In this issue of Blood, Nanbakhsh et al show that leukemic cell lines rendered resistant to cytarabine display a higher expression of the natural killer (NK) group 2, member D (NKG2D) ligands (NKG2DL) UL-16 binding proteins 1-3 (ULBP1-3). This expression is c-Myc dependent as it is abrogated by c-Myc inhibitors or small interfering RNA. Consequently, upregulation of NKG2DL renders the cell lines more sensitive to NK-cell–mediated lysis.1 

Cytarabine-resistant AML cell lines or blasts from refractory AML express some of the ligands for the NK-activating receptor NKG2D. This expression is associated with c-Myc induction. Hence, at least ULBP1 and ULBP3 have c-Myc docking sites in their gene promoter. The upregulation of ULBPs by leukemic cells renders the cells more susceptible to in vitro NK-cell–mediated lysis. The study by Nanbakhsh et al raises several questions. Usually, cytarabine is not used as a single reagent but in combination with anthracyclines; this combination has not been tested by the authors. As chemotherapy drugs usually trigger the DDR pathway, it would be interesting to replace the ATM/ATR pathway in that context. Furthermore, AML-NK recognition has also been attributed to NCRs and DNAM-1. It would then be interesting to study the expression of the ligands for these receptors by AML cells after chemotherapy treatment. HLA-I expression by leukemic cells is also an escape mechanism. Is c-Myc or simply cytarabine associated with HLA upregulation or downregulation? Finally, phenotypically and functionally defective NK cells have often been described in AML. Consequently, the expression of NKG2DL might not result in an increased sensitivity to NK-cell killing in patients. These questions summarize the up-to-date challenges in drug-resistant AML treatment. ATM, ataxia telangiectasia mutated; ATR, ataxia telangiectasia and Rad3 related; DNAM-1-L, DNAX accessory molecule-1-ligands; NCR-L, natural cytotoxicity receptors-ligands. Professional illustration by Xavier Studio.

Cytarabine-resistant AML cell lines or blasts from refractory AML express some of the ligands for the NK-activating receptor NKG2D. This expression is associated with c-Myc induction. Hence, at least ULBP1 and ULBP3 have c-Myc docking sites in their gene promoter. The upregulation of ULBPs by leukemic cells renders the cells more susceptible to in vitro NK-cell–mediated lysis. The study by Nanbakhsh et al raises several questions. Usually, cytarabine is not used as a single reagent but in combination with anthracyclines; this combination has not been tested by the authors. As chemotherapy drugs usually trigger the DDR pathway, it would be interesting to replace the ATM/ATR pathway in that context. Furthermore, AML-NK recognition has also been attributed to NCRs and DNAM-1. It would then be interesting to study the expression of the ligands for these receptors by AML cells after chemotherapy treatment. HLA-I expression by leukemic cells is also an escape mechanism. Is c-Myc or simply cytarabine associated with HLA upregulation or downregulation? Finally, phenotypically and functionally defective NK cells have often been described in AML. Consequently, the expression of NKG2DL might not result in an increased sensitivity to NK-cell killing in patients. These questions summarize the up-to-date challenges in drug-resistant AML treatment. ATM, ataxia telangiectasia mutated; ATR, ataxia telangiectasia and Rad3 related; DNAM-1-L, DNAX accessory molecule-1-ligands; NCR-L, natural cytotoxicity receptors-ligands. Professional illustration by Xavier Studio.

Close modal

Although not described as a major receptor involved in the recognition of acute myeloid leukemic cells (in contrast to others such as natural cytotoxicity receptors [NCRs] NKp30, NKp44, and NKp46 or DNAM-1), NKG2D is one of the main activating receptors expressed by NK cells and displays some role in the recognition of AML after histone deacetylase inhibitor (HDACi) therapy.2,3  Noteworthy, NKG2DLs include ULBP1-4 and the nonclassical HLA class I (HLA-I) molecules MICA/B.

This work, in line with previous studies, confirms that it is possible to modify leukemic cells to render them more susceptible to NK-cell recognition. Hence, cytarabine, a drug that interferes with DNA synthesis and kills cycling cells, is used as a classical treatment of leukemia. However, especially in acute myeloid leukemia (AML), it is usually used in combination with anthracyclines. It would be interesting to analyze the effect of cotreatment of leukemic cells with cytarabine and anthracyclines (such as daunorubicine) on not only the induction of c-Myc but also the expression of NKG2DL.

As mentioned by Nanbakhsh et al, c-Myc regulates the expression of ∼15% of all cellular genes. In the cellular model used by the authors, induction of c-Myc is involved directly or indirectly in the upregulation of NKG2DL. Cytarabine and c-Myc both trigger the DNA damage response (DDR) pathway, here independently of p53. Although no clear hints were obtained from gene expression from cytarabine-resistant cell lines, it would be interesting to check whether other ligands for activating NK receptors, such as NCR and DNAM-1 ligands, are modified. For instance PVR, a ligand for DNAM-1, can be expressed after DNA damage.4  Unfortunately, little is known about the ligands for NCR. On a global point of view, it would be interesting to verify whether cytarabine (alone or in combination with anthracyclines) would also upregulate other ligands of NK receptors (see figure). Another question that rises is whether this is the toxicity of the drugs that induces the expression of NKG2DL or whether this is due to a bystander effect generated consequently to c-Myc induction and other genes linked to DNA damage or cell arrest.5 

Nanbakhsh et al also confirm their finding in primary leukemic cells. Hence, in 8 patients suffering from refractory AML (ie, resistant to chemotherapy treatment), leukemic cells are ULBP1-3 positive and this expression is reduced after ex vivo treatment with c-Myc inhibitor. These data suggest that, although AML blasts are resistant to cytarabine, they are sensitive to in vitro NK-cell–mediated killing thanks to the expression of NKG2DL. As mentioned above, DNAM-1 and NKp46 are also involved in AML killing. Therefore, it would be interesting to test whether ligands for these receptors are also induced by chemotherapy. It is noteworthy that other factors may explain the inability of NK cells to definitely clear the tumor cells. As noticed by the authors, one should remember that many factors might alter the successful recognition of leukemic cells by autologous NK cells. As mentioned above, the other NK receptors may be involved, but the expression of HLA-I molecules may also be a critical parameter. Hence, a major limitation for antitumor activity of autologous NK cells remains the inhibition by HLA-I expressed by leukemic cells. Although HLA downmodulation has been observed in many cancers including AML, unless there is specific HLA loss, HLA expression usually remains high enough to ensure NK-cell inhibition. In that context, it would be then interesting to test whether HLA-matched normal NK cells could kill cytarabine-treated AML blasts (see figure).

Last but not least, another parameter to take into account for the failure of NK cells to clear AML cells positive for NKG2DL, albeit resistant to chemotherapy, is the overall responsiveness of autologous NK cells. Several studies have highlighted defects of NK cells in AML both at diagnosis and in refractory and relapse stages. Among the defects identified, NCR, NKG2D, and DNAM-1 (Chretien et al,6  Sanchez-Correa et al,7  and C.F. and D.O., unpublished data) are often downregulated in patients. Based on this knowledge, it is easy to understand why the effects generated by cytarabine and c-Myc on leukemic cells in patients may not be sufficient to clear refractory or relapsing AML. As the authors mentioned, the current study emphasizes the potential of combinatorial therapy using, for instance, donor–NK-cell infusion, that is, cells that have not been altered by AML development in drug-resistant patients.

As c-Myc is an oncogene that promotes tumor growth and resistance to drugs, this study raises the question of whether, in refractory AML, inhibiting c-Myc with inhibitors would be a good strategy. Studies are required to decipher the impact of chemotherapies on the immunogenicity of remaining AML cells. This strategy will have to be carefully examined because ongoing clinical studies have embarked novel chemotherapy combinations together with the methyltransferase inhibitor decitabine and HDACi that display complex functional consequences on AML functions in vitro.8-10 

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

1
Nanbakhsh
 
A
Pochon
 
C
Mallavialle
 
A
Amsellem
 
S
Bourhis
 
JH
Chouaib
 
S
c-Myc regulates expression of NKG2D ligands ULBP1/2/3 in AML and modulates their susceptibility to NK-mediated lysis.
Blood
2014
, vol. 
123
 
23
(pg. 
3585
-
3595
)
2
Diermayr
 
S
Himmelreich
 
H
Durovic
 
B
, et al. 
NKG2D ligand expression in AML increases in response to HDAC inhibitor valproic acid and contributes to allorecognition by NK-cell lines with single KIR-HLA class I specificities.
Blood
2008
, vol. 
111
 
3
(pg. 
1428
-
1436
)
3
Kato
 
N
Tanaka
 
J
Sugita
 
J
, et al. 
Regulation of the expression of MHC class I-related chain A, B (MICA, MICB) via chromatin remodeling and its impact on the susceptibility of leukemic cells to the cytotoxicity of NKG2D-expressing cells.
Leukemia
2007
, vol. 
21
 
10
(pg. 
2103
-
2108
)
4
Soriani
 
A
Zingoni
 
A
Cerboni
 
C
, et al. 
ATM-ATR-dependent up-regulation of DNAM-1 and NKG2D ligands on multiple myeloma cells by therapeutic agents results in enhanced NK-cell susceptibility and is associated with a senescent phenotype.
Blood
2009
, vol. 
113
 
15
(pg. 
3503
-
3511
)
5
Cerboni
 
C
Fionda
 
C
Soriani
 
A
, et al. 
The DNA damage response: a common pathway in the regulation of NKG2D and DNAM-1 ligand expression in normal, infected, and cancer cells.
Front Immunol
2014
, vol. 
4
 pg. 
508
 
6
Chretien
 
AS
Le Roy
 
A
Vey
 
N
, et al. 
Cancer-induced alterations of NK-mediated target recognition: current and investigational pharmacological strategies aiming at restoring NK-mediated anti-tumor activity.
Front Immunol
2014
, vol. 
5
 pg. 
122
 
7
Sanchez-Correa
 
B
Morgado
 
S
Gayoso
 
I
, et al. 
Human NK cells in acute myeloid leukaemia patients: analysis of NK cell-activating receptors and their ligands.
Cancer Immunol Immunother
2011
, vol. 
60
 
8
(pg. 
1195
-
1205
)
8
Schmiedel
 
BJ
Arélin
 
V
Gruenebach
 
F
Krusch
 
M
Schmidt
 
SM
Salih
 
HR
Azacytidine impairs NK cell reactivity while decitabine augments NK cell responsiveness toward stimulation.
Int J Cancer
2011
, vol. 
128
 
12
(pg. 
2911
-
2922
)
9
Elias
 
S
Yamin
 
R
Golomb
 
L
, et al. 
Immune evasion by oncogenic proteins of acute myeloid leukemia.
Blood
2014
, vol. 
123
 
10
(pg. 
1535
-
1543
)
10
Fiegler
 
N
Textor
 
S
Arnold
 
A
, et al. 
Downregulation of the activating NKp30 ligand B7-H6 by HDAC inhibitors impairs tumor cell recognition by NK cells.
Blood
2013
, vol. 
122
 
5
(pg. 
684
-
693
)
Sign in via your Institution