Purpose: Acute myelogenous leukemia (AML) is a disease originating from normal hematopoietic CD34+CD38 progenitor cells. Modulation of the multidrug ATP-binding cassette transporter ABCB1 has not resulted in improved outcome in AML, raising the question whether leukemic CD34+CD38 cells are targeted by this strategy.

Experimental Design: ABCB1-mediated transport in leukemic CD34+CD38 cells compared with their normal counterparts was assessed by quantitating the effect of specific ABCB1 modulators (verapamil and PSC-833) on mitoxantrone retention [defined as efflux index (EI), intracellular mitoxantrone fluorescence intensity in the presence/absence of inhibitor].

Results: ABCB1 was the major drug transporter in CD34+CD38 cells in normal bone marrow (n = 16), as shown by the abrogation of mitoxantrone extrusion by ABCB1 modulators (EI, 1.99 ± 0.08). Surprisingly, ABCB1-mediated drug extrusion was invariably reduced in CD34+CD38 cells in AML (n = 15; EI, 1.21 ± 0.05; P < 0.001), which resulted in increased intracellular mitoxantrone retention in these cells (mitoxantrone fluorescence intensity, 4.54 ± 0.46 versus 3.08 ± 0.23; P = 0.004). Active drug extrusion from these cells occurred in the presence of ABCB1 modulators in the majority of samples, pointing in the direction of redundant drug extrusion mechanisms. Residual normal CD34+CD38 cells could be identified by their conserved ABCB1-mediated extrusion capacity.

Conclusion: ABCB1-mediated drug extrusion is reduced in leukemic CD34+CD38 progenitor cells compared with their residual normal counterparts. Redundant drug transport mechanisms confer mitoxantrone transport from leukemic progenitors. These data argue that ABCB1 modulation is not an effective strategy to circumvent drug extrusion from primitive leukemic progenitor cells and may preferentially target residual normal progenitors in AML.

Acute myelogenous leukemia (AML) is considered a disease originating from CD34+CD38 hematopoietic stem cells (1). This has been conclusively shown in studies using the nonobese diabetic-severe combined immunodeficient mouse model showing that cells with leukemic engraftment and self-renewal potential in AML are found in the CD34+CD38 subpopulation (2). This CD34+CD38 phenotype of leukemia-initiating cells was observed regardless of the lineage markers expressed by the leukemic blasts, percentage of cells expressing the CD34 surface antigen, or the French-American-British subtype. These studies stress that leukemia-initiating transformation and progression-associated genetic events occur at the level of these primitive CD34+CD38 cells. Consequently, incomplete chemotherapeutic eradication of these cells may ultimately result in disease relapse. Therefore, elucidation of the mechanisms conferring resistance against chemotherapy to these cells is of crucial importance (3).

The ABCB1-encoded P-glycoprotein is a highly conserved membrane-bound ATP-binding cassette (ABC) transporter, which extrudes a wide variety of structurally unrelated chemotherapeutic compounds across the cell membrane (4), conferring the multidrug resistance phenotype in cancer cells. ABCB1 is highly expressed in normal hematopoietic CD34+CD38 stem cells (5, 6).

The observation that it is frequently expressed in AML and that expression is associated with adverse treatment outcome (7) has led to the introduction of ABCB1 modulators to enhance chemotoxicity of various chemotherapeutic agents in clinical trials. Phase III clinical trials using PSC-833 (valspodar), the most potent and specific inhibitor of ABCB1 function today, however, failed to reach their intended end points of improved complete remission and survival and have been complicated by increased toxicity (812). In contrast, a clinical trial using cyclosporin A, which is a broad-spectrum ABC transporter inhibitor, showed clinical benefit (13). The reasons for the lack of benefit of ABCB1 modulation in AML remain to be elucidated. In interpreting these negative results, it is important to know whether the leukemia-initiating CD34+CD38 hematopoietic subpopulation is targeted by ABCB1 modulation and whether additional transporters confer drug extrusion from these cells, which may explain the beneficial effect of broad-spectrum inhibition. The vast amount of studies on ABCB1 expression and function in AML has been done on general blast populations. The goal of the current study was to investigate ABCB1 expression and function in leukemic CD34+CD38 cells and the effect of ABCB1 modulators on drug retention in these cells compared with their normal counterparts.

Bone marrow samples. Bone marrow was obtained after informed consent from healthy bone marrow donors and patients with AML at diagnosis. Mononuclear cells were isolated by Ficoll (1.077 g/mL density centrifugation; Pharmacy Biotech, Uppsala, Sweden). Isolation, cryopreservation, and thawing procedures of cells have been described previously (14) and were identical for normal and leukemic bone marrow samples.

CD34+CD38 hematopoietic cells. CD34+CD38 cells were defined flow cytometrically as described previously (15) and are shown in Fig. 1A.

Figure 1.

ABCB1-mediated drug extrusion is reduced in CD34+CD38 progenitor cells in AML compared with their normal counterparts. A, representative examples of normal bone marrow (NBM; top) and AML (bottom). CD34+CD38 cells were defined flow cytometrically as the CD34-FITC + cells (blue) with CD38-PE expression within the first decade of fluorescence emission (red) and compared with CD34+CD38+ cells (blue gated) with exclusion of a decade between CD38 and CD38+ cells. Gray, CD34 cells. CD34+CD38 cells exhibited restricted light-scattering characteristics (SSC), confirming the lymphoid appearance of these cells (inset). The median frequency of CD34+CD38 cells was 0.1% of mononuclear cells (range, 0.1-0.3%) in normal bone marrow and 0.2% of mononuclear cells (range, 0.1-10%) in AML. No difference existed in average CD38 density between normal and leukemic CD34+CD38 cells (MFI, 0.57 ± 0.05 SD; range, 0.40-0.70; MFI, 0.53 ± 0.12 SD; range, 0.30-0.70). B, representative examples of flow cytometric assessment of ABCB1-mediated mitoxantrone transport in CD34+CD38 cells in normal bone marrow (top) and AML (bottom). Mean intracellular MFI was assessed in CD34+CD38 cells as described in Materials and Methods. Normal CD34+CD38 cells display low intracellular mitoxantrone fluorescence compared with CD34+CD38+ cells (inset), which can be increased significantly by blockage of ABCB1-mediated transport by verapamil/PSC-833 (EI, 2.29). CD34+CD38 cells in AML display increased intracellular mitoxantrone retention compared with CD34+CD38 cells from normal bone marrow due to reduced ABCB1-mediated transport as illustrated by a lack of effect of verapamil/PSC-833 on intracellular mitoxantrone fluorescence (EI, 0.98). C, representative examples of the effect of ABCB1 inhibition by verapamil on rhodamine 123 fluorescence in CD34+CD38 cells assessed by single-cell image analysis. For experimental procedure, see Materials and Methods. Encircled, cells. Normal CD34+CD38 cells (top) display low retention of rhodamine 123, which can be significantly increased when verapamil is added (EI, 3.24). ABCB1-mediated rhodamine 123 extrusion from CD34+CD38 cells is reduced in AML (bottom), illustrated by the lack of effect of ABCB1 inhibition by verapamil on intracellular rhodamine 123 fluorescence (EI, 1.00).

Figure 1.

ABCB1-mediated drug extrusion is reduced in CD34+CD38 progenitor cells in AML compared with their normal counterparts. A, representative examples of normal bone marrow (NBM; top) and AML (bottom). CD34+CD38 cells were defined flow cytometrically as the CD34-FITC + cells (blue) with CD38-PE expression within the first decade of fluorescence emission (red) and compared with CD34+CD38+ cells (blue gated) with exclusion of a decade between CD38 and CD38+ cells. Gray, CD34 cells. CD34+CD38 cells exhibited restricted light-scattering characteristics (SSC), confirming the lymphoid appearance of these cells (inset). The median frequency of CD34+CD38 cells was 0.1% of mononuclear cells (range, 0.1-0.3%) in normal bone marrow and 0.2% of mononuclear cells (range, 0.1-10%) in AML. No difference existed in average CD38 density between normal and leukemic CD34+CD38 cells (MFI, 0.57 ± 0.05 SD; range, 0.40-0.70; MFI, 0.53 ± 0.12 SD; range, 0.30-0.70). B, representative examples of flow cytometric assessment of ABCB1-mediated mitoxantrone transport in CD34+CD38 cells in normal bone marrow (top) and AML (bottom). Mean intracellular MFI was assessed in CD34+CD38 cells as described in Materials and Methods. Normal CD34+CD38 cells display low intracellular mitoxantrone fluorescence compared with CD34+CD38+ cells (inset), which can be increased significantly by blockage of ABCB1-mediated transport by verapamil/PSC-833 (EI, 2.29). CD34+CD38 cells in AML display increased intracellular mitoxantrone retention compared with CD34+CD38 cells from normal bone marrow due to reduced ABCB1-mediated transport as illustrated by a lack of effect of verapamil/PSC-833 on intracellular mitoxantrone fluorescence (EI, 0.98). C, representative examples of the effect of ABCB1 inhibition by verapamil on rhodamine 123 fluorescence in CD34+CD38 cells assessed by single-cell image analysis. For experimental procedure, see Materials and Methods. Encircled, cells. Normal CD34+CD38 cells (top) display low retention of rhodamine 123, which can be significantly increased when verapamil is added (EI, 3.24). ABCB1-mediated rhodamine 123 extrusion from CD34+CD38 cells is reduced in AML (bottom), illustrated by the lack of effect of ABCB1 inhibition by verapamil on intracellular rhodamine 123 fluorescence (EI, 1.00).

Close modal

Flow cytometric assessment of ABCB1 expression. ABCB1 expression was analyzed using the ABCB1-specific antibody MRK16, recognizing an external epitope of the protein in three-color flow cytometric assays as described previously (7). ABCB1 protein expression was quantitated as the median fluorescence channel shift (MRK16/IgG2a isotype control) in CD34+CD38, CD34+CD38+, and CD34 cell populations. Differences in fluorescence are assessed using the Kolmogorov-Smirnov statistic, denoted as D. This method accurately identifies small differences in fluorescence and is useful in detection of low-level ABCB1 expression (7). MRK16 staining intensity is categorized as follows: negative (D < 0.10), dim (0.10 < D < 0.15), moderate (0.15 < D > 0.25), and bright (D > 0.25).

Flow cytometric assessment of ABCB1-mediated mitoxantrone transport. Cells were stained with CD34-FITC and CD38-PE membrane markers, washed in HBSS 1% FCS, and preincubated with or without verapamil (20 μg/mL; Knoll AG, Ludwigshafen, Germany; ref. 16) or PSC-833 (2 μmol/L; Novartis Pharma AG, Basel, Switzerland; ref. 17) as inhibitors for ABCB1-mediated transport for 20 minutes in Iscove's modified Dulbecco's medium supplemented with 1% FCS. Verapamil at the concentration used in this study is regarded ABCB1 specific without inhibition of ABCG2 [breast cancer resistance protein (BCRP); ref. 18] and ABCC1 (multidrug resistance protein; ref. 19), although inhibition of ABCG2 by verapamil has been described at higher concentrations (20). Mitoxantrone (Novantrone;10 μmol/L; Lederle, Etten-Leur, the Netherlands) was added, and cells were incubated for 2 hours at 37°C, 5% CO2, with or without verapamil or PSC-833. Subsequently, cells were allowed an additional 1-hour efflux in drug-free medium with or without inhibitor. The combined uptake and efflux with or without inhibitor assay was chosen because it has higher sensitivity for ABC transporter function compared with assays using only uptake with or without inhibitor (15).

Cellular mitoxantrone fluorescence was measured at a single time point after the additional hour of efflux in drug-free medium on a flow cytometer (Coulter Elite, Beckman Coulter, Fullerton, CA) equipped with an argon laser. Fluorescence was assessed at an excitation wavelength of 635 nm through a 670-nm bandpass filter in a three-color protocol with CD34-FITC and CD38-PE. At least 200 CD34+CD38 cells were analyzed in each sample.

ABCB1-mediated transport was depicted as the ratio of intracellular mitoxantrone fluorescence intensity (MFI) in the presence or absence of inhibitor [defined as efflux index (EI)]. To assess interexperimental reproducibility, ABCB1-mediated transport in CD34+CD38 cells was assessed in a normal bone marrow sample in completely independent experiments done on separate days (n = 7). ABCB1-mediated EI was 2.10 ± 0.09 (mean ± SE), indicating good reproducibility of the assay. This sample was subsequently included in other experiments as a control. Preliminary experiments showed similar ABCB1-mediated efflux in cryopreserved samples when compared with fresh cells in normal bone marrow and AML, in line with a previous report addressing this issue (21).

Single-cell image analysis for assessment of ABCB1-mediated transport of rhodamine 123 in CD34+CD38 cells. ABCB1-mediated transport of rhodamine 123 was assessed in CD34+CD38 cells sorted on adhesive biolayers as comprehensively described elsewhere (22). Briefly, 500 CD34+CD38 cells were sorted in 8 μL washing buffer solution containing 0.9% NaCl supplemented with sodium diphosphate (6.6%, v/v), albumin (1%, v/v), and glucose (1%, v/v) applied on a glass coverslip coated with mussel adhesive protein (10 μL/cm2). Cells were allowed to adhere for 10 minutes in a perfusion chamber constructed to enable cell environment manipulation (incubation and washing steps) within the microscope environment. The microscope was placed in an incubator to allow all procedures to run at 37°C, which is essential for physiologic efflux studies.

After immobilization, cells were incubated with rhodamine 123 dissolved in Iscove's modified Dulbecco's medium containing 0.5% (v/v) heat-inactivated FCS (Hyclone, Logan, UT) with or without efflux modulator during 30 minutes. Cells were washed with washing buffer at 37°C, again in the presence or absence of efflux modulators, and allowed to efflux the dye for an additional 15 minutes. Fluorescence images were acquired after 15 minutes of efflux. Cells were excited with a mercury arc lamp using bandpass filter of 440 to 490 nm for rhodamine 123 or bandpass filter of 510 to 560 nm for propidium iodide. Emission was measured with bandpass filter of 515 to 565 nm and longpass filter of 610 nm. Cell viability typically exceeded 90% of cells. Dead cells were excluded from analysis.

Fluorescence intensity analysis of individual cells was done on TCL-Image 4.6 software package (TNO, Delft, the Netherlands). At least 30 cells were evaluated per sample. Fluorescence intensity of the overall cell population is expressed as MFI and depicted in arbitrary fluorescence units. ABCB1-mediated efflux was quantitated as the ratio of mean fluorescence intensity in the presence or absence of verapamil and depicted as “EI.”

Fluorescence in situ hybridization. CD34+CD38 cells were sorted and lysed in 15 μL KCl (75 μmol/L) on a glass coverslip. Cells were fixed in methanol/acetic acid and stored at 4°C until analysis. Fluorescence in situ hybridization was done using the LSI AML1/ETO dual color, dual-fusion t(8;21) probe, or LSI EGR1 (5q31) dual-color probe (Vysis, Downers Grove, IL) according to the manufacturer's instructions. Cells (100) were analyzed microscopically for t(8;21) or chromosome 5 (5q31) cytogenetic abnormalities as appropriate.

Adapted real-time quantitative reverse transcription-PCR for assessment of CBFB-MYH11 gene expression in CD34+CD38 cells. A linear real-time quantitative reverse transcription-PCR approach has been developed for quantitation of gene expression in low-frequency hematopoietic stem cells and was described comprehensively elsewhere (6). Primer and probe sequences were as follows: glyceraldehyde-3-phosphate dehydrogenase, glyceraldehyde-3-phosphate dehydrogenase control reagents (Applied Biosystems, Foster City, CA; part 402869); CBFB-MYH11 (type D) fusion gene, 5′-CATTAGCACAACAGGCCTTTGA-3′ (forward), 5′-CCTCGTTAAGCATCCCTGTGA-3′ (reverse), and TET-ATAGAGACAGGTCTCATCG (probe).

Normalized gene expression to the internal standard glyceraldehyde-3-phosphate dehydrogenase is given by the following equation: To/Ro(Xn) = K (1 + E)(CT,r − CT,t), where To is the initial number of target gene copies, Ro is the initial number of standard gene copies, E is the efficiency of amplification, CT,t is the threshold cycle of target gene, CT,r is the threshold cycle of standard gene, and K is constant.

Statistical analysis. Differences in ABCB1 protein expression (D of MRK16/isotype control) and function (EI) between different normal and leukemic cell populations were calculated using the Student's t test with a level of significance of P < 0.05.

ABCB1 is the major determinant of mitoxantrone extrusion in CD34+CD38 cells in normal bone marrow. CD34+CD38 hematopoietic cells were flow cytometrically defined as shown in Fig. 1A. ABCB1 protein expression was assessed in hematopoietic cell populations using the monoclonal antibody MRK16. ABCB1 was differentially expressed in CD34+CD38 cells compared with more differentiated cell populations (Fig. 2). A decrease in ABCB1 protein expression in CD34+CD38+ and CD34 cells was observed in all samples examined.

Figure 2.

ABCB1 is differentially expressed in CD34+CD38 cells in both normal bone marrow and AML. ABCB1 expression was assessed flow cytometrically using the MRK16 monoclonal antibody and valued using the Kolmogorov-Smirnov statistic, denoted as D, as described in the Materials and Methods. ABCB1 was preferentially expressed in CD34+CD38 cells compared with more differentiated CD34+CD38+ progenitors in all normal (n = 10; black dots) and leukemic (n = 10; white dots) samples examined. MRK16 staining intensity is categorized as follows: negative (D < 0.10), dim (0.10 < D < 0.15), moderate (0.15 < D > 0.25), and bright (D > 0.25). No difference existed in levels of expression in CD34+CD38 cells between normal bone marrow and AML (median D, 0.13 ± 0.03 and 0.12 ± 0.04, respectively).

Figure 2.

ABCB1 is differentially expressed in CD34+CD38 cells in both normal bone marrow and AML. ABCB1 expression was assessed flow cytometrically using the MRK16 monoclonal antibody and valued using the Kolmogorov-Smirnov statistic, denoted as D, as described in the Materials and Methods. ABCB1 was preferentially expressed in CD34+CD38 cells compared with more differentiated CD34+CD38+ progenitors in all normal (n = 10; black dots) and leukemic (n = 10; white dots) samples examined. MRK16 staining intensity is categorized as follows: negative (D < 0.10), dim (0.10 < D < 0.15), moderate (0.15 < D > 0.25), and bright (D > 0.25). No difference existed in levels of expression in CD34+CD38 cells between normal bone marrow and AML (median D, 0.13 ± 0.03 and 0.12 ± 0.04, respectively).

Close modal

CD34+CD38 cells in normal bone marrow were mitoxantrone “dull” compared with CD34+CD38+ cells (MFI, 3.08 ± 0.23 versus 7.29 ± 0.50; n = 16; P < 0.001). Modulation of ABCB1-mediated transport by verapamil significantly increased mitoxantrone fluorescence in CD34+CD38 cells (Fig. 1B). In line with expression profiles, ABCB1-mediated efflux was invariably higher in CD34+CD38 cells (mean EI, 1.99 ± 0.08; range, 1.64-2.92) compared with more differentiated CD34+CD38+ cells (1.45 ± 0.06; Fig. 3A). Serial measurements of mitoxantrone fluorescence in CD34+CD38 cells both after 2-hour exposure with mitoxantrone and after an additional hour of efflux in drug-free medium done in a subpanel of samples (n = 7) showed complete abrogation of mitoxantrone extrusion by ABCB1 modulation (Fig. 4A).

Figure 3.

Reduced ABCB1-mediated drug transport in CD34+CD38 cells is a biological commonality in AML. ABCB1-mediated transport in hematopoietic CD34+CD38 cells from normal bone marrow and AML samples depicted as an EI of substrate fluorescence in the presence or absence of verapamil. Black dot, each sample. Average ABCB1 efflux index ±SEM is indicated. A, ABCB1-mediated mitoxantrone transport as determined by flow cytometry. B, ABCB1-mediated rhodamine 123 transport as determined by single-cell image analysis.

Figure 3.

Reduced ABCB1-mediated drug transport in CD34+CD38 cells is a biological commonality in AML. ABCB1-mediated transport in hematopoietic CD34+CD38 cells from normal bone marrow and AML samples depicted as an EI of substrate fluorescence in the presence or absence of verapamil. Black dot, each sample. Average ABCB1 efflux index ±SEM is indicated. A, ABCB1-mediated mitoxantrone transport as determined by flow cytometry. B, ABCB1-mediated rhodamine 123 transport as determined by single-cell image analysis.

Close modal
Figure 4.

ABCB1 modulation abrogates mitoxantrone extrusion from normal but not leukemic CD34+CD38 cells. To assess whether mitoxantrone efflux occurs in CD34+CD38 cells in the presence of ABCB1 modulation, serial measurements were done of mitoxantrone (MITO) fluorescence both after 2 hours of exposure to mitoxantrone and after an additional hour of efflux in drug-free medium, both in the presence and absence of ABCB1 modulation. Inhibition of ABCB1 by verapamil (VERA) completely abrogates mitoxantrone efflux during the additional hour of efflux in drug-free medium in CD34+CD38 cells from normal bone marrow (MFI, 6.74 ± 0.34 versus 6.74 ± 0.49, respectively; A) but not in AML (MFI, 6.90 ± 0.55 versus 6.26 ± 0.62, respectively, difference using t test for paired samples; P = 0.006; B). Bars, average value of mitoxantrone fluorescence ±SEM. When cells were put on ice during the additional hour of efflux, no decrease in mitoxantrone fluorescence was observed.

Figure 4.

ABCB1 modulation abrogates mitoxantrone extrusion from normal but not leukemic CD34+CD38 cells. To assess whether mitoxantrone efflux occurs in CD34+CD38 cells in the presence of ABCB1 modulation, serial measurements were done of mitoxantrone (MITO) fluorescence both after 2 hours of exposure to mitoxantrone and after an additional hour of efflux in drug-free medium, both in the presence and absence of ABCB1 modulation. Inhibition of ABCB1 by verapamil (VERA) completely abrogates mitoxantrone efflux during the additional hour of efflux in drug-free medium in CD34+CD38 cells from normal bone marrow (MFI, 6.74 ± 0.34 versus 6.74 ± 0.49, respectively; A) but not in AML (MFI, 6.90 ± 0.55 versus 6.26 ± 0.62, respectively, difference using t test for paired samples; P = 0.006; B). Bars, average value of mitoxantrone fluorescence ±SEM. When cells were put on ice during the additional hour of efflux, no decrease in mitoxantrone fluorescence was observed.

Close modal

Verapamil is regarded ABCB1 specific, although inhibition of ABCG2 (BCRP) has been observed at concentrations considerably higher than used in our experiments. To definitely confirm that the drug efflux from normal CD34+CD38 cells was indeed ABCB1 mediated, additional experiments were done.

First, similar results were found when the ABCB1-specific inhibitor PSC-833 was used in a subgroup of samples (n = 4; mean EI ± SE, 2.03 ± 0.12 and 1.38 ± 0.09 for CD34+CD38 and CD34+CD38+ cells, respectively; Fig. 1B), showing that the observed efflux is ABCB1 rather than BCRP mediated because PSC833 is not an inhibitor of BCRP. This was further confirmed using rhodamine 123 as a substrate, which is not a substrate for BCRP (23), in an independent assay in single CD34+CD38 cells sorted on an adhesive biolayer (Fig. 1C). Rhodamine 123 retention was significantly lower in CD34+CD38 cells compared with CD34+CD38+ cells in normal bone marrow (P < 0.001), similar to the mitoxantrone dull phenotype of CD34+CD38 cells. Blocking of ABCB1-mediated transport by verapamil increased rhodamine 123 fluorescence significantly. ABCB1-mediated efflux was found in all examined (n = 5) CD34+CD38 samples (mean EI, 3.74 ± 1.81; Fig. 3B).

Together, these results show that ABCB1-mediated transport is the major determinant of the mitoxantrone dull phenotype of CD34+CD38 cells in human normal bone marrow and that its modulation completely abrogates mitoxantrone extrusion from these cells.

ABCB1-mediated transport is reduced in CD34+CD38 hematopoietic cells in AML. Conserved ABCB1-mediated drug extrusion from CD34+CD38 cells after malignant transformation would be an important obstacle for chemotherapeutic eradication of these cells in AML. We therefore investigated ABCB1 expression and function of this cell population in AML compared with their counterparts in normal bone marrow.

Investigations were done on a panel of bone marrow samples of 20 untreated AML patients (Table 1). The panel consisted predominantly of CD34+ leukemias, with CD34+ cells median percentage of 31% (range, 1-91). CD34+CD38 cells in AML were immunophenotypically defined as indicated in Fig. 1A. Fluorescence in situ hybridization analyses in AML patients who carried a cytogenetic abnormality and from whom sufficient cells were available for analysis showed a predominantly leukemic character of CD34+CD38 cells in these samples (n = 5; Table 2) as described previously (24).

Table 1.

Patient characteristics

N 20 
Sex  
    Male 
    Female 11 
Age, median (range) 50 (15-71) 
% CD34 31 (1-91) 
French-American-British  
    M1 
    M2 
    M4 
    M5 
Cytogenetics  
    Normal 
    t(8;21) 4 (+2 complex) 
    (6;11) 
    45XO 
    Complex 
N 20 
Sex  
    Male 
    Female 11 
Age, median (range) 50 (15-71) 
% CD34 31 (1-91) 
French-American-British  
    M1 
    M2 
    M4 
    M5 
Cytogenetics  
    Normal 
    t(8;21) 4 (+2 complex) 
    (6;11) 
    45XO 
    Complex 
Table 2.

CD34+CD38 cells in AML are predominately of leukemic origin

Patient no.Cytogenetic aberrancy% Cytogenetic aberrant CD34+CD38 cellsABCB1 EI
Rhodamine 123 [3.74 (1.76-5.66)*]Mitoxantrone [1.99 (1.64-2.94)*]
−5, −7, −15, −17, t(2;3) 56 0.84 1.32 
t(8;21) 90 NA 0.92 
t(8;21) 95 0.81 1.15 
t(8;21) 90 1.39 NA 
t(8;21) 100 NA 1.28 
Patient no.Cytogenetic aberrancy% Cytogenetic aberrant CD34+CD38 cellsABCB1 EI
Rhodamine 123 [3.74 (1.76-5.66)*]Mitoxantrone [1.99 (1.64-2.94)*]
−5, −7, −15, −17, t(2;3) 56 0.84 1.32 
t(8;21) 90 NA 0.92 
t(8;21) 95 0.81 1.15 
t(8;21) 90 1.39 NA 
t(8;21) 100 NA 1.28 

Abbreviation: NA, material not available for analysis.

*

Values normal bone marrow: mean (range).

ABCB1 expression in CD34+CD38 cells in AML as assessed by the MRK16 monoclonal antibody was comparable with expression of their counterparts in normal bone marrow (Fig. 2). Similar to the expression in normal bone marrow, decreased protein expression in CD34+CD38+ and CD34 subsets was observed in all samples.

Surprisingly, ABCB1-mediated mitoxantrone transport was invariably reduced in CD34+CD38 cells in AML compared with their counterparts in normal bone marrow as shown by a decreased effect of ABCB1 modulation by verapamil on intracellular mitoxantrone fluorescence (mean EI, 1.21 ± 0.05; range, 0.93-1.51; P < 0.0001; Figs. 1B and 3A). Similar results were found when PSC-833 was used as an inhibitor (n = 4; mean EI in CD34+CD38 cells, 1.23 ± 0.16; Fig. 1B). Reduced ABCB1 function was not restricted to CD34+CD38 cells in AML; reduced ABCB1 function compared with normal counterparts was also found for CD34+CD38+ cells (mean EI, 1.17 ± 0.03; range, 1.05-1.31; P < 0.001). Reduced ABCB1-mediated transport in CD34+CD38 cells in AML was confirmed using rhodamine 123 as a substrate in single cells (Figs. 1C and 3B). CD34+CD38 cells in AML (n = 8) were rhodamine 123 dull compared with the more differentiated CD34+CD38+ cells, but ABCB1 modulation by verapamil did not increase intracellular retention (mean EI, 0.99 ± 0.07; Fig. 3B). Of interest, analysis of mitoxantrone efflux mediated by the ABC transporters ABCG2 (BCRP), done in a series of normal and leukemic bone marrow samples, including those described in this article, using the ABCG2-specific fumitremorgin C analogue KO143 as inhibitor, showed no difference between normal and leukemic CD34+CD38 cells (15), further showing that the observed decrease in mitoxantrone efflux is indeed due to deficient ABCB1 rather than ABCG2 activity.

Serial measurements both after 2 hours of drug exposure and after an additional hour in drug-free medium in a subpanel of AML samples (n = 10; Fig. 4B) showed significant extrusion of mitoxantrone in the presence of verapamil in the majority of patients, pointing in the direction of redundant drug extrusion mechanisms.

These redundant drug transport mechanisms, however, did not compensate for the reduced ABCB1-mediated transport from leukemic CD34+CD38 cells as suggested by the significantly higher mitoxantrone retention in leukemic CD34+CD38 cells compared with their counterparts in normal bone marrow after drug exposure and subsequent efflux in drug-free medium (MFI, 4.54 ± 0.46 versus 3.08 ± 0.23; P = 0.004). These findings strongly suggest that the decreased efficacy of ABCB1 modulation on mitoxantrone retention of leukemic CD34+CD38 cells is indeed due to reduced ABCB1 activity rather than increased activity of other transport mechanisms, which would have resulted in similar or lower retention of mitoxantrone compared with their normal CD34+CD38 counterparts.

Translated to the clinical context, these data indicate that modulation of ABCB1 has limited effect on mitoxantrone retention in leukemic CD34+CD38 cells due to reduced activity of ABCB1 in leukemic CD34+CD38 cells. Drug efflux from these cells occurs in the presence of ABCB1 modulation due to the activity of promiscuous transport mechanisms.

ABCB1-mediated drug transport identifies a subpopulation of residual normal CD34+CD38 hematopoietic stem cells in a patient with CBFB-MYH11-positive AML. The finding that impairment of ABCB1 function is biological commonality in CD34+CD38 cells in AML compared with normal bone marrow implies that, in AML, residual normal CD34+CD38 hematopoietic cells may be identified by their conserved ABCB1-mediated transport capacity.

Indeed, during additional monitoring, one patient was identified with a clearly discernable CD34+CD38 mitoxantrone dull population caused by ABCB1-mediated drug efflux within the range observed in normal bone marrow (Fig. 5). Because the intracellular presence of mitoxantrone precluded the use of fluorescence in situ hybridization, we used real-time quantitative reverse transcription-PCR to quantify the number of CBFB-MYH11 transcripts in these CD34+CD38 subpopulations. The number of transcripts in the CD34+CD38 ABCB1 efflux ± cell population was similar to that in the positive control (blast population from CBFB-MYH11, type D–positive patient; Xn, 4.3 × 10−3 versus 3.5 × 10−3 copies CBFB-MYH11/copy glyceraldehyde-3-phosphate dehydrogenase, respectively), whereas no transcripts were detected in the CD34+CD38 ABCB1 efflux ++ subpopulation. This sample provides proof of principle that conserved ABCB1-mediated drug efflux may indeed identify a subpopulation of CD34+CD38 cells that harbor residual normal cells at least in a subgroup of AML patients. This implies that modulation of ABCB1 preferentially targets these residual normal cells as shown in Fig. 5.

Figure 5.

ABCB1-mediated mitoxantrone transport identifies a subpopulation that harbors residual normal CD34+CD38 cells in a patient with CBFB-MYH11 AML. A patient was identified in which mitoxantrone discriminated a population of CD34+CD38 mitoxantrone dull cells. The dull phenotype was determined by strong ABCB1-mediated efflux (CD34+CD38 ABCB1 efflux ++) within the range observed in CD34+CD38 cells observed in normal bone marrow (EI, 2.50). Real-time PCR confirmed the residual normal character of these cells.

Figure 5.

ABCB1-mediated mitoxantrone transport identifies a subpopulation that harbors residual normal CD34+CD38 cells in a patient with CBFB-MYH11 AML. A patient was identified in which mitoxantrone discriminated a population of CD34+CD38 mitoxantrone dull cells. The dull phenotype was determined by strong ABCB1-mediated efflux (CD34+CD38 ABCB1 efflux ++) within the range observed in CD34+CD38 cells observed in normal bone marrow (EI, 2.50). Real-time PCR confirmed the residual normal character of these cells.

Close modal

In this report, we show that ABCB1-mediated drug transport is reduced in leukemic CD34+CD38 progenitor cells compared with their normal counterparts in AML.

This finding was initially unanticipated because AML is generally considered to be an ABCB1-overexpressing malignancy with conserved physiologic function (25). This assumption, however, is based on the observation that ABCB1 is preferentially expressed in CD34+ blasts in AML (26) similar to preferential expression of ABCB1 in these cells in normal bone marrow (27), thus reflecting similar patterns of expression rather than direct comparison of ABCB1 expression and function of normal and malignant cell subpopulations. Expression data in our study confirm the preferential expression of ABCB1 in CD34+ cells in AML (26) at levels comparable with those observed in normal bone marrow. ABCB1 transport capacity, however, was markedly impaired in both CD34+CD38 and CD34+CD38+ cells in AML.

The occurrence of ABCB1 protein+ efflux leukemic blasts has been observed in a minority of AML cases (28) earlier, pointing in the direction of the presence of nonfunctional forms of the ABCB1 protein, but the current report, aimed at directly comparing ABCB1 function between normal and leukemic primitive progenitor cells, shows that impaired ABCB1 function is a biological commonality rather than an occasional finding in AML.

The molecular determinants behind impaired ABCB1-mediated transport in hematopoietic cells in AML remain to be elucidated and may comprise both downstream event of oncoproteins central in leukemogenesis, such as AML1-ETO (29) and TEL-AML1 (30), and factors governed by the cellular environment, such as disruption of cellular ATP metabolism, membrane integrity, or cell cycle regulation (31).

Regardless of the underlying mechanism, the finding that ABCB1-mediated transport is impaired in CD34+CD38 cells in AML has important clinical implications. The recognition of AML as a disease originating from these cells dictates a paradigm shift in the treatment toward the eradication of this crucial stem cell population in AML.

Our results indicate that ABCB1 modulation does not circumvent drug extrusion from these cells and that additional, redundant drug extrusion mechanisms determine the relatively dull mitoxantrone appearance of these cells. We have recently documented ABCG2/BCRP as one of these additional drug transporters in leukemic CD34+CD38 cells (15). Additionally, using low-density real-time PCR arrays, we recently showed differential expression of all 13 ABC transporters currently associated with drug extrusion in CD34+CD38 cells in AML (32). These studies, however, did not show higher expression or function of these transporters in leukemic CD34+CD38 cells compared with their normal counterparts. This may explain the observation in this study that intracellular retention of mitoxantrone is increased in leukemic CD34+CD38 cells compared with their normal counterparts, suggesting that redundant mechanisms confer drug extrusion from leukemic cells but that this redundant action does not completely compensate for the reduced ABCB1 function in these cells.

The combined data suggest that the prognostic value of ABCB1, one of the main rationales for ABCB1 targeting in AML, may be pleiotropoic, identifying leukemias with a primitive CD34+ (CD38) character and innate drug resistance due to differential expression of many, promiscuous ABC transporters and possibly other mechanisms protecting primitive progenitor cells. Modulation of additional transporters is therefore likely to be required to abrogate drug extrusion and to enhance drug-induced eradication of leukemic stem cells in AML. These laboratory findings provide an alternative explanation for the poor results of ABCB1 modulation by the highly potent ABCB1 inhibitor PSC-833 and other inhibitors on long-term disease outcome in clinical trials (810). The report of improved clinical outcome in a trial using cyclosporin as an ABCB1 inhibitor in AML (13) may reflect these findings because cyclosporin is a more promiscuous ABC transporter inhibitor with effects on ABCB1 and other ABC transporters (33).

Additionally, the current study shows that residual normal CD34+CD38 cells are likely to harbor within the ABCB1 efflux ++ subpopulation. This seems congruent with the report (34) that the capacity to efflux Hoechst 33342 (side population) identifies a subpopulation of CD34+CD38 residual normal cells in AML, whereas CD34+CD38 Hoechst bright cells were leukemic. Although these authors could not relate Hoechst 33342 efflux with ABCB1 protein expression in the overall side population of cells, this was not investigated in the CD34+CD38 subset. The current report shows that differences in ABCB1-mediated efflux discriminate normal from leukemic CD34+CD38 cells. This finding implies that ABCB1 modulation may preferentially target residual normal hematopoietic cells, whereas drug extrusion from leukemic CD34+CD38 cells occurs through the activity of redundant mechanisms. Together, these data argue against the use of ABCB1 modulation as a strategy to eradicate leukemic stem cells in AML but prompt further research to elucidate the mechanisms involved in the extrusion of drugs from leukemic CD34+CD38 cells to provide novel targets for the eradication of this crucial cell population in AML.

Grant support: Dutch Cancer Society grant (M.H.G.P. Raaijmakers) and Vanderes Cancer Foundation.

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

We thank L. Heuver, A. Pennings, and G. Vierwinden for technical assistance; Hematology Data Base Center Nijmegen for providing material and clinical data; and F. Russel for critically reviewing the article.

1
Huntly BJ, Gilliland DG. Leukaemia stem cells and the evolution of cancer-stem-cell research.
Nat Rev Cancer
2005
;
5
:
311
–21.
2
Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell.
Nat Med
1997
;
3
:
730
–7.
3
Dean M, Fojo T, Bates S. Tumour stem cells and drug resistance.
Nat Rev Cancer
2005
;
5
:
275
–84.
4
Ambudkar SV, Dey S, Hrycyna CA, et al. Biochemical, cellular, and pharmacological aspects of the multidrug transporter.
Annu Rev Pharmacol Toxicol
1999
;
39
:
361
–98.
5
Chaudhary PM, Roninson IB. Expression and activity of P-glycoprotein, a multidrug efflux pump, in human hematopoietic stem cells.
Cell
1991
;
66
:
85
–94.
6
Raaijmakers MH, van Emst L, De Witte T, Mensink E, Raymakers RA. Quantitative assessment of gene expression in highly purified hematopoietic cells using real-time reverse transcriptase polymerase chain reaction.
Exp Hematol
2002
;
30
:
481
–7.
7
Leith CP, Kopecky KJ, Chen IM, et al. Frequency and clinical significance of the expression of the multidrug resistance proteins MDR1/P-glycoprotein, MRP1, and LRP in acute myeloid leukemia: a Southwest Oncology Group Study.
Blood
1999
;
94
:
1086
–99.
8
van der Holt B, Lowenberg B, Burnett AK, et al. The value of the MDR1 reversal agent PSC-833 in addition to daunorubicin and cytarabine in the treatment of elderly patients with previously untreated acute myeloid leukemia (AML), in relation to MDR1 status at diagnosis.
Blood
2005
;
106
:
2646
–54.
9
Baer MR, George SL, Dodge RK, et al. Phase 3 study of the multidrug resistance modulator PSC-833 in previously untreated patients 60 years of age and older with acute myeloid leukemia: Cancer and Leukemia Group B Study 9720.
Blood
2002
;
100
:
1224
–32.
10
Greenberg PL, Lee SJ, Advani R, et al. Mitoxantrone, etoposide, and cytarabine with or without valspodar in patients with relapsed or refractory acute myeloid leukemia and high-risk myelodysplastic syndrome: a phase III trial (E2995).
J Clin Oncol
2004
;
22
:
1078
–86.
11
Mahadevan D, List AF. Targeting the multidrug resistance-1 transporter in AML: molecular regulation and therapeutic strategies.
Blood
2004
;
104
:
1940
–51.
12
Tallman MS, Gilliland DG, Rowe JM. Drug therapy for acute myeloid leukemia.
Blood
2005
;
106
:
1154
–63.
13
List AF, Kopecky KJ, Willman CL, et al. Benefit of cyclosporine modulation of drug resistance in patients with poor-risk acute myeloid leukemia: a Southwest Oncology Group study.
Blood
2001
;
98
:
3212
–20.
14
Smeets M, Raymakers R, Vierwinden G, et al. A low but functionally significant MDR1 expression protects primitive haemopoietic progenitor cells from anthracycline toxicity.
Br J Haematol
1997
;
96
:
346
–55.
15
Raaijmakers MH, de Grouw EP, Heuver LH, et al. Breast cancer resistance protein in drug resistance of primitive CD34+38 cells in acute myeloid leukemia.
Clin Cancer Res
2005
;
11
:
2436
–44.
16
Yusa K, Tsuruo T. Reversal mechanism of multidrug resistance by verapamil: direct binding of verapamil to P-glycoprotein on specific sites and transport of verapamil outward across the plasma membrane of K562/ADM cells.
Cancer Res
1989
;
49
:
5002
–6.
17
Boesch D, Gaveriaux C, Jachez B, et al. In vivo circumvention of P-glycoprotein-mediated multidrug resistance of tumor cells with SDZ.
Cancer Res
1991
;
51
:
4226
–33.
18
Ozvegy C, Litman T, Szakacs G, et al. Functional characterization of the human multidrug transporter, ABCG2, expressed in insect cells.
Biochem Biophys Res Commun
2001
;
285
:
111
–7.
19
Laupeze B, Amiot L, Payen L, et al. Multidrug resistance protein (MRP) activity in normal mature leukocytes and CD34-positive hematopoietic cells from peripheral blood.
Life Sci
2001
;
68
:
1323
–31.
20
Scharenberg CW, Harkey MA, Torok-Storb B. The ABCG2 transporter is an efficient Hoechst 33342 efflux pump and is preferentially expressed by immature human hematopoietic progenitors.
Blood
2002
;
99
:
507
–12.
21
Broxterman HJ, Sonneveld P, Feller N, et al. Quality control of multidrug resistance assays in adult acute leukemia: correlation between assays for P-glycoprotein expression and activity.
Blood
1996
;
87
:
4809
–16.
22
Raaijmakers MHGP, Van den Bosch G, Boezeman J, De Witte T, Raymakers RA. Single-cell image analysis to assess ABC-transporter-mediated efflux in highly purified hematopoietic progenitors.
Cytometry
2002
;
49
:
135
–42.
23
Robey RW, Honjo Y, van de LA, et al. A functional assay for detection of the mitoxantrone resistance protein, MXR (ABCG2).
Biochim Biophys Acta
2001
;
1512
:
171
–82.
24
Haase D, Feuring-Buske M, Konemann S, et al. Evidence for malignant transformation in acute myeloid leukemia at the level of early hematopoietic stem cells by cytogenetic analysis of CD34+ subpopulations.
Blood
1995
;
86
:
2906
–12.
25
List AF, Spier CM. Multidrug resistance in acute leukemia: a conserved physiologic function.
Leuk Lymphoma
1992
;
8
:
9
–14.
26
te Boekhorst PA, de Leeuw K, Schoester M, et al. Predominance of functional multidrug resistance (MDR-1) phenotype in CD34+ acute myeloid leukemia cells.
Blood
1993
;
82
:
3157
–62.
27
Drach D, Zhao S, Drach J, et al. Subpopulations of normal peripheral blood and bone marrow cells express a functional multidrug resistant phenotype.
Blood
1992
;
80
:
2729
–34.
28
Leith CP, Chen IM, Kopecky KJ, et al. Correlation of multidrug resistance (MDR1) protein expression with functional dye/drug efflux in acute myeloid leukemia by multiparameter flow cytometry: identification of discordant MDR−/efflux+ and MDR1+/efflux− cases.
Blood
1995
;
86
:
2329
–42.
29
Lutterbach B, Sun D, Schuetz J, Hiebert SW. The MYND motif is required for repression of basal transcription from the multidrug resistance 1 promoter by the t(8;21) fusion protein.
Mol Cell Biol
1998
;
18
:
3604
–11.
30
Miyazaki K, Uchida H, Miyachi H, et al. TEL/AML1, the leukemia-related chimeric protein overcomes drug resistance through transcriptional repression of multidrug resistance-1 (MDR1) gene expression [abstract 242]. Blood Suppl 2002;100.
31
Smeets ME, Raymakers RA, Vierwinden G, et al. Triggering noncycling hematopoietic progenitors and leukemic blasts to proliferate increases anthracycline retention and toxicity by downregulating multidrug resistance.
Blood
1999
;
94
:
2414
–23.
32
de Grouw EP, Raaijmakers MHGP, Boezeman J, et al. Preferential expression of a high number of ATP binding cassette transporters in both normal and leukemic CD34+CD38 cells.
Leukemia
2006
;
20
:
750
–4.
33
Qadir M, O'Loughlin KL, Fricke SM, et al. Cyclosporin A is a broad-spectrum multidrug resistance modulator.
Clin Cancer Res
2005
;
11
:
2320
–6.
34
Feuring-Buske M, Hogge DE. Hoechst 33342 efflux identifies a subpopulation of cytogenetically normal CD34(+)CD38(−) progenitor cells from patients with acute myeloid leukemia.
Blood
2001
;
97
:
3882
–9.