Cancer stem cells in solid tumors Laurie E Ailles1 and Irving L Weissman2

Page created by Nelson Hammond
 
CONTINUE READING
Cancer stem cells in solid tumors Laurie E Ailles1 and Irving L Weissman2
Cancer stem cells in solid tumors
Laurie E Ailles1 and Irving L Weissman2

Cancer stem cells (CSCs) are cells that drive tumorigenesis, as         several important biological properties of CSCs: first,
well as giving rise to a large population of differentiated progeny     what is the cell of origin for a given tumor? Second, what
that make up the bulk of the tumor, but that lack tumorigenic           are the signaling pathways that drive self renewal and/or
potential. CSCs have been identified in a variety of human              differentiation of CSCs? Third, are there molecules
tumors, as assayed by their ability to initiate tumor growth in         uniquely expressed on CSCs, regardless of whether they
immunocompromised mice. Further characterization studies                are functional, that will allow targeted therapies to be
have demonstrated that gene expression profiles in breast               developed? Fourth, what are the mechanisms by which
cancer correlate with patient prognosis, and brain CSCs are             CSCs escape conventional therapies and can we defeat
specifically resistant to radiation through DNA damage repair.          these mechanisms? Answers to these questions should
In addition, specific signaling pathways play a functional role in      lead to the development of therapies that target the CSC
CSC self renewal and/or differentiation, and early studies              population and eliminate the ‘engine’ that drives tumors
indicate that CSCs are associated with a microenvironmental             to grow, invade, and seed metastatic lesions. The chal-
niche. Thus the biological properties of CSCs are just beginning        lenges involved in identifying a CSC population from a
to be revealed, and the continuation of these studies should            solid tumor, and recent successes in this area are
lead to the development of CSC-targeted therapies for cancer            described in this review, as well as early studies that
treatment.                                                              represent the first steps toward understanding the bio-
Addresses                                                               logical properties of these cells.
1
  Institute for Stem Cell Biology and Regenerative Medicine, Stanford
University School of Medicine, 1050 Arastradero Road, Palo Alto, CA     Identification of markers for the prospective
94304, United States
2
  Department of Pathology and Institute for Stem Cell Biology and
                                                                        isolation of cancer stem cells
Regenerative Medicine, Stanford University School of Medicine, B257     A recent AACR workshop stated that ‘cancer stem cells can
Beckman Center, Stanford, CA 94305, United States                       . . . only be defined experimentally by their ability to
                                                                        recapitulate the generation of a continuously growing
Corresponding author: Ailles, Laurie E (lailles@stanford.edu) and
                                                                        tumor’ [1]. The most widely accepted assay to validate a
Weissman, Irving L (irv@stanford.edu)
                                                                        candidate CSC population is therefore tumor initiation and
                                                                        serial transplantation in immunocompromised mice, where
  Current Opinion in Biotechnology 2007, 18:460–466                     the tumor that grows in the mice recapitulates the hetero-
  This review comes from a themed issue on
                                                                        geneity of the primary patient tumor. This was first
  Tissue and cell engineering                                           achieved for human acute myeloid leukemia (AML).
  Edited by Alan Trounson and Andrew Elefanty                           The availability of cell surface markers allowing the iso-
                                                                        lation of cells at distinct stages of hematopoietic devel-
                                                                        opment, including hematopoietic stem cells (HSCs) [2,3],
  0958-1669/$ – see front matter                                        combined with specific and quantitative assays, greatly
  Published by Elsevier Ltd.                                            facilitated the identification of AML stem cells. In this
                                                                        case, it was found that a population of cells with the
  DOI 10.1016/j.copbio.2007.10.007
                                                                        phenotype CD34+CD38 [4] and CD90 [5], that
                                                                        represented 0.1–1% of the total cells, were the only cells
                                                                        able to initiate AML that resembled the human disease in
Introduction                                                            NOD/SCID mice. Adding in vitro assays led to the finding
The cancer stem cell (CSC) model of tumor development                   that the CSCs were in the blood lineage panel marker
and progression states that tumors, like normal adult                   negative fraction [Lin ], while true CD34+38-90+lin
tissues, contain a subset of cells that both self renew                 HSCs contains the AML1/ETO translocation in nonleu-
and give rise to differentiated progeny. As in other tissues,           kemic, or preleukemic, HSC [6]. The CD34+38-90-
the stem cells are a minority of the whole organ, and are               CD45RA Lin population has recently been identified
the only cells that can maintain tumor growth indefinitely.             in normal human hematopoietic tissues as a multipotent
The remaining cells, though actively proliferating and                  progenitor, but not a HSC [7]. Thus, the phenotype of the
making up the majority of the cells in the tumor, are also              AML stem cell indicates that they probably originate in the
differentiating and destined to die. The self renewal                   hematopoietic stem/progenitor cell compartment, possibly
properties of the CSCs are thus the real driving force                  from the multipotent progenitor. It has been demonstrated
behind tumor growth. The identification of markers that                 in CML that has progressed to blast crisis, the progression
allow the prospective isolation of CSCs from whole tumor                step is linked to the activation of a self renewal program in
tissues will allow us to develop an understanding of                    the granulocyte-macrophage progenitor population, thus

Current Opinion in Biotechnology 2007, 18:460–466                                                               www.sciencedirect.com
Cancer stem cells in solid tumors Ailles and Weissman 461

conferring cells that normally cannot self renew with CSC       Table 1
properties [8]. Thus while CSCs may derive from normal          Cell surface phenotype of CSCs identified in human cancers
stem cells, this may not be the case in all tumors. More
                                                                Tumor type                CSC phenotype            Reference
differentiated cells could acquire self renewal capacity
through multiple mutagenic events, leading them to              Breast                CD44+ CD24 /low              [9]
                                                                CNS                   CD133+                       [10]
become the CSC that is driving tumor growth.                    Multiple myeloma      CD138                        [11]
                                                                Melanoma              CD20+                        [22]
In order to identify CSCs, patient-derived tumor cells are      Prostate              CD44+ a2b1+ CD133+           [23,24]
stained with labeled antibodies to various cell surface         HNSCC                 CD44+                        [12]
                                                                Colon                 CD133+                       [15,16]
markers, either singly or in combination, and techniques
                                                                Colon                 CD44+ EpCam+ CD166+          [14]
such as magnetic columns or fluorescence-activated cell         Pancreatic            CD44+ EpCam+ CD24+           [13]
sorting (FACS) are used to separate labeled versus
unlabeled populations of cells. These populations are
then assayed for their ability to initiate tumor growth
in immunocompromised mice. Immunostaining and flow             was demonstrated that as few as 100 CD133+ cells from
cytometric analysis of the engrafted tumor must then           human brain tumors could initiate new tumors in the
show recapitulation of all of the original populations that    brains of immunocompromised mice, while 100 000
were in the primary tumor, and upon resorting of the CSC       CD133 cells did not contain any tumor-initiating
phenotype and retransplantation, a fully heterogeneous         activity [11]. In 2004 it was also shown that multiple
tumor must again be initiated. Ultimately, clonal mark-        myeloma contains a rare subset of cells, defined by their
ing, for example by retroviral insertion sites, and the        lack of expression of the plasma cell marker CD138, that
demonstration of multiple secondary tumors containing          are clonogenic in vitro and tumorigenic in vivo [12]. More
the same clone will be necessary to truly demonstrate self     recently, similar findings have been made for HNSCC
renewal. To date this has only been demonstrated in            [13], pancreatic cancer [14], and colon cancer [15–17].
AML [9].                                                       The cell surface phenotypes of CSC populations from
                                                               these human solid tumors are described in Table 1.
In the case of solid tumors, the challenge of identifying
and characterizing CSCs is significant. In most cases,         There have also been compelling studies in which
unlike the hematopoietic system, the normal tissue             putative CSCs have been identified based on their ability
developmental hierarchy has not been identified or             to form colonies in vitro. Sphere-forming ability as a
characterized. This makes the selection of candidate           measure of stem cells was first developed for central
markers more difficult, and provides no basis for com-         nervous system (CNS) cells, where it has been shown
parison between normal and malignant cells, or means to        that a subset of cells isolated from human fetal brain, and
identify the cell of origin. In vivo models of human solid     subsequently from human CNS tumors, can form spheres
tumors can also be challenging to achieve, as in some          when cultured under the appropriate conditions [11,18–
cases orthotopic injection is not feasible or is technically   22]. These spheres can self renew in vitro, and differen-
challenging (e.g. lung, colon, and bladder cancers). In        tiate into all of the neuronal lineages, both in vitro and in
these cases, subcutaneous injection or implantation under      vivo. Importantly, it was subsequently demonstrated for
the kidney capsule can be successful, but it is essential to   brain tumors that the neurosphere-forming cells could be
confirm that these xenografts recapitulate the histology of    prospectively isolated from fresh tissue using the cell
the patient tumors.                                            surface marker CD133, and that the CD133+ cells did
                                                               indeed initiate brain tumors in vivo, without any in vitro
Al-Hajj et al. [10] were the first to identify and prospec-    manipulation, indicating that they do in fact represent
tively isolate a minority subpopulation of cells from a        CSCs [11]. Fang et al. [23] have shown that upon culturing
human solid tumor (breast cancer) that contained all of        of metastatic melanoma cell suspensions under appropri-
the in vivo tumor-forming ability, while the remaining         ate conditions, a subset of cells could be propagated as
bulk of cells from these tumors had none, even when            nonadherent spheres, which could then be induced to
injected at many-fold higher cell doses. The tumorigenic       differentiate in vitro and to generate tumors in vivo.
cell population was identified based on its cell surface       However, a marker has not definitively been identified
phenotype, which was Lineage-CD44+CD24 /low. This              to allow the prospective isolation of melanoma sphere-
population could initiate tumors in immunocompromised          forming cells from fresh tumors, though the authors
mice with as few as 200 cells, while as many as 500 000 or     provided some evidence to suggest that CD20 can enrich
more of the remaining cells in the tumor did not initiate      for the multipotent fraction from established sphere
new tumors in mice. A similar finding was subsequently         cultures and melanoma cell lines. However, the ability
made in human brain tumors (GBMs and medulloblas-              to acutely isolate and assay subpopulations of cells from
tomas). In this case, it was already known that normal         tumors that behave as CSCs is essential before perform-
neuronal stem cells express the marker CD133 [19], and it      ing characterizations such as gene expression profiling, to

www.sciencedirect.com                                                        Current Opinion in Biotechnology 2007, 18:460–466
462 Tissue and cell engineering

avoid artifacts introduced by culturing cells for extended        cells that are unique and that may otherwise not have
periods of time. Another tumor for which CSCs have been           been revealed. They also found that CD24+ cells in one
potentially identified based on in vitro assays is prostate       patient sample had an additional genetic change not
cancer. Cells that are CD44+ a2b1+ CD133+ possess a               present in the CD44+CD24 cells. The authors state
significant capacity for self renewal, differentiation, and       that it has not been proven that the CD24+ cells are the
invasiveness in vitro [24]. In this case, it also remains to be   progeny of the CD44+CD24 cells; however this is not
seen whether these cells can reproduce the original tumor         the case. In the study by Al-Hajj et al. [10], it was shown
in vivo.                                                          that tumors grown from the CD44+CD24 tumorigenic
                                                                  population gave rise again to the original tumor hetero-
Understanding the biological properties of                        geneity, including the CD24+ cells. Thus the finding of
CSC                                                               genetic changes present in the CD24+ cells but not in the
Once CSCs have been identified, the real work begins,             CD44+CD24 population, but not vice versa, does not
which is the characterization of their molecular and              question the validity of the CSC hypothesis as Shipitsin
biological properties, in hopes of identifying ways to            et al. suggest. It is conceivable that additional genetic
specifically target and eradicate these cells in cancer           changes could take place in differentiating progenitors
patients. This is a new field in which we have barely             that are the progeny of the CSC, or that clonal evolution
begun to scratch the surface because of the only very             could occur within the CSC population itself as the tumor
recent ability to prospectively isolate CSCs from various         grows and the CSC population expands. Thus although
tumors. Some of the approaches towards gaining a deeper           the authors state that their data calls the CSC hypothesis
understanding of CSC biology that have been reported              into question, in our opinion their data very nicely sup-
are described below.                                              ports it.

Global gene expression profiling                                  Identification of self renewal signaling pathways in
One approach is to perform global gene expression profil-         CSCs
ing, and compare the CSC profile to that of the non-CSCs          In addition to gene expression profiling, CSCs have been
within the tumor, or to known profiles from the corre-            demonstrated to express known stem-cell-associated
sponding normal tissue. Such a study was performed for            genes by other methods, including qRT-PCR and immu-
breast cancer, in which the gene expression profile of the        nostaining. HNSCC CSCs differentially express BMI-1
CSC was compared to that of normal breast epithelium,             in the CD44+ population compared to the CD44 popu-
leading to the identification of 186 genes that were              lation, and pancreatic CSCs differentially express SONIC
differentially expressed in the CSC population. Upon              HEDGEHOG. Studies have yet to be done to determine
comparison with previously reported gene signatures in            whether these genes are functionally important in these
breast cancer, there was a significant association between        human tumor types, but their known roles in the self-
the CSC signature and both overall and metastasis-free            renewal of normal tissue stem cells and other CSC popu-
survival [25]. The finding that detectable expression of a        lations suggest that they may also be important in these
specific set of CSC-related genes upon analysis of whole          tumors. For example, in mice, BMI-1 is essential for the
tumor tissue correlates with prognosis suggests that more         self renewal of hematopoietic and neural stem cells
aggressive tumors may contain a relatively higher per-            [27,28] and has been shown to be required for the self-
centage of CSC, possibly caused by mutations that arrest          renewal of leukemic stem cells [29], and BMI-1 is over-
cells in an immature state of differentiation. Indeed,            expressed in human AML compared to normal bone
differentiation status correlates with prognosis in many          marrow. The Hedgehog pathway has long been impli-
cancers.                                                          cated in many different kinds of cancer [30] and more
                                                                  recently has been related specifically to human CSCs
A similar approach was taken by Shipitsin et al. [26], in         from multiple myeloma [31], breast cancer [32], and
which breast tumor cells with the CSC phenotype were              gliomas [33,34]. The Wnt signaling pathway has also
analyzed for their global gene expression profile com-            been implicated in both stem cell self renewal and cancer
pared to the nontumorigenic cells from the same tumors.           [35–37], and has been shown specifically to be active in
The profiles found indicated that cells of the CSC phe-           the CSC population of human CML blast crisis patients
notype expressed stem/progenitor-associated genes while           [8]. BMPs and their antagonists are known to play import-
the non-CSC expressed differentiation-associated genes.           ant roles in regulating homeostasis of various organs and
They also found a correlation of their gene expression            tissues via the control of differentiation, proliferation, and
signature with patient clinical outcome. Furthermore,             apoptosis [38,39]. They have also been implicated in
they identified a signaling pathway (TGF-b) specifically          several cancers [39,40]. Piccirillo et al. [41] recently
active in the CSC population, and found that inhibition of        demonstrated that BMPs, in particular BMP4, depleted
this pathway in vitro led to differentiation. These obser-        the brain tumor stem-cell population in vitro via a pro-
vations once again demonstrate that purification of CSC           differentiation effect, and inhibited tumorigenicity in
allows the identification of biological properties of those       vivo [41]. There are several other signaling pathways that

Current Opinion in Biotechnology 2007, 18:460–466                                                          www.sciencedirect.com
Cancer stem cells in solid tumors Ailles and Weissman 463

have known roles in maintenance and/or control of normal         state. Finally, increasing the number of endothelial cells
and CSC compartments in mice, as well as being impli-            or blood vessels in xenografts expanded the CSC popu-
cated in cancer, including the Notch, PTEN, and Hox              lation and accelerated their growth, while antiangiogenic
pathways [42,43], but to date no studies have been               therapies depleted the CSCs from the xenografts and
performed to demonstrate a possible functional role              arrested tumor growth. Thus the concept of the role of a
specifically in human CSC populations.                           CSC niche and the potential of inhibiting its interactions
                                                                 with CSC populations appears to be an approach with a
The discovery of signaling pathways that play a functional       great deal of therapeutic potential, both for disease treat-
role in CSC selfrenewal is extremely important from a            ment and for chemoprevention [50]. Furthermore, the
therapeutic perspective, as some of these pathways have          identification of the CSC niche should allow the devel-
known chemical inhibitors (e.g. the Hh pathway can be            opment of in vitro assay systems that mimic the in vivo
inhibited by cyclopamine treatment), or function as indu-        environment, providing more rapid results than the cur-
cers of differentiation (e.g. BMPs).                             rently cumbersome and time-consuming in vivo assays, as
                                                                 well as allowing manipulations that are currently not
Identification of a CSC niche                                    feasible with in vivo systems. Ultimately, an understand-
A well-known property of normal stem cells is their              ing of the CSC niche in addition to the CSCs themselves
dependence upon their microenvironment, or ‘niche’ to            for each different type of cancer should lead to a better
maintain their quiescent and undifferentiated state, while       understanding of the signals that are important for CSC
maintaining their proliferation and differentiation poten-       self renewal and/or differentiation in those cancers.
tials [44]. It is well known that a large component of solid
tumors is made up of nonepithelial stromal cells, which          Are CSCs selectively resistant to conventional
include inflammatory cells, vascular endothelial cells, and      therapies?
fibroblasts, and that in fact these stromal components are       Normal stem cells are known to be relatively quiescent,
essential for tumor growth, most probably through direct         resistant to drugs and toxins through the expression of
cell contacts and/or secreted factors [45,46]. In an effort to   drug efflux pumps, and have an active DNA-repair
better understand the role of the microenvironment in            capacity and resistance to apoptosis [51]. If CSCs share
tumor growth, Sneddon et al. [39] described a study in           many of the properties of normal stem cells, as is hypoth-
which they compared gene expression profiles of squa-            esized, this means that conventional chemo and radiation
mous cell carcinoma (SCC)-derived stromal cells versus           therapies, which target rapidly cycling cells, will lead to
stromal cells from nontumor skin and identified two BMP          reduction of the tumor through killing of the progeny of
antagonists (GREMLIN1 and FOLLISTATIN), that                     the CSCs, but the CSCs themselves will remain unaf-
were differentially expressed in the tumor-derived               fected. Furthermore, the expression of drug efflux pumps
stroma. Furthermore, BMP2 and BMP4 were expressed                and DNA-repair mechanisms will make them addition-
in SCC tumor cell nests, and GREMLIN1 was able to                ally resistant to these forms of therapy. Several studies
prevent inhibition of SCC growth by BMP4 in vitro.               have been done which support this hypothesis. In the
GREMLIN1 is expressed by stromal cells in diverse                hematopoietic system, both normal and leukemic stem
human carcinomas, but not in corresponding normal                cells are largely quiescent and express drug efflux mol-
tissues. This study suggests that the expression of factors      ecules such as ABCG2 [52,53], and AML stem cells are
by the tumor microenvironment that regulate self renewal         selectively resistant to both daunorubicin and Ara-C
may be a general feature of human cancer, and identifi-          [54,55]. More recently, it was shown that CD133+ brain
cation of such factors may lead to the identification of         tumor stem cells are selectively resistant to radiation, both
potential therapeutic targets. Prince et al. [13] demon-         in vitro and in vivo, and the mechanism for this resistance
strated that the CSC-containing population in well and           was shown to be through preferential activation of the
moderately differentiated HNSCC is physically located            DNA damage checkpoint response and an increase in
adjacent to stroma, further indicating that there may be         DNA-repair capacity [56]. By understanding the mech-
important interactions between the CSC and the stroma            anisms that allow CSCs to resist conventional therapies, it
in carcinomas.                                                   may be possible to find ways to manipulate them to
                                                                 become sensitive to these therapies.
Brain tumor stem cells also appear to interact with a niche.
Mouse neural stem cells have been shown to lie within a          Conclusions
vascular niche in which endothelial cells regulate stem          The CSC field, particularly in the area of solid tumors, is a
cell self renewal [47,48]. Therefore Calabrese et al. [49]       very young one. A model of solid tumor stem-cell biology
investigated whether the CD133+Nestin+ cells that con-           is shown in Figure 1, highlighting the properties that are
tain the CSC fraction are associated with vasculature and        currently viewed as important for tumor growth, and that
found that indeed they are. Furthermore, endothelial             may represent therapeutic targets. We are still in the very
cells supply secreted factors that maintain the brain tumor      technically challenging stage of identifying the methods
stem cells in vitro in a self renewing and undifferentiated      that will allow us to prospectively isolate CSCs from

www.sciencedirect.com                                                          Current Opinion in Biotechnology 2007, 18:460–466
464 Tissue and cell engineering

Figure 1

Model of cancer stem cells (CSCs) in solid tumors. CSCs are associated with the stromal components of the tumor, including fibroblasts and/or
blood vessels, which make up the CSC ‘niche’. The niche cells secrete factors that support CSC self renewal. CSCs retain differentiation
potential, giving rise to non-self renewing tumor cells that make up the bulk of the tumor. CSCs may be drug and/or radiation resistant, and may
express CSC-specific antigens. Current research focuses on identification of the molecular mechanisms regulating these properties of CSCs
(highlighted in green), as they represent potential targets for therapy.

various solid tumors, and most importantly, to demon-                      Conflict of interest
strate in vivo that the populations we are looking at are                  ILW has stock in Amgen, Inc., was a member of the Amgen scientific
                                                                           advisory board, and is a cofounder and member of the Board of Directors of
truly CSCs. In the cases where CSCs have been ident-                       Cellerant, Inc., and Stem Cells, Inc.
ified, early studies have already shown that they possess
important biological properties that directly relate to                    Acknowledgements
patient outcome. The breast CSC gene expression profile                    This work was supported in part by National Institutes of Health Grants
                                                                           R01CA086017 and R01CA086065 and an anonymous gift fund for Cancer
was shown to correlate with patient prognosis, and the                     Stem Cell research at Stanford University. In addition LEA is a recipient of
brain CSC has been shown to be specifically resistant to                   a Stanford Comprehensive Cancer Center Developmental Research Award.
radiation through DNA damage repair. In addition,
specific signaling pathways have been shown to play a                      References
functional role in CSC self renewal and/or differentiation,                1.   Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL,
and early studies indicate that CSCs are associated with a                      Visvader J, Weissman IL, Wahl GM: Cancer stem cells —
                                                                                perspectives on current status and future directions: AACR
microenvironmental niche. The mechanisms by which                               workshop on cancer stem cells. Cancer Res 2006, 66:9339-9344.
CSCs evade current therapies are being elucidated.                         2.   Baum CM, Weissman IL, Tsukamoto AS, Buckle AM, Peault B:
These types of studies should lead to a better under-                           Isolation of a candidate human hematopoietic stem-cell
standing of the molecular mechanisms controlling self-                          population. Proc Natl Acad Sci U S A 1992, 89:2804-2808.
renewal and/or differentiation. The advent of new high-                    3.   Uchida N, Sutton RE, Friera AM, He D, Reitsma MJ, Chang WC,
                                                                                Veres G, Scollay R, Weissman IL: HIV, but not murine leukemia
throughput technologies and the ability to apply them to                        virus, vectors mediate high efficiency gene transfer into
small numbers of cells should lead to the identification of                     freshly isolated G0/G1 human hematopoietic stem cells.
                                                                                Proc Natl Acad Sci U S A 1998, 95:11939-11944.
CSC-specific targets. The development of methods for
the prospective isolation of CSCs is thus the first step,                  4.   Bonnet D, Dick JE: Human acute myeloid leukemia is organized
                                                                                as a hierarchy that originates from a primitive hematopoietic
which then opens the door to a variety of approaches that                       cell. Nat Med 1997, 3:730-737.
could ultimately lead to CSC-specific therapies for cancer                 5.   Blair A, Hogge DE, Ailles LE, Lansdorp PM, Sutherland HJ: Lack of
treatment.                                                                      expression of Thy-1 (CD90) on acute myeloid leukemia cells

Current Opinion in Biotechnology 2007, 18:460–466                                                                            www.sciencedirect.com
Cancer stem cells in solid tumors Ailles and Weissman 465

     with long-term proliferative ability in vitro and in vivo. Blood   25. Liu R, Wang X, Chen GY, Dalerba P, Gurney A, Hoey T, Sherlock G,
     1997, 89:3104-3112.                                                    Lewicki J, Shedden K, Clarke MF: The prognostic role of a gene
                                                                            signature from tumorigenic breast-cancer cells. N Engl J Med
6.   Miyamoto T, Weissman IL, Akashi K: AML1/ETO-expressing                 2007, 356:217-226.
     nonleukemic stem cells in acute myelogenous leukemia with
     8;21 chromosomal translocation. Proc Natl Acad Sci U S A 2000,     26. Shipitsin M, Campbell LL, Argani P, Weremowicz S, Bloushtain-
     97:7521-7526.                                                          Qimron N, Yao J, Nikolskaya T, Serebryiskaya T, Beroukhim R,
                                                                            Hu M et al.: Molecular definition of breast tumor heterogeneity.
7.   Majeti R, Park CY, Weissman IL: Identification of a hierarchy of       Cancer Cell 2007, 11:259-273.
     multipotent hematopoietic progenitors in human cord blood.
     Cell Stem Cell 2007, in press.                                     27. Molofsky AV, Pardal R, Iwashita T, Park IK, Clarke MF,
                                                                            Morrison SJ: Bmi-1 dependence distinguishes neural stem cell
8.   Jamieson CH, Ailles LE, Dylla SJ, Muijtjens M, Jones C,                self-renewal from progenitor proliferation. Nature 2003,
     Zehnder JL, Gotlib J, Li K, Manz MG, Keating A et al.:                 425:962-967.
     Granulocyte-macrophage progenitors as candidate leukemic
     stem cells in blast-crisis CML. N Engl J Med 2004, 351:657-667.    28. Park IK, Qian D, Kiel M, Becker MW, Pihalja M, Weissman IL,
                                                                            Morrison SJ, Clarke MF: Bmi-1 is required for maintenance of
9.   Hope KJ, Jin L, Dick JE: Acute myeloid leukemia originates from        adult self-renewing haematopoietic stem cells. Nature 2003,
     a hierarchy of leukemic stem cell classes that differ in self-         423:302-305.
     renewal capacity. Nat Immunol 2004, 5:738-743.
                                                                        29. Lessard J, Sauvageau G: Bmi-1 determines the proliferative
10. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ,                   capacity of normal and leukaemic stem cells. Nature 2003,
    Clarke MF: Prospective identification of tumorigenic breast             423:255-260.
    cancer cells. Proc Natl Acad Sci U S A 2003, 100:3983-3988.
                                                                        30. Kasper M, Regl G, Frischauf AM, Aberger F: GLI transcription
11. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T,            factors: mediators of oncogenic Hedgehog signalling. Eur J
    Henkelman RM, Cusimano MD, Dirks PB: Identification of human            Cancer 2006, 42:437-445.
    brain tumour initiating cells. Nature 2004, 432:396-401.
                                                                        31. Peacock CD, Wang Q, Gesell GS, Corcoran-Schwartz IM, Jones E,
12. Matsui W, Huff CA, Wang Q, Malehorn MT, Barber J, Tanhehco Y,
                                                                            Kim J, Devereux WL, Rhodes JT, Huff CA, Beachy PA et al.:
    Smith BD, Civin CI, Jones RJ: Characterization of clonogenic
                                                                            Hedgehog signaling maintains a tumor stem cell compartment
    multiple myeloma cells. Blood 2004, 103:2332-2336.
                                                                            in multiple myeloma. Proc Natl Acad Sci U S A 2007, 104:4048-
13. Prince ME, Sivanandan R, Kaczorowski A, Wolf GT, Kaplan MJ,             4053.
    Dalerba P, Weissman IL, Clarke MF, Ailles LE: Identification of a
                                                                        32. Liu S, Dontu G, Mantle ID, Patel S, Ahn NS, Jackson KW, Suri P,
    subpopulation of cells with cancer stem cell properties in head
                                                                            Wicha MS: Hedgehog signaling and Bmi-1 regulate self-
    and neck squamous cell carcinoma. Proc Natl Acad Sci U S A
    2007, 104:973-978.                                                      renewal of normal and malignant human mammary stem cells.
                                                                            Cancer Res 2006, 66:6063-6071.
14. Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, Wicha M,
    Clarke MF, Simeone DM: Identification of pancreatic cancer          33. Bar EE, Chaudhry A, Lin A, Fan X, Schreck K, Matsui W,
    stem cells. Cancer Res 2007, 67:1030-1037.                              Vescovi AL, Piccirillo S, Dimeco F, Olivi A et al.: Cyclopamine-
                                                                            mediated hedgehog pathway inhibition depletes stem-like
15. Dalerba P, Dylla SJ, Park IK, Liu R, Wang X, Cho RW, Hoey T,            cancer cells in glioblastoma. Stem Cells 2007, 25:2524-2533.
    Gurney A, Huang EH, Simeone DM et al.: Phenotypic
    characterization of human colorectal cancer stem cells. Proc        34. Clement V, Sanchez P, de Tribolet N, Radovanovic I, Ruiz i
    Natl Acad Sci U S A 2007, 104:10158-10163.                              Altaba A: HEDGEHOG-GLI1 signaling regulates human glioma
                                                                            growth, cancer stem cell self-renewal, and tumorigenicity.
16. O’Brien CA, Pollett A, Gallinger S, Dick JE: A human colon cancer       Curr Biol 2007, 17:165-172.
    cell capable of initiating tumour growth in immunodeficient
    mice. Nature 2007, 445:106-110.                                     35. Reguart N, He B, Taron M, You L, Jablons DM, Rosell R: The role
                                                                            of Wnt signaling in cancer and stem cells. Future Oncol 2005,
17. Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M,           1:787-797.
    Peschle C, De Maria R: Identification and expansion of human
    colon-cancer-initiating cells. Nature 2007, 445:111-115.            36. Reya T, Duncan AW, Ailles L, Domen J, Scherer DC, Willert K,
                                                                            Hintz L, Nusse R, Weissman IL: A role for Wnt signalling in self-
18. Tamaki S, Eckert K, He D, Sutton R, Doshe M, Jain G, Tushinski R,       renewal of haematopoietic stem cells. Nature 2003, 423:409-
    Reitsma M, Harris B, Tsukamoto A et al.: Engraftment of sorted/         414.
    expanded human central nervous system stem cells from fetal
    brain. J Neurosci Res 2002, 69:976-986.                             37. Reya T, Morrison SJ, Clarke MF, Weissman IL: Stem cells,
                                                                            cancer, and cancer stem cells. Nature 2001, 414:105-111.
19. Uchida N, Buck DW, He D, Reitsma MJ, Masek M, Phan TV,
    Tsukamoto AS, Gage FH, Weissman IL: Direct isolation of human       38. Ye L, Lewis-Russell JM, Kyanaston HG, Jiang WG: Bone
    central nervous system stem cells. Proc Natl Acad Sci U S A             morphogenetic proteins and their receptor signaling in
    2000, 97:14720-14725.                                                   prostate cancer. Histol Histopathol 2007, 22:1129-1147.

20. Hemmati HD, Nakano I, Lazareff JA, Masterman-Smith M,               39. Sneddon JB, Zhen HH, Montgomery K, van de Rijn M, Tward AD,
    Geschwind DH, Bronner-Fraser M, Kornblum HI: Cancerous stem             West R, Gladstone H, Chang HY, Morganroth GS, Oro AE et al.:
    cells can arise from pediatric brain tumors. Proc Natl Acad Sci         Bone morphogenetic protein antagonist gremlin 1 is widely
    U S A 2003, 100:15178-15183.                                            expressed by cancer-associated stromal cells and can
                                                                            promote tumor cell proliferation. Proc Natl Acad Sci U S A 2006,
21. Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J,          103:14842-14847.
    Dirks PB: Identification of a cancer stem cell in human brain
    tumors. Cancer Res 2003, 63:5821-5828.                              40. Waite KA, Eng C: From developmental disorder to heritable
                                                                            cancer: it’s all in the BMP/TGF-beta family. Nat Rev Genet 2003,
22. Reynolds BA, Weiss S: Clonal and population analyses                    4:763-773.
    demonstrate that an EGF-responsive mammalian embryonic
    CNS precursor is a stem cell. Dev Biol 1996, 175:1-13.              41. Piccirillo SG, Reynolds BA, Zanetti N, Lamorte G, Binda E,
                                                                            Broggi G, Brem H, Olivi A, Dimeco F, Vescovi AL: Bone
23. Fang D, Nguyen TK, Leishear K, Finko R, Kulp AN, Hotz S, Van            morphogenetic proteins inhibit the tumorigenic potential of
    Belle PA, Xu X, Elder DE, Herlyn M: A tumorigenic subpopulation         human brain tumour-initiating cells. Nature 2006, 444:761-765.
    with stem cell properties in melanomas. Cancer Res 2005,
    65:9328-9337.                                                       42. Lobo NA, Shimono Y, Qian D, Clarke MF: The biology of cancer
                                                                            stem cells. Annu Rev Cell Dev Biol 2007, 23:675-699.
24. Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ:
    Prospective identification of tumorigenic prostate cancer           43. Rossi DJ, Weissman IL: Pten, tumorigenesis, and stem cell self-
    stem cells. Cancer Res 2005, 65:10946-10951.                            renewal. Cell 2006, 125:229-231.

www.sciencedirect.com                                                                   Current Opinion in Biotechnology 2007, 18:460–466
466 Tissue and cell engineering

44. Fuchs E, Tumbar T, Guasch G: Socializing with the neighbors:         52. Steinbach D, Legrand O: ABC transporters and drug resistance
    stem cells and their niche. Cell 2004, 116:769-778.                      in leukemia: was P-gp nothing but the first head of the hydra?
                                                                             Leukemia 2007, 21:1172-1176.
45. Kenny PA, Lee GY, Bissell MJ: Targeting the tumor
    microenvironment. Front Biosci 2007, 12:3468-3474.                   53. Zhou S, Schuetz JD, Bunting KD, Colapietro AM, Sampath J,
                                                                             Morris JJ, Lagutina I, Grosveld GC, Osawa M, Nakauchi H et al.:
46. Egeblad M, Littlepage LE, Werb Z: The fibroblastic                       The ABC transporter Bcrp1/ABCG2 is expressed in a wide
    coconspirator in cancer progression. Cold Spring Harb Symp               variety of stem cells and is a molecular determinant of the
    Quant Biol 2005, 70:383-388.                                             side-population phenotype. Nat Med 2001, 7:1028-1034.
47. Palmer TD, Willhoite AR, Gage FH: Vascular niche for adult           54. Costello RT, Mallet F, Gaugler B, Sainty D, Arnoulet C, Gastaut JA,
    hippocampal neurogenesis. J Comp Neurol 2000, 425:479-494.               Olive D: Human acute myeloid leukemia CD34+/CD38S
48. Shen Q, Goderie SK, Jin L, Karanth N, Sun Y, Abramova N,                 progenitor cells have decreased sensitivity to chemotherapy
    Vincent P, Pumiglia K, Temple S: Endothelial cells stimulate self-       and Fas-induced apoptosis, reduced immunogenicity, and
    renewal and expand neurogenesis of neural stem cells.                    impaired dendritic cell transformation capacities. Cancer Res
    Science 2004, 304:1338-1340.                                             2000, 60:4403-4411.

49. Calabrese C, Poppleton H, Kocak M, Hogg TL, Fuller C, Hamner B,      55. Guzman ML, Neering SJ, Upchurch D, Grimes B, Howard DS,
    Oh EY, Gaber MW, Finklestein D, Allen M et al.: A perivascular           Rizzieri DA, Luger SM, Jordan CT: Nuclear factor-kappaB is
    niche for brain tumor stem cells. Cancer Cell 2007, 11:69-82.            constitutively activated in primitive human acute
                                                                             myelogenous leukemia cells. Blood 2001, 98:2301-2307.
50. Albini A, Sporn MB: The tumour microenvironment as a target
    for chemoprevention. Nat Rev Cancer 2007, 7:139-147.                 56. Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB,
                                                                             Dewhirst MW, Bigner DD, Rich JN: Glioma stem cells promote
51. Dean M, Fojo T, Bates S: Tumour stem cells and drug                      radioresistance by preferential activation of the DNA damage
    resistance. Nat Rev Cancer 2005, 5:275-284.                              response. Nature 2006, 444:756-760.

Current Opinion in Biotechnology 2007, 18:460–466                                                                        www.sciencedirect.com
You can also read