Anda di halaman 1dari 20

ONCH-1372; No.

of Pages 20
ARTICLE IN PRESS

Critical Reviews in Oncology/Hematology xxx (2010) xxx–xxx

Neoplastic stem cells: Current concepts and clinical perspectives


Axel Schulenburg a,d,∗ , Kira Brämswig b , Harald Herrmann c,d , Heidrun Karlic d ,
Irina Mirkina c,d , Rainer Hubmann c,d , Sylvia Laffer d , Brigitte Marian e , Medhat Shehata c,d ,
Clemens Krepler d,f , Hubert Pehamberger d,f , Thomas Grunt b,d , Ulrich Jäger c,d ,
Christoph C. Zielinski b,d , Peter Valent c,d
aBone Marrow Transplantation Unit, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
b Division of Clinical Oncology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
c Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
d Ludwig Boltzmann Cluster Oncology, Vienna, Austria
e Department of Internal Medicine I, Institute for Cancer Research, Medical University of Vienna, Vienna, Austria
f Department of Dermatology, Medical University of Vienna, Vienna, Austria

Accepted 6 January 2010

Contents

1. Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1.1. Identification of putative CSC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2. Definition of CSC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3. Limitations of in vivo CSC assays . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4. In vitro assays . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5. Antigens commonly expressed on CSC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
6. General problems with the so-called ‘stem cell markers’ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
7. Myeloid neoplasms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
8. Lymphoid neoplasms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
8.1. Acute lymphoblastic leukemia (ALL) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
8.2. Multiple myeloma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
9. Solid tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
9.1. Head and neck squamous cell cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
9.2. Colon CSC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
9.3. Liver CSC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
9.4. Pancreas CSC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
9.5. Breast CSC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
9.6. Brain tumor stem cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
9.7. Prostate CSC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
9.8. Sarcoma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
9.9. Other solid tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
10. Melanoma stem cells (MSC) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
11. CSC plasticity—the Hydra Model of CSC development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
12. Strategies for the successful elimination of CSC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
13. Concluding remarks and future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Reviewers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

∗ Corresponding author at: Department of Medicine I, Stem Cell Transplantation Unit, Medical University of Vienna, Währinger Gürtel 18-20,

A-1090 Wien, Austria. Tel.: +43 1 404006085; fax: +43 1 404005701.


E-mail address: axel.schulenburg@meduniwien.ac.at (A. Schulenburg).

1040-8428/$ – see front matter © 2010 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.critrevonc.2010.01.001

Please cite this article in press as: Schulenburg A, et al. Neoplastic stem cells: Current concepts and clinical perspectives. Crit Rev
Oncol/Hematol (2010), doi:10.1016/j.critrevonc.2010.01.001
ONCH-1372; No. of Pages 20
ARTICLE IN PRESS
2 A. Schulenburg et al. / Critical Reviews in Oncology/Hematology xxx (2010) xxx–xxx

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Biographies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

Abstract
Neoplastic stem cells have initially been characterized in myeloid leukemias where NOD/SCID mouse-repopulating progenitors supposedly
reside within a CD34+/Lin− subset of the malignant clone. These progenitors are considered to be self-renewing cells responsible for the in
vivo long-term growth of neoplastic cells in leukemic patients. Therefore, these cells represent an attractive target of therapy. In some lymphoid
leukemias, NOD/SCID mouse-repopulating cells were also reported to reside within the CD34+/Lin− subfraction of the clone. More recently,
several attempts have been made to transfer the cancer stem cell concept to solid tumors and other non-hematopoietic neoplasms. In several of
these tumors, the cell surface antigens AC133 (CD133) and CD44 are considered to indicate the potential of a cell to initiate permanent tumor
formation in vivo. However, several questions concerning the phenotype, self-renewal capacity, stroma-dependence, and other properties of
cancer- or leukemia-initiating cells remain to be solved. The current article provides a summary of our current knowledge on neoplastic
(cancer) stem cells, with special emphasis on clinical implications and therapeutic options as well as a discussion about conceptual and
technical limitations.
© 2010 Elsevier Ireland Ltd. All rights reserved.
Keywords: Cancer stem cells; Targeted therapy; Drug resistance

1. Introduction which subpopulations with variable capacity of long-term


survival can be detected and separated using monoclonal anti-
The principle concept of cancer stem cells (CSC) has bodies. Such antibodies are directed against organ-specific
gained acceptance in recent years [1–4]. The CSC concept or/and lineage-specific antigens or so-called ‘stem cell mark-
postulates the existence of subfractions of “tumor stem cells” ers’, and can be employed to enrich (separate) stem cells
within each neoplasm. These CSC exhibit the capacity for by fluorescence-activated cell sorting (FACS) from primary
self-renewal and unlimited growth, and in this regard dif- cell samples [1–4,7]. The different subsets of cancer cells
fer from more mature neoplastic cells (progeny) that have (putative stem cells and more mature cells) are then investi-
only a limited capacity to divide and to survive. The con- gated for their capacity to repopulate immunodeficient mice
cept of tumor stem cells may provide explanations for the with the tumor/leukemia (stem cell function). In fact, CSC
failure of certain treatments to induce long-term remission. should be able to reproducibly establish the original cancer or
In fact, in many instances, conventional chemotherapy may leukemia (all or most components of the disease) in a xeno-
act only on more mature cells, whereas immature neoplastic transplant model (also in secondary recipient mice). In most
stem cells exhibit resistance, so that these drugs fail to target studies, non-obese diabetic severe combined immunodefi-
and eliminate CSC [1–4]. Thereby, the CSC concept points to cient (NOD/SCID) mice have been used [1–4,8,9]. However,
the need to develop new treatment strategies through which depending on the type of tumor, other mouse systems may
CSC can be eliminated. A prerequisite for the evaluation of provide an even better engraftment [10,11]. Despite limita-
CSC as “target-cell” in oncology is their identification and tions (non-human microenvironment, slowly growing tumors
knowledge about target expression profiles. Therefore, sub- may not establish during the lifetime of mice), immun-
stantial efforts have recently been made to identify CSC in odeficient mouse models remain a widely used approach
various types of cancer and to identify molecular targets and and are considered the best available standard-model for
expression profiles in these cells. the identification of cancer- and leukemia-initiating cells
A number of different monoclonal antibodies directed in primary tissue samples. Depending on the type of neo-
against various cell surface antigens have been used to iden- plasm, primary neoplastic cells are injected intravenously
tify CSC-enriched cell populations in various neoplasms, and (leukemias, metastatic carcinomas), subcutaneously (skin
to purify these cells for molecular and functional studies [5,6]. tumors, solid tumors), or directly into solid organs [12–17].
These experiments focus on the identification and character- In case of leukemias, NOD/SCID mice are usually irradi-
ization of molecular targets, and effects of natural ligands, ated sublethally in order to provide proper engraftment of
response modifiers, and targeted drugs on these cells. leukemic cells in the bone marrow cavities of mice [18,19].
The current article provides a summary of our current An unresolved question is whether and what cytokines and
knowledge about cancer- and leukemia-initiating cells, with what human microenvironmental cells are required to facil-
special focus on clinical implications and perspectives. itate optimal engraftment and growth/survival of neoplastic
cells in various disease models.
1.1. Identification of putative CSC After injection, tumor/leukemia cell growth is monitored
over several weeks. When a tumor or leukemia has devel-
The identification of CSC is usually based on differen- oped, the mouse is sacrificed and the neoplasm examined
tial expression of cell surface antigens (markers) through for histologic and molecular features [1–4]. Key questions in

Please cite this article in press as: Schulenburg A, et al. Neoplastic stem cells: Current concepts and clinical perspectives. Crit Rev
Oncol/Hematol (2010), doi:10.1016/j.critrevonc.2010.01.001
ONCH-1372; No. of Pages 20
ARTICLE IN PRESS
A. Schulenburg et al. / Critical Reviews in Oncology/Hematology xxx (2010) xxx–xxx 3

each experiment are whether the xenotransplant tumor indeed the lifetime of the mouse. Second, the microenvironment is
resembles the original neoplasm and whether indeed most or usually species-specific and often tumor-specific. In fact, the
all elements (subclones) of the original neoplasm are found in microenvironment of the normal mouse may differ in several
the xenotransplant tumor [2]. To further document long-term aspects from the tumor microenvironment that supports the
engraftment and thus to confirm the stem cell function of CSC growth of neoplastic (stem) cells in the natural (human) host.
in xenotransplant tumors, these tumors can be recovered from Likewise, microenvironment receptors and cytokines in the
mouse tissues and can be transplanted into secondary recipi- mouse may not cross-interact in all cases with the respec-
ent mice, where self-renewing CSC should again form tumor tive receptors expressed on human tumor/leukemia (stem)
lesions and all components of the primary tumor/leukemia as cells. To overcome this problem, NOD/SCID mice have been
well as a new CSC pool [1,2]. It is important in each project to treated with human cytokines or are cotransplanted with
demonstrate that the more mature cells are unable to repop- microenvironmental cells in order to facilitate better growth
ulate leukemias/tumors in the same mice [20] which may be of neoplastic (stem) cells [9]. Another important limitation
difficult to demonstrate in slowly growing/developing neo- is that most mouse models that have been used in the past,
plasms, as the time of development of the human neoplasm including NOD/SCID mice, harbour a residual immune sys-
may exceed the lifetime of the mouse. Moreover, in such neo- tem through which these mice can eliminate subfractions of
plasms, it may be difficult to delineate between engraftment injected cancer/leukemia cells, especially when these cells
of “real” stem cells (CSC) and the persistence of more mature display numerous immunogenic antigens (more mature cells)
progenitors that have only a limited capacity to divide. There- or when cells are antibody-laden (antibody-stained) cells.
fore, in these neoplasms (e.g. chronic leukemias), it may be The same cells may, however, grow and form tumors in
of particular importance to confirm engraftment and growth mice with a more severely impaired immune system [22,23].
of neoplastic cells in secondary recipient mice in order to Finally, most neoplasms may grow in permanently estab-
learn whether these cells exhibit or lack stem cell function. lished subclones with subclone-specific stem cells. In other
Another important question is whether engrafted cells indeed words, the stem cell pool in most neoplasms is composed of
are derived from neoplastic stem cells (CSC) or derive from several (many) different subsets of stem cells with varying
normal stem cells. biological properties and diverse growth characteristics, and
it remains unknown whether all (relevant) subclones can be
grown in one xenotransplant model.
2. Definition of CSC

Cancer/leukemia stem cells (CSC) are undifferentiated


4. In vitro assays
cells and are defined by three key features [1,21]: first, these
cells can differentiate into most or all types of cells that are
As stem cell research using NOD/SCID mice is expen-
produced by the original tumor. Second, CSC have the ability
sive and time-consuming and may have several limitations,
to self-renew. Finally, CSC maintain the stem cell pool and
in vitro long-term growth assays are often used in order to
most (or even all) mature elements of the tumor/leukemia for
screen for stem cell fractions or CSC-regulating compounds.
unlimited time periods by balancing between self-renewal
Such in vitro long-term growth assays have been established
(proliferation without maturation) and proliferation plus dif-
for myeloid and lymphoid neoplasms as well as various solid
ferentiation and maturation by asymmetrical cell division(s)
tumors [24–28]. Interestingly, in most instances, a stroma
[1,4]. The process(es) of cell division, of self-renewal, and
cell layer supports the long-term growth of immature neo-
of differentiation of CSC are considered to be regulated by
plastic cells in these assays, which is in line with the concept
a network of cytokines and by the microenvironment, simi-
that the (tumor) microenvironment essentially contributes to
lar to normal stem cells [1,4]. In many instances, the same
growth and survival of CSC and thus the development and
cytokine that regulates growth of normal (stem) cells in a cer-
biology of these neoplasms (stem cell niche). In myeloid neo-
tain organ will also promote the growth and self-renewal of
plasms, the Dexter type long-term culture system, originally
neoplastic stem cells [1,4].
established for normal pluripotent hematopoietic progenitor
cells [29] has been employed and found to support the long-
3. Limitations of in vivo CSC assays term growth of leukemias [30]. However, interestingly, some
of these leukemias may even have a “growth-disadvantage”
Despite the obvious value of an in vivo model that is compared to normal stem cells (long-term culture initiating
sufficient to demonstrate the tumor-initiating potential of cells) in these cultures [31]. In lymphoid neoplasms, includ-
distinct subpopulations of neoplastic cells, a number of lim- ing CLL and ALL, it is well known that stromal cells can
itations of the xenotransplant assay have to be considered. inhibit apoptosis and support the growth of leukemic cells in
First, neoplasms with a low growth rate (e.g. indolent neo- vitro [32,33]. In various solid tumors and melanomas, iso-
plasms, low-grade malignancies, preneoplasm) are difficult lated cells are found to form three-dimensional spheres that
to analyze in a mouse xenotransplant model as in most presumably are composed of immature neoplastic cells and
instances, the development phase of the neoplasm exceeds supporting/nutritive stromal cells, and thus may resemble an

Please cite this article in press as: Schulenburg A, et al. Neoplastic stem cells: Current concepts and clinical perspectives. Crit Rev
Oncol/Hematol (2010), doi:10.1016/j.critrevonc.2010.01.001
ONCH-1372; No. of Pages 20
ARTICLE IN PRESS
4 A. Schulenburg et al. / Critical Reviews in Oncology/Hematology xxx (2010) xxx–xxx

in vitro model of the so-called stem cell niche [34]. Such ulated by the WNT pathway [13,16,45,51]. The frequently
sphere formation has been described for neural cancer (stem) observed expression of CD44 and CD133 on CSC suggests
cells, especially glioblastomas, colon cancer, breast cancer, a functional role for these receptors in CSC biology. How-
and melanoma cells [24–28]. The culture system is based on ever, it remains unknown whether these receptors are indeed
the ability of cancer cells to form three dimensional spheres necessary for stem cell functions or are just expressed on
in vitro, and the ability of sphere-derived cells to induce long- CSC without a specific function. Whatever the answer to this
term growth of tumors [24–28]. In fact, at least some of question is, these markers are commonly found on immature
the tumor cells within these spheres are considered to have neoplastic cells (CSC) and may thereby help in the identifi-
self-renewal capacity and tumor-initiating (CSC) potential. cation of CSC in various organs and tumors. More recently,
it has been reported that in a colon adenoma strain, LT97,
the CD44+ subpopulation exhibits a faster growth rate and
5. Antigens commonly expressed on CSC expresses higher levels of other stem cell antigens (such
as Musashi-1 and EphB2) compared to the CD44-negative
Neoplastic stem cells are considered to express a simi- fraction of adenoma cells [52]. Similarly, the CD133+ frac-
lar antigen pattern, to display similar functional properties, tion of the HCT116 colon cancer cell line has recently been
and to be regulated by similar receptor ligands when com- described to display higher levels of stem cell antigens and
pared to normal stem cells (derived from the same organ a faster growth rate in vitro and in vivo compared to the
system). Therefore, many stem cell/progenitor cell markers CD133− fraction [53].
are also markers of neoplastic stem cells. These antigens Another basic requirement for CSC is to protect them-
include cytokine receptors, homing receptors, and various selves against various external toxic stimuli and drugs, which
drug transporters (Table 1). makes them often drug-resistant. One example for such a
To identify potential surface markers of CSC, it is drug transporter protein is ABCG2 which is able to pump
helpful to look for interactions of these cells with the sur- out not only cell-specific substances but also exogenous tox-
rounding microenvironment. Based on the behaviour of ins and cytotoxic drugs, and thus is responsible (in part) for
normal stem cells, CSC should interact with the supporting resistance of CSC against various drugs. Other drug trans-
microenvironment via several biologically relevant surface porters that have been discussed as indicating the long-term
receptors mediating specific functions. One specific stem cell repopulation-potential of malignant cells include MDR-1
function related to the microenvironment is “stem cell hom- and ABCB5 [54,55]. Various specific dyes like the Hoechst
ing” [35–38]. Major homing receptors discussed as being 33342 dye, are also transported from the cell into the extra-
expressed on CSC are integrins, selectin-ligands, chemokine cellular space via specific transporters, which allows the
receptors, other cytoadhesion molecules (CAMs), and lig- definition of the so-called “side population” which seems
ands of matrix molecules such as L1 or the hyaluronic closely related to the stem cell fraction in many tumors
acid receptor CD44 [39,40]. L1 is also involved in the [56]. A similar distribution and association with stem cells
epithelial–mesenchymal transition (EMT) and is found on has been described for the enzyme aldehyde dehydrogenase
the edge of invasive colon cancer and its metastases [41,42]. (aldolase) [57–61]. This enzyme has been described to be
Together with L1, CD133 appears to be necessary for tumor involved in the metabolization of several cytostatic drugs
growth of gliomas [43]. CD133 and CD44 show overlapping including cyclophosphamide and thereby may be associated
expression in various tumors and CSC [26,44,45]. CD133, a with chemoresistance [60,62,63].
glycoprotein also known as Prominin 1 (PROM1), is a mem- Lastly, CSC are considered to respond to various exter-
ber of the pentaspan family of transmembrane glycoproteins nal physiologic (sometimes paracrine or autocrine) stimuli
(5-transmembrane, 5-TM) which specifically bind to cellular including various cytokines. In line with this assumption,
protrusions [46–48]. CD133 is expressed in hematopoietic CSC express certain cytokine and chemokine receptors
stem cells, endothelial progenitors, as well as on glioblas- on their cell surface. For example the IL-3 receptor
toma, colon CSC, and other solid tumor CSC [46–48]. (CD123/CD131), SCF receptor (KIT) and the G-CSF-
The CD44 protein is a cell-surface glycoprotein involved receptor are usually expressed on leukemic CSC in AML
in cell–cell interactions, adhesion, migration, and homing [64,65]. It has also been described that EGF receptor fam-
[49]. It is a receptor for hyaluronic acid and can also inter- ily members including HER2 are expressed on epithelial
act with other ligands, such as osteopontin, collagen species, CSC including mammary CSC [66,67]. There is also evi-
and matrix metalloproteinases (MMPs) [49]. A specialized dence that IGF- and FGF-receptors play an important role
sialofucosylated glycoform of CD44, called HCELL, is usu- in solid tumors and may be expressed on solid tumor CSC
ally expressed on human hematopoietic stem cells, and is a [68,69]. More recently, it has been described that pancreatic
highly potent E-selectin and L-selectin ligand [50]. HCELL CSC express CXCR4 [12]. A clinically important question
functions as a “bone marrow homing receptor” directing is whether solid tumor CSC or leukemic CSC express recep-
migration of human hematopoietic stem cells and mesenchy- tors for erythropoietin (EPO), G-CSF, or GM-CSF, as these
mal stem cells to the bone marrow [50]. HCELL is expressed cytokines are often applied in cancer/leukemia patients [70].
on breast-, prostate-, pancreatic-, and colon CSC, and is reg- At least for AML and some tumor cell types it has been

Please cite this article in press as: Schulenburg A, et al. Neoplastic stem cells: Current concepts and clinical perspectives. Crit Rev
Oncol/Hematol (2010), doi:10.1016/j.critrevonc.2010.01.001
ONCH-1372; No. of Pages 20
ARTICLE IN PRESS
A. Schulenburg et al. / Critical Reviews in Oncology/Hematology xxx (2010) xxx–xxx 5

Table 1
Cell surface molecules detectable on stem and progenitor cells.
Marker/antigen Background and function of marker/antigen
CD133 CD133 antigen is a transmembrane molecule expressed on hematopoietic stem and progenitor cells, on circulating
endothelial progenitor cells, neural stem cells, renal and prostate stem cells [215].
CD34 CD34 is an adhesion molecule expressed on human hematopoietic stem and progenitor cells, endothelial progenitor
cells and vascular endothelial cells [142].
CD44 CD44 is a cell adhesion receptor, and its ligands are hyaluronate and the cytokine osteopontin. CD44 is expressed in
lymphoid, mieloid and erythroid cells, mesenchymal stem cells and may be useful predictor of lymph node metastasis
[1].
CD29 CD29 is a beta1 integrin involved in cell adhesion embryogenesis, tissue repair, immune response and metatastatic
diffusion of tumor cells reacting with thrombocytes, monocytes and a T and B lymphocytes and is also expressed on
mesenchymal stem cells [216].
CD24 CD24 antigen is a glycosylphosphatidylinositol-linked membrane sialoglycoprotein. CD24 is present on B cells, from
the stage pre-B to the mature B cell stage, but not on plasma cells. It is expressed on mature granulocytes and on a
variety of epithelial cell types [1].
CD166 CD166 molecule, a mesenchymal stem cell marker that displayed heterogeneous expression patterns in CRC epithelial
cells (15, 16) and whose increased expression levels were previously associated with poor clinical outcome in CRC
patients [45].
CD326 EpCAM (CD326) is a pan-epithelial differentiation antigen expressed on the basolateral surface of various carcinomas
to varying degrees. As a homotypic cell adhesion molecule, it is intimately integrated within the Cadherin–Catenin and
WNT pathways. It has recently been shown to modulate the expression of proto-oncogenes such as c-myc [217].
CD90 Thy-1 (CD90) is expressed on many cell types, including T cells, thymocytes, neurons, endothelial cells, and fibroblasts.
Activation of Thy-1 can promote T cell activation, and this role of Thy-1 is reviewed elsewhere. Thy-1 also affects
numerous nonimmunologic biological processes, including cellular adhesion, neurite outgrowth, tumor growth,
migration, and cell death [218].
CD123 The specific [alpha] subunit of the interleukin-3 receptor (IL-3R-alpha, CD123) is expressed on hematopoietic cells,
including monocytes, neutrophils, basophils, and megakaryocytes but not on peripheral T cells, natural killer cells,
platelets, and red blood cells [219].
CD9 CD9 belongs to a tetraspanin superfamily and is expressed in a variety of blood cells including pre-B lymphocytes but
not in HSCs. It is also expressed in many types of solid tumors, and is involved in a various kinds of cell processes, such
as cell adhesion, motility, and signalling events through an association with integrin family proteins [220].
CD20 CD20 (human B-lymphocyte-restricted differentiation antigen, Bp35), is a hydrophobic transmembrane protein with a
molecular weight of approximately 35 kDa located on pre-B and mature B lymphocytes. The antigen is expressed on
most B-cell non-Hodgkin’s lymphomas but is not found on stem cells, pro-B cells, normal plasma cells or other normal
tissues. Plasma blasts and stimulated plasma cells may express CD20. CD20 regulates an early step(s) in the activation
process for cell cycle initiation and differentiation, and possibly functions as a calcium ion channel. CD20 is not shed
from the cell surface and does not internalize upon antibody binding. Free CD20 antigen is not found in the circulation;
thus a drug that reacts with CD20, such as an antibody, would not be neutralized before binding to its target cell [221].
ABCB1 (MDR1) and ABCG2 ABCB1 and ABCG2 are recognized as belonging to a family of at least 48 human ABC transporters involved in a
variety of essential cellular transport processes. They are the products of MDR genes and confer multidrug resistance by
pumping out chemotherapy. They also pump out Hoechst dye and rhodamine. Reversal of MDR in vitro was easily
attained with a variety of inhibitors like verapamil [222].
ABCB5 ABCB5 [subfamily B (MDR/TAP)] is a novel human ABC transporter encoded on chromosome 7p15.3 [223]. ABCB5,
like ABCB1, acts as an energy-dependent drug efflux transporter for the fluorescent probe rhodamine-123 [224].
CLL-1 Human CLL-1 (also known as MICL or CLEC12A), is a type II transmembrane glycoprotein and member of the large
family of C-type lectin-like receptors involved in immune regulation. The intracellular domain of CLL-1 contains both
an immunotyrosine-based inhibition motif as well as a YXXM motif, suggesting a role for CLL-1 as a signalling
receptor [86,225].
Erythropoietin receptor The erythropoietin receptor (EpoR) consists of two peptide chains and is a member of the cytokine receptor family. The
interaction of erythropoietin with its cell surface receptor induces a conformational change of receptor homodimers
leading to the activation of intracellular signal transduction that mediates the ability of erythropoietin to support the
proliferation, terminal differentiation and survival [226].

described that CSC indeed express receptors for SCF, G-CSF, Rather, most of these antigens are broadly expressed on var-
and sometimes also for GM-CSF [65]. ious mesenchymal cells. Likewise, CD44 is expressed not
only on hematopoietic and non-hematopoietic stem cells
but also on most mature cells, including monocytes, lym-
6. General problems with the so-called ‘stem cell phocytes, granulocytes, epithelial cells, and melanocytes
markers’ [50,71]. Similarly, in most leukemias and solid neoplasms,
CD44 is expressed on mature cells. Prominin (CD133) is
One general problem is that the so-called stem cell mark- expressed on myeloid progenitor cells and on various (imma-
ers are by far not specific for stem cells or progenitor cells. ture and mature) mesenchymal cells, including endothelial

Please cite this article in press as: Schulenburg A, et al. Neoplastic stem cells: Current concepts and clinical perspectives. Crit Rev
Oncol/Hematol (2010), doi:10.1016/j.critrevonc.2010.01.001
ONCH-1372; No. of Pages 20
ARTICLE IN PRESS
6 A. Schulenburg et al. / Critical Reviews in Oncology/Hematology xxx (2010) xxx–xxx

cells [46–48]. Even the CD34 antigen (hematopoietic pre- more severely immunodeficient mice (NOG or NSG mice)
cursor cell antigen-1, HPCA1) is not only expressed on may be required for optimal engraftment of CSC [10,11]. It
hematopoietic stem cells but also on more mature progen- can therefore be expected that stem cell research will employ
itors and also on endothelial cells [72]. All in all, no stem NOG or NSG mice in various CSC models in the future.
cell specific antigen has been identified so far. Therefore, it Recently, Kim et al. found that a polyclonal anti-human
is essential to apply combinations of markers and antibodies CD24 rabbit antibody initiates cross-linking of CD24 on
and to define stem cell-enriched fractions on the basis of typ- tumor cells, and that this cross-link induces apoptosis in
ical antigen combinations. Usually, one or two markers are breast cancer cells in a cell culture system using MCF-7
employed by investigators to gate for or to exclude a germ cells [74]. This may have implications for the characteriza-
layer or an organ system (e.g. CD45 as pan-hematopoietic cell tion of breast cancer stem cells, since these cells supposedly
marker). In certain instances a cocktail of antibodies is used reside within the CD44+CD24low (CD24-negative) fraction
to exclude more mature cells in a certain organ system, e.g. of tumor cells. If CD24+ tumor cells would just be elimi-
more mature hematopoietic cells by lineage-specific mark- nated (faster than CD24low cells) by the antibody-induced
ers (“Lin-cocktail” defining Lin-negative progenitor cells) cross-linking of CD24, the CD24-negativity could no longer
[19,73]. Here, one problem may be that in certain leukemias, be regarded as a stem cell-related feature. Whether indeed
CSC may aberrantly express lineage-related marker antigens. CD24-induced stem cell depletion occurs after antibody-
In these leukemias, application of the “Lin-cocktail” would binding remains to be shown.
lead to a loss of CSC (sub)fractions. Another major limitation may be that CSC receptors and
Finally, markers that are typically expressed on immature their ligands and homing receptors in the tissue environment
cells of a given neoplasm or organ, are applied, e.g. CD34 for are sometimes species-specific. Therefore, CSC are often
lymphohematopoietic cells and most myeloid leukemias. In injected directly into a certain target organ (e.g. bone marrow,
these defined progenitor fractions, further subpopulations in pancreas, brain, others) in these mice [12–17,44,75,76]. Nev-
which CSC reside, are defined. This is performed by antibod- ertheless, it is difficult to know whether some of the CSC will
ies that positively or negatively identify these subfractions. not (cannot) grow in a xenotransplant model simply because
Based on antibody-binding patterns and identification of sub- essential mouse-derived ligands are not cross-reacting with
fractions enriched in CSC, these cells can be isolated by human receptors expressed on CSC. A possible solution to
multi-color flow cytometry and cell sorting. The read out that this problem may be to co-transplant human stroma cells,
is then used to confirm the presence of CSC is either an in to humanize mice (organ-specific microenvironment), or to
vitro long-term culture system or – preferably – the immun- inject mice with all important (human) ligands in order to
odeficient mouse model. In most instances, the NOD/SCID guarantee proper engraftment of CSC.
mouse has been employed in these assays. However, there are Probably the most important problem with CSC markers
a number of caveats that have to be considered when using and CSC-reactive antibodies is stem cell plasticity. In fact,
sorted cells in these mouse models and bioassays. The most it has been described that leukemic and solid tumor CSC
important caveat may be that the antibody itself may stimu- fractions are usually composed of several different subclones
late or may interfere with in vitro growth, engraftment, or/and defined by varying profiles of molecular markers, point muta-
long-term repopulation of progenitor cells in NOD/SCID tions in critical target genes (often causing drug resistance in
mice [22]. Therefore, it has to be excluded by appropriate subclones), and expression of cell surface receptors including
control experiments, that the antibodies used to positively stem cell markers. This means, that CSC in a given tumor may
or negatively define CSC would per se induce or inhibit display varying combinations of cell surface antigens which
engraftment of CSC in NOD/SCID mice, or would interfere makes it difficult to define all CSC subfractions (subclones)
with in vitro CSC growth in the bioassay applied or with by monoclonal antibodies. In some instances, the phenotypic
in vivo engraftment [22]. Recently, Taussig et al. revealed a heterogeneity may also be associated with functional hetero-
procedure-related problem in the description of AML CSC geneity. However, so far very little is known about cell surface
defined by expression of CD34 and lack of CD38 [22]. In their antigens defining subfractions of CSC in various solid tumors
paper, they were able to show that the original description and leukemias. Likewise, in AML, leukemic CD34+/Lin−
of leukemic stem cells as CD38-negative cells in the mouse (stem) cells are often composed of a CD133+ and a CD133−
model may be due to the clearance of leukemic CSC together subfraction [77].
with the CD38 antibody by the residual immune system of In the following paragraphs, we review human neoplasms
NOD/SCID mice. Clearance was not observed when mice where CSC have recently been identified.
were further immunosuppressed by drugs or when the anti-
body was degraded into fab fragments which allowed CD38+
leukemic cells to repopulate these mice [22]. Moreover, 7. Myeloid neoplasms
CD34+/CD38+ grew well in more severely immunodeficient
IL2rgamma(null) mice (NSG mice) [22]. All in all, it appears It is generally assumed that most if not all myeloid
that NOD/SCID mice may not be the most suitable strain to neoplasms derive from a clonal immature hematopoietic
study CSC growth in solid tumors and leukemias. Rather, progenitor (stem) cell. Therefore, myeloid neoplasms are

Please cite this article in press as: Schulenburg A, et al. Neoplastic stem cells: Current concepts and clinical perspectives. Crit Rev
Oncol/Hematol (2010), doi:10.1016/j.critrevonc.2010.01.001
ONCH-1372; No. of Pages 20
ARTICLE IN PRESS
A. Schulenburg et al. / Critical Reviews in Oncology/Hematology xxx (2010) xxx–xxx 7

optimal models to study the CSC hypothesis. Normal may be CD34-negative cells [87]. In many of these leukemias
hematopoietic stem cells are considered to reside within it even remains to be demonstrated that AML stem cells reside
the Lin-negative and CD38-negative portion of CD34- within the (small) CD34+ subpopulation of clonal leukemic
positive progenitor cells. Leukemic stem cells have first cells. It is also important to note that AML types in which the
been described in AML, and later in CML [19,73,78]. In stem cell origin of the leukemia can be demonstrated using a
other myeloid neoplasms, neoplastic stem cells are less well NOD/SCID mouse xenotransplant model are those variants
defined. The phenotype of leukemic stem cells is considered where blast cells exhibit a high proliferative potential and
to be similar to that of normal stem cells (Lin−, CD34+, resistance against chemotherapy [88].
CD38− and in part CD38+ ) [8,19,79] (Table 2). Several In CML, most clonal cells are CD34-negative cells. In
attempts have been made to identify markers that distin- these patients, a complex multi-step enrichment technique
guish between normal and neoplastic meyloid stem cells. for the isolation of putative CD34+/Lin− stem cells has been
This would enable therapeutic approaches that might be proposed [73]. Indeed, when these cells were injected into
able to spare normal stem cells [80]. Indeed, several surface NOD/SCID or NOD/SCID-beta2microglobulin−/− mice,
molecules may be expressed abundantly on CD34+/Lin− they were found to repopulate these mice with a CML-like
AML stem cells, whereas normal hematopoietic stem cells disease. By contrast, more mature CD34+ CML cells only
lack or express only low levels of these antigens. Exam- produced an early transient leukemic repopulation that was
ples for antigens preferentially expressed on leukemic CSC no longer detectable after 6 weeks [89]. Similar to the AML
in AML are the alpha chain of the IL-3 receptor (CD123), stem cell, the CML stem cell was found to express the IL-
the Mylotarg-receptor Siglec-3 (CD33), CD96, CXCR4, and 3 receptor [90]. Other target antigens like CD33, CD44, or
CLL-1 [64,81–86]. CD33 is of special interest as CD33- CD117 were also found to be expressed on CD34+/CD38−
targeting drugs are available and are used to treat patients stem cells in CML patients [64]. CML stem cells also
with (refractory) AML. Fig. 1 shows the effects of Mylotarg express ABCB1 and ABCG2, whereas the levels of OCT-
on survival of CD34+/CD38+ and CD34+/CD38− AML 1 expressed on CML stem cells are rather low [55,91]. Both
cells. Neoplastic stem cells have been defined in several ABCG molecules are known to mediate the efflux of ima-
but not all variants of AML [8,19]. Likewise, in a group tinib and other drugs and thus confer resistance. OCT-1,
of AML patients, leukemic blast cells and immature pro- on the other hand, mediates the uptake of imatinib, and
genitors are CD34-negative. In these patients, it is very low expression of these transporter molecules is associ-
difficult to define stem cell compartments. Other examples ated with low drug-uptake and thus low intracellular levels
are promyelocytic leukemia, NPM-mutated AML variants, of imatinib. All these mechanisms may contribute to the
and monoblastic leukemias where most of the neoplastic cells intrinsic resistance of CML CSC against imatinib [55,92].

Table 2
Published cell surface phenotype of neoplastic stem cells in various malignancies.
Disease Surface marker Refs.
AML CD34+, CD38−, CD44, CD123+ Bonnet and Dick [19]
ALL Ph+ CD34+, CD38− Cobaleda et al. [105]
TEL-AML1–positive CD34+/CD38−/low/CD19+ Hong et al. [106]
c-ALL CD34+/CD19+ Kong et al. [107]
Childhood B-ALL CD133(+)/CD19(−) and CD38(−) Cox et al. [108]
B lineage ALL CD34+/CD9+ Nishida et al. [109]
CML CD34+, CD38−, CD123+ Holyoake et al. [73]
Breast CD44+, CD24−/low, ESA+ Al-Hajj et al. [13]
CD44+ Li et al. [16]
Pancreas
CD133+ Hermann et al. [12]
CD133+ Ma et al. [131]
Liver
CD90+ Yang et al. [17]
CD133 Ricci-Vitian et al. [26], O’Brien et al. [44]
Colon
CD44 Dalerba et al. [45]
CD133+/alpha 2 beta 1 integrin/CD44+ Collins et al. [142]
Prostate
CD44+/CD24− Hurt et al. [51]
Sarcoma CD133+ Suva et al. [151]
CD20+ Fang et al. [24]
Melanoma
CD133+ Monzani et al. [162]
CD133+ Singh et al. [15]
CNS
Hemm et al. [27]

Please cite this article in press as: Schulenburg A, et al. Neoplastic stem cells: Current concepts and clinical perspectives. Crit Rev
Oncol/Hematol (2010), doi:10.1016/j.critrevonc.2010.01.001
ONCH-1372; No. of Pages 20
ARTICLE IN PRESS
8 A. Schulenburg et al. / Critical Reviews in Oncology/Hematology xxx (2010) xxx–xxx

Fig. 1. Apoptosis in AML stem cells induced by Mylotarg (GO). Blast cells obtained from a patient with AML (FAB M0) were incubated with control medium
(upper graphs) or with gemtuzumab/ozogamicin (GO = Mylotarg), 1 ␮g/ml, at 37 ◦ C for 48 h (lower graphs). Then, apoptosis was analyzed by combined flow
cytometry using antibodies against CD34 and CD38, and AnnexinV-FITC. The percentage of apoptotic cells was analyzed in CD34+/CD38+ cells (right panel)
and in the CD34+/CD38− fraction of the clone (left panel) after gating for vital (7-AAD negative) cells.

More recently, imatinib-resistance of CML stem cells has nalling is initiated by specific oncoproteins, like BCR-ABL in
been employed to screen for stem cell-related markers in CML, or PML-RAR-alpha in acute promyelocytic leukemia
BCR/ABL transformed (imatinib-exposed) murine cells [93]. [96–100].
In these studies, several antigens potentially involved in In other myeloid neoplasms, very little is known about
leukemic stem cell growth and survival have been identi- the biology and phenotype of neoplastic stem cells. Florian
fied. One of these CML stem cell-related genes is the Alox5 et al. examined the phenotype of CD34+/CD38− cells in
gene that encodes the 5-lipoxygenase (5-LO) [87]. In the various myeloid neoplasms including myelodysplastic syn-
absence of Alox5/5-LO, BCR/ABL failed to induce a CML- dromes, CML, and systemic mastocytosis [64]. In all these
like disease in mice, whereas Alox5-deficiency showed no myeloid neoplasms, the phenotype, determined by antibody-
effects on growth of normal stem cells [93]. These data staining, appeared to be very similar, and included major
suggest that growth and survival of leukemic stem cells surface targets such as CD123 and CD33 [64]. In classi-
in CML are regulated by specific gene products, and the cal JAK2 V617F-mutated myeloproliferative disorders, the
same may hold true for AML. Also, recent data suggest JAK2 mutant is usually detectable in the CD34+/CD38−
that neoplastic stem cells in various myeloid neoplasms fraction of clonal cells [101,102]. However, JAK2 V617F
may use similar if not identical signalling pathways for may not be detectable in all neoplastic stem cell subclones
growth and survival in vivo. These pathways include the PI3 in these patients [103], and leukemic progression (sec-
kinase-mTOR pathway, WNT-␤-catenin pathway, and Notch ondary AML) is often accompanied by a loss of the JAK2
signalling-pathway [94]. However, all these pathways may mutant, which may be explained by expansion of a more
be shared by normal and leukemic stem cells, and therefore immature JAK2 V617F-negative subclone during disease
may not be optimal targets, at least when treatment should progression [103]. Otherwise, very little is known about the
spare normal (stem) cells. Also, Hedgehog signalling seems biology, target expression profiles, and phenotypes of neo-
to be important for the development of myeloid leukemias plastic stem cells in JAK2+ MPN. Recently, a defective stem
and may be a promising target pathway [95]. Other path- cell niche has been discussed as an important factor con-
ways found in leukemic stem cells are the JAK2-STAT5 tributing to the pathogenesis of JAK2-mutated neoplasms
pathway and NF-kappa-B pathway. In most instances, sig- [104].

Please cite this article in press as: Schulenburg A, et al. Neoplastic stem cells: Current concepts and clinical perspectives. Crit Rev
Oncol/Hematol (2010), doi:10.1016/j.critrevonc.2010.01.001
ONCH-1372; No. of Pages 20
ARTICLE IN PRESS
A. Schulenburg et al. / Critical Reviews in Oncology/Hematology xxx (2010) xxx–xxx 9

8. Lymphoid neoplasms arise from clonogenic CD138-negative B cells [112]. In par-


ticular, CD138− cells were found to act clonogenic in vitro
8.1. Acute lymphoblastic leukemia (ALL) and to produce myelomas in NOD/SCID mice [119]. In
2008 it was found that a combination of dexamethasone,
Little is known about subpopulations of CD34+ ALL lenalidomide, bortezomib, and 4-hydroxycyclophosphamide
cells that display stem cell function. In 2000, Cobaleda et inhibits the growth of CD138+ multiple myeloma cells but
al. showed that in Ph+ (BCR/ABL+) ALL, the NOD/SCID not the growth of CD138-negative myeloma precursor cells
mouse-repopulating ALL stem cell resides within the in vitro. Several lines of evidence suggest that the phenotype
CD34+/CD38− fraction of the clone, similar to AML of myeloma stem cells is similar to that of normal memory
stem cells [105]. When ≥100 CD34-positive/CD38-negative B cells (CD138−/CD20+/CD27+) [119–121].
cells were injected intravenously, leukemias developed after
4–6 weeks in these mice [105]. Hong et al. recently
described that the CD34+/CD38−/low/CD19+ cells in TEL- 9. Solid tumors
AML1–positive c-ALL (0.002% of total mononuclear cells)
propagate leukemias in NOD/SCID mice [106]. In contrast, 9.1. Head and neck squamous cell cancer
Kong et al. found that CD38 is not useful for the identi-
fication of a c-ALL stem cell [107]. They observed that Despite combination therapy head and neck squamous cell
CD34+/CD38−/CD19+ as well as CD34+/CD38+/CD19+ cancer (HNSCC) remains one of the most difficult challenges
cells are able to establish leukemias after 4–15 weeks in oncology. HNSCC resistance to various drugs has limited
when at least 5 × 103 cells were intravenously injected into the usefulness of chemotherapy in this disease. Recently,
NOD/SCID/IL2r null mice [107]. Recently, Cox et al. found CD44 has been identified as a potential marker of CSC in
evidence that the pediatric B-ALL stem cell resides within HNSCC [122]. When 5 × 103 CD44-positive poorly to well
a CD133+/CD19− subpopulation [108]. After injection of differentiated primary HNSCC cells were injected subcuta-
743–50,000 CD133+/CD19− cells into NOD/SCID mice, neously into NOD/SCID mice or Rag2␥DKO mice, these
engraftment was observed after 8–10 weeks [108]. Another cells gave rise to tumors within 10–16 weeks [122]. The
potential B-ALL stem cell marker may be CD9. Nishida et stem cell-related gene BMI-1 was demonstrated to be overex-
al. injected at least 1 × 104 CD9+ (cultured) ALL cells intra- pressed in the CD44+ subpopulation of HNSCC tumor cells
venously into a total of 20 NOG mice, and all mice died compared to CD44− cells [122]. In vitro experiments per-
within 45 days due to leukemia [109]. Despite the com- formed with various types of cancer cells suggest that CD133
mon notion that the ALL stem cell should be CD34+ cells, may also be a marker for long-term proliferating cells, but
this may not hold true for all B-lineage ALL variants and it remains unknown whether CD133 is indeed a stem cell
also not for T ALL [110,111]. Recent data suggest that the marker for HNSCC [123].
Notch pathway may be involved in CSC function in patients
with T-ALL [112]. Notch is already known to play a role in 9.2. Colon CSC
normal T-cell development and T-ALL [113–116]. Whereas
in ALL, at least first attempts have been made to identify Since the gastrointestinal tract is an organ-system with
a leukemic stem cells, there is very limited if any infor- a high turnover of cells where proliferation and cell self-
mation about NOD/SCID mouse-repopulating cells derived renewal take place, it was of great interest to learn about
from other lymphoproliferative neoplasms, such as chronic the location, biology, and phenotype of colon CSC. Sev-
lymphocytic leukemia (CLL) or other Non-Hodgkin’s Lym- eral lines of evidence suggest that immature colon cells (and
phomas (NHL) [117]. In one study, Nowakowski et al. found a presumably also CSC) are located in colon crypt bottoms.
small CD5+/CD19+/ABCG+ population of CLL cells [111]. With regard to the phenotype, first reports pointed to CD133
From gene expression profiling data and an increase of this as a potential CSC marker antigen colon cancer [26,44]
subpopulation of CLL cells after therapy, the authors con- (Table 2). When CD133-positive cells were injected into
cluded that these cells may have stem cell like properties the kidney capsule or subcutaneously into NOD/SCID mice,
[118]. More recent data suggest that the CLL cell with stem colon tumors developed after several weeks [26,44]. Later,
cell-like properties may also reside within a small CD34+ Dalerba et al. found that CD44 is another potential marker for
subset of leukemic cells that co-express CD19 and CD5 CSC in colon cancer patients [45]. When CD44+/EpCAM+
(unpublished observation). cells were injected subcutaneously into NOD/SCID mice
tumors developed after 20 weeks in these mice [45]. Fur-
8.2. Multiple myeloma thermore, they found that the CSC population is further
characterized by coexpression of CD166 [45]. Recently, it
Matsui et al. were the first to report on the existence of was reported that CD133+ cells derived from colon spheres
myeloma-initiating stem cell fractions in multiple myeloma are clonogenic cells and can form adenocarcinomas in a
patients [119]. They showed that that CD138+ fraction of mouse xenotransplant model [28]. These CD133+ sphere-
myeloma cells cannot undergo long-term proliferation but derived cells in part co-expressed CD24, CD29, CD44, and

Please cite this article in press as: Schulenburg A, et al. Neoplastic stem cells: Current concepts and clinical perspectives. Crit Rev
Oncol/Hematol (2010), doi:10.1016/j.critrevonc.2010.01.001
ONCH-1372; No. of Pages 20
ARTICLE IN PRESS
10 A. Schulenburg et al. / Critical Reviews in Oncology/Hematology xxx (2010) xxx–xxx

CD166. Interestingly, of all markers tested, only CD24 was cells by maturation arrest. Functional analysis confirmed
found to enrich for cells with even higher clonogenic activ- that distinct cell subsets present in HCC exhibit stem
ity. All in all, several surface markers, including CD44 and cell-like properties including long-term survival (immor-
CD133 have been discussed as potential markers of colon tality), transplantability, and resistance to (chemo)therapy.
CSC (Table 2). With regard to CD44, this may also hold true More recently, several markers that may indicate stem cell
for colon adenomas. In fact, it has been shown that the colon function in HCC cells have been identified. These mark-
adenoma cell strain LT97 consists of a CD44+ and a CD44− ers include CD133 and CD90 [131,132]. When 5 × 103
portion, and that both subfractions differ in their growth CD45− /CD90+ cells, isolated from tumor specimens or blood
kinetics and expression of stem cell markers [52]. In particu- samples of HCC patients, were injected into the liver of
lar, CD44-positive LT97 cells attach and grow for unlimited SCID/Beige mice, tumors developed within 12–16 weeks in
time periods, whereas CD44− cells are slowly growing cells these mice.
(Fig. 2) [52]. Moreover, in contrast to CD44− LT97 cells,
CD44+ LT97 cells display nuclear beta-catenin and express 9.4. Pancreas CSC
beta-catenin target genes, such as ephrin B receptor (ephB2)
and the musashi1 antigen (msi1) [52]. These stem cell anti- In pancreatic cancer, CSC have also been described.
gens are also detectable in colon CSC [124]. All in all, CD44 These pancreatic cancer stem cells express the cell sur-
may not only be a CSC marker for established colon ade- face markers CD44, CD24, and EpCAM, and represent
nocarcinomas, but also for CSC in preneoplastic lesions, i.e. 0.5–1.0% of all pancreatic cancer cells [16]. When ≥100
colon adenomas which may have implications for the biol- CD44+/CD24+/EpCAM+ cells were injected into the pan-
ogy of tumor development. In fact, colon carcinoma CSC creas of NOD/SCID mice, tumors developed within 4 weeks
may develop from adenoma CSC by subclone selection after in these mice [16]. Another report suggested that CD133
accumulation of further hits [125,126]. is expressed on pancreatic CSC [12]. When 500 CD133-
positive pancreas tumor cells were injected into the pancreas
9.3. Liver CSC of NMRI-nu/nu mice, tumors developed within 3 weeks [12].
Pancreatic CSC display high levels of genes involved in self-
A number of previous studies suggest that hepatic can- renewal, such as the Sonic Hedgehog (SHH) antigen and
cer cells derive from immature progenitor cells in the BMI-1 [133,134]. In addition, CD133+/CXCR4+ cells have
liver [127]. There are four candidates for liver CSC: bone been described to be responsible for metastasis formation in
marrow derived cells, oval cells, hepatocytes, and hep- pancreatic cancer in a model employing the L3.6pl cell line
atopancreatic stem cells [128]. Recent data suggest that [12].
the so-called small oval cells in hepatocellular carcinomas
(HCC) are involved in the carcinogenic process and dis- 9.5. Breast CSC
play stem cell markers [129,130]. Since these small oval
cells are also found in the normal liver, the hypothesis In 2003, Al-Hajj et al. identified a NOD/SCID mouse-
was raised that HCC arise from normal liver progenitor repopulating breast cancer cell subpopulation. These cells
express CD44, the breast/ovarian cancer-specific antigen
B38.1, and epithelial-specific antigen ESA (= EpCAM) [13].
In contrast to non-repopulating breast cancer cells, breast
CSC coexpress CD44 but display only low amounts of or
lack CD24. When ≥100 CD44+/B38.1+/CD24− cells were
injected into the mammary fat pad of NOD/SCID mice, pal-
pable tumors developed within 12 weeks, whereas none of the
CD44−/B38.1− cells developed tumors in these mice. Other
studies have suggested that high aldehyde dehydrogenase lev-
els may be indicative for enhanced malignant and metastatic
potential and thus may help in the identification of breast CSC
[60]. Most consecutive studies were performed in cell line
models, whereas only very little is known about expression of
surface antigens or targets in primary breast CSC. One inter-
esting question will be to learn whether breast CSC express
Fig. 2. Correlation between expression of CD44 and proliferative poten- members of the ERBB family of oncogenic receptors. In fact,
tial of LT97 cells. The adenoma cell strain LT97 was cultured in complete ERBB receptors are major targets of therapy in this type of
medium and passaged once every 10–14 days. Expression of CD44 on LT97
cancer. Data published so far suggest that mammary CSC
cells was determined by flow cytometry in early (passage up to 20), inter-
mediate (passage 20–30), and late (passage above 30) cultures. The figure express EGFR and ERBB2/Her2 but lack estrogen receptor
shows the percentage of CD44+ cells (black bars) and the plating efficiency alpha [135,136]. This may be due to several different mecha-
(hatched bars) over time (early, intermediate, late passage). nisms such as gene silencing by epigenetic events [137,138].

Please cite this article in press as: Schulenburg A, et al. Neoplastic stem cells: Current concepts and clinical perspectives. Crit Rev
Oncol/Hematol (2010), doi:10.1016/j.critrevonc.2010.01.001
ONCH-1372; No. of Pages 20
ARTICLE IN PRESS
A. Schulenburg et al. / Critical Reviews in Oncology/Hematology xxx (2010) xxx–xxx 11

Whether this holds true for all breast cancer variants remains were also able to show that these tumors again contained
at present unknown. Ewing sarcoma-initiating stem cells when re-transplanted
into secondary recipient mice [151].
9.6. Brain tumor stem cells
9.9. Other solid tumors
Hemmati et al. characterized and isolated stem cells from
human paediatric brain tumors of different pathologic sub- In gastric cancer up to now only CD44 is described as
types, including glioblastoma and medulloblastoma [27]. The a potential CSC marker [152]. Another group postulated
authors reported that a minority of the brain tumor cells that some gastric tumors may even arise from bone mar-
were able to form neurospheres and to grow to tumors when row cells [153]. Lung CSC are not well characterized so
xenotransplanted into rat brain. These neurosphere-derived far. In one paper, Kim et al. describe bronchioalveolar stem
cells were found to be Lin-negative and expressed CD133 cells (BASCs) as potential lung CSC, whereas in another
as well as nestin [27]. More recently, Singh et al. confirmed paper, lung CSC are identified as CD133+ cells [154,155].
that brain CSC reside within the CD133+/nestin+ fraction When 1 × 104 EpCAM+/CD133+ cells from small and non-
of brain tumor cells in a NOD/SCID mouse model [15]. In small-cell lung cancers were injected subcutaneously, tumors
fact, when as few as 100 CD133+ glioblastoma cells were developed within 3 weeks in SCID mice [155]. This obser-
injected into the brain of NOD/SCID mice, tumors devel- vation has recently been confirmed for non-small-cell lung
oped after 12 to 14 weeks in most animals. Bao et al. found cancer cells by Tirino et al. [156]. As in many other organs,
that L1 CAM may also serve as a marker for brain CSC there is also evidence for the expression of CD44 as well as
since L1 CAM is overexpressed on CD133+ cells, and tar- CD133 on the surface of ovarian CSC [157–159]. Baba et
geting of L1 CAM reduced the tumorigenicity of CD133+ al. injected 4–5-week-old BALB/cAnNCr-nu/nu mice with
cells [43]. Ehtesham et al. showed that glioblastoma pro- sorted CD133− (left flank) and CD133+ (right flank) A2780
genitors in glioma-derived spheres may express CXCR4 cells. They observed that CD133+ cells formed tumors that
and that binding of its ligand CXCL12 results in increased grew to a larger size and appeared within shorter time than
growth of neurosphere cells [139]. Recently, Read et al. tumors from CD133− cells isolated from the same parental
found that CD15+ cells are enriched for CSC in medulloblas- cell line [158]. On the other hand, Kusumbe et al. showed
tomas [140]. After intracranial implantation of 3 × 105 cells, that CD133+ cells in ovarian cancer are only responsible for
Ptc+/− mice develop tumors within 50 days. the development of the tumor vasculature by giving rise to
endothelial cells. By contrast, CD133+ ovarian cells were not
9.7. Prostate CSC tumorigenic in this study [160]. In one report CD133+ cells
from anaplastic thyroid cancer showed reconstitution of the
The isolation of CSC in prostate cancer is difficult due tumor in NOD/SCID mice after injection of at least 10,000
to the heterogeneity of prostate tumors and the small sam- CD133+ cells [161].
ple size. It is unclear whether prostate CSC derive from
the basal or luminal layer [141]. However, selection of
cells with a CD133+/alpha 2 beta 1 integrin+/CD44+ phe- 10. Melanoma stem cells (MSC)
notype resulted in enrichment for prostate cancer-initiating
cells [142–145]. Moreover, CD44+ prostate cancer cells So far, little is known about tumor-initiating cells in skin
have recently been described as more invasive and shown cancer patients. In fact, CSC have only been investigated
to have increased Hedgehog signalling and activation of the and partly characterized in malignant melanomas. Fang et al.
PI3K/AKT signalling pathway compared to CD44− cells described that melanoma spheres can be grown from pri-
[146–148]. In 2008, Hurt et al. observed stem cell-like prop- mary melanoma cells, and that melanoma sphere-derived
erties of CD44+/CD24− cells derived from prostate cell lines cells exhibit long-term growth, multilineage potential, and
(LNCaP and DU145) [51]. The expression of androgen recep- tumor-initiating potential in SCID mice [24]. They found that
tors on prostate CSC is controversial [142,147,149]. The gene a subpopulation of cells in these spheres is CD20+/CD45−
expression profile of prostate cancer stem cells can also be cells that co-express melanoma antigens. This CD20+ frac-
related to Gleason grade and patient outcome [150]. tion of melanoma cells, when cultured separately, was found
to form larger spheres than CD20− cells, and showed
9.8. Sarcoma long-term proliferation in vitro. Based on these data, it
is tempting to speculate that melanoma stem cells reside
Recently, Suva et al. identified for the first time a Ewing within the CD20+ fraction of cells, although this has not
sarcoma cancer stem cell by sorting with magnetic beads for formally been proven by mouse experiments using primary
CD133+ cells [151]. After cell isolation from primary Ewing melanoma cells sorted for CD20+ and CD20− cells. In addi-
sarcoma tissue, these CD133+ cells were found to establish tion, the expression of CD20 on melanoma cells has not
Ewing sarcomas with parental tumor phenotype and hierar- been confirmed in other studies. Another marker that has
chical organization in NOD/SCID mice [151]. The authors been discussed as a potential stem cell marker for human

Please cite this article in press as: Schulenburg A, et al. Neoplastic stem cells: Current concepts and clinical perspectives. Crit Rev
Oncol/Hematol (2010), doi:10.1016/j.critrevonc.2010.01.001
ONCH-1372; No. of Pages 20
ARTICLE IN PRESS
12 A. Schulenburg et al. / Critical Reviews in Oncology/Hematology xxx (2010) xxx–xxx

melanoma is CD133 [162]. It has been described that only been described that EPO receptor expression is associated
0.2–0.8% of primary metastatic melanoma cells express with disease progression and the metastasizing potential of
CD133 [162]. Monzani et al. observed that 40–50 days melanomas [169,172]. Finally, EPO has been described to
after subcutaneous injection of 3.5 × 105 CD133-positive initiate signalling and promote survival in human melanomas
cells into NOD/SCID mice, these mice develop detectable [171]. Whether indeed the EPO receptor is a functionally rev-
melanoma lesions, whereas the CD133-negative fraction did elant antigen expressed specifically on melanoma initiating
not form tumors in NOD/SCID mice [162]. These data sug- cells is currently under investigation.
gest that melanoma stem cells may reside within a CD133+
fraction of the clone, at least in metastatic melanomas [163].
In line with this hypothesis, several melanoma cells lines 11. CSC plasticity—the Hydra Model of CSC
display CD133 [164,165]. Other studies have shown that cer- development
tain drug-transporters that (when expressed) are indicative
of chemoresistance, may be expressed on melanoma stem A number of previous and more recent data suggest that
cells (and stem cells in other tumors). Among these trans- neoplastic stem cell clones display substantial plasticity and
porters are MDR1, ABCG2 and ABCB5 [166]. Schatton et al. are often composed of several different subclones. In many
described that the ABCB5+ fraction of primary (freshly iso- cases, subclone formation may precede the development of
lated) melanoma cells is enriched for melanoma-repopulating a frank neoplasm, and only a few (or even only one) of
stem cells, whereas ABCB5-negative cells are less capable these subclones may progress to an overt malignancy. This
of initiating melanomas in NOD/SCID mice [54]. ABCB5+ assumption is consistent with the multi-hit theory of cancer
melanoma cells were found to co-express other potential development [173–176] and would predict that neoplastic
stem cell markers including ABCB1, TIE1, nestin, and CD44 stem cells detectable in the NOD/SCID or NOG (NSG)
[54,167]. mouse assay would not all be able to repopulate all com-
However, the melanoma-initiating potential of single ponents (cell subclones) in a given neoplasm. Rather, this
(stem) cells may greatly depend on the microenvironment model predicts early formation of subclones with different
and the mouse strain employed to demonstrate repopulation. transformation potential, and each subclone must be expected
Quintana et al. have recently shown that up to 30% of all to contain subclone-specific stem cells. A good example
melanoma cells are melanoma-repopulating cells (indepen- for the presence of multiple subclones in one neoplasm is
dent of their phenotype) when interleukin-2 receptor gamma the coexistence of two histologically different hematopoi-
null NOD/SCID mice are used, whereas tumorigenicity of etic disorders in one patient. Another example is CML,
the same cells is much lower when NOD/SCID mice with where during treatment with imatinib, one or more different
an intact interleukin-2 receptor are employed [23]. These subclones exhibiting imatinib-resistant BCR/ABL mutants
data suggest that the frequency of melanoma-initiating cells may be selected (by treatment), and each of these subclones
may be relatively high, at least in a severely immunocom- may progress into a clinically relevant leukemia (subclone-
promised host. Whether the same cells (all these cells) are specific progress) [91,177,178]. Whereas many of these
also able to repopulate melanomas in an immunocompetent subclones bearing (drug-resistant) BCR/ABL mutations may
host or in patients remains unclear. In fact, the stem cell be present (as small clones) before imatinib is started, some
function of a given tumor/melanoma cell may not only be of these subclones may progress to leukemia during imatinib
predetermined by intrinsic factors (genetic and epigenetic therapy which may point to clonal instability and a potential
stem cell programs) but also be microenvironmental factors effect of the drug on subclone formation. Similar observations
(factors defining the stem cell niche) and also the immune of stem cell plasticity have been made in other hematopoi-
system (immunosurveillance, apoptosis-induction by killer etic malignancies and may also apply to non-hematopoietic
cells, tumor cell phagocytosis). neoplasms [179].
The observation that up to 30% of melanoma cells may Recent data suggest the stem cell plasticity may even
have the potential to repopulate melanomas in NOG mice involve the supportive stroma or tumor/leukemia-associated
and thus have stem cell function, has changed our views on angiogenesis. In particular, it has been described that
what types and subpopulations of melanoma cells indeed are endothelial cells in lymphomas and other neoplasms are of
repopulating and non-repopulating cells. Unfortunately, only monoclonal origin [180]. Other studies suggest that cultured
a few markers can clearly discriminate larger subpopulations bone marrow stroma cells and mesenchymal progenitors in
of melanoma cells with different potential to metastasize and patients with Ph+ CML are of clonal origin [181]. However,
to form new tumor lesions in patients. One such marker is other studies suggest that stromal cells and mesenchymal pro-
the erythropoietin (EPO) receptor. In fact, the EPO receptor genitor cells in patients with CML are non-clonal cells and
is only expressed in trace amounts in normal melanocytes, not derived from the Ph+ clone [182,183].
but is expressed in melanoma cells [168–171]. Whereas in The above described heterogeneity and plasticity of CSC
primary melanomas, only a small subpopulation of cells dis- populations in various neoplasms may be one reason for
play the EPO receptor, in metastatic melanomas up to 30% of the complexity of CSC evolution and function, and may
all cells express this antigen [168]. Moreover, it has recently also explain why it is difficult to design effective treatment

Please cite this article in press as: Schulenburg A, et al. Neoplastic stem cells: Current concepts and clinical perspectives. Crit Rev
Oncol/Hematol (2010), doi:10.1016/j.critrevonc.2010.01.001
ONCH-1372; No. of Pages 20
ARTICLE IN PRESS
A. Schulenburg et al. / Critical Reviews in Oncology/Hematology xxx (2010) xxx–xxx 13

approaches sufficient to eliminate all these cells and sub- found to induce apoptosis in CD34+/CD38−/CD33+ stem
clones in cancer/leukemia patients [184–186]. cells in AML [65] (Fig. 1). A novel and promising marker
for stem cell targeting in AML may be CLL-1 that is able
to discriminate between normal and leukemic stem cells in
12. Strategies for the successful elimination of CSC the bone marrow [86]. Another promising marker is CD96
since Hosen et al. showed that CD96 is specific for AML
Several different strategies have been considered to inhibit stem cells. He showed that CD96+ AML cells can engraft
growth and/or survival of CSC, with the ultimate goal to elim- irradiated Rag2(−/−) gamma(c)(−/−) mice [85]. However,
inate all CSC in these malignancies. These concepts include so far no targeted drugs specific for CD96 or CLL-1 have
relevant surface targets, signal transduction molecules, and been developed. Several different CD antigens are employed
certain survival molecules expressed in CSC (Fig. 3). Many as targets of therapy in B-cell Non-Hodgkin’s lymphomas,
concepts still relate to myeloid or lymphoid neoplasms, including CD20, CD22, CD23, CD25, or CD52. Clinical tri-
whereas so far, only a few treatment strategies have been pre- als have shown that antibodies directed against some of these
sented for solid tumors and melanomas. Moreover, whereas antigens (especially CD20 and CD52) can improve treat-
much is known about the expression of various surface and ment and prognosis in these patients. It is therefore tempting
cytoplasmic drug targets in solid tumors and leukemias, only to speculate that some of these antigens are also expressed
very little is known about expression of the same targets in on immature lymphoma-initiating tumor cells. Studies are
neoplastic stem cells (Fig. 3). ongoing to define the CD antigen profile in lymphoid stem
In myeloid neoplasms, potential surface targets include cells in various NHLs. An interesting aspect is that some
the IL-3R alpha chain CD123, the Mylotarg-receptor Siglec- of the lymphoid markers, like CD20, are also expressed on
3 (CD33), CD44, and the tyrosine kinase receptor KIT non-hematopoietic CSC. In solid tumors, cell surface targets
(CD117). Target expression can be exploited by selecting include members of the ErbB receptor family, IGF receptors,
blocking antibodies or by constructing antibody–toxin con- TGF␤ receptors, and FGF receptors [190–193]. A general
jugates, cytokine–ligand–toxin conjugates, or antibody–drug problem with surface targets is that CSC subclones often
conjugates. A good example for a ligand–toxin con- exhibit or develop resistance against antibody-based or other
jugate is the IL-3-diphteria toxin fusion protein [187]. drugs. Several different mechanisms of drug resistance have
An example for an antibody–drug conjugate is the been discussed, including expression of multi-drug resistance
CD33-targeting fusion molecule gemtuzumab–ozogamcin gene products like MDR1 or reduced antibody-binding. In
(Mylotarg) [81–83] (Fig. 1). It has also been reported that addition, subclones that do not express the surface CD anti-
an unconjugated blocking CD123 antibody can effectivity gen may be selected by antibody-based therapy. Therefore,
reduce leukemia cell growth in mice [188] and even in a few antibody-therapy is usually combined with conventional or
patients with AML [189]. So far, only a few studies have with other targeted drugs.
attempted to demonstrate functional significance of target Apart from surface markers, also intracellular targets
expression in leukemic stem cells. In one study, Mylotarg was have been identified in CSC. Among those, signal trans-

Fig. 3. Expression of molecular targets in and on neoplastic cells and their progenitors. Whereas much is known about target expression profiles in more mature
neoplastic cells (left part of panel), little is known about expression of molecular targets in leukemic (neoplastic) stem cells (right panel part “?”). ST, signal
transduction; TK, tyrosine kinase(s); TF, transcription factor(s); TSG, tumor suppressor gene(s); DR, death receptors.

Please cite this article in press as: Schulenburg A, et al. Neoplastic stem cells: Current concepts and clinical perspectives. Crit Rev
Oncol/Hematol (2010), doi:10.1016/j.critrevonc.2010.01.001
ONCH-1372; No. of Pages 20
ARTICLE IN PRESS
14 A. Schulenburg et al. / Critical Reviews in Oncology/Hematology xxx (2010) xxx–xxx

duction molecules and survival molecules may represent Reviewers


most promising target antigens. These drugs include tyro-
sine kinase inhibitors, farnesyl transferase inhibitors, and Dr. Dominique Bonnet, Cancer Research UK, London
other kinase inhibitors including PI3 kinase and mTOR Research Institute, Haematopoietic Stem Cell Laboratory, 44
blockers [194–196]. In some myeloid neoplasms like CML, Lincoln’s Inn Fields, London WC2A 3PX, United Kingdom.
BCR/ABL tyrosine kinase inhibitors have been used with
considerable success [197]. More recently tyrosine kinase
inhibitors have also been employed in clinical trials in solid References
tumors and melanomas [98,196,198–209]. Among survival- [1] Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer,
related targets, most promising molecules may be members and cancer stem cells. Nature 2001;414:105–11.
of the BCL-2 family (BCL-2, MCL-1, BCL-xL, others) and [2] Ailles LE, Weissman IL. Cancer stem cells in solid tumors. Curr Opin
various Heat shock proteins (HSP32, HSP70, HSP90, others) Biotechnol 2007;18:460–6.
[210–212]. Studies are ongoing to define whether these tar- [3] Cho RW, Clarke MF. Recent advances in cancer stem cells. Curr Opin
Genet Dev 2008;18:48–53.
gets are expressed in CSC in various hematopoietic or solid [4] Dalerba P, Cho RW, Clarke MF. Cancer stem cells: models and con-
tumors. Likewise, it has been described that the HSP32, also cepts. Annu Rev Med 2007;58:267–84.
known as heme oxygenase 1, is expressed in CSC in various [5] Korkaya H, Wicha MS. Selective targeting of cancer stem cells: a new
leukemias and solid tumor models [213,214]. concept in cancer therapeutics. BioDrugs 2007;21:299–310.
Despite intensive research and some progress in targeted [6] Besancon R, Valsesia-Wittmann S, Puisieux A, de Fromentel CC,
Maguer-Satta V. Cancer stem cells: the emerging challenge of drug
therapy in solid tumors, only a few targeted drugs have pro- targeting. Curr Med Chem 2009;16:394–416.
duced convincing results in clinical trials, which may be due [7] Polyak K, Hahn WC. Roots and stems: stem cells in cancer. Nat Med
to resistance and the complexity of the signalling cascades 2006;12:296–300.
in cancer cells and CSC. This is mainly due to (the various [8] Lapidot T, Sirard C, Vormoor J, et al. A cell initiating human acute
forms of) resistance as well as CSC plasticity with target- myeloid leukaemia after transplantation into SCID mice. Nature
1994;367:645–8.
negative subclone formation. Therefore, it seems reasonable [9] Bonnet D, Bhatia M, Wang JC, Kapp U, Dick JE. Cytokine treatment
to combine targeted drugs (antibodies, small molecules) with or accessory cells are required to initiate engraftment of purified prim-
each other or with conventional therapy (chemotherapy) in itive human hematopoietic cells transplanted at limiting doses into
order to overcome drug resistance in CSC. NOD/SCID mice. Bone Marrow Transplant 1999;23:203–9.
Finally, provided that specific antigens of CSC are iden- [10] Pearson T, Greiner DL, Shultz LD. Creation of “humanized” mice to
study human immunity. Curr Protoc Immunol 2008 [Chapter 15:Unit
tified, an attractive approach would be to induce specific 15 21].
immune responses and to establish vaccination therapy [11] Ishikawa F, Saito Y, Yoshida S, Harada M, Shultz LD. The dif-
approaches in these malignancies. ferentiative and regenerative properties of human hematopoietic
stem/progenitor cells in NOD-SCID/IL2rgamma(null) mice. Curr Top
Microbiol Immunol 2008;324:87–94.
13. Concluding remarks and future directions [12] Hermann PC, Huber SL, Herrler T, et al. Distinct populations of cancer
stem cells determine tumor growth and metastatic activity in human
The emerging concept of neoplastic stem cells and CSC- pancreatic cancer. Cell Stem Cell 2007;1:313–23.
[13] Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF.
related targets may offer new insights into the biology and the Prospective identification of tumorigenic breast cancer cells. Proc Natl
pathogenesis of various malignant disorders and new possi- Acad Sci USA 2003;100:3983–8.
bilities for the design of targeted drug therapies. Since CSC [14] Bao S, Wu Q, McLendon RE, et al. Glioma stem cells promote radiore-
display considerable heterogeneity and plasticity, eradication sistance by preferential activation of the DNA damage response.
of these cells and thus cure may only be reached when combi- Nature 2006;444:756–60.
[15] Singh SK, Hawkins C, Clarke ID, et al. Identification of human brain
nations of anticancer drugs (therapies) are applied. Therefore, tumour initiating cells. Nature 2004;432:396–401.
treatment designs aiming at cancer/leukemia stem cell elim- [16] Li C, Heidt DG, Dalerba P, et al. Identification of pancreatic cancer
ination need to take all potential targets and all relevant stem stem cells. Cancer Res 2007;67:1030–7.
cell subclones into account. There is hope for the future [17] Yang ZF, Ho DW, Ng MN, et al. Significance of CD90+ cancer stem
that such novel treatment approaches will improve therapy cells in human liver cancer. Cancer Cell 2008;13:153–66.
[18] Bhatia M, Wang JC, Kapp U, Bonnet D, Dick JE. Purification
in cancer patients. of primitive human hematopoietic cells capable of repopulating
immune-deficient mice. Proc Natl Acad Sci USA 1997;94:5320–5.
[19] Bonnet D, Dick JE. Human acute myeloid leukemia is organized as
Conflict of interest a hierarchy that originates from a primitive hematopoietic cell. Nat
Med 1997;3:730–7.
All the authors declare that they have no proprietary, finan- [20] Al-Hajj M. Cancer stem cells and oncology therapeutics. Curr Opin
cial, professional or other personal interest of any nature or Oncol 2007;19:61–4.
kind in any product, service and/or company that could be [21] Till JE, Mc CE. A direct measurement of the radiation sensitivity of
normal mouse bone marrow cells. Radiat Res 1961;14:213–22.
construed as influencing the position presented in the review [22] Taussig DC, Miraki-Moud F, Anjos-Afonso F, et al. Anti-CD38
entitled “Neoplastic stem cells: Current concepts and clinical antibody-mediated clearance of human repopulating cells masks the
perspectives”. heterogeneity of leukemia-initiating cells. Blood 2008;112:568–75.

Please cite this article in press as: Schulenburg A, et al. Neoplastic stem cells: Current concepts and clinical perspectives. Crit Rev
Oncol/Hematol (2010), doi:10.1016/j.critrevonc.2010.01.001
ONCH-1372; No. of Pages 20
ARTICLE IN PRESS
A. Schulenburg et al. / Critical Reviews in Oncology/Hematology xxx (2010) xxx–xxx 15

[23] Quintana E, Shackleton M, Sabel MS, Fullen DR, Johnson TM, Mor- [47] Corbeil D, Roper K, Hellwig A, et al. The human AC133 hematopoi-
rison SJ. Efficient tumour formation by single human melanoma cells. etic stem cell antigen is also expressed in epithelial cells and targeted
Nature 2008;456:593–8. to plasma membrane protrusions. J Biol Chem 2000;275:5512–20.
[24] Fang D, Nguyen TK, Leishear K, et al. A tumorigenic sub- [48] Shmelkov SV, St Clair R, Lyden D, Rafii S. AC133/CD133/Prominin-
population with stem cell properties in melanomas. Cancer Res 1. Int J Biochem Cell Biol 2005;37:715–9.
2005;65:9328–37. [49] Gunthert U. CD44: a multitude of isoforms with diverse functions.
[25] Dontu G, Abdallah WM, Foley JM, et al. In vitro propagation and Curr Top Microbiol Immunol 1993;184:47–63.
transcriptional profiling of human mammary stem/progenitor cells. [50] Sackstein R, Merzaban JS, Cain DW, et al. Ex vivo glycan engineer-
Genes Dev 2003;17:1253–70. ing of CD44 programs human multipotent mesenchymal stromal cell
[26] Ricci-Vitiani L, Lombardi DG, Pilozzi E, et al. Identification trafficking to bone. Nat Med 2008;14:181–7.
and expansion of human colon-cancer-initiating cells. Nature [51] Hurt EM, Kawasaki BT, Klarmann GJ, Thomas SB, Farrar WL.
2007;445:111–5. CD44+ CD24(−) prostate cells are early cancer progenitor/stem cells
[27] Hemmati HD, Nakano I, Lazareff JA, et al. Cancerous stem cells that provide a model for patients with poor prognosis. Br J Cancer
can arise from pediatric brain tumors. Proc Natl Acad Sci USA 2008;98:756–65.
2003;100:15178–83. [52] Schulenburg A, Cech P, Herbacek I, et al. CD44-positive colorectal
[28] Vermeulen L, Todaro M, de Sousa Mello F, et al. Single-cell cloning of adenoma cells express the potential stem cell markers musashi antigen
colon cancer stem cells reveals a multi-lineage differentiation capac- (msi1) and ephrin B2 receptor (EphB2). J Pathol 2007;213:152–60.
ity. Proc Natl Acad Sci USA 2008;105:13427–32. [53] Botchkina IL, Rowehl RA, Rivadeneira DE, et al. Phenotypic sub-
[29] Dexter TM, Allen TD, Lajtha LG. Conditions controlling the populations of metastatic colon cancer stem cells: genomic analysis.
proliferation of haemopoietic stem cells in vitro. J Cell Physiol Cancer Genomics Proteomics 2009;6:19–29.
1977;91:335–44. [54] Schatton T, Murphy GF, Frank NY, et al. Identification of cells initi-
[30] Mayani H, Flores-Figueroa E, Chávez-González A. In vitro biology ating human melanomas. Nature 2008;451:345–9.
of human myeloid leukemia. Leuk Res 2009;33:624–37. [55] White DL, Saunders VA, Dang P, et al. OCT-1-mediated influx is a
[31] Coulombel L, Eaves C, Kalousek D, Gupta C, Eaves A. Long- key determinant of the intracellular uptake of imatinib but not nilo-
term marrow culture of cells from patients with acute myelogenous tinib (AMN107): reduced OCT-1 activity is the cause of low in vitro
leukemia. Selection in favor of normal phenotypes in some but not all sensitivity to imatinib. Blood 2006;108:697–704.
cases. J Clin Invest 1985;75:961–9. [56] Wu C, Alman BA. Side population cells in human cancers. Cancer
[32] Lagneaux L, Delforge A, De Bruyn C, Bernier M, Bron D. Adhesion Lett 2008;268:1–9.
to bone marrow stroma inhibits apoptosis of chronic lymphocytic [57] Chen YC, Chen YW, Hsu HS, et al. Aldehyde dehydrogenase 1 is
leukemia cells. Leuk Lymphoma 1999;35:445–53. a putative marker for cancer stem cells in head and neck squamous
[33] Panayiotidis P, Jones D, Ganeshaguru K, Foroni L, Hoffbrand AV. cancer. Biochem Biophys Res Commun 2009;385:307–13.
Human bone marrow stromal cells prevent apoptosis and support [58] Jiang F, Qiu Q, Khanna A, et al. Aldehyde dehydrogenase 1 is a
the survival of chronic lymphocytic leukaemia cells in vitro. Br J tumor stem cell-associated marker in lung cancer. Mol Cancer Res
Haematol 1996;92:97–103. 2009;7:330–8.
[34] Reynolds BA, Weiss S. Clonal and population analyses demonstrate [59] Ucar D, Cogle CR, Zucali JR, et al. Aldehyde dehydrogenase activ-
that an EGF-responsive mammalian embryonic CNS precursor is a ity as a functional marker for lung cancer. Chem Biol Interact
stem cell. Dev Biol 1996;175:1–13. 2009;178:48–55.
[35] Scadden DT. The stem-cell niche as an entity of action. Nature [60] Croker AK, Goodale D, Chu J, et al. High aldehyde dehydrogenase
2006;441:1075–9. and expression of cancer stem cell markers selects for breast cancer
[36] Torok-Storb B. Cellular interactions. Blood 1988;72:373–85. cells with enhanced malignant and metastatic ability. J Cell Mol Med
[37] Greenberger JS. The hematopoietic microenvironment. Crit Rev 2008;13:2236–52.
Oncol Hematol 1991;11:65–84. [61] Cheung AM, Wan TS, Leung JC, et al. Aldehyde dehydrogenase activ-
[38] Mayani H, Guilbert LJ, Janowska-Wieczorek A. Biology of the ity in leukemic blasts defines a subgroup of acute myeloid leukemia
hemopoietic microenvironment. Eur J Haematol 1992;49:225–33. with adverse prognosis and superior NOD/SCID engrafting potential.
[39] Rosel M, Khaldoyanidi S, Zawadzki V, Zoller M. Involvement of Leukemia 2007;21:1423–30.
CD44 variant isoform v10 in progenitor cell adhesion and maturation. [62] Ma S, Chan KW, Lee TK, et al. Aldehyde dehydrogenase discrimi-
Exp Hematol 1999;27:698–711. nates the CD133 liver cancer stem cell populations. Mol Cancer Res
[40] Alakel N, Jing D, Muller K, Bornhauser M, Ehninger G, Ordemann 2008;6:1146–53.
R. Direct contact with mesenchymal stromal cells affects migratory [63] Douville J, Beaulieu R, Balicki D. ALDH1 as a functional marker
behavior and gene expression profile of CD133+ hematopoietic stem of cancer stem and progenitor cells. Stem Cells Dev 2009;18:17–
cells during ex vivo expansion. Exp Hematol 2009;37:504–13. 26.
[41] Raveh S, Gavert N, Spiegel I, Ben-Ze’ev A. The cell adhesion nectin- [64] Florian S, Sonneck K, Hauswirth AW, et al. Detection of molecular
like molecules (Necl) 1 and 4 suppress the growth and tumorigenic targets on the surface of CD34+/CD38− stem cells in various myeloid
ability of colon cancer cells. J Cell Biochem 2009;108:326–36. malignancies. Leuk Lymphoma 2006;47:207–22.
[42] Gavert N, Ben-Shmuel A, Raveh S, Ben-Ze’ev A. L1-CAM in can- [65] Herrmann H. Phenotypic and functional characterization of
cerous tissues. Expert Opin Biol Ther 2008;8:1749–57. CD34+/CD38−/CD123+ leukemic progenitor (stem) cells in AML:
[43] Bao S, Wu Q, Li Z, et al. Targeting cancer stem cells through L1CAM a flow cytometric approach. Blood (ASH Annual Meeting Abstracts)
suppresses glioma growth. Cancer Res 2008;68:6043–8. 2008:483.
[44] O’Brien CA, Pollett A, Gallinger S, Dick JE. A human colon cancer [66] Morimoto K, Kim SJ, Tanei T, et al. Stem cell marker aldehyde
cell capable of initiating tumour growth in immunodeficient mice. dehydrogenase 1-positive breast cancers are characterized by negative
Nature 2007;445:106–10. estrogen receptor, positive human epidermal growth factor receptor
[45] Dalerba P, Dylla SJ, Park IK, et al. Phenotypic characterization type 2, and high Ki67 expression. Cancer Sci 2009;100:1062–8.
of human colorectal cancer stem cells. Proc Natl Acad Sci USA [67] Korkaya H, Paulson A, Iovino F, Wicha MS. HER2 regulates the
2007;104:10158–63. mammary stem/progenitor cell population driving tumorigenesis and
[46] Corbeil D, Fargeas CA, Huttner WB. Rat prominin, like its mouse and invasion. Oncogene 2008;27:6120–30.
human orthologues, is a pentaspan membrane glycoprotein. Biochem [68] Hewish M, Chau I, Cunningham D. Insulin-like growth factor 1
Biophys Res Commun 2001;285:939–44. receptor targeted therapeutics: novel compounds and novel treatment

Please cite this article in press as: Schulenburg A, et al. Neoplastic stem cells: Current concepts and clinical perspectives. Crit Rev
Oncol/Hematol (2010), doi:10.1016/j.critrevonc.2010.01.001
ONCH-1372; No. of Pages 20
ARTICLE IN PRESS
16 A. Schulenburg et al. / Critical Reviews in Oncology/Hematology xxx (2010) xxx–xxx

strategies for cancer medicine. Recent Patents Anticancer Drug Dis- tosis induced by Bcr-Abl tyrosine kinase inhibitors. Leukemia
cov 2009;4:54–72. 2002;16:1589–95.
[69] Gotoh N. Control of stemness by fibroblast growth factor signal- [91] Jordanides NE, Jorgensen HG, Holyoake TL, Mountford JC. Func-
ing in stem cells and cancer stem cells. Curr Stem Cell Res Ther tional ABCG2 is overexpressed on primary CML CD34+ cells and is
2009;4:9–15. inhibited by imatinib mesylate. Blood 2006;108:1370–3.
[70] Phillips TM, Kim K, Vlashi E, McBride WH, Pajonk F. Effects of [92] Jiang X, Zhao Y, Smith C, et al. Chronic myeloid leukemia stem cells
recombinant erythropoietin on breast cancer-initiating cells. Neopla- possess multiple unique features of resistance to BCR-ABL targeted
sia 2007;9:1122–9. therapies. Leukemia 2007;21:926–35.
[71] Ponta H, Sherman L, Herrlich PA. CD44: from adhesion molecules [93] Chen Y, Hu Y, Zhang H, Peng C, Li S. Loss of the Alox5 gene
to signalling regulators. Nat Rev Mol Cell Biol 2003;4:33–45. impairs leukemia stem cells and prevents chronic myeloid leukemia.
[72] Benedetti F. CD34+ cells: biological aspects. Tumori 1996;82:S3–13. Nat Genet 2009;41:783–92.
[73] Holyoake TL, Jiang X, Jorgensen HG, et al. Primitive quiescent [94] Jamieson CH. Chronic myeloid leukemia stem cells. Hematol Am
leukemic cells from patients with chronic myeloid leukemia sponta- Soc Hematol Educ Program 2008;2008:436–42.
neously initiate factor-independent growth in vitro in association with [95] Zhao C, Chen A, Jamieson CH, et al. Hedgehog signalling is essential
up-regulation of expression of interleukin-3. Blood 2001;97:720–8. for maintenance of cancer stem cells in myeloid leukaemia. Nature
[74] Kim JB, Ko E, Han W, et al. CD24 cross-linking induces apoptosis in, 2009;458:776–9.
and inhibits migration of, MCF-7 breast cancer cells. BMC Cancer [96] Yee KW, Zeng Z, Konopleva M, et al. Phase I/II study of the mam-
2008;8:118. malian target of rapamycin inhibitor everolimus (RAD001) in patients
[75] Yu F, Yao H, Zhu P, et al. let-7 regulates self renewal and tumorigenic- with relapsed or refractory hematologic malignancies. Clin Cancer
ity of breast cancer cells. Cell 2007;131:1109–23. Res 2006;12:5165–73.
[76] Ginestier C, Hur MH, Charafe-Jauffret E, et al. ALDH1 is a marker [97] Guzman ML, Neering SJ, Upchurch D, et al. Nuclear factor-kappaB
of normal and malignant human mammary stem cells and a predictor is constitutively activated in primitive human acute myelogenous
of poor clinical outcome. Cell Stem Cell 2007;1:555–67. leukemia cells. Blood 2001;98:2301–7.
[77] Vercauteren SM, Sutherland HJ. CD133 (AC133) expression on AML [98] Recher C, Beyne-Rauzy O, Demur C, et al. Antileukemic activity of
cells and progenitors. Cytotherapy 2001;3:449–59. rapamycin in acute myeloid leukemia. Blood 2005;105:2527–34.
[78] Fialkow PJ, Jacobson RJ, Papayannopoulou T. Chronic myelocytic [99] Druker BJ, Tamura S, Buchdunger E, et al. Effects of a selective
leukemia: clonal origin in a stem cell common to the granulo- inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl positive
cyte, erythrocyte, platelet and monocyte/macrophage. Am J Med cells. Nat Med 1996;2:561–6.
1977;63:125–30. [100] Chen ZX, Xue YQ, Zhang R, et al. A clinical and experimental
[79] Dick JE. Complexity of the human acute myeloid leukemia stem cell study on all-trans retinoic acid-treated acute promyelocytic leukemia
compartment: implications for therapy. Biol Blood Marrow Trans- patients. Blood 1991;78:1413–9.
plant 2005;11:9–11. [101] Moliterno AR, Williams DM, Rogers O, Isaacs MA, Spivak JL.
[80] Majeti R, Becker MW, Tian Q, et al. Dysregulated gene expression Phenotypic variability within the JAK2 V617F-positive MPD: roles
networks in human acute myelogenous leukemia stem cells. Proc Natl of progenitor cell and neutrophil allele burdens. Exp Hematol
Acad Sci USA 2009;106:3396–401. 2008;36:1480–6.
[81] Sievers EL, Appelbaum FR, Spielberger RT, et al. Selective ablation [102] Catani L, Zini R, Sollazzo D, et al. Molecular profile of CD34+
of acute myeloid leukemia using antibody-targeted chemotherapy: stem/progenitor cells according to JAK2V617F mutation status in
a phase I study of an anti-CD33 calicheamicin immunoconjugate. essential thrombocythemia. Leukemia 2009;23:997–1000.
Blood 1999;93:3678–84. [103] Li S, Kralovics R, De Libero G, Theocharides A, Gisslinger H,
[82] Sievers EL, Larson RA, Stadtmauer EA, et al. Efficacy and safety Skoda RC. Clonal heterogeneity in polycythemia vera patients
of gemtuzumab ozogamicin in patients with CD33-positive acute with JAK2 exon12 and JAK2-V617F mutations. Blood 2008;111:
myeloid leukemia in first relapse. J Clin Oncol 2001;19:3244–54. 3863–6.
[83] Feldman E, Kalaycio M, Weiner G, et al. Treatment of relapsed or [104] Lataillade JJ, Pierre-Louis O, Hasselbalch HC, et al. Does primary
refractory acute myeloid leukemia with humanized anti-CD33 mon- myelofibrosis involve a defective stem cell niche? From concept to
oclonal antibody HuM195. Leukemia 2003;17:314–8. evidence. Blood 2008;112:3026–35.
[84] Jin L, Hope KJ, Zhai Q, Smadja-Joffe F, Dick JE. Targeting of [105] Cobaleda C, Gutierrez-Cianca N, Perez-Losada J, et al. A primitive
CD44 eradicates human acute myeloid leukemic stem cells. Nat Med hematopoietic cell is the target for the leukemic transformation in
2006;12:1167–74. human philadelphia-positive acute lymphoblastic leukemia. Blood
[85] Hosen N, Park CY, Tatsumi N, et al. CD96 is a leukemic stem cell- 2000;95:1007–13.
specific marker in human acute myeloid leukemia. Proc Natl Acad [106] Hong D, Gupta R, Ancliff P, et al. Initiating and cancer-
Sci USA 2007;104:11008–13. propagating cells in TEL-AML1-associated childhood leukemia.
[86] van Rhenen A, van Dongen GA, Kelder A, et al. The novel AML stem Science 2008;319:336–9.
cell associated antigen CLL-1 aids in discrimination between normal [107] Kong Y, Yoshida S, Saito Y, et al. CD34+CD38+CD19+ as
and leukemic stem cells. Blood 2007;110:2659–66. well as CD34+CD38-CD19+ cells are leukemia-initiating cells
[87] Falini B, Nicoletti I, Martelli MF, Mecucci C. Acute myeloid leukemia with self-renewal capacity in human B-precursor ALL. Leukemia
carrying cytoplasmic/mutated nucleophosmin (NPMc+ AML): bio- 2008;22:1207–13.
logic and clinical features. Blood 2007;109:874–85. [108] Cox CV, Diamanti P, Evely RS, Kearns PR, Blair A. Expression
[88] Pearce DJ, Taussig D, Zibara K, et al. AML engraftment in of CD133 on leukemia-initiating cells in childhood ALL. Blood
the NOD/SCID assay reflects the outcome of AML: implica- 2009;113:3287–96.
tions for our understanding of the heterogeneity of AML. Blood [109] Nishida H, Yamazaki H, Yamada T, et al. CD9 correlates with cancer
2006;107:1166–73. stem cell potentials in human B-acute lymphoblastic leukemia cells.
[89] Eisterer W, Jiang X, Christ O, et al. Different subsets of pri- Biochem Biophys Res Commun 2009;382:57–62.
mary chronic myeloid leukemia stem cells engraft immunodeficient [110] Cox CV, Martin HM, Kearns PR, Virgo P, Evely RS, Blair A. Char-
mice and produce a model of the human disease. Leukemia acterization of a progenitor cell population in childhood T-cell acute
2005;19:435–41. lymphoblastic leukemia. Blood 2007;109:674–82.
[90] Dorsey JF, Cunnick JM, Lanehart R, et al. Interleukin-3 protects [111] Yamazaki H, Nishida H, Iwata S, Dang NH, Morimoto C. CD90
Bcr-Abl-transformed hematopoietic progenitor cells from apop- and CD110 correlate with cancer stem cell potentials in human T-

Please cite this article in press as: Schulenburg A, et al. Neoplastic stem cells: Current concepts and clinical perspectives. Crit Rev
Oncol/Hematol (2010), doi:10.1016/j.critrevonc.2010.01.001
ONCH-1372; No. of Pages 20
ARTICLE IN PRESS
A. Schulenburg et al. / Critical Reviews in Oncology/Hematology xxx (2010) xxx–xxx 17

acute lymphoblastic leukemia cells. Biochem Biophys Res Commun [134] Lee CJ, Dosch J, Simeone DM. Pancreatic cancer stem cells. J Clin
2009;383:172–7. Oncol 2008;26:2806–12.
[112] Armstrong F, de la Grange PB, Gerby B, et al. NOTCH is a key [135] Schmidt C. Lapatinib study supports cancer stem cell hypothe-
regulator of human T-cell acute leukemia initiating cell activity. Blood sis, encourages industry research. J Natl Cancer Inst 2008;100:
2009;113:1730–40. 694–5.
[113] De Keersmaecker K, Marynen P, Cools J. Genetic insights in the [136] O’Brien CS, Howell SJ, Farnie G, Clarke RB. Resistance to endocrine
pathogenesis of T-cell acute lymphoblastic leukemia. Haematologica therapy: are breast cancer stem cells the culprits? J Mammary Gland
2005;90:1116–27. Biol Neoplasia 2009;14:45–54.
[114] Radtke F, Wilson A, Mancini SJ, MacDonald HR. Notch regulation of [137] Ushijima T, Okochi-Takada E. Aberrant methylations in cancer cells:
lymphocyte development and function. Nat Immunol 2004;5:247–53. where do they come from? Cancer Sci 2005;96:206–11.
[115] Grabher C, von Boehmer H, Look AT. Notch 1 activation in the molec- [138] Watts CK, Handel ML, King RJ, Sutherland RL. Oestrogen receptor
ular pathogenesis of T-cell acute lymphoblastic leukaemia. Nat Rev gene structure and function in breast cancer. J Steroid Biochem Mol
Cancer 2006;6:347–59. Biol 1992;41:529–36.
[116] Weng AP, Ferrando AA, Lee W, et al. Activating mutations of [139] Ehtesham M, Mapara KY, Stevenson CB, Thompson RC. CXCR4
NOTCH1 in human T cell acute lymphoblastic leukemia. Science mediates the proliferation of glioblastoma progenitor cells. Cancer
2004;306:269–71. Lett 2009;274:305–12.
[117] Durig J, Ebeling P, Grabellus F, et al. A novel nonobese diabetic/severe [140] Read TA, Fogarty MP, Markant SL, et al. Identification of CD15 as a
combined immunodeficient xenograft model for chronic lymphocytic marker for tumor-propagating cells in a mouse model of medulloblas-
leukemia reflects important clinical characteristics of the disease. toma. Cancer Cell 2009;15:135–47.
Cancer Res 2007;67:8653–61. [141] Kurita T, Medina RT, Mills AA, Cunha GR. Role of p63 and basal
[118] Nowakowski. ABCG2 is expressed in a small population of cir- cells in the prostate. Development 2004;131:4955–64.
culating B-cells characterized by expression of self-renewal and [142] Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ. Prospective
early B-cell development genes and resistance to therapy. Blood identification of tumorigenic prostate cancer stem cells. Cancer Res
2008;112(11):1–1444, 1082. 2005;65:10946–51.
[119] Matsui W, Huff CA, Wang Q, et al. Characterization of clonogenic [143] Maitland NJ, Collins AT. Prostate cancer stem cells: a new target for
multiple myeloma cells. Blood 2004;103:2332–6. therapy. J Clin Oncol 2008;26:2862–70.
[120] Rasmussen T, Lodahl M, Hancke S, Johnsen HE. In multiple [144] Kasper S. Exploring the origins of the normal prostate and prostate
myeloma clonotypic CD38−/CD19+/CD27+ memory B cells recir- cancer stem cell. Stem Cell Rev 2008;4:193–201.
culate through bone marrow, peripheral blood and lymph nodes. Leuk [145] Gu G, Yuan J, Wills M, Kasper S. Prostate cancer cells with stem cell
Lymphoma 2004;45:1413–7. characteristics reconstitute the original human tumor in vivo. Cancer
[121] Matsui W, Wang Q, Barber JP, et al. Clonogenic multiple myeloma Res 2007;67:4807–15.
progenitors, stem cell properties, and drug resistance. Cancer Res [146] Klarmann GJ, Hurt EM, Mathews LA, et al. Invasive prostate cancer
2008;68:190–7. cells are tumor initiating cells that have a stem cell-like genomic
[122] Prince ME, Sivanandan R, Kaczorowski A, et al. Identification signature. Clin Exp Metast 2009;26:433–46.
of a subpopulation of cells with cancer stem cell properties in [147] Patrawala L, Calhoun T, Schneider-Broussard R, et al. Highly puri-
head and neck squamous cell carcinoma. Proc Natl Acad Sci USA fied CD44+ prostate cancer cells from xenograft human tumors are
2007;104:973–8. enriched in tumorigenic and metastatic progenitor cells. Oncogene
[123] Zhou L, Wei X, Cheng L, Tian J, Jiang JJ. CD133, one of the 2006;25:1696–708.
markers of cancer stem cells in Hep-2 cell line. Laryngoscope [148] Dubrovska A, Kim S, Salamone RJ, et al. The role of PTEN/Akt/PI3K
2007;117:455–60. signaling in the maintenance and viability of prostate cancer stem-
[124] van de Wetering M, Sancho E, Verweij C, et al. The beta-catenin/TCF- like cell populations. Proc Natl Acad Sci USA 2009;106:268–
4 complex imposes a crypt progenitor phenotype on colorectal cancer 73.
cells. Cell 2002;111:241–50. [149] Sharifi N, Hurt EM, Farrar WL. Androgen receptor expression in
[125] Fearon ER, Vogelstein B. A genetic model for colorectal tumorigen- prostate cancer stem cells: is there a conundrum? Cancer Chemother
esis. Cell 1990;61:759–67. Pharmacol 2008;62:921–3.
[126] Knudson Jr AG. Hereditary cancer, oncogenes, and antioncogenes. [150] True L, Coleman I, Hawley S, et al. A molecular correlate to the
Cancer Res 1985;45:1437–43. Gleason grading system for prostate adenocarcinoma. Proc Natl Acad
[127] Sell S, Pierce GB. Maturation arrest of stem cell differentiation is Sci USA 2006;103:10991–6.
a common pathway for the cellular origin of teratocarcinomas and [151] Suvà ML, Riggi N, Stehle JC, et al. Identification of cancer stem cells
epithelial cancers. Lab Invest 1994;70:6–22. in Ewing’s sarcoma. Cancer Res 2009;69:1776–81.
[128] Sell S, Leffert HL. Liver cancer stem cells. J Clin Oncol [152] Takaishi S, Okumura T, Tu S, et al. Identification of gastric can-
2008;26:2800–5. cer stem cells using the cell surface marker CD44. Stem Cells
[129] Factor VM, Radaeva SA, Thorgeirsson SS. Origin and fate of oval 2009;27:1006–20.
cells in dipin-induced hepatocarcinogenesis in the mouse. Am J Pathol [153] Houghton J, Stoicov C, Nomura S, et al. Gastric cancer originating
1994;145:409–22. from bone marrow-derived cells. Science 2004;306:1568–71.
[130] Petersen BE, Goff JP, Greenberger JS, Michalopoulos GK. Hepatic [154] Kim CF, Jackson EL, Woolfenden AE, et al. Identification of
oval cells express the hematopoietic stem cell marker Thy-1 in the bronchioalveolar stem cells in normal lung and lung cancer. Cell
rat. Hepatology 1998;27:433–45. 2005;121:823–35.
[131] Ma S, Lee TK, Zheng BJ, Chan KW, Guan XY. CD133+ HCC cancer [155] Eramo A, Lotti F, Sette G, et al. Identification and expansion of
stem cells confer chemoresistance by preferential expression of the the tumorigenic lung cancer stem cell population. Cell Death Differ
Akt/PKB survival pathway. Oncogene 2008;27:1749–58. 2008;15:504–14.
[132] Yang ZF, Ngai P, Ho DW, et al. Identification of local and circulating [156] Tirino V, Camerlingo R, Franco R, et al. The role of CD133 in the
cancer stem cells in human liver cancer. Hepatology 2008;47:919– identification and characterisation of tumour-initiating cells in non-
28. small-cell lung cancer. Eur J Cardiothorac Surg 2009;36:446–53.
[133] Berman DM, Karhadkar SS, Maitra A, et al. Widespread requirement [157] Santin AD. Prospective identification and characterization of ovarian
for Hedgehog ligand stimulation in growth of digestive tract tumours. cancer stem cells: implications for the treatment of chemotherapy
Nature 2003;425:846–51. resistant/recurrent ovarian disease. Cell Cycle 2009:8.

Please cite this article in press as: Schulenburg A, et al. Neoplastic stem cells: Current concepts and clinical perspectives. Crit Rev
Oncol/Hematol (2010), doi:10.1016/j.critrevonc.2010.01.001
ONCH-1372; No. of Pages 20
ARTICLE IN PRESS
18 A. Schulenburg et al. / Critical Reviews in Oncology/Hematology xxx (2010) xxx–xxx

[158] Baba T, Convery PA, Matsumura N, et al. Epigenetic regulation of myelogenous leukemia: role of malignant stromal macrophages.
CD133 and tumorigenicity of CD133+ ovarian cancer cells. Oncogene Blood 1995;85:3636–45.
2009;28:209–18. [182] Wohrer S, Rabitsch W, Shehata M, et al. Mesenchymal stem cells in
[159] Ferrandina G, Bonanno G, Pierelli L, et al. Expression of CD133-1 and patients with chronic myelogenous leukaemia or bi-phenotypic Ph+
CD133-2 in ovarian cancer. Int J Gynecol Cancer 2008;18:506–14. acute leukaemia are not related to the leukaemic clone. Anticancer
[160] Kusumbe AP, Mali AM, Bapat SA. CD133-expressing stem cells Res 2007;27:3837–41.
associated with ovarian metastases establish an endothelial hierarchy [183] Jootar S, Pornprasertsud N, Petvises S, et al. Bone marrow derived
and contribute to tumor vasculature. Stem Cells 2009;27:498–508. mesenchymal stem cells from chronic myeloid leukemia t(9;22)
[161] Friedman S, Lu M, Schultz A, Thomas D, Lin RY. CD133+ anaplastic patients are devoid of Philadelphia chromosome and support cord
thyroid cancer cells initiate tumors in immunodeficient mice and are blood stem cell expansion. Leuk Res 2006;30:1493–8.
regulated by thyrotropin. PLoS ONE 2009;4:e5395. [184] Valent P. Emerging stem cell concepts for imatinib-resistant chronic
[162] Monzani E, Facchetti F, Galmozzi E, et al. Melanoma contains CD133 myeloid leukaemia: implications for the biology, management, and
and ABCG2 positive cells with enhanced tumourigenic potential. Eur therapy of the disease. Br J Haematol 2008;142:361–78.
J Cancer 2007;43:935–46. [185] Valent P, Deininger M. Clinical perspectives of concepts on neoplastic
[163] Rappa G, Fodstad O, Lorico A. The stem cell-associated antigen stem cells and stem cell-resistance in chronic myeloid leukemia. Leuk
CD133 (Prominin-1) is a molecular therapeutic target for metastatic Lymphoma 2008;49:604–9.
melanoma. Stem Cells 2008;26:3008–17. [186] Clarke MF. Chronic myelogenous leukemia—identifying the hydra’s
[164] Dou J, Pan M, Wen P, et al. Isolation and identification of cancer heads. N Engl J Med 2004;351:634–6.
stem-like cells from murine melanoma cell lines. Cell Mol Immunol [187] Frankel A, Liu JS, Rizzieri D, Hogge D. Phase I clinical study of
2007;4:467–72. diphtheria toxin-interleukin 3 fusion protein in patients with acute
[165] Jaksch M, Munera J, Bajpai R, Terskikh A, Oshima RG. Cell myeloid leukemia and myelodysplasia. Leuk Lymphoma 2008;49:
cycle-dependent variation of a CD133 epitope in human embry- 543–53.
onic stem cell, colon cancer, and melanoma cell lines. Cancer Res [188] Jin L, Lee EM, Ramshaw HS, et al. Monoclonal antibody-mediated
2008;68:7882–6. targeting of CD123, IL-3 receptor alpha chain, eliminates human acute
[166] Keshet GI, Goldstein I, Itzhaki O, et al. MDR1 expression identifies myeloid leukemic stem cells. Cell Stem Cell 2009;5:31–42.
human melanoma stem cells. Biochem Biophys Research Commun [189] Roberts AW, He S, Bradstock KF, et al. A phase 1 and correla-
2008;368:930–6. tive biological study of CSL360(anti-CD123 mAb) in AML. Blood
[167] Hendrix MJ, Seftor EA, Hess AR, Seftor RE. Molecular plasticity of 2008;112:1015–6.
human melanoma cells. Oncogene 2003;22:3070–5. [190] Sharma SV, Bell DW, Settleman J, Haber DA. Epidermal growth factor
[168] Selzer E, Wacheck V, Kodym R, et al. Erythropoietin receptor expres- receptor mutations in lung cancer. Nat Rev Cancer 2007;7:169–81.
sion in human melanoma cells. Melanoma Res 2000;10:421–6. [191] Giovannucci E. Insulin, insulin-like growth factors and colon cancer:
[169] Mirmohammadsadegh A, Marini A, Gustrau A, Delia D, et al. Role of a review of the evidence. J Nutr 2001;131:3109S–20S.
erythropoietin receptor expression in malignant melanoma. J Invest [192] Wakefield LM, Roberts AB. TGF-beta signaling: positive and nega-
Dermatol 2010;130:201–10. tive effects on tumorigenesis. Curr Opin Genet Dev 2002;12:22–9.
[170] Kumar SM, Acs G, Fang D, Herlyn M, Elder DE, Xu X. Func- [193] Korc M, Friesel RE. The role of fibroblast growth factors in tumor
tional erythropoietin autocrine loop in melanoma. Am J Pathol growth. Curr Cancer Drug Targets 2009;9:639–51.
2005;166:823–30. [194] Xu Q, Simpson SE, Scialla TJ, Bagg A, Carroll M. Survival of
[171] Kumar SM, Yu H, Fong D, Acs G, Xu X. Erythropoietin activates the acute myeloid leukemia cells requires PI3 kinase activation. Blood
phosphoinositide 3-kinase/Akt pathway in human melanoma cells. 2003;102:972–80.
Melanoma Res 2006;16:275–83. [195] Recher C, Dos Santos C, Demur C, Payrastre B. mTOR, a new ther-
[172] Tas F, Oguz H, Argon A, et al. The value of serum levels of IL-6, TNF- apeutic target in acute myeloid leukemia. Cell Cycle (Georgetown,
alpha, and erythropoietin in metastatic malignant melanoma: serum Tex) 2005;4:1540–9.
IL-6 level is a valuable prognostic factor at least as serum LDH in [196] Böhm A, Aichberger KJ, Mayerhofer M, et al. Targeting of mTOR is
advanced melanoma. Med Oncol 2005;22:241–6. associated with decreased growth and decreased VEGF expression in
[173] Nordling CO. A new theory on cancer-inducing mechanism. Br J acute myeloid leukaemia cells. Eur J Clin Invest 2009;39:395–405.
Cancer 1953;7:68–72. [197] Weisberg E, Banerji L, Wright RD, et al. Potentiation of
[174] Armitage P, Doll R. The age distribution of cancer and a multi-stage antileukemic therapies by the dual PI3K/PDK-1 inhibitor, BAG956:
theory of carcinogenesis. Br J Cancer 1954;8:1–12. effects on BCR-ABL- and mutant FLT3-expressing cells. Blood
[175] Knudson Jr AG. Mutation and cancer: statistical study of retinoblas- 2008;111:3723–34.
toma. Proc Natl Acad Sci USA 1971;68:820–3. [198] Ratain MJ, Eisen T, Stadler WM, et al. Phase II placebo-controlled
[176] Cairns J. Mutation selection and the natural history of cancer. Nature randomized discontinuation trial of sorafenib in patients with
1975;255:197–200. metastatic renal cell carcinoma. J Clin Oncol 2006;24:2505–12.
[177] Gorre ME, Mohammed M, Ellwood K, et al. Clinical resistance to [199] Escudier B, Eisen T, Stadler WM, et al. Sorafenib in advanced clear-
STI-571 cancer therapy caused by BCR-ABL gene mutation or ampli- cell renal-cell carcinoma. N Engl J Med 2007;356:125–34.
fication. Science 2001;293:876–80. [200] Motzer RJ, Michaelson MD, Redman BG, et al. Activity of SU11248,
[178] Shah NP, Nicoll JM, Nagar B, et al. Multiple BCR-ABL kinase a multitargeted inhibitor of vascular endothelial growth factor receptor
domain mutations confer polyclonal resistance to the tyrosine kinase and platelet-derived growth factor receptor, in patients with metastatic
inhibitor imatinib (STI571) in chronic phase and blast crisis chronic renal cell carcinoma. J Clin Oncol 2006;24:16–24.
myeloid leukemia. Cancer Cell 2002;2:117–25. [201] Llovet JM, Ricci S, Mazzaferro V, et al. Sorafenib in advanced hepa-
[179] Turley EA, Veiseh M, Radisky DC, Bissell MJ. Mechanisms of dis- tocellular carcinoma. N Engl J Med 2008;359:378–90.
ease: epithelial-mesenchymal transition—does cellular plasticity fuel [202] Cheng AL, Kang YK, Chen Z, et al. Efficacy and safety of sorafenib
neoplastic progression? Nat Clin Pract Oncol 2008;5:280–90. in patients in the Asia-Pacific region with advanced hepatocellular
[180] Streubel B, Chott A, Huber D, et al. Lymphoma-specific genetic aber- carcinoma: a phase III randomised, double-blind, placebo-controlled
rations in microvascular endothelial cells in B-cell lymphomas. N trial. Lancet Oncol 2009;10:25–34.
Engl J Med 2004;351:250–9. [203] Cunningham D, Humblet Y, Siena S, et al. Cetuximab monother-
[181] Bhatia R, McGlave PB, Dewald GW, Blazar BR, Verfaillie CM. apy and cetuximab plus irinotecan in irinotecan-refractory metastatic
Abnormal function of the bone marrow microenvironment in chronic colorectal cancer. N Engl J Med 2004;351:337–45.

Please cite this article in press as: Schulenburg A, et al. Neoplastic stem cells: Current concepts and clinical perspectives. Crit Rev
Oncol/Hematol (2010), doi:10.1016/j.critrevonc.2010.01.001
ONCH-1372; No. of Pages 20
ARTICLE IN PRESS
A. Schulenburg et al. / Critical Reviews in Oncology/Hematology xxx (2010) xxx–xxx 19

[204] Rosell R, Robinet G, Szczesna A, et al. Randomized phase II Biographies


study of cetuximab plus cisplatin/vinorelbine compared with cis-
platin/vinorelbine alone as first-line therapy in EGFR-expressing
advanced non-small-cell lung cancer. Ann Oncol 2008;19:362–9. Axel Schulenburg graduated at and is now working at the
[205] Mohi MG, Boulton C, Gu TL, et al. Combination of rapamycin Medical University of Vienna, Bone Marrow Transplantation
and protein tyrosine kinase (PTK) inhibitors for the treatment of Unit. His research focus is Cancer stem cells.
leukemias caused by oncogenic PTKs. Proc Natl Acad Sci USA
2004;101:3130–5. Kira Brämswig studied medicine in Würzburg and Vienna
[206] Noh WC, Mondesire WH, Peng J, et al. Determinants of rapamycin and has M.D. and Ph.D. degrees. She is now working
sensitivity in breast cancer cells. Clin Cancer Res 2004;10:1013– at the Oncology Department of the Medical University
23.
of Vienna. Scientifically she is focused on tumorangio-
[207] Chan S. Targeting the mammalian target of rapamycin (mTOR): a
new approach to treating cancer. Br J Cancer 2004;91:1420–4. genesis.
[208] Hartog H, Wesseling J. Boezen HM, van der Graaf WT, The insulin- Harald Herrmann obtained his M.D. in 2008 and is now
like growth factor 1 receptor in cancer: old focus, new future. Eur J
Cancer 2007;43:1895–904.
a Ph.D. student at the Medical University of Vienna. He is
[209] Weroha SJ, Haluska P. IGF-1 receptor inhibitors in clini- enrolled in the Ph.D.-program malignant diseases and focuses
cal trials—early lessons. J Mammary Gland Biol Neoplasia his research on stem cells in myeloid leukemias.
2008;13:471–83.
[210] Flandrin P, Guyotat D, Duval A, et al. Significance of heat-shock Heidrun Karlic studied biology and biochemistry at
protein (HSP) 90 expression in acute myeloid leukemia cells. Cell the University of Vienna with a Ph.D. graduation and
Stress Chaperones 2008;13:357–64. is since July 1987 senior scientist at the Ludwig Boltz-
[211] Thomas X, Campos L, Mounier C, et al. Expression of heat-shock mann Institute for Leukemia Research and Hematology,
proteins is associated with major adverse prognostic factors in acute
myeloid leukemia. Leukemia Res 2005;29:1049–58.
Hanusch Hospital, Vienna Austria. Her research focus is
[212] Thomas X, Campos L, Le QH, Guyotat D. Heat shock pro- epigenetics.
teins and acute leukemias. Hematology (Amsterdam, Netherlands)
2005;10:225–35.
Irina Mirkina studied molecular biology at the Russian
[213] Mayerhofer M, Gleixner KV, Mayerhofer J, et al. Targeting of heat Academy of Sciences (RAS), Moscow, Russia from 1996
shock protein 32 (Hsp32)/heme oxygenase-1 (HO-1) in leukemic cells to 1999 with a Ph.D. She is now working on melanoma
in chronic myeloid leukemia: a novel approach to overcome resistance stem cells as a postdoctoral scientist in the laboratory of the
against imatinib. Blood 2008;111:2200–10. Medical University of Vienna.
[214] Gleixner KV, Mayerhofer M, Vales A, et al. Targeting of Hsp32 in
solid tumors and leukemias: a novel approach to optimize anticancer Rainer Hubmann studied molecular biology at the Uni-
therapy. Curr Cancer Drug Targets 2009;9:675–89. versity of Vienna and holds a Ph.D. He is now a postdoc
[215] Al-Hajj M, Clarke MF. Self-renewal and solid tumor stem cells. Onco-
gene 2004;23:7274–82.
at the Medical University of Vienna and is working on the
[216] Graziano A, d’Aquino R, Tirino V, Desiderio V, Rossi A, Pirozzi G. regulation and function of NOTCH2 in B-CLL.
The stem cell hypothesis in head and neck cancer. J Cell Biochem
2008;103:408–12.
Sylvia Laffer studied biology at the University of Vienna
[217] Chaudry MA, Sales K, Ruf P, Lindhofer H, Winslet MC. EpCAM from 1986 to 1992 and has a Ph.D. She is now working on
an immunotherapeutic target for gastrointestinal malignancy: current cancer stem cells for the Ludwig Boltzmann Cluster Oncol-
experience and future challenges. Br J Cancer 2007;96:1013–9. ogy.
[218] Rege TA, Hagood JS. Thy-1 as a regulator of cell-cell and cell-matrix
interactions in axon regeneration, apoptosis, adhesion, migration, can- Brigitte Marian studied pharmacy from 1972 to 1977 at
cer, and fibrosis. FASEB J 2006;20:1045–54. the University of Vienna and has a M.Sc. and a Ph.D. in
[219] Moretti S, Lanza F, Dabusti M, et al. CD123 (interleukin 3 receptor science. She was a research fellow at the Memorial Sloan
alpha chain). J Biol Regul Homeost Agents 2001;15:98–100.
Kettering Cancer Center, New York, USA and is now an
[220] Tarrant JM, Robb L, van Spriel AB, Wright MD. Tetraspanins:
molecular organisers of the leukocyte surface. Trends Immunol Asc. Prof. at the Institute of Cancer Research at the Med-
2003;24:610–7. ical University Vienna. Her research is focused on Colon
[221] Pescovitz MD. Rituximab, an anti-cd20 monoclonal antibody: history Cancer.
and mechanism of action. Am J Transplant 2006;6:859–66.
[222] Donnenberg VS, Donnenberg AD. Multiple drug resistance in can- Medhat Shehata is a postdoctoral scientist at the Medical
cer revisited: the cancer stem cell hypothesis. J Clin Pharmacol University of Vienna and is involved in research of Chronic
2005;45:872–7. lymphocytic leukemia.
[223] Frank NY, Pendse SS, Lapchak PH, et al. Regulation of progenitor
cell fusion by ABCB5 P-glycoprotein, a novel human ATP-binding Clemens Krepler obtained his M.D. in 2000 and is now
cassette transporter. J Biol Chem 2003;278:47156–65. a specialist degree in dermatology and venereology at the
[224] Frank NY, Margaryan A, Huang Y, et al. ABCB5-mediated doxoru- Medical University of Vienna. He is enrolled in the Ph.D.-
bicin transport and chemoresistance in human malignant melanoma.
program malignant diseases and focuses his research on stem
Cancer Res 2005;65:4320–33.
[225] Bakker AB, van den Oudenrijn S, Bakker AQ, et al. C-type lectin-like cells in melanoma.
molecule-1: a novel myeloid cell surface marker associated with acute Hubert Pehamberger is full professor and head of the Der-
myeloid leukemia. Cancer Res 2004;64:8443–50.
[226] Maiese K, Li F, Chong ZZ. New avenues of exploration for erythro-
matology Department of the Medical University of Vienna.
poietin. JAMA 2005;293:90–5. His research focus is melanoma.

Please cite this article in press as: Schulenburg A, et al. Neoplastic stem cells: Current concepts and clinical perspectives. Crit Rev
Oncol/Hematol (2010), doi:10.1016/j.critrevonc.2010.01.001
ONCH-1372; No. of Pages 20
ARTICLE IN PRESS
20 A. Schulenburg et al. / Critical Reviews in Oncology/Hematology xxx (2010) xxx–xxx

Thomas Grunt studied biology from 1976 to 1982 at Christoph Zielinski is full professor and head of the Oncol-
the University of Salzburg. He has Ph.D. and master ogy Department at the Medical University of Vienna.
degrees. He is now professor at the Oncology Laboratory in
Peter Valent is Asc. Professor for experimental hematol-
Vienna.
ogy at the Medical University of Vienna. He is working in
Ulrich Jäger is full professor and head of the Hematology the research of stem cells in hematologic malignancies and
Department of the Medical University of Vienna. His research mast cells.
interests are lymphatic malignancies.

Please cite this article in press as: Schulenburg A, et al. Neoplastic stem cells: Current concepts and clinical perspectives. Crit Rev
Oncol/Hematol (2010), doi:10.1016/j.critrevonc.2010.01.001

Anda mungkin juga menyukai