Download Eradication of Leukemia Stem Cells as a New Goal of Therapy in

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts
no text concepts found
Transcript
Review
Eradication of Leukemia Stem Cells as a New Goal of
Therapy in Leukemia
Farhad Ravandi and Zeev Estrov
Abstract
Leukemias have traditionally been classified and treated on the basis of phenotypic characteristics,
such as morphology and cell-surface markers, and, more recently, cytogenetic aberrations. These
classification systems are flawed because they do not take into account cellular function. The
leukemia cell population is functionally heterogeneous: it consists of leukemia stem cells (LSC)
and mature leukemia cells that differentiate abnormally to varying extents. Like normal hematopoietic stem cells, LSCs are quiescent and have self-renewal and clonogenic capacity. Because
they are quiescent, LSCs do not respond to cell cycle ^ specific cytotoxic agents used to treat
leukemia and so contribute to treatment failure.These cells may undergo mutations and epigenetic
changes, further leading to drug resistance and relapse. Recent data suggest that mature leukemia cells may acquire LSC characteristics, thereby evading chemotherapeutic treatment and
sustaining the disease. Ongoing research is likely to reveal the molecular mechanisms responsible
for LSC characteristics and lead to novel strategies for eradicating leukemia.
The hematopoietic system is thought to originate from
pluripotent hematopoietic stem cells (HSC) capable of
producing a hierarchy of downstream multilineage and unilineage progenitor cells that differentiate into mature cells
(1, 2). HSCs have self-renewal and clonogenic abilities and
can differentiate into multiple lineages. HSCs with the capacity for both long-term and short-term repopulation of the
mouse hematopoietic system have been characterized. These
rare cells give rise to progenitor cells, which are destined to
generate fully differentiated cells. HSC self-renewal is thought
to be either symmetrical, producing two daughter HSCs, or
asymmetrical, producing an identical HSC and a progenitor
with diminished self-renewal capacity but with the ability to
enact clonal expansion and maintain the circulating blood cell
population (1 – 3).
It has recently been hypothesized that leukemogenesis, and,
for that matter, carcinogenesis, arises from neoplastic stem
cells. Leukemia stem cells (LSC) exhibit characteristics similar
to those of normal HSCs and are thought to arise from normal
stem cells through the accumulation of oncogenic insults (4, 5).
However, LSCs may also arise from differentiated progenitor
cells that have reacquired the capacity for self-renewal (6 – 8).
Like normal HSCs, LSCs give rise to differentiated daughter cells
that lose their self-renewal capacity. However, defects in their
cellular machinery usually eliminate their ability to differentiate
fully into morphologically and phenotypically mature cells.
As a result, the leukemic population consists of undifferentiated
and variably differentiated leukemia cells. The degree of
differentiation of leukemia cells has traditionally formed the
basis of the morphologic classification of leukemias (9, 10).
The generally accepted paradigm of leukemogenesis rests on
the theory that leukemia arises from a single cell and is
maintained by a small population of LSCs (5, 11, 12). Studies
of acute myeloid leukemia (AML) have shown that only 0.1%
to 1% of AML cells have the capacity to initiate leukemia when
injected into severe combined immunodeficient mice (4, 13). It
has long been recognized that, like solid tumors, AML consists
of a heterogeneous population of cells with a small percentage
of noncycling, quiescent cells (14, 15). Two models of leukemogenesis have long been proposed. According to the stochastic model, leukemia consists of a homogeneous population
of immature cells and a few cells that can either self-renew or
proliferate in a stochastic manner (3, 16, 17). According to
the hierarchy model, leukemia consists of a heterogeneous
population, within which only a small percentage of LSCs
sustain the disease. Recently, a third model was proposed.
According to this model, mature leukemia cells can dedifferentiate and regain LSC capacity (6, 8, 18). Whereas the first two
models hold that only LSCs sustain leukemia, the third model
allows that mature cells can regain self-renewal capacity,
thereby sustaining the disease.
Despite the development of multiple new agents that are
effective at reducing the tumor burden in patients with
leukemia, relapse continues to be the most common cause
of death, particularly in patients with AML. Current efforts
directed at detecting and quantifying minimal residual disease
are based on the assumption that eradication of all cells capable
of sustaining the disease will improve treatment outcomes.
Modern techniques for detecting minimal residual disease, such
Authors’ Affiliation: Department of Leukemia, The University of Texas M.D.
Anderson Cancer Center, Houston,Texas
Received 8/29/05; revised 10/12/05; accepted 11/3/05.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
Requests for reprints: Farhad Ravandi, Department of Leukemia, Unit 428, The
University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard,
Houston, TX 77030. Phone : 713-745-0394; Fax : 713-745-4612; E-mail:
fravandi@ mdanderson.org.
F 2006 American Association for Cancer Research.
doi:10.1158/1078-0432.CCR-05-1879
Clin Cancer Res 2006;12(2) January 15, 2006
340
www.aacrjournals.org
Downloaded from clincancerres.aacrjournals.org on June 18, 2017. © 2006 American Association for Cancer
Research.
Leukemia Stem Cells as Therapeutic Targets
as PCR, are not sensitive enough to detect all residual leukemic
cells. Therefore, a state of absolute disease eradication cannot
be determined with the available tests. For example, disease
recurrence has been reported after discontinuation of therapy
with imatinib mesylate in patients with chronic myelogenous
leukemia who have achieved a complete molecular remission
(19). On the other hand, minimal residual disease will not
lead to disease recurrence if the residual leukemic cells are not
disease-sustaining or if the residual LSCs remain dormant
(20, 21). Here, we examine the role of LSCs as applied to
treatment strategies aimed at curing leukemia.
Defining LSCs
The phenotypic and functional properties of normal HSCs
have been extensively studied (22, 23). Several combinations of
cell-surface markers, such as Lin Thy+CD34+CD38 /low, have
been used to identify populations that are enriched with
HSCs (24). Other techniques include the extrusion of Hoechst
dye 33342 to identify side population cells, the use of
immature cell-surface markers such as CD133 and CD150,
and the use of Bodipy aminoacetaldehyde to assess aldehyde
dehydrogenase activity (25 – 30). It was shown that, for most
AML subtypes (with the exception of acute promyelocytic
leukemia), the cells that initiate the disease in non-obese
diabetic/severe combined immunodeficient mice have a
CD34+CD38 or CD34+CD38+/low phenotype (4, 31). However, leukemic cells bearing these phenotypes are dissimilar
functionally and include both short-term and long-term
leukemia-initiating cells (Fig. 1). Several studies have suggested
that HSCs and LSCs share some cell-surface markers but not
others. For example, HSCs and LSCs both express CD34
but not CD71 and HLA-DR. However, Thy-1 (CD90) and c-Kit
(CD117) are expressed on HSCs but not on LSCs, and CD123
and interleukin-3 receptor-a are expressed on LSCs but not
on HSCs (31 – 33). Remarkably, cytogenetically abnormal
leukemia-initiating cells have been found in the CD34+CD90+
population from several patients with AML and, in rare
cases, CD34 cells as well as CD34+ cells have successfully
engrafted and initiated human leukemia in mice (34 – 36). In
acute promyelocytic leukemia, unlike in other myeloid leukemias, the characteristic translocation has been observed in
CD34 CD38+ but not in CD34+CD38 cells (4, 37). Therefore,
a universal phenotype for LSCs may not exist and patientto-patient variations in cell-surface protein expression may be
the rule.
Of note, a similar heterogeneity exists in HSC gene
expression: genes thought to form a stem cell gene signature
have been identified; however, dissimilar data suggest that the
existence of such a signature may be premature (38 – 40).
Because stem cell – specific properties, such as self-renewal,
quiescence, and proliferation, are not governed by genes that
are specific to stem cells, it was proposed that there is a ‘‘stem
cell state’’ rather than a ‘‘stem cell portrait’’ in the hematopoietic
system (41). The stem cell state may represent a transient and
potentially reversible state that cells can assume in response to
the correct trigger. Using a similar concept, one can argue that
there is an ‘‘LSC state’’ rather than an ‘‘LSC phenotype,’’ a
concept that is supported by the well-described lineage
‘‘infidelity’’ in human leukemias (42). Although defining the
sets of conditions that pertain to the LSC state may allow the
www.aacrjournals.org
Fig. 1. Proposed concepts and properties of leukemic stem cells (2, 5). SL-IC,
SCID-leukemia initiating cell; AML-CFU, AML colony-forming units.
development of strategies to eliminate LSCs or render them
inconsequential, the identification of specific genes and surface
markers expressed solely by LSCs may be difficult or
impossible.
Molecular Pathways Regulating HSCs and LSCs
It is generally accepted that self-renewal is the hallmark
property of stem cells in both normal and neoplastic tissues.
Recent research has delineated molecular pathways that
regulate the self-renewal capacity of HSCs. Overexpression
and knockout experiments have identified several genes,
transcription factors, and cell cycle regulators that modulate
the self renewal and differentiation of HSCs (43, 44).
Genes such as SCL, GATA-2, LMO-2, and AML-1 (also known
as CBFA2 or RUNX1) govern the transcriptional regulation
of early hematopoiesis, and the deregulation of these genes
through chromosomal aberrations leads to the genesis of
several hematopoietic malignancies. For example, the gene
encoding the transcription factor SCL is the most frequent target
of chromosomal rearrangements in children with T-cell acute
lymphoblastic leukemia (45). SCL is normally expressed in
HSCs and immature progenitors and is down-regulated as
differentiation proceeds. As a result of chromosomal translocations, SCL is inappropriately expressed and, through
collaboration with other oncoproteins, initiates malignant
transformation (46). Similarly, transcriptional activation of
the AML-1 gene is required for definitive hematopoiesis. As a
result of translocation t(8;21), the fusion protein AML-ETO,
one of the most frequent chromosomal abnormalities in AML,
is generated (47). Constitutive expression of AML-ETO has been
shown to increase the frequency of self-renewal in stem cells
(48). Of interest, such increased self-renewal is of no apparent
pathogenic consequence, presumably because secondary mutations are necessary for the expression of the leukemic
phenotype (49).
Other transcription factors such as the Homeobox (Hox)
genes, including HoxB4, and the Wnt signaling pathway have
341
Clin Cancer Res 2006;12(2) January 15, 2006
Downloaded from clincancerres.aacrjournals.org on June 18, 2017. © 2006 American Association for Cancer
Research.
Review
recent years suggest that a small population of LSCs, through
their HSC-like properties, survive chemotherapy and sustain
the disease (3, 66). High levels of ATP-binding cassette
transporters have been reported in both normal and cancer
stem cells (68, 69). Indeed, HSCs, but not lineage-committed
progenitors, express high levels of genes responsible for
multidrug resistance-related transporters (70). As mentioned
earlier, the efflux of fluorescent dyes such as Hoechst 33342 has
been used to isolate HSCs (25, 71). Similarly, LSCs either
inherently possess drug resistance mechanisms or acquire them
through mutations (66).
Other properties of LSCs are also likely to contribute to
drug resistance and relapse. Stem cell progenies such as longterm culture-initiating cells and AML colony-forming units
are in an active cell cycle (72, 73) whereas several studies
have clearly shown that LSCs remain quiescent (65, 74 – 76).
For example, a study by Guzman et al. (75) showed that as
much as 96% of the LSC population, as defined by the phenotype CD34+CD38 CD123+, were in the G0 phase of the cell
cycle. This resting status of the putative LSCs protects them
from the commonly used cell cycle – specific chemotherapeutic
agents.
It is likely that secondary events, such as the development of
mutations, further contribute to the intrinsic resistant properties of LSCs. In a multistep pathogenic process, quiescent
LSCs may carry the initial mutagenic event leading to genomic
instability and the induction of secondary mutations that are
responsible for a more resistant phenotype. Alternatively,
random secondary mutations or mutations occurring as a
result of selective pressure caused by therapy may contribute to
disease progression or resistance. This has been seen in patients
with chronic myelogenous leukemia (CML) treated with
imatinib mesylate, in whom mutations of the ATP-binding site
of BCR-ABL are well documented (77).
well-described roles in regulating the self-renewal and differentiation of HSCs (35, 50). HoxB4 promotes the expansion
of HSCs whereas they retain their ability to differentiate into
normal lymphoid and myeloid cells (51). It is abundantly
expressed in HSCs but declines as terminal differentiation
proceeds (51). Of note, deregulated expression of Hox family
members such as HoxA9 is commonly observed in AML
(52, 53). The Wnt signaling pathway has been shown to be
critical to the development of several organs and recent studies
have illustrated its important role in the regulation of hematopoietic stem and progenitor cell function (35, 54). Overexpression of h-catenin, a downstream activator of the Wnt
signaling pathway, expands the transplantable HSC pool in
long-term cultures (3). Furthermore, activation of Wnt signaling also increases the expression of other transcription factors
and cell cycle regulators important in HSC renewal, such as
HoxB4 and Notch-1 (35, 55).
The Notch/Jagged pathway is important in regulating the
integration of extracellular regulatory signals controlling
HSC fate (56). Ligand binding leads to proteolytic cleavage
and transport of the intracellular domain of Notch into the
nucleus, where it functions as a transcription activator.
Members of the Notch family have critical roles in keeping
HSCs in an undifferentiated state and may act as a gatekeeper
for factors governing self-renewal and lineage commitment
(57). Of interest, the gene encoding the Notch receptor was
originally identified as the gene rearranged by recurrent
chromosomal translocations in some patients with T-cell acute
lymphoblastic leukemia (58). Other transcription factors and
cell cycle regulators associated with oncogenesis, such as Bmi-1
and Sonic hedgehog (Shh), may play roles in the regulation
of proliferation of both HSCs and LSCs (59, 60). Bmi-1 is
a member of the Polycomb family of genes thought to be
responsible for the preservation of gene silencing (61). It is
highly expressed in purified HSCs and its expression declines
with differentiation (62). It seems to regulate stem cell renewal
by modulating other genes that are important in cellular
functions such as proliferation, survival, and lineage commitment (62). Bmi-1 has an essential role in regulating the
proliferative potential of leukemic stem cells (63). Although
direct evidence for the role of Shh in the regulation of stem cell
renewal is lacking, in vitro studies have shown increased selfrenewal of HSCs in response to Shh albeit in combination with
other growth factors (64).
Therefore, differential expression of several transcription
factors controls the fate of HSCs and plays a critical role in
the determination of self-renewal, differentiation, and lineage
commitment. These pathways are under the control of various
intracellular stimuli as well as cytokines and stromal factors
from adjacent cells in the bone marrow microenvironment.
Further studies of these transcription factors in HSCs and the
mechanisms causing their deregulation are likely to provide us
with better targets for development of disease-specific therapies.
Strategies to Overcome LSC Resistance to Therapy
As implied by the multistep theory of carcinogenesis, LSCs
are likely to have the fewest number of molecular aberrations in
the population of the malignant cells and, as such, biologically
most similar to normal HSCs. The primary challenge in
developing treatment strategies targeted toward LSCs is to
identify proapoptotic stimuli that spare the normal HSCs while
exerting the desired effect on LSCs.
A primary concern in the development of tumor stem cell –
specific drugs is to overcome the inherent drug efflux pumps
that are highly expressed in LSCs. Several agents effective
in inhibiting the ATP-binding cassette transporters have been
studied and found to have limited clinical efficacy (78, 79). The
biggest obstacle to this approach is the similarly high
expression of these transporters in normal HSCs, making them
equally susceptible to the inhibitors (66). As such, strategies
directed at pathways that specifically regulate LSC survival
would probably be more fruitful (80). The search for
identification of survival pathways that are preferentially
overexpressed in LSCs is ongoing and several recent studies
have described means of differential activation of apoptosis
mechanisms in LSCs (75, 81, 82).
The transcription factor nuclear factor nB was found to
be constitutively activated in LSCs but not in normal HSCs
(75). Therefore, nuclear factor nB inhibitors were added to
The Role of LSCs in Drug Resistance and Relapse
Normal HSCs possess several characteristics that protect
them from potential insults. LSCs have similar properties,
including quiescence (65), resistance to drugs and toxins
through the expression of ATP-associated transporters (66),
and resistance to apoptotic stimuli (67). Data accumulated in
Clin Cancer Res 2006;12(2) January 15, 2006
342
www.aacrjournals.org
Downloaded from clincancerres.aacrjournals.org on June 18, 2017. © 2006 American Association for Cancer
Research.
Leukemia Stem Cells as Therapeutic Targets
used to monitor minimal residual disease. The clinical benefits
of early identification of minimal residual disease using these
techniques remain uncertain and are under investigation.
Despite significant progress, relapse continues to be the
most prominent factor responsible for the failure of therapy
in leukemia, particularly in AML. Recent insights into the
nature of normal and malignant stem cells have led to the
identification of quiescent and drug-resistant LSCs as the likely
minimal residual disease candidates responsible for relapse.
In the appropriate setting, assays such as PCR may identify
residual cells expressing leukemia-specific molecular abnormality; however, these techniques cannot currently distinguish
LSCs from their more mature progeny. As such, further research
should be focused on better defining the LSC state.
Characterization of the molecular and biological features of
the cells that initiate and maintain leukemia is an essential step
in the development of novel agents effective against the disease.
Skeptics may ignore the above-mentioned evidence and argue
that LSCs are simply leukemia cells previously referred to as
resting in the G0 phase of the cell cycle. Others may concentrate
their efforts on identifying a ‘‘stem cell phenotype,’’ thus
ignoring the ‘‘stem cell state’’ and the potential fluidity of
LSCs (a process in which the leukemia progenitors regain stem
cell properties). Future studies of LSC biology, concentrating
on the function of LSCs rather than strictly on their phenotype,
are likely to identify new molecular targets and effective
treatment modalities directed at LSCs.
antileukemic agents such as idarubicin in experimental models
(75, 81). Recently, single-agent parthenolide, a potent
inhibitor of nuclear factor nB, was found to induce apoptosis
in AML and CML LSCs and progenitors while sparing normal
HSCs (82). Notably, parthenolide was much more selective
in eliminating LSCs and sparing normal hematopoietic cells
than was the standard chemotherapy agent cytarabine (82).
Constitutive activation of the phosphatidylinositide-3 kinase is
also necessary for the survival of LSCs and its pharmacologic
inhibition by LY294002 leads to a dose-dependent decrease in
survival (83).
The fate of LSCs depends on the relative expression of
transcription factors and their regulation, usually by aberrant
signaling pathways (84, 85). Although it has been proposed
that recruitment of LSCs from the G0 to the S phase of the cell
cycle might contribute to their eradication by cell cycle – specific
cytotoxic agents, it is possible that prolonging the ‘‘quiescent
phase’’ could be beneficial. One could envision a scenario in
which LSCs are maintained in a state of hibernation, thereby
prolonging relapse-free survival. The best evidence for this
possibility is the existence of patients with clinically distinct
‘‘indolent’’ and ‘‘proliferative’’ forms of AML as well as the wide
variation in the duration of relapse-free interval in individual
patients.
Future Directions
Over the past several decades, the mainstay of leukemia
therapy (and treatment of other tumors) has been to induce a
complete remission and to consolidate this with further courses
of chemotherapy. Recently, flow cytometry and PCR have been
Acknowledgments
We thank David Galloway for editing this manuscript.
References
1. Dick JE. Stem cells: self-renewal writ in blood. Nature
2003;423:231 ^ 3.
2. Warner JK, Wang JC, Hope KJ, Jin L, Dick JE. Concepts of human leukemic development. Oncogene
2004;23:7164 ^ 77.
3. Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem
cells, cancer, and cancer stem cells. Nature 2001;414:
105 ^ 11.
4. 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.
5. 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.
6. Jamieson CH, Ailles LE, Dylla SJ, et al. Granulocytemacrophage progenitors as candidate leukemic stem
cells in blast-crisis CML. N Engl J Med 2004;351:
657 ^ 67.
7. Korbling M, Estrov Z. Adult stem cells for tissue
repairDa new therapeutic concept ? N Engl J Med
2003;349:570 ^ 82.
8. Cozzio A, Passegue E, Ayton PM, Karsunky H,
Cleary ML, Weissman IL. Similar MLL-associated leukemias arising from self-renewing stem cells and
short-lived myeloid progenitors. Genes Dev 2003;17:
3029 ^ 35.
9. Bennett JM, Catovsky D, Daniel MT, et al. Proposed
revised criteria for the classification of acute myeloid
leukemia. A report of the French-American-British
Cooperative Group. Ann Intern Med1985;103:620 ^ 5.
10. Vardiman JW, Harris NL, Brunning RD. The World
Health Organization (WHO) classification of the myeloid neoplasms. Blood 2002;100:2292 ^ 302.
11. McCulloch EA, Howatson AF, Buick RN, Minden
www.aacrjournals.org
MD, Izaguirre CA. Acute myeloblastic leukemia considered as a clonal hemopathy. Blood Cells 1979;5:
261 ^ 82.
12. Fialkow PJ, Singer JW, Raskind WH, et al. Clonal
development, stem-cell differentiation, and clinical
remissions in acute nonlymphocytic leukemia. N Engl
J Med 1987;317:468 ^ 73.
13. Lapidot T, Sirard C, Vormoor J, et al. A cell initiating
human acute myeloid leukaemia after transplantation
into SCID mice. Nature 1994;367:645 ^ 8.
14. Clarkson B, Strife A, Fried J, et al. Studies of cellular
proliferation in human leukemia. IV. Behavior of normal
hemotopoietic cells in 3 adults with acute leukemia
given continuous infusions of 3H-thymidine for 8 or
10 days. Cancer 1970;26:1 ^ 19.
15. Wantzin GL. Nuclear labelling of leukaemic blast
cells with tritiated thymidine triphosphate in 35
patients with acut leukaemia. Br J Haematol 1977;37:
475 ^ 82.
16. Till JE, McCulloch EA, Siminovitch L. A stochastic
model of stem cell proliferation, based on the growth
of spleen colony-forming cells. Proc Natl Acad Sci
U S A 1964;51:29 ^ 36.
17. Korn AP, Henkelman RM, Ottensmeyer FP, Till JE.
Investigations of a stochastic model of haemopoiesis.
Exp Hematol 1973;1:362 ^ 75.
18. Passegue E, Jamieson CH, Ailles LE, Weissman IL.
Normal and leukemic hematopoiesis: are leukemias a
stem cell disorder or a reacquisition of stem cell characteristics? Proc Natl Acad Sci U S A 2003;100 Suppl
1:11842 ^ 9.
19. Cortes J, O’Brien S, Kantarjian H. Discontinuation of
imatinib therapy after achieving a molecular response.
Blood 2004;104:2204 ^ 5.
20. Talpaz M, Estrov Z, Kantarjian H, Ku S, Foteh A,
343
Kurzrock R. Persistence of dormant leukemic progenitors during interferon-induced remission in chronic
myelogenous leukemia. Analysis by polymerase chain
reaction of individual colonies. J Clin Invest 1994;94:
1383 ^ 9.
21. Faderl S,Talpaz M, Kantarjian HM, Estrov Z. Should
polymerase chain reaction analysis to detect minimal
residual disease in patients with chronic myelogenous
leukemia be used in clinical decision making? Blood
1999;93:2755 ^ 9.
22. Domen J,Weissman IL. Self-renewal, differentiation
or death: regulation and manipulation of hematopoietic stem cell fate. Mol Med Today 1999;5:201 ^ 8.
23. Weissman IL. Translating stem and progenitor cell
biology to the clinic : barriers and opportunities.
Science 2000;287:1442 ^ 6.
24. Morrison SJ, Uchida N,Weissman IL.The biology of
hematopoietic stem cells. Annu Rev Cell Dev Biol
1995;11:35 ^ 71.
25. Goodell MA, Brose K, Paradis G, Conner AS,
Mulligan RC. Isolation and functional properties of
murine hematopoietic stem cells that are replicating
in vivo. J Exp Med 1996;183:1797 ^ 806.
26. Kania G, Corbeil D, Fuchs J, et al. Somatic stem cell
marker prominin-1/CD133 is expressed in embryonic
stem cell-derived progenitors. Stem Cells 2005;23:
791 ^ 804.
27. Bornhauser M, Eger L, Oelschlaegel U, et al. Rapid
reconstitution of dendritic cells after allogeneic transplantation of CD133+ selected hematopoietic stem
cells. Leukemia 2005;19:161 ^ 5.
28. Kiel MJ,Yilmaz OH, IwashitaT,Terhorst C, Morrison
SJ. SLAM family receptors distinguish hematopoietic
stem and progenitor cells and reveal endothelial niches
for stem cells. Cell 2005;121:1109 ^ 21.
Clin Cancer Res 2006;12(2) January 15, 2006
Downloaded from clincancerres.aacrjournals.org on June 18, 2017. © 2006 American Association for Cancer
Research.
Review
29. Storms RW,Trujillo AP, SpringerJB, et al. Isolation of
primitive human hematopoietic progenitors on the
basis of aldehyde dehydrogenase activity. Proc Natl
Acad Sci U S A 1999;96:9118 ^ 23.
30. Hess DA, MeyerroseTE,Wirthlin L, et al. Functional
characterization of highly purified human hematopoietic repopulating cells isolated according to aldehyde
dehydrogenase activity. Blood 2004;104:1648 ^ 55.
31. Blair A, Hogge DE, Ailles LE, Lansdorp PM, Sutherland HJ. Lack of expression of Thy-1 (CD90) on acute
myeloid leukemia cells with long-term proliferative
ability in vitro and in vivo. Blood 1997;89:3104 ^ 12.
32. Blair A, Sutherland HJ. Primitive acute myeloid leukemia cells with long-term proliferative ability in vitro
and in vivo lack surface expression of c-kit (CD117).
Exp Hematol 2000;28:660 ^ 71.
33. Jordan CT, Upchurch D, Szilvassy SJ, et al. The
interleukin-3 receptor a chain is a unique marker for
human acute myelogenous leukemia stem cells. Leukemia 2000;14:1777 ^ 84.
34. Brendel C, Mohr B, Schimmelpfennig C, et al.
Detection of cytogenetic aberrations both in CD90
(Thy-1)-positive and (Thy-1)-negative stem cell
(CD34) subfractions of patients with acute and chronic
myeloid leukemias. Leukemia1999;13:1770 ^ 5.
35. Reya T, Duncan AW, Ailles L, et al. A role for Wnt
signalling in self-renewal of haematopoietic stem cells.
Nature 2003;423:409 ^ 14.
36. Terpstra W, Prins A, Ploemacher RE, et al. Longterm leukemia-initiating capacity of a CD34 subpopulation of acute myeloid leukemia. Blood 1996;
87:2187 ^ 94.
37. Grimwade D, Enver T. Acute promyelocytic leukemia: where does it stem from? Leukemia 2004;18:
375 ^ 84.
38. Phillips RL, Ernst RE, Brunk B, et al.The genetic program of hematopoietic stem cells. Science 2000;288:
1635 ^ 40.
39. Ivanova NB, Dimos JT, Schaniel C, Hackney JA,
Moore KA, Lemischka IR. A stem cell molecular signature. Science 2002;298:601 ^ 4.
40. Ramalho-Santos M, Yoon S, Matsuzaki Y, Mulligan
RC, Melton DA. ‘‘Stemness’’: transcriptional profiling
of embryonic and adult stem cells. Science 2002;
298:597 ^ 600.
41. Zipori D. The nature of stem cells: state rather than
entity. Nat Rev Genet 2004;5:873 ^ 8.
42. Smith LJ, Curtis JE, Messner HA, Senn JS,
Furthmayr H, McCulloch EA. Lineage infidelity in
acute leukemia. Blood 1983;61:1138 ^ 45.
43. Zhu J, Emerson SG. Hematopoietic cytokines, transcription factors and lineage commitment. Oncogene
2002;21:3295 ^ 313.
44. Stein MI, Zhu J, Emerson SG. Molecular pathways
regulating the self-renewal of hematopoietic stem
cells. Exp Hematol 2004;32:1129 ^ 36.
45. Begley CG, Green AR. The SCL gene: from case
report to critical hematopoietic regulator. Blood 1999;
93:2760 ^ 70.
46. Lecuyer E, HoangT. SCL: from the origin of hematopoiesis to stem cells and leukemia. Exp Hematol 2004;
32:11 ^ 24.
47. Licht JD. AML1and the AML1-ETO fusion protein in
the pathogenesis of t(8;21) AML. Oncogene 2001;20:
5660 ^ 79.
48. Mulloy JC, Cammenga J, MacKenzie KL, Berguido
FJ, Moore MA, Nimer SD. The AML1-ETO fusion protein promotes the expansion of human hematopoietic
stem cells. Blood 2002;99:15 ^ 23.
49. de Guzman CG,Warren AJ, Zhang Z, et al. Hematopoietic stem cell expansion and distinct myeloid developmental abnormalities in a murine model of the
AML1-ETO translocation. Mol Cell Biol 2002;22:
5506 ^ 17.
50. Zhu J, Giannola DM, Zhang Y, Rivera AJ, Emerson
SG. NF-Ycooperates with USF1/2 to induce the hematopoietic expression of HOXB4. Blood 2003;102:
2420 ^ 7.
51. Sauvageau G, Thorsteinsdottir U, Eaves CJ, et al.
Overexpression of HOXB4 in hematopoietic cells
causes the selective expansion of more primitive
populations in vitro and in vivo. Genes Dev 1995;
9:1753 ^ 65.
52. Golub TR, Slonim DK, Tamayo P, et al. Molecular
classification of cancer: class discovery and class prediction by gene expression monitoring. Science 1999;
286:531 ^ 7.
53. Lawrence HJ, Rozenfeld S, Cruz C, et al. Frequent
co-expression of the HOXA9 and MEIS1 homeobox
genes in human myeloid leukemias. Leukemia 1999;
13:1993 ^ 9.
54. Staal FJ, Clevers HC. WNT signalling and haematopoiesis: a WNT-WNT situation. Nat Rev Immunol
2005;5:21 ^ 30.
55. Duncan AW, Rattis FM, DiMascio LN, et al. Integration of Notch and Wnt signaling in hematopoietic stem
cell maintenance. Nat Immunol 2005;6:314 ^ 22.
56. Artavanis-Tsakonas S, Rand MD, Lake RJ. Notch
signaling: cell fate control and signal integration in
development. Science 1999;284:770 ^ 6.
57. Pui JC, Allman D, Xu L, et al. Notch1 expression in
early lymphopoiesis influences B versus T lineage determination. Immunity 1999;11:299 ^ 308.
58. Ellisen LW, Bird J,West DC, et al. TAN-1, the human
homolog of the Drosophila notch gene, is broken by
chromosomal translocations in T lymphoblastic neoplasms. Cell 1991;66:649 ^ 61.
59. van der Lugt NM, Alkema M, Berns A, DeschampsJ.
The Polycomb-group homolog Bmi-1is a regulator of
murine Hox gene expression. Mech Dev 1996;58:
153 ^ 64.
60. Taipale J, Beachy PA. The Hedgehog and Wnt signalling pathways in cancer. Nature 2001;411:349 ^ 54.
61. Lessard J, Baban S, Sauvageau G. Stage-specific
expression of polycomb group genes in human bone
marrow cells. Blood 1998;91:1216 ^ 24.
62. Park IK, Qian D, Kiel M, et al. Bmi-1 is required for
maintenance of adult self-renewing haematopoietic
stem cells. Nature 2003;423:302 ^ 5.
63. Lessard J, Sauvageau G. Bmi-1determines the proliferative capacity of normal and leukaemic stem cells.
Nature 2003;423:255 ^ 60.
64. Bhardwaj G, Murdoch B,Wu D, et al. Sonic hedgehog induces the proliferation of primitive human
hematopoietic cells via BMP regulation. Nat Immunol
2001;2:172 ^ 80.
65. Guan Y, Gerhard B, Hogge DE. Detection, isolation,
and stimulation of quiescent primitive leukemic progenitor cells from patients with acute myeloid leukemia
(AML). Blood 2003;101:3142 ^ 9.
66. Dean M, FojoT, Bates S.Tumour stem cells and drug
resistance. Nat Rev Cancer 2005;5:275 ^ 84.
67. Konopleva M, Zhao S, HuW, et al.The anti-apoptotic
genes Bcl-X(L) and Bcl-2 are over-expressed and
contribute to chemoresistance of non-proliferating
leukaemic CD34+ cells. Br J Haematol 2002;118:
521 ^ 34.
68. Chaudhary PM, Roninson IB. Expression and activ-
Clin Cancer Res 2006;12(2) January 15, 2006
344
ity of P-glycoprotein, a multidrug efflux pump, in
human hematopoietic stem cells. Cell 1991;66:85 ^ 94.
69. Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATP-dependent transporters.
Nat Rev Cancer 2002;2:48 ^ 58.
70. Scharenberg CW, Harkey MA, Torok-Storb B. The
ABCG2 transporter is an efficient Hoechst 33342
efflux pump and is preferentially expressed by immature human hematopoietic progenitors. Blood 2002;
99:507 ^ 12.
71. Feuring-Buske M, Hogge DE. Hoechst 33342 efflux
identifies a subpopulation of cytogenetically normal
CD34(+)CD38( ) progenitor cells from patients with
acute myeloid leukemia. Blood 2001;97:3882 ^ 9.
72. Guan Y, Hogge DE. Proliferative status of primitive
hematopoietic progenitors from patients with acute
myelogenous leukemia (AML). Leukemia 2000;14:
2135 ^ 41.
73. Ailles LE, Humphries RK, Thomas TE, Hogge DE.
Retroviral marking of acute myelogenous leukemia
progenitors that initiate long-term culture and growth
in immunodeficient mice. Exp Hematol 1999;27:
1609 ^ 20.
74. Terpstra W, Ploemacher RE, Prins A, 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.
75. Guzman ML, Neering SJ, Upchurch D, et al. Nuclear
factor-nB is constitutively activated in primitive human
acute myelogenous leukemia cells. Blood 2001;98:
2301 ^ 7.
76. Holyoake T, Jiang X, Eaves C, Eaves A. Isolation of
a highly quiescent subpopulation of primitive leukemic cells in chronic myeloid leukemia. Blood 1999;
94:2056 ^ 64.
77. Chu S, Xu H, Shah NP, et al. Detection of BCR-ABL
kinase mutations in CD34+ cells from chronic myelogenous leukemia patients in complete cytogenetic
remission on imatinib mesylate treatment. Blood
2005;105:2093 ^ 8.
78. List AF, Kopecky KJ, Willman CL, et al. Benefit of
cyclosporine modulation of drug resistance in patients
with poor-risk acute myeloid leukemia: a Southwest
Oncology Group study. Blood 2001;98:3212 ^ 20.
79. Baer MR, George SL, Dodge RK, et al. Phase 3
study of the multidrug resistance modulator PSC833 in previously untreated patients 60 years of age
and older with acute myeloid leukemia: Cancer and
Leukemia Group B Study 9720. Blood 2002;100:
1224 ^ 32.
80. Jordan CT, Guzman ML. Mechanisms controlling
pathogenesis and survival of leukemic stem cells.
Oncogene 2004;23:7178 ^ 87.
81. Guzman ML, Swiderski CF, Howard DS, et al. Preferential induction of apoptosis for primary human leukemic stem cells. Proc Natl Acad Sci U S A 2002;99:
16220 ^ 5.
82. Guzman ML, Rossi RM, Karnischky L, et al. The
sesquiterpene lactone parthenolide induces apoptosis
of human acute myelogenous leukemia stem and
progenitor cells. Blood 2005;105:4163 ^ 9.
83. Xu Q, Simpson SE, Scialla TJ, Bagg A, Carroll M.
Survival of acute myeloid leukemia cells requires PI3
kinase activation. Blood 2003;102:972 ^ 80.
84. TaniT,Ylanne J,Virtanen I. Expression of megakaryocytic and erythroid properties in human leukemic cells.
Exp Hematol 1996;24:158 ^ 68.
85. Blair A, Pamphilon DH. Leukaemic stem cells. Transfus Med 2003;13:363 ^ 75.
www.aacrjournals.org
Downloaded from clincancerres.aacrjournals.org on June 18, 2017. © 2006 American Association for Cancer
Research.
Eradication of Leukemia Stem Cells as a New Goal of
Therapy in Leukemia
Farhad Ravandi and Zeev Estrov
Clin Cancer Res 2006;12:340-344.
Updated version
Cited articles
Citing articles
E-mail alerts
Reprints and
Subscriptions
Permissions
Access the most recent version of this article at:
http://clincancerres.aacrjournals.org/content/12/2/340
This article cites 85 articles, 36 of which you can access for free at:
http://clincancerres.aacrjournals.org/content/12/2/340.full.html#ref-list-1
This article has been cited by 4 HighWire-hosted articles. Access the articles at:
/content/12/2/340.full.html#related-urls
Sign up to receive free email-alerts related to this article or journal.
To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Department at [email protected].
To request permission to re-use all or part of this article, contact the AACR Publications
Department at [email protected].
Downloaded from clincancerres.aacrjournals.org on June 18, 2017. © 2006 American Association for Cancer
Research.