Artigo 02 - cancer stem cells.pdf

Embed Size (px)

Citation preview

  • 8/15/2019 Artigo 02 - cancer stem cells.pdf

    1/8

    Die Hard: Are Cancer Stem Cells the Bruce Willises ofTumor Biology?

    Ákos Fábián,1 Márk Barok,1 György Vereb,1* János Szöllo†si1,2*

    AbstractIn recent years, an exponentially growing number of studies have focused on identify-ing cancer stem cells (CSC) in human malignancies. The rare CSCs could be crucial incontrolling and curing cancer: through asymmetric division CSCs supposedly drive tu-mor growth and evade therapy with the help of traits shared with normal stem cells

    such as quiescence, self-renewal ability, and multidrug resistance pump activity. Here,we give a brief overview of techniques used to confirm the stem cell-like behavior of pu-tative CSCs and discuss markers and methods for identifying, isolating, and culturingthem. We touch on the limitations of each marker and why the combined use of CSCmarkers, in vitro and in vivo assays may still fail to identify all relevant CSC popula-tions. Finally, the various experimental findings supporting and contradicting the CSChypothesis are summarized. The large number of tumor types thus far with a subpopu-lation of uniquely tumorigenic and therapy resistant cells suggests that despite theunanswered questions and inconsistencies, the CSC hypothesis has a legitimate role toplay in tumor biology. At the same time, experimental evidence supporting the estab-lished alternative theory of clonal evolution can be found as well. Therefore, a modelthat describes cancer initiation and progression should combine elements of clonal evo-lution and CSC theory.   ' 2008 International Society for Advancement of Cytometry 

    Key terms

    cancer stem cells; progenitor cells; stem cell markers; clonal evolution; therapy resist-ance

    THE  cancer stem cell hypothesis in its most accepted form states that tumorousgrowth is sustained by only a portion of the tumor cells. These cells uniquely possess

    the self-renewal and differentiation capabilities of normal tissue stem cells and are

    hence referred to as cancer stem cells (CSC) (1,2). According to the CSC hypothesis

    all cancer cells are derived from the multipotent CSCs. These, through asymmetric

    division give rise to progenitor cells, which are fast cycling and have limited self-

    renewal and differentiation capacity. Mirroring the hierarchical structure of normal

    tissues, the daughter cells of progenitors then differentiate to form the heterogeneous

    tumor mass. As CSCs would be the only cells attributed with long-term self-renewal

    potential, they are the ones supposedly responsible for the occurrence of distant me-

    tastases and also for recurrence of malignant disease after initial successful treatment.

    A further implication in therapy resistance is that CSCs are thought to possess the

    defense mechanisms of normal stem cells, such as multidrug resistance (MDR) pump

    activity (3) and by virtue of their stem cell nature are slow cycling, rendering them

    immune to traditional chemotherapy targeting fast cycling tumor cells (2).

    The cancer stem cell hypothesis witnessed a revival in the last 15 years [for a

    detailed timeline, see Ref. (4)], mostly fueled by the recognition that tumor cells are

    not equal with respect to their ability to regrow their tumors of origin when trans-

    planted into immune compromised mice. It was first shown for acute myeloid leuke-

    mia (AML) that significantly fewer malignant cells with a specific cell surface phenotype

    1Department of Biophysics and Cell

    Biology, Research Center for Molecular

    Medicine, Medical and Health Science

    Center, University of Debrecen, Hungary

    2Cell Biology and Signaling Research

    Group of the Hungarian Academy of

    Sciences, Research Center for

    Molecular Medicine, Medical and Health

    Science Center, University of Debrecen,

    Hungary

    Received 22 October 2008; Accepted 5

    November 2008

    Grant sponsor: Hungarian National

    Research Fund; Grant numbers: OTKA

    K68763, K62648, K75752; Grant sponsor:

    European Community; Grant numbers: EU

    FP6 LSHB-CT-2004-503467, EU FP6 LSHC-

    CT-2005-018914, EU FP6 MCRTN-CT-

    035946-2, EU FP6 MRTN-CT-2005-019481

    *Correspondence to: János Szöl lo†si or

    György Vereb, Department of

    Biophysics and Cell Biology, Faculty of

    Medicine, Medical and Health Science

    Center, University of Debrecen, P.O.Box

    39, Nagyerdei krt. 98, Debrecen H-4012,

    Hungary

    Email: [email protected] or [email protected]

    Published online 2 December 2008 in

    Wiley InterScience (www.interscience.

    wiley.com)

    DOI: 10.1002/cyto.a.20690

    © 2008 International Society forAdvancement of Cytometry

    Review Article

    Cytometry Part A    75A: 6774, 2009

  • 8/15/2019 Artigo 02 - cancer stem cells.pdf

    2/8

    are required for transferring leukemia to NOD/SCID (non-

    obese diabetic/severe combined immune deficient) mice than

    when injecting unsorted cells (5). Interestingly, the only sub-

    population of cells capable of initiating the disease in recipi-

    ents had a surface marker signature (CD3411CD382) closely 

    resembling that of normal hematopoietic stem cells (HSC)

    (5,6). Since ground breaking work with hematologic malig-

    nancies, putative cancer stem cells have been identified in a

    plethora of solid tumors, including breast (7), brain (8), lung

    (9), prostate (10), colon (11,12), liver (13,14), ovarian (15,16),

    head and neck (17), melanoma (18,19), and pancreas (20)

    cancers.

    Stem cells are found in most adult tissues (21–27) and

    might contribute to tissue repair as well as play a role in

    malignancies (28). The CSC concept originally stated that

    CSCs are mutated versions of these tissue-derived stem cells.

    Stem cells seemed a natural source of cancer initiating cells

    because they persist for a long time and thus are more likely to

    accumulate the genetic changes required for malignant trans-

    formation (1,29). The active self-renewal pathway also means

    that they require fewer such mutations than differentiated cells

    (1), which originally were believed to require at least six muta-

    tions to acquire all properties of cancer cells (30). However,

    later on it has been shown that retroviral insertion of just three

    genes is enough to convert human mammary epithelial cells

    (HMEC) to cancer cells (31) and four specific genetic modifi-

    cations are sufficient to reprogram diploid human fibroblasts

    and convert them to embryonic stem cells (32). Ectopic over-

    expression of Wnt-1 in HMECs results in oncogenic transfor-

    mation by eliciting a DNA damage response that leads to func-

    tional inactivation of p53 and loss of the G1/S checkpoint

    (33). Although a further mutation is required to increase

    Notch signaling, in this setting essentially one initial mutation

    is sufficient for a differentiated human cell to become a tu-

    mor-forming cancer cell. These findings further support that

    CSCs indeed need not be of stem cell origin. Instead, the CSC

    phenotype is most likely an ‘‘All roads lead to Rome’’-type

    endpoint of malignant transformation, where irrespective of 

    the cell of origin, certain common pathways have to be active/

    deregulated for self-renewal and tumor growth (Fig. 1).

    IDENTIFYING CSCSNo general protocol has been established to confirm pu-

    tative CSCs as such, but any study’s aim is to prove the main

    Figure 1.   The role of CSCs in tumor biology. The CSC phenotype could be a common phenotype acquired by normal tissue stem, progeni-tor or differentiated cells through mutations, which activate/deregulate certain signaling pathways, with the key changes depending onthe cell of origin (1–3). The CSC then fosters progeny that follow an aberrant differentiation pathway. As the cancer progresses any cell

    may undergo clonal evolution and thus, influence tumor behavior and potentially mask or disrupt the underlying hierarchical organizationof the cancer cells.

    REVIEW ARTICLE

    68   Cancer Stem Cells 

  • 8/15/2019 Artigo 02 - cancer stem cells.pdf

    3/8

    identifying criteria of CSCs—self-renewal and the capacity to

    give rise to the heterogeneous lineages that comprise the origi-

    nal tumor (2). CSCs should also have a phenotype that is con-

    sistently unique to the self-renewing fraction of cells and

    allows separation from the rest of the tumor cells.

    In vitro assays for confirming self-renewal include serial

    colony forming unit (CFU) assays and propagation as tumor-

    spheres (34) in stem cell culturing conditions (Table 1) (2). In

    the case of culturing tumorspheres, verification is needed that

    sphere formation is a result of clonal growth (35) and not sec-ondary aggregation of cells (36). Injection of spheres into

    immune compromised mice can confirm whether the cultured

    cells also possess the tumor-forming potential required of 

    CSCs. As stem cell culturing conditions might influence the

    biological behavior of cells (37), the cells corresponding to the

    surface marker phenotype of sphere-forming cells ideally need

    to be isolated from the original tumors and injected without

    prior culturing. Differentiation capacity of spheres can be

    examined by culturing in differentiating medium and obser-

    ving morphological and expression pattern changes, most

    notably loss of CSC markers and ‘‘stemness’’-associated gene

    expression patterns (23). Asymmetric division can be con-

    firmed by culturing separated CSCs and identifying non-CSC

    phenotype cells among the progeny. In vitro proliferation and

    invasion assays also provide valuable details about the proper-

    ties of investigated CSC and non-CSC populations, but on

    their own do not predict in vivo behavior (38).

    Self-renewal, tumor propagation, and multilineage differ-

    entiation can be demonstrated in vivo by xenotransplantation

    into immune compromised mice. The putative CSC popula-

    tion, the only cell type capable of propagating the tumor

    according to CSC theory, gives rise to tumors from fewer cells

    than bulk tumor or non-CSC populations. Self-renewal allows

    the tumors to be serially transplanted (also known as in vivo

    passaging), thus CSCs isolated from secondary tumors are

    able to form tumors when injected into mice. As a conse-

    quence of multilineage differentiation, the original histological

    phenotype of the parent tumor is maintained and recreated in

    all tumors grown from CSCs during serial transplantation.

    Special care must be taken to assure before inoculation that

    differences in tumor-forming capabilities of CSC and non-

    CSC populations are not due to differences in cell cycle, viabil-

    ity after cell separation or fraction of tumor cells relative to all

    injected cells. Verifying viability of cells after injection canprove that non-CSCs engraft and survive in the mouse envir-

    onment, but are unable to form tumors on their own (8,12).

    WHAT TO LOOK FORThe list of potential markers of CSCs is long (Table 2).

    Thus far, CD133 (3,8,10–15,39–43), CD44 (10,16,17,44,45),

    CD24 (in combination with CD44) (7,20,35,38), efflux of 

    Hoechst or Rhodamine dyes [also referred to as Side Popula-

    tion (SP)] (3,46–49), CD90 (3,50), CD117 (3,16), CD34 (5,6),

    CD20 (18), and aldehyde dehydrogenase (ALDH) (51,52) have

    all been used to identify putative CSCs in one or multiple tu-

    mor types [a comprehensive overview of normal tissue stem

    cell and CSC markers can be found in Ref. (53)]. However,

    these markers have certain limitations; most notably they fail

    to identify all CSCs (marker negative cells can also have

    tumorigenic and clonogenic properties), and merely designate

    a subpopulation that is enriched for clonogenic and tumori-

    genic activity (54). Also, not all cells with a CSC marker phe-

    notype behave as CSCs. Most markers for separating CSCs

    were chosen due to their expression on normal stem cells of 

    certain tissues. A recent study in a mouse model of  H. pylori -

    induced gastric tumors found that infection of gastric mucosa

    induced migration of nonresident, bone marrow derived cells

    to the gastric epithelium (55). The neoplastic lesions that

    Table 1.   Culturing conditions for tumorsphere formation

    TUMOR TYPE MEDIUM SUPPLEMENTS CULTURED IN REF.

    Breast Serum-free DMEM-12 bFGF, EGF, insulin, BSA 96-well culture dish (35)

    Breast RPMI 1640 Glutamine, 10% FCS Low-binding plates (61)

    Colon DMEM-F12 EGF, FGF-2, glucose, glutamine, insulin,

    progesterone, putrescin, sodiumselenite, apotransferrin, Hepes, heparin,

    BSA

    NA (42)

    Colon DMEM 10% Fetal bovine serum 24 well plate (60)

    Colon Serum-free medium EGF, FGF-2 NA (11)

    Pancreatic NS-A basal serum-free

    medium

    EGF, FGF-2, glucose glutamine, insulin,

    progesterone, putrescin, sodium

    selenite, transferin

    Hanging drops (40)

    Melanoma Mouse embryonic fibroblast

    Conditioned human

    embryonic stem cell medium

    bFGF Noncoated flasks (18)

    Ovarian Serum-free DMEM-F12 bFGF, EGF, insulin, BSA Ultra low attachment plates (16)

    bFGF, basic fibroblast growth factor; EGF, epidermal growth factor; BSA, bovine serum albumin; FCS, fetal calf serum; NA, notavailable.

    REVIEW ARTICLE

    Cytometry Part A    75A: 6774, 2009   69

  • 8/15/2019 Artigo 02 - cancer stem cells.pdf

    4/8

    formed were of the same origin. This indicates that the origi-

    nating cell of a malignancy must not necessarily be from the

    site of formation and therefore might have a different expres-

    sion profile from the resident stem cells.Another question that remains to be answered is how reli-

    ably the CSCs are defined by these markers. For instance, in an

    experiment in which mammospheres were cultured from

    mouse mammary gland cells, cells from fresh tissue dissocia-

    tions that could form spheres were CD241, but 6 days of 

    preculturing led to mammosphere formation from only the

    CD242 subpopulation (36). This indicates that the cell popula-

    tion exhibiting stem cell properties may well depend on the

    conditions under which the tumors grow (56,57) and the envi-

    ronmental influences they encounter during separation and

    culture. As obtaining single cell suspensions from solid tumors

    often involves mechanical and enzymatic disaggregation lasting

    hours, the possibility of altering surface marker expressionprofiles has to be taken into consideration. In our own trials,

    incubation with digesting enzymes (Collagenase I, Dispase)

    substantially downregulated the expression of CD44 even after

    1 h (unpublished observation). Therefore, immunohistochem-

    istry or fluorescent labeling of both the original tumor tissue

    and the isolated cells is necessary to confirm that surface

    expression patterns are not an artifact of cancer cell isolation.

    The unreliability of the side population to identify CSCs

    has been discussed extensively, with the conclusions that nei-

    ther do all tumors or cell lines have a consistently identifiable

    SP (48) nor are all CSCs necessarily within the SP when there

    is one (2,4). Also, the potential cytotoxicity of retained

    Hoechst inside the cell is of concern. The marker CD133 has

    been used in CSC experiments to identify CSCs in brain, liver,

    colon, prostate, and ovarian cancer. Recent papers have shown

    that expression of CD133 can be epigenetically regulated

    (41,58) and that CD133 expression may be associated with cell

    cycle phase (59), although this was previously not seen (15).

    Another group found that contrary to previous experiments,

    where CD1331 cells were a rare, CSC-enriched population in

    colon cancer, basically all colon cancer cells from primary 

    tumors they studied were CD1331 (60). In the same study,

    only a portion of cells from liver metastasis were CD1331, but

    both CD1331 and CD1332 cells were capable of tumorsphere

    formation in vitro and tumor initiation in vivo. Most interest-

    ingly, CD1332 cells had a CD441CD242 phenotype, whereas

    CD1331 cells presented a CD44low CD241 phenotype. Upon

    investigating primary glioblastomas, Beier et al. demonstratedthat in roughly 25% of tumors the CSC fraction was in the

    CD1332 population (43). These cells were capable of asym-

    metric division, sphere formation and had similar in vivo

    tumorigenicity as the CD1331 CSCs from the other 75%

    of examined tumors. In another study of cell lines derived

    from BRCA1 deficient breast tumors in mice, CD1331 and

    CD441CD242 phenotypes were two non-overlapping popula-

    tions that were both enriched for CSCs (61). These experi-

    ments suggest that depending on the tumor of origin the

    CSCs might be within different phenotypic subpopulations

    and that more of these subpopulations can coexist.

    CD44 is unique in the list of markers, because it has been

    shown to play an active role in tumorigenesis and xenograftformation (62). In experiments conducted with hepatocellular

    carcinoma (HCC) cells, putative CD901 HCC CSCs were also

    CD441 (50). Treatment of these cells with an anti-CD44 anti-

    body induced apoptosis in a dose-dependent manner. In a

    strain of intestinal tumor prone mice, CD44 knockouts had a

    reduced incidence rate of adenoma, probably by regulating the

    amount of DNA damage that the cell attempts to repair before

    initiating apoptosis (63). Finally, in a mouse model of chronic

    myeloid leukemia (CML), BCR-ABL-1 positive progenitors

    required CD44 for efficient bone marrow homing (although

    B-cell acute lymphocytic leukemia (B-ALL) initiating cells

    with the same mutation did not) (64).

    The isolation of CSCs can prove a challenge, even when

    working with verified, stably expressed CSC markers. Because

    of the relatively small percentage of CSCs in the tumor, large

    numbers of bulk cells have to be investigated to acquire

    enough CSCs for an experiment. At the same time non-CSC

    populations have to be of high purity, since even a small per-

    centage of contaminating highly tumorigenic CSCs can dra-

    matically influence the results of xenotransplantation experi-

    ments. The diversity of investigated malignancies and markers

    means that for each sample and source tissue, the appropriate

    isolation, labeling and gating strategy has to be optimized

    individually and must incorporate the use of proper positive

    Table 2.   Commonly used markers of CSCs

    CSC MARKER ASSOCIATED TUMOR TYPE(S)

    CD133 Brain (8,39,43), colon (11,12,42), liver (13,14), lung (3), ovarian (15,41), pancreatic (40), prostate (10)

    CD44 Colorectal (44,45), head and neck squamous cell carcinoma (17), liver (50), ovarian (16), prostate (10)

    CD24a (with CD44

    coexpression)

    Breast (7,35,38), pancreatic (20)

    Side population Brain (48), breast (48), lung (3), ovarian (46) prostate (48), thyroid (47)

    CD90 Breast (3), liver (50), lung (3)

    CD34 AML (5,6), lung (9)

    CD117 Lung (3), ovarian (16)

    CD20 Melanoma (18)

    ALDH Breast (52), liver (51)

    a In breast tumors CD242, whereas in pancreatic tumors CD241 phenotype is associated with CSCs.

    REVIEW ARTICLE

    70   Cancer Stem Cells 

  • 8/15/2019 Artigo 02 - cancer stem cells.pdf

    5/8

    and negative controls (25,65). For examples of a prudent

    approach please refer to figures 1 and 2 of Ref. (25) in this

    issue.

    INCONSISTENCIES OF THE CSC HYPOTHESISXenotransplantation models, in particular those used for

    CSC experiments have received a lot of criticism. Most of 

    these revolve around the fact that the mouse is a foreign envir-

    onment for human cancer cells and therefore, the full tumori-

    genetic potential that would be seen with stromal elements

    and cytokine signaling more closely resembling human tissues

    cannot be revealed. Indeed, in several studies involving con-

    genic transplantation into mice, the required number of tu-

    mor cells drastically drops as compared to xenografting

    (61,66) and transplantability of the malignancy is no longer

    confined to a subset of cells (66). The notion that the mouse

    environment is somehow selecting which cells can propagate

    is further reinforced by findings that Matrigel, co-injected nor-

    mal stromal cells or irradiated cancer cells (feeder cells) all

    reduce the number of tumor cells needed for reproducible tu-mor growth (31,67). These findings have prompted the use of 

    a more careful nomenclature, in which the CSCs are referred

    to as tumor initiating cells (TIC). This highlights their most

    prominent biological feature and at the same time acknowl-

    edges tumor initiation may be more characteristic of the host-

    graft interaction than the cancer cells themselves.

    Another phenomenon the CSC hypothesis has trouble

    explaining is why so many bulk cells are needed for tumor for-

    mation. More specifically, if a nonselected tumor cell popula-

    tion is implanted, the CSC content of the minimum cell dose

    for tumor growth is usually 10 times that required when the

    same CSCs are used after separation from the bulk. One expla-

    nation for this was that somehow the non-CSC population isnegatively regulating the growth of the CSCs (12,68). It is,

    however, unclear how injecting more cells overcomes this inhi-

    bition, as the relative proportion of CSCs and non-CSCs does

    not change with the number of cells injected and an increase

    of non-CSCs in the finite volume of a mouse should result in

    higher concentrations of hypothetical inhibitory substances.

    Along another line of thought, Hill proposed that the small

    number of CSCs injected may not provoke an immune

    response (56), whereas intermediate numbers from bulk tu-

    mor would lead to a tumor rejecting immune response and

    high numbers of bulk tumor cells could overwhelm the

    immune system and form tumors. This scenario fails to

    explain how the growing tumors from CSCs evade rejection

    (the extracellular matrix synthesized by the tumor cells may 

    play a role (69)) and why co-injection of feeder cells (essen-

    tially increasing immunogenicity without increasing the num-

    ber of cells capable of proliferation) reduces the number of tu-

    mor cells required. It is also contradicted by findings of 

    engrafted, but not proliferating non-CSC tumor cells at the

    injection site (8,12).

    The enhancing effects of Matrigel and co-injected feeder

    cells on tumor growth in the mouse model indicate that can-

    cer cells may be lacking some extracellular signals required for

    effective proliferation. Hill has proposed that cancer cells capa-

    ble of tumor formation after xenotransplantation may possess

    an autocrine growth loop (54). A recent study showed that in

    colon cancer signaling through IL-4 has a significant role in

    tumor growth (42). The ability of CSCs to resist cell death is

    mediated by IL-4 and IL-4 is produced by the colon cancer

    cells. Breast, thyroid, and lung cancers also produce IL-4 (70).

    Another study with melanomas suggested that CSCs might

    drive the proliferation of non-CSCs (19). Calculations by Kern

    and Shibata imply that some, if not most of the tumorigenic

    potential of a tumor must lie in the non-CSC fraction (68).

    The discrepancies in the number of cells needed for tumor

    growth can be explained if we suppose that non-CSCs have

    tumorigenic potential as well, however, they have a higher

    threshold for tumor initiating signaling and/or lack effective

    autocrine signaling. This higher threshold becomes more

    accentuated in mice where normal environmental cues are

    sparse and therefore they are dependent on signaling from

    other sources. Tumors from bulk cells then can only form

    when a critical level of stimulation is provided by the auto/

    paracrine signaling of the co-injected CSCs. The non-CSCs atthe same time might essentially act in the way a regular stem

    cell niche would and negatively regulate the proliferation of 

    CSCs.

    Serial transplantations have revealed significant genetic

    instability in tumors originating from CSCs (39,71,72). One

    clear manifestation of this phenomenon is that tumors

    become more aggressive with in vivo passaging, with earlier

    tumor presentation and faster tumor growth rates

    (11,40,71,72). As the number of injected CSCs does not

    change, the CSCs re-injected during serial transplantation

    must gain additional traits, which the original CSCs from the

    primary tumors did not have. The logical conclusion is that

    CSCs themselves are not genetically stable over time, whichallows them to evolve and adapt to the mouse environment.

    RETHINKING THE CONCEPT OF CSCSDespite the obvious shortcomings of the CSC hypothesis

    (Table 3), several lines of experimental evidence suggest that

    CSCs might have a crucial role in tumor biology. Several stu-

    dies identified cancers that can be clonally initiated and sus-

    tained (44,73,74). Disruption of a single signaling pathway has

    resulted in malignant transformation (73,75) and was shown

    to change the behavior of progenitors and stem cells (73,76).

    Asymmetric division of CSCs also explains why cells with cap-

    abilities of the putative CSC population do not become the

    dominant cell type in tumors, although it can be argued that

    CSCs only have a selectional advantage over other tumor cells

    under very harsh conditions, such as chemotherapy or xeno-

    translpantation.

    The methods used to identify putative CSCs are by no

    means perfect. However, tumor subpopulations that have rela-

    tively low stromal dependence when compared with other tu-

    mor cells were readily identified and exhibited more efficient

    tumor-forming in mice. They are more therapy resistant in

    vitro (16,41,42,61,77) and in vivo (78), effectively increasing

    their relative numbers after cytoreductive therapy (40,42,44).

    Why CSCs have an intrinsic therapy resistance even in pre-

    REVIEW ARTICLE

    Cytometry Part A    75A: 6774, 2009   71

  • 8/15/2019 Artigo 02 - cancer stem cells.pdf

    6/8

    viously untreated tumors is hard to explain with clonal evolu-

    tion, since no prior selective pressure was applied to produce

    this phenotype. Although clonal evolution would allow this

    phenotype to occur as a byproduct of constant mutation, it is

    unclear why it would be so consistently found as a small frac-

    tion of so many tumors.

    When designing therapy, it should always be considered

    that one aspect of therapy resistance of CSCs is their quiescent

    nature. Selective targeting of putative CSCs to reduce tumor

    growth and resistance to chemotherapy has proved promising.Antibody targeting of ABCB5 on melanoma cells significantly 

    reduced tumor growth and tumor formation rate (19). Phar-

    macological inhibition of CXCR4 in pancreatic cancer signifi-

    cantly reduced tumor metastasis in xenografted mice (40). Yil-

    maz et al induced leukemia in mice by deletion of PTEN (73).

    Administration of rapamycin—an inhibitor of mTOR in the

    PI3K downstream pathway (which is normally negatively 

    regulated by PTEN)—blocked leukemogenesis, and prolonged

    survival of mice with established leukemia. Rapamycin also

    restored long-term self-renewal capability of hematopoietic

    stem cells with the PTEN deletion. Both in vitro and in vivo

    chemotherapy resistance of colon CSCs was reduced by anti-

    IL-4 antibody treatment (42). Bauerschmitz et al. identified

    tumor specific promoters in CD441CD242 breast tumor cells

    (79). By targeting oncolytic viruses to these promoters they 

    achieved significant in vitro and in vivo killing of the putative

    breast CSCs and reduced the size of xenograft tumors. Though

    these targeting strategies still require refinement, they also

    show that specific knowledge on the particular molecular mar-

    kers and/or signaling profile of a given person’s cancer stem

    cells may aid in the design of individualized and effective ther-

    apy. The specific targeting of CSCs in this setting could help

    eradicate a cancer cell subpopulation capable of evading tradi-

    tional therapy and so increase disease free survival.

    The theories proposed among others by Campbell and

    Polyak (80) and Adams and Strasser (81) have tried to unify 

    the competing models of clonal evolution and CSCs. Accord-

    ing to these, evidence can be found for both models and their

    prevalence is probably unique to every tumor and may actu-

    ally change as the tumor progresses. Although CSCs seem to

    be a special subset of tumor cells, newer studies show that

    CSCs in themselves are still a heterogeneous population with

    different biological properties and that multiple populations

    with CSC characteristics can coexist in the same tumor. Maet al. investigated HCC cell lines and were able to separate sub-

    populations with different tumorigenic potential based on

    CD133 and ALDH expression (51), which however contradicts

    the original CSC hypothesis of just one population with tu-

    mor-forming capabilities. Herman et al found that

    CD1331CXCR42 and CD1331CXCR41 pancreatic cancer

    cells do not differ in tumorigenicity, but only the

    CD1331CXCR41 population migrates and metastasizes (40).

    Work with AML found leukemia-inducing cells to be hetero-

    geneous in self-renewal potential (74). In lung cancers, several

    subpopulations with different profiles of CSC marker expres-

    sion can be identified (3). The CSC hypothesis explains this

    heterogeneity with the existence of cancer progenitor cells,

    which still possess some residual stem cell traits. It has been

    proposed that tumor grade depends on the relative proportion

    of progenitors within the tumor. Accordingly, dedifferentia-

    tion and aggressiveness may reflect expansion or increased

    self-renewal of the fast cycling progenitor population (3).

    Whether the heterogeneity of the CSC population is

    caused by clonal evolution or partial differentiation of the can-

    cer initiating cell is probably more a question of faith at this

    time, than an evidence-backed scientific decision. To further

    obscure the picture, recent findings indicate that clonal diver-

    sity is beneficial for tumor progression (82). It is also becom-

    Table 3.   Evidence supporting and contradicting the CSC hypothesis

    EXPERIMENTAL EVIDENCE

    Supporting the CSC hypothesis Contradicting the CSC hypothesis

    Large number of cells are needed for xenotransplantation

    of tumors

    In congenic transplantations substantially fewer cells are needed

    The required number of putative CSCs for

    xenotransplantation of tumors is relatively small

    Xenograft tumors can be serially transplanted, but only 

    with the CSC subpopulation

    Transplantability of malignancies is not restricted to one

    subpopulation in congenic transplantations

    Non-CSC populations do not initiate tumor growth in

    vivo, or require more cells than the CSC population to

    do so

    A small fraction of tumors cells are capable of sustained

    growth under stem cell culturing conditions

    CSC markers do not identify a pure CSC population

    CSCs have higher clonogenicity in vitro The CSC population is heterogeneous in itself, with differences in

    metastatic and tumorigenic potential

    Cultured CSCs can give rise to progeny with non-CSC

    phenotypesCSCs have intrinsic in vitro and in vivo therapy resistance

    REVIEW ARTICLE

    72   Cancer Stem Cells 

  • 8/15/2019 Artigo 02 - cancer stem cells.pdf

    7/8

    ing clear that several properties, which we thought were

    intrinsic to CSCs are modulated by the microenvironment of 

    the cancer cells (83), and such key traits as metastasizing (84)

    and growth (85) may depend on the normal stromal cells that

    interact with the cancer cells (86).

    CONCLUSIONSThe CSC hypothesis has come a long way since its first

    inception. Putative CSCs have been identified in many solid

    tumors. Though the methods to identify CSCs have their

    uncertainties, the isolated CSCs are a consistently tumorigenic

    and therapy-resistant subpopulation of tumor cells. Studies

    have proved again and again that putative CSCs are capable of 

    self-renewal and multilineage differentiation. Selective target-

    ing of CSCs seems to be an effective strategy to reduce therapy 

    resistance and growth of xenograft tumors. However, the CSC

    hypothesis faces new challenges. Recent findings indicate that

    CSCs themselves may be a heterogeneous population, with

    differences in tumor-forming and metastasizing capabilities.

    Coexistence of multiple CSC populations with different phe-notypes can also not be ruled out. Data suggest that CSCs may 

    not be stable over time and could undergo clonal evolution as

    well. The stromal environment and CSC niche has a verified

    crucial role in the behavior of cancer cells in some cases. The

    concerns about the relevance of mouse xenotransplant experi-

    ments to human cancers, as well as substantial evidence for

    clonal evolution should also not be overlooked. We have to

    accept that neither the CSC hypothesis nor clonal evolution

    can explain all experimental evidence by itself. By acknowled-

    ging the respective strengths and weaknesses of both, a com-

    bined model can be constructed in which tumor behavior and

    progression can be dictated by CSC biology and modulated or

    dominated by clonal evolution and effects of the microenvir-onment. The great challenge of the years to come will be how 

    we determine the contribution of each model to tumor growth

    in a given patient and how we can use that information to

    design more effective and hopefully curative therapies.

    LITERATURE CITED1. Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem

    cells. Nature 2001;414:105–111.

    2. Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL, Visvader J, Weiss-man IL, Wahl GM. Cancer stem cells—Perspectives on current status and futuredirections: AACR Workshop on cancer stem cells. Cancer Res 2006;66:9339–9344.

    3. Donnenberg VS, Landreneau RJ, Donnenberg AD. Tumorigenic stem and progenitorcells: Implications for the therapeutic index of anti-cancer agents. J Control Release2007;122:385–391.

    4. Ma S, Chan KW, Guan XY. In search of liver cancer stem cells. Stem Cell Rev 

    2008;4:179–192.5. Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, Minden M,

    Paterson B, Caligiuri MA, Dick JE. A cell initiating human acute myeloid leukaemiaafter transplantation into SCID mice. Nature 1994;367:645–648.

    6. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy thatoriginates from a primitive hematopoietic cell. Nat Med 1997;3:730–737.

    7. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospectiveidentification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA2003;100:3983–3988.

    8. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusi-mano MD, Dirks PB. Identification of human brain tumour initiating cells. Nature2004;432:396–401.

    9. Kim CF, Jackson EL, Woolfenden AE, Lawrence S, Babar I, Vogel S, Crowley D, Bron-son RT, Jacks T. Identification of bronchioalveolar stem cells in normal lung and lungcancer. Cell 2005;121:823–835.

    10. Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ. Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res 2005;65:10946–10951.

    11. Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, De MariaR. Identification and expansion of human colon-cancer-initiating cells. Nature2007;445:111–115.

    12. O’Brien CA, Pollett A, Gallinger S, Dick JE. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 2007;445:106–110.

    13. Ma S, Chan KW, Hu L, Lee TK, Wo JY, Ng IO, Zheng BJ, Guan XY. Identification andcharacterization of tumorigenic liver cancer stem/progenitor cells. Gastroenterology 2007;132:2542–2556.

    14. Yin S, Li J, Hu C, Chen X, Yao M, Yan M, Jiang G, Ge C, Xie H, Wan D, Yang S,Zheng S, Gu J. CD133 positive hepatocellular carcinoma cells possess high capacity for tumorigenicity. Int J Cancer 2007;120:1444–1450.

    15. Ferrandina G, Bonanno G, Pierelli L, Perillo A, Procoli A, Mariotti A, Corallo M,Martinelli E, Rutella S, Paglia A, Zannoni G, Mancuso S, Scambia G. Expression of CD133-1 and CD133-2 in ovarian cancer. Int J Gynecol Cancer 2008;18:506–514.

    16. Zhang S, Balch C, Chan MW, Lai HC, Matei D, Schilder JM, Yan PS, Huang TH,Nephew KP. Identification and characterization of ovarian cancer-initiating cellsfrom primary human tumors. Cancer Res 2008;68:4311–4320.

    17. Prince ME, Sivanandan R, Kaczorowski A, Wolf GT, Kaplan MJ, Dalerba P, WeissmanIL, Clarke MF, Ailles LE. Identification of a subpopulation of cells with cancer stemcell properties in head and neck squamous cell carcinoma. Proc Natl Acad Sci USA2007;104:973–978.

    18. Fang D, Nguyen TK, Leishear K, Finko R, Kulp AN, Hotz S, Van Belle PA, Xu X, ElderDE, Herlyn M. A tumorigenic subpopulation with stem cell properties in melanomas.Cancer Res 2005;65:9328–9337.

    19. Schatton T, Murphy GF, Frank NY, Yamaura K, Waaga-Gasser AM, Gasser M, ZhanQ, Jordan S, Duncan LM, Weishaupt C, Fuhlbrigge RC, Kupper TS, Sayegh MH,Frank MH. Identification of cells initiating human melanomas. Nature 2008;451:345–349.

    20. Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, Wicha M, Clarke MF,Simeone DM. Identification of pancreatic cancer stem cells. Cancer Res 2007;67:1030–1037.

    21. Challen GA, Boles N, Lin KK-Y, Goodell MA. Mouse hematopoietic stem cell identifica-tionand analysis. Cytometry A 2009;75A: in press.DOI: 10.1002/cyto.a.20674 (thisissue).

    22. Möbius-Winkler S, Höllriegel R, Schuler G, Adams V. Endothelial progenitor cells:Implications for cardiovascular disease. Cytometry A 2009;75A: in press. DOI:10.1002/cyto.a.20669 (this issue).

    23. Takács L, Tóth L, Berta A, Vereb G. Stem Cells of the Adult cornea: From cytometricmarkers to therapeutic applications. Cytometry A 2009;75A: in press. DOI: 10.1002/cyto.a.20671 (this issue).

    24. Trujillo CA, Schwindt TT, Martins AH, Alves JM, Mello LE, Ulrich H. Novel perspec-tives of neural stem cell differentiation: From neurotransmitters to therapeutics.Cytometry A 2009;75A:in press. DOI: 10.1002/cyto.a.20666 (this issue).

    25. Zuba-Surma EK, Kucia M, Ratajczak J, Ratajczak MZ. ‘‘Small Stem Cells’’ in adult tis-sues: Very small embryonic-like stem cells (VSELs) Stand up! Cytometry A 2009;75A:in press. DOI: 10.1002/cyto.a.20665 (this issue).

    26. Parker GC, Anastassova-Kristeva M, Broxmeyer HE, Dodge WH, Eisenberg LM,Gehling UM, Guenin LM, Huss R, Moldovan NI, Rao M, Srour EF, Yoder MC. Stemcells: Shibboleths of development. Stem Cells and Development 2004;13:579–584.

    27. Young HE, Duplaa C, Katz R, Thompson T, Hawkins KC, Boev AN, Henson NL,Heaton M, Sood R, Ashley D, Stout C, Morgan JH, 3rd, Uchakin PN, Rimando M,Long GF, Thomas C, Yoon JI, Park JE, Hunt DJ, Walsh NM, Davis JC, Lightner JE,Hutchings AM, Murphy ML, Boswell E, McAbee JA, Gray BM, Piskurich J, Blake L,Collins JA, Moreau C, Hixson D, Bowyer FP 3rd, Black AC Jr. Adult-derived stemcells and their potential for use in tissue repair and molecular medicine. J Cell MolMed 2005;9:753–769.

    28. Ratajczak MZ, Zuba-Surma EK, Wysoczynski M, Wan W, Ratajczak J, Wojakowski W,Kucia M. Hunt for pluripotent stem cell—Regenerative medicine search for almighty cell. J Autoimmun 2008;30:151–162.

    29. Wicha MS, Liu S, Dontu G Cancer stem cells: An old idea—A paradigm shift. CancerRes 2006;66:1883–1890; discussion 1895–1896.

    30. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100:57–70.

    31. Elenbaas B, Spirio L, Koerner F, Fleming MD, Zimonjic DB, Donaher JL, PopescuNC, Hahn WC, Weinberg RA. Human breast cancer cells generated by oncogenictransformation of primary mammary epithelial cells. Genes Dev 2001;15:50–65.

    32. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, Nie J,Jonsdottir GA, Ruotti V, Stewart R, Slukvin II, Thomson JA. Induced pluripotentstem cell lines derived from human somatic cells. Science 2007;318:1917–1920.

    33. Ayyanan A, Civenni G, Ciarloni L, Morel C, Mueller N, Lefort K, Mandinova A, Raf-foul W, Fiche M, Dotto GP, Brisken C. Increased Wnt signaling triggers oncogenic

    conversion of human breast epithelial cells by a Notch-dependent mechanism. ProcNatl Acad Sci USA 2006;103:3799–3804.

    34. Ladman AJ, Martinez AO. Cell contacts and surface features of three murine tumorsgrown as multicellular spheroids. Eur J Cell Biol 1988;45:224–229.

    35. Ponti D, Costa A, Zaffaroni N, Pratesi G, Petrangolini G, Coradini D, Pilotti S, Pier-otti MA, Daidone MG. Isolation and in vitro propagation of tumorigenic breast can-cer cells with stem/progenitor cell properties. Cancer Res 2005;65:5506–5511.

    36. Liao MJ, Zhang CC, Zhou B, Zimonjic DB, Mani SA, Kaba M, Gifford A, ReinhardtF, Popescu NC, Guo W, Eaton EN, Lodish HF, Weinberg RA. Enrichment of a popu-lation of mammary gland cells that form mammospheres and have in vivo repopulat-ing activity. Cancer Res 2007;67:8131–8138.

    37. Shipitsin M, Polyak K. The cancer stem cell hypothesis: In search of definitions, mar-kers, and relevance. Lab Invest 2008;88:459–463.

    38. Sheridan C, Kishimoto H, Fuchs RK, Mehrotra S, Bhat-Nakshatri P, Turner CH,Goulet R Jr, Badve S, Nakshatri H CD441/CD242   breast cancer cells exhibitenhanced invasive properties: An early step necessary for metastasis. Breast CancerRes 2006;8:R59.

    REVIEW ARTICLE

    Cytometry Part A    75A: 6774, 2009   73

  • 8/15/2019 Artigo 02 - cancer stem cells.pdf

    8/8

    39. Shu Q, Wong KK, Su JM, Adesina AM, Yu LT, Tsang YT, Antalffy BC, Baxter P, Perlaky L, Yang J, Dauser RC, Chintagumpala M, Blaney SM, Lau CC, Li XN. Direct orthotopictransplantation of fresh surgical specimen preserves CD1331  tumor cells in clinically relevant mouse models of medulloblastoma and glioma. Stem Cells 2008;26:1414–1424.

    40. Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, Guba M, Bruns CJ,Heeschen C. Distinct populations of cancer stem cells determine tumor growth andmetastatic activity in human pancreatic cancer. Cell Stem Cell 2007;1:313–323.

    41. Baba T, Convery PA, Matsumura N, Whitaker RS, Kondoh E, Perry T, Huang Z,Bentley RC, Mori S, Fujii S, Marks JR, Berchuck A, Murphy SK. Epigenetic regulationof CD133 and tumorigenicity of CD1331  ovarian cancer cells. Oncogene 2008: inpress. DOI:10.1038/onc.2008.374.

    42. Todaro M, Alea MP, Di Stefano AB, Cammareri P, Vermeulen L, Iovino F, Tripodo C,Russo A, Gulotta G, Medema JP, Stassi G. Colon cancer stem cells dictate tumor growthand resist cell death by production of interleukin-4. Cell Stem Cell 2007;1:389–402.

    43. Beier D, Hau P, Proescholdt M, Lohmeier A, Wischhusen J, Oefner PJ, Aigner L,Brawanski A, Bogdahn U, Beier CP. CD133(1) and CD133(2) glioblastoma-derivedcancer stem cells show differential growth characteristics and molecular profiles.Cancer Res 2007;67:4010–4015.

    44. Dylla SJ, Beviglia L, Park IK, Chartier C, Raval J, Ngan L, Pickell K, Aguilar J, Lazetic S,Smith-Berdan S, Clarke MF, Hoey T, Lewicki J, Gurney AL. Colorectal cancer stem cellsare enriched in xenogeneic tumors following chemotherapy. PLoS ONE 2008;3:e2428.

    45. Dalerba P, Dylla SJ, Park IK, Liu R, Wang X, Cho RW, Hoey T, Gurney A, Huang EH,Simeone DM, Shelton AA, Parmiani G, Castelli C, Clarke MF. Phenotypic characteri-zation of human colorectal cancer stem cells. Proc Natl Acad Sci USA 2007;104:10158–10163.

    46. Szotek PP, Pieretti-Vanmarcke R, Masiakos PT, Dinulescu DM, Connolly D, Foster R,Dombkowski D, Preffer F, Maclaughlin DT, Donahoe PK. Ovarian cancer side popu-lation defines cells with stem cell-like characteristics and mullerian inhibiting sub-stance responsiveness. Proc Natl Acad Sci USA 2006;103:11154–11159.

    47. Mitsutake N, Iwao A, Nagai K, Namba H, Ohtsuru A, Saenko V, Yamashita S. Charac-terization of side population in thyroid cancer cell lines: Cancer stem-like cells areenriched partly but not exclusively. Endocrinology 2007;148:1797–1803.

    48. Patrawala L, Calhoun T, Schneider-Broussard R, Zhou J, Claypool K, Tang DG. Sidepopulation is enriched in tumorigenic, stem-like cancer cells, whereas ABCG21 andABCG2- cancer cells are similarly tumorigenic. Cancer Res 2005;65:6207–6219.

    49. Benchaouir R, Picot J, Greppo N, Rameau P, Stockholm D, Garcia L, Paldi A,Laplace-Builhe C. Combination of quantification and observation methods for study of ‘‘Side Population’’ cells in their ‘‘in vitro’’ microenvironment. Cytometry A2007;71:251–257.

    50. Yang ZF, Ngai P, Ho DW, Yu WC, Ng MN, Lau CK, Li ML, Tam KH, Lam CT, PoonRT, Fan ST. Identification of local and circulating cancer stem cells in human livercancer. Hepatology 2008;47:919–928.

    51. Ma S, Chan KW, Lee TK, Tang KH, Wo JY, Zheng BJ, Guan XY. Aldehyde dehydro-genase discriminates the CD133 liver cancer stem cell populations. Mol Cancer Res2008;6:1146–1153.

    52. Ginestier C, Hur MH, Charafe-Jauffret E, Monville F, Dutcher J, Brown M, Jacquemier J,Viens P, Kleer CG, Liu S, Schott A, Hayes D, Birnbaum D, Wicha MS, Dontu G.ALDH1 is a marker of normal and malignant human mammary stem cells and a predic-tor of poor clinical outcome. Cell Stem Cell 2007;1:555–567.

    53. Klonisch T, Wiechec E, Hombach-Klonisch S, Ande SR, Wesselborg S, Schulze-Osth-off K, Los M. Cancer stem cell markers in common cancers—Therapeutic implica-tions. Trends Mol Med 2008;14:450–460.

    54. Hill RP. Identifying cancer stem cells in solid tumors: Case not proven. Cancer Res2006;66:1891–1895; discussion 1890.

    55. Houghton J, Stoicov C, Nomura S, Rogers AB, Carlson J, Li H, Cai X, Fox JG, Gold-enring JR, Wang TC. Gastric cancer originating from bone marrow-derived cells.Science 2004;306:1568–1571.

    56. Hill RP, PerrisR. ‘‘Destemming’’ cancer stemcells. J Natl Cancer Inst2007;99:1435–1440.

    57. Axelson H, Fredlund E, Ovenberger M, Landberg G, Pahlman S. Hypoxia-induceddedifferentiation of tumor cells—A mechanism behind heterogeneity and aggressive-ness of solid tumors. Semin Cell Dev Biol 2005;16:554–563.

    58. Shmelkov SV, Jun L, St Clair R, McGarrigle D, Derderian CA, Usenko JK, Costa C,Zhang F, Guo X, Rafii S. Alternative promoters regulate transcription of the gene thatencodes stem cell surface protein AC133. Blood 2004;103:2055–2061.

    59. Jaksch M, Munera J, Bajpai R, Terskikh A, Oshima RG. Cell cycle-dependent varia-tion of a CD133 epitope in human embryonic stem cell, colon cancer, and melanomacell lines. Cancer Res 2008;68:7882–7886.

    60. Shmelkov SV, Butler JM, Hooper AT, Hormigo A, Kushner J, Milde T, St Clair R, Bal- jevic M, White I, Jin DK, Chadburn A, Murphy AJ, Valenzuela DM, G ale NW, Thur-

    ston G, Yancopoulos GD, D’Angelica M, Kemeny N, Lyden D, Rafii S. CD133 expres-sion is not restricted to stem cells, and both CD1331 and CD1332 metastatic coloncancer cells initiate tumors. J Clin Invest 2008;118:2111–2120.

    61. Wright MH, Calcagno AM, Salcido CD, Carlson MD, Ambudkar SV, Varticovski L.Brca1 breast tumors contain distinct CD441/CD242 and CD1331 cells with cancerstem cell characteristics. Breast Cancer Res 2008;10:R10.

    62. Palyi-Krekk Z, Barok M, Isola J, Tammi M, Szollosi J, Nagy P. Hyaluronan-inducedmasking of ErbB2 and CD44-enhanced trastuzumab internalisation in trastuzumabresistant breast cancer. Eur J Cancer 2007;43:2423–2433.

    63. Zeilstra J, Joosten SP, Dokter M, Verwiel E, Spaargaren M, Pals ST. Deletion of theWNT target and cancer stem cell marker CD44 in Apc(Min/1) mice attenuates intes-tinal tumorigenesis. Cancer Res 2008;68:3655–3661.

    64. Krause DS, Lazarides K, von Andrian UH, Van Etten RA. Requirement for CD44 inhoming and engraftment of BCR-ABL-expressing leukemic stem cells. Nat Med2006;12:1175–1180.

    65. Cammareri P, Lombardo Y, Francipane MG, Bonventre S, Todaro M, Stassi G. Isola-tion and culture of colon cancer stem cells. Methods Cell Biol 2008;86:311–324.

    66. Kelly PN, Dakic A, Adams JM, Nutt SL, Strasser A. Tumor growth need not be drivenby rare cancer stem cells. Science 2007;317:337.

    67. Hewitt HB, Blake E, Proter EH. The effect of lethally irradiated cells on the trans-plantability of murine tumours. Br J Cancer 1973;28:123–135.

    68. Kern SE, Shibata D. The fuzzy math of solid tumor stem cells: A perspective. CancerRes 2007;67:8985–8988.

    69. Barok M, Isola J, Palyi-Krekk Z, Nagy P, Juhasz I, Vereb G, Kauraniemi P, Kapanen A,Tanner M, Vereb G, Szollosi J. Trastuzumab causes antibody-dependent cellular cyto-toxicity-mediated growth inhibition of submacroscopic JIMT-1 breast cancer xeno-grafts despite intrinsic drug resistance. Mol Cancer Ther 2007;6:2065–2072.

    70. Francipane MG, Alea MP, Lombardo Y, Todaro M, Medema JP, Stassi G. Crucial roleof interleukin-4 in the survival of colon cancer stem cells. Cancer Res 2008;68:4022–4025.

    71. Conway AE, Lindgren A, Galic Z, Pyle AD, Wu H, Zack JA, Pelligrini M, Teitell MA,Clark A. A Pluripotency and self-renewal program controls the expansion of geneti-cally unstable cancer stem cells in pluripotent stem cell-derived tumors. Stem Cells2008: in press. DOI: 10.1634/stemcells.2008-0529.

    72. Odoux C, Fohrer H, Hoppo T, Guzik L, Stolz DB, Lewis DW, Gollin SM, GamblinTC, Geller DA, Lagasse E. A stochastic model for cancer stem cell origin in metastaticcolon cancer. Cancer Res 2008;68:6932–6941.

    73. Yilmaz OH, Valdez R, Theisen BK, Guo W, Ferguson O, Wu H, Morrison SJ. Ptendependence distinguishes haematopoietic stem cells from leukaemia-initiating cells.Nature 2006;441:475–482.

    74. Hope KJ, Jin L, Dick JE. Acute myeloid leukemia originates from a hierarchy of leuke-mic stem cell classes that differ in self-renewal capacity. Nat Immunol 2004;5:738–743.

    75. Thayer SP, di Magliano MP, Heiser PW, Nielsen CM, Roberts DJ, Lauwers GY, Qi YP,Gysin S, Fernandez-del Castillo C, Yajnik V, Antoniu B, McMahon M, Warshaw AL,Hebrok M. Hedgehog is an early and late mediator of pancreatic cancer tumorigene-sis. Nature 2003;425:851–856.

    76. Wang S, Garcia AJ, Wu M, Lawson DA, Witte ON, Wu H. Pten deletion leads to theexpansion of a prostatic stem/progenitor cell subpopulation and tumor initiation.Proc Natl Acad Sci USA 2006;103:1480–1485.

    77. Ma S, Lee TK, Zheng BJ, Chan KW, Guan XY. CD1331HCC cancer stem cells conferchemoresistance by preferential expression of the Akt/PKB survival pathway. Onco-gene 2008;27:1749–1758.

    78. Valent P. Emerging stem cell concepts for imatinib-resistant chronic myeloid leukae-

    mia: Implications for the biology, management, and therapy of the disease. Br J Hae-matol 2008;142:361–378.

    79. Bauerschmitz GJ, Ranki T, Kangasniemi L, Ribacka C, Eriksson M, Porten M,Herrmann I, Ristimaki A, Virkkunen P, Tarkkanen M, Hakkarainen T, Kanerva A,Rein D, Pesonen S, Hemminki A. Tissue-specific promoters active inCD441CD24-/low breast cancer cells. Cancer Res 2008;68:5533–5539.

    80. Campbell LL, Polyak K. Breast tumor heterogeneity: Cancer stem cells or clonal evo-lution? Cell Cycle 2007;6:2332–2338.

    81. Adams JM, Strasser A. Is tumor growth sustained by rare cancer stem cells or domi-nant clones? Cancer Res 2008;68:4018–4021.

    82. Polyak K. Is breast tumor progression really linear? Clin Cancer Res 2008;14:339–341.

    83. Rak J, Milsom C, Yu J. Vascular determinants of cancer stem cell dormancy—Do ageand coagulation system play a role? Acta Pathol Microbiol Immunol Scand2008;116:660–676.

    84. Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, Bell GW, Richardson AL,Polyak K, Tubo R, Weinberg RA. Mesenchymal stem cells within tumour stroma pro-mote breast cancer metastasis. Nature 2007;449:557–563.

    85. Calabrese C, Poppleton H, Kocak M, Hogg TL, Fuller C, Hamner B, Oh EY, GaberMW, Finklestein D, Allen M, Frank A, Bayazitov IT, Zakharenko SS, Gajjar A, David-off A, Gilbertson RJ. A perivascular niche for brain tumor stem cells. Cancer Cell

    2007;11:69–82.86. Bhatia B, Multani AS, Patrawala L, Chen X, Calhoun-Davis T, Zhou J, Schroeder L,

    Schneider-Broussard R, Shen J, Pathak S, Chang S, Tang DG. Evidence that senescenthuman prostate epithelial cells enhance tumorigenicity: Cell fusion as a potentialmechanism and inhibition by p16INK4a and hTERT. Int J Cancer 2008;122:1483–1495.

    REVIEW ARTICLE

    74   Cancer Stem Cells