To the editor:

The human organic cation transporter (OCT1) has been implicated in the uptake of imatinib into chronic myeloid leukemia (CML) cells.1-4  Moreover, OCT1 expression or activity is thought to be a critical determinant of imatinib response and is considered to predict the long-term outcome of chronic-phase CML.5-7  Assessment of OCT1 activity was derived from the intracellular uptake and retention (IUR) of imatinib with and without inhibitors such as prazosin or amantadine.1,3  These functional OCT1 activity assays, as originally devised by Thomas et al and later on also used by White and colleagues, solely rely on transporter inhibition.1,3  For the correct interpretation of these measurements, it is essential that this inhibitor-based assay truly reflect OCT1 transporter activity. This is a clinically relevant issue in view of the ongoing debate on how to identify patients with chronic-phase CML whose disease is likely to respond poorly to imatinib. White and Hughes argue that functional OCT1 activity is the best classifier for stratification, whereas others consider the BCR-ABL transcript levels to be the superior prognostic marker.8 

In a recent paper in Blood, Giannoudis et al provided evidence that OCT1 M420del modifies clinical outcome in patients with CML treated with imatinib.9  In vitro studies using OCT1-transfected KCL22 cells showed that M420del is associated with impaired IUR of imatinib, suggesting that patients carrying this M420del allele might do better if treated with a tyrosine kinase inhibitor that does not rely on OCT1 transporter activity. Functional OCT1 activity was determined by subtracting the uptake in the presence of amantadine, a specific OCT1 inhibitor according to the authors, from the uptake observed in unmanipulated cells. Examination of the data presented in their Figure 2 suggest that the IUR of imatinib in the absence of 500 µM of amantadine was increased by ∼50% in OCT1-transfected KCL22 cells compared with mock-transfected cells.9  It was assumed that this difference in imatinib uptake could be attributed to OCT1 activity. Remarkably, although mock-transfected KCL22 cells did not express detectable OCT1 mRNA and protein (Figure 4, Giannoudis et al9 ), a substantial IUR of imatinib was observed, which could be partially inhibited by amantadine. According to the authors (Figure 4, Giannoudis et al9 ), this decrease in IUR of imatinib is attributable to OCT1 activity. On the basis of these observations and our experimental data (see below), we argue that these inhibitor-based assays, designed to quantify functional OCT1 transporter activity, are in fact measuring additional, as yet unidentified, processes involved in imatinib IUR. Therefore, we have serious concerns about the use of “specific” OCT1 inhibitors to demonstrate OCT1-mediated uptake of imatinib. Using HEK293 (human embryonal kidney) cells, we show that overexpression of OCT1 resulted in a limited (∼25%) increase in imatinib IUR (Figure 1A-C). These results coincide with other imatinib uptake studies using OCT1-transfected leukemic cells or oocytes.2,10  Furthermore, we show a dose-dependent decrease of imatinib uptake by amantadine in HEK293/OCT1 cells, but importantly, amantadine also decreased the imatinib uptake in HEK293/Neo control cells having an almost 4-log lower OCT1 expression (Figure 1D-E). Similar, even more profound results were obtained with prazosin (Figure 1F-G). Analogous inhibitory effects were also seen in the leukemic K562 cell line (Figure 1H-I). Importantly, both amantadine and prazosin decreased the IUR of imatinib independent of OCT1 expression levels.

Figure 1

IUR of imatinib in HEK293 and K562 cells. (A) OCT1 mRNA expression levels in arbitrary units (a.u.) in OCT1-transfected HEK293 cells (HEK293/hOCT1) compared with mock (pcDNA3)-transfected control cells (HEK293/Neo). (B) HEK293/hOCT1 cells display increased uptake of ASP+, a fluorescent substrate of OCT1, confirming that OCT1 is functional. ASP+-associated fluorescence (a.u.) was measured by FACScan flow cytometry as described previously.11  (C) The IUR of imatinib was slightly (∼25%), but significantly (P < .05), increased in the HEK293/hOCT1 cells compared with the HEK293/Neo control cells. Cells were exposed to 30 µM of imatinib for 1 hour. Effect of amantadine (D,E,H) and prazosin (F,G,I) on the IUR of imatinib in HEK293/hOCT1 (D,F), HEK293/Neo control (E,G) cells and K562 cells (H-I). Cells were pretreated for 30 minutes with the indicated concentrations of inhibitor followed by an additional 30 minutes of exposure of the cells to 10 µM of imatinib. Uptake data are expressed as the mean ± standard deviation (derived from at least 3 replicates), and the Student t test was used to detect significant differences as indicated by an asterisk (*P < .05, **P < .01, ***P < .001) in IUR between HEK293/hOCT1 and HEK293/Neo cells (C) or between the IUR ± inhibitor (D-I). Amantadine showed a maximal inhibition of ∼65% at 1000 µM, whereas prazosin markedly inhibited the IUR of imatinib by ∼50% at 30 µM and up to ∼90% at 100 µM. It should be noted that the inhibitory effect of these compounds on the IUR of imatinib in HEK293 cells did not differ between OCT1-transfected and Neo control cells and was comparable with that seen in K562 cells having low OCT1 expression. Apparently, the inhibitory effects of these compounds do not depend on the level of OCT1 expression.

Figure 1

IUR of imatinib in HEK293 and K562 cells. (A) OCT1 mRNA expression levels in arbitrary units (a.u.) in OCT1-transfected HEK293 cells (HEK293/hOCT1) compared with mock (pcDNA3)-transfected control cells (HEK293/Neo). (B) HEK293/hOCT1 cells display increased uptake of ASP+, a fluorescent substrate of OCT1, confirming that OCT1 is functional. ASP+-associated fluorescence (a.u.) was measured by FACScan flow cytometry as described previously.11  (C) The IUR of imatinib was slightly (∼25%), but significantly (P < .05), increased in the HEK293/hOCT1 cells compared with the HEK293/Neo control cells. Cells were exposed to 30 µM of imatinib for 1 hour. Effect of amantadine (D,E,H) and prazosin (F,G,I) on the IUR of imatinib in HEK293/hOCT1 (D,F), HEK293/Neo control (E,G) cells and K562 cells (H-I). Cells were pretreated for 30 minutes with the indicated concentrations of inhibitor followed by an additional 30 minutes of exposure of the cells to 10 µM of imatinib. Uptake data are expressed as the mean ± standard deviation (derived from at least 3 replicates), and the Student t test was used to detect significant differences as indicated by an asterisk (*P < .05, **P < .01, ***P < .001) in IUR between HEK293/hOCT1 and HEK293/Neo cells (C) or between the IUR ± inhibitor (D-I). Amantadine showed a maximal inhibition of ∼65% at 1000 µM, whereas prazosin markedly inhibited the IUR of imatinib by ∼50% at 30 µM and up to ∼90% at 100 µM. It should be noted that the inhibitory effect of these compounds on the IUR of imatinib in HEK293 cells did not differ between OCT1-transfected and Neo control cells and was comparable with that seen in K562 cells having low OCT1 expression. Apparently, the inhibitory effects of these compounds do not depend on the level of OCT1 expression.

Close modal

In conclusion, we demonstrated that these inhibitors are not exclusively specific for OCT1 and, therefore, we would like to emphasize to be very cautious in the interpretation of inhibitor-based OCT1 data. In fact, our data indicate that prazosin and amantadine may also interfere with other imatinib uptake processes evidently distinct from that of OCT1-mediated IUR.

Acknowledgments: The authors thank Xander den Dekker, Sandra Segeletz, and Peter de Bruijn for technical assistance with the IUR studies. The authors are grateful to Novartis for providing imatinib.

Contributions: H.B. designed and performed the experiments, analyzed the data, produced the figure, and wrote the manuscript; R.H.J.M. and A.S. wrote the manuscript; and E.A.C.W. designed the research and wrote the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: H. Burger, Department of Medical Oncology, Room Ae 306, Erasmus MC Cancer Institute, Rotterdam, The Netherlands, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands; e-mail: h.burger@erasmusmc.nl.

1
White
 
DL
Saunders
 
VA
Dang
 
PO
, et al. 
OCT-1-mediated influx is a key determinant of the intracellular uptake of imatinib but not nilotinib (AMN107): reduced OCT-1 activity is the cause of low in vitro sensitivity to imatinib.
Blood
2006
, vol. 
108
 
2
(pg. 
697
-
704
)
2
Wang
 
L
Giannoudis
 
A
Lane
 
S
Williamson
 
P
Pirmohamed
 
M
Clark
 
RE
Expression of the uptake drug transporter hOCT1 is an important clinical determinant of the response to imatinib in chronic myeloid leukemia.
Clin Pharmacol Ther
2008
, vol. 
83
 
2
(pg. 
258
-
264
)
3
Thomas
 
J
Wang
 
L
Clark
 
RE
Pirmohamed
 
M
Active transport of imatinib into and out of cells: implications for drug resistance.
Blood
2004
, vol. 
104
 
12
(pg. 
3739
-
3745
)
4
Eechoute
 
K
Sparreboom
 
A
Burger
 
H
, et al. 
Drug transporters and imatinib treatment: implications for clinical practice.
Clin Cancer Res
2011
, vol. 
17
 
3
(pg. 
406
-
415
)
5
White
 
DL
Saunders
 
VA
Dang
 
P
, et al. 
Most CML patients who have a suboptimal response to imatinib have low OCT-1 activity: higher doses of imatinib may overcome the negative impact of low OCT-1 activity.
Blood
2007
, vol. 
110
 
12
(pg. 
4064
-
4072
)
6
White
 
DL
Radich
 
J
Soverini
 
S
, et al. 
Chronic phase chronic myeloid leukemia patients with low OCT-1 activity randomized to high-dose imatinib achieve better responses and have lower failure rates than those randomized to standard-dose imatinib.
Haematologica
2012
, vol. 
97
 
6
(pg. 
907
-
914
)
7
White
 
DL
Dang
 
P
Engler
 
J
, et al. 
Functional activity of the OCT-1 protein is predictive of long-term outcome in patients with chronic-phase chronic myeloid leukemia treated with imatinib.
J Clin Oncol
2010
, vol. 
28
 
16
(pg. 
2761
-
2767
)
8
White
 
DL
Hughes
 
TP
Classification of patients with chronic myeloid leukemia on basis of BCR-ABL transcript level at 3 months fails to identify patients with low organic cation transporter-1 activity destined to have poor imatinib response.
J Clin Oncol
2012
, vol. 
30
 
10
(pg. 
1144
-
1145; author reply 1145-1146
)
9
Giannoudis
 
A
Wang
 
L
Jorgensen
 
AL
, et al. 
The hOCT1 SNPs M420del and M408V alter imatinib uptake and M420del modifies clinical outcome in imatinib-treated chronic myeloid leukemia.
Blood
2013
, vol. 
121
 
4
(pg. 
628
-
637
)
10
Hu
 
S
Franke
 
RM
Filipski
 
KK
, et al. 
Interaction of imatinib with human organic ion carriers.
Clin Cancer Res
2008
, vol. 
14
 
10
(pg. 
3141
-
3148
)
11
Burger
 
H
Zoumaro-Djayoon
 
A
Boersma
 
AW
, et al. 
Differential transport of platinum compounds by the human organic cation transporter hOCT2 (hSLC22A2).
Br J Pharmacol
2010
, vol. 
159
 
4
(pg. 
898
-
908
)
Sign in via your Institution