Complete Stem Cells and Cancer 1st Edition Kimberly E. Foreman Ebook Download PDF &
Complete Stem Cells and Cancer 1st Edition Kimberly E. Foreman Ebook Download PDF &
https://ebookfinal.com/download/cancer-stem-cells-1st-edition-
vinagolu-k-rajasekhar/
https://ebookfinal.com/download/cancer-stem-cells-methods-and-
protocols-1st-edition-gianpaolo-papaccio/
https://ebookfinal.com/download/cancer-stem-cells-1st-edition-william-
l-farrar-phd/
https://ebookfinal.com/download/cancer-stem-cells-methods-and-
protocols-1st-edition-michael-c-dovey/
Mesenchymal Stem Cells in Cancer Therapy 1st Edition
Khalid Shah (Auth.)
https://ebookfinal.com/download/mesenchymal-stem-cells-in-cancer-
therapy-1st-edition-khalid-shah-auth/
https://ebookfinal.com/download/therapeutic-potential-of-
differentiation-in-cancer-and-normal-stem-cells-1st-edition-juan-
antonio-marchal/
https://ebookfinal.com/download/patient-specific-stem-cells-1st-
edition-lamba/
https://ebookfinal.com/download/induced-stem-cells-1st-edition-
patrick-j-sullivan/
https://ebookfinal.com/download/germline-stem-cells-1st-edition-david-
a-dansereau/
Stem Cells and Cancer 1st Edition Kimberly E. Foreman
Digital Instant Download
Author(s): Kimberly E. Foreman, Paola Rizzo, Clodia Osipo, Lucio Miele
(auth.), Beverly A. Teicher, Rebecca G. Bagley (eds.)
ISBN(s): 9781603279338, 1603279334
Edition: 1
File Details: PDF, 5.99 MB
Year: 2009
Language: english
Cancer Drug Discovery and Development
Series Editor: Beverly A. Teicher, Genzyme Corporation
Framingham, MA, USA
Edited by
REBECCA G. BAGLEY, MS
Genzyme Corporation,
Framingham, MA, USA
The recent surge in stem cell research has ignited a field of discovery into many human
diseases including diabetes, neuropathologies, and cancer. Stem cell therapy is a prom-
ising approach to the treatment of many debilitating diseases to replace specific differ-
entiated cells that have been lost or died. Although stem cells may provide therapeutic
benefit under certain conditions, stem cells are often implicated in the initiation, pro-
gression, and therapeutic resistance of malignant disease.
This first edition of Stem Cells and Cancer is intended to give a current perspective
on the role of stem cells in cancer and strategies for novel therapies directed toward
tumor stem cells. Cancer stem cells remain a controversial topic and the criteria that
define cancer stem cells are continuing to evolve. The current cancer stem cell hypoth-
esis is presented in several chapters with distinctions made between the hierarchical and
stochastic models of tumor cell development. “Stemness,” self-renewal, pluripotency,
clonality, and tumorigenicity are important concepts applied toward defining cancer
stem cells. Signaling pathways such as Wnt, Sonic Hedgehog, Notch, and Bmi-1 that
are involved in differentiation, proliferation, and survival are implicated in the malignant
process. Additional chapters address the identification of cancer stem cell populations
through the evaluation of molecular markers such as CD133, CD44, and CD24, for
example, or by Hoechst dye exclusion to recognize “side populations.” Mesenchymal
and hematopoietic stem cells are described as well as mouse models that are employed
to elucidate the properties and functionality of stem cells in cancer and the stem cell
niche. This book encompasses a wide variety of human cancers that include but are not
limited to leukemia, gliomas, breast, and prostate cancers. Resistance to conventional
therapies, genetic vs. epigenetic changes that affect therapeutic response, and strategies
to prevent disease recurrence are challenges that have been incorporated into this volume.
Stem Cells and Cancer represents a compendium of cutting edge research by experts
in the field and will be instrumental in the study of this intriguing line of investigation
for many years to come.
Framingham, MA Rebecca G. Bagley
Beverly A. Teicher
v
Contents
vii
viii Contents
10 Mouse Mammary Tumor Virus: Stem Cells and Mammary Cancer .............. 127
Gilbert H. Smith
11 Tumor Dormancy, Metastasis, and Cancer Stem Cells ................................... 141
Alysha K. Croker, Jason L. Townson, Alison L. Allan,
and Ann F. Chambers
12 Cancer Stem Cells: Gastrointestinal Cancers ................................................. 155
Hideshi Ishii, Naotsugu Haraguchi, Keisuke Ieta,
Koshi Mimori, and Masaki Mori
13 Cancer Stem Cells: Hepatocellular Carcinoma............................................... 165
Thomas Shupe and Bryon E. Petersen
14 Cancer Stem Cells: Lung Cancer .................................................................... 177
Jaclyn Y. Hung
15 Cancer Stem Cells: Pancreatic Cancer ............................................................ 185
Joseph Dosch, Cheong Jun Lee, and Diane M. Simeone
16 Prostate Stem Cells and Cancer in Animals ................................................... 199
Alexander Yu. Nikitin, Melia G. Nafus, Zongxiang Zhou,
Chun-Peng Liao, and Pradip Roy-Burman
17 Prostate Cancer Stem/Progenitor Cells ........................................................... 217
Sofia Honorio, Hangwen Li, and Dean G. Tang
18 Adult Prostate Epithelium Renewal, Stem Cells and Cancer ......................... 231
Chiara Grisanzio and Sabina Signoretti
19 Stem Cells, Angiogenesis, and Neurogenesis in Tumors ............................... 247
Judith A. Varner
Part V Targeting Cancer Stem Cells with Therapy
20 Implications of Cancer Stem Cells for Cancer Therapy ................................. 255
Liang Cheng, Shaobo Zhang, Darrell D. Davidson,
Rodolfo Montironi, and Antonio Lopez-Beltran
24 Therapeutic Index and the Cancer Stem Cell Paradigm ................................. 309
Vera S. Donnenberg and Albert D. Donnenberg
Index........................................................................................................................ 327
Contributors
Alison L. Allan, Ph.D. • London Regional Cancer Program, London, ON, Canada
Department of Anatomy and Cell Biology, University of Western Ontario, London,
ON, Canada; Department of Oncology, University of Western Ontario, London,
ON, Canada
Rebecca G. Bagley, M.S. • Genzyme Corporation, Framingham, MA, USA
Surinder K. Batra, Ph.D. • Department of Biochemistry & Molecular Biology,
Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska
Medical Center, Omaha, NE, USA
Stephen B. Baylin, MD • Cancer Biology Division, The Sidney Kimmel
Comprehensive Cancer Center, The Johns Hopkins University Medical Institutions,
Baltimore, MD, USA
Johan Bengzon, M.D., Ph.D. • Division of Neurosurgery, Rausing Laboratory,
Department of Clinical Sciences, University Hospital, Lund, Sweden
Lung Strategic Research Center for Stem Cell Biology and Cell Therapy,
University Hospital, Lund, Sweden; Department of Pathology, Division of Neuropa-
thology, University Hospital, Lund, Sweden
Keith L. Black, M.D. • Department of Neurosurgery, Cedars-Sinai Medical Center,
Los Angeles, CA, USA
Angelika M. Burger, Ph.D. • Department of Pharmacology, Wayne State
University, Karmanos Cancer Institute, Hudson-Webber Cancer Research Center,
Detroit, MI, USA
Ann F. Chambers, Ph.D. • London Regional Cancer Program, London, ON,
Canada; Department of Oncology, University of Western Ontario, London, ON,
Canada; Department of Medical Biophysics, University of Western Ontario,
London, ON, Canada
Liang Cheng, M.D. • Department of Pathology and Urology, Indiana School
of Medicine, Indianapolis, IN, USA; School of Medicine, Polytechnic University
of the Marche Region, United Hospitals, Ancona, Italy; Department of Surgery,
Cordoba University School of Medicine, Cordoba, Spain
Alysha K. Croker, B.Sc. • London Regional Cancer Program, London, ON,
Canada; Department of Anatomy and Cell Biology, University of Western
Ontario, London, ON, Canada; Department of Oncology, University of Western
xi
xii Contributors
Abstract
The “cancer stem cell” hypothesis is receiving increasing interest and has become the object of considerable
debate among cancer biologists and clinicians. This ongoing debate is focusing attention on the very
definition of stemness and its significance in the context of a malignancy. From a therapeutic standpoint,
the cancer stem cell hypothesis emphasizes the cellular heterogeneity in cancers, and the need to specifi-
cally target small cell populations that resemble tissue stem cells and are phenotypically different from the
majority of cancer cells. Regardless of their origin, these cells divide slowly, have the ability to undergo
asymmetric cell division and are highly resistant to conventional chemotherapeutics. These characteristics
make them prime suspects as potential causes of disease recurrence and metastasis, which are the main
causes of morbidity and mortality in oncology. This chapter provides an introduction to the cancer stem
cell hypothesis, briefly summarizes the evidence supporting this theory and the aspects that remain contro-
versial. Finally, we present a brief discussion of the possible therapeutic significance of cancer stem cells
and the current efforts to target developmental pathways on which these cells depend.
Key Words: Cancer stem cells, Tumor-initiating cells, Stem cell niche, Targeted therapies
From: Cancer Drug Discovery and Development: Stem Cells and Cancer,
Edited by: R.G. Bagley and B.A. Teicher, DOI: 10.1007/978-1-60327-933-8_1,
© Humana Press, a part of Springer Science + Business Media, LLC 2009
3
4 Part I / Introduction to Cancer Stem Cells
“Differentiation”
Self-replication
Proliferation
Asymmetric
Cell
Division
CSC
“Progenitor”
CSC
“Progenitor”
Bulk
Cancer cells
Fig. 1. The CSC hypothesis. CSCs are thought to maintain their numbers by slow self-replication, and produce other
tumor cells by asymmetric cell division. In this process, cell division of a CSC generates a CSC and a transformed
“progenitor-like” cell, which has limited self-renewal ability but are highly proliferative, similar to a transit-amplifying
population in normal tissue. These progenitors give rise to more or less partially differentiated bulk tumor cells through
a combination of proliferation and abortive differentiation.
tumor cells that are ultimately responsible for tumor formation and growth. These cells, defined as
“cancer stem cells” (CSCs) or tumor initiating cells (TICs), possess several key properties of normal
tissue stem cells including self-renewal (i.e., the ability of a cell to renew itself indefinitely in an
undifferentiated state), unlimited proliferative potential, infrequent or slow replication, resistance to toxic
xenobiotics, high DNA repair capacity, and the ability to give rise to daughter cells that differentiate.
However, unlike highly regulated tissue stem cells, CSCs demonstrate dysregulated self-renewal/dif-
ferentiation programs and produce daughter cells that arrest at various stages of differentiation. The
daughter cells make up the bulk of the tumor and are characterized by rapid replication, limited pro-
liferative potential, and the inability to form a new tumor. Only the CSC is able to initiate tumor
formation as it is solely capable of self-renewal. A diagrammatic representation of the CSC hypoth-
esis is shown in Fig. 1.
The strongest evidence for the CSC theory comes from studies in acute myelogenous leukemia
(AML). Landmark studies by Dick and colleagues demonstrated that only rare cells in AML were able
to initiate leukemia in murine models, and serial transplantation studies revealed these cells had a high
self-renewal capacity (4, 5). The cell responsible for tumor initiation was identified as having a
CD34+CD38− phenotype, which was particularly interesting as bulk AML samples tend to be CD34−.
Furthermore, CD34+CD38− is a phenotype characteristic of normal hematopoietic stem cells (HSC)
indicating the putative CSCs may have a primitive phenotype. Bonnet et al. found that as few as 5 ×
103 CD34+CD38− cells could engraft an immunocompromised mouse, while 100 times more CD34− or
CD34+CD38+ cells from the same donor could not (5). Importantly, the tumors derived from injection of
the CD34+CD38− cells was heterogeneous and composed of a mixture of tumorigenic and nontumori-
genic cells similar to the donor sample (5). Since then, stem-like cells have been identified in a variety
Chapter 1 / The Cancer Stem Cell Hypothesis 5
of human malignancies including other leukemias and solid tumors such as breast, colon, brain, head
and neck, lung, pancreatic, nasopharyngeal cancers, and melanomas (4–18). In many cases, a tumori-
genic subset of cells could be reproducibly identified and isolated based on a distinct set of cell mark-
ers separating it from the nontumorigenic subset (19). Attempts to isolate CSCs from other malignancies
are underway in laboratories worldwide, and this list is likely to grow. Remarkably, even established
cancer cell lines that have been grown in vitro for many years appear to contain CSC-like populations
that can be isolated and are highly tumorigenic (20, 21). The surface markers of CSCs from different
tumor types are diverse, suggesting that their biological behaviors may be different as well.
One reason the CSC theory has generated such enthusiasm is that it may help explain long-standing
problems in cancer biology. It is well-recognized that tumors are heterogeneous in terms of both
functional heterogeneity and cellular composition. Functional heterogeneity refers to the observation
that only a small portion of tumor cells can give rise to colonies in clonogenic assays in vitro or
tumors in vivo. Under the traditional theory of tumor formation (also called the stochastic model),
every tumor cell should be equally capable of forming a tumor. As tens to hundreds of thousands of
tumor cells are needed to reproducibly initiate tumors in animal models, investigators concluded that
the process was inefficient. However, with the CSC theory, the number of cells needed to form a
tumor would simply be determined by the relative frequency of CSC in the tumor population. A suf-
ficient number of CSCs must be present in the inoculum, since most cells in the line are proliferative
but nontumorigenic. The phenotypic heterogeneity of tumors is also more easily explained by the CSC
theory. Mutations in the CSC would be passed on to each daughter cell, and as the daughter cell dif-
ferentiates, it may arrest at any one of numerous points prior to full maturation. In the stochastic
model, the tumor cell would need to dedifferentiate to different degrees to form a phenotypically
heterogeneous but genetically clonal population. Although the genomic instability associated with
cancer clearly makes this possible, it is easier to envision an abortive version of the normal hierarchi-
cal differentiation program in a tissue as opposed to a random back-differentiation process affecting
individual cells to different extents.
It has also been postulated that the CSC theory may explain why it is so difficult to treat cancer. If
this model is correct, then directing cancer therapeutics at the bulk of rapidly replicating tumor cells
is not likely to achieve tumor eradication, unless the CSCs are eliminated. This could explain the vex-
ing problem faced by oncologists worldwide, who often can achieve complete clinical and pathologi-
cal remissions of cancers with chemotherapy, only to see the cancers recur, often in metastatic and
ultimately lethal forms. This clinical phenomenon implies that very small numbers of cells, some-
times undetectable even by sophisticated molecular diagnostic tools, are capable of causing tumor
relapses. Standard chemotherapeutic strategies using mitotic poisons, DNA-damaging agents, antime-
tabolites, or even modern “targeted” agents such as growth factor receptor kinase inhibitors often are
aimed at actively proliferating cells resulting in growth arrest and/or cell death. This strategy effi-
ciently kills the daughter cells, but is much less effective against CSCs, which can remain quiescent
for extended periods of time. Thus, tumors may shrink in response to traditional chemotherapy, even
to the point where they are undetectable, yet CSCs often persist and eventually cause relapsed and
metastatic disease (2). Furthermore, when CSCs are exposed to and escape from chemotherapy-in-
duced death, they may become more resistant to these insults and pass this on to their daughter cells.
This may explain why recurrent cancers are often more resistant to treatment than primary disease.
An additional characteristic of CSCs that makes them more difficult to eradicate than “bulk” cancer
cells is their high level expression of ABC family transporters, which catalyze the ATP-dependent
transport of toxic chemicals from the cell (22). These molecules were originally identified as one of
the main cause of multidrug resistance in cancers (23). Evolutionarily, it is plausible that normal tissue
stem cells would be particularly well protected against toxic insults, because of their fundamental role
6 Part I / Introduction to Cancer Stem Cells
in tissue regeneration. Unfortunately, this property also makes the neoplastic counterparts of tissue
stem cells highly resistant to many common chemotherapeutic drugs. Indeed, one of the most popular
ways of isolating putative CSC population takes advantage of their ability to rapidly efflux DNA-
binding fluorescent dyes such as Hoechst or 7-AAD, which is due to high level expression of ABC
transporters. Cells that retain less dye appear as a “side population” (SP) in flow cytometry experi-
ments. In several cases, SP cells have been shown to be enriched in putative CSCs (24–26).
the transformation process could reprogram a cell to a stem-like phenotype. It is important to note that
these studies were conducted in fibroblasts and not epithelial cells. Can a similar process of reprogram-
ming occur in common epithelial malignancies? A process of partial dedifferentiation has been known
for years in epithelial cancers as epithelial-mesenchymal transition (EMT) (31–34). This consists in
loss of epithelial markers, such as tissue-specific cytokeratins, and adhesion molecules, such as
E-cadherin, and acquisition of markers typical of mesenchymal cells, such as vimentin and N-cadherin.
The process of EMT is thought to contribute to the ability of transformed epithelial cells to metasta-
size. In this model, cancer cells need to undergo EMT to migrate through the body, and once they seed
distant metastatic sites, they can revert to a more or less “epithelial” phenotype through a process of
mesenchymal-epithelial transition (MET). Several transcription factors such as Twist, Snail, or Slug
and secretory proteins of the TGF-β family, including some bone morphogenetic proteins (BMPs), can
induce the EMT program (34). Vascular mimicry is thought to be a specialized form of EMT in which
tumor cells can acquire an endothelial phenotype (35, 36).
Thus, the question seems to be not whether or not differentiation plasticity is possible in epithelial
cancer cells, but whether this process can go as far as generating a cell that has the functional
characteristics of a stem cell. What a simple dedifferentiation model does not immediately explain is
the hierarchical organization of cells in malignancies. If dedifferentiation is a secondary event that
arises through selection and confers a selective advantage to less differentiated cells, why is there a
hierarchical organization among neoplastic cells with a highly tumorigenic, dededifferentiated popula-
tion capable of generating less tumorigenic, more differentiated cells? One possible explanation is that
dedifferentiation is a highly improbable event, which produces a cell fate program that includes func-
tional “stemness.” Thus, only a few cells or even a single cell would have to undergo this process to
generate a small population of CSCs. These then give rise to the rest of the cancer cell population
through a process of hierarchical abortive differentiation that imperfectly recapitulates that of a
normal tissue.
An intermediate possibility is that the CSC could originate not exclusively from tissue stem cells,
but from a restricted number of cell populations including tissue stem cells and immature progenitor
cells, which are immediately below tissue stem cells in the differentiation hierarchy and are capable
of short-term self-replication. Experimental support for this hypothesis comes from several studies in
leukemia where the introduction of oncogenic fusion gene products into hematopoietic progenitor cells
resulted in AML in animal models. Cozzio et al. found that expression of the MLL-ENL fusion gene
product in hematopoietic progenitor cells resulted in leukemia, albeit with less efficiency than when it
was expressed in true hematopoietic stem cells (37). Similar results were also found with the MOZ-
TIF2 fusion gene product (38). More recently, Somervaille and Cleary enforced MLL-AF9 expression
in normal murine HSC and progenitor cells (39). Using serial transplantation in mice, they discovered
that the functional CSC expressed MAC-1 and Gr1, two markers associated with more mature cells
(39). Interestingly, the cells also expressed the stem cell marker c-kit, suggesting CSCs may express an
unusual combination of cell markers (39). Taken together, these studies clearly support the notion that
AML may arise from either stem or progenitor cells in a mouse model; however, caution should be
used in interpreting this data. Murine cells are generally easier to transform than human cells; hence,
it is unclear if these findings are relevant to human disease (40). A similar theory has been proposed
for breast cancer (41, 42). According to Dontu et al. (41), the existence of ERa-negative breast cancers
and ERa-positive breast cancers of variable biological aggressiveness may be explained by postulating
that CSCs in these cancers originate from different cell populations. The most aggressive, undifferenti-
ated ERa-negative cancers and poor-prognosis ERa-positive cancers would arise from the most primi-
tive mammary stem cells, which are ERa-negative, while less aggressive ERa-positive cancers would
arise from CSCs derived from intermediate progenitors that are ERa-positive. These can generate
8 Part I / Introduction to Cancer Stem Cells
Differentiation
Self-replication
Proliferation
Asymmetric
Cell
TSC
Division
TSC Progenitor
1 2
“De - differentiation”
CSC CSC
Fig. 2. Possible origins of CSCs. Three different but not mutually exclusive models are schematically presented.
“Lightning” symbols indicate transforming mutations. CSCs may originate exclusively from the transformation of
primitive tissue stem cells (TSC, model 1), or from the transformation of either TSC or progenitor cells (model 2).
Alternatively, CSCs may originate from the transformation and dedifferentiation of more mature cells, which reac-
quire stem cell properties as a consequence of transforming mutations (model 3) (see Color Plates).
are not necessary for continued in vitro propagation of the cell line. Strasser and colleagues have
proposed that the reason so many human cancer cells are needed to initiate tumor formation is that
the murine microenvironment is not appropriate for development of human cancers, and only a few
cells are capable of overcoming this hostile environment (43). These authors have taken the approach
of genetically engineered mouse cells to develop lymphoma (primary Em-myc lymphomas), isolating
subpopulations of the tumor cells based on the murine stem cell markers Sca-1 and AA4.1 (CD93),
and examining tumor formation in syngeneic naïve, immunocompetent mice (43). They report iden-
tify a small subpopulation (2–5%) of cells with stem-like characteristics, but found that Sca-1+AA4.1hi
and Sca-1+AA4.1lo cells were equally capable of forming tumors (43). These data have been inter-
preted as evidence against the universal validity of the cancer stem cell model. It should be pointed
out that although xenograft models are certainly artificial, transgenic mouse models of carcinogenesis
have important limitations of their own, and may or may not faithfully recapitulate human carcino-
genesis. Typically, in these models a very potent oncogene is overexpressed in a target cell population,
and the whole process of carcinogenesis and tumor progression is dramatically accelerated compared
with human disease. Mouse cells are far more susceptible to transformation than human cells, and
may be able to more easily reacquire functional “stemness.” It is interesting to notice that the oncogene
used in this particular experimental model, c-Myc, is also one of the stemness-inducing genes that can
reprogram human fibroblasts to an embryonic stem cell-like phenotype. Thus, an alternate explana-
tion for these data is that both Sca-1+AA4.1hi and Sca-1+AA4.1lo cells in this transgenic model have
acquired functional “stemness” through a process of dedifferentiation, and can behave as CSCs. More
sophisticated animal models will be required to gain further insights into this issue. These models
should be based on human cells, but attempt to recapitulate as much as possible the human microen-
vironment. Such a humanized xenograft model has been generated for the mammary gland (45), and
should provide valuable information on putative breast cancer stem cells.
The controversy on the human relevance of CSC data obtained in xenograft models underscores
the importance of tumor microenvironment in the biology of CSCs. Tumor-stroma interactions may
indeed be critical in reprogramming cancer cell developmental pathways. Transforming growth factor
(TGF)-b and bone morphogenetic proteins (BMPs) can be produced by tumor stroma, as can several
other mediators of intercellular communication such as Wnt, Hedgehog, and Notch ligands. There is
growing interest in studying the CSC “niche” as a potential therapeutic target. Normal stem cells are
well known to require signals from their immediate environment, including stromal cells, microvas-
cular endothelial cells, and extracellular matrix for their long-term survival and self-renewal. This
specialized microenvironment is commonly referred to as the stem cell niche, and it is best understood
in the hematopoietic system (46). There is increasing evidence that CSCs also require microenviron-
mental signals from specialized niches (47–49) (Fig. 3). Autocrine and paracrine mediators secreted
by the CSCs themselves or by other tumor cells may also play an important role, at least in some
malignancies (50, 51). How much autocrine or paracrine interactions contribute to the CSC niche is
still unclear. However, at least under some circumstances CSCs can recreate a niche-like environment
in the absence of other cell types. Putative breast cancer stem cells can form spheroids called “mam-
mospheres” in suspension culture (52, 53). Other putative CSCs can also form similar spheroids.
Mammospheres contain few CSCs, and mostly consist of precursors and partially differentiated cells.
Mammospheres can propagate in vitro and form secondary and tertiary mammospheres, which retain
the original cellular composition. This implies that at least under some culture conditions, the CSCs
themselves and their immediate progeny can form a functional niche that is capable of sustaining
self-renewal, asymmetric cell division and partial differentiation.
Undoubtedly, much remains to be clarified and further studies are needed. These may well reveal
that the origin of the functional CSC may vary based on the cell type involved and the specific nature
of the oncogenic events leading to transformation.
10 Part I / Introduction to Cancer Stem Cells
Endothelial
PG cells
SC CSC
ECM
Fig. 3. The CSC “niche”. In vivo, CSCs may require signals from their microenvironment to maintain their properties,
as is the case for normal tissue stem cells. Microenvironmental signals may be received from endothelial cells, from
various types of stromal cells (SC), such as fibroblasts, bone marrow stromal cells, or immunocytes infiltrating the
tumor, from progenitor cells (PG) derived from the CSCs themselves, and/or from the extracellular matrix (ECM). It
is likely that the cross-talk between CSCs and other cells is bidirectional. These signals may be therapeutically tar-
geted to deprive CSCs of indispensable microenvironmental signals.
far behind. Interest in using Notch inhibitors to target CSCs is growing. In glioblastomas, elevated
Notch expression has been associated with high nestin levels and is linked to a poor prognosis (62, 63).
Furthermore, Notch inhibition reduced the ability of brain CSCs to form tumors (64). In breast cancer,
Notch expression and activation has been associated with a poor prognosis, and studies indicate that
Notch inhibitors can kill breast cancer cells in vitro (65–67). As CSCs have been identified in primary
breast cancers, there has been much interest in Notch signaling in breast CSCs (6). Farnie et al. recently
compared mammospheres derived from normal mammary tissue and human ductal carcinoma in situ
(DCIS) and reported that activated Notch-1, Notch-4, and the downstream target Hes-1 were expressed
in mammospheres from DCIS samples, but not those derived from normal breast tissue (68). Notch
inhibition with a g-secretase inhibitor or a neutralizing Notch-4 antibody significantly reduced the ability
of DCIS derived cells to form mammospheres (68). These results suggest that Notch inhibition may be
able to preferentially target breast CSCs, while sparing normal mammary stem cells. Laboratories
around the world, including ours, are exploring the development of therapeutic regimens including
Notch, Hedgehog, or Wnt inhibitors to target CSCs. Figure 4 shows a simplified representation of
pathways that have been associated with CSC maintenance.
Notch
ligands
Hh Wnt
BMPs ECM
DNA repair
ABC
xenobiotics Slow replication
Self-renewal
Pluripotency
Bmi-1
Musashi
Sox2
Oct4
Fig. 4. Molecular pathways affecting CSCs. The figure shows a list, not meant to be all-inclusive, of pathways that have
been shown to modulate the CSC phenotype. Extracellular signals delivered through the Hedgehog (Hh), Notch, Wnt
pathways or through TGF-b and the related BMPs, or from ECM proteins and from growth factors such as hepatocyte
growth factor (Met ligand) may all participate in regulating the maintenance, self-renewal, and differentiation of CSCs.
Slow replication, ability to generate partially differentiated progenies (pluripotency) highly effective DNA repair, abil-
ity to eliminate xenobiotics through ABC family transporters (ABC), and expression of primitive membrane markers
(CD133, Met) have been documented in many putative CSC populations isolated from tumors or cell lines. Transcription
factors such as Bmi-1, Musashi, Sox2, Oct4, and others have been shown to be commonly expressed in putative CSCs
and participate in controlling their phenotype.
12 Part I / Introduction to Cancer Stem Cells
CONCLUSIONS
The CSC hypothesis has sparked a tremendous increase in scientific interest in the hierarchical
organization of cancer cells, the isolation of rare cellular subpopulations that may be responsible for
treatment failures, and the role of microenvironmental niches in the maintenance of these populations.
There are still many questions that remain unanswered, particularly surrounding the origin of CSC
populations in human tumors and the interpretation of data generated by current experimental models.
Yet, looking at cancers from the perspective offered by the CSC hypothesis may answer fundamental
questions in tumor biology and open the way to paradigm shifts in our therapeutic approach to malig-
nancies. Thus, it is reasonable to take the view that studying the mechanisms regulating the survival,
self-renewal, and differentiation of normal and transformed stem cells could potentially lead to tre-
mendous advances in the treatment of neoplastic diseases.
REFERENCES
1. Wicha MS, Liu S, Dontu G. Cancer stem cells: an old idea–a paradigm shift. Cancer Res 2006;66:1883–1890.
2. Song LL, Miele L. Cancer stem cells-an old idea that’s new again: implications for the diagnosis and treatment of breast
cancer. Expert Opin Biol Ther 2007;7:431–438.
3. Pardal R, Clarke MF, Morrison SJ. Applying the principles of stem-cell biology to cancer. Nat Rev Cancer
2003;3:895–902.
4. 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 leukaemia after transplantation into SCID mice. Nature 1994;367:645–648.
5. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hemat-
opoietic cell. Nat Med 1997;3:730–737.
6. Al Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast
cancer cells. Proc Natl Acad Sci USA 2003;100:3983–3988.
7. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusimano MD, Dirks PB. Identification
of human brain tumour initiating cells. Nature 2004;432:396–401.
8. Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB. Identification of a cancer stem cell in
human brain tumors. Cancer Res 2003;63:5821–5828.
9. Prince ME, Sivanandan R, Kaczorowski A, Wolf GT, Kaplan MJ, Dalerba P, Weissman IL, Clarke MF, Ailles LE.
Identification of a subpopulation of cells with cancer stem cell properties in head and neck squamous cell carcinoma.
Proc Natl Acad Sci USA 2007;104:973–978.
10. O’Brien CA, Pollett A, Gallinger S, Dick JE. A human colon cancer cell capable of initiating tumour growth in immuno-
deficient mice. Nature 2007;445:106–110.
11. Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, De Maria R. Identification and expansion of
human colon-cancer-initiating cells. Nature 2007;445:111–115.
12. Schatton T, Murphy GF, Frank NY, Yamaura K, Waaga-Gasser AM, Gasser M, Zhan Q, 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.
13. Seigel GM, Hackam AS, Ganguly A, Mandell LM, Gonzalez-Fernandez F. Human embryonic and neuronal stem cell
markers in retinoblastoma. Mol Vis 2007;13:823–832.
14. Ho MM, Ng AV, Lam S, Hung JY. Side population in human lung cancer cell lines and tumors is enriched with stem-like
cancer cells. Cancer Res 2007;67:4827–4833.
15. Zen Y, Fujii T, Yoshikawa S, Takamura H, Tani T, Ohta T, Nakanuma Y. Histological and culture studies with respect to
ABCG2 expression support the existence of a cancer cell hierarchy in human hepatocellular carcinoma. Am J Pathol
2007;170:1750–1762.
16. Wang J, Guo LP, Chen LZ, Zeng YX, Lu SH. Identification of cancer stem cell-like side population cells in human
nasopharyngeal carcinoma cell line. Cancer Res 2007;67:3716–3724.
17. Olempska M, Eisenach PA, Ammerpohl O, Ungefroren H, Fandrich F, Kalthoff H. Detection of tumor stem cell markers
in pancreatic carcinoma cell lines. Hepatobiliary Pancreat Dis Int 2007;6:92–97.
18. Haraguchi N, Inoue H, Tanaka F, Mimori K, Utsunomiya T, Sasaki A, Mori M. Cancer stem cells in human gastrointes-
tinal cancers. Hum Cell 2006;19:24–29.
Chapter 1 / The Cancer Stem Cell Hypothesis 13
19. 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 characterization of human colorectal cancer stem cells. Proc Natl Acad
Sci USA 2007;104:10158–10163.
20. Kondo T. Stem cell-like cancer cells in cancer cell lines. Cancer Biomark 2007;3:245–250.
21. Setoguchi T, Taga T, Kondo T. Cancer stem cells persist in many cancer cell lines. Cell Cycle 2004;3:414–415.
22. Lou H, Dean M. Targeted therapy for cancer stem cells: the patched pathway and ABC transporters. Oncogene
2007;26:1357–1360.
23. Donnenberg VS, Donnenberg AD. Multiple drug resistance in cancer revisited: the cancer stem cell hypothesis. J Clin
Pharmacol 2005;45:872–877.
24. Hadnagy A, Gaboury L, Beaulieu R, Balicki D. SP analysis may be used to identify cancer stem cell populations. Exp
Cell Res 2006;312:3701–3710.
25. Hirschmann-Jax C, Foster AE, Wulf GG, Nuchtern JG, Jax TW, Gobel U, Goodell MA, Brenner MK. A distinct “side popu-
lation” of cells with high drug efflux capacity in human tumor cells. Proc Natl Acad Sci USA 2004;101:14228–14233.
26. Hirschmann-Jax C, Foster AE, Wulf GG, Goodell MA, Brenner MK. A distinct “side population” of cells in human
tumor cells: implications for tumor biology and therapy. Cell Cycle 2005;4:203–205.
27. Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ. Prospective identification of tumorigenic prostate cancer stem
cells. Cancer Res 2005;65:10946–10951.
28. Hemmati HD, Nakano I, Lazareff JA, Masterman-Smith M, Geschwind DH, Bronner-Fraser M, Kornblum HI.
Cancerous stem cells can arise from pediatric brain tumors. Proc Natl Acad Sci USA 2003;100:15178–15183.
29. 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 pluripotent stem cell lines derived from human somatic cells. Science
2007;318:1917–1920.
30. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S. Induction of pluripotent stem cells
from adult human fibroblasts by defined factors. Cell 2007;131:861–872.
31. Hugo H, Ackland ML, Blick T, Lawrence MG, Clements JA, Williams ED, Thompson EW. Epithelial-mesenchymal and
mesenchymal–epithelial transitions in carcinoma progression. J Cell Physiol 2007;213:374–383.
32. Peinado H, Olmeda D, Cano A. Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial
phenotype? Nat Rev Cancer 2007;7:415–428.
33. Gupta PB, Mani S, Yang J, Hartwell K, Weinberg RA. The evolving portrait of cancer metastasis. Cold Spring Harb
Symp Quant Biol 2005;70:291–297.
34. Yang J, Mani SA, Weinberg RA. Exploring a new twist on tumor metastasis. Cancer Res 2006;66:4549–4552.
35. Hendrix MJ, Seftor RE, Seftor EA, Gruman LM, Lee LM, Nickoloff BJ, Miele L, Sheriff DD, Schatteman GC.
Transendothelial function of human metastatic melanoma cells: role of the microenvironment in cell-fate determination.
Cancer Res 2002;62:665–668.
36. Hess AR, Margaryan NV, Seftor EA, Hendrix MJ. Deciphering the signaling events that promote melanoma tumor cell vas-
culogenic mimicry and their link to embryonic vasculogenesis: role of the Eph receptors. Dev Dyn 2007;236:3283–3296.
37. Cozzio A, Passegue E, Ayton PM, Karsunky H, Cleary ML, Weissman IL. Similar MLL-associated leukemias arising
from self-renewing stem cells and short-lived myeloid progenitors. Genes Dev 2003;17:3029–3035.
38. Huntly BJ, Shigematsu H, Deguchi K, Lee BH, Mizuno S, Duclos N, Rowan R, Amaral S, Curley D, Williams IR,
Akashi K, Gilliland DG. MOZ-TIF2, but not BCR-ABL, confers properties of leukemic stem cells to committed murine
hematopoietic progenitors. Cancer Cell 2004;6:587–596.
39. Somervaille TC, Cleary ML. Identification and characterization of leukemia stem cells in murine MLL-AF9 acute
myeloid leukemia. Cancer Cell 2006;10:257–268.
40. Rangarajan A, Weinberg RA. Opinion: Comparative biology of mouse versus human cells: modelling human cancer in
mice. Nat Rev Cancer 2003;3:952–959.
41. Dontu G, El Ashry D, Wicha MS. Breast cancer, stem/progenitor cells and the estrogen receptor. Trends Endocrinol
Metab 2004;15:193–197.
42. Kalirai H, Clarke RB. Human breast epithelial stem cells and their regulation. J Pathol 2006;208:7–16.
43. Kelly PN, Dakic A, Adams JM, Nutt SL, Strasser A. Tumor growth need not be driven by rare cancer stem cells. Science
2007;317:337.
44. Hill RP. Identifying cancer stem cells in solid tumors: case not proven. Cancer Res 2006;66:1891–1895.
45. Kuperwasser C, Chavarria T, Wu M, Magrane G, Gray JW, Carey L, Richardson A, Weinberg RA. Reconstruction of
functionally normal and malignant human breast tissues in mice. Proc Natl Acad Sci USA 2004;101:4966–4971.
46. Scadden DT. The stem cell niche in health and leukemic disease. Best Pract Res Clin Haematol 2007;20:19–27.
47. Gilbertson RJ, Rich JN. Making a tumour’s bed: glioblastoma stem cells and the vascular niche. Nat Rev Cancer
2007;7:733–736.
14 Part I / Introduction to Cancer Stem Cells
48. Baguley BC. Tumor stem cell niches: a new functional framework for the action of anticancer drugs. Recent Patents
Anticancer Drug Discov 2006;1:121–127.
49. Yang ZJ, Wechsler-Reya RJ. Hit ‘em where they live: targeting the cancer stem cell niche. Cancer Cell 2007;11:3–5.
50. Hoelzinger DB, Demuth T, Berens ME. Autocrine factors that sustain glioma invasion and paracrine biology in the brain
microenvironment. J Natl Cancer Inst 2007;99:1583–1593.
51. Fodde R, Brabletz T. Wnt/beta-catenin signaling in cancer stemness and malignant behavior. Curr Opin Cell Biol
2007;19:150–158.
52. Dontu G, Wicha MS. Survival of mammary stem cells in suspension culture: implications for stem cell biology and
neoplasia. J Mammary Gland Biol Neoplasia 2005;10:75–86.
53. Liu S, Dontu G, Wicha MS. Mammary stem cells, self-renewal pathways, and carcinogenesis. Breast Cancer Res
2005;7:86–95.
54. Hosen N, Park CY, Tatsumi N, Oji Y, Sugiyama H, Gramatzki M, Krensky AM, Weissman IL. CD96 is a leukemic stem
cell-specific marker in human acute myeloid leukemia. Proc Natl Acad Sci USA 2007;104:11008–11013.
55. Katoh M. Networking of WNT, FGF, Notch, BMP, and Hedgehog signaling pathways during carcinogenesis. Stem Cell
Rev 2007;3:30–38.
56. Tung DC, Chao KS. Targeting hedgehog in cancer stem cells: how a paradigm shift can improve treatment response.
Future Oncol 2007;3:569–574.
57. Cho RW, Wang X, Diehn M, Shedden K, Chen GY, Sherlock G, Gurney A, Lewicki J, Clarke MF. Isolation and molecu-
lar characterization of cancer stem cells in MMTV-Wnt-1 murine breast tumors. Stem Cells 2008;26:364–371.
58. Korkaya H, Wicha MS. Selective targeting of cancer stem cells: a new concept in cancer therapeutics. BioDrugs
2007;21:299–310.
59. Dontu G, Jackson KW, McNicholas E, Kawamura MJ, Abdallah WM, Wicha MS. Role of Notch signaling in cell-fate
determination of human mammary stem/progenitor cells. Breast Cancer Res 2004;6:R605–R615.
60. Bailey JM, Singh PK, Hollingsworth MA. Cancer metastasis facilitated by developmental pathways: Sonic hedgehog,
Notch, and bone morphogenic proteins. J Cell Biochem 2007;102:829–839.
61. Rizzo P, Osipo C, Foreman KE, Miele L. Rational targeting of Notch signaling in cancer. Oncogene 2008;27:5124–31.
62. Shih AH, Holland EC. Notch signaling enhances nestin expression in gliomas. Neoplasia 2006;8:1072–1082.
63. Phillips HS, Kharbanda S, Chen R, Forrest WF, Soriano RH, Wu TD, Misra A, Nigro JM, Colman H, Soroceanu L,
Williams PM, Modrusan Z, Feuerstein BG, Aldape K. Molecular subclasses of high-grade glioma predict prognosis, deline-
ate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell 2006;9:157–173.
64. Fan X, Matsui W, Khaki L, Stearns D, Chun J, Li YM, Eberhart CG. Notch pathway inhibition depletes stem-like cells
and blocks engraftment in embryonal brain tumors. Cancer Res 2006;66:7445–7452.
65. Reedijk M, Odorcic S, Chang L, Zhang H, Miller N, McCready DR, Lockwood G, Egan SE. High-level coexpression
of JAG1 and NOTCH1 is observed in human breast cancer and is associated with poor overall survival. Cancer Res
2005;65:8530–8537.
66. Dickson BC, Mulligan AM, Zhang H, Lockwood G, O’Malley FP, Egan SE, Reedijk M. High-level JAG1 mRNA and
protein predict poor outcome in breast cancer. Mod Pathol 2007;20:685–693.
67. Zang S, Ji C, Qu X, Dong X, Ma D, Ye J, Ma R, Dai J, Guo D. A study on Notch signaling in human breast cancer.
Neoplasma 2007;54:304–310.
68. Farnie G, Clarke RB, Spence K, Pinnock N, Brennan K, Anderson NG, Bundred NJ. Novel cell culture technique for
primary ductal carcinoma in situ: role of Notch and epidermal growth factor receptor signaling pathways. J Natl Cancer
Inst 2007;99:616–627.
2 Tumor Stem Cells and Malignant Cells,
One and the Same
Beverly A. Teicher
Abstract
Cancer is a proliferative, invasive, and metastatic disease often caused by repeated tissue insults result-
ing in accumulation of genetic abnormalities that rarely produce malignant cells. The survival of mouse
L1210 leukemia was determined for inoculations of 1 cell up to 106 cells. The survival times varied in
a log-linear manner with the inoculum cell number from 19 days with 1 cell to 7 days with 106 cells
implanted. In preclinical tumor models or in patients, tumor nodules of 108–109 cells are advanced can-
cer. Malignant cells frequently secrete growth modulatory substances that regulate their growth and alter
growth of normal cells. Whether the metastatic malignant cell is the same or significantly different from
the primary lesion malignant cell remains a topic of active investigation. Reaching a detectable lesion takes
10 years. Genetic instability produces variants in the primary tumor and metastases that are more hetero-
geneous than the early disease. The argument that cancer arises only from the tissue stem cell populations
and that cancer stem cells comprise perhaps 1 in 100,000 or 1 in 10,000 cells within the tumor leads to
the notion that agents that selectively kill cancer stem cells will not decrease the tumor mass. The cells
that initiate, sustain, and populate cancers are malignant cells. Cancer stem cell notion is useful if it leads
to important research questions and to better therapeutics.
Key Words: Colony forming units, Malignant cells, Genetic instability, Metastasis, L1210 leukemia
INTRODUCTION
Cancer is a proliferative, invasive, and metastatic disease that is frequently caused by repeated
insults to a tissue resulting in accumulation of genetic abnormalities that, by rare chance, produce a
malignant cell. Cancer cells are genetically aberrant and instable. Some cancers begin as a single
clone (and a few remain clonal) and other arise from a field of repeatedly damaged cells. The search
for an understanding of cancer and for the key as to how to control and ablate malignant disease often
returns to the remarkable processes of normal tissue/embryo development and normal tissue repair.
The “well-behaved” proliferative and self-limiting biology of wound repair, gut lining replacement,
liver regeneration, skin renewal, and bone marrow generation of hematopoietic cells has taught us that
cell proliferation and differentiation are a constant process in complex organisms and are well-con-
trolled under normal circumstances.
The concept of a stem cell was put forth by Till and McCulloch to describe the ability of a single
mouse bone marrow cell to produce a colony of cells in the mouse spleen and later to describe the
From: Cancer Drug Discovery and Development: Stem Cells and Cancer,
Edited by: R.G. Bagley and B.A. Teicher, DOI: 10.1007/978-1-60327-933-8_2,
© Humana Press, a part of Springer Science + Business Media, LLC 2009
15
16 Part I / Introduction to Cancer Stem Cells
ability of similar single bone marrow cells to give to colonies of varied types in cell culture (1, 2). A
colony-forming unit (CFU) is an individual cell that is able to clone itself into a colony of identical
cells. A CFU is a measure of viable bacterial numbers or a measure of viable mammalian malignant
cells in a culture. In reconstituting the immune system of lethally irradiated mice, bone marrow cells
from syngeneic donors are intravenously injected into the recipient animals and colonies form in the
spleen. Each colony is the progeny of a pluripotent stem cell; therefore, the number of colonies is a
measure of the number of stem cells. These findings led to the notion that cancer can arise from mul-
tiply insulted cells that by rare chance have aberrantly turned on genes that normally are expressed only
by normal tissue “stem” cells. Thus, cancer cells have aberrantly reverted to a dedifferentiated prolif-
erative state. These malignant cells are trying to build a tissue but they are abnormal and lethal.
Indeed, an area of therapeutic investigation has a goal to discover agents that can terminally dif-
ferentiate malignant cells to a quiescent nonproliferative state.
EARLY OBSERVATIONS
An interesting aspect of the current cancer stem cell debate regards the number of human tumor
cells required to initiate the growth of a subcutaneous nodule in immunodeficient mice. A very large
number of variables would need to be optimized to achieve reliable data from such observations. A
historical perspective looking at syngeneic mouse tumors may help. The L1210 and P388 mouse
leukemias were developed in 1948 and 1955, respectively (3–5). L1210 and P388 leukemias were
both chemically induced in a DBA/2 mouse by painting the skin with methylcholanthrene. The leuke-
mias have been propagated in DBA/2 mice by implanting intraperitoneally 0.1 mL of a diluted ascetic
fluid containing either 105 L1210 cells or 106 P388 cells. These mouse leukemias were the first tumors
used for large-scale drug discovery screening programs by the national drug development program
instituted in 1954 by Congress, which directed the National Cancer Institute to start a program. The
Cancer Chemotherapy National Service Center (CCNSC) screen consisted of three mouse tumors:
L1201 leukemia, SA-180 sarcoma, and mammary adenocarcinoma 755 (6). Over the years, the pri-
mary screen varied from the original three tumors to L1210 plus two arbitrarily selected tumors to
L1210 plus Walker 256 carcinosarcoma to L1210 plus P388 leukemia to L1210 plus B16 melanoma
or Lewis lung carcinoma. In 1976, a change occurred in the NCI primary screen. The new screen
included a panel of colon, breast, and lung tumor models (mouse and human); however, compounds
were initially screened in P388 leukemia (7).
Skipper and Schabel and colleagues explored the growth characteristics of the L1210 and P388 leuke-
mias in mice (8, 9). The testing was conducted in a hybrid of DBA/2 hosts. Tumor cell implant sites
were intraperitoneal injection, subcutaneous implant, intravenous injection, and intracranial injection.
For L1210 leukemia with an inoculum of 105 cells, the mean days of survival and tumor cell doubling
times for these implant sites were 8.8, 9.9, 6.4, and 7.0 days and 0.34, 0.46, 0.45, and 0.37 days, respec-
tively (Fig. 1). The mean survival times of mice implanted with L1210 cells by these various routes was
determined for inoculations of 1 cell up to 106 cells. The survival times varied in a log-linear manner
with the inoculum cell number. Thus, when the mice were implanted with 1 L1210 cell by intraperito-
neal injection, they survived 19 days and when the mice were implanted with 106 L1210 cells intraperi-
toneally, they survived 7 days (Fig. 1). Similar studies were conducted with P388 leukemia. For P388
leukemia (106 cells), the mean days of survival and the tumor doubling times for the same implant sites
were 10.3, 13.0, 8.0, and 8.0 days and 0.44, 0.52, 0.68, and 0.63 days, respectively. From these studies,
it must be concluded that every L1210 and P388 cell is a cancer stem cell.
Skipper and Schabel applied similar analyses to solid tumors especially the mouse Ridgway osteo-
genic sarcoma (10). In preclinical tumor models or in patients, tumor nodules of 108–109 cells are
Chapter 2 / Tumor Stem Cells and Malignant Cells, One and the Same 17
1e+6
1e+4
1e+3
1e+2
1e+1 IP IMPLANT
IV IMPLANT
IC IMPLANT
1e+0
0 2 4 6 8 10 12 14 16 18 20
Fig. 1. Mean survival times of mice inoculated with various numbers of murine L1210 leukemia cells injected intra-
peritoneally, intravenously, or intracranially. These data form the basis for the in vivo bioassay method for determin-
ing the number of L1210 cells surviving after treatment of L1210 tumor-bearing mice with therapy. From these
survival curves, it was determined that from: (1) intraperitoneal inoculation of L1210 cell-generation time = 0.55
days; the lethal number of L1210 cells = 1.5 × 109; (2) intravenous inoculation the L1210 cell-generation time = 0.43
days; and (3) intracranial inoculation the L1210 cell-generation time = 0.46 days (8,9).
advanced cancer (Fig. 2) (10, 11). One source of variability in the response of drug-sensitive tumor
cells to a drug is the heterogeneity of the blood supply such that the drug does not reach the tumor
cells distal from the blood supply in sufficient concentration to be lethal. Thus, the pharmacokinetics
and concentration of a drug required to kill tumor cells distal from vasculature should be documented.
In addition, the physiologic heterogeneity of tumor masses as a source of varied treatment response,
Skipper and Schabel considered the heterogeneity of tumor stem cells, defined as cells capable of unlim-
ited proliferative thrust, caused by the inherent genetic instability of malignant cells to be a source of variable
treatment response. Skipper and Schabel considered various types of tumor stem cells that might account
for fluctuation in response to chemotherapy in similarly treated individuals bearing a specific cancer, and
classifications of cancers by chemotherapeutic effect. Fluctuating ratios of treatment responsive to treat-
ment resistant stem cells, as predicted by the mutation theory, could account for one patient responding
to a drug and the next not responding. Differences in tumor growth fraction and differences in tumor
distribution into pharmacologic sanctuaries could also strongly influence a patient’s response to therapy.
Treatment resistant stem cells are primarily responsible for the failure of the best available chemotherapy
to cure responsive, refractory, and very refractory experimental neoplasms. These data examined suggest
that differences in the resistant to responsive stem cell ratios in different types of cancer may account for
their being classified as responsive, refractory, or very refractory (12).
18 Part I / Introduction to Cancer Stem Cells
1.00E+14
Tumor Mass
1.00E+13
1012 = 1 kg
1.00E+12
1.00E+11
1010 = 10 g
1.00E+10
Number of Tumor Cells
109 = 1 g
1.00E+09
1.00E+08 108 = 130 mg
1.00E+07
106 = 1 mg
1.00E+06
1.00E+05
1.00E+04
1.00E+03
1.00E+02
1.00E+01
1.00E+00
0 5 10 15 20 25 30 35 40 45 50
Number of Population Doublings
Fig. 2. Tumor cell numbers and weight of the tumor mass are shown. In patients, tumors are advanced at first
presentation or at recurrence after initial noncurative therapy (10,11).
Autocrine Factors
Paracrine Factors
Malignant Cell
Cancer Stem Cell
Blood Vessel
Cells mobilized
from distal sites
Vascular Cells
Stromal Cells
Fig. 3. Malignant cells can have abscopal effects on the host through secretion of paracrine factors and can produced
autocrine factors that can sustain proliferation of the malignancy.
HEMATOPOIESIS AS A MODEL
Analogies for the development of malignancy have been sought in the processes of normal aging
and in the differentiation of cells in the hematopoietic system (22, 23). The incidence of many cancers
increase with age because of increased probability of DNA changes that may allow occurrence of a
malignant cell and because some of the alterations associated with normal aging increase the suscep-
tibility of cells to carcinogenic events. In normal aging, there is a decrease in DNA repair capacity
and a decline in cellular immune reactivity that could contribute to permitting malignant growth (22).
Normal hematopoiesis, the formation of the many cell types in blood, is a process of development,
self-renewal through mitosis, and differentiation of hematopoietic stem cells, the source cell of all
blood cell lineages (24). Because most blood cells have relatively short lifespans, hematopoietic stem
cells continuously replicate themselves through self-renewal to prevent depletion of the stem cell
pool while simultaneously differentiating into multiple lineages of the varied blood cell types. The
fate choice of hematopoietic cells to either self-renew or differentiate is controlled by intrinsic mecha-
nisms and extrinsic signals from the environment or the stem cell niche (25). In adults, the hematopoi-
etic stem cell number is relatively constant under normal conditions. Bone marrow hematopoietic
stem cells appear quiescent; however, the majority divide regularly as shown by their slow constant
incorporation of radio-labeled nucleotides (26, 27). There are two proposed mechanisms by which
asymmetric cell division may be achieved called divisional asymmetry and environmental asymmetry.
In divisional asymmetry, specific cell fate determinants in the genome, RNA, and proteins are distrib-
uted unequally during cell division. After cell division, only one daughter cell receives the deter-
minants, thus retaining the hematopoietic stem cell fate while the other daughter differentiates.
20 Part I / Introduction to Cancer Stem Cells
In environmental asymmetry, one hematopoietic stem cell niche and retains the stem cell identity,
while the other enters a different environment favoring its differentiation (24).
The first human leukemia-lymphoma cell lines were Burkitt’s lymphoma lines developed in 1963
(28). The most widely used of these lines is the Raji Burkitt’s lymphoma (29). The term “cell line”
indicates that the population of cells grew continuously in culture (to this day); therefore, the cells were
“immortal” or capable of continuous self-renewal. Some the leukemia-lymphoma cell lines were pre-
sumed to be monoclonal that is derived from one malignant cell and that the current multiclonal lines
emerged during extended culture. The differentiation of the cells arrested at a discrete stage during
maturation of the lineage. The early cell lines were grown autonomously in basic nutrient media sup-
plemented with fetal calf (or other serum) serum independent of external growth factors, although, in
some cases, exogenously added growth factors could stimulate proliferation. Later cell lines were
selected to be growth factor dependent (30). Nearly all of the established leukemia and lymphoma cell
lines are genetically abnormal. Of 429 lines analyzed only two had normal karyotypes (31). In addition
to gross cytogenetic alterations, many of the lines harbor point mutations deletions and amplifications
of specific genes (32). However, despite the evidence of genetic instability and abnormality, these cell
lines have remained “stable” in long-term culture for nearly 50 years.
An interesting case is the impact of stem cell “dose” on hematopoietic recovery in autologous
blood stem cell recipients (23). Hematopoietic cells collected from blood and reinfused into patients
following high-dose chemotherapy are generally termed “stem cells,” even though the population of
cells contains true stem cells and differentiated progenitor cells (33, 34). Mobilized blood stem cell
quantity, identified by expression of CD34, may be the strongest predictor of days to hematopoietic
recovery (i.e., platelets and neutrophils) in autologous blood stem cell recipients (35). The majority
of patients will recover if a stem cell dose of ³5 × 106/kg stem cells are infused, which corresponds
to 3.5 × 108 cells (36). Bone marrow engraftment is the result of early undifferentiated stem and pro-
genitor cell self-renewal and differentiation. The rate of engraftment is dependent, in part, on the
number and type of stem cells infused. Using a genetically engineered model cell to represent human
leukemia stem cells, Hope et al. tracked these human leukemia stem cells in SCID mice and found
that the leukemia stem cells were not functionally homogeneous (37). Like normal hematopoietic
stem cells, some human leukemia stem cells divided rarely and underwent self-renewal rather than
commitment after cell division and others demonstrated heterogeneity in self-renewal potential.
METASTASIS
The detection of circulating tumor cells in the blood and lymph system and micrometastases has
been investigated through multiple eras of cancer research because of the potential importance of
these cells to prognosis and therapeutic approaches (38). The first publication of circulating tumor
cells was in 1869 when Ashworth reported cancer cells in the blood of a patient at autopsy (39). In
the decade 1955–1965, 5,000 cancer patients were tested for circulating tumor cells using 20 different
cytological methods; however, it was finally realized that the tests used were not sufficiently discrimi-
natory (40). When immunohistochemical detection methods were developed and more recently with
the development of PCR techniques, interest in documenting a connection between circulating tumor
cell numbers and stage of disease and in detecting occult metastases renewed (41–43). Although the
accuracy of methods for detecting tumor cells in circulation has markedly improved over the years,
the application of these data to determination of the most appropriate treatment regimen for individual
patients has yet to occur. It is not clear whether circulating tumor cells are enriched for cancer “stem
cells,” malignant cells that can initiate metastasis or whether circulating tumor cells reflect the quality
of the primary lesion vasculature or other properties of the malignant disease (44, 45). There is no
doubt that malignant cells must be plastic and adaptive to altering environments and stress. The capacity
Chapter 2 / Tumor Stem Cells and Malignant Cells, One and the Same 21
Clonal Selection
Parallel Evolution
Dynamic
Heterogeneity
Clonal Dominance
Stem Cell
Fig. 4. The various patterns of cell division that can lead to the cellular heterogeneity found in most malignant disease.
of some malignant cells to make epithelial to mesenchymal transition and then return to an epithelial
phenotype is an example of the plasticity of malignant cells (44). During wound healing processes
normal cells can undergo these changes as well.
Metastasis is a critical characteristic of malignant disease (45) (Fig. 4). The placement of a cancer stem
cell in the metastatic process and understanding whether the metastatic malignant cell is the same or
significantly different from the malignant cell of the primary lesion remains a topic of active investigation.
The clonal selection hypothesis is that cell populations with metastatic capacity are subpopulations within
the primary lesion. The parallel evolution model theorizes that metastasis occurs early in malignant dis-
ease and evolves along a different path than the primary lesion. The dynamic heterogeneity hypothesis
indicates that metastatic variants arise within the primary tumor, and these variants spread and evolve
further at secondary sites. The notion of clonal dominance is that more virulent metastatic subclones
occur within the primary tumor and outgrow the original malignant cells and eventually dominate the
primary lesion and metastases. The stem cell model indicates that only cancer stem cells and not the
majority population of the tumor have the capacity to metastasize and establish distant lesions (46–48).
Gatenby and Gillies proposed a model for the somatic evolution of invasive cancer as overcoming
a series of barriers to proliferation (49). Tumor development and the genotypic and phenotypic hetero-
geneity of cancer cell populations is described using an equivalence principle such that multiple
alterations in the cell may allow to successfully adapt to and overcome a critical barrier moving
toward malignancy. During tumor initiation, progression and metastasis, approximately 30 generation
22 Part I / Introduction to Cancer Stem Cells
Primary
Tumor
Doublings 0 3 30 40
Tumor cells 1 8 109 1012
Time 0 1yr 10yr 13yr
Metastases
Metastasis Primary
Tumor
1 cm tumor
Fig. 5. Two possible patterns of malignant tumor growth and spread of metastatic disease over time and malignant
cell doublings is shown.
times or about 10 years are required for a single malignant cell to produce a palpable mass of about
1 cm3 composed of about 109 tumor cells (45) (Fig. 5). Over the course of these 10 years to reach a
clinically detectable lesion, genetic instability produces variants in the primary tumor and metastases
that are more heterogeneous than the early disease. A lethal tumor burden of 1012 tumor cells could
be expected after 10 cell doublings or 3 years from detection of the disease without treatment.
CARCINOGENESIS
Chemical carcinogen-induced rat mammary tumors provided an in vivo model for the early cancer
stem cell hypothesis (50). In the rat mammary gland, epithelial stem cells insulted by a chemical
targeting DNA can give rise to more-rapidly proliferating benign stem cells, which retain some
response to normal growth-controlling factors. These altered but benign stem cells can also differenti-
ate to a degree. The genetic instability of these cells results in the generation of malignant cells in
subsequent generations with loss of response to growth-controlling factors, decreased capacity for
differentiation and escape from immune surveillance. The cells which then compose the malignant
disease have departed in critical ways from the normal epithelial stem cells of origin and continue to
suffer from genetic instability and to evolve.
Colony formation is a demonstration of self-renewal capacity. It was noted early that colony-
forming human epidermal cells were heterogeneous in capacity for sustained growth and that the
potential for continued growth from a single cell could be predicted from the phenotype of the colony
produced (51). Three types of colonies were described: (1) holocolonies characterized as large with a
smooth perimeter containing mainly small cells that may be considered the most undifferentiated
population; (2) paracolonies characterized as being small with a highly irregular perimeter containing
cells that are large and flattened and appear terminally differentiated; and (3) merocolonies characterized
Chapter 2 / Tumor Stem Cells and Malignant Cells, One and the Same 23
by medium to large size and a wrinkled perimeter with heterogeneous cells. In the case of normal cells,
the transitions from holoclone (stem-like) to meroclone to paraclone (terminally differentiated) are
thought to be unidirectional and result in progressively restricted growth potential. However, malig-
nant cells have increased plasticity and may dedifferentiate toward holoclone states or abnormally
differentiate forming giant multinucleated cells that no longer proliferate. Locke et al. studied seven
well-established human cancer cell lines from varied histology and found that cell lines generated
from carcinomas produced cell culture colony patterns similar to those produced by the stem and
amplifying cells of normal epithelia as described earlier (52). The cancer cell lines appeared to main-
tain a subpopulation of “stem cells” during passage of the line. The heterogeneity of the colonies
produced from the cancer cell lines indicates that the stem cell property of asymmetrical division
persists in cancer cell lines but is shifted toward stem cell self-renewal. Thus, malignant cells attempt
to recapitulate the behavior of normal tissue cells; however, their behavior is aberrant and detrimental.
Malignant cells are damaged cells that have dedifferentiated, returned to the behavior of embryonic
cells rather than dying or being recognized as abnormal by the immune system.
The skin epidermis is a normal tissue that is continuously confronted with physiochemical traumas
from the environment (53). The skin undergoes continual rejuvenation through homeostasis and
is primed to undergo wound repair in response to injury. The epidermis has tissue “stem cells,”
which both self-perpetuate and give rise to the differentiating cells that constitute the tissue. The
TGFb pathway is involved in the control of proliferation of many cells. The loss of response to TGFb
proliferation control is a necessary step in malignant transformation (54–56). In the absence of
TGFb, proliferation control, squamous cell carcinomas spontaneously develop in mice (57). These
tumors have many of the characteristics of invasive squamous cell carcinomas.
Although some may argue that the embryological, stem-like properties of tumors and tumor cell
lines indicate that the malignant cell originates only from tissue stem cells, this argument seems
unnecessary to explain the dedifferentiated properties of the malignant cell (58). The argument that
cancer arises only from the stem cell populations of tumor that comprise perhaps 1 in 100,000 or 1 in
10,000 cells within the tumor leads to the notion that treatment agents that selectively kill the cancer
stem cell will not result in decrease in the tumor mass. Classic studies and recent studies show that
with some tumor cell lines inoculation of mice with few cells from 1 to 10 can produce tumors that
are lethal to the host (59). Most transplantable tumor models are carried out with the implant for
105–107 tumor cells primarily due to the impatience of the experimentalist, who would like to see a
palpable nodule in 1 or 2 weeks and not in several months or years. Generally, if a tumor nodule is
not palpable in 2 or 3 weeks, the tumor is considered a no take. The timeline for these studies is that
of the experimentalist not reflective of the nature of cell proliferation. Although it appears that only a
minute proportion of human tumor cells can grow readily and rapidly in mice, syngeneic tumor mod-
els indicate that inoculums of many fewer cells that divide quite rapidly in the murine host yield lethal
malignant disease in a short time (60).
CONCLUSIONS
In most incarnations, the cancer stem cell is very similar to a normal tissue stem cell. The cancer
stem hypothesis has been advanced to describe how a cancer originates, how it is sustained, and what
makes it drug resistant. Among the properties of tissue stem cells in the adult are the relative quies-
cence manifested by infrequent cell divisions, resistance to drugs/toxins mediated by ABC transport-
ers, active DNA repair pathways and resistance to apoptosis. The cancer stem cell has been defined
as a cell involved in malignant disease process that possess the capacity to self-renew and give rise to
the heterogeneous lineages of cancer cells that comprise the tumor. Whether the malignant cells with
24 Part I / Introduction to Cancer Stem Cells
long term self-renewal capacity that sustain the cancer arose from tissue stem cells that acquired
mutations leading to a differentiation block and malignancy or whether more differentiated cells
acquired mutations that dedifferentiated them and resulted in a malignant cell capable of self-renewal
has not been determined. Thus, the use of the term “stem cell” may not be accurate. The cells that
initiate, sustain, and populate cancers are malignant cells (61, 62). The notion of the cancer stem cell
is useful, if it leads important research questions being addressed and to better therapeutics being
developed. The self-renewal, pluripotency, and drug resistance of malignant cells is a product of their
abnormality and genetic instability.
REFERENCES
1. Till JE, McCulloch EA. Early repair processes in marrow cells irradiated and proliferating in vivo. Radiat Res 1963; 18:
96–105.
2. McCulloch EA, Till JE. Perspectives on the properties of stem cells. Nature Med 2005; 11: 10276–8.
3. Law LW, Dunn TB, Boyle PJ, Miller JH. Observations on the effect of a folic acid antagonist on transplantable lymphoid
leukemias in mice. J Natl Cancer Inst 1949; 10: 179–92.
4. Dawe CJ, Potter M. Morphologic and bioloigc progression of a lymphoid neoplasm of the mouse in vivo and in vitro.
Amer J Pathol 1957; 33: 603.
5. Waud WR. Murine L1210 and P388 leukemias. In: Anticancer Drug Development Guide: Preclinical Screening, Clinical
Trials and Approval. BA Teicher Ed. Humana Press Inc, Totowa, NJ 1997; pp59–74.
6. Goldin A, Serpick AA, Mantel NA. A commentary, experimental screening procedures and clinical predictability value.
Cancer Chemother Rep 1966; 50: 173–218.
7. Goldin A, Vendetti JM, Muggia FM, Rozencweig M, DeVita VT. New animal models in cancer chemotherapy. In: Fox
BW (ed.) Advances in Medical Oncology, Research and Education, Vol 5. Basis for Cancer Therapy I. New York:
Pergamon. 1979; 113–22.
8. Skipper HE, Schabel FM, Wilcox WS, Laster WR, Trader MW, Thompson SA. Experimental evaluation of potential
anticancer agents. XVIII. Effects of therapy on viability and rate of proliferation of leukemia cells in various anatomic
sites. Cancer Chemother Reps 1965; 47: 41–65.
9. Schabel FM Jr, Griswold DP Jr, Laster WR Jr, Corbett TH, Lloyd HH. Quantitative evaluation of anticancer agent activ-
ity in experimental animals. Pharmacol Ther (A) 1977; 1: 411–435.
10. Schabel FM Jr, Griswold DP Jr, Corbett TH, Laster WR Jr. Increasing the therapeutic response rates to anticancer drugs
by applying the basic principles of pharmacology. Cancer 1984; 54(6 suppl): 1160–7.
11. De Vita VT Jr, Young RC, Canellos GP. Combination versus single agent chemostherapy: a review of the basis for selec-
tion of drug treatment of cancer. Cancer 1975; 35: 98–110.
12. Skipper HE, Schabel FM Jr. Tumor stem cell heterogeneity: implications with respect to classification of cancers by
chemotherapeutic effect. Cancer Treat Reps 1984; 68: 43–61.
13. Martin DS, Balis ME, Fisher B, Frei E, Freireich EJ, Heppner GH, Holland JF, Houghton JA, Houghton PJ, Johnson
RK, Mittelman A, Rustum Y, Sawyer RC, Schmid FA, Stolfi RL, Young CW. Role of murine tumor models in cancer
treatment research. Cancer Res 1986; 46: 2189–92.
14. Nicolson GL. Tumor cell instability, diversification and progression to the metastatic phenotype: from oncogene to
oncofetal expression. Cancer Res 1987; 47: 1473–87.
15. Foulds L. Neoplastic Development. New York: Academic Press, 1975.
16. Nowell PC. The clonal evolution of tumor cell populations. Science 1976; 194: 23–8.
17. Nowell PC. Mechanisms of tumor progression. Cancer Res 1986; 46: 2203–7.
18. Heppner GH, Miller BE, Miller FR. Tumor subpopulation interactions in neoplasms. Biochim Biophys Acta 1984; 695:
215–26.
19. Miller FR. Intratumor immunologic heterogeneity. Cancer Metastasis Rev 1982; 1: 319–34.
20. Miller FR. Tumor subpopulations intereactions in metastasis. Invasion Metastasis 1983; 3: 234–42.
21. Walsh JH, Karnes WE, Cuttitta F, Walker A. Autocrine growth factors and solid tumor malignancy. West J Med 1991;
155: 152–63.
22. Ebbesen P. Cancer and normal ageing. Mech Ageing Develop 1984; 25: 269–83.
23. Pecora AL. Impact of stem cell dose on hematopoietic recovery in autologous blood stem cell recipients. Bone Marrow
Transplant 1999; 23 (suppl 2): S7–S12.
24. Huang X, Cho S, Spangrude GJ. Hematopoietic stem cells: generation and self-renewal. Cell Death Differentiation
2007; 14: 1851–9.
Chapter 2 / Tumor Stem Cells and Malignant Cells, One and the Same 25
25. Moore KA, Lemischka IR. Stem cells and their niches. Science 2006; 311: 1880–5.
26. Bradford GB, Williams B, Rossi R, Bertoncello I. Quierscence, cycling and turnover in the primitive hematopoietic stem
cell compartment. Exp Hematol 1997; 25: 445–53.
27. Cheshier SH, Morrison SJ, Liao X, Weissman IL. In vivo proliferation and cell cycle kinetics of long-term self-renewing
hematopoietic stem cells. Proc Natl Acad Sci USA 1999; 96: 3120–5.
28. Drexler HG, Matsuo Y, MacLeod AF. Continuous hematopoietic cell lines as model systems for leukemia-lymphoma
research. Leukemia Res 2000; 24: 881–911.
29. Pulvertaft RJV. Cytology of Burkitt’s tumor (African lymphoma). Lancet 1964; i: 238–40.
30. Drexler HG, Zaborski M, Quentmeier H. Cytokine response profiles of human myeloid factor-dependent human leukemia
cell lines. Leukemia 1997; 11: 701–8.
31. Dexler HG (editor). The Leukemia-lymphoma Cll Lines Factsbook. San Diego, CA: Academic Press, 2000.
32. Drexler HG, Fombonne S, Matsuo Y, Hu ZB, Hamaguchi H, Uphoff CC .P53 alterations in human leukemia-lymphoma
cell lines: in vitro artifact or prerequisite for cell immortalization ? Leukemia 2000; 14: 198–206.
33. Krause DS, Fackler MJ, Civin CI, Stratford May W. CD34: structure, biology and clinical utility. Blood 1996; 87: 1–13.
34. Berardi AC, Wang A, Levine JD, Lopez P, Scadden DT. Functional isolation and characterization of human hematopoi-
etic stem cells. Science 1995: 267: 104–8.
35. Furness SGB, McNagny K. Beyond mere markers: functions for CD34 family of sialomucins in hematopoiesis. Immunol
Res 2006; 34: 13–32.
36. Pecora AL, Preti RA, Gleim GW, Jennis A, Zahos K, Cantwell S, Doria L, Isaacs R, Gillio AP, Michelis MA, Brochstein
JA. CD34+CD33− cells influence days to engraftment and transfusion requirements in autologous blood stem cell
recipients. J Clin Oncol 1998; 16: 2093–104.
37. Hope KJ, Jin L, Dick JE. Acute myeloid leukemia originates from a hierarchy of leukemia stem cell classes that differ
in self-renewal capacity. Nature Immunol 2004; 5: 738–43.
38. Ghossein RA, Bhattacharya S, Rosai J. Molecular detection of micrometastases and circulating tumor cells in solid
tumors. Clin Cancer Res 1999; 5: 1950–60.
39. Ashworth TR. A case of cancer in which cells similar to those in the tumors were seen in the blood after death.
Australian Med J 1869; 14: 146.
40. Christopherson W. Cancer cells in the peripheral blood: a second look. Acta Cytol 1965; 9: 169–74.
41. Moss TJ, Sanders DG. Detection of neuroblastoma cells in blood. J Clin Oncol 1990; 8: 736–40.
42. Pelkey TJ, Frierson HF, Bruns DE. Molecular and immunological detection of circulating tumor cells and micrometas-
eses from solid tumors. Clin Chem 1996; 42: 1369–81.
43. Campana D, Pui CH. Detection of minimal residual disease in acute leukemias: methodological advances and clinical
significance. Blood 1995; 85: 1416–34.
44. Scheel C, Onder T, Karnoub A, Weinberg RA. Adaptation versus selection: the origins of metastatic behavior. Cancer
Res 2007; 67: 11476–80.
45. Talmadge JE. Clonal selection of metastasis within the life history of a tumor. Cancer Res 2007; 67: 11471–5.
46. Gray JW. Evidence emerges for early metstasis and parallel evolution of primary and meststatic tumors. Cancer Cell
2003; 4: 4–6.
47. Schmidt-Kittler O, Ragg T, Daskalakis A, Granzow M, Ahr A, Blankenstein TJ, Kaufmann M, Diebold J, Arnholdt H,
Mullor P, Bischoff J, Harich D, Schlimok G, Riethmuller G, Eils R, Klein CA. From latent disseminated cells to overt
metastasis: genetic analysis of systemic breast cancer progression. Proc Natl Acad Sci USA 2003; 100: 7737–42.
48. Weigelt B, Glas AM, Wessels LF, Witteveen AT, Peterse JL, Van’t Veer LJ. Gene expression profiles of primary breast
tumors maintained in distant metastases. Proc Natl Acad Sci USA 2003; 100: 15901–5.
49. Gatenby RA, Gillies RJ. A microenvironmental model of carcinogenesis. Nature 2008; 8: 56–61.
50. Rudland PS. Stem cells and the development of mammary cancers in experimental rats and in humans. Cancer
Mestastasis Rev 1987; 6: 55–83.
51. Barrandon Y, Green H. Three clonal types of keratinocyte with different capacities for multiplication. Proc Natl Acad
Sci USA 1987; 84: 2302–6.
52. Locke M, Heywood M, Fawell S, Mackenzie IC. Retention of intrinsic stem cell hierarchies in carcinoma-derived cell
lines. Cancer Res 2005; 65: 8944–50.
53. Fuchs E. Skin stem cells: rising to the surface. J Cell Biol 2008; 180: 273–84.
54. Teicher BA. Malignant cells, directors of the malignant process: role of transforming growth factor-beta. Cancer
Metastasis Rev 20: 133–143, 2001.
55. Pinkas J, Teicher BA. TGF-b in cancer and as a therapeutic target. Biochem Pharmacol 72: 523–529, 2006.
56. Teicher BA. Transforming growth factor-b and the immune response to malignant disease. Clin Cancer Res 2007; 13:
6247–51.
26 Part I / Introduction to Cancer Stem Cells
57. Guasch G, Schober M, Pasolli HA, Conn EB, Polak L, Fuchs E. Loss of TGFb signaling destabilizes homeostasis and
promotes squamous cell carcinomas in stratified epithelia. Cancer Cell 2007; 12: 313–27.
58. Wicha MS, Liu S, Dontu G. Cancer stem cells: an old idea – a paradigm shift. Cancer Res 2006; 66: 1883–90.
59. Kelly PN, Dakic A, Adams JM, Nutt SL, Strasser A. Tumor growth need not be driven by rare cancer stem cells. Science
2007; 317: 337.
60. Adams JM, Kelly PN, Dakic A, Nutt SL, Strasser A. Response to comment on: “Tumor growth need not be driven by
rare cancer stem cells”. Science 2007; 318: 1722d.
61. Hill RP, Perris R, “Destemming” cancer stem cells. J Natl Cancer Inst 2007; 99: 1435–40.
62. Hill RP. Identifying cancer stem cells in solid tumors: case not proven. Cancer Res 2006; 66: 1891–6.
II The Stem Cell Niche
3 Mesenchymal Stem Cells in Tumor
Stroma
Abstract
Mesenchymal stem cells (MSC) are defined, minimally, as cells that display a fibroblastic morphology in
cell culture, exhibit a robust self-renewal capacity, and retain the ability to undergo trilineage differentia-
tion into adipocytes, chondrocytes, and osteoblasts. MSC can be isolated from diverse tissues but are most
commonly isolated from red bone marrow. Accumulating evidence suggests that bone marrow MSC can
be mobilized into the periphery to serve as regenerative stem cells at sites of injury and inflammation.
Although the in vivo biology of MSC is poorly understood, several studies have demonstrated that MSC
can be selectively recruited into tumors. Following engraftment within tumor stroma, MSC proliferate
and acquire an activated phenotype similar tumor-associated fibroblasts (TAF). Tumor-homing properties
of MSC have lead to their utility as therapeutic cell-based antitumor protein delivery vehicles. However,
with a greater appreciation for the influential role that the tumor microenvironment can serve during tumor
initiation, promotion, and progression, MSC may enhance tumor progression following acquisition of
TAF-like characteristics. A more comprehensive delineation of the biological role of MSC within tumor
stroma will improve our understanding pf tumor-stroma interactions and facilitate future development of
MSC-based clinical therapies.
Key Words: Mesenchymal stem cell (MSC), Tumor microenvironment, Tumor-associated fibroblast
(TAF), Stromagenesis, Myofibroblast, Desmoplasia, Tumor stroma
INTRODUCTION
Carcinomas are solid tumors that arise from malignant epithelial cells and represent the most com-
mon type of cancer in humans. However, it is becoming increasingly clear that malignant cells are
supported by nonepithelial tumor stroma that shape a given tumor microenvironment. Tumor stroma
can be generally divided into four main components: (1) tumor vasculature, (2) inflammatory immune
cells, (3) extracellular matrix (ECM)/soluble growth factors, and (4) tumor-associated fibroblasts
(TAF) (Fig. 1). Bidirectional paracrine communications between connective tissue fibroblasts and
epithelial cells are vital for normal tissue homeostasis, and a comparable but progressive intimacy
continues throughout tumor development. Although mesenchymal cell fibroblasts (herein referred to
as fibroblasts) can enhance tumor growth and metastasis by means of promoting angiogenesis (1),
epithelial–mesenchymal transition (2), and progressive genetic instability (3, 4), there is also evidence
that healthy tumor microenvironments can act in a dominant manner to inhibit tumor growth (5).
From: Cancer Drug Discovery and Development: Stem Cells and Cancer,
Edited by: R.G. Bagley and B.A. Teicher, DOI: 10.1007/978-1-60327-933-8_3,
© Humana Press, a part of Springer Science + Business Media, LLC 2009
29
Exploring the Variety of Random
Documents with Different Content
back
back
back
back
back
back
back
back
back
back
back
back
back
back
back
back
back
back
back
back
Welcome to our website – the ideal destination for book lovers and
knowledge seekers. With a mission to inspire endlessly, we offer a
vast collection of books, ranging from classic literary works to
specialized publications, self-development books, and children's
literature. Each book is a new journey of discovery, expanding
knowledge and enriching the soul of the reade
Our website is not just a platform for buying books, but a bridge
connecting readers to the timeless values of culture and wisdom. With
an elegant, user-friendly interface and an intelligent search system,
we are committed to providing a quick and convenient shopping
experience. Additionally, our special promotions and home delivery
services ensure that you save time and fully enjoy the joy of reading.
ebookfinal.com