed squares represent a median TSI50,short of >25 μmol/L.

Close modal

Because a continuous exposure to MTX can bypass some potential mechanisms of MTX resistance,12,39,40 cells were continuously exposed to MTX. TSI50,cont. values for MTX were higher for relapsed c/preB-ALL cells compared with initial c/preB-ALL cells. In contrast, TSI50,cont. values for MTX in T-ALL and AML samples were comparable to those in c/preB-ALL (Table 4 and Fig 1A through D [open squares]).

Table 4.

Median TSI50,cont. Values (μmol/L) for MTX and Novel Antifolates and Normalized TSI50,cont. Ratios in Childhood Leukemia Subtypes

Initial c/preB-ALLRelapsed c/preB-ALL (rALL)T-ALL AML
TSI50,cont. (μmol/L)TSI50,cont. (μmol/L) Normalized TSI50,cont. Ratio TSI50,cont. (μmol/L) Normalized TSI50,cont. RatioTSI50,cont. (μmol/L) Normalized TSI50,cont. Ratio
MTX  0.058  0.174-150 3.0 0.080  1.4  0.051  0.88  
TMQ  0.022  0.022 1.0 0.012  0.564-151 0.00464-151 0.214-151 
GW1843U89 0.0017  0.0023  1.44-150 0.0036  2.14-153 0.0021  1.2 
MTA  0.076  0.11  1.5  0.414-151 5.44-151 0.11 1.5 
Raltitrexed  0.0047  0.011 2.3  0.0274-151 5.8 0.0134-150 2.8 
ZD9331  0.017  0.045  2.6 0.060  3.54-151 0.017  1.0 
Initial c/preB-ALLRelapsed c/preB-ALL (rALL)T-ALL AML
TSI50,cont. (μmol/L)TSI50,cont. (μmol/L) Normalized TSI50,cont. Ratio TSI50,cont. (μmol/L) Normalized TSI50,cont. RatioTSI50,cont. (μmol/L) Normalized TSI50,cont. Ratio
MTX  0.058  0.174-150 3.0 0.080  1.4  0.051  0.88  
TMQ  0.022  0.022 1.0 0.012  0.564-151 0.00464-151 0.214-151 
GW1843U89 0.0017  0.0023  1.44-150 0.0036  2.14-153 0.0021  1.2 
MTA  0.076  0.11  1.5  0.414-151 5.44-151 0.11 1.5 
Raltitrexed  0.0047  0.011 2.3  0.0274-151 5.8 0.0134-150 2.8 
ZD9331  0.017  0.045  2.6 0.060  3.54-151 0.017  1.0 

TSI50,cont. values were measured by the in situ TS inhibition assay after 21 hours of continuous drug exposure. Mann-Whitney U test was applied to analyze differences between untreated c/preB-ALL and relapsed c/preB-ALL (rALL), T-ALL, or AML. Normalized TSI50,cont. was calculated as (TSI50,cont. for individual sample)/(median TSI50,cont. for group of c/preB-ALL). Wilcoxon signed ranks test was applied to analyze differences between normalized TSI50,cont. ratios for a novel antifolate and for MTX.

F4-150

P ≤ .01;

F4-151

P ≤ .001;

P ≤ .05;

F4-153

P < .1.

Potency of Novel Antifolates to Inhibit In Situ TS Activity in c/preB-ALL

Of the novel antifolates, only the polyglutamatable TS-inhibitors (GW1843U89 and Raltitrexed) displayed lower TSI50,shortvalues than MTX (Table 3 and Fig 1A). MTA, an inhibitor of both DHFR and TS displayed similar TSI50,short values. Both nonpolyglutamatable antifolates, TMQ, and ZD9331 showed a lower activity than MTX; the median TSI50,short for the DHFR inhibitor TMQ was 4-fold higher than for MTX, but for ZD9331, TSI50,short values were generally higher than the highest concentration used in the assay (25 μmol/L).

Prolonged exposure to the antifolates showed a completely different pattern. For all drugs, except for MTA, median TSI50,cont. values were lower than for MTX (Table 4 and Fig1A).

Potency of Novel Antifolates to Circumvent MTX Resistance in Relapsed c/preB-ALL (rALL)

Relapsed c/preB-ALL compared with initial c/preB-ALL.

Because several of the novel antifolates displayed a differential better activity than MTX in c/preB-ALL, we also tested these compounds in the MTX-resistant subtypes. No significant differences in TSI50,short values were observed between rALL and initial c/preB-ALL for TMQ, GW1843U89, MTA, or ZD9331 (Table 3 and Fig 1B). When exposed to Raltitrexed, the median TSI50,short was higher for rALL compared with initial c/preB-ALL.

Relapsed c/preB-ALL is the only subgroup in this study that remained more resistant to MTX during continuous incubation compared with initial c/preB-ALL (Table 4 and Fig 1B). In contrast, no significant differences in TSI50,cont. values between rALL and initial c/preB-ALL cells were observed for TMQ, GW1843U89, MTA, or ZD9331. For Raltitrexed, however, the median TSI50,cont. was higher for rALL compared with c/preB-ALL.

Novel antifolates compared with MTX (relapsed c/preB-ALL).

To evaluate the relative potency of each antifolate compared with MTX within individual samples, normalized TSI50 ratios were calculated by dividing the TSI50 values obtained for each individual rALL sample by the median TSI50 value obtained for the group of initial c/preB-ALL samples. The median normalized TSI50,short ratio for rALL was 3 for MTX (Table 3). The median normalized TSI50,short ratio was lower for TMQ and for GW1843U89 compared with MTX. For MTA and Raltitrexed, the normalized TSI50,short ratios were not different from the normalized TSI50,short ratios calculated for MTX. For ZD9331, again the TSI50,short values were higher than the highest concentration tested.

The normalized TSI50,cont. ratio for MTX was 3 for rALL. The median normalized TSI50,cont. ratios were lower for TMQ and GW1843U89 than for MTX (Table 4). No significant decrease in normalized TSI50,cont. ratios was observed for MTA, Raltitrexed, and for ZD9331 compared with that for MTX.

Potency of Novel Antifolates to Circumvent MTX Resistance in T-ALL

T-ALL compared with initial c/preB-ALL.

TSI50,short values for GW1843U89 and Raltitrexed were higher for T-ALL samples compared with c/preB-ALL samples, while no significant differences between T-ALL and c/preB-ALL were observed for TMQ and for MTA (Table 3 and Fig 1C). As for MTX, no differences in TSI50,cont. values were observed between the group of T-ALL samples and the group of c/preB-ALL samples for TMQ, GW1843U89, and ZD9331 (Table 4). T-ALL samples, however, were more resistant to MTA and to Raltitrexed compared with c/preB-ALL after continuous exposure.

Novel antifolates compared with MTX (T-ALL).

The median normalized TSI50,short was 10 for MTX in the group of T-ALL samples (Table 3). The median normalized TSI50,short ratio was significantly lower for TMQ and tended to be lower for GW1843U89 and MTA. For Raltitrexed, normalized TSI50,short ratios were not significantly different from those obtained for MTX. The median normalized TSI50,cont.was 1.4 for MTX in the group of T-ALL samples (Table 4). Normalized TSI50,cont. ratios were lower for TMQ, equal for GW1843U89, but higher for MTA, Raltitrexed, and ZD9331 compared with MTX.

Potency of Novel Antifolates to Circumvent MTX Resistance in AML

AML compared with c/preB-ALL.

The median TSI50,short value was lower for TMQ and higher for Raltitrexed in the group of AML compared with c/preB-ALL samples (Table 3 and Fig 1D). TSI50,cont. values were also lower for TMQ and higher for Raltitrexed for AML compared with c/preB-ALL (Table 4). No significant differences between AML and c/preB-ALL were observed for GW1843U89 and MTA after short exposure or during continuous exposure.

Novel antifolates compared with MTX (AML).

The median normalized TSI50,short ratio for MTX was 6 for AML samples (Table 3). Median normalized TSI50,short ratios were lower for TMQ, GW1843U89, and MTA than for MTX. The median normalized TSI50,cont. for MTX was 0.9 for AML samples (Table 4). Normalized TSI50,cont. ratios were lower for TMQ, equal for GW1843U89 and ZD9331, and higher for MTA and Raltitrexed compared with MTX.

Molecular Biological Correlates

To investigate whether antifolate sensitivity was correlated with mRNA levels of proteins involved in folate metabolism, we developed competitive templates to determine the mRNA levels of RFC, DHFR, TS, FPGS, and FPGH. No correlations were observed for antifolate sensitivity and mRNA levels of RFC, DHFR, or TS in a group of 16 samples for which these parameters were determined. However, for the enzymes involved in polyglutamylation, we observed a significant correlation between a high ratio of FPGH/FPGS mRNA levels and high TSI50,short values for MTX (r = .64, P = .007) and for MTA (r = .58, P = .030). For MTX, this could not be explained by a correlation with the mRNA levels of one of the enzymes independently. For MTA, however, high FPGH mRNA levels were correlated with high TSI50,short values (r = .58,P = .024), whereas low FPGS mRNA levels were correlated with high TSI50,cont. values (r = −.44, P= .051). Also for Raltitrexed and ZD9331, low FPGS mRNA levels were correlated with high TSI50,cont. values (r = −.77, P = .009 and r = −.49, P = .055, respectively). For GW1843U89, a trend was observed between low TSI50,short and high FPGS mRNA levels (r = −.45, P = .056).

In the present study, ex vivo experiments showed relative MTX resistance in relapsed c/preB-ALL, T-ALL, and AML samples compared with initial c/preB-ALL after short-term drug exposure. This MTX resistance could be circumvented by continuous drug exposure for T-ALL and AML. Thus, with respect to long-term continuous drug exposure, ALL at relapse was the only leukemia subgroup for which resistance to MTX was still observed. Five rationally designed folate antagonists (TMQ, GW1843U89, MTA, Raltitrexed, and ZD9331) displayed a marked potency to circumvent the observed MTX resistance in childhood leukemia cells.

We previously demonstrated that for cell lines, sensitivity to MTX and to novel antifolates, as measured via conventional cytotoxicity assays, correlated with sensitivity measured via the in situ TS inhibition assay, both for short and continuous exposure conditions.12,28 As antifolate sensitivity of primary leukemia samples cannot be determined using conventional assays, as demonstrated and discussed previously,12 we determined the potency of novel antifolates versus MTX on the basis of in situ TS inhibition for all leukemia subgroups. The specific TS inhibitors GW1843U89, Raltitrexed, and ZD9331 showed markedly lower TSI50,cont. values than observed for MTX, which reflects their enhanced RFC-mediated membrane transport and more potent TS inhibition.30-32 Moreover, GW1843U89 and Raltitrexed were more efficiently retained during the drug-free period, as illustrated by a diminished difference between median values of TSI50,short and TSI50,cont compared with this difference for MTX (Fig 1). This result is consistent with a superior polyglutamylation of these compounds by FPGS as compared with MTX.31,32 ZD9331 as a nonpolyglutamatable drug required continuous exposure to inhibit TS; after a drug-free period, inhibition of TS activity was not sustained.28 

To show whether MTX resistance can be circumvented by novel antifolates, we analyzed the differential potency of antifolates between the leukemia subtypes. rALL cells were resistant to MTX both after short-term exposure and after continuous exposure as compared with initial c/preB-ALL samples. Resistance to MTX after continuous drug exposure was not observed for T-ALL and AML, suggesting a different mechanism of MTX resistance in rALL compared with T-ALL and AML. Both after short-term and after continuous exposure, MTX resistance could be circumvented by TMQ and GW1843U89, but not by MTA and Raltitrexed in rALL. RFC transport defects, which have been reported to contribute to MTX resistance in rALL cells,7can also reduce transport of Raltitrexed.32,41 On the other hand, residual RFC activity may still allow sufficient accumulation of GW1843U89 in rALL cells to confer drug sensitivity.

T-ALL was MTX resistant only after short-term exposure as compared with initial c/preB-ALL. This resistance could be circumvented by continuous MTX exposure, while the nonpolyglutamatable antifolate, TMQ, was equally potent in both subtypes. Because polyglutamylation defects could be overcome by continuous exposure in cell line experiments,40 the circumvention of MTX resistance in these primary leukemia samples is in agreement with the observation that T-ALL samples are less capable of polyglutamylation compared with initial c/preB-ALL samples.13,14,40 For TMQ, the circumvention of MTX resistance is of particular interest because increased DHFR levels have been reported as an additional mechanism of MTX resistance in T-ALL cells.8 Although DHFR is the target enzyme of TMQ, these small elevations of DHFR apparently do not result in cross-resistance to TMQ. The improved activity of TMQ over MTX might reflect its sufficient intracellular accumulation42 to levels that can sustain DHFR inhibition both in c/preB-ALL and T-ALL cells. T-ALL tended to be less resistant to GW1843U89 and MTA, but not to Raltitrexed, as compared with MTX. Despite the low number of samples tested for Raltitrexed sensitivity, this suggests that resistance due to a low FPGS activity, as reported for T-ALL cells previously,14,17 cannot be circumvented by the increased affinity of FPGS for Raltitrexed. This is consistent with studies on MTX-resistant human leukemia cell lines.43 AML samples, which displayed MTX resistance only after short-term drug exposure, were sensitive to TMQ, GW1843U89, and MTA after short-term exposure. In contrast, Raltitrexed was not able to circumvent MTX resistance ex vivo in these AML samples. Whether this is solely due to the lower activity of FPGS14,17 along with altered kinetic properties,26 as reported for AML cells, or a multifactorial process involving decreased transport7 and increased FPGH activity,14 is presently unknown.

We and others previously described a good correlation between TSI50,short values, MTX polyglutamylation, and the ratio of FPGH/FPGS activities12,14,44 or FPGH/FPGS mRNA levels (present report). The present study shows no correlations for TSI50 values and mRNA expression of the target enzymes DHFR and TS, or of RFC, for any of the antifolates. This suggests that mutations or posttranslational modifications are of more importance in determining the overall response to these drugs. For GW1843U89 and MTA, correlations were observed between TSI50,short and mRNA expression levels of FPGH and FPGS, respectively. This might suggest that MTX resistance due to high or low mRNA expression of FPGH or FPGS, respectively, could not be circumvented by these antifolates. Despite these correlations, however, GW1843U89 and MTA demonstrated good potency to circumvent MTX resistance in childhood T-ALL and AML cells.

To date, only TMQ45 and Raltitrexed46 have been administered in pediatric clinical trials. Peak plasma levels obtained during these trials, together with plasma levels of GW1843U89 and MTA obtained in adults, are provided in Table 2. Although these values suggest that the presented TSI50 values for the novel antifolates are clinically achievable, some considerations have to be kept in mind. Binding of drugs to serum proteins (in vivo) or to plastic (in vitro) and technical parameters, such as incubation time, make it difficult to extrapolate in vitro data to in vivo values.

Although the poor prognosis of AML and ALL at relapse warrants novel treatment modalities, low numbers of pediatric patients are eligible for entry into phase I/II trials. This is due to the low incidence of childhood cancer and the relatively good prognosis of the majority of the cases. Therefore, the agents to be chosen for further studies need to be prioritized.22 The availability of biochemical assays, such as the in situ TS inhibition assay, and molecular probes7,16,47 to analyze MTX-resistance parameters, may facilitate the initiation of phase I/II clinical trials in childhood leukemia with novel antifolates.22,23,48 

In conclusion, these ex vivo results suggest that novel antifolates could be promising candidates for further evaluation in clinical trials with T-ALL, rALL, and AML.

Supported by Grant No. VU 94-679 from the Dutch Cancer Society.

The publication costs of this article were defrayed in part by page charge payment. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. section 1734 solely to indicate this fact.

1
Pui
CH
Evans
WE
Acute lymphoblastic leukemia.
N Engl J Med
339
1998
605
2
Evans
WE
Relling
MV
Rodman
JH
Crom
WR
Boyett
JM
Pui
C-H
Conventional compared with individualised chemotherapy for childhood acute lymphoblastic leukemia.
N Engl J Med
338
1998
499
3
Masson
E
Relling
MV
Synold
TW
Liu
Q
Schuetz
JD
Sandlund
JT
Pui
C-H
Evans
WE
Accumulation of methotrexate polyglutamates in lymphoblasts is a determinant of antileukemic effects in vivo, a rationale for high-dose methotrexate.
J Clin Invest
97
1996
73
4
Whitehead
VM
Rosenblatt
DS
Vuhich
M-J
Shuster
JJ
Witte
A
Beaulieu
D
Accumulation of methotrexate and methotrexate polyglutamates in lymphoblasts at diagnosis of childhood acute lymphoblastic leukemia: A pilot prognostic factor analysis.
Blood
76
1990
44
5
Sadowitz
PD
Smith
SD
Shuster
J
Wharam
MD
Buchanan
GR
Rivera
GK
Treatment of late bone marrow relapse in children with acute lymphoblastic leukemia: A Pediatric Oncology Group study.
Blood
81
1993
602
6
Henze
G
Fengler
R
Hartmann
R
Kornhuber
B
Janka-Schaub
G
Niethammer
D
Riehm
H
Six-year experience with a comprehensive approach to the treatment of recurrent childhood acute lymphoblastic leukemia (ALL-REZ BFM 85). A relapse study of the BFM group.
Blood
78
1991
1166
7
Gorlick
R
Göker
E
Trippett
T
Steinherz
P
Elisseyeff
Y
Mazumdar
M
Flintoff
WF
Bertino
JR
Defective transport is a common mechanism of acquired methotrexate resistance in acute lymphocytic leukemia and is associated with decreased reduced folate carrier expression.
Blood
89
1997
1013
8
Matherly
LH
Taub
JW
Wong
SC
Simpson
PM
Ekizian
R
Buck
S
Williamson
M
Amylon
M
Pullen
J
Camitta
B
Ravindranath
Y
Increased frequency of expression of elevated dihydrofolate reductase in T-cell versus B-precursor acute lymphoblastic leukemia in children.
Blood
90
1997
578
9
Göker
E
Waltham
M
Kheradpour
A
Trippett
T
Mazumdar
M
Elisseyeff
Y
Schnieders
B
Steinherz
P
Tan
C
Berman
E
Bertino
JR
Amplification of the dihydrofolate reductase gene is a mechanism of acquired resistance to methotrexate in patients with acute lymphoblastic leukemia and is correlated with p53 gene mutations.
Blood
86
1995
677
10
Spencer
HT
Sorrentino
BP
Pui
CH
Chunduru
SK
Sleep
SE
Blakley
RL
Mutations in the gene for human dihydrofolate reductase: An unlikely cause of clinical relapse in pediatric leukemia after therapy with methotrexate.
Leukemia
10
1996
439
11
Gorlick
R
Göker
E
Trippett
T
Waltham
M
Banerjee
D
Bertino
JR
Intrinsic and acquired resistance to methotrexate in acute leukemia.
N Engl J Med
335
1996
1041
12
Rots
MG
Pieters
R
Kaspers
GJL
Van Zantwijk
CH
Noordhuis
P
Mauritz
R
Veerman
AJP
Jansen
G
Peters
GJ
Differential methotrexate resistance in childhood T- versus common/preB-acute lymphoblastic leukemia can be measured by an in situ thymidylate synthase inhibition assay, but not by the MTT assay.
Blood
93
1999
1067
13
Göker
E
Lin
JT
Trippett
T
Elisseyeff
Y
Tong
WP
Niedzwiecki
D
Tan
C
Steinherz
P
Schweitzer
BI
Bertino
JR
Decreased polyglutamylation of methotrexate in acute lymphoblastic leukemia blasts in adults compared to children with this disease.
Leukemia
7
1993
1000
14
Rots
MG
Pieters
R
Peters
GJ
Noordhuis
P
Van Zantwijk
CH
Hählen
K
Creutzig
U
Kaspers
GJL
Veerman
AJP
Jansen
G
The role of folylpolyglutamate synthetase and folylpolyglutamate hydrolase in methotrexate accumulation and polyglutamylation in childhood leukemia.
Blood
93
1999
1677
15
Synold
TW
Relling
MV
Boyett
JM
Rivera
GK
Sandlund
JT
Mahmoud
H
Crist
WM
Pui
CH
Evans
WE
Blast cell methotrexate-polyglutamate accumulation in vivo differs by lineage, ploidy, and methotrexate dose in acute lymphoblastic leukemia.
J Clin Invest
94
1994
1996
16
Galpin
AJ
Schuetz
JD
Masson
E
Yanishevski
Y
Synold
TW
Barredo
JC
Pui
CH
Relling
MV
Evans
WE
Differences in folylpolyglutamate synthetase and dihydrofolate reductase expression in human B-lineage versus T-lineage leukemic lymphoblasts: Mechanisms for lineage differences in methotrexate polyglutamylation and cytotoxicity.
Mol Pharmacol
52
1997
155
17
Barredo
JC
Synold
TW
Laver
J
Relling
MV
Pui
CH
Priest
DG
Evans
WE
Differences in constitutive and post-methotrexate folylpolyglutamate synthetase activity in B-lineage and T-lineage leukemia.
Blood
84
1994
564
18
Goldman
DI
Matherly
LH
The cellular pharmacology of methotrexate.
Pharmacol Ther
28
1985
77
19
Bender
RA
Anti-folate resistance in leukemia: Treatment with “high-dose” methotrexate and citrovorum factor.
Cancer Treat Rev
2
1975
215
20
Pui
C-H
Childhood Leukemias.
N Engl J Med
332
1995
1618
21
Kaspers
GJL
Kardos
G
Pieters
R
Van Zantwijk
CH
Klumper
E
Hählen
K
De Waal
FC
Van Wering
ER
Veerman
AJP
Different cellular drug resistance profiles in childhood lymphoblastic and non-lymphoblastic leukemia: A preliminary report.
Leukemia
8
1994
1224
22
Estlin
EJ
Ablett
S
Newell
DR
Lewis
IJ
Lashford
L
Pearson
AD
Phase I trials in paediatric oncology—-the European perspective. The New Agents Group of the United Kingdom Childrens Cancer Study Group.
Invest New Drugs
14
1996
23
23
Weitman
S
Carlson
L
Pratt
CB
New drug development for pediatric oncology.
Invest New Drugs
14
1996
1
24
Lin
JT
Tong
WP
Trippett
TM
Niedzwiecki
D
Tao
Y
Tan
C
Steinherz
P
Schweitzer
BI
Bertino
JR
Basis for natural resistance to methotrexate in human acute non-lymphocytic leukemia.
Leuk Res
15
1991
1191
25
Yamauchi
H
Iwata
N
Omine
M
Maekawa
T
In vitro methotrexate polyglutamate formation is elevated in acute lymphoid leukemia cells compared with acute myeloid leukemia and normal bone marrow cells.
Acta Haematol Jpn
51
1988
766
26
Longo
GS
Gorlick
R
Tong
WP
Ercikan
E
Bertino
JR
Disparate affinities of antifolates for folylpolyglutamate synthetase from human leukemia cells.
Blood
90
1997
1241
27
Dedhar
S
Hartley
D
Fitz-Gibbons
D
Phillips
G
Goldie
JH
Heterogeneity in the specific activity and methotrexate sensitivity of dihydrofolate reductase from blast cells of acute myelogenous leukemia patients.
J Clin Oncol
3
1985
1545
28
Mauritz
R
Bekkenk
M
Rots
MG
Pieters
R
Mini
E
Van Zantwijk
CH
Veerman
AJP
Peters
GJ
Jansen
G
Ex vivo activity of methotrexate versus novel antifolate inhibitors of dihydrofolate reductase and thymidylate synthase against childhood leukemia cells.
Clin Cancer Res
4
1998
2399
29
Kamen
BA
Eibl
B
Cashmore
AR
Bertino
JR
Uptake and efficiency of trimetrexate (TMQ,2,4-diamino-5-methyl-6-[(3,4,5-trimethoxyanilino)methyl] quinazoline), a non-classical antifolate in methotrexate resistant leukemia cells in vitro.
Biochem Pharmacol
33
1984
1697
30
Jackman
AL
Kimbell
R
Aherne
GW
Brunton
L
Jansen
G
Stephens
TC
Smith
MN
Wardleworth
JM
Boyle
FT
Cellular pharmacology and in vivo activity of a new anticancer agent, ZD9331—a water-soluble, nonpolyglutamatable, quinazoline-based inhibitor of thymidylate synthase.
Clin Cancer Res
3
1997
911
31
Duch
DS
Banks
S
Dev
IK
Dickerson
SH
Ferone
R
Heath
LS
Humphreys
J
Knick
V
Pendergast
W
Singer
S
Smith
GK
Waters
K
Wilson
HR
Biochemical and cellular pharmacology of 1843U89, a novel benzoquinazoline inhibitor of thymidylate synthase.
Cancer Res
53
1993
810
32
Jackman
AL
Taylor
GA
Gibson
W
Kimbell
R
Brown
M
Calvert
AH
Judson
IR
Hughes
LR
ICI D1694, a quinazoline antifolate thymidylate synthase inhibitor that is a potent inhibitor of L1210 tumor cell growth in vitro and in vivo: A new agent for clinical studies.
Cancer Res
51
1991
5579
33
Shih
C
Chen
VJ
Gossett
LS
Gates
SB
MacKellar
WC
Habeck
LL
Shackelford
KA
Mendelsohn
LG
Soose
DJ
Patel
VF
Andis
SL
Bewley
JR
Rayl
EA
Moroson
BA
Beardsley
GP
Kohler
W
Ratnam
M
Schultz
RM
LY231514, a pyrrolo[2,3-d]pyrimidine-based antifolate that inhibits multiple folate-requiring enzymes.
Cancer Res
57
1997
1116
34
Kaspers
GJL
Veerman
AJP
Pieters
R
Broekema
GJ
Huismans
DR
Kazemier
KM
Loonen
AH
Rottier
MMA
Van Zantwijk
CH
Hählen
K
Van Wering
ER
Mononuclear cells contaminating acute lymphoblastic leukaemic samples tested for cellular drug resistance using the methyl-thiazol-tetrazolium assay.
Br J Cancer
70
1994
1047
35
Lin
JT
Bertino
JR
Trimetrexate: A second generation folate antagonist in clinical trial.
J Clin Oncol
5
1987
2032
36
Rodenhuis
S
McGuire
JJ
Narayanan
R
Bertino
JR
Development of an assay system for the detection and classification of methotrexate resistance in fresh human leukemic cells.
Cancer Res
46
1986
6513
37
Peters
GJ
Laurensse
EJ
Leyva
A
Lankelma
J
Pinedo
HM
Sensitivity of human, murine and rat cells to 5-fluorouracil and 5’-deoxy-5-fluorouridine in relation to drug-metabolizing enzymes.
Cancer Res
46
1986
20
38
Willey
JC
Crawford
EL
Jackson
CM
Weaver
DA
Hoban
JC
Khuder
SA
DeMuth
JP
Expression measurement of many genes simultaneously by quantitative RT-PCR using standardized mixtures of competitive templates.
Am J Resp Cell Mol Biol
19
1998
6
39
Braakhuis
BJM
Jansen
G
Noordhuis
P
Kegel
A
Peters
GJ
Importance of pharmacodynamics in the in vitro antiproliferative activity of the antifolates methotrexate and 10-ethyl-10-deazaaminopterin against human head and neck squamous cell carcinoma.
Biochem Pharmacol
46
1993
2155
40
McGloskey
DE
McGuire
JJ
Russell
CA
Rowan
BG
Bertino
JR
Pizzorno
G
Mini
E
Decreased folylpolyglutamate synthetase activity as a mechanism of methotrexate resistance in CCRF-CEM human leukemia sublines.
J Biol Chem
266
1991
6181
41
Westerhof
GR
Kathmann
I
Schornagel
JH
Jackman
AL
Peters
GJ
Boyle
FT
Pinedo
HM
Jansen
G
Carrier- and receptor-mediated transport of folate antagonists targetting folate dependent enzymes: Correlates of molecular structure and biological activity.
Mol Pharmacol
48
1995
459
42
Rodenhuis
S
McGuire
JJ
Sawicki
WL
Bertino
JR
Effects of methotrexate and of the “nonclassical” folate antagonist trimetrexate on human leukemia cells.
Leukemia
1
1987
116
43
McGuire
JJ
Heitzman
KJ
Haile
WH
Russell
CA
McCloskey
DE
Piper
JR
Cross-resistance studies of folylpolyglutamate synthase-deficiency, methotrexate-resistant CCRF-CEM human leukemia sublines.
Leukemia
7
1993
1996
44
Longo
GS
Gorlick
R
Tong
WP
Lin
S
Steinherz
P
Bertino
JR
Gamma-glutamyl hydrolase and folylpolyglutamate synthetase activities predict polyglutamylation of methotrexate in acute leukemias.
Oncol Res
9
1997
259
45
Hum
M
Holcenberg
JS
Tkaczewski
I
Weaver
JW
Wilson
J
Kamen
B
High-dose trimetrexate and minimal-dose leucovorin: A case for selective protection?
Clin Cancer Res
4
1998
2981
46
Widemann
BC
Balis
FM
Reaman
GH
Sato
J
O’Brian
M
Krailo
M
Adamson
PC
Pediatric phase I trial and phamacokinetic (PK) study of Raltitrexed (ZD1694, Tomudex).
Proc Am Soc Clin Oncol
18
1999
563a
(abstr)
47
Rots
MG
Noordhuis
P
Willey
JC
Van Zantwijk
CH
DeMuth
JP
Veerman
AJP
Pieters
R
Peters
GJ
Jansen
G
Expression of mRNA encoding for the reduced folate carrier and folate metabolizing enzymes in childhood leukemia cells as measured with a competitive template RT-PCR method.
Ann Oncol
9
1998
162
(abstr)
48
Smith
A
Hum
M
Winick
N
Kamen
BA
A case for the use of aminopterin in treatment of patients with leukemia based on metabolic studies of blasts in vitro.
Clin Cancer Res
2
1996
69
49
Evans
WE
Crom
WR
Abromowitch
M
Dodge
R
Look
AT
Bowman
WP
George
SL
Pui
CH
Clinical pharmacodynamics of high-dose methotrexate in acute lymphocytic leukemia. Identification of a relation between concentration and effect.
N Engl J Med
314
1986
471
50
Burris
HA
Kisor
DF
Smetzer
LA
Eckardt
JR
Rodriguez
GI
Rinaldi
DA
Lampkin
TA
Bigley
JW
Von Hoff
DD
A phase I pharmacokinetic (pk) study of the thymidylate synthase inhibitor 1843U89 during a daily × 5 bolus administration schedule.
Proc Am Soc Clin Oncol
15
1996
477
(abstr)
51
Rinaldi
DA
Overview of phase 1 trials of multitargeted antifolate (MTA, LY231514).
Semin Oncol
26
1999
82

Author notes

Address reprint requests to Marianne G. Rots, Department of Pediatric Hematology/Oncology, University Hospital Vrije Universi- teit, PO Box 7057, 1007 MB Amsterdam, The Netherlands; e-mail: marianne.rots@azvu.nl.

Sign in via your Institution