In adult acute myeloid leukemia (AML), the weight of the contribution of the combined activity of Pgp and MRP1 to drug resistance is not known. To address this question, we compared the activity of these proteins to the in vitro resistance to daunorubicin (DNR), etoposide, and cytosine arabinoside (Ara-C), using the calcein-AM uptake and the 3-[4, 5-di-methyl-thiazol-2, 5-diphenyl] tetrazolium bromide (MTT) assay in 80 adult AML patients. We found no correlation or only a weak correlation between the in vitro drug resistance to DNR and etoposide and MRP1 or Pgp expression or function when tested separately. However, a strong correlation was observed between the simultaneous activity of MRP1 and Pgp (quantified as the modulation of calcein-AM uptake by cyclosporin A and probenecid) and the LC50 of DNR (r = .77, P < .0001). This emphasized the role of these two proteins, not separately, but together in the resistance to DNR. In contrast, Mvp/LRP expression did not correlate with the LC50 of DNR. A high level of simultaneous activity of Pgp and MRP1 was predictive of a poor treatment outcome (for achievement of CR [P = .008], duration of relapse-free survival [RFS; P = .01], and duration of overall survival [OS; P = .02]). In addition, high LC50 of DNR and high LC50 of etoposide together were also predictive of a poor treatment outcome (for duration of RFS [P= .02] and duration of OS [P = .02]). The unfavorable cytogenetic category was more closely associated with the combined activity of both MRP1 and Pgp (P = .002) than with the activity of Pgp or MRP1 separately. This could explain the poor prognosis and the in vitro resistance to daunorubicin in this group of patients. These data suggest that treatment outcome may be improved when cellular DNR and etoposide resistance can be circumvented or modulated. Modulation of not only Pgp but also MRP1 could be essential to attain this aim in adult AML.

MULTIDRUG RESISTANCE (MDR) of some cancers, particularly acute myeloid leukemia (AML), remains a major obstacle to successful chemotherapy. The best-characterized resistance mechanism in AML which has been shown to be associated with poor outcome is mediated by the MDR1 gene and expression of membrane P glycoprotein.1 But alternative proteins, such as the more recently recognized multidrug-associated protein (MRP1)2 or the lung-resistance protein (Mvp/LRP),3 may also contribute to the resistance to anthracyclines and etoposide in AML. However, the role of these two proteins are still under discussion.4-10In several publications, the expression of Pgp did not correlate with its function of drug efflux.1,11 For this reason, determining the functional role appears to be more informative than quantification of MDR proteins. In previous studies, we have shown in cell lines and in AML that the quantification of calcein-acetoxymethylester (calcein-AM) uptake (with or without specific modulator[s] of MRP1 and/or Pgp) can be used to assess the activity of both MRP1 and Pgp.10,12 Calcein-AM, which is a substrate of both Pgp and MRP1, becomes fluorescent after the cleavage of calcein-AM by cellular esterases, producing a measurable fluorescent derivate calcein in flow cytometry.10,12-16 

Despite its association with clinical resistance, MDR1expression was not correlated with in vitro resistance to daunorubicin (DNR) and etoposide, using the quick and semi-automatized 3-[4, 5-di-methyl-thiazol-2, 5-diphenyl] tetrazolium bromide (MTT) assay, in several studies.17-19 However, both we and others have shown that MDR1 and MRP1 gene overexpression emerged in a sequential manner during selection of different leukemic cell lines by drugs.20-22 The overexpression of the MRP1 gene preceded that of the MDR1 gene; afterward, MRP1 andMDR1 were co-overexpressed. In light of these results,MRP1 gene overexpression is probably an early event in the development of drug resistance, and clinical trials that modulated only Pgp might have limited or no success. In addition, both we and van der Kolk et al have shown that MRP1 was functional in fresh leukemic blast cells.10,23 Therefore, it is important to study the combined activity of Pgp and MRP1. To date, no study has analyzed the correlations between the simultaneous activity of Pgp and MRP1 and in vitro and in vivo drug resistance to DNR or etoposide, even though coexpression of these two proteins is common in adult AML.4Understanding of these relationships can help unravel the mechanisms of resistance to anthracyclines and etoposide which are clinically relevant in adult AML. In addition, such studies can emphasize the contribution of the combined activity of Pgp and MRP1 in comparison to other mechanisms.

Therefore, we have studied the contribution of Pgp, MRP1, and Mvp/LRP expression and Pgp and MRP1 function (using calcein-AM) to the in vitro resistance to DNR and etoposide and to the in vivo treatment result in 80 adult AML patients.

Patients.

Between July 1995 and December 1997, 80 samples from adult AML patients (60 de novo and 20 relapsed AML) were successfully tested. The diagnosis was based on French-American-British (FAB) criteria.24,25 Immunophenotyping was performed by using flow cytometry. Promyelocytic leukemia (AML3) patients were excluded from the study (because of retinoic acid treatment). For each patient, several clinical and biological characteristics were analyzed (age, white blood cell [WBC] count at diagnosis, CD34 expression, and karyotype). Unfavorable karyotypes were defined as t(9;22) or abnormalities of chromosomes 5 or 7, abnormalities of 11q2.3 band, or complex abnormalities. Inversion in chromosome 16 (inv 16) or t(8;21) indicated good prognosis, and the other karyotypes, including normal, indicated intermediate prognosis.26 Only untreated de novo AML patients (60 patients) were analyzed for treatment outcome. De novo AML patients, in our department, were included in the European Organization for the Research and Treatment of Cancer (EORTC) leukemia cooperative group protocols (AML-13 for patients ≥60 years old, and AML10 for patients <60 years old). In induction phase, all the patients received a standard dose of cytosine arabinoside (Ara-C) (100 mg/m2/d × 10 days), etoposide, and one anthracycline (DNR, idarubicin, or mitoxantrone × 3 days) at random.

Level of MDR1, MRP1, and Mvp/LRP mRNA expression.

The level of MDR1, MRP1, and Mvp/LRP mRNA expression measured by reverse transcriptase-polymerase chain reaction (RT-PCR) was described elsewhere.4,10,27 The variations between samples in the cDNA synthesis were normalized by their relative quantities of β2 microglobulin (β2m) amplified by 23 cycles of PCR. The normalized yield of MDR products relative to β2m were then compared with those of A549 cells for MRP1 and Mvp/LRP (a cell line that expressed a high level of MRP1 and Mvp/LRP) and to those of HL60/DNR for MDR1 (a cell line that expressed a high level of MDR1), which were defined as 1 arbitrary unit. All samples contained more than 80% of leukemic cells. Percentage of blast cells was determined by the May-Grünwald-Giemsa staining and by immunophenotyping performed by flow cytometry. We performed this test in 75 of the 80 patients. Correlations with clinical outcome were largely performed using results of RT-PCR as a continuous variable, in accordance with consensual recommendations.28-30 

Levels of Pgp, MRP1, and Mvp/LRP protein expression.

Pgp, MRP1, and Mvp/LRP protein expression was measured by labeling fresh viable cells with the UIC2, MRPm6, and LRP56 monoclonal antibodies (MoAbs), respectively, and phycoerythrin (PE)-labeled second antibody as described before.10 The expression of MDR proteins was established with blast cells selected by CD34 antibody (HPCA2 clone; Becton Dickinson, Le Pont de Claix, France) (two-color assays) or other markers (for example CD33/CD7, CD33/CD2, CD33/CD19, or CD33/CD22 by three-color assays) whenever possible, or with physical characteristics only if blast cells did not express characteristic markers. Fluorescence was analyzed on a FACSORT flow cytometer (Becton Dickinson). Values were expressed as adjusted for control, ie, the ratio of MoAb fluorescence/control antibody fluorescence. We performed this test in 75 of the 80 patients. Correlations with clinical outcome were largely performed using the fluorescence ratio as a continuous variable, in accordance with consensual recommendations.28-30 

Functional analysis of Pgp and MRP1 using calcein-AM.

Cells exposed to the nonfluorescent calcein-AM become fluorescent after the intracytoplasmic cleavage of calcein-AM by cellular esterases which produced the fluorescent derivate calcein. Both Pgp and MRP1 actively extruded calcein-AM.12,13 When we measured calcein-AM uptake by flow cytometry, we assessed the amount of fluorescent calcein that had been converted from nonfluorescent calcein-AM. When the Pgp and/or MRP1 proteins were active, less calcein-AM was retained and less was converted to fluorescent calcein. Therefore, calcein-AM uptake (with specific modulators of Pgp and/or MRP1) could be used to assess whether Pgp and/or MRP1 were functional.10,12-15,31,32 In our previous studies, calcein-AM uptake ± cyclosporin A (CsA) provided in AML cells a functional test as specific and sensitive as Rh123 ± CsA,10 the most specific and sensitive Pgp functional test.15,31 Calcein-AM uptake ± probenecid also provided a functional test for MRP1 in leukemic cells. Probenecid was used as specific modulator of MRP1 activity.10,12,33 

We performed this functional test in 40 adult AML patients among the 60 de novo AML patients. Cells were incubated with 0.1 μmol/L of calcein-AM for 15 minutes at 37°C in RPMI medium without or with modulators (only CsA [2 μmol/L] for Pgp function, only probenecid [2 mmol/L] for MRP1 function, or both CsA and probenecid together to assess the simultaneous activity of Pgp and MRP1). Cells were washed twice in cold phosphate-buffered saline (PBS) and samples were analyzed with a FACSORT flow cytometer. One example is shown in Fig 1. All samples were analyzed without fixation. All the data were calculated as the ratio of drug fluorescence with modulator(s) divided by drug fluorescence without modulator after subtraction of the fluorescence of the control. Dead cells were gated out following scatter characteristics.34The function of MDR proteins was established with blast cells selected as above. Correlations with clinical outcome were largely performed using fluorescence ratio as a continuous variable, in accordance with several consensual recommendations.28-30 

Fig. 1.

One example of Pgp and MRP1 activity quantified by the effect of probenecid ± CsA (modulators of MRP1 and Pgp, respectively) on the level of calcein-AM uptake. Cell fluorescence (A) without modulator, (B) with probenecid, (C) with CsA, and (D) with both probenecid and CsA together. The results were calculated as the ratio of drug fluorescence with modulator divided by drug fluorescence without modulator after subtraction of the fluorescence of the control. For this example the ratios were 1.51 with probenecid (which quantified MRP1 activity); 1.7 with CsA (which quantified Pgp activity); and 2.66 with both probenecid and CsA (which quantified the combined activity of MRP1 and Pgp).

Fig. 1.

One example of Pgp and MRP1 activity quantified by the effect of probenecid ± CsA (modulators of MRP1 and Pgp, respectively) on the level of calcein-AM uptake. Cell fluorescence (A) without modulator, (B) with probenecid, (C) with CsA, and (D) with both probenecid and CsA together. The results were calculated as the ratio of drug fluorescence with modulator divided by drug fluorescence without modulator after subtraction of the fluorescence of the control. For this example the ratios were 1.51 with probenecid (which quantified MRP1 activity); 1.7 with CsA (which quantified Pgp activity); and 2.66 with both probenecid and CsA (which quantified the combined activity of MRP1 and Pgp).

Close modal
MTT cytotoxicity test.

In vitro sensitivity of cells to DNR, Ara-C, and etoposide was determined by planting 2 × 105 cells in a 200-μL growth medium, without any specific growth factor, containing several dilutions of the drug in 96-well microtiter plates. Each concentration of drugs was repeated in six wells. After incubation for 3 days at 37°C with 5% CO2, cell viability was determined using the MTT assay as described by Plumb et al.35 Briefly, 20 μL of MTT (5 mg/mL in PBS) was added to each well and incubated for 6 hours. The medium and MTT were then removed from the wells by centrifugation, and formazan crystals were dissolved in 200 μL of dimethyl sulfoxide (DMSO). The absorbance was recorded in a microplate reader (Model MR5000; Dynatech Laboratories, France) at the wavelength of 550 nm. The effect of drug on growth inhibition could be assessed as: % of Growth Inhibition = 1 − [(Absorbance of Drug-Treated Cells/Absorbance of Untreated Cells) × 100]. The lethal concentration 50% (LC50) was determined as the drug concentration that resulted in a 50% growth inhibition. Samples were considered evaluable if the drug-free control wells contained more than 80% of leukemic cells before and more than 70% of leukemic cells after 3 days of culture. The MTT assay gave reliable results under these conditions.36 Percentage of blast cells was determined by the May-Grünwald-Giemsa stain and by immunophenotyping that was performed by flow cytometry. In vitro drug resistance was defined as the LC50 more than the plasma peak concentration achieved in pharmacologic studies (etoposide 60 μmol/L, DNR 0.85 μmol/L, Ara-C 4 μmol/L).37-39 

Statistical analysis.

Clinical and biological factors were investigated for their influence on remission rate by the χ2 or Fisher’s exact tests for binary variables and by the Mann Whitney U test for continuous variables. Correlations among levels of expression of continuous variables were estimated using the Spearman rank coefficient. The rate of (1) relapse-free survival (RFS) was measured from establishment of complete remission (CR) until relapse or death from any cause, with observation censored for patients last known alive without report of relapse; and (2) overall survival (OS) was measured from diagnosis until death from any cause, with observation censored for patients last known alive. RFS and OS were estimated by the method of Kaplan and Meier40 and compared by the log-rank test and the Breslow-Gehan-Wilcoxon test. However, data were largely reported and analyzed as continuous variables, as in the consensus recommendations.28-30 Analyses of prognostic factors for treatment outcomes were based on proportional hazards regression models for RFS and OS.41 Significance was defined as a two-tailedP value of ≤.05.

Expression and function of MDR variables.

We have performed both mRNA detection by RT-PCR and protein detection by flow cytometry in 70 of 80 patients. The correlation between RT-PCR and flow cytometry was good for MRP1 gene expression (r= .87, P < .0001) (Fig 2A). All the negative samples in RT-PCR (a sensitive technique) had a fluorescence ratio of MRP1 protein expression ≤1.4 and all samples with a fluorescence ratio >1.4 expressed MRP1 mRNA (Fig 2A). Therefore, we have used this threshold of positivity (1.4) for MRP1 protein expression. With this cut-off, 34% of patients expressed MRP1 protein. There was also a strong correlation between MRP1 expression by RT-PCR or flow cytometry and MRP1 activity (Fig 2B, r = .81,P < .0001; Fig 2C, r = .81, P < .0001, respectively). In some cases, the ratios of fluorescence in the MRP1 protein activity assay ranged from 0.72 to 1. This was probably caused by variations in cellular uptake of calcein-AM in the experiments. Conversely, fluorescence ratios of up to 1.28 can also represent some experimental variability. Therefore, we have used this threshold of positivity (1.28) for MRP1 protein activity (Fig 2B and C). With this cut-point, 27% of patients expressed a functional MRP1 protein.

Fig. 2.

(A) Correlation between MRP1 mRNA expression by RT-PCR and MRP1 protein expression by flow cytometry. All patients negative by RT-PCR (a very sensitive technique) expressed a fluorescence ratio from 0.81 to 1.4 in the MRP1 protein detection assay. Therefore, the threshold of positivity of 1.4 (horizontal dotted line) was used (with this cut-off of fluorescence ratio, 34% of patients expressed MRP1 protein). (B) Correlation between the effect of probenecid on calcein-AM uptake and MRP1 mRNA expression by RT-PCR in 40 patients. Three patients positive by RT-PCR assay were negative in the MRP1 activity assay. (C) Correlation between the effect of probenecid on calcein-AM uptake and MRP1 protein expression by flow cytometry in 40 patients. One patient positive by flow cytometry assay was negative in the MRP1 activity assay. In some cases, the ratios of fluorescence in MRP1 protein activity assay ranged from 0.72 to 1 (B and C). This was probably caused by variations in cellular uptake of calcein-AM in the experiments. Conversely, the fluorescence ratios of up to 1.28 can also represent some experimental variability (area between the two vertical dotted lines) (B and C). With these thresholds of positivity, there was 7.5% discordance between RT-PCR and functional assays (3 of 40 samples were MRP1+/activity−) and 2.5% between protein detection and functional assays (1 of 40 samples was MRP1+/activity−).

Fig. 2.

(A) Correlation between MRP1 mRNA expression by RT-PCR and MRP1 protein expression by flow cytometry. All patients negative by RT-PCR (a very sensitive technique) expressed a fluorescence ratio from 0.81 to 1.4 in the MRP1 protein detection assay. Therefore, the threshold of positivity of 1.4 (horizontal dotted line) was used (with this cut-off of fluorescence ratio, 34% of patients expressed MRP1 protein). (B) Correlation between the effect of probenecid on calcein-AM uptake and MRP1 mRNA expression by RT-PCR in 40 patients. Three patients positive by RT-PCR assay were negative in the MRP1 activity assay. (C) Correlation between the effect of probenecid on calcein-AM uptake and MRP1 protein expression by flow cytometry in 40 patients. One patient positive by flow cytometry assay was negative in the MRP1 activity assay. In some cases, the ratios of fluorescence in MRP1 protein activity assay ranged from 0.72 to 1 (B and C). This was probably caused by variations in cellular uptake of calcein-AM in the experiments. Conversely, the fluorescence ratios of up to 1.28 can also represent some experimental variability (area between the two vertical dotted lines) (B and C). With these thresholds of positivity, there was 7.5% discordance between RT-PCR and functional assays (3 of 40 samples were MRP1+/activity−) and 2.5% between protein detection and functional assays (1 of 40 samples was MRP1+/activity−).

Close modal

The correlation between RT-PCR and flow cytometry was also strong forMDR1 gene expression (r = .95, P < .0001) (Fig 3A). All the negative samples in RT-PCR had a fluorescence ratio of Pgp expression ≤1.3 and all samples with a fluorescence ratio >1.3 expressed MDR1 mRNA (Fig 3A). Therefore, we have used this threshold of positivity (1.3) for Pgp expression. With this cut-point, 51% of patients expressed MDR1. There was also a correlation between MDR1 mRNA expression and Pgp function (Fig 3B, r = .63, P < .0001) and between Pgp expression and Pgp function (Fig 3C, r = .66, P < .0001). In some cases, the ratios of fluorescence in Pgp activity assay ranged from 0.61 to 1. This was probably because of variations in cellular uptake of calcein-AM in the experiments. Conversely, the fluorescence ratios of up to 1.39 can also represent some experimental variability. Therefore, we have used this threshold of positivity (1.39) for Pgp activity (Fig 3B and C). With this cut-point, 35% of patients expressed a functional Pgp.

Fig. 3.

(A) Correlation between MDR1 mRNA expression by RT-PCR and Pgp expression by flow cytometry (r = .95, P < .0001). All patients negative by RT-PCR expressed a fluorescence ratio from 0.7 to 1.3 in the Pgp detection assay. Therefore, the threshold of positivity of 1.3 (horizontal dotted line) was used (with this cut-off of fluorescence ratio, 51% of patients expressed Pgp). (B) Correlation between the effect of CsA on calcein-AM uptake and MDR1 mRNA expression by RT-PCR in 40 patients (r = .63, P < .0001). (C) Correlation between the effect of CsA on calcein-AM uptake and Pgp expression by flow cytometry in 40 patients (r = .66,P < .0001). In some cases, the ratios of fluorescence in Pgp activity assay ranged from 0.61 to 1. This was probably caused by variations in cellular uptake of calcein-AM in the experiments. Conversely, the fluorescence ratios of up to 1.39 can also represent some experimental variability (area between the two vertical bold lines) (B and C). With these thresholds of positivity, 14 patients (35%) were Pgp+/activity+, 12 patients (30%) were Pgp−/activity−. However, as previously described,1,6,11discrepant cases were identified, including 13 samples (32.5%) Pgp+/activity− and 1 sample (2.5%) Pgp−/activity+.

Fig. 3.

(A) Correlation between MDR1 mRNA expression by RT-PCR and Pgp expression by flow cytometry (r = .95, P < .0001). All patients negative by RT-PCR expressed a fluorescence ratio from 0.7 to 1.3 in the Pgp detection assay. Therefore, the threshold of positivity of 1.3 (horizontal dotted line) was used (with this cut-off of fluorescence ratio, 51% of patients expressed Pgp). (B) Correlation between the effect of CsA on calcein-AM uptake and MDR1 mRNA expression by RT-PCR in 40 patients (r = .63, P < .0001). (C) Correlation between the effect of CsA on calcein-AM uptake and Pgp expression by flow cytometry in 40 patients (r = .66,P < .0001). In some cases, the ratios of fluorescence in Pgp activity assay ranged from 0.61 to 1. This was probably caused by variations in cellular uptake of calcein-AM in the experiments. Conversely, the fluorescence ratios of up to 1.39 can also represent some experimental variability (area between the two vertical bold lines) (B and C). With these thresholds of positivity, 14 patients (35%) were Pgp+/activity+, 12 patients (30%) were Pgp−/activity−. However, as previously described,1,6,11discrepant cases were identified, including 13 samples (32.5%) Pgp+/activity− and 1 sample (2.5%) Pgp−/activity+.

Close modal

As previously reported,10 there was a weak correlation between Mvp/LRP and MRP1 protein expression (r = .29, P= .04), but no correlation between Mvp/LRP and Pgp (r = .03,P = .84) and between MRP1 and Pgp expression (r = .25, P = .10) (data not shown). In contrast, there was a weak correlation between MRP1 and Pgp activity (r = .39,P = .008) (Fig 4).

Fig. 4.

Graphic representation of samples (A) with simultaneous activity of both Pgp and MRP1 (6 patients; 15%); (B) with MRP1 activity, without activity of Pgp (5 patients; 12.5%); (C) with Pgp activity, without activity of MRP1 (7 patients; 17.5%); and (D) without activity of both Pgp and MRP1 (22 patients; 55%). The area included between the two horizontal bold lines represents the samples with a negative activity of Pgp (fluorescence ratios of samples which ranged between 0.61 and 1.39 represent experimental variability). The area included between the two vertical dotted lines represented the samples without activity of MRP1 (fluorescence ratios of samples which ranged between 0.72 and 1.28 represent experimental variability). There was a weak correlation (r = .39, P = .008) between MRP1 and Pgp activity. *Two samples.

Fig. 4.

Graphic representation of samples (A) with simultaneous activity of both Pgp and MRP1 (6 patients; 15%); (B) with MRP1 activity, without activity of Pgp (5 patients; 12.5%); (C) with Pgp activity, without activity of MRP1 (7 patients; 17.5%); and (D) without activity of both Pgp and MRP1 (22 patients; 55%). The area included between the two horizontal bold lines represents the samples with a negative activity of Pgp (fluorescence ratios of samples which ranged between 0.61 and 1.39 represent experimental variability). The area included between the two vertical dotted lines represented the samples without activity of MRP1 (fluorescence ratios of samples which ranged between 0.72 and 1.28 represent experimental variability). There was a weak correlation (r = .39, P = .008) between MRP1 and Pgp activity. *Two samples.

Close modal

We also analyzed 40 patients for combined Pgp and MRP1 activity (Fig4). In these patients, we found 6 (15%) samples with simultaneous activity of Pgp and MRP1, 22 (55%) samples without Pgp and MRP1 activity, 13 samples (32%) with Pgp activity, and 11 samples (27%) with MRP1 activity. Therefore, 18 (45%) samples had functional activity of one or both proteins (Fig 4).

The percentage of patients who expressed Pgp or MRP1 (protein or function) is shown in Table 1.

Table 1.

Summary of Expression and Activity of Pgp and/or MRP1 in Adult AML

% of Positive Patients
Pgp expression  51  
MRP1 expression  34  
Pgp function  35  
MRP1 function  27 
Functional activity of Pgp and MRP1 together  15  
Functional activity of one or both proteins  45-52* 
% of Positive Patients
Pgp expression  51  
MRP1 expression  34  
Pgp function  35  
MRP1 function  27 
Functional activity of Pgp and MRP1 together  15  
Functional activity of one or both proteins  45-52* 
*

According to the functional assay (see Figs 4 and 5).

MDR parameters and other in vitro resistance variables.

No statistically significant correlation was found between the level of Pgp expression and the LC50 of DNR (r = .29, P = .10), etoposide (r = .09, P = .62), and Ara-C (r = .12, P = .37) and between the level of MRP1 expression and the LC50 of DNR (r = .33, P = .07), etoposide (r = .23, P = .12) and Ara-C (r = .14, P = .50) (data not shown). There was also no correlation between the level of Mvp/LRP expression and the LC50 of DNR, etoposide, and Ara-C (data not shown).

Similarly, we found no correlation or only a weak correlation between the LC50 and both the effect of CsA on the level of calcein-AM uptake, which quantified only Pgp activity (r = .36, P = .02 for DNR; r = .03, P = .85 for etoposide; and r= .25, P = .16 for Ara-C) (Fig 5A and B) and the effect of probenecid on the level of calcein-AM uptake, which quantified only MRP1 activity (r = .47, P = .002 for DNR; r = .24, P= .14 for etoposide and r = −.08, P = .61 for Ara-C) (Fig 5C and D). In contrast, there was a strong correlation between the LC50 of DNR and the combined effect of probenecid and CsA (which quantified the simultaneous activity of Pgp and MRP1) on the level of calcein-AM uptake (r = .77, P < .0001) (Fig 5E), a weak correlation between the LC50 of etoposide and the combined effect of probenecid and CsA on the level of calcein-AM uptake (r = .54, P = .007) (Fig 5F) and no correlation between the LC50 of Ara-C and the combined effect of probenecid and CsA on the level of calcein-AM uptake (r = .27, P = .20) (data not shown).

Fig. 5.

Correlations between Pgp activity (measured by the effect of CsA on calcein-AM uptake) (y-axis, A and B) and the LC50 of DNR (x-axis, A) and the LC50 of etoposide (x-axis, B); between MRP1 activity (which was measured by the effect of probenecid on calcein-AM uptake) (y-axis, C and D) and the LC50 of DNR (x-axis, C) and the LC50 of etoposide (x-axis, D); and between the simultaneous activity of MRP1 and Pgp (which was quantified by the combined effect of probenecid and CsA on calcein-AM uptake) (y-axis, E and F) and the LC50 of DNR (x-axis, E) and the LC50 of etoposide (x-axis, F). When the combined effect of CsA/probenecid on calcein-AM uptake is analyzed, some cases had a fluorescence ratio which ranged from 0.59 to 1 (E and F). As for protein detection, this was probably caused by variations in cellular uptake of calcein-AM in the experiments. A similar variation in the opposite direction (1.41) can also represent some experimental variability. Therefore, we have used this threshold of positivity (1.41) for the simultaneous activity of Pgp and MRP1 (horizontal dotted line). With this cut-off, 52% of patients expressed combined activity of both MRP1 and Pgp.

Fig. 5.

Correlations between Pgp activity (measured by the effect of CsA on calcein-AM uptake) (y-axis, A and B) and the LC50 of DNR (x-axis, A) and the LC50 of etoposide (x-axis, B); between MRP1 activity (which was measured by the effect of probenecid on calcein-AM uptake) (y-axis, C and D) and the LC50 of DNR (x-axis, C) and the LC50 of etoposide (x-axis, D); and between the simultaneous activity of MRP1 and Pgp (which was quantified by the combined effect of probenecid and CsA on calcein-AM uptake) (y-axis, E and F) and the LC50 of DNR (x-axis, E) and the LC50 of etoposide (x-axis, F). When the combined effect of CsA/probenecid on calcein-AM uptake is analyzed, some cases had a fluorescence ratio which ranged from 0.59 to 1 (E and F). As for protein detection, this was probably caused by variations in cellular uptake of calcein-AM in the experiments. A similar variation in the opposite direction (1.41) can also represent some experimental variability. Therefore, we have used this threshold of positivity (1.41) for the simultaneous activity of Pgp and MRP1 (horizontal dotted line). With this cut-off, 52% of patients expressed combined activity of both MRP1 and Pgp.

Close modal
MDR parameters and in vivo resistance.

Sixty untreated AML patients were evaluable for clinical response. Sixty-five percent of patients achieved CR. Variables influencing CR are shown Table 2. CR rate significantly decreased with increasing MDR1 gene expression (0.197 ± 0.022 v 0.092 ± 0.087,P = .04 by RT-PCR; 3.93 ± 2.27 v 2.10 ± 0.59,P = .04 by flow cytometry) and with increasing MRP1 gene expression (0.756 ± 0.312 v 0.212 ± 0.341,P = .05 by RT-PCR; 1.90 ± 0.43 v 1.38 ± 0.53, P = .05 by flow cytometry). However, CR rate was not associated with the level of Mvp/LRP expression by both assays (RT-PCR and flow cytometry) (Table 2). Patients who achieved CR also had a lower activity of Pgp (1.17 ± 0.27 v 1.55 ± 0.37,P = .05), a lower activity of MRP1 (1.12 ± 0.36 v1.39 ± 0.22, P = .05) and a lower simultaneous activity of MRP1 and Pgp (1.35 ± 0.47 v 2 ± 0.54, P = .008) than patients who did not (Table 2). While the CR rate significantly decreased with increasing LC50 of DNR (0.56 ± 1.32 v 0.29 ± 0.28, P = .05), it was not associated with the level of LC50 of etoposide and Ara-C. When the threshold of positivity was used for in vitro MDR variables, we obtained the same results (Table 3).

Table 2.

Parameters Influencing Achievement of CR

Parameters Patients Who Achieved CR (patients)Patients Refractory to Treatment (patients)P Value
In Vitro Resistance Parameters  
MDR1 expression  
 RT-PCR* 0.092 ± 0.087  0.197 ± 0.022  .04#  
 Flow cytometry 2.10 ± 0.59  3.93 ± 2.27  .04#  
MRP1 expression  
 RT-PCR* 0.212 ± 0.341 0.756 ± 0.312  .05#  
 Flow cytometry 1.38 ± 0.53  1.90 ± 0.43  .05#  
Mvp/LRP expression  
 RT-PCR* 0.57 ± 0.51  0.69 ± 0.66 NS  
 Flow cytometry 3.7 ± 1.7 4.4 ± 3.1  NS  
Modulatory effect of CsA on calcein-AM uptake,2-153 1.17 ± 0.27  1.55 ± 0.37  .05# 
Modulatory effect of probenecid on calcein-AM uptake,2-155 1.12 ± 0.36  1.39 ± 0.22  .04#  
Modulatory effect of CsA + probenecid on calcein-AM uptake,2-154 1.35 ± 0.47 2 ± 0.54  .008#  
LC50 DNR (μmol/L)  0.29 ± 0.28 0.56 ± 1.32  .05#  
LC50 AraC (μmol/L)  5.0 ± 8.8 17.2 ± 28.5  .06#  
LC50 etoposide (μmol/L) 14.9 ± 17.3  20.4 ± 28.5   NS# 
Clinical and Biological Parameters  
Age (yr)  48 ± 16  62 ± 22  .03#  
WBC at diagnosis (×109/L)  77 ± 72  139 ± 111 .03#  
CD34 (% of positive patients)  54 64   NS2-160 
Cytogenetic (%)    .052-164 
 Good  88  12 
 Intermediate  58  42 
 Poor  40  60 
Parameters Patients Who Achieved CR (patients)Patients Refractory to Treatment (patients)P Value
In Vitro Resistance Parameters  
MDR1 expression  
 RT-PCR* 0.092 ± 0.087  0.197 ± 0.022  .04#  
 Flow cytometry 2.10 ± 0.59  3.93 ± 2.27  .04#  
MRP1 expression  
 RT-PCR* 0.212 ± 0.341 0.756 ± 0.312  .05#  
 Flow cytometry 1.38 ± 0.53  1.90 ± 0.43  .05#  
Mvp/LRP expression  
 RT-PCR* 0.57 ± 0.51  0.69 ± 0.66 NS  
 Flow cytometry 3.7 ± 1.7 4.4 ± 3.1  NS  
Modulatory effect of CsA on calcein-AM uptake,2-153 1.17 ± 0.27  1.55 ± 0.37  .05# 
Modulatory effect of probenecid on calcein-AM uptake,2-155 1.12 ± 0.36  1.39 ± 0.22  .04#  
Modulatory effect of CsA + probenecid on calcein-AM uptake,2-154 1.35 ± 0.47 2 ± 0.54  .008#  
LC50 DNR (μmol/L)  0.29 ± 0.28 0.56 ± 1.32  .05#  
LC50 AraC (μmol/L)  5.0 ± 8.8 17.2 ± 28.5  .06#  
LC50 etoposide (μmol/L) 14.9 ± 17.3  20.4 ± 28.5   NS# 
Clinical and Biological Parameters  
Age (yr)  48 ± 16  62 ± 22  .03#  
WBC at diagnosis (×109/L)  77 ± 72  139 ± 111 .03#  
CD34 (% of positive patients)  54 64   NS2-160 
Cytogenetic (%)    .052-164 
 Good  88  12 
 Intermediate  58  42 
 Poor  40  60 

Abbreviation: NS, not significant.

*

Variations between samples in cDNA synthesis were normalized by their relative quantities of β2m amplified by 23-cycle PCR. The normalized yield of MDR products relative to β2m were then compared with those of A549 cells for MRP1 and LRP and with those of HL60 Pgp for MDR1, which were defined as 1 arbitrary unit.

Values were expressed as adjusted for control, ie, the ratio of MoAbs fluorescence divided by control antibody fluorescence.

Data were calculated as the ratio of drug fluorescence with modulator divided by drug fluorescence without modulator after subtraction of the fluorescence of the control.

F2-153

Quantified Pgp activity.

F2-155

Quantified MRP1 activity.

F2-154

Quantified the simultaneous activity of Pgp and MRP1.

#Using U Mann Whitney test.

F2-160

Using χ2 test.

F2-164

Using Kruskal Wallis test.

Table 3.

In Vitro MDR Variables Influencing Achievement of CR (using the threshold of positivity)

Variables3-150 (thresholds of positivity)3-151Response to Treatment P Value3-152
No. % CR
Pgp expression (1.3)  
 Positive 31  51  .02  
 Negative  29  79  
MRP1 expression (1.4)  
 Positive  20  45  .02  
 Negative  40 75  
Modulatory effect of CsA on calcein-AM uptake (1.39) 
 Positive  14  42  .03  
 Negative  26  76 
Modulatory effect of probenecid on calcein-AM uptake (1.28) 
 Positive  11  36  .02  
 Negative  29  75 
Modulatory effect of CsA + probenecid on calcein-AM uptake (1.41)  
 Positive  18  33  .0001  
 Negative 22  90 
Variables3-150 (thresholds of positivity)3-151Response to Treatment P Value3-152
No. % CR
Pgp expression (1.3)  
 Positive 31  51  .02  
 Negative  29  79  
MRP1 expression (1.4)  
 Positive  20  45  .02  
 Negative  40 75  
Modulatory effect of CsA on calcein-AM uptake (1.39) 
 Positive  14  42  .03  
 Negative  26  76 
Modulatory effect of probenecid on calcein-AM uptake (1.28) 
 Positive  11  36  .02  
 Negative  29  75 
Modulatory effect of CsA + probenecid on calcein-AM uptake (1.41)  
 Positive  18  33  .0001  
 Negative 22  90 
F3-150

None of the other in vitro resistance parameters, Mvp/LRP expression, LC50 DNR, LC50 etoposide, or LC50 Ara-C, had prognostic significance.

F3-151

The thresholds of positivity were defined in Figs 2, 3, and 5.

F3-152

Using χ2 test.

RFS and OS decreased significantly with increasing LC50 of Ara-C (P = .02 and P = .001, respectively), and with increasing simultaneous activity of Pgp and MRP1 (P = .01 andP = .02, respectively) (Table 4). However, the expression (by both RT-PCR and flow cytometry) and the activity of Pgp and MRP1 separately did not influence RFS or OS (data not shown).

Table 4.

Significant Prognostic Variables for RFS and OS

Variables4-150RFS (P value) OS (Pvalue)
In Vitro Resistance Parameters  
Effect of both probenecid and CsA on calcein-AM uptake  .01  .02  
LC50 Ara-C  .02 .001  
Clinical and Biological Parameters  
Age  .03  .01  
Cytogenetics  .04 .01  
WBC  .03  .04 
Variables4-150RFS (P value) OS (Pvalue)
In Vitro Resistance Parameters  
Effect of both probenecid and CsA on calcein-AM uptake  .01  .02  
LC50 Ara-C  .02 .001  
Clinical and Biological Parameters  
Age  .03  .01  
Cytogenetics  .04 .01  
WBC  .03  .04 
F4-150

Only variables with P values ≤.05 on log regression were recorded in the table. The other variables with a P value >.05 were MDR1, MRP1, and Mvp/LRP expression (by RT-PCR and flow cytometry); Pgp and MRP1 activity (the effect of either CsA or probenecid on calcein-AM uptake, respectively); LC50 DNR and LC50 etoposide; and CD34 expression.

Interestingly, while the LC50 of DNR and LC50 of etoposide were not separately associated with RFS and OS, patients with both high LC50 DNR (≥0.85 μmol/L) and high LC50 etoposide (≥60 μmol/L) had higher risks of relapse or death than other patients (Fig 6a and c). Similarly, patients with high combined activities of Pgp and MRP1 also had a higher risk of relapse or death than other patients (Fig 6b and d).

Fig. 6.

Relation between the in vitro drug resistance to DNR and etoposide together (a and b) or the simultaneous effect of both CsA and probenecid on calcein-AM uptake (b and d) and the probability (which analyzed the combined activity of Pgp and MRP1) of RFS and OS in untreated de novo AML patients. †Compared by the log-rank test; ††compared by the BreslowGehan-Wilcoxon test.

Fig. 6.

Relation between the in vitro drug resistance to DNR and etoposide together (a and b) or the simultaneous effect of both CsA and probenecid on calcein-AM uptake (b and d) and the probability (which analyzed the combined activity of Pgp and MRP1) of RFS and OS in untreated de novo AML patients. †Compared by the log-rank test; ††compared by the BreslowGehan-Wilcoxon test.

Close modal
Other prognostic factors and correlation with in vitro resistance variables.

The effect of other well-known variables such as age, cytogenetics, WBC count at diagnosis, and CD34 expression on clinical response were also analyzed. RFS and OS were significantly poorer for patients with unfavorable cytogenetics (P = .04 and P = .01, respectively) and decreased significantly with increasing age (P = .03 and P = .01, respectively) and increasing WBC (P = .03 and P = .04, respectively) (Table 4). CD34 expression was not a prognostic factor for RFS and OS.

Table 5 shows significant associations between older age and high mRNA MDR1 expression (P= .01), high Pgp expression (P = .01), high effect of CsA on calcein-AM uptake (P = .009), high level of LC50 of DNR (P = .05), and high level of LC50 of Ara-C (P = .04). Unfavorable cytogenetic effect was correlated with a high effect of both probenecid and CsA on calcein-AM uptake (P = .002), a high level of LC50 of DNR (P = .03), and a high level of LC50 of Ara-C (P = .02). High WBC count at diagnosis was associated with a high level of Mvp/LRP (P = .02).

Table 5.

Significant Associations Between Well-Known Clinical and Biological Variables on Clinical Response in AML and In Vitro Resistance Variables

Characteristics (clinical and biological variables)Associated With5-150P Value5-151
Older age High MDR1 expression (RT-PCR)  .01  
 High Pgp expression (flow cytometry)  .01  
 High effect of CsA on calcein-AM uptake  .009  
 High LC50 of Ara-C  .04 
 High LC50 of DNR  .05  
High WBC at diagnosis  High Mvp/LRP expression (flow cytometry)  .02  
Unfavorable cytogenetics  Effect of both CsA and probenecid on calcein-AM uptake .002  
 High LC50 of Ara-C  .02  
 High LC50 of DNR .03 
Characteristics (clinical and biological variables)Associated With5-150P Value5-151
Older age High MDR1 expression (RT-PCR)  .01  
 High Pgp expression (flow cytometry)  .01  
 High effect of CsA on calcein-AM uptake  .009  
 High LC50 of Ara-C  .04 
 High LC50 of DNR  .05  
High WBC at diagnosis  High Mvp/LRP expression (flow cytometry)  .02  
Unfavorable cytogenetics  Effect of both CsA and probenecid on calcein-AM uptake .002  
 High LC50 of Ara-C  .02  
 High LC50 of DNR .03 
F5-150

The variables analyzed were: MDR1, MRP1, andMvp/LRP gene expression (measured by RT-PCR and flow cytometry), Pgp function (measured by the effect of CsA on calcein-AM uptake), MRP1 function (measured by the effect of probenecid on calcein-AM uptake), the simultaneous function of both Pgp and MRP1 (measured by the combined effect of CsA and probenecid on calcein-AM uptake), and the in vitro resistance to DNR, etoposide, and Ara-C (measured by the MTT assay).

F5-151

Only associations with P value ≤.05 on log regression were recorded in the table.

While both MRP1 and Pgp may confer resistance to different families of drugs in AML, including anthracyclines and etoposide, the relative importance of these two genes is not known. Several studies reported the sequential expression of MRP1 and Pgp in drug-selected cell lines.20-22 At clinically relevant concentrations of doxorubicin or homoharringtonine, resistance to these drugs was related to MRP1 overexpression, but not to MDR1 expression in human myeloid leukemia cell lines.20,22 Only when cell lines were exposed to drugs for a prolonged time period or selected for relatively high-level drug resistance did Pgp/MDR1 overexpression become apparent. Similar findings occur in other cell lines (murine leukemia, small cell lung cancer cells).21,42,43 In light of these results,MRP1 gene overexpression is probably an early event in the development of drug resistance, and MRP1 and Pgp could be coexpressed in AML cells exposed to pharmacological doses of cytotoxic drugs. In clinical samples of AML, MRP1 overexpression ranged from 7% to 30%.4,7,8,44-49 MRP1 overexpression was more frequent in drug-refractory or relapsed patients than in drug-sensitive patients in one study,4 but not in others.46,47 Similarly, the coexpression and correlation between MRP1 and MDR1 are under debate. These contradictory results might be partially caused by differences in the composition of samples and experimental methods, as well as differences in the definition of overexpression. To date, few data on the coexpression of these two genes in AML cells have been reported, and the combined functionality of these two proteins in clinical samples has not been studied. We have shown, in previous studies, that calcein-AM uptake can be used to assess whether MRP1 and/or Pgp are functional and to assess the simultaneous activity of MRP1 and Pgp in fresh leukemic cells.10,12 In our present study, 45% of the AML samples studied exhibited a functional activity of one or both proteins. Taken together, these previous and present reports suggest that MRP1 and Pgp need to be considered together and that clinical trials that selectively modulate Pgp are likely to achieve limited success. Therefore, we analyzed the contribution of the combined activity of Pgp and MRP1 to in vitro and in vivo resistance to chemotherapy in AML patients.

In our study, the absence of or only weak correlations between MRP1 or Pgp expression or function (tested separately) and in vitro drug resistance to DNR and etoposide were in agreement with other data17-19 and could be partly explained by the separate analysis for the two resistance genes. In agreement with this, we have found a good correlation between the simultaneous activity of MRP1 and Pgp and in vitro resistance to DNR, which emphasizing the role of these two proteins together in the resistance to DNR in adult AML. In addition, probenecid, the modulator of MRP1 used in this study, has been associated with an increased accumulation of DNR and with the correction of the altered distribution of DNR in leukemic cell lines.33 

Similarly, we have shown that the combined activity of MRP1 and Pgp was a prognostic factor for treatment outcome (achievement of CR, and duration of RFS and OS), but not MRP1 or Pgp separately (for RFS and OS). In several other studies, the prognostic value of MRP1 expression is discussed.4,7,8,44-49 However, in these studies only one technique was used and functionality was not assessed. As for MDR1,15,28-30 an elaboration of consensus recommendations would be required.

In contrast, preliminary studies using the MTT assay for the prediction of chemoresistance in adult AML suggest that it may be helpful for risk-group stratification in adult AML.50,51 In addition, this test has a strong value in the prediction of clinical response in childhood leukemias.52,53 It was also shown that in vitro drug resistance determined with the differential staining cytotoxicity (DiSC) assay, based on the same concept as the MTT assay, was related to survival in adult AML.54 Therefore, we used the MTT assay to assess the in vitro resistance to drugs. In our study, patients who exhibited both high LC50 of DNR and high LC50 of etoposide, but not etoposide alone, had a poorer prognosis than other patients, underlying the importance of these two drugs. In clinical trials, it was unclear whether the addition of etoposide improved treatment outcome in adult AML.55-57 But in Bishop’s randomized study, which included 264 patients, there was an additional benefit from the use of etoposide specifically confined to patients ages less than 55 years.58 There has not been another large randomized trial comparing anthracycline + Ara-C ± etoposide as induction chemotherapy. Nevertheless, our finding is in accordance with the fact that etoposide could be effective in the treament of AML. We also showed that the combined activity of MRP1 and Pgp in resistance to etoposide appears less important than their role in resistance to DNR.

We have shown that the well-known prognostic factors in AML,59 age and cytogenetics, were associated with both Pgp and MRP1 expression and function. Age has already been correlated with Pgp expression and function.6,7 In our study, the unfavorable cytogenetic category was better associated with the combined activity of both MRP1 and Pgp than with the activity of Pgp or MRP1 separately. This could explain the poor prognosis and the in vitro resistance to daunorubicin in this group of patients. Modulation of not only Pgp but also MRP1 could be essential, in this category of patients, to improve the results of treatment.

As in our previous report,10 we found a good correlation between mRNA expression detected by RT-PCR and protein expression detected by flow cytometry for both MDR1 and MRP1genes. As previously described, we have identified discrepant cases between Pgp expression and function.1,6,11 In contrast, for MRP1, discrepancy between protein expression and function were uncommon. 28-30 We recommend the detection of MRP1 expression by flow cytometry. In addition, a functional test using calcein-AM uptake assay could be used to assess the activity of MRP1 (we found 7% of cases with a discrepancy between MRP1 expression and function) and the simultaneous activity of both MRP1 and Pgp. Other functional tests can assess the activity of MRP1.60 A critical evaluation of these different assays would be useful.

Unlike the results obtained with MRP1 and Pgp, we found, as recently reported by both Leith et al7 and us,10 that the level of Mvp/LRP expression is not correlated with treatment outcome in adult AML, in contrast to other studies.5,9 In our study, the level of Mvp/LRP expression was not correlated with in vitro resistance to DNR or etoposide. Therefore, a causal mechanistic relationship of Mvp/LRP with drug resistance is still lacking. But, as we recently reported, the discrepancy in the clinical significance of Mvp/LRP expression may be related to the methodology used.61 

In conclusion, all the data presented here support the hypothesis that the modulation of both Pgp and MRP1, by agents such as probenecid and PSC833, may simultaneously increase the percentage of CR, the percentage of RFS, and survival duration in adult AML patients by increasing blast cell DNR ± etoposide cytotoxicity. The concentration of probenecid that reverses MRP1 function is clinically achievable in vivo.33 However, because of the relatively small patient numbers in our study, a multicenter study will be required to evaluate all of these resistance parameters.

Supported in part by a grant from ARC (Grant No. 9637).

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
Marie
JP
Zhou
DC
Gurbuxani
S
Legrand
O
Zittoun
R
MDR1/P-glycoprotein in haematological neoplasms.
Eur J Cancer
32A
1996
1034
2
Cole
SPC
Bhardwaj
G
Gerlach
JH
Mackie
JE
Grant
CE
Almquist
KC
Kurz
EU
Duncan
AM
Deeley
RG
Overexpression of a transporter gene in a multidrug-resistant human lung cancer cell line.
Science
258
1992
1650
3
Scheper
RJ
Broxterman
HJ
Scheffer
GL
Kaaijk
P
Dalton
WS
van Heijningen
THM
van Kalken
GK
Slovak
ML
de Vries
EGE
van der Valk
P
Meijer
CJLM
Pinedo
HM
Over-expression of a Mr 110,000 vesicular protein in non-P-glycoprotein-mediated multidrug resistance.
Cancer Res
53
1993
1475
4
Zhou
DC
Zittoun
R
Marie
J-P
Expression of multidrug resistance-associated protein (MRP) and multidrug resistance (MDR1) genes in acute myeloid leukemia.
Leukemia
9
1995
1661
5
List
AF
Spier
CS
Grogan
TM
Jonhson
C
Roe
DJ
Greer
JP
Wolff
SN
Broxterman
HJ
Scheffer
GL
Scheper
RJ
Dalton
WS
Overexpression of the major vault transporter protein lung-resistance protein predicts treatment outcome in acute myeloid leukemia.
Blood
87
1996
2464
6
Leith
CP
Kopecky
KJ
Godwin
J
McConnell
T
Slovak
ML
Ming-Chen
I
Head
DR
Appelbaum
FR
Willman
CL
Acute myeloid leukemia in the elderly: Assessment of multidrug resistance (MDR1) and cytogenetics distinguishes biologic subgroups with remarkably distinct responses to standard chemotherapy. A Southwest Oncology Group study.
Blood
89
1997
3323
7
Leith
CP
Kopecky
KJ
Chen
IM
Slovak
M
Head
DR
Weick
J
Appelbaum
FR
Wiilman
CL
Frequency and clinical significance of expression of the multidrug resistance proteins, MDR1, MRP and LRP in acute myeloid leukemia patients less than 65 yrs old. A Southwest Oncology Group study.
Blood
90
1997
389a
(abstr, suppl 1)
8
Filipits
M
Suchomel
RW
Zöchbauer
S
Brunner
R
Lechner
K
Pirker
R
Multidrug resistance-associated protein in acute myeloid leukemia: No impact on treatment outcome.
Clin Cancer Res
3
1997
1419
9
Filipits
M
Pohl
G
Stranzl
T
Suchomel
RW
Scheper
RJ
Jäger
U
Geissler
K
Lechner
K
Pirker
R
Expression of the lung resistance protein predicts poor outcome in de novo acute myeloid leukemia.
Blood
91
1998
1508
10
Legrand
O
Simonin
G
Perrot
JY
Zittoun
R
Marie
J-P
Pgp and MRP activities using calcein-AM are prognostic factors in adult acute myeloid leukemia patients.
Blood
91
1998
4480
11
Bailly
JD
Muller
C
Jaffrézou
JP
Demur
C
Gassar
G
Bordier
C
Laurent
G
Lack of correlation between expression and function of Pgp in acute myeloid leukemia cells.
Leukemia
9
1995
799
12
Legrand
O
Simonin
G
Zittoun
R
Marie
J-P
Both P-gp and MRP contribute to drug resistance in AML.
Leukemia
12
1998
1327
(letter)
13
Homolya
L
Hollo
Z
Germann
UA
Pastan
I
Gottesman
MM
Sarkadi
B
Fluorescent cellular indicators are extruded by the multidrug resistance protein.
J Biol Chem
268
1993
21493
14
Feller
N
Broxterman
HJ
Wahrer
DC
Pinedo
HM
ATP-dependent efflux of calcein by the multidrug resistance protein (MRP): No inhibition by intracellular glutathione depletion.
FEBS Lett
368
1995
385
15
Broxterman
HJ
Sonneveld
P
Feller
N
Osenkoppele
GJ
Währer
DCR
Eekman
CA
Schoester
M
Lankelma
J
Pinedo
HM
Löwenberg
B
Schuurhuis
GJ
Quality control of multidrug resistance assays in adult acute leukemia: Correlation between assays for P-glycoprotein expression and activity.
Blood
87
1996
4809
16
Barrand
MA
Bagrij
T
Neo
SY
Multidrug resistance-associated protein: A protein distinct from P-glycoprotein involved in cytotoxic drug expulsion.
Gen Pharmacol
28
1997
639
17
Chitnis
M
Hegde
U
Chavan
S
Juvekar
A
Advani
S
Expression of the multidrug transporter P-glycoprotein and in vitro chemosensitivity: Correlation with in vivo response to chemotherapy in acute myeloid leukemia.
Sel Cancer Ther
7
1991
165
18
Noogaard
JM
Bukh
A
Langkjer
ST
Clausen
N
Palshof
T
Pedersen
B
Hokland
P
MDR1 gene expression and drug resistance of AML cells.
Br J Haematol
100
1998
534
19
Efferth
T
Fabry
U
Osieka
R
Apoptosis and resistance to daunorubicin in human leukemic cells.
Leukemia
11
1997
1180
20
Slapak
CA
Mizunuma
N
Kufe
DW
Expression of the multidrug resistance associated protein and P-glycoprotein in doxorubicin-selected human myeloid leukemia cells.
Blood
84
1994
3113
21
Brock
I
Hipfner
DR
Nielsen
BS
Jensen
PB
Deeley
RG
Cole
SPC
Sehested
M
Sequential coexpression of the multidrug resistance gene MRP and mdr1 and their products in VP-16 (etoposide)-selected H69 small cell lung cancer cells.
Cancer Res
55
1995
459
22
Zhou
DC
Ramond
S
Viguié
F
Faussat
AM
Zittoun
R
Marie
J-P
Progressive resistance to homoharringtonine in human myeloleukemia K562 cells: Relationship to sequential emergence of MRP and MDR1 gene overexpression and MDR1 gene translation.
Int J Cancer
65
1996
365
23
van der Kolk
DM
de Vries
EG
Koning
JA
van den Berg
E
Muller
M
Vellenga
E
Activity and expression of the multidrug resistance proteins MRP1 and MRP2 in acute myeloid leukemia cells, tumor cell lines, and normal hematopoietic CD34+ peripheral cells.
Clin Cancer Res
4
1998
1727
24
Bennett
JM
Catovsky
D
Daniel
MT
Flandrin
G
Galton
DAG
Gralnick
NR
Sultan
C
The morphological classification of acute lymphoblastic leukaemia: Concordance among observers and clinical correlations.
Br J Haematol
47
1981
553
25
Bennett
JM
Catovsky
D
Daniel
MT
Flandrin
G
Galton
DAG
Gralnick
NR
Sultan
C
Proposed revised criteria for the classification of acute myeloid leukemia.
Ann Intern Med
103
1985
620
26
Arthur
DC
Berger
R
Golomb
HM
Swanbury
GJ
Reeves
BR
Alimena
G
Van Den Berghe
H
Bloomfield
CD
De la Chapelle
A
Dewald
GW
Garson
OM
Hagemeijer
A
Kaneko
Y
Mitelman
F
Pierre
RV
Ruutu
T
Sakurai
M
Lawler
SD
Rowley
JD
The clinical significance of karyotype in acute myelogenous leukemia.
Cancer Genet Cytogenet
40
1989
203
27
Legrand
O
Perrot
J-Y
Tang
RP
Simonin
G
Gurbuxani
S
Zittoun
R
Marie
J-P
Expression of the multidrug resistance-associated protein (MRP) mRNA and protein in normal peripheral blood and bone marrow haematopoietic cells.
Br J Haematol
94
1996
23
28
Beck
WT
Grogan
TM
Willman
CL
Cordon-Cardo
C
Parham
DM
Kuttesch
JF
Andreeff
M
Bates
SE
Berard
CW
Boyett
JM
Brophy
NA
Broxterman
HJ
Chan
HSL
Dalton
WS
Dielt
M
Fojo
AT
Gascoyne
RD
Head
D
Houghton
PJ
Kumar Svrivastava
D
Lehnert
M
Leith
CP
Paietta
E
Pavelic
ZP
Rimsza
L
Roninson
IB
Sikic
BI
Twentyman
PR
Warnke
R
Weinstein
R
Methods to detect P-glycoprotein-associated multidrug resistance in patient tumors: Consensus recommendations.
Cancer Res
56
1996
3010
29
Marie
J-P
Huet
S
Faussat
A-M
Perrot
J-Y
Chevillard
S
Barbu
V
Bayle
C
Boutonnat
J
Calvo
F
Campos-Guyotat
L
Colosetti
P
Cazin
J-L
De Cremoux
P
Delvincourt
C
Demur
C
Drenou
B
Fenneteau
O
Feuillard
J
Garnier-Suillerot
A
Genne
P
Gorisse
M-C
Gosselin
P
Jouault
H
Lacave
R
Le Calvez
G
Léglise
M-C
Léonce
S
Manfait
M
Maynadié
M
Merle-Béral
H
Merlin
J-L
Mousseau
M
Morjani
H
Picard
F
Pinguet
F
Poncelet
P
Racadot
E
Raphael
M
Richard
B
Rossi
J-F
Schlegel
N
Vielh
P
Zhou
DC
Robert
J
French Network of the Drug Resistance Intergroup, and Drug Resistance Network of ‘Assistance Publique-Hôpitaux de Paris’: Multicentric evaluation of the MDR phenotype in leukemia.
Leukemia
11
1997
1086
30
Marie
J-P
Legrand
O
Perrot
J-Y
Chevillard
S
Huet
S
Robert
J
Measuring multidrug resistance expression in human malignancies: Elaboration of concensus recommendations.
Semin Hematol
34
1997
63
31
Hollo
Z
Homolya
L
Hegedus
T
Sarkadi
B
Transport properties of the multidrug resistance-associated protein (MRP) in human tumour cells.
FEBS Lett
383
1996
99
32
Hollo
Z
Homolya
L
Davis
CW
Sarkadi
B
Calcein accumulation as a fluorometric functional assay of the multidrug transporter.
Biochim Biophys Acta
1191
1994
384
33
Gollapudi
S
Kim
CH
Tran
BN
Sangha
S
Gupta
S
Probenecid reverses multidrug resistance in resistance-associated protein-overexpressing HL60/AR and H69/AR cells but not in P-glycoprotein-overexpressing HL60/Tax and P388/ADR cells.
Cancer Chemother Pharmacol
40
1997
150
34
Maslak
P
Hegewisch-Becker
S
Godfrey
L
Andreeff
M
Flow cytometric determination of the multidrug-resistant phenotype in acute leukemia.
Cytometry
17
1994
84
35
Plumb
JA
Milroy
R
Kaye
SB
Effect of the pH dependence of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide-formazan absorption on chemosensitivity determined by a novel tetrazolium-based assay.
Cancer Res
49
1989
4435
36
Kaspers
GJL
Veerman
AJP
Pierters
R
Broekema
GH
Huismans
DR
Kasimier
KM
Loonen
AH
Van Zantwijk
CH
Hälen
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
37
Albert
DS
Bachur
NR
Holtzman
JL
The pharmacokinetics of daunomycin in man.
Clin Pharmacol Ther
12
1971
96
38
Farina
P
Marzillo
G
D’Incalci
M
High-performance liquid chromatography determination of 4′-demethylepipodophyllotoxin-9 (4,6-O-ethylidene β-D-glucopyranoside) (VP16-213) in human plasma.
J Chromatogr
222
1981
141
39
Damon
LE
Plunkett
W
Linker
CA
Plasma and cerebrospinal fluid pharmacokinetics of 1-β-D-arabinofuranosylcytosine and 1-β-D arabinofuranosyluracil following the repeated intravenous administration of high and intermediate dose of 1-β-D-arabinofuranosylcytosine.
Cancer Res
51
1991
4141
40
Kaplan
EI
Meier
P
Non-parametric estimation from incomplete observations.
J Am Stat Assoc
53
1976
457
41
Cox
DR
Regression models and life tables (with discussion).
J Royal Stat Soc B
34
1972
187
42
Slapak
CA
Daniel
JC
Levy
SB
Sequential emergence of distinct resistance phenotypes in murine erythroleukemia cells under adriamycin selection: Decreased anthracycline uptake precedes increased P-glycoprotein expression.
Cancer Res
50
1990
7895
43
Richon
VM
Weich
N
Leng
L
Kiyokawa
H
Ngo
L
Rifkind
RA
Marks
PA
Characteristics of erythroleukemia cells selected for vincristine resistance that have accelerated inducer-mediated differentiation.
Proc Natl Acad Sci USA
88
1991
1666
44
Hart
SM
Ganeshaguru
K
Hoffbrand
AV
Prentice
HG
Mehta
AB
Expression of the multidrug resistance-associated protein (MRP) in acute leukaemia.
Leukemia
8
1994
2163
45
Nooter
K
Westerman
AM
Flens
MJ
Zaman
GJR
Scheper
RJ
van Wingerden
KE
Burger
H
Oostrum
R
Boersma
T
Sonneveld
P
Gratama
JW
Kok
T
Eggermont
AMM
Bosman
FT
Stoter
G
Expression of the multidrug resistance associated protien (MRP) gene in human cancer.
Clin Cancer Res
1
1995
1301
46
Schneider
E
Cowan
KH
Bader
H
Toomey
S
Schwartz
GN
Karp
JE
Burke
PJ
Kaufmann
SH
Increased expression of the multidrug resistance-associated protein gene in relapsed acute leukemia.
Blood
85
1995
186
47
Ross
DD
Doyle
LA
Schiffer
CA
Lee
EJ
Grant
CE
Cole
SPC
Deeley
RG
Yang
W
Tong
Y
Expression of multidrug resistance-associated protein (MRP) mRNA in blast cells from acute myeloid leukemia patients.
Leukemia
10
1996
48
48
Burger
H
Nooter
K
Zaman
GJR
Sonneveld
P
van Wingerden
KE
Oostrum
RG
Stoter
G
Expression of the multidrug resistance-associated protein (MRP) in acute and chronic leukemias.
Leukemia
8
1994
990
49
Schuurhuis
GJ
Broxterman
HJ
Ossenkeppele
GJ
Baak
JPA
Eekman
CA
Kuiper
CM
Feller
N
van Heijningen
THM
Klumper
E
Pierters
R
Lankelma
J
Pinedo
HM
Functional multidrug resistance phenotype associated with combined overexpression of Pgp/MDR1 and MRP together with -beta-d-arabinosylcytosine sensitivity may predict clinical response in acute myeloid leukemia.
Clin Cancer Res
1
1995
81
50
Klumper
E
Ossenkoppele
GJ
Pieters
R
Huismans
DR
Loonen
AH
Rottier
A
Wertra
G
Veerman
JP
In vitro resistance to cytosine arabinoside, not to daunorubicin, is associated with the risk of relapse in de novo acute myeloid leukemia.
Br J Haematol
3
1996
903
51
Salmon
SE
Chemosensitivity testing: Another chapter.
J Natl Cancer Inst
82
1990
32
52
Pierters
R
Huismans
DR
Loonen
AH
Hälen
K
van der Does van den Berg
A
van Wering
ER
Veerman
AJP
Relation of cellular drug resistance to long-term clinical outcome in childhood acute lymphoblastic leukaemia.
Lancet
338
1990
399
53
Kaspers
GJL
Veerman
AJP
Pierters
R
Van Zantwijk
CH
Smets
LA
Van Wering
ER
Van Der Does-Van Den Berg
A
In vitro cellular drug resistance and prognosis in newly diagnosed childhood acute lymphoblastic leukemia.
Blood
90
1997
2723
54
Tidefelt
U
Sundman-Engberg
B
Rhedin
AS
Paul
C
In vitro drug testing in patients with acute leukaemia with incubation mimicking in vivo intracellular drug concentrations.
Eur J Haematol
43
1989
374
55
Vogler
WR
McCarley
DL
Stagg
M
Bartolucci
AA
Moore
J
Martelo
O
Omura
GA
A phase III trial of high-dose cytosine arabinoside with or without etoposide in relapsed and refractory acute myelogenous leukemia. A Southeastern Cancer Study Group Trial.
Leukemia
8
1994
1847
56
Hann
IM
Stevens
RF
Goldstone
AH
Rees
JKH
Wheatley
K
Gray
RG
Burnett
AK
on behalf of the Adult and Childhood Leukaemia Working Parties of the Medical Research Council
Randomized comparison of DAT versus ADE as induction chemotherapy in children and younger adult with acute myeloid leukemia. Results of the Medical Research Council’s 10th AML trial (MRC AML10).
Blood
89
1997
2311
57
Odom
LF
Gordon
EM
Acute monoblastic leukaemia in infancy and early childhood: Successful treatment with an epipodophyllotoxin.
Blood
64
1984
875
58
Bishop
JF
Lowenthal
RM
Joshua
D
Matthews
JP
Todd
D
Cobcroft
R
Whiteside
MG
Kronenberg
H
Ma
D
Dodds
A
Herrmann
R
Szer
J
Wolf
MM
Young
G
for the Australian Leukemia Study Group
Etoposide in acute nonlymphoblastic leukemia.
Blood
75
1990
27
59
Rowe
JM
Liesveld
JL
Trearment and prognostic facrtors in acute myeloid leukaemia.
Baillieres Clin Haematol
9
1996
87
60
Legrand
O
Zittoun
R
Marie
JP
Role of MRP1 in multidrug resistance in acute myeloid leukemia.
Leukemia
13
1999
578
61
Legrand
O
Simonin
G
Zittoun
R
Marie
JP
Lung resistance protein (LRP) gene expression in adult acute myeloid leukemia: A critical evaluation by three techniques.
Leukemia
12
1998
1367

Author notes

Address reprint requests to Ollivier Legrand, MD, PhD, Hôpital Hôtel Dieu, 1 place du parvis Notre Dame, Service d’hématologie, 181 Paris Cedex 04, France; e-mail:olivier.legrand@htd.ap-hop-paris.fr.

Sign in via your Institution