ArticlePDF AvailableLiterature Review

Targeted Elimination of Leukemia Stem Cells; a New Therapeutic Approach in Hemato-Oncology

Authors:

Abstract and Figures

Despite recent advances, treatment of leukemia is often not curative. New insights indicate that this may be attributable to a small population of therapy-resistant malignant cells with self-renewal capacity and the ability to generate large numbers of more differentiated leukemia cells. These leukemia-initiating cells are commonly referred to as Leukemia Stem Cells (LSCs). LSCs are regarded as the root of leukemia origin and leukemia recurrence after seemingly successful therapy. Not surprisingly therefore, contemporary leukemia research has focused on ways to specifically eliminate LSCs, leading to the identification of several promising anti-LSC strategies. Firstly, LSCs may be eliminated by antibody- or ligand-based cell surface delivery of therapeutics such as naked antibodies, immunotoxins, and immunocytokines. This approach exploits LSC-associated surface antigens, such as CD33, CD44, CD96, CD123 and CLL-1 for LSC-selective therapy and aims to spare normal hematopoietic stem cells. A second strategy aims to disrupt the interactions between LSCs and their highly specialized niche. These interactions appear to be pivotal for maintenance of the stem cell-like characteristics of LSCs. A third strategy centers on the selective modulation of aberrantly activated signaling pathways central to LSC biology. A fourth strategy, dubbed 'epigenetic reprogramming', aims to selectively reverse epigenetic alterations that are implicated in ontogeny and maintenance of LSCs. In this review, we will discuss the rationale for these LSCs-targeted strategies and highlight recent advances that may ultimately help pave the way towards selective LSCs-elimination.
Antibody-and ligand-based approaches for the elimination of LSCs. The cell surface molecules CD96, CD123 (IL3R), CLL-1, CD33 and TRAIL-R1/R2 may be exploited for the selective elimination of LSCs. CD123 targeted strategies include mAb7G3, a fusion protein comprising an CD123-specific antibody fragment linked to pseudomonas endotoxin A (26292(Fv)-PE38-KDEL) and an IL3: diphtheria toxin (IL-3:DT) fusion protein. CLL-1 has been targeted with naked antibodies resulting in Complement Dependent Cytotoxicity. CD33 has been targeted with several experimental therapeutics, including radionuclides, a scFvCD33: sTRAIL fusion protein and the clinically applied immunotoxin Gemtuzumab Ozogamicin (GO). After CD33-selective binding, GO internalizes and ends up in the lysosomes. In the acidic milieu of the lysosomes, the calicheamicin moiety is hydrolytically released. Next, calicheamicin translocates to the nucleus and intercalates with the DNA, resulting in apoptotic cell death. Therapeutic strategies targeting the death-receptors, TRAIL-R1/R2, include activating anti-TRAIL-R1 and anti-TRAIL-R2 antibodies, recombinant preparations of soluble TRAIL (sTRAIL) and scFv:sTRAIL fusion proteins. Activation of TRAIL-R1/-R2 receptors results in the recruitment of the adaptor molecule FADD and caspase-8 to the death domains of these receptors. Assembly of this so-called death-inducing signaling complex (DISC) leads to sequential activation of caspase-8 and caspase-3, ultimately resulting in DNA damage and apoptosis. In some cell types, caspase-8 can cleave Bid (tBid) and thereby activate a mitochondrial amplification loop. This loop is characterized by cytochrome C release from the mitochondria and subsequent the sequential activation of caspase-9 and caspase-3.
… 
Targeting aberrantly regulated pathways and epigenetic reprogramming Aberrantly regulated Wnt and Hedgehog pathways have been identified in LSCs and therefore might be suitable candidates for therapeutic intervention. At the cell surface, binding of Wnt to LRP5/6 and Frizzled destabilizes the Multiprotein Destruction Complex (MDC), which in the absence of Wnt signaling facilitates the degradation of -catenin. As a result of Wnt binding to its receptors, -catenin accumulates and translocates to the nucleus where it complexes with TCF, Bcl-9 and pygo to induce the expression of several genes including c-myc, cyclin-D and CD44. Extracellularly, the Wnt signaling pathway can be inhibited by the scavengers sFRP, Wif1 and Cerberus and the DKK-family members which prevent Wnt binding to its receptor. Intracellularly, Wnt signaling can be inhibited by the compounds CGP049090 and PKF115-584, which disrupt the TCF/-catenin complex. The Hedgehog pathway is initiated by binding of a ligand (Shh, Ihh or Dhh) to the receptor Patched. Hereupon, Smoothened becomes activated and via the Gli family of transcription factors induces the expression of several target genes including CyclinD/E and Bmi-1. The Hedgehog pathway can be inhibited by cyclopamine which stabilizes Smoothened in its inactive form. Binding of Growth factors (GF) to Growth Factor Receptors (GFR) or binding of VLA-4 to fibronectin induces activation of PI3K. This pro-survival signaling pathway is characterized by the conversion of PIP2 to PIP3 and subsequent downstream signaling via several proteins, including mTOR. Normally, PTEN functions as an inhibitor of the PI3K pathway. Pharmacologically, both rapamycin and arsenic trioxide (via PML) can alter mTOR activation. The enzymes ALDH1A1 and ALDH3A1 have been implicated in Multi Drug Resistance (MDR) and can be inhibited by DEAB and ATRA. The anti-leukemia activity of parthenolide (PTL) is characterized by the inhibition of NF-B and the generation of ROS. The compound TDZD-8 can also inhibit NF-B activation and might influence LSCs membrane integrity. Frequently, gene expression in leukemic cells is altered by epigenetic reprogramming. Therefore, reprogramming the epigenetic status using decitabine or HDACi appears promising for the elimination of LSCs.
… 
Content may be subject to copyright.
Current Drug Targets, 2010, 11, 95-110 95
1389-4501/10 $55.00+.00 © 2010 Bentham Science Publishers Ltd.
Targeted Elimination of Leukemia Stem Cells; a New Therapeutic
Approach in Hemato-Oncology
B. ten Cate1, M. de Bruyn1, Y. Wei1,2, E. Bremer1 and W. Helfrich*,1,2
1Department of Surgery, Surgical Research Laboratories, University Medical Center Groningen, University of
Groningen, Groningen, The Netherlands; 2Department of General Surgery, The First Clinical Hospital of Harbin
Medical University, Harbin, China
Abstract: Despite recent advances, treatment of leukemia is often not curative. New insights indicate that this may be
attributable to a small population of therapy-resistant malignant cells with self-renewal capacity and the ability to generate
large numbers of more differentiated leukemia cells. These leukemia-initiating cells are commonly referred to as
Leukemia Stem Cells (LSCs). LSCs are regarded as the root of leukemia origin and leukemia recurrence after seemingly
successful therapy. Not surprisingly therefore, contemporary leukemia research has focused on ways to specifically
eliminate LSCs, leading to the identification of several promising anti-LSC strategies. Firstly, LSCs may be eliminated by
antibody- or ligand-based cell surface delivery of therapeutics such as naked antibodies, immunotoxins, and
immunocytokines. This approach exploits LSC-associated surface antigens, such as CD33, CD44, CD96, CD123 and
CLL-1 for LSC-selective therapy and aims to spare normal hematopoietic stem cells. A second strategy aims to disrupt
the interactions between LSCs and their highly specialized niche. These interactions appear to be pivotal for maintenance
of the stem cell-like characteristics of LSCs. A third strategy centers on the selective modulation of aberrantly activated
signaling pathways central to LSC biology. A fourth strategy, dubbed ‘epigenetic reprogramming’, aims to selectively
reverse ep igenetic alterations that are implicat ed in ontogeny and maintenance of LSCs. In this review , we will discuss the
rationale for these LSCs-targeted strategies and highlight recent advances that may ultimately help pave the way towards
selective LSCs-elimination.
Keywords: Leukemia stem cells, AML, CML, targeting, apoptosis, antibody, epigenetics.
INTRODUCTION
To a varying degree most leukemia types initially
respond well to therapy with partial or even complete
remissions. Unfortunately, after a period of minimal residual
disease (MRD) many patients succumb to refractory relapses
of the disease. Recent insight indicates that the development
of these relapses may be due to the selective continued
survival of a small, but distinct, population of therapy-
resistant tumor-initiating cells, commonly referred to as
Leukemia Stem Cells (LSCs). LSCs are thought to originate
either from normal hematopoietic stem cells (HSCs) or from
more differentiated progenitor cells that have acquired
malignant features. In the latter case, the progenitor cells
have de-differentiated and re-acquired stem cell-like
characteristics via as yet undefined pathways [1, 2]. Both
LSCs and HSCs possess self-renewal capacity but are
relatively quiescent compared to more mature progenitors
cells (see F ig. 1). However, LSCs typically have a much
stronger capacity for cellular expansion than normal HSCs,
probably due to an increase in symmetric self-renewal
activity of LSCs [3].
LSCs were first identified in Acute Myeloid Leukemia
(AML) by John Dick and colleagues [3, 4] and have by now
been found in all major forms of leukemia [3-6]. LSCs are
*Address correspondence to this author at the Department of Surgery,
Surgical Research Laboratories, University Medical Center Groningen,
University of Groningen, Groningen, The Netherlands; Hanzeplein 1, BA44,
9713 GZ Groningen, The Netherlands; Tel:+31 50 361 3733; Fax:+ 31 50
36 32 796; E-mail: w.helfrich@med.umcg.nl
experimentally defined as cells capable of serial engraftment
in immunocompromised mice (see Fig. 1). Importantly, mice
engrafted with these LSCs develop leukemia which resem-
bles the patient’s original disease [7]. During this last decade
substantial progress in the field of LSC research has led to
the identification of many of the hallmark characteristics of
LSCs. Therapy-resistance is typical for LSCs which is
thought to arise from (1) their relative quiescent cellular
activity [8, 9], (2) their array of self-protecting mechanisms
[10], and (3) the highly protective microenvironment in
which the LSCs resides [11].
Since curative treatment of leukemia will most likely
require th e elimination of LSCs, new strategies need to be
designed that overcome the above-described features.
Importantly, in this design care should be taken to spare
normal HSCs. In this respect, strategies that target a distinct
Achilles' heel of the LSC biology appear to be particularly
promising. Here, we review the rationale of different LSCs-
specific strategies and highlight recent advances that may
ultimately lead to selective LSCs-elimination.
LSC CELL SURFACE TARGETING APPROACHES
Cell surface antigens expressed on LSCs may be exp-
loited for antibody- or ligand-based therapeutic approaches.
The ideal target antigen is exclusively and abundantly
expressed on LSCs, thereby maximizing the therapeutic
effect and minimizing off-target effects. Unfortunately, such
an antigen has not been uncovered. However, a series of
surrogate cell surface mark ers have been identified which
have favorable LSCs-restricted expression profiles. These
96 Current Drug Targets, 2010, Vol. 11, No. 1 ten Cate et al.
surrogate markers can be targeted with naked antibodies,
which typically eliminate targeted cells via Antibody
Dependent Cellular Cytotoxicity (ADCC) or Complement
Dependent Cytotoxicity (CDC). However, some antibodies
can directly interfere with the signaling activity by binding to
receptor molecules and sensitize targeted cells to apoptosis
induction [12] or directly induce apoptosis as reported for the
anti-CD20 monoclonal antibody (mAb) Rituximab [13]. The
tumoricidal potential of antibodies can be further enhanced
by conjugating them to cytotoxic drugs or radionuclides.
Alternatively, cell surface molecules may also be targeted
using recombinant forms of their corresponding natural
ligand. Ligand-based approaches have been used to deliver
cytotoxic drugs to the target cell and to trigger apoptosis-
inducing receptors. However, an obvious potential limitation
to antibody-based and ligand-based therapy is the selective
loss of the target antigen during therapy or the presence of
target antigen-negative leukemia cells at the start of therapy.
In this respect, fluctuations in the expression of cell surface
markers on HSCs have been reported [14]. Therefore, the
rational design of combinatorial antibody-based or ligand-
based strategies that target two or more different LSC anti-
gens simultaneously may prove the best way to prevent
escape from therapy. Below, a selection of the most promi-
nent cell surface molecules on LSCs and approaches for
targeting these molecules are briefly reviewed (summarized
in Fig. 2).
LSC Target Antigens
CD33 belongs to immunoglobulin (Ig) superfamily and is
a member of the sialoadhesin family of cellular interaction
Fig. (1). Hypotheses for the ontogeny of leukemia stem cells. LSCs can arise by somatic mutations in normal hematopoietic stem cells
(HSC) or more mature progenitor cells can become malignant, de-differentiate and re-acquire stem cell-lik e characteristics. Currently, LSCs
(typically CD34+CD38-) are defined by the ability to engraft in immunocompromised mice and initiate leukemia resembling the original
patient’s diseas e.
Targeted Elimination of Leukemia Stem Cells Current Drug Targets, 2010, Vo l. 11, No. 1 97
molecules [15, 16]. The intracellular domain of CD33
contains an immuno-receptor tyrosine-based inhibitory motif
(ITIM) domain which may function as a negative regulator
of cellular differentiation [17]. CD33 expression is largely
restricted to the myeloid lineage, however some expression
is detected on certain peripheral blood lymphocytes. In
leukemia, CD33 is expressed on blasts in the majority of
AML patients. Importantly, it has been noted that CD33 is
also expressed on AML-LSCs, but not on normal HSCs [18,
19].
CD123 (IL-3R) is the alpha subunit that hetero-
dimerizes with the IL3 chain to form the interleukin-3
receptor. CD123 is constitutively expressed on hematopoie-
tic progenitors cells (HPCs), on which it is involved in
proliferation and differentiation. Furthermore, CD123 is
expressed on LSCs in AML [20], Chronic Myeloid Leuke-
mia (CML), Myelodysplastic Syndrome (MDS) and
Systemic Mastocytosis [18], but not on HSCs [21]. This
expression profile indicates that CD123 might be a suitable
target for the selective elimination of LSCs in various
hematological malignancies.
C-type Lectin-Like Molecule-1 (CLL-1) is a heavily N-
glycosylated transmembrane protein with an intracellular
ITIM domain [22]. CLL-1 is exclusively expressed on cells
of the hematopoietic lineage, including dendritic cells,
granulocytes and monocytes [23]. In leukemia, CLL-1
expression is detected on 92% of all AML cases [23] on
which a high expression at diagnosis correlates with an early
relapse [24]. Importantly, CLL-1 is also expressed on AML-
LSCs, but not on HSCs [24, 25]. In line with these findings,
Fig. (2). Antibody- and ligand-based approaches for the elimination of LSCs. The cell surface molecules CD96, CD123 (IL3R), CLL-1,
CD33 and TRAIL-R1/R2 may be exploited for the selective elimination of LSCs. CD123 targeted strategies include mAb7G3, a fusion
protein comprising an CD123-specific antibody fragment linked to pseudomonas endotoxin A (26292(Fv)-PE38-KDEL) and an IL3:
diphtheria toxin (IL-3:DT) fusion protein. CLL-1 has been targeted with naked antibodies resulting in Complement Dependent Cytotoxicity.
CD33 has been targeted with several experimental therapeutics, including radionuclides, a scFvCD33: sTRAIL fusion protein and the
clinically applied immunotoxin Gemtuzumab Ozogamicin (GO). After CD33-selective binding, GO internalizes and ends up in the
lysosomes. In the acidic milieu of the lysosomes, the calicheamicin moiety is hydrolytically released. Next, calicheamicin translocates to the
nucleus and intercalates with the DNA, resulting in apoptotic cell death. Therapeutic strategies targeting the death-receptors, TRAIL-R1/R2,
include activating anti-TRAIL-R1 and anti-TRAIL-R2 antibodies, recombinant preparations of soluble TRAIL (sTRAIL) and scFv:sTRAIL
fusion proteins. Activation of TRAIL-R1/-R2 receptors results in the recruitment of the adaptor molecule FADD and caspase-8 to the death
domains of these receptors. Assembly of this so-called death-inducing signaling complex (DISC) leads to sequential activation of caspase-8
and caspase-3, ultimately resulting in DNA damage and apoptosis. In some cell types, caspase-8 can cleave Bid (tBid) and thereby activate a
mitochondrial amplification loop. This loop is characterized by cytochrome C release from the mitochondria and subsequent the sequential
activation of caspase-9 and caspase-3.
98 Current Drug Targets, 2010, Vol. 11, N o. 1 ten Cate et al.
it was demonstrated that sorted CD34+/CLL-1+ AML cells
are able to engraft and generate CLL-1+ blasts after trans-
plantation in Non-Obese Diabetic/Severe Combined
Immuno-Deficient (NOD/SCID) mice [24].
CD96 is a transmembrane protein belonging to the Ig
super-family. The extracellular domain of CD96 has three
Ig-like regions and a stalk-like region which can be heavily
O-glycosylated. The only ligand for CD96 identified thus far
is CD155, also known as polio virus receptor [26]. On
normal cells CD96 protein expression appears to be
restricted to T and NK cells, while its expression was also
detected on T cell -Acute Lymphoid Leukemia (ALL) cells
and AML cells [27, 28]. Recently, Hosen and colleagues
identified that comp ared with normal HSCs in AML-LSCs
CD96 mRNA levels are at least 200-fold increased [28].
These mRNA expression data suggests that CD96 protein
may be selectively over-expressed on AML-LSCs. In line
with this finding, they demonstrated that CD96+ AML cells
have a significantly higher engraftment potential then CD96-
AML cells [28]. Therefore, the development of antibody-
based or CD96 ligand-based therapeutics may have potential
for the targeted elimination of CD96+ AML-LSCs.
Targeting LSCs with Naked Antibodies
Indications for the efficacy of targeting CD123+ AML-
LSCs with naked antibodies is demonstrated by a preclinical
study using the CD123 neutralizing antibody, mAb7G3 [29].
Treatment with mAb7G3 inhibited engraftment and homing
of human CD123+ AML-LSCs when injected in NOD/SCID
mice. This effect of mAb7G3 was restricted to AML cells,
since there was only a minor effect towards normal bone
marrow (BM) cells. Furthermore, mAb7G3 treatment of
NOD/SCID mice with pre-established AML reduced both
the disease burden and the ability to transplant AML in
secondary recipient mice (ASH Annual Meeting Abstracts,
20071).
Recently, in vitro treatment with naked anti-CLL-1 mAbs
in the presen ce of serum resulted in efficient Complement
Dependent Cytotoxicity (CDC) towards various CLL-1+
AML cell lines. In lin e with these findings, ex vivo treatment
of primary AML blasts with naked anti-CLL1 mAbs resulted
in CDC in all samples tested. Furthermore, in a xenograft
model one particular anti-CLL-1 mAb reduced the growth of
established tumors by 38% (ASH annual meeting abstracts,
20082). Together, these data indicate that targeting both
CD123 and CLL-1 by naked antibodies might be a promising
approach for the selective elimination of LSCs.
Targeting LSCs with Immunoconjugates
The expression of CD33 in AML has resulted in the
development of several CD33-targeted experimental
therapeutics [30, 31] and the clinically applied anti-AML
1 Lock R, Jin L, Lee E, Ramshaw HS, Busfield S, Peoppl AG, et al. CD123 (IL-3
Receptor alpha Chain) Neutralization by a monoclonal antibody selectively eliminates
human acute myeloid leukemic stem cells. ASH Annual Meeting Abstracts 2007; 110:
161.
2 Korver W, Zhao X, Singh S, Pardoux C, Zhao C, Sen S, et al. CLL-1, a receptor
expressed on myeloid leukemic stem cells, is a potential target for Immunotherapy of
AML. ASH Annual Meeting Abstracts 2008; 112: 4003.
immunotoxin Gemtuzumab Ozogamicin (GO). GO consists
of an anti-CD33 mAb that is chemically coupled to a
derivative of the calicheamicintoxin. After CD33-selective
binding, GO internalizes and ends up in the acidic milieu of
lysosomes where the calicheamicin moiety is hydrolytically
released. Subsequently, calicheamicin translocates to the
nucleus and intercalates with the DNA, causing site-specific
double-strand breaks resulting in apoptotic cell death [32]. In
the United States, GO was approved by the FDA for use in
patients over the age of 60 with relapsed AML who are not
considered candidates for standard chemotherapy [33]. In
this subset of patients, GO treatment resulted in a 26%
remission rate, albeit with sometimes considerable tox icity
[34]. Recently, we have demonstrated that the activity of GO
is strongly enhanced when treatment is combined with the
epigenetic modifying drug valproic acid (VPA), a histone
deacetylase inhibitor (HDACi). The co-treatment of AML
cells with GO and VPA f acilitated the interca-lation o f
calicheamicin with the DNA, followed by synergistic
apoptosis induction [35].
It has been reported that in vitro treatment of AML-LSCs
with GO results in a median decrease in cellular viability
with 46%, while normal HSCs were largely unaffected (ASH
Annual Meeting Abstract, 20073). Therefore, it would be
interesting to assess whether the anti-LSCs activ ity of GO
can be further enhanced when treatment is combined with
VPA.
One potential resistance mechanism of AML-LSCs
towards treatment with GO involves the increased expression
and/or activity of drug efflux pumps. In this respect, it
appears that P-glycoprotein (Pgp) expression confers resis-
tance to GO and as such is asso ciated with a worse clinical
response [36]. Therefore, combination treatment of GO and
Pgp inhibitors may enhance the therapeutic effect of GO
towards Pgp-expressing LSCs.
Alternatively, efflux pump-related resistance in AML-
LSCs may be overcome using an anti-CD33 antibody conju-
gated to cytolytic radionuclides like 111In. In vitro expe-
riments w ith an 111In-conjugated anti-CD33 mAb resul-ted in
potent apoptosis in both chemo-resistant CD33+, HL-60
AML cells and in primary AML cells. Indeed, efflux pump-
related resistance was overcome by 111In since these AML
cells expressed several drug efflux pumps including Pgp,
Breast Cancer Resistance Protein 1 and Multidrug Resistance
Protein 1 [37].
Currently, the efficacy of various CD123-targeted immu-
noconjugates is under investigation. In one particular
approach CD123 is targeted by a fusion protein comprising a
CD123-specific single chain fragment of the variable regions
(scFv) antibody fragment that is genetically fused to a
fragment of pseudomonas exotoxin A (PE38). This fusion
protein, designated 26292(Fv)-PE38-KDEL, potently ind-
uced apoptosis in a variety of CD123+ leukemia cell lines
[38]. It remains to be investigated whether 26292(Fv)-PE38-
KDEL can also eliminate CD123+ LSCs.
3 Jawad M, Mony U, Russell N, Pallis M. In vitro chemosensitivity of leukaemic stem
and progenitor cells to gemtuzumab ozogamicin (mylotarg) in AML. ASH Annual
Meeting Abstracts 2007; 110: 650
Targeted Elimination of Leukemia Stem Cells Current Drug Targets, 2010, Vo l. 11, No. 1 99
Targeting LSCs with Ligand-Based Approaches
The cytokine IL3 has been used in fusion protein format
to selectively deliver diphtheria toxin (DT) to CD123+ AML
cells [39]. This IL3-DT fusion protein also selectively
eliminated primitive A ML progenitors, while normal
progenitors where not affected [40]. Importantly, the IL3-DT
fusion protein inhibited engraftment of human CD123+
AML-LSC cells when injected in NOD/SCID mice [40, 41].
Recently, it was demonstrated that the pro-apoptotic activity
of IL3-DT is enhanced when treatment is combined with
Tumor necrosis factor Related Apoptosis Inducing Ligand
(TRAIL) [42]. Currently, a phase I clinical trial with this
IL3-DT fusion protein for a subset of AML and MDS
patients is initiated [41].
Activation of Apoptosis Inducing Receptors
The agonistic TRAIL death receptors, TRAIL-R1 and
TRAIL-R2 (also known as DR4 and DR5, respectively)
belong to the Tumor Necrosis Factor receptor family of
death receptors. Ligation of these death receptors by TRAIL
activates the extrinsic apoptotic pathway (reviewed in [43]).
Hereupon, the adaptor protein FAS associated death domain
(FADD) and the initiator caspase-8 are recruited to the
intracellular death domains of these receptors. Assembly of
this so-called death-inducing signaling complex (DISC)
leads to sequential activation of initiator and effector
caspases and, ultimately, results in apoptotic cell death. In
some cell types, the death-receptor pathway relies on an
additional mitochondrial amplification loop that is activated
by caspase-8-mediated cleavage of the BH3-only interacting
domain death agonist (Bid) to a truncated form. Next,
truncated BID (tBid) activates the mitochondrial pathway
which involves cytochrome C and caspase-9. Intriguingly,
apoptosis resulting from ligation of agonistic TRAIL-Rs
appears to be restricted to malignant cells only, since normal
cells appear to be resistant to TRAIL-R ligation. The mode
of action of this tumor-selectivity of TRAIL-Rs ligation is
still enigmatic.
Currently, activating anti-TRAIL-R1 and anti-TRAIL-R2
antibodies, as well as recombinant preparations of solub le
TRAIL (sTRAIL) are evaluated in clinical trials [44-47]. In
our laboratory, we have developed a series of fusion
proteins, consisting of a tumor-selective scFv antibody
fragment genetically fused to sTRAIL. These scFv:sTRAIL
fusion proteins are designed to enhance tumor-selective cell
surface delivery of sTRAIL (reviewed in [48]). Pre-
clinically, these scFv:sTRAIL fusion proteins demons-trate
potent and target antigen-restricted apoptosis induction. To
this end, we have exploited both antigens expressed on solid
tumors including Epidermal Growth Factor Receptor [49-51]
and Epithelial Cell Adhesion Molecule [52, 53], as well as
antigens selectiv ely expressed on leukemia cells including
CD7 [54], CD19 [55] and CD33 [30].
Very recently, the expression of both TRAIL-R1 and
TRAIL-R2 has been reported on LSCs. Comparing the gene
expression profiles of AML-LSCs and HSCs, Majeti and
colleagues demonstrated an increased expression of TRAIL-
R1 and TRAIL-R2 in LSCs [56]. Furthermore, Yong et al.
demonstrated that in CML non-cycling CD34+ cells have a
significant higher expression of TRAIL-R1 and TRAIL-R2
than cycling CD34+ CML cells [57]. In line with this finding,
sTRAIL significantly inhibited the clonogenic capacity of
CD34+ AML cells without affecting the clonogenic capacity
of normal CD34+ cells [58]. Together, these studies indicate
that TRAIL-R1 and TRAIL-R2 receptors might be suitable
candidates for the selective elimination of LSCs by targeted
apoptosis induction. At present, we are assessing the efficacy
of CD33- and CLL-1-selective scFv:sTRAIL fusion proteins
for targeted apoptosis induction in AML-LSCs.
DISRUPTION OF THE LSCS-NICHE INTERACTION
In 1978, Schofield proposed that HSCs receive
regulatory factors and signals within a specialized niche [59].
Currently, there are indications that these specialized niches
are located in the bone marrow (BM), near the endosteum
[60]. The endosteum is the inner surface located at the bone-
BM interface and consisting mainly of osteoblasts and
osteoclasts. More recent evidence indicates that also vascular
and perivascular cells appear to contribute to these niche
interactions as well (see review [61]). Signals within the
HSCs nich e regulate the normal survival, proliferation and
differentiation of the HSCs. Recently, it was demonstrated
that in leukemia the micromilieu in the HSCs niche appears
to disturbed by the presence of Stem Cell Factor that is
locally secreted by leukemia cells [62]. Furthermore, it has
been demonstrated that intravenously injected AML-LSCs
home to, engraft and subsequently reside in the endosteal
region [63]. Such cellular tropism of AML-LSCs for a
dedicate niche typically involves the coordinated signaling
by various chemokines and adhesion molecules. Neutraliza-
tion or inhibition of these signals may be exploited for the
selective elimination of LSCs. Below, a selection of recent
studies is reviewed that collectively indicate that disruption
of the LSCs-niche interactions may indeed form a
therapeutic option for the elimination of LSCs (summarized
in Fig. 3).
Targeting the Stroma-Derived Factor-1(SDF-1)/CXCR4
Axis
Previously, it has been demonstrated that the chemokine
SDF-1 (CXCL12) as expressed by reticular stromal cells in
the BM is pivotal for homing of HSCs to their niche [64].
SDF-1 binds to the transmembrane receptor CXCR4 which
is expressed on HSCs [64, 65], leukemic blasts and AML-
LSCs [66]. Of note, a high expression of CXCR4 in AML
correlates with a negative prognosis [67, 68]. The pivotal
role of CXCR4 in retention of HSCs in the niche is clearly
demonstrated by the mobilization of HSCs by the CXCR4
antagonist AMD3100 [69].
The interaction of leukemia cells with cell adhesion
molecules in the niche typically reduces their sensitivity to
cytotoxic therapies, a process known as cell adhesion-
mediated drug resistance (CAM-DR). Inhibition of CXCR4
by the specific inhibitors AMD3100 and AMD3465 sensi-
tized AML cells for cytotoxic therapies [70-72]. Most likely,
treatment with the CXCR4 inhibitors mobilizes AML cells,
thereby reducing CAM-DR and sensitizing the AML cells
for cytotoxic therapy.
100 Current Drug Targets, 2010, Vol. 11, No. 1 ten Cate et al.
Tavor and colleagues demonstrated that the anti-CXCR4
mAb12G5 significantly inhibits homing of AML cells to the
BM of recipient mice. Notably, treatment of mice with
established AML with mAb12G5 reduced the leukemic
burden in the BM, blood and spleen. Importantly, similar
treatment did not affect the levels of engrafted HPCs [66].
Additionally, lowering available SDF-1 levels in leuke-
mia might offer a therapeutic strategy. Recently, the medi-
cinal herb-derived compound berberine was shown to inhibit
SDF-1 production by stromal cells, thereby impairing the
migration of AML blasts and AML-LSCs in vitro [73].
Together these results indicate the importance of a properly
functioning SDF-1/CXCR4 axis for homing and retention of
AML blasts and AML-LSCs. Therefore, the SDF-1/CXCR4
axis appears to be a suitable targ et candidate for the selective
elimination of A ML-LSCs.
Targeting Very Late Antigen 4 (VLA-4)
The 1-integr in, VLA-4, has a vital role in the retention
of HPCs in the BM niche by immobilizing these cells [74].
The interaction of VLA-4 on AML cells with fibronectin on
stromal cells initiates the phosphatidylinositol-3-OH kinase
(PI3K) pro-survival pathway. Activation of the PI3K
pathway in AML cells contributes to CAM-DR, which might
explain the observation that VLA-4 expression on AML cells
is correlated with a negative prognosis for AML patients. In
line with this, Matsunaga and colleagues demonstrated that
inhibition of VLA-4 interaction with fibronectin sensitized
AML cells to cytosine arabinoside (Ara-C). This was
demonstrated in a mice model for MRD in which they
applied VLA-4 neutralizing antibodies to prevent the
interaction with fibronectin [75]. Using the same MRD
model, they recently demonstrated that a fibronectin-based
synthetic peptide, designated FNIII14 [76], also sensitized
AML cells to Ara-C [77]. Both approaches may be of
considerable interest for disruption of the LSCs-niche
interaction.
Targeting CD44
CD44 is a transmembrane receptor capable of binding
several ligands, including hyaluronic acid which is abun-
dantly expressed in the endosteal region [78]. On HSCs,
CD44 serves as a homing receptor by interacting with E-
selectin and L-selectin expressed in the BM [79]. Since
CD44 is expressed on both CML-LSCs and AML-LSCs [80,
Fig. (3). D isruption of the LSC-niche interaction. The LSC-niche interaction can be disrupted by targeting VLA-4, CXCR4, CD44 and
Rac1. Binding of LSCs-expressed VLA-4 to fibronectin in the BM initiates the PI3K pro-survival pathway and cell adhesion-mediated drug
resistance (CAM-DR). The VLA-4-fibronectin interaction has been disrupted by mAbs and the synthetic peptide FNIII14. CXCR4 has been
targeted with mAb12G5 and the specific inhibitors AMD3100 and AMD3465. Furthermore, the concentration of the CXCR4 ligand, SDF-1,
can be lowered using berberine. The interaction of CD44 with the niche has been disrupted by mAbH90 and mAbIM7. The compound,
NSC23766, has been utilized to specifically inhibit Rac1.
Targeted Elimination of Leukemia Stem Cells Current Drug Targets, 2010, Vo l. 11, No. 1 101
81], neutralization of CD44-binding might impair the
homing and retention of LSCs in the BM.
In CML, about 95% of the cases is characterized by the
presence of the oncogenic fusion protein Bcr-Abl, a con-
stitutively active mutant tyrosine kinase that causes
uncontrolled cellular proliferation. The Bcr-Abl inhibitor
Imatinib Mesylate induces remissions in the majority of
CML patients. However, Imatinib appears to be unable to
eradicate quiescent CML-LSCs [82], which might explain
why the majority of CML patients relapse upon treatment
discontinuation. Therefore, novel therapeutics for the
selective elimination of Bcr-Abl+ CML-LSCs are needed.
In a series of elegant exper iments, Krause and colleagues
demonstrated that CD44 expressed on Bcr-Abl+ CML-LSCs
is essential for homing of these cells in recipient mice.
Thereto, they performed a competitive homing assay in
which the relative contribution of Bcr-Abl-transduced
CD44+ BM cells and Bcr-Abl-transduced CD44- BM cells in
the development of CML was analyzed. Both cells types
were mixed and injected intravenously in irradiated
recipients mice. After CML development, the animals were
sacrificed and the relative contribution of the two distinct
cell populations was analyzed. This revealed that Bcr-Abl-
transduced CD44+ BM cells contributed 5 times more to the
development of CML than Bcr-Abl-transduced CD44- BM
cells. CML development by the two different cell types was
similar when injected intrafemorally. These results indicate
that CD44 is necessary for homing of intravenously injected
Bcr-Abl transduced cells to th e BM. Importantly, anti-CD4 4
antibody treatment significantly prolonged survival of
NOD/SCID recipient mice upon transplantation with CML-
LSCs [80].
Simultaneously, the group of John Dick obtained similar
results by targeting CD44 in AML. Treatment with the
CD44-activating mAbH90 inhibited AML-LSCs homing to
the BM niche and altered the fate of the AML-LSCs
resulting in an efficient and selective elimination of th e
AML-LSCs [81]. Together, these studies clearly indicate a
rationale for targeting CD44 to selectively eliminate CML-
LSCs and AML-LSCs. Therefore, an evaluation of the
efficacy of targeting CD44 in both CML and AML patients
seems warranted.
Targeting Rac
The Rac subfamily of the Rho-family of GTP-ases is
implicated in cell adhesion and motility. The family consists
of three members Rac1, Rac2 and Rac3. Rac1 is ubiquitously
expressed [83], while Rac2 expression is limited to the
hematopoietic system [84] and Rac3 is selectively expressed
in the brain [85]. Rac1-/- HSCs and progenitors are unable to
reconstitute hematopoiese in irradiated recipient mice upon
transplantation [86]. Moreover, it has been shown that Rac is
overexpressed on CD34+ cells derived from AML patients.
Rac appears to contribute to an enhanced migration and
adhesion capacity of CD34+ cells to BM stromal cells.
Gao and colleagues [87] generated a Rac1 inhibitor,
designated NSC23766, that significantly delayed leukemia
development in a murine tumor model [88] and strongly
reduced the clonal expansion of CD34+ AML cells [89].
Together, these promising data warrants further pre-clinical
assessment of Rac1 inhibition for the elimination of LSCs.
TARGETING ABERRANT MOLECULAR PATH-
WAYS
Both, HSCs and LSCs rely on similar molecular
pathways for their maintenance and self-renewal. However,
in LSCs these pathways are often aberrantly regulated. LSCs
appear to be particular dependent on these deregulated
pathways to maintain their malignant phenotype. This strong
dependence indicates that therapeutic intervention in these
pathways might be exploitab le for the selectiv e elimination
of LSCs. Thus far, several of these deregulated pathways
have been identified (e.g. the Hedgehog and Wnt pathways).
The selective targeting of these pathways may yield
promising anti-LSCs activity.
Compounds such as arsenic trioxide and parthenolide
have been assessed for their capacity to induce apoptosis in
LSCs. However, the pathways in which these compounds
intervene are currently not completely understood. There-
fore, further delineation of their specific molecular targets
may contribute to development of more and rationally
designed LSCs targeted therapeutics. Below, we provide a
selection of aberrantly regulated pathways present in LSCs
and a selection of chemical compounds that may have
potential for the elimination of LSCs (summarized in Fig. 4).
The Hedgehog (Hh) Pathway
The Hh pathway is involved in HSCs self-renewal [90]
and is initiated by binding of one of the three Hh ligands,
Sonic Hedgehog (Shh), Indian Hedgehog (Ihh) or desert
Hedgehog (Dhh) to the transmembrane receptor Patched.
Upon binding of Hh to Patched the inhibiting effect of
Patched on the transmembrane receptor Smoothened (Smo)
is relieved (reviewed in [91]). Next, Smo can activate Hh
targets g enes via the Gli family of transcription factors.
Amongst these Hh targets genes are CyclinD/E [92] and B-
cell-specific Moloney murine leukemia virus insertion site 1
(Bmi-1) [93] which are involved in cell proliferation and
self-renewal, respectively. The Hh pathway can be inhibited
by the chemical compound cyclopamine, which stabilizes
Smo in its inactive form [94].
In a very recent study conducted by Zhao and colleagues
the role of Smo in Bcr-Abl+ CML-LSCs was analyzed [95].
Thereto, Bcr-Abl-transduced normal HPCs and Bcr-Abl-
transduced Smo-/- HPCs were transplanted in irradiated
recipient mice after which the development of CML was
compared. More than 90% of the mice transplanted with
Bcr-Abl-transduced normal HPCs developed CML. In
contrast, only 47% of the mice developed CML when
transplanted with Bcr-Abl-transduced Smo-/- HPCs. When
the Bcr-Abl-transduced HSCs were transplanted in mice, all
recipient mice died from CML within four weeks. Howev er,
when these transplanted mice were treated with cyclo-
pamine, 60% of the animals were still alive after 7 weeks.
Furthermore, CML cells freshly isolated from cyclopamine-
treated mice were unable to effectively repopulate secondary
recipients. This corroborates with the strong reduction (up to
14-fold) of CML-LSCs in cyclopamine-treated mice.
Addition ally, cyclopamine treatment of human CML cell
102 Current Drug Targets, 2010, Vol. 11, No. 1 ten Cate et al.
lines and primary CML samples reduced their clonogenic
capacity. Importantly, the authors indicate that Smo inhibi-
tion by cyclopamine may also impaired the propagation of
Imatinib-resistant human CML cells in xenografts [95].
The Wnt pathway
Wnt is implicated in HSCs self-renewal and prolif eration
[96] and is overexpressed in many different types of human
cancers [97]. Thus far, three distinct Wnt signaling pathways
have been described of which the canonical pathway is most
prominently involved in leukemia. The key mediator of the
canonical pathway is -catenin. Normally, -catenin is deg-
raded rapidly after synthesis by the multiprotein destruction
complex (MDC), which tags -catenin for proteasomal deg-
radation. The MDC consists of axin, adenomatous polyposis
coli (APC) and glycogen-synthase kinase 3 (GSK3). Upon
binding of Wnt to its receptors Frizzled and LRP5/6, the
MDC becomes destabilized and therefore unable to tag -
catenin for degradation. Consequently, -catenin accumu-
lates and is able to translocate to the nucleus. In the nucleus
-catenin complexes with T-cell factor (TCF), B cell
Fig. (4). Targeting aberrantly regulated pathways and epigenetic reprogramming Aberrantly regulated Wnt and Hedgehog pathways
have been identified in LSCs and therefore might be suitable candidates for therapeutic intervention. At the cell surface, binding of Wnt to
LRP5/6 and Frizzled destabilizes the Multiprotein Destruction Complex (MDC), which in the absence of Wnt signaling facilitates the
degradation of -catenin. As a result of Wnt binding to its receptors, -catenin accumulates and translocates to the nucleus where it
complexes with TCF, Bcl-9 and pygo to induce the expression of several genes including c-myc, cyclin-D and CD44. Extracellularly, the
Wnt signaling pathway can be inhibited by the scavengers sFRP, Wif1 and Cerberus and the DKK-family members which prevent Wnt
binding to its receptor. Intracellularly, Wnt signaling can be inhibited by the compounds CGP049090 and PKF115-584, which disrupt the
TCF/-catenin complex. The Hedgehog pathway is initiated by binding of a ligand (Shh, Ihh or Dhh) to the receptor Patched. Hereupon,
Smoothened becomes activated and via the Gli family of transcription factors induces the expression of several target genes including
CyclinD/E and Bmi-1. The Hedgehog pathway can be inhibited by cyclopamine which stabilizes Smoothened in its inactive form. Binding of
Growth factors (GF) to Growth Factor Receptors (GFR) or binding of VLA-4 to fibronectin induces activation of PI3K. This pro-survival
signaling pathway is characterized by the conversion of PIP2 to PIP3 and subsequent downstream signaling via several proteins, including
mTOR. Normally, PTEN functions as an inhibitor of the PI3K pathway. Pharmacologically, both rapamycin and arsenic trioxide (via PML)
can alter mTOR activ ation. The enzymes ALDH1A1 and ALDH3A1 have been implicated in Multi D rug Resistance (MDR) and can be
inhibited by DEAB and ATRA. The anti-leukemia activity of parthenolide (PTL) is characterized by the inhibition of NF-B and the
generation of ROS. The compound TDZD-8 can also inhibit NF-B activation and might influence LSCs membrane integrity. Frequently,
gene expression in leukemic cells is altered by epigenetic reprogramming. Therefore, reprogramming the epigenetic status using decitabine
or HDACi appears promising for the elimination of LSCs.
Targeted Elimination of Leukemia Stem Cells Current Drug Targets, 2010, Vo l. 11, No. 1 103
lymphoma 9 (Bcl-9) and pygo to induce TCF target genes
expression [98], including the oncogenes C-MYC [99],
Cyclin-D1 [100] and the cell surface antigen CD44 [101].
In AML, several oncogenic fusion proteins, such as
AML-1⁄eight twenty one (AML1-ETO), promyelocytic
leukemia ⁄ retinoic acid receptor alpha (PML⁄RAR) and
promyelocytic leukemia zinc finger (PLZF⁄RAR) [102] can
activate the Wnt signaling pathway. Furthermore, evidence is
emerging that indicate an aberrant Wnt signaling pathway in
LSCs [56] (extensively reviewed in ref. [103]). In this
respect, it has been shown that -catenin plays a pivotal role
in self-renewal of both HSCs as well as CML-LSCs [104].
Furthermore, very recently -catenin has been implicated in
the survival of Imatinib resistant CML-LSCs in Bcr-Abl
induced CML [105]. Therefore, modulation of Wnt signaling
in the LSCs may contribute to their selective elimination.
There are several possibilities to inhibit the Wnt
signaling pathway. Wnt signaling can be inhibited by
extracellular proteins (reviewed in [106]) which scavenge
Wnt and thereby prevent Wnt from binding to its receptors.
These Wnt binding proteins include secreted Frizzled-related
proteins (sFRP), Wnt inhibitory factor 1 (wif-1) and
Cerberus. Moreover, members of the Dickkopf (DKK)
family can directly bind the Wnt receptor and thereby
prevent Wnt-induced signaling. In this respect, very recently
it appeared that the anti-proliferative effects of human
mesenchymal stem cells towards CML cells is at least partly
attributable to secretion of DKK-1[107]. Additionally, also
very recently two natural compounds, CGP049090 and PKF
115-584 [108], have been identified that disrupt the TCF/-
catenin complex. In AML these compounds possess potent
tumoricidal activity [109]. It would be worthwhile to assess
the possibilities of inhibition of the Wnt signaling pathway
for the elimination of LSCs.
Aldehyde Dehydrogenases (ALDH) Enzymes
ALDH are a group of cytosolic enzymes that catalyzes
the conversion of intracellular aldehydes. The ALDH
superfamily consists of seventeen members [110] of which
two members, cytosolic aldehyde dehydrogenase class-1A1
(ALDH1A1) and class-3A1 (ALDH3A1), have been impli-
cated in tumor drug resistance against oxazaphosphorines
(e.g. cyclophosphamide) [111]. ALDH enzyme activity,
including the activity of ALDH1A1 and ALDH3A1, can be
assessed using activatable fluorescent substrates. ALDH
enzyme activity measurements enables the identification and
isolation of HSCs [112], breast cancer stem cells [113], liver
cancer stem cells [114] and AML-LSCs [115]. Manipulation
of ALDH1A1 and ALDH3A1 expression or activity may be
used to sensitize AML-LSCs to chemotherapy, especially to
oxazaphosphorines. In this respect, the ALDH1 inhibitor,
diethylaminobenzaldehyde (DEAB) might be utilized for
sensitizing AML-LSCs to treatment with chemotherapeutic
agents. Unfortunately, DEAB appeared to b e very instable in
vivo [116]. Therefore, the development of novel DEAB-
analogues with enhanced stability is warranted. All-trans
Retinoic Acid (ATRA), an established clinically applied
anti-leukemia agent, appears to downregulate both
ALDH1A1 and ALDH3A1 which may be beneficial for the
selective elimination of LSCs [111]. In this respect,
treatment of Acute Promyelocytic Leukemia (APL) and
AML cells with ATRA resulted in differentiation of tumor
cells and stimulation of cell cycling [117]. Therefore, we
suggest that the combination treatment of oxazaphosphorines
and ATRA may have potential for the elimination of LSCs.
Phosphatase and Tensin Homologue (PTEN)
PTEN is the second most commonly mutated gene in
human malignancies, including hematopoietic malignancies
[118, 119]. Normally, PTEN inhibits cell proliferation and
cell survival by negatively regulating the PI3K pathway
[120]. In this respect, Yilmaz and colleagues recently
compared th e effect of PTEN inactivation in HSCs and LSCs
[121]. Conditional deletion of PTEN resulted in the
development of myeloproliferative disorder in 17 out of 19
mice which in the majority culminated in leukemia. In th e
HSCs, deletion of PTEN was characterized by an elevated
cell cycle progression and an impaired replenishment of the
HSCs pool, thus finally resulting in a depletion of HSCs.
Apparently, PTEN deletion facilitates leukemogenesis while
it depletes HSCs. These effects of PTEN deletion appear to
be mediated by mammalian target of rapamycin (mTOR),
since treatment with the mTOR inhibitor rapamycin reversed
the effects. Therefore, rapamycin treatment seems to be a
promising approach for the selective elimination of LSCs
while sparing HSCs.
However, Ito and colleagues recently reported on a
contradictory effect of rapamycin [122]. They investigated
the role of the tumor-suppressor promyelocytic leukemia
protein (PML) in CML. Their results demonstrated that
PML-deficient CML-LSCs are less quiescent than wildtype
CML-LSCs, resulting in LSCs exhaustion. Furthermore,
deletion of PML in HSCs also results in exhaustion and con-
sequently impairment in long-term hematopoietic recons-
titution. Since PML deficiency in both HSCs and LSCs is
characterized by an increased mTOR activity they assessed
the effect of rapamycin treatment. Here, rapamycin rescued
the typical PML-deficient phenotype, thus exhaustion of the
CML-LSCs and HSCs was prevented. Together, these
seemingly contradictory results of mTOR inhibition in the
elimination of LSCs indicate that further delineation of the
role of mTOR in LSCs is needed.
Of note, since arsenic trioxide is known to decrease PML
levels, Ito and colleagues continued with assessing arsenic
trioxide for the elimination of CML-LSCs [122]. Treatment
with arsenic trioxide resulted in the exit of the CML-LSCs
from quiescence and therefore the CML-LSCs might become
exhausted and more vulnerable to chemotherapeutics. Impor-
tantly, this cell cycle entry appeared to be more profound in
LSCs than in HSCs, thereby generating a possible
therapeutic window for the treatment of CML-LSCs by
arsenic trioxide.
4-benzyl, 2-methyl, 1,2,4-thiadiazolidine, 3,5 dione
(TDZD-8)
TDZD-8 was designed as a non-ATP competitive
inhibitor of GSK3 for the treatment of Alzheimer’s disease
[123]. However, Guzman and colleagues uncovered potent
tumoricidal activity of TDZD-8 towards leukemia cells. In a
series of experiments they demonstrated that TDZD-8
exhibit strong anti-leukemia effects towards AML, CLL,
104 Current Drug Targets, 2010, Vol. 11, N o. 1 ten Cate et al.
ALL and blast crisis CML cells, without apparent toxic
effects towards normal BM cells. Since a panel of other
GSK3 inhibitors did not possess any comparable activity,
the anti-leukemia effect is probably not governed by the
inhibition of GSK3. This notion is supported by the
observation that -catenin, of which the degradation is in
part mediated by GSK3, is elevated in CML-LSCs.
Moreover, very recently GSK3 missplicing was observed in
CML-LSCs and not in HSCs [124], together indicating that
GSK3 is dispensable for leukemogenesis.
Importantly, TDZD-8 induced apoptosis in AML-LSCs
and CML-LSCs without affecting HSCs [125]. In line with
this, TDZD-8 specifically reduced the engraftment potential
of AML-LSCs and not of HSCs. TDZD-8 is a highly
hydrophobic agent which may result in a rapid loss of
membrane integrity in leukemic cells by TDZD-8 insertions
in the cellular membrane. The absence of this eff ect in HSCs
might indicate that there is crucial difference in the
membrane composition of LSCs and HSCs [125], which
might open novel avenues for the selective elimination of
LSCs.
Parthenolide (PTL)
PTL is a compound, derived from the plant feverfew,
which eliminates cancer cells via reactive oxygen species
and the inhibition of NF-kB [126-128]. Guzman and
colleagues analyzed the effects of PTL and Ara-C on AML
blasts and AML-LSCs. Apoptosis induction in the AML
blasts by Ara-C was more pronounced than by PTL.
However, PTL potently induced apoptosis in AML-LSCs,
whereas Ara-C exhibited only minimal apoptotic activity.
Importantly, the PTL concentration necessary for significant
AML-LSCs elimination had only a modest effect on HSCs
[129]. Moreover, PTL treatment significantly reduced the
engraftment potential of A ML-LSCs, while PTL treatment
did not affected the engraftment of HSCs [129].
Despite the favorable characteristics, the clinical appli-
cation of PTL is probably hampered by its poor water
solubility. Therefore, Guzman and colleagues generated a
series of parthenolide analogues aiming to identify an active
compound with improved pharmacological properties [130].
Recently, they identified a novel compound, designated
dimethylamino analog of parthenolide (DMAPT), which
exhibits similar activity as PTL. However, when formulated
as a fumarate salt, DMAPT had 1000-fold enhanced water
solubility, resulting in a 70% bioavailability of orally applied
DMAPT [130]. In contrast to PTL, oral administration of
DMAPT did result in plasma concentrations sufficient to
exert an anti-leukemia effect in both rodent and canine
models.
EPIGEN ETIC REPROGRAMMING OF LSCS
The step-wise conversion of a normal cell to a leukemic
cell typically involves the activation of oncogenes and anti-
apoptotic genes and the inactivation of tumor suppressor and
proapoptotic genes [131]. Frequently, gene expression in
leukemic cells is altered by epigenetic reprogramming
(reviewed in [132]).
Epigenetic reprogramming is characterized by various
distinct chromatin modifications. Chromatin consists of
histone proteins and DNA. The central unit of the chromatin
structure is an octamer of core histone proteins, referred to as
the nucleosome. The distance between adjacent nucleosomes
and thus the compaction of the chromatin in between,
defines two types of chromatin. Euchromatin is the open
type of chromatin with a relative low compaction which
allows access of the transcription machinery to the D NA,
while heterochromatin is the condensed form in which the
transcription machinery can not access the DNA and reflects
an inactive state. There are several types of modifications
that can alter the accessibility of the transcription machinery
to the DNA. The best studied are DNA and histone
methylation which are associated with heterochromatin and
histone acetylation which is associated with euchromatin.
Normal stems cells rely on Polycomb group (PcG)
proteins to repress genes encoding transcription factors
required for differentiation including the homeotic (HOX)
genes [133]. It is postulated that methylation of promoter
regions of those repressed genes could lock in stem cell
phenotypes and initiate abnormal clonal expansion and
thereby predispose to cancer [134, 135]. Repression of genes
by the PcG proteins is initiated by the Polycomb Repressor
Complex 2 (PRC2) containing the histone methyltransferase
EZH2, after which the Polycomb Repressor Complex 1
(PRC1) further maintains the repression. The PRC1
comprises amongst others Bmi-1, which is essential for self-
renewal of HSCs [136]. Of note, in both AML and CML
patients Bmi-1 is overexpressed compared to normal CD34+
BM cells. Patients with an elevated Bmi-1 expression have
less chance of achieving complete remission, higher chance
of relapse and reduced duration of survival [137, 138].
Moreover, very recently Wang and colleagues have shown
that the dysregulation of a chromatin-binding plant homeo-
domain (PHD) finger can cause hematological malignancies
[139]. Normally, PHD fingers respond to certain histone
methylation states [140, 141]. In AML however, chromo-
somal translocations are observed which results in the fusion
of a PHD finger to a common leukemia fusion partner called
NUP98 [142-144]. Wang and colleagues compared the
transcriptome profile of BM progenitor cells transduced with
such a fusion protein with control cells. This analysis
indicated that in the cells transduced with this fusion protein
several genes were upregulated including HOXA9 and
MEIS1 [145]. Lessard and Sauvageau have shown that
enforced expression of HOXA9/MEIS1 can induce
leukemia, which is dependent on the expression of Bmi-1
[146], suggesting that Bmi-1 inhibitors may contribute to the
elimination of LSCs.
However, also drugs with a more general epigenetic
modification activity may have clinical relevance in
attacking LSCs. This is evident from the clinical application
of the demethylating agent decitabine, which is approved for
the treatment of MDS. Decitabine inhibits DNA methyltrans-
ferases (DNMT) and therefore has a broad range of action by
tipping the balance towards euchromatin. Intriguingly,
decitabine exerts differential effects in LSCs and HSCs,
which is probably due to differences in their epigenomic
status. In LSCs treatment with decitabine results in
differentiation, whereas decitabine treatment of HSCs results
Targeted Elimination of Leukemia Stem Cells Current Drug Targets, 2010, Vo l. 11, No. 1 105
in enhanced self-renewal (ASH Annual Meeting Abstracts,
20084). Furthermore, it was demonstrated that decitabine
treatment reactivated more genes in bladder cancer cells than
in normal fibroblasts [147]. These studies indicate that
general epigenetic modifications can exert differential effects
in LSCs and HSCs, which might be utilized for the selective
elimination of LSCs.
Other general epigenetic modulators are HDACi such as
suberoylanilide hydroxamic acid (SAHA), 4-phenylbutyric
acid (PBA) and VPA. HDACi can also tip the balance
toward euchromatin and have shown to induce apoptosis in
leukemia (see Fig. 4). In clinical trials VPA monotherapy
was well tolerated, but showed disappointing anti-leukemia
effects (reviewed in [148]). However, combination treatment
of VPA with other anti-cancer agents showed significan tly
enhanced and sometimes synergistic anti-tumor effects [35,
149-151].
MicroRNA’s (miRs) are small, non-coding, single stran-
ded RNA sequences that regulate gene expression by binding
to mRNA molecules. Interestingly, upon co-treatment with
decitabine and PBA, miRs appear to be differential reac-
tivated in cancer cells and normal cells [152]. Saito and
colleagues have shown that the transcription of one parti-
cular miR, miR-127, was increased upon combination treat-
ment of decitabine and PBA. Subsequently, they identified
that a predicted target of miR-127, BCL6, was downre-
gulated [152]. This indicates that not only epigenetic
silenced protein-coding sequences, but also miRs can be
reactivated upon treatment with demethylating agents and
HDACi. This opens novel avenues for modifying proteins
levels in LS Cs and therefore may be exploited to eliminate
LSCs.
CONCLUSIONS AND PERSPECTIVES
The specific elimination of LSCs in hemato-oncology is
a research area still in its infancy. Currently, researchers are
at the brink of identifying biological processes imperative to
LSCs biology. Elucidation of these LSCs-specific charac-
teristics is of eminent importance for the rational design of
novel LSCs targeted therapies. It is anticipated that the
successful elimination of LSCs will have tremendous impact
on leukemia therapy. Several strategies appear promising for
the elimination of LSCs, includ ing targeting LSC-selective
cell surface antigens, disrupting LSCs-niche interactions,
targeting aberrantly regulated pathways and epigenetic
reprogramming of LSCs. Several studies utilizing these
strategies have generated promising pre-clinical results.
However, the vast majority of the data is obtained by xeno-
grafting LSCs in severely immunocompromised recipient
mice. It is evident that the data obtained using these models
can not be extrapolated directly to the clinical situation.
Furthermore, it is likely that multifaceted therapy combining
several strategies will be the most effectiv e approach.
Therefore, rational designed combinatorial approaches and
multimodality targeting of LSCs are warranted. Subsequent,
these combinatorial strategies have to prove to be safe and
effective in clinical trials.
4 Negrotto S, Hu Z, Link K, Duong H, Schade AE, Maciejewski JP, et al. Differentia-
tion-chronology specific function of DNMT1 and selective anti-leukemia stem-cell
therapy. ASH Annual Meeting Abstracts 2008; 112: 201.
ACKNOWLEDGEMENTS
Supported by grants from the Dutch Cancer Society
(2005-3358 and 2007-3784) to W.H.
ABBREVIATION S
ALDH = Aldehyde dehydrogenases
ALDH1A1 = Aldehyde Dehydrogenase class-1A1
ALDH3A1 = Aldehyde Dehydrogenase class-3A1
ALL = Acute Lymphoid Leukemia
AML = Acute Myeloid Leukemia
AML1-ETO = AML-1⁄ eight twenty one
AML-LSCs = Acute Myeloid Leukemia Leukemia
Stem Cells
APC = Adenomatous Polyposis Coli
APL = Acute Promyelocytic Leukemia
Ara-C = Cytosine Arabinoside
ATRA = All-trans Retinoic Acid
Bcl-9 = B Cell Lymphoma 9
Bcr-Abl = Breakpoint cluster region-abelson
Bid = BH3-only interacting domain death
agonist
BM = bone marrow
Bmi-1 = B-cell-specific Moloney murine
leukemia virus insertion site-1
CAM-DR = Cell Adhesion-Mediated Drug
Resistance
CD = Cluster of Differentiation
CDC = Complement Dependent Cytotoxicity
CLL = Chronic Lymphoid Leukemia
CLL-1 = C-type Lectin-Like Molecule-1
CML = Chronic Myeloid Leukemia
CML-LSCs = Chronic Myeloid Leukemia Leukemia
Stem Cells
DEAB = Diethylamino-benzaldehyde
Dhh = Desert Hedgehog
DISC = Death Induced Signaling Complex
DKK = Dickkopf
DMAPT = Dimethylamino Analog of Parthenolide
DNA = Deoxyribonucleic acid
DNMT = DNA methyltransferase
DR = Death Receptor
DT = Diphtheria Toxin
FDA = Federal Drug Administration
FADD = FAS associated death domain
GO = Gemtuzumab Ozogamicin
106 Current Drug Targets, 2010, Vol. 11, N o. 1 ten Cate et al.
GSK3 = Glycogen-synthase kinase 3
HAT = Histone Acetyltransferase
HDACi = Histone Deactylase inhibitor
Hh = Hedgehog
HPCs = Hematopoietic Progenitor Cells
HSCs = Hematopoietic Stem Cells
Ig = Immunoglobulin
Ihh = Indian Hedgehog
IL-3R = Interleukin 3 Receptor
ITIM = Immunoreceptor tyrosine-based
inhibitory motif
LRP5/6 = Lipoprotein receptor-related protein 5/6
LSCs = Leukemia Stem Cells
mAb = Monoclonal Antibody
MDC = Multiprotein Destruction Complex
MDS = Myelodysplastic Syndrome
miRs = microRNA’s
mTOR = Mammalian target of rapamycin
NF-kB = Nuclear Factor kappa B
NK cell = Natural Killer cell
NOD/SCID = Non-Obese Diabetic/ Severe Combined
Immunodeficient
PBA = 4-phenylbutyric acid
PcG = Polycomb group
PE38 = Pseudomonas Exotoxin Fragment of
38 kilodalton
Pgp = P-glycoprotein
PIP2 = Phosphatidylinositol (3,4)-bisphosphate
PIP3 = Phosphatidylinositol (3,4,5)-
trisphosphate
PI3K = Phosphatidylinositol-3-OH kinase
PLZF ⁄RAR = Promyelocytic leukemia zinc finger⁄
retinoic acid receptor alpha
PML = Promyelocytic leukem ia protein
PML⁄RAR = Promyelocytic leukemia retinoic acid
receptor alpha
PRC1 = Polycomb Repressor Complex 1
PRC2 = Polycomb Repressor Complex 2
PTEN = Phosphatase and tensin homologue
PTL = Parthenolide
SAHA = Suberoylanilide hydroxamic acid
scFv = Single chain fragment of the variable
regions
SDF-1 = Stroma-derived factor-1
Shh = Sonic Hedgehog
Smo = Smoothened
SP = Side Population
tBid = Truncated form of Bid
TCF = T-cell factor
TDZD-8 = 4-benzyl, 2-methyl, 1,2,4-
thiadiazolidine, 3,5 dione
TNFR = Tumor Necrosis Factor Receptor
TRAIL = Tumor necrosis factor Related Apoptosis
Inducing Ligand
TRAIL-R1 = TRAIL Receptor 1
TRAIL-R2 = TRAIL Receptor 2
VLA-4 = Very Late Antigen 4
wif-1 = Wnt inhibitory factor 1
REFERENCES
[1] Jordan CT, Guzman ML, Noble M. Cancer stem cells. N Engl J
Med 2006; 355: 1253-61.
[2] Krause DS, Van Etten RA. Right on target: eradicating leukemic
stem cells. Trends Mol Med 2007; 13: 470-81.
[3] 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.
[4] Lapidot T, Sirard C , Vormoor J, Murdoch B, Hoang T, Caceres-
Cortes J, et al. A cell initiating human acute myeloid leukae mia
after transplantation into SCID mice. Nature 1994; 367: 645-8.
[5] Castor A, Nilsson L, Astrand-Grundstrom I, Buitenhuis M,
Ramirez C, Anderson K, et al. Distinct p atterns of hematopoietic
stem cell involvement in acute lymphoblastic leukemia. Nat Med
2005; 11: 630-7.
[6] Misaghian N, Ligresti G, Steelman LS, Bertrand FE, Basecke J,
Libra M, et al. Targeting the leukemic stem cell: the Holy Grail of
leukemia therapy. Leukemia 2009; 23 : 25-42.
[7] Dalerba P, Cho RW, Clarke MF. Cancer stem cells: models and
concepts. Annu Rev Med 2007; 58: 267-84.
[8] Hope KJ, Jin L, Dick JE. Acute myeloid leukemia originates from a
hierarchy of leukemic stem cell classes that differ in self-renewal
capacity. Nat Immunol 2004; 5: 738-43.
[9] Terpstra W, Ploemacher RE, Prins A, van Lom K, Pouwels K,
Wognum AW, et al. Fluorouracil selectively spares acute myeloid
leukemia cells with long-term growth abilities in immunodeficient
mice and in culture. Blood 1996; 88: 1944-50.
[10] Dean M, Fojo T, Bates S. Tumour stem cells and drug resistance.
Nat Rev Cancer 2005; 5: 275-84.
[11] Lane SW, Scadden DT, Gilliland DG. The leukemic stem cell
niche - current concepts and therapeutic opportunities. Blood 2009
[Epub ahead of print].
[12] Stel AJ, Ten Cate B, Jacobs S, Kok JW, Spierings DC, Don dorff
M, et al. Fas receptor clustering and involvement of the death
receptor p athway in rituximab-mediated apoptosis with
concomitant sensitization of lymphoma B cells to fas-induced
apoptosis. J Immunol 2007; 178: 2287-95.
[13] Byrd JC, Kitada S, Flinn IW, Aron JL, Pearson M, Lucas D, et al.
The mechanism of tumor cell clearance by rituximab in vivo in
patients with B-cell chronic lymphocytic leukemia: evidence of
caspase activation and apoptosis induction. Blood 2002; 99: 1038-
43.
[14] Sato T, Laver JH, Ogawa M. Reversible expression of CD34 by
murine hematopoietic stem cells. Blood 1999; 94: 2548-54.
[15] Freeman SD, Kelm S, Barber EK, Crocker PR. Characterization o f
CD33 as a new member of the sialoadhesin family of cellular
interaction molecules. Blood 1995; 85: 2005-12.
[16] Wellhausen SR, Peiper SC. CD33: biochemical and biological
characterization and evaluation of clinical relevance. J Biol Regul
Homeost Agents 2002; 16: 139-43.
Targeted Elimination of Leukemia Stem Cells Current Drug Targets, 2010, Vo l. 11, No. 1 107
[17] Tchilian EZ, Beverley PC, Young BD, Watt SM. Molecular
cloning of two isoforms of the murine homolog of the myeloid
CD33 antigen. Blood 1994; 83: 3188-98.
[18] Florian S, Sonneck K, Hauswirth AW, Krauth MT, Schernthaner
GH, Sperr WR, et al. Detection of molecular targets on the surface
of CD34+/CD38-- stem cells in various myeloid malignancies.
Leuk Lymphoma 2006; 47: 207-22.
[19] Hauswirth AW, Florian S, Printz D, Sotlar K, Krauth MT, Fritsch
G, et al. Expression of the target receptor CD33 in CD34+/CD38-
/CD123+ AML stem cells. Eur J Clin Invest 2007; 37: 73-82.
[20] Jordan CT, Upchurch D, Szilvassy SJ, Guzman ML, Howard DS,
Pettigrew AL , et al. The interleukin-3 receptor alpha chain is a
unique marker for human acute myelogenous leukemia stem cells.
Leukemia 2000; 14: 1777-84.
[21] Moretti S, Lanza F, Dabusti M, Tieghi A, Camp ioni D, Dominici
M, et al. CD123 (interleukin 3 receptor alpha chain). J Biol Regul
Homeost Agents 2001; 15: 98-100.
[22] Marsha ll AS, Willment JA, Lin HH, Williams DL, Gordon S,
Brown GD. Identification and characterization of a novel human
myeloid inhibitory C-type lectin-like receptor (MICL) that is
predominantly expressed on granulocytes and monocytes. J Biol
Chem 2004; 279: 14792-802.
[23] Bakker AB, van den Oudenrijn S, Bakker AQ, Feller N, van Meijer
M, Bia JA, et al. C-type lectin-like molecule-1: a novel myeloid
cell surface marker associated with acute myeloid leukemia.
Cancer Res 2004; 64: 8443-50.
[24] van Rhenen A, van Dongen GA, Kelder A, Rombouts EJ, Feller N,
Moshaver B, et al. The novel AML stem cell associated antigen
CLL-1 aids in discrimination between normal and leukemic stem
cells. Blood 2007; 110: 2659-66.
[25] Moshaver B, van Rhenen A, Kelder A, van der Pol M, Terwijn M,
Bachas C, et al. Identification of a small subpopulation of
candidate leukemia-initiating cells in the side population of patients
with acute myeloid leukemia. Stem Cells 2008; 26: 3059-67.
[26] Fuchs A, Cella M, Giurisato E, Shaw AS, Colonna M. Cutting
edge: CD96 (tactile) promotes NK cell-targe t cell adhesion by
interacting with the poliovirus receptor (CD155). J Immunol 2004;
172: 3994-8.
[27] Gramatzki M, Ludwig WD, Burger R, Moos P, Roh wer P, Grunert
C, et al. Antibodies TC-12 ("unique") and TH-111 (CD96)
characterize T-cell acute lymphoblastic leukemia and a subgroup of
acute myeloid leukemia. Exp Hematol 1998; 26: 1209-14.
[28] Hosen N, Park CY, Tatsumi N, Oji Y, Sugiyama H, Gramatzki M,
et al. CD96 is a leukemic stem cell-specific marker in human acute
myeloid leukemia. Proc Natl Acad Sci U S A 2007; 104: 11008-13.
[29] Sun Q, Woodcock JM, Rapoport A, Stomski FC, Korpelainen EI,
Bagley CJ, et al. Monoclonal antibody 7G3 recognizes the N-
terminal domain of the human interleukin-3 (IL-3) receptor alpha-
chain and functions as a specific IL-3 receptor antagonist. Blood
1996; 87: 83-92.
[30] Ten Cate B, Bremer E, de Bruyn M, Bijma T, Samplonius D,
Schwemmlein M, et al. A novel AML-selective TRAIL fusion
protein that is superior to Gemtuzumab Ozogamicin in terms of in
vitro selectivity, activity and stability. Leukemia 2009 Mar 5.
[31] Schwemmlein M, Peipp M, Barbin K, Saul D, Stockmeyer B, Repp
R, et al. A CD33-specific single-chain immunotoxin mediates
potent apoptosis of cultured human myeloid leukaemia cells. Br J
Haematol 2006; 133: 141-51.
[32] van Der Velden VH, te Marvelde JG, Hoogeveen PG, Bernstein ID,
Houtsmuller AB, Berger MS, et al. Targeting of the CD33-
calicheam icin immunoconjugate Mylotarg (CMA-676) in a cute
myeloid leukemia: in vivo and in vitro satu ration and
internalization by leukemic and normal myeloid cells. Blood 2001;
97: 3197-204.
[33] Bross PF, Beitz J, Chen G, Chen XH, Duffy E, Kieffer L, et al.
Approval summary: gemtuzumab ozogamicin in relapsed acute
myeloid leukemia. Clin Cancer Res 2001; 7: 1490-6.
[34] Larson RA, Sievers EL, Stadtmaue r EA, Lowenberg B, Estey EH,
Dombret H, et al. Final report of the efficacy and safety of
gemtuzumab ozogamicin (Mylotarg) in patients with CD33-
positive acute myeloid leukemia in first recurrence. Cancer 2005;
104: 1442-52.
[35] Ten Cate B, Samplonius DF, Bijma T, de Leij LF, Helfrich W,
Bremer E. The histone deacetylase inhibitor valproic acid potently
augments gemtuzumab ozogamicin-induced apoptosis in acute
myeloid leukemic cells. Leukemia 2007; 21: 248-52.
[36] Walter RB, Gooley TA, van Der Velden VH, Loken MR, van
Dongen JJ, Flowers DA, et al. CD33 expression and P-
glycoprotein-mediated drug efflux inversely correlate and predict
clinical outcome in patients with acute myeloid leukemia treated
with gemtuzumab ozogamicin monotherapy. Blood 2007; 109:
4168-70.
[37] Kersemans V, Cornelissen B, Mind en MD, Bran dwein J, Reilly
RM. Drug-resistant AML cells and primary AML specimens are
killed by 111In-anti-CD33 monoclonal antibodies modified with
nuclear localizing peptide sequences. J Nucl Med 2008; 49: 1546-
54.
[38] Du X, Ho M, Pastan I. New immunotoxins targeting CD123, a
stem cell antigen on acute myeloid leukemia cells. J Immunother
2007; 30: 607-13.
[39] Testa U, Riccioni R, Biffoni M, Diverio D, Lo-Coco F, Foa R, et
al. Diphtheria toxin fused to variant human interleukin-3 induces
cytotoxicity of blasts fro m patients with acute myelo id leukemia
according to the level of interleukin-3 receptor expression. Blood
2005; 106: 2527-9.
[40] Feuring-Buske M, Frankel AE, Alexander RL, Gerhard B, Hogge
DE. A diphtheria toxin-interleukin 3 fusion protein is cytotoxic to
primitive acute myeloid leuke mia progenitors but spares normal
progenitors. Cancer Res 2002; 62: 1730-6.
[41] Yalcintepe L, Frankel AE, Hogge DE. Expression of interleukin-3
receptor subunits on defined subpopulations of acute myeloid
leukemia blasts predicts the cytotox icity of diphtheria toxin
interleukin -3 fusion protein against malignant progenitors that
engraft in immunodeficient mice. Blood 2006; 108: 3530-7.
[42] Horita H, Frankel AE, Thorburn A. Acute-myeloid-leukemia-
targeted toxins kill tumor cells b y cell-type-specific mechanisms
and synergize with TRAIL to allow manipulation of the extent and
mechanism of tumor cell death. Leukemia 2008; 22: 652-5.
[43] Ashkenazi A. Targeting the extrinsic apoptosis pathway in cancer.
Cytokine Growth Factor Rev 2008; 19: 325-31.
[44] Tolcher AW, Mita M, Meropol NJ, von Mehren M, Patnaik A,
Padavic K, et al. Phase I ph armacokinetic and biologic correlative
study of mapatumumab, a fully human monoclonal antibody with
agonist activity to tumor necrosis factor-related apoptosis-inducing
ligand receptor-1. J Clin Oncol 2007; 25: 1390-5.
[45] Hotte SJ, Hirte H W, Chen EX, Siu LL, Le LH, Corey A, et al. A
phase 1 study of mapatumumab (fu lly huma n monoclonal antibody
to TRAIL-R1) in patients with advanc ed solid malignancies. Clin
Cancer Res 2008; 14: 3450-5.
[46] Plummer R, Attard G, Pacey S, Li L, Razak A, Perrett R, et al.
Phase 1 and pharmacokinetic study of lexatumumab in patients
with advanced cancers. Clin Cancer Res 2007; 13: 6187-94.
[47] Ashkenazi A. Directing cancer cells to se lf-destruct with pro-
apoptotic receptor agonists. Nat Rev Drug Discov 2008; 7: 1001-
12.
[48] Bremer E, de Bru yn M, Wajant H, Helfrich W . Targeted cancer
immunotherapy using ligand s of the tumor necrosis facto r super-
family. Curr Drug Targets 2009; 10: 94-103.
[49] Bremer E, van Dam GM, de Bruyn M, van Riezen M, Dijkstra M,
Kamps G, et al. Potent systemic anticancer activity of adenovirally
expressed EGFR-selective TRAIL fusion protein. Mol Ther 2008;
16: 1919-26.
[50] Bremer E, Samplonius DF, van Genne L, Dijkstra MH, Kroesen
BJ, de Leij LF, et al. Simultaneous inhibition of epid ermal growth
factor receptor (EGFR) signaling and enhanced activation of tumor
necrosis factor-related apoptosis-inducing ligand (TRAIL)
receptor-mediated apoptosis induction by an scFv:sTRAIL fusion
protein with specificity for human EGFR. J Biol Chem 2005; 280:
10025-33.
[51] Bremer E, de Bruyn M, Samplonius DF, Bijma T, Ten Cate B, de
Leij LF, et al. Targeted delivery of a designed sTRAIL mutant
results in superior apoptotic activity towards EGFR-positive tumor
cells. J Mol Med 2008; 86: 909-24.
[52] Bremer E, Samplonius D, Kroesen BJ, van Genne L, de Leij L,
Helfrich W. Exceptionally potent anti-tumor bystander activity of
an scFv:sTRAIL fusion protein with specificity for EGP2 toward
target antigen-negative tumor cells. Neoplasia 2004; 6: 636-45.
[53] Bremer E, Kuijlen J, Samplonius D, Walczak H, de Leij L, Helfrich
W. Target cell-restricted and -enhanced apoptosis induction by a
scFv:sTRAIL fusion protein with specificity for the pancarcinoma-
associated antigen EGP2. Int J Cancer 2004; 109: 281-90.
108 Current Drug Targets, 2010, Vol. 11, N o. 1 ten Cate et al.
[54] Bremer E, Samplonius DF, Peipp M, van Genne L, Kroesen BJ,
Fey GH, et al. Target cell-restricted apoptosis induction of acute
leukemic T cells by a recombinant tumor necrosis factor-related
apoptosis-inducing ligand fusion protein with specificity for human
CD7. Cancer Res 2005; 65: 3380-8.
[55] Stieglmaier J, Bremer E, Kellner C, Liebig TM, Ten Cate B, Peipp
M, et al. Selective induction of apoptosis in leukemic B-lymphoid
cells by a CD19-specific TRAIL fusion protein. Cancer Immunol
Immunother 2008; 57: 233-46.
[56] Majeti R, Becker MW, Tian Q, Lee TL, Yan X, Liu R, et al.
Dysregulated gene expression networks in human acute
myelogenous leukemia stem cells. Proc Natl Acad Sci USA 2009
[Epub ahead of print].
[57] Yong A, Keyvanfar K, Hensel N, Eniafe R, Savani B, Berg M, et
al. Primitive quiescent CD34+ cells in chronic myeloid leukemia
are targeted by in vitro expanded natural killer cells, which are
functionally enhanced by bortezomib. Blood 2009; 113: 875-82.
[58] Tazzari PL, Tabellini G, Ricci F, Papa V, Bortu l R, Chiarini F, et
al. Synergistic proapoptotic activity of recombinant TRAIL plus
the Akt inhibitor Perifosine in acute myelogenous leukemia cells.
Cancer Res 2008; 68: 9394-403.
[59] Schofield R. The relationship between the spleen colony-forming
cell and the haemopoietic stem cell. Blood Cells 1978; 4: 7-25.
[60] Nilsson SK, Johnston HM, Coverdale JA. Spatial localization of
transplanted hemopoietic stem cells: inferences for the localization
of stem cell niches. Blood 2001; 97: 2293-9.
[61] Kiel MJ, Morrison SJ. Unce rtainty in the niches that maintain
haematopoietic stem cells. Nat Rev Immunol 2008; 8: 290-301.
[62] Colmone A, Amorim M, Pontier AL, Wang S, Jablonski E, Sipkins
DA. Leukemic cells create bone marrow niches that disrupt the
behavior of normal hematopoietic progenitor cells. Science 2008;
322: 1861-5.
[63] Ishikawa F, Yoshida S, Saito Y, Hijikata A, Kitamura H, Tanaka S,
et al. Chemotherapy-resistant human AML stem cells home to and
engraft within the bone-marrow endosteal region. Nat Biotechnol
2007; 25: 1315-21.
[64] Peled A, Petit I, Kollet O, Magid M, Ponomaryov T, Byk T, et al.
Dependence of human stem cell engraftment and repopulation of
NOD/SCID mice on CXCR4. Science 1999; 283: 845-8.
[65] Sugiyama T, Kohara H, Noda M, Nagasawa T. Maintenance of the
hematopoietic stem cell pool by CXCL12-CXCR4 chemokine
signaling in bone marrow stromal cell niches. Immunity 2006; 25:
977-88.
[66] Tavor S, Petit I, Porozov S, Avigdor A, Dar A, Leider-Trejo L, et
al. CXCR4 regulates migration and development of human acute
myelogenous leukemia stem cells in transplanted NOD/SCID mice.
Cancer Res 2004; 64: 2817-24.
[67] Spoo AC, Lubbert M, Wierda WG, Burger JA. CXCR4 is a
prognostic marker in acute myelogenous leukemia. Blood 2007;
109: 786-91.
[68] Rombouts EJ, Pavic B, Lowenberg B, Ploemacher RE. Relation
between CXCR-4 expression, Flt3 mutations, and unfavorable
prognosis of adult acute myeloid leukemia. Blood 2004; 104: 550-
7.
[69] Broxmeyer HE, Orschell CM, Clapp DW, Hangoc G, Cooper S,
Plett PA, et al. Rapid mobilization of murine and human
hematopoietic stem and progenitor cells with AMD3100, a CXCR4
antagonist. J Exp Med 2005; 201: 1307-18.
[70] Nervi B, Ramire z P, Rettig MP, Uy GL, Holt MS, Ritchey JK, et
al. Chemosensitization of acute myeloid leukemia (AML)
following mobilization by the CXCR4 antagonist AMD3100.
Blood 2009; 113: 6206-14.
[71] Zeng Z, Shi YX, Samudio IJ, Wang RY, Ling X, Frolova O, et al.
Targeting th e leukemia microenvironment by CXCR4 inh ibition
overcomes resistance to kinase inhibitors and chemotherapy in
AML. Blood 2009; 113: 6215-24.
[72] Burger JA, Peled A. CXCR4 antagonists: targeting the
microenvironment in leukemia and other cancers. Leukemia 2009;
23: 43-52.
[73] Li H, Guo L, Jie S, Liu W, Zhu J, Du W, et al. Berberine inhibits
SDF-1-induced AML cells and leukemic stem cells migration via
regulation of SDF-1 level in bone marrow stromal cells. Biomed
Pharmacother 2008; 62: 573-8.
[74] Papayannopoulou T, Priestley GV, Nakamoto B, Zafiropoulos V,
Scott LM. Molecular pathways in bone marrow homing: dominant
role of alpha(4)beta(1) over beta(2)-integrins and selectins. Blood
2001; 98: 2403-11.
[75] Matsunaga T, Takemoto N, Sato T, Takimoto R, Tanaka I, Fujimi
A, et al. Interaction be tween leukemic-cell VLA-4 and stromal
fibronectin is a decisive factor for minimal residual disease of acute
myelogenous leukemia. Nat Med 2003; 9: 1158-65.
[76] Kato R, Ishikawa T, Kamiya S, Oguma F, Ueki M, Goto S, et al. A
new type of antimetastatic peptide derived from fibronectin. Clin
Cancer Res 2002; 8: 2455-62.
[77] Matsunaga T, Fukai F, Miura S, Nakane Y, Owaki T, Kodama H,
et al. Combination therapy of an anticancer drug with the FNIII14
peptide of fibronectin effectively overcomes cell adhesion-
mediated drug resistance of acute myelogenous leukemia.
Leukemia 2008; 22: 353-60.
[78] Avigdor A, Goichberg P, Shivtiel S, Dar A, Peled A, Samira S, et
al. CD44 and hyaluronic acid cooperate with SDF-1 in the
trafficking of human CD34+ stem/progenitor cells to bone marrow.
Blood 2004; 103: 2981-9.
[79] Sackstein R, Merzaban JS, Cain DW, Dagia NM, Spencer JA, Lin
CP, et al. Ex vivo glycan engineering of CD44 programs human
multipotent mesenchyma l stromal cell traffick ing to bone. Nat Med
2008; 14: 181-7.
[80] Krause DS, Lazarides K, von Andrian UH, Van Etten RA.
Requirement for CD44 in homing and engraftment of BCR-ABL-
expressing leukemic stem cells. Nat Med 2006; 12: 1175-80.
[81] Jin L, Hope KJ, Zhai Q, Smadja-Joffe F, Dick JE. Targeting of
CD44 eradicates human acute myeloid leukemic stem cells. Nat
Med 2006; 12: 1167-74.
[82] Graham SM, Jorgensen HG, Allan E, Pearson C, Alcorn MJ,
Richmond L, et al. Primitive, quiesc ent, Philadelphia-positive stem
cells from patients with chronic myeloid leukemia are insensitive to
STI571 in vitro. Blood 2002; 99: 319-25.
[83] Moll J, Sansig G, Fattori E, van der Putten H. The murine rac1
gene: cDNA cloning, tissue distribution and regulated expression
of rac1 mRNA by disassembly of actin microfilaments. Oncogene
1991; 6: 863-6.
[84] Shirsat NV, Pignolo RJ, Kreider BL, Rovera G. A member of the
ras gene superfamily is ex pressed sp ecifically in T, B and myeloid
hemopoietic cells. Oncogene 1990; 5: 769-72.
[85] Haataja L, Groffen J, Heisterkamp N. Characterization of RAC3, a
novel member of the Rho family. J Biol Chem 1997; 272: 20384-8.
[86] Gu Y, Filippi MD, Cancelas JA, Siefrin g JE, Williams EP, Jasti
AC, et al. Hematopoietic cell re gulation by Rac1 and Rac2
guanosine triphosphatases. Science 2003; 302: 445-9.
[87] Gao Y, Dickerson JB, Guo F, Zheng J, Zheng Y. Rational design
and characterization of a Rac GTPase-specific small molecule
inhibitor. Proc Natl Acad Sci U S A 2004; 101: 7618-23.
[88] Muller LU, Schore RJ, Zheng Y, Thomas EK, Kim MO, Cancelas
JA, et al. Rac guanosine triphosphatases represent a potential target
in AML. Leukemia 2008; 22: 1803-6.
[89] Rozenveld-Geugien M, Baas IO, van Gosliga D, Vellenga E,
Schuringa JJ. Expansion of normal and leukemic human
hematopoietic stem/progenitor cells requires rac-mediated
interaction with stromal cells. Exp Hematol 2007; 3 5: 782-92.
[90] Beachy PA, Karhadkar SS, Berman DM. Tissue repair and stem
cell renewal in carcinogenesis. Nature 2004; 432: 324-31.
[91] Rohatgi R, Scott MP. Patch ing the gaps in Hedgehog signallin g.
Nat Cell Biol 2007; 9: 1005-9.
[92] Duman-Scheel M, Weng L, Xin S, Du W. Hedgehog regulates cell
growth and proliferation by inducing Cyclin D and Cyclin E.
Nature 2002; 417: 299-304.
[93] Liu S, Dontu G, Mantle ID, Patel S, Ahn NS, Jackson KW, et al.
Hedgehog signaling and Bmi-1 regulate self-renewal of normal and
malignant human mammary stem cells. Cancer Res 2006; 66:
6063-71.
[94] Chen JK, Taipale J, Cooper MK, Beachy PA. Inhibition of
Hedgehog signaling by direct binding of cyclopamine to
Smoothened. Genes Dev 2002; 16: 2743-8.
[95] Zhao C, Chen A, Jamieson CH, Fereshteh M, Abrahamsson A,
Blum J, et al. Hedgehog signalling is e ssential for maintenance of
cancer stem cells in myeloid leukaemia. Nature 2009; 458: 776-9.
[96] Rey a T, Duncan AW, Ailles L, Domen J, Scherer DC, Willert K, et
al. A role for Wnt signalling in self-renewal of haematopoietic
stem cells. Nature 2003; 423: 409-14.
[97] Reya T, Clevers H. Wnt signalling in stem cells an d cancer. Nature
2005; 434: 843-50.
Targeted Elimination of Leukemia Stem Cells Current Drug Targets, 2010, Vo l. 11, No. 1 109
[98] Giles RH, van Es JH, Clevers H. Caught up in a Wnt storm: Wnt
signaling in cancer. Biochim Biophys Acta 2003; 1653: 1-24.
[99] He TC, Sparks AB, Rag o C, Hermeking H, Zawel L, da Costa LT,
et al. Identification of c-MYC as a target of the APC pathway.
Science 1998; 281: 1509-12.
[100] Tetsu O, McCormick F. Beta-catenin regulates expression of cyclin
D1 in colon carcinoma cells. Nature 1999; 398: 422-6.
[101] Wielenga VJ, Smits R, Korinek V, Smit L, Kielman M, Fodde R, et
al. Expression of CD44 in Apc and Tcf mutant mice implies
regulation by the WNT pathway. Am J Pathol 1999; 154: 515-23.
[102] Muller-Tidow C, Steffen B, Cauvet T, Tickenbrock L, Ji P,
Diederichs S, et al. Translocation products in acute myeloid
leukemia activate the Wnt signaling pathway in hematopoietic
cells. Mol Cell Biol 2004; 24: 2890-904.
[103] Mikesch JH, Steffen B, Berdel WE, Serve H, Muller- Tidow C. The
emerging role of Wnt signaling in the pathogenesis of acute
myeloid leukemia. Leukemia 2007; 21: 1638-47.
[104] Zhao C, Blum J, Chen A, Kwon HY, Jung SH, Cook JM, et al.
Loss of beta-catenin impairs the renewal of normal and CML stem
cells in vivo. Cancer Cell 2007; 12: 528-41.
[105] Hu Y, Chen Y, Douglas L, Li S. beta-Catenin is essential for
survival of leukemic stem cells insensitive to kinase inhibition in
mice with BCR-ABL-induced chronic myeloid leukemia.
Leukemia 2009; 23: 109-16.
[106] Kawano Y, Kypta R. Secreted antagonists of the Wnt signalling
pathway. J Cell Sci 2003; 116: 2627-34.
[107] Zhu Y, Sun Z, Han Q, Liao L, Wang J, Bian C, et al. Human
mesenchymal stem cells inhibit cancer cell proliferation by
secreting DKK-1. Leukemia 2009; 23: 925-33.
[108] Lepourcelet M, Chen YN, France DS, Wang H, Crews P, Petersen
F, et al. Small-molecule antagonists of the oncogenic Tcf/beta-
catenin protein complex. Cancer Cell 2004; 5: 91-102.
[109] Minke KS, Staib P, Puetter A, Gehrke I, Gandhirajan RK,
Schlosser A, et al. Small molecule inhibitors of WNT signaling
effectively induce apoptosis in acute myeloid leukemia cells. Eur J
Haematol 2009; 82: 165-75.
[110] Sophos NA, Vasiliou V. Aldehyde dehydrogenase gene
superfamily: the 2002 update. Chem Biol Interact 2003; 143-144:
5-22.
[111] Moreb JS, Mohuczy D, Ostmark B, Zucali JR. RNAi-mediated
knockdown of aldehyde dehydrogenase class-1A1 and class-3A1 is
specific and reveals that each contributes equally to the resistance
against 4-hydroperoxycyclophosphamide. Cancer Chemother
Pharmacol 2007; 59: 127-36.
[112] Pearce DJ, Taussig D, Simpson C, Allen K, Rohatiner AZ, Lister
TA, et al. Characterization of cells with a high aldehyde
dehydrogenase activity from cord blood and acute myeloid
leukemia samples. Stem Cells 2005; 23: 752-60.
[113] Ginestier C, Hur MH, Charafe-Jauffret E, Monville F, Dutcher J,
Brown M, et al. ALDH1 is a mark er of normal and malignant
human mammary stem cells and a predictor of poor clinical
outcome. Cell Stem Cell 2007; 1: 555-67.
[114] Ma S, Chan KW, Lee TK, Tang KH, Wo JY, Zheng BJ, et al.
Aldehyde dehydrogenase discriminates the CD133 liver cancer
stem cell populations. Mol Cancer Res 2008; 6: 1146-53.
[115] Cheung AM, Wan TS, Leung JC, Chan LY, Huang H, Kwong YL,
et al. Aldehyde dehydrogenase activity in leukemic blasts defines a
subgroup of acute myeloid leukemia with adverse prognosis and
superior NOD/SCID engrafting potential. Leukemia 2007; 21:
1423-30.
[116] Dylla SJ, Beviglia L, Park IK, Chartier C, Raval J, Ngan L, et al.
Colorectal cancer stem cells are enriched in xenogeneic tumors
following chemotherapy. PLoS ONE 2008; 3: e2428.
[117] Ryningen A, Stapnes C, Paulsen K, Lassalle P, Gjertsen BT,
Bruserud O. In vivo biological effects of ATRA in the treatment of
AML. Expert Opin Investig Drugs 2008; 17: 1623-33.
[118] Dahia PL, Aguiar RC, Alberta J, Kum JB, Caron S, Sill H, et al.
PTEN is inversely correlated with the cell survival factor Akt/PKB
and is inactivated via multiple mechanismsin haematological
malignancies. Hum Mol Genet 1999; 8: 185-93.
[119] Cheong JW, Eom JI, Maeng HY, Lee ST, Hahn JS, Ko YW, et al.
Phosphatase and tensin homologue phosphorylation in the C-
terminal regulatory domain is frequently observed in acute myeloid
leukaemia and associated with poor clinical outcome. Br J
Haematol 2003; 122: 454-6.
[120] Maehama T, Dixon JE. The tumor suppressor, PTEN/MMAC1,
dephosphorylates the lipid second messenger, phosphatidylinositol
3,4,5-trisphosphate. J Biol Chem 1998; 273: 13375-8.
[121] Yilmaz OH, Valdez R, Theisen BK, Guo W, Ferguson DO, Wu H,
et al. Pten dependence distinguishes haematopoietic stem cells
from leukaemia-initiating cells. Nature 2006; 441: 475-82.
[122] Ito K, Bernardi R, Morotti A, Matsuoka S, Saglio G, Ikeda Y, et al.
PML targeting eradicates quiescent leukaemia-initiating cells.
Nature 2008; 453: 1072-8.
[123] Martinez A, Alonso M, Castro A, Perez C, Moreno FJ. First non-
ATP competitive glycogen synthase kinase 3 beta (GSK-3beta)
inhibitors: thiadiazolidinones (TDZD) as potential drugs for the
treatment of Alzheimer's disease. J Med Chem 2002; 45: 1292-9.
[124] Abrahamsson AE, Geron I, Gotlib J, Dao KH, Barroga CF, Newton
IG, et al. Glycogen synthase kinase 3beta missplicing contributes
to leukemia stem cell gen eration. Proc Natl Acad Sci U S A 2009;
106: 3925-9.
[125] Guzman ML, Li X, Corbett CA, Rossi RM, Bushnell T, Liesveld
JL, et al. Rapid and selective death of leukemia stem and
progenitor cells induced by the compound 4-benzyl, 2-methyl,
1,2,4-thiadiazolidine, 3,5 dione (TDZD-8). Blood 2007; 110: 4436-
44.
[126] Steele AJ, Jones DT, Ganeshaguru K, Duke VM, Yogashangary
BC, North JM, et al. The sesquiterpene lactone parthenolide
induces selective apoptosis of B-chronic lymphocytic leukemia
cells in vitro. Leukemia 2006; 20: 1073-9.
[127] Wen J, You KR, Lee SY, Song CH, Kim DG. Oxidative stress-
mediated apoptosis. The anticancer effect of the sesquiterpene
lactone parthenolide. J Biol Chem 2002; 277: 38954-64.
[128] Oka D, Nishimura K, Shiba M, Nakai Y, Arai Y, Nakayama M, et
al. Sesquiterpen e lactone p arthenolide suppresses tumor growth in
a xenograft model of renal cell carcinoma by inhibiting the
activation of NF-kappaB. Int J Cancer 2007; 120: 2576-81.
[129] Guzman ML, Rossi RM, Karnischky L, Li X, Peterson DR,
Howard DS, et al. The sesquiterpene lactone parthenolide induces
apoptosis of human acute myelogenous leukemia stem and
progenitor cells. Blood 2005; 105: 4163-9.
[130] Guzman ML, Rossi RM, Neelakantan S, Li X, Corbett CA,
Hassane DC, et al. An orally bioavailable parthenolide analog
selectively eradicates acute myelogenous leukemia stem and
progenitor cells. Blood 2007; 110: 4427-35.
[131] Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;
100: 57-70.
[132] Jones PA, Baylin SB. The epigenomics of cancer. Cell 2007; 128:
683-92.
[133] Ringro se L, Paro R. Epigenetic regulation of cellu lar memory by
the Polycomb and Trithorax group proteins. Annu Rev Genet 2004;
38: 413-43.
[134] Schuebel K, Chen W, Baylin SB. CIMPle origin for promoter
hypermethylation in colorectal cancer? Nat Genet 2006; 38: 738-
40.
[135] Widschwendter M, Fiegl H, Egle D, Mueller-Holzner E, Spizzo G,
Marth C, et al. Epigenetic stem cell signature in cancer. Nat Genet
2007; 39: 157-8.
[136] Park IK, Qian D, Kiel M, Becker MW, Pihalja M, Weissman IL, et
al. Bmi-1 is required for maintenance of adult self-renewing
haematopoietic stem cells. Nature 2003; 423: 302-5.
[137] Chowdhury M, Mihara K, Yasunaga S, Ohtaki M, Takihara Y,
Kimura A. Expression of Polycomb-group (PcG) protein BMI-1
predicts prognosis in patients with acute myeloid leuk emia.
Leukemia 2007; 21: 1116-22.
[138] Mohty M, Yong AS, Szydlo RM, Apperley JF, Melo JV. The
polycomb group BMI1 gene is a molecular marker for pred icting
prognosis of chronic myeloid leukemia. Blood 2007; 110: 380-3.
[139] Wang GG, Song J, Wang Z, Dormann HL, Casadio F, Li H, et al.
Haematopoietic malignancies caused by dysregulation of a
chromatin-binding PHD finger. Nature 2009; 459: 847-51.
[140] Taverna SD, Li H, Ruthenburg AJ, Allis CD, Patel DJ. How
chromatin-binding modules interpret histone modifications: lessons
from professional pocket pickers. Nat Struct Mol Biol 2007; 14:
1025-40.
[141] Baker LA, Allis CD, Wang GG. PHD fingers in human diseases:
disorders arising from misinterpreting epigenetic marks. Mutat Res
2008; 647: 3-12.
[142] van Zutven LJ, Onen E, Velthuizen SC, van Drunen E, von Bergh
AR, van den Heuvel-Eibrink MM et al. Identification of NUP98
110 Current Drug Targets, 2010, Vol. 11, N o. 1 ten Cate et al.
abnormalities in acute leukemia: JARID1A (12p 13) as a new
partner gene. Genes Chromosomes Cancer 2006; 45: 437-46.
[143] Wang GG, Cai L, Pasillas MP, Kamp s MP. NUP98-NSD1 links
H3K36 methylation to Hox-A gene activation and
leukaemogenesis. Nat Cell Biol 2007; 9: 804-12.
[144] Moore MA, Chung KY, Plasilova M, Schuringa JJ, Shieh JH, Zhou
P, et al. NUP98 dysregulation in myeloid leukemogenesis. Ann N
Y Acad Sci 2007; 1106: 114-42.
[145] Wang GG, Song J, Wang Z, Dormann HL, Casadio F, Li H, et al.
Haematopoietic malignancies caused by dysregulation of a
chromatin-binding PHD finger. Nature 2009.
[146] Lessard J, Sauvageau G. Bmi-1 determines the proliferative
capacity of normal and leukaemic stem cells. Nature 2003; 423:
255-60.
[147] Liang G, Gonzales FA, Jones PA, Orntoft TF, Thykjaer T. Analysis
of gene induction in human fibroblasts and bladder cancer cells
exposed to the methylation inhibitor 5-aza-2'-deoxycytidine.
Cancer Res 2002; 62: 961-6.
[148] Kuendgen A, Gattermann N. Valproic acid for the treatment of
myeloid malignancies. Cancer 2007; 110: 943-54.
[149] Guo F, Sigua C, Tao J, Bali P, George P, Li Y, et al. Cotreatment
with histone deacetylase inhibitor LAQ824 enhances Apo-
2L/tumor necrosis factor-related apoptosis inducing ligand-induced
death inducing signaling complex activity and apoptosis of h uman
acute leukemia cells. Cancer Res 2004; 64: 2580-9.
[150] Morotti A, Ci lloni D, Messa F, Ar ruga F, Defilipp i I, Carturan S, et
al. Valproate enhances imatin ib-induced growth arrest and
apoptosis in chronic myeloid leukemia cells. Cancer 2006; 106:
1188-96.
[151] Yu C, Rahmani M, Conrad D, Subler M, Dent P, Grant S. The
proteasome inhibitor bortezomib interacts sy nergistically with
histone deacetylase inhibitors to indu ce apoptosis in Bcr/Abl+ cells
sensitive a nd resistan t to STI571. Blood 2003; 102: 3765-74.
[152] Saito Y, Liang G, Egger G, Friedman JM, Chuang JC, Coetzee GA,
et al. Specific activation of microRNA-127 with downregulation of
the proto-oncogene BCL6 by chromatin-modifying drugs in human
cancer cells. Cancer Cell 2006; 9: 435-43.
Received: May 19, 2009 Revised: June 19, 2009 Accepted: August 24, 2009
... Dysregulation of this process results in the development of diseases such as acute myeloid leukemia (AML), an aggressive clonal disease of abnormal HSCs, and primitive progenitors that blocks their myeloid differentiation to generate self-renewing leukemia stem cells (LSCs) (Testa, 2011). Furthermore, the presence of leukemia stem/initiating cells (LSCs/LICs) can lead to the occurrence or relapse of myeloid leukemia, which is likely to be a major cause of drug-resistant disease and relapse in AML patients (ten Cate et al., 2010). ...
... (1) Antibodies targeting surface markers of CSCs. Based on the identification of specific surface markers for CSCs, such as CD34 + /CD38 − , CD33, and CD44 + /CD24 − (Al-Hajj et al., 2003;ten Cate et al., 2010), antibodies against specific surface markers have been developed and even used in clinical settings. For example, as 80-90% of stem cells in AML express CD33, antibodies targeting CD33, such as gemtuzumab, became an important drug for the treatment of AML (Laing et al., 2017). ...
Article
Full-text available
N6-methyladenosine (m6A) is one of the most common internal RNA modifications in eukaryotes. It is a dynamic and reversible process that requires an orchestrated participation of methyltransferase, demethylase, and methylated binding protein. m6A modification can affect RNA degradation, translation, and microRNA processing. m6A plays an important role in the regulation of various processes in living organisms. In addition to being involved in normal physiological processes such as sperm development, immunity, fat differentiation, cell development, and differentiation, it is also involved in tumor progression and stem cell differentiation. Curiously enough, cancer stem cells, a rare group of cells present in malignant tumors, retain the characteristics of stem cells and play an important role in the survival, proliferation, metastasis, and recurrence of cancers. Recently, studies demonstrated that m6A participates in the self-renewal and pluripotent regulation of these stem cells. However, considering that multiple targets of m6A are involved in different physiological processes, the exact role of m6A in cancer progression remains controversial. This article focuses on the mechanism of m6A and its effects on the differentiation of cancer stem cells, to provide a basis for elucidating the tumorigenesis mechanisms and exploring new potential therapeutic approaches.
... Also, these strategies are favorable since they lead to smaller number of complications and target the bulk of blasts compared to chemotherapy [1]. For this purpose, much attention has been paid to the identification of the cell marker targets on the LSC cell surface [6]. Among the cell surface markers, CD33 antigens are highly promising candidates for AML targeted therapy. ...
... AML LSCs specifically possess CD34 + CD38 cell surface markers [19]. CD123 and CD33 markers are extremely promising candi-dates for AML targeted therapy among the abovementioned cell surface markers [6]. ...
Article
Full-text available
Acute myeloblastic leukemia (AML) is the most frequent acute leukemia in adulthood with very poor overall survival rates. In the past few decades, significant progresses had led to the findings of new therapeutic approaches and the better understanding of the molecular complexity of this hematologic malignancy. Leukemic stem cells (LSCs) play a key role in the initiation, progression, regression, and drug resistance of different types of leukemia. The cellular and molecular characteristics of LSCs and their mechanism in the development of leukemia had not yet been specified. Therefore, determining their cellular and molecular characteristics and creating new approaches for targeted therapy of LSCs is crucial for the future of leukemia research. For this reason, the recognition of surface maker targets on the cell surface of LSCs has attracted much attention. CD33 has been detected on blasts in most AML patients, making them an interesting target for AML therapy. Genetic engineering of T cells with chimeric antigen receptor (CAR-T cell therapy) is a novel therapeutic strategy. It extends the range of antigens available for use in adoptive T-cell immunotherapy. This review will focus on CAR-T cell approaches as well as monoclonal antibody (mAB)-based therapy, the two antibody-based therapies utilized in AML treatment.
... Toxin-based drugs have been mostly utilized in the treatment of hematological tumors because, being injected in the bloodstream, they can more easily reach target cancer cells. Numerous ITs formed by a toxic RIP domain like saporin, PAP, momordin and RTA, combined with a monoclonal antibody against markers of hematological malignancies such as CD2, CD19, CD22, CD25, CD30, CD38 and CD123 have been tested alone or in combination in animal models of lymphoma and leukemia [97][98][99][100][101][102] and some underwent early clinical phase studies. The anti-cancer activity of several ITs have been assayed also against solid tumors, but did not display the same successful results, likely due to the limited penetration within the tumor mass and relevant immunogenicity, the latter greatly reduced in hematological cancer patients. ...
... The clinical evaluation of the most promising targeted toxins is ongoing and, by exploiting novel discoveries and technologies in the biomedical fields, new toxin-related therapeutic options could be envisaged. Noteworthy examples are described below and summarized in Figure 7. Toxins 2017, 9, 314 14 of 33 formed by a toxic RIP domain like saporin, PAP, momordin and RTA, combined with a monoclonal antibody against markers of hematological malignancies such as CD2, CD19, CD22, CD25, CD30, CD38 and CD123 have been tested alone or in combination in animal models of lymphoma and leukemia [97][98][99][100][101][102] and some underwent early clinical phase studies. The anti-cancer activity of several ITs have been assayed also against solid tumors, but did not display the same successful results, likely due to the limited penetration within the tumor mass and relevant immunogenicity, the latter greatly reduced in hematological cancer patients. ...
Article
Full-text available
Plant ribosome-inactivating protein (RIP) toxins are EC3.2.2.22 N-glycosidases, found among most plant species encoded as small gene families, distributed in several tissues being endowed with defensive functions against fungal or viral infections. The two main plant RIP classes include type I (monomeric) and type II (dimeric) as the prototype ricin holotoxin from Ricinus communis that is composed of a catalytic active A chain linked via a disulphide bridge to a B-lectin domain that mediates efficient endocytosis in eukaryotic cells. Plant RIPs can recognize a universally conserved stem-loop, known as the α-sarcin/ ricin loop or SRL structure in 23S/25S/28S rRNA. By depurinating a single adenine (A4324 in 28S rat rRNA), they can irreversibly arrest protein translation and trigger cell death in the intoxicated mammalian cell. Besides their useful application as potential weapons against infected/tumor cells, ricin was also used in bio-terroristic attacks and, as such, constitutes a major concern. In this review, we aim to summarize past studies and more recent progresses made studying plant RIPs and discuss successful approaches that might help overcoming some of the bottlenecks encountered during the development of their biomedical applications.
... Zhao et al. generated specific mAbs against CLL-1 and demonstrated their direct cytotoxic and anticancer activity in vitro and in vivo [63]. Furthermore, among LSC-associated surface antigens exploited for LSC-selective therapy, CD44, CD47, and CD123 are the best known [64]. ...
Article
Full-text available
Acute myeloid leukemia (AML) is a malignant disease of hematopoietic precursors at the earliest stage of maturation, resulting in a clonalproliferation of myoblasts replacing normal hematopoiesis. AML represents one of the most common types of leukemia, mostly affecting elderly patients. To date, standard chemotherapy protocols are only effective in patients at low risk of relapse and therapy-related mortality. The average 5-year overall survival (OS) is approximately 28%. Allogeneic hematopoietic stem cell transplantation (HSCT) improves prognosis but is limited by donor availability, a relatively young age of patients, and absence of significant comorbidities. Moreover, it is associated with significant morbidity and mortality. However, increasing understanding of AML immunobiology is leading to the development of innovative therapeutic strategies. Immunotherapy is considered an attractive strategy for controlling and eliminating the disease. It can be a real breakthrough in the treatment of leukemia, especially in patients who are not eligible forintensive chemotherapy. In this review, we focused on the progress of immunotherapy in the field of AML by discussing monoclonal antibodies (mAbs), immune checkpoint inhibitors, chimeric antigen receptor T cells (CAR-T cells), and vaccine therapeutic choices.
Chapter
CD33 is a myeloid differentiation marker antigen with immune functions associated with anti-inflammatory signaling, cell adhesion, and endocytosis. CD33 is expressed by the earliest myeloid progenitors and present during myelomonocytic differentiation. Thus, it is an immune function protein with anti-inflammatory signaling, cell adhesion, and endocytosis functions with sialic acid-modified proteins or lipids as ligands. Despite CD33’s importance in hematological cancers, the information is limited to the CD33 expression. CD33 (Siglec-3) is also a type I TM glycoprotein with 2 Ig-like extracellular domains [1, 2], as the smallest group of the siglecs with a 67 kDa transmembrane glycoprotein and recognizes both α2,3-SA and α2,6-SAs [3, 4]. For the protein sequence and structure of Siglec-3, the Ig-like-V-type region is suggested to be located on amino acid sequence No. 19–135. In the Ig-like-V-type region, amino acid No. 119, Arginine, is suggested to be the sialic acid-binding site. From amino acids 145–228 consists of Ig-like-C2 type region. The transmembrane region is located within amino acids 260–282. ITIM motif is in amino acids 338–343 region and ITIM-like motif is in 356–361 region, respectively (Fig. 12.1).
Chapter
Tumors of the uterine corpus include endometrial and mesenchymal tumors. Carcinoma of the endometrium is the most common female pelvic malignancy, the fourth most common cancer in women in the United States, and the sixth most common malignancy in women worldwide. This chapter summarizes important concepts regarding the pertinent anatomy, epidemiology, clinical presentation, staging, imaging evaluation, and treatment of these malignancies, which are essential to the successful multidisciplinary management of these patients. Imaging remains integral to the diagnosis, staging, and response assessment of these cancers, with the radiologist playing an essential role in patient care.
Article
Acute myeloid leukemia (AML) is an uncommon but potentially catastrophic diagnosis with historically high mortality rates. The standard of care treatment remained unchanged for decades; however, recent discoveries of molecular drivers of leukemogenesis and disease progression have led to novel therapies for AML. Ongoing research and clinical trials are actively seeking to personalize therapy by identifying molecular targets, discovering patient specific and disease specific risk factors, and identifying effective combinations of modalities and drugs. This review focuses on important updates in diagnostic and disease classifications that reflect new understanding of the biology of AML, its mutational heterogeneity, some important genetic and environmental risk factors, and new treatment options including cytotoxic chemotherapy, novel targeted agents, and cellular therapies.
Article
Acute myeloid leukemia (AML) remains the most lethal of leukemias and a small population of cells called leukemic stem cells (LSCs) has been associated with disease relapses. Some cell signaling pathways play an important role in AML survival, proliferation and self-renewal properties and are abnormally activated or suppressed in LSCs. This includes the NF-κB, Wnt/β-catenin, Hedgehog, Notch, EGFR, JAK/STAT, PI3K/AKT/mTOR, TGF/SMAD and PPAR pathways. This review aimed to discuss these pathways as molecular targets for eliminating AML LSCs. Herein, inhibitors/activators of these pathways were summarized as a potential new anti-AML therapy capable of eliminating LSCs to guide future researches. The clinical use of cell signaling pathways data can be useful to enhance the anti-AML therapy.
Chapter
For the treatment of different leukemia different chemotherapies are available. However the success rate of any particular drug scheduling may vary with leukemic condition. In general, low dose of chemotherapy is suggested for chronic leukemia, whereas application of high dose (myeloablative) chemotherapy is applied for acute and vigorous type of leukemia. In present work we have shown that chronic type of leukemia is controlled; however, for controlling vigorously growing leukemia is a challenge due to chemotherapeutic toxicity to the normal cells of the hematopoietic system. Hence for its management, we developed a control analysis model. This model may help to design an optimal chemotherapeutic schedule so that the controlling of the vigorously growing leukemic growth can be possible in one hand with the sustenance of the normal non-leukemic cell population on the other hand. This work shows that for long-term chemotherapeutic success in individual leukemic patients demands a judicious choice of drug dosing strategy that may determine the trade-off between leukemic growth and restoration time of normal cell population of the hematopoietic system.
Article
Acute myeloid leukemia (AML) patients show high relapse rates and some develop conventional chemotherapy resistance. Leukemia Stem Cells (LSCs) are the main player for AML relapses and drug resistance. K562/ADM cell, a leukemia resistant cell subtype, was used as leukemia multidrug resistant model to explore drug resistance of leukemia cells. We found that CD34⁺CD38⁻, CD34⁺CD38⁻CD123⁺, cell surface markers of LSCs were significantly high expressed in K562/ADM cell, not in K562 cell with less drug resistance; compared with LSCs from K562, multidrug resistance protein P-gp and BCRP expression were significantly higher in K562/ADM cell. Compared to the patients with primary leukemia, the subpopulation of LSCs from patients with AML relapses and drug resistance is significantly increased. Arsenic trioxide (As2O3) as an inhibitor markedly inhibited proliferation and induced apoptosis of LSCs from K562/ADM cell in a dose dependent way. The results of colony formation assay showed that As2O3 obviously inhibited colony formation of LSCs from K562/ADM cell. Collectively, our study showed that LSCs take an important role in the drug resistance of leukemia therapies. As2O3, as an inhibition agent, may be a potential drug targeting LSCs for improving the cure rate.
Article
Full-text available
Recent studies suggest that the population of malignant cells found in human acute myelogenous leukemia (AML) arises from a rare population of leukemic stem cells (LSCs). LSCs have been documented for nearly all AML subtypes and have been phenotypically described as CD34+/CD38- or CD34+/HLA-DR-. Given the potentially critical role of these primitive cells in perpetuating leukemic disease, we sought to further investigate their molecular and cellular characteristics. Flow cytometric studies using primary AML tissue showed that the interleukin-3 receptor alpha chain (IL-3Ralpha or CD123) was strongly expressed in CD34+/CD38- cells (98 +/- 2% positive) from 16 of 18 primary specimens. Conversely, normal bone marrow derived CD34+/CD38- cells showed virtually no detectable expression of the CD123 antigen. To assess the functional role of IL-3Ralpha positive cells, purified CD34+/CD123+ leukemia cells were transplanted into immune deficient NOD/SCID mice. These experiments showed that CD123+ cells were competent to establish and maintain leukemic populations in vivo. To begin to elucidate a biological role for CD123 in leukemia, primary AML samples were analyzed with respect to signal transduction activity in the MAPK, Akt, and Stat5 pathways. Phosphorylation was not detected in response to IL-3 stimulation, thereby suggesting CD123 is not active in conventional IL-3-mediated signaling. Collectively, these data indicate that CD123 represents a unique marker for primitive leukemic stem cells. Given the strong expression of this receptor on LSCs, we propose that targeting of CD123 may be a promising strategy for the preferential ablation of AML cells.
Article
Full-text available
The specific retention of intravenously administered hemopoietic cells within bone marrow is a complex multistep process. Despite recent insights, the molecular mechanics governing this process remain largely undefined. This study explored the influence of β2-integrins on the homing to bone marrow and repopulation kinetics of progenitor cells. Both antifunctional antibodies and genetically deficient cells were used. In addition, triple selectin-deficient mice were used as recipients of either deficient (selectin or β2) or normal cells in homing experiments. The homing patterns of either β2 null or selectin null cells into normal or selectin-deficient recipients were similar to those of normal cells given to normal recipients. Furthermore, spleen colony-forming units and the early bone marrow repopulating activity for the first 2 weeks after transplantation were not significantly different from those of control cells. These data are in contrast to the importance of β2-integrin and selectins in the adhesion/migration cascade of mature leukocytes. The special bone marrow flow hemodynamics may account for these differences. Although early deaths after transplantation can be seen in recipients deficient in CD18 and selectin, these are attributed to septic complications rather than homing defects. However, when β2- or selectin-null donor cells are treated with anti-α4 antibodies before their transplantation to normal or selectin-deficient recipients, a dramatic inhibition of homing (>90%) was found. The data suggest that the α4β1/vascular cell adhesion molecule-1 pathway alone is capable of providing effective capture of cells within the bone marrow, but if its function is compromised, the synergistic contribution of other pathways, that is, β2-integrins or selectins, is uncovered.
Article
Full-text available
Antibody-targeted chemotherapy is a promising therapy in patients with acute myeloid leukemia (AML). In a phase II study of Mylotarg (CMA-676, gemtuzumab ozogamicin), which consists of a CD33 antibody linked to calicheamicin, satura- tion and internalization by leukemic and normal myeloid cells were analyzed in 122 patients with relapsed AML. Periph- eral blood samples were obtained just before and 3 and 6 hours after the start of the first and second Mylotarg treatment cycles. Within 3 to 6 hours after infusion, near complete saturation of CD33 anti- genic sites by Mylotarg was reached for AML blasts, monocytes, and granulo- cytes, whereas Mylotarg did not bind to lymphocytes. Saturation levels prior to the start of the second Mylotarg treat- ment cycle were significantly increased compared with background levels before the start of the first cycle. This apparently was caused by remaining circulating My- lotarg from the first treatment cycle (;2 weeks earlier). On binding of Mylotarg to the CD33 antigen, Mylotarg was rapidly internalized, as determined by the de- crease in maximal surface membrane My- lotarg binding. Internalization of Mylotarg was also demonstrated in myeloid cells in vitro and was confirmed by confocal laser microscopy. In vitro studies using pulse labeling with Mylotarg showed a continuous renewed membrane expression of CD33 an- tigens, which can significantly increase the internalization process and thereby the intra- cellular accumulation of the drug. Finally, Mylotarg induced dose-dependent apopto- sis in myeloid cells in vitro. These data indicate that Mylotarg is rapidly and specifi- cally targeted to CD331 cells, followed by internalization and subsequent induction of cell death. (Blood. 2001;97:3197-3204)
Article
Full-text available
Purpose: To assess the safety and tolerability, pharmacokinetics, and early evidence of antitumor activity of escalating doses of lexatumumab (HGS-ETR2), a fully human agonistic monoclonal antibody which targets and activates the tumor necrosis factor-related apoptosis-inducing ligand receptor 2 (TRAIL-R2) in patients with advanced solid malignancies. Experimental design: In this phase 1, open label study, patients with advanced solid malignancies were treated with escalating doses of lexatumumab administered i.v. over 30 to 120 min every 21 days. A cohort of four patients, which could be expanded to six patients, was studied at each dose level. The dose-limiting toxicity (DLT) dose was defined as the dose at which the incidence of DLT in the first two cycles was >or=33%. The maximum tolerated dose was defined as the highest dose at which <33% of subjects experienced DLT. The pharmacokinetics and immunogenicity of lexatumumab were also characterized. Tumor specimens from historical or current biopsies, when available, were stained for TRAIL-R2 using immunohistochemistry techniques. Results: Thirty-seven patients received 120 cycles of lexatumumab at doses ranging from 0.1 to 20 mg/kg every 21 days as of May 2006. The 20 mg/kg dose was identified as the DLT dose based on DLTs in three of seven patients treated with this dose; DLTs included asymptomatic elevations of serum amylase, transaminases, and bilirubin. The 10 mg/kg dose cohort was expanded to 12 patients and the 10 mg/kg dose was identified as the maximum tolerated dose. The mean (+/-SD) clearance and apparent terminal half-life values at the 10 mg/kg dose averaged 6.0 (2.9) mL/d/kg and 16.4 (10.9) days, respectively. Twelve patients had durable stable disease that lasted a median of 4.5 months, including three patients with sarcoma having prolonged stable disease (>or=6.7 months). Immunohistochemistry for TRAIL-R2 showed specific staining in >10% of tumor cells for 16 of the 20 evaluable specimens submitted (80%). Conclusions: Lexatumumab was safe and well tolerated at doses up to and including 10 mg/kg every 21 days. Lexatumumab was associated with sustained stable disease in several patients. Pharmacokinetics were linear over the dose range studied, and consistent with a two-compartment model with first-order elimination from the central compartment. Additional evaluation of this novel apoptosis-inducing agent, particularly in combination with chemotherapy agents, is warranted and ongoing.
Article
Present studies demonstrate that treatment with the histone deacetylases inhibitor LAQ824, a cinnamic acid hydroxamate, increased the acetylation of histones H3 and H4, as well as induced p21WAF1 in the human T-cell acute leukemia Jurkat, B lymphoblast SKW 6.4, and acute myelogenous leukemia HL-60 cells. This was associated with increased accumulation of the cells in the G1 phase of the cell cycle, as well as accompanied by the processing and activity of caspase-9 and -3, and apoptosis. Exposure to LAQ824 increased the mRNA and protein expressions of the death receptors DR5 and/or DR4, but reduced the mRNA and protein levels of cellular FLICE-inhibitory protein (c-FLIP). As compared with treatment with Apo-2L/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) or LAQ824 alone, pretreatment with LAQ824 increased the assembly of Fas-associated death domain and caspase-8, but not of c-FLIP, into the Apo-2L/TRAIL-induced death-inducing signaling complex. This increased the processing of caspase-8 and Bcl-2 interacting domain (BID), augmented cytosolic accumulation of the prodeath molecules cytochrome-c, Smac and Omi, as well as led to increased activity of caspase-3 and apoptosis. Treatment with LAQ824 also down-regulated the levels of Bcl-2, Bcl-xL, XIAP, and survivin. Partial inhibition of apoptosis due to LAQ824 or Apo-2L/TRAIL exerted by Bcl-2 overexpression was reversed by cotreatment with LAQ824 and Apo-2L/TRAIL. Significantly, cotreatment with LAQ824 increased Apo-2L/TRAIL-induced apoptosis of primary acute myelogenous leukemia blast samples isolated from 10 patients with acute myelogenous leukemia. Taken together, these findings indicate that LAQ824 may have promising activity in augmenting Apo-2L/TRAIL-induced death-inducing signaling complex and apoptosis of human acute leukemia cells.
Article
In clinical trials, the tyrosine kinase inhibitor STI571 has proven highly effective in reducing leukemic cell burden in chronic myeloid leukemia (CML). The overall sensitivity of CML CD34+ progenitor cells to STI571 and the degree to which cell death was dependent on cell cycle status were determined. Stem cells (Lin−CD34+) from the peripheral blood of patients with CML in chronic phase and from granulocyte–colony-stimulating factor–mobilized healthy donors were labeled with carboxy-fluorescein diacetate succinimidyl diester dye to enable high-resolution tracking of cell division. Then they were cultured for 3 days with and without growth factors ± STI571. After culture, the cells were separated by fluorescence-activated cell sorting into populations of viable quiescent versus cycling cells for genotyping. For healthy controls, in the presence of growth factors, STI571 affected neither cell cycle kinetics nor recovery of viable cells. In the absence of growth factors, normal cells were unable to divide. For CML samples, in the presence or absence of growth factors, the response to STI571 was variable. In the most sensitive cases, STI571 killed almost all dividing cells; however, a significant population of viable CD34+ cells was recovered in the undivided peak and confirmed to be part of the leukemic clone. STI571 also appeared to exhibit antiproliferative activity on the quiescent population. These studies confirm that CML stem cells remain viable in a quiescent state even in the presence of growth factors and STI571. Despite dramatic short-term responses in vivo, such in vitro insensitivity to STI571, in combination with its demonstrated antiproliferative activity, could translate into disease relapse after prolonged therapy.
Article
Valproic acid (VPA) has been used as an anticonvulsant for decades. Recently, it was demonstrated that VPA also acts as a histone deacetylase inhibitor and induces differentiation and apoptosis in a variety of malignant cells in vitro. The effect of VPA on tumor cells differs according to cell type, degree of differentiation, and underlying genetic alterations. Clinical trials with VPA have focused on acute myeloid leukemia and the myelodysplastic syndromes. When it was used as monotherapy or in combination with all-trans retinoic acid, which synergizes in vitro, VPA achieved hematologic improvement in a subset of patients. Similar to other inhibitors of histone deacetylases, complete or partial remissions rarely were observed. In this report, the authors reviewed the in vitro and in vivo data obtained with VPA, and they considered possible combination regimens aimed at improving therapeutic efficacy. Cancer 2007;110:943-54. (c) 2007 American Cancer Society.
Article
Philadelphia chromosome-positive (Ph+) chronic myeloid leukemia (CML) induced by the BCR-ABL oncogene is believed to be developed from leukemic stem cells (LSCs), and we have previously shown in mice that LSCs for CML express the same cell surface markers that are also expressed on normal hematopoietic stem cells (HSCs). Although the inhibition of BCR-ABL kinase activity by imatinib is highly effective in treating human Ph+ CML in chronic phase, it is difficult to achieve molecular remission of the disease, suggesting that LSCs remain in patients. In this study, we find that following imatinib treatment, LSCs not only remained but also accumulated increasingly in bone marrow of CML mice. This insensitivity of LSCs to imatinib was not because of the lack of BCR-ABL kinase inhibition by imatinib, and proliferating leukemic cells derived from LSCs were still sensitive to growth inhibition by imatinib. These results identify an LSC survival pathway that is not inhibited by imatinib. Furthermore, we show that β-catenin in the Wnt signaling pathway is essential for survival and self-renewal of LSCs, providing a new strategy for targeting these cells.