Sie sind auf Seite 1von 15

ASH 50th anniversary review

Stem cell concepts renew cancer research


John E. Dick1
1Division of Cell and Molecular Biology, University Health Network, and Department of Molecular Genetics, University of Toronto, Toronto, ON

Although uncontrolled proliferation is a says. Understanding the molecular and chy with cancer stem cells at the apex.
distinguishing property of a tumor as a cellular basis of this heterogeneity has This review provides a historical over-
whole, the individual cells that make up important implications in the design of view of the influence of hematology on
the tumor exhibit considerable variation therapeutic strategies. The mechanistic the development of stem cell concepts
in many properties, including morphol- basis of tumor heterogeneity has been and their linkage to cancer. (Blood. 2008;
ogy, proliferation kinetics, and the ability uncertain; however, there is now strong 112:4793-4807)
to initiate tumor growth in transplant as- evidence that cancer is a cellular hierar-

Introduction
Tumors from different patients, whether leukemic or solid, exhibit to the development of effective anticancer therapies still exist. It is
significant heterogeneity in terms of morphology, cell surface important to understand the historical underpinnings of the CSC
markers, genetic lesions, cell proliferation kinetics, and response to field to recognize the challenges we still face. By understanding
therapy. Heterogeneity of all these features is also seen within an where we came from, we will have a clearer view of the way
individual tumor that is clonal (ie, initiated from a single cell). forward. The history of stem cell research, and cancer stem cell
Although individual cells of a tumor all share common genetic research in particular, is long and better suited for a book rather
aberrations reflective of their clonal origin, single-cell analysis has than a short review. Thus, I only touch on a few highlights that I
shown the existence of variation in genetic and epigenetic abnor- find significant and that played a role in guiding our own work in
malities between different cells or locations within a tumor. The this field; most of my comments revolve around studies in
cellular and molecular basis for this heterogeneity represents a hematopoiesis and acute myeloid leukemia (AML) specifically. My
fundamental problem that has interested cancer researchers for apologies go to those whose work I was not able to include or could
many decades. One possible explanation is that all tumor cells only be treated in a superficial fashion.
could be biologically equivalent and heterogeneity derives from
extrinsic or intrinsic influences that result in random or stochastic
responses. Alternatively, it is possible that the tumor is a caricature
of normal tissue development and retains hierarchical organization Tumor heterogeneity
with (cancer) stem cells at the apex.1 This review will focus on
50 years of hematology research that played a role in garnering The notion that cancer is a disease of abnormal or uncontrolled
evidence to support the hierarchy model. proliferation has governed cancer research for more than 7 decades.
There is mounting evidence from cell purification studies that a Because normal tissues also undergo continuous regeneration,
subset of cells, termed cancer stem cells (CSCs), or cancer- albeit at rates specific to each organ, the historical thrust of cancer
initiating cells, that are distinct from the bulk of the tumor are research was to develop drugs that targeted the enhanced rate of
responsible for long-term maintenance of tumor growth in several neoplastic proliferation. Clinicians used doses and schedules that
cancers.2 Evidence is strongest for the acute leukemias, although achieved many log orders of cell kill while maintaining acceptable
recent studies have identified the existence of CSCs in an increas- levels of toxicity to normal tissues. Based on a deep understanding
ingly longer list of solid tumors, including brain, breast, and colon.3 of the molecular basis of cancer, current therapeutic strategies now
This conceptual shift has important implications, not only for focus on inhibiting the molecular drivers of cancer (eg, trastuzumab-
researchers seeking to understand mechanisms of tumor initiation Her2/Neu, imatinib-bcr/abl, erlotonib, epidermal growth factor
and progression but also for developing and evaluating effective receptor). With an increased appreciation of the extrinsic microen-
anticancer therapies. Although the clinical relevance of CSCs vironmental factors that support tumor growth, such as the
beyond experimental models is still lacking, the high frequency of vasculature, approaches that interfere with these factors (ie,
relapse after conventional cytotoxic chemotherapies predicts that bevacizumab, vascular endothelial growth factor) are also being
CSCs survive standard treatments. Many papers, reviews, and brought forward. What all these strategies, whether from the past or
funding bodies are making strong predictions that targeting CSCs present, have in common is the tacit perspective of cancer as a
more effectively will lead to improved patient outcomes. Indeed, homogeneous, abnormal entity. Therefore, drugs targeting molecu-
there has been almost breathless excitement that this “new” field of lar lesions should be equally effective against all tumor cells,
CSC biology will solve the stubbornly high relapse rates that barring the emergence of resistant subclones. The goal is to ensure
continue to exist for many cancers. However, stem cell concepts that enough drug(s) is given to target the primary molecular
and how they apply to cancer are decades old, and many obstacles pathway and then combined with drugs that target other relevant or

Submitted August 11, 2008; accepted September 9, 2008. DOI 10.1182/blood- © 2008 by The American Society of Hematology
2008-08-077941.

BLOOD, 15 DECEMBER 2008 䡠 VOLUME 112, NUMBER 13 4793


4794 DICK BLOOD, 15 DECEMBER 2008 䡠 VOLUME 112, NUMBER 13

compensatory pathways to enable killing of every tumor cell. last from weeks to months. In particular, Clarkson, Rubinow,
Indeed, historically, even research to understand the basic biology Skipper and their colleagues inferred that the slow cycling fraction
of cancer uses strategies that treat an individual cancer as function- was actually generating the fast cycling fraction and suggested
ally homogeneous. Much cancer research, even to the present day, these represented a leukemic stem cell (LSC) population because
follows a paradigm where the entire tumor or an entire dish of they had similar cytokinetic properties to those observed for
tissue culture cells is used for genetic, proteomic, or biochemical normal hematopoietic stem cells (HSCs).18 They noted that the
studies, mostly because the technology used to interrogate biologic dormant cells were much smaller in size and had less granularity
processes (ie, proteomics, biochemistry) cannot be done on single than the rapidly proliferating fraction, which led to speculation that
cells and requires large cell numbers. Clearly, this has been a these cells could represent distinct cellular fractions. Of course,
productive approach as evidenced by the thorough understanding such studies could not prove whether the cycling fractions repre-
gained on the genetic and epigenetic basis of the neoplastic sented the same population, existing in either a proliferative or a
process. However, an inability to deal meaningfully with limiting nonproliferative state, or whether these observations point to a
cell numbers has forced the cancer research enterprise to operate hierarchical relationship representing 2 functionally distinct cell
within a dichotomy because it is also well known that tumors are types. Even so, based on these observations, reviews of this era
not homogeneous but exhibit significant heterogeneity. predicted that the inability to eradicate the theoretical LSCs
Since the time of the great experimental pathologists (Virchow, represented the cause of relapse and the ultimate failure for
Maximov, and others), it has been recognized that tumors, be they antiproliferative chemotherapies, which formed the main chemo-
liquid or solid, exhibit marked morphologic heterogeneity. With the therapeutic armament at the time.14,19 Investigators then combined
advent of transplantation of murine cancer cell lines into mice, in vivo cytokinetic studies with drug treatment and tailored new
Furth and Kahn in 1937 were able to establish that a single cell, and therapies based on these findings. They made the interesting
not a transmissible agent, could initiate a cancer graft.4 As others observation that LSCs respond to depletion of the leukemia cell
applied transplantation assays to a variety of solid tumors and mass that occurs when antiproliferative drugs are administered to
leukemias throughout the next several decades, it became clear that AML patients. LSCs begin to go into cycle and expand in the same
there was wide variation in the frequency of cells that initiate way normal stem cells go into cycle after chemotherapy-induced
cancer growth.5-7 Remarkably, human studies have been carried out cytopenias. Thus, they suggested that one way to eliminate dormant
since the late 1800s (reviewed by Southam et al8) where tumors LSCs would be to find the window when they cycle to kill them
were excised, single-cell suspensions made and autotransplanted when they are most vulnerable. Indeed, the cytokinetic findings
into the thigh at different cell doses. These studies showed that represented the underpinning for pioneering clinical trials carried
tumor reinitiation was variable and rare, often requiring more than out by Clarkson and his colleagues at Memorial Sloan-Kettering
106 cells. Finally, in an elegant series of experiments, Pierce et al Cancer Center that used multiple drugs.20 The fundamental prob-
showed that malignant teratocarcinoma cells can spontaneously lem investigators faced at this time was the inability to directly
differentiate into mature benign cells.9 Thus, a tumor can be identify and assay LSCs, making it impossible to characterize them
considered a hierarchy defined by a maturation process in the same and harness the acquired knowledge for therapeutic approaches in the
way that normal tissue development occurs. In 1961, Pierce and future. Pointed comments made at least as early as 1970 predicted that
Speers proposed the idea that tumors are caricatures of normal antiproliferative chemotherapy agents would not effectively eradicate
development and that this maturation process could be harnessed as these dormant LSCs and that future progress must focus on understand-
a means of therapy.10 ing their properties.17 These remarkably prescient observations retain
The concept of cellular heterogeneity reached its zenith with the their potency and freshness even today, nearly 40 years later.
functional cell proliferation studies of the 1940s, 1950s, and 1960s. The cytokinetic studies drove interest away from the study of
The advent of radiolabeling cells, combined with autoradiography bulk leukemias because they were mostly composed of postmitotic
by Belanger and Leblond in 1946, enabled precise measurements blasts. Because there was little that could be done to assay the
of the proliferation, lifespan, and hierarchical relationships in dormant LSC, attention focused on the development of a clono-
normal and neoplastic tissues.11 Clermont and Leblond showed, for genic assay for the rapidly proliferating leukemic cells (AML-
example, that the testis contained proliferating cells that took up a colony-forming unit [CFU]) that the in vivo studies had uncovered.
pulse of label and then by tracking the label showed their McCulloch et al,21,22 Moore and Metcalf et al23,24, Griffin and
differentiation to mature cells.12; they also noted that these cells Löwenberg,25 and Dicke et al26 pioneered these assays and made
maintained themselves. In the 1960s and 1970s, several prominent detailed insights into their properties. Parallel clonal assays were
hematologists, including Clarkson et al,13,14 Killmann et al,15 developed for solid tumor clonogenic progenitors.27 In a key series
Gavosto,16 and others, carried out pioneering studies that were of experiments, Sabbath et al developed a series of monoclonal
(perhaps more than any other findings) the studies establishing that antibodies (mAb) directed against myeloid differentiation antigens
tumors exhibited functional heterogeneity. These were cytokinetic and assessed the phenotype of the AML-CFU itself by complement
studies carried out first in cell lines and murine models of the acute mediated lysis followed by clonogenic assay.28 Parallel studies
leukemias and then, remarkably by in vivo examination of were done with normal cells. The AML-CFU from different
leukemia blast proliferation kinetics in human AML and acute patients could be classified into 3 groups: primitive (similar to
lymphoblastic leukemia (ALL) patients. The in vivo data could not multipotent normal CFU), early lineage committed (similar to
have been clearer; the majority of leukemic blasts were postmitotic normal day 14 CFU-GM), and late (similar to day 7 CFU-GM).
and needed to be continuously replenished from a relatively small This work established that the AML-CFU were distinct from most
proliferative fraction17 (Figure 1). Only a small fraction (⬃ 5%) of leukemic blasts and the leukemic cells within an individual colony
leukemic blasts was rapidly cycling in vivo. However, careful could be more differentiated than the individual AML-CFU that
analysis demonstrated that there were 2 proliferative fractions in originated the colony. These clonal data provided the first strong
patients: a larger, fast cycling subset with a 24-hour cell cycle time proof for a hierarchy in AML. Moreover, this work suggested that
and a smaller, slow cycling fraction with a dormancy estimated to the different classes of AML-CFU might originate from normal
BLOOD, 15 DECEMBER 2008 䡠 VOLUME 112, NUMBER 13 CANCER STEM CELLS 4795

Figure 1. Labeling pattern of leukemic cells in marrow of patient 1.


Patient 1, a patient with acute myelomonocytic leukemia, received a
continuous 10-day infusion of tritiated thymidine. Leukemic cells were
arbitrarily divided into types I, II, and III based on increasing levels of
morphologic maturity (type I indicates primitive blast forms; type III, most
differentiated cells). At the end of the 10-day infusion, most type II and type
III cells were labeled in both marrow (shown here) and blood (not shown),
but only 40% of type I cells were labeled, reflecting their slow proliferative
rate. Many of the type I cells remained highly labeled for over 3 weeks after
the infusion. Reprinted from Clarkson17 by permission.

cells at different stages of differentiation.25 McCulloch et al showed sive models could explain tumor heterogeneity. The stochastic
additional evidence for heterogeneity with the finding that the model predicts that a tumor is biologically homogeneous and the
AML-CFU were heterogeneous in their serial replating ability, behavior of the cancer cells is influenced by intrinsic (eg, levels of
some unable to generate secondary colonies whereas others had transcription factors, signaling pathways) or extrinsic (eg, host
high capacity.22,29,30 Because this is an indicator of self-renewal, factors, microenvironment, immune response) factors. These influ-
they predicted that some AML-CFU were more primitive than ences are unpredictable or random and result in heterogeneity in the
others. Although the properties of AML-CFU (number, prolifera- expression of cell surface markers or other markers of maturation,
tion, drug sensitivity) on their own did not predict clinical disease in entry to cell cycle, or in tumor initiation capacity. A key tenet of
properties or response to therapy, the secondary replating capacity this model is that all cells of the tumor are equally sensitive to such
had better predictive power. The AML-CFU assay was used stochastic influences and also that tumor cells can revert from one
intensively from the late 1960s to the early 1980s and resulted in many state to another because these influences do not induce permanent
important insights into AML biology. However, the limitations of the changes.1 In contrast, the hierarchy model predicts that the tumor is
assay and the lessons that had been learned from the study of normal a caricature of normal tissue development where stem cells
hematopoiesis underscored the need for functional in vivo assays to maintain normal tissue hierarchies (skin, colon, blood). In this
determine whether an LSC, earlier than AML-CFU, existed in AML. model, the “leukemic stem cells” are biologically distinct, they
maintain themselves and the clone by self-renewal, and they
mature to generate progeny that lack stem cell properties (Figure
2). Thus, both the stochastic and hierarchy models accommodate
Models of tumor heterogeneity the existence of “functional leukemic stem cells,” cells that behave
as leukemia stem cells. The essential difference is that, in the
Collectively, the data described in “Tumor heterogeneity” firmly stochastic model, LSCs arise randomly and every tumor cell has
established that tumors are not only morphologically heteroge- the potential to behave like an LSC given the right influences;
neous as observed more than 100 years ago, but they are whereas in the hierarchy model, there is only a distinct subset of
functionally heterogeneous in terms of cell proliferation or even cells that has the potential to behave like LSCs. The only means to
tumor initiation based on transplant assays. Two mutually exclu- determine which model is correct is prospective isolation of cell
4796 DICK BLOOD, 15 DECEMBER 2008 䡠 VOLUME 112, NUMBER 13

Figure 2. Models of tumor heterogeneity. Tumors are composed of phenotypically and functionally heterogeneous cells. There are 2 theories as to how this heterogeneity
arises. According to the stochastic model, tumor cells are biologically equivalent, but their behavior is influenced by intrinsic and extrinsic factors and is therefore both variable
and unpredictable. Thus, tumor-initiating activity cannot be enriched by sorting cells based on intrinsic characteristics. In contrast, the hierarchy model postulates the existence
of biologically distinct classes of cells with differing functional abilities and behavior. Only a subset of cells can initiate tumor growth; these cancer stem cells possess
self-renewal and give rise to nontumorigenic progeny that make up the bulk of the tumor. This model predicts that tumor-initiating cells can be identified and purified from the
bulk nontumorigenic population based on intrinsic characteristics.

fractions followed by clonal analysis with a functional LSC assay. However, I do not think the absence of technology can fully explain
If the hierarchy model is correct, then it should be possible to sort why lags existed from the mid-1970s when all the theoretical
tumors into fractions that have LSC activity and those that do not. principles pointing to the existence of LSCs and a description of
In contrast, the stochastic model predicts that LSC activity may be their properties were well worked out, to the mid-1990s when
observed in any fraction and cannot be prospectively isolated direct proof for the existence of LSC was finally discovered, and
because all tumor cells have the same potential (even if it is low) then to the most recent years when CSC concepts have been
for tumor initiation. extended to solid tumors. The explanation also rests in the time it
took for stem cell concepts to permeate the broader cancer research
community and to become more accepted. The research environ-
Conditions for discovery ment was primed with the groundbreaking studies of normal HSCs,
eventually leading to HSC transplant as therapy and to the
The term “leukemia stem cell” had been in use for many decades, crossover of universal stem cell concepts into many areas of
and a large part of the hematology community assumed that the biology. This created a backdrop in which technologies were
hierarchy model was true and the leukemia clone existed in a developing, including the use of flow cell sorters in combination
hierarchy sustained by an LSC, with obvious parallels to the role with mAb that bind cell surface markers and the development of
that HSCs play in sustaining the normal hematopoietic hierarchy. human xenograft assays for human HSCs and LSCs. Because these
However, formal proof that LSCs had stem cell attributes and were conditions set the stage for discovery of LSCs and eventually CSCs
distinct from the rest of the leukemic cells that could never behave from solid tumors, it is instructive to review the key highlights that
as LSCs was still lacking. Clearly, technologies that were not culminated to identify HSCs and the universal concepts these stem
available in the 1970s were required to move the field forward. cell studies generated.
BLOOD, 15 DECEMBER 2008 䡠 VOLUME 112, NUMBER 13 CANCER STEM CELLS 4797

Identification of normal HSCs in the mouse Applying universal stem cell concepts to
other fields
The modern era of stem cell research can be traced back to the
seismic shift in the 1960s when Till and McCulloch moved HSC While these concepts were solidifying, stem cell concepts began to
research from a descriptive science to a quantitative science. percolate into other research fields through a series of culminating
Although careful studies using microscopic analysis pointed to the events and set the stage for further discovery. Perhaps the most
idea of a common stem cell for all blood lineages,31 equally robust influential factor that drove stem cell concepts beyond the domain
was the view that independent stem cells existed for each major of a small number of stem cell biologists into a wider sphere of
blood lineage, which was promulgated by textbooks from the biomedical research was the development of HSC transplantation
1940s.32 The experimental finding by Till and McCulloch that the for treatment of leukemia and other hematologic diseases. Through
“regeneration nodules” on the spleen of lethally irradiated mice a series of elegant studies in the 1950s, proof was provided that
(termed CFU-spleen [CFU-S])33 transplanted with BM cells were cells (as opposed to a humoral factor) from the bone marrow of
clonal (based on X-ray induced chromosomal lineage tracking34) mice could protect against lethal doses of radiation by effectively
and contained multiple blood lineages proved that the hematopoi- regenerating all of the blood lineages in transplant recipients.52,58 In
etic system derives from a multipotent HSC rather than from a 1956, Thomas et al carried out the first human bone marrow
transplantations,59 although initially a failure, they brought a
series of lineage-specific stem cells. With a clonal, quantitative, and
decade of improvements in tissue typing and methods for control-
functional assay in hand, in short order they established all of the
ling graft-vs-host disease. Finally, the pioneering clinical trials of
major stem cell principles under which we continue to operate
Good, Thomas, and others showed that human BM could be used to
today: HSCs have a high capacity for self-renewal, proliferation,
cure immune deficiency by sibling donor transplants in 196860 and
and multipotential differentiation. Their observations that the
neoplastic disease in 1969,61 beginning the modern era of human
self-renewal capacity of individual CFU-S was highly variable35 allogeneic marrow grafting. During the same time period, the new
led them to propose the concept that the control of HSC self- field of molecular biology was established. This made it possible
renewal was governed by stochastic principles,36 a hypothesis that not only to use genetics to characterize biologic processes in a
is still being tested and still able to provoke strong opinions among much more rapid fashion, but also to produce biologic regulators
stem cell biologists.37-41 Their focus on the need for quantitative on an industrial scale. All of these research paths collided in the
functional assays prompted the development of in vitro clonal 1980s with the birth of the biotechnology industry, which was
progenitor assays by Pluznik and Sachs42 and Bradley and Met- mostly focused on the commercialization of products to stimulate
calf43 that were central to the later discovery of hematopoietic blood cells. Thus, the arcane fields of blood diseases, HSC transplanta-
colony stimulating factors (as reviewed in the 50th Anniversary tion, and stem cells in general had moved from a backwater to front
Review by Metcalf44). With the use of progenitor assays, it was stage. The final influence in this fertile period was the discovery of
possible to describe precisely the lineage relationships that occur embryonic stem cells by Evans and Kaufman62 and Martin63 in 1981
during maturation. Despite widespread use of the spleen colony with the attendant promise of regenerative medicine. Stem cell biology
assay as a surrogate HSC assay, by the mid 1980s it was clear that a had finally become a hot topic, and it had begun to capture the attention
more primitive HSC must exist. First, CFU-S were heterogeneous of a broad swath of the research community.
in terms of their spleen repopulation kinetics.45,46 Second, careful
lineage examination showed that the spleen colony was devoid of
lymphoid cells and therefore not multipotential.47,48 Third, after Technology and assay developments
5-fluorouracil treatment, CFU-S were killed whereas other dormant
cells capable of CFU-S replenishment survived, suggesting the It was within this milieu that 2 important technologic advances
existence of a pre-CFU-S stem cell.49,50 Harrison, in particular, took place that directly led to the renewal of interest in the concepts
drove the paradigm that cells with long-term repopulation capacity of LSC from 20 years earlier. The first was the development of
could be distinguished from those with short-term potential.51 fluorescence-activated cell sorting (FACS), and the second was the
Using chromosomal translocation as a means of cellular marking development of xenotransplantation assays for normal and leuke-
mic human stem cells. Although Miller and Phillips had developed
(first developed by Ford et al52), in 1968 the first clonal evidence
the first large-scale methods for cell separation based on sedimenta-
established multilineage repopulation, including CFU-S and
tion,64 these methods lacked the precision needed for meticulous
B cells.53 Using the same approach, Abramson et al showed that
cell fractionation. The development of FACS by Herzenberg’s
repopulation of myeloid and lymphoid lineages could derive from a
group at Stanford in 1972,65 combined with technology to create
long-term multipotential stem cell or from lineage-restricted stem
specific mAbs against cell surface differentiation antigens, finally
cells.54 However, the efficiency of marking was low and the need to provided the means to fractionate cells with high purity. As applied
irradiate the bone marrow to induce chromosomal translocation to stem cell fractionation, Visser et al were the first to use flow
meant that it was difficult to establish whether the identified HSC sorting based on wheat germ agglutinin binding as a means to
classes truly existed or were experimentally induced artifacts. With enrich (6-fold) CFU-S compared with unfractionated bone marrow
the development of more benign methods for HSC clonal tracking cells.66 Later it was possible to sort on the basis of cell cycle status
using retroviral insertion sites, it was possible to establish that using Hoechst dyes, and in this way CFU-S were distinguished
long-term, multipotential HSCs existed.55-57 Collectively, these from the more actively dividing granulocyte-macrophage-CFU
studies established that clonal assays are essential to uncover progenitor cells.67 Improved cell fractionation based on cell surface
stem cell properties; and when individual HSCs are assessed, markers and longer-term functional assays culminated in an
they exhibit wide variations in repopulation dynamics and important paper by Spangrude et al who purified mouse multipoten-
self-renewal capacity. tial HSCs (although mostly those capable of relatively short-term
4798 DICK BLOOD, 15 DECEMBER 2008 䡠 VOLUME 112, NUMBER 13

repopulation) by combining depletion of several markers of SCID mice establish the importance of controlling macrophages to
maturation with expression of the Sca1 and c-Kit proteins (LSK achieve HSC engraftment.91 Finally, direct intrafemoral delivery of
cells).68 Improvements in markers and the use of single-cell cells resulted in more sensitive SRC detection, as well as assess-
transplantations have now resulted in essentially pure populations ment of progenitors that possess transient repopulation potential, or
of HSCs capable of multipotential repopulation.40,69-72 However, are lineage restricted and would not read out in classic intravenous-
only 20% to 50% of these clonal HSCs are capable of long-term based HSC transplantation assays.90,92-95
repopulation; it remains unclear whether these HSC fractions are
still not pure and that additional markers may be found to
prospectively isolate HSCs with high capacity for self-renewal away Characterization of normal human HSCs
from those with more limited capacity, or whether this remaining
variability is because of stochastic influences on an otherwise pure HSC The SRC xenotransplantation assay has emerged as the “gold
population.39,40 The intense efforts to purify murine HSCs and their standard” surrogate assay for human HSCs.96 Combined with
downstream progeny established a pathway of discovery that could be clonogenic and stromal-based assays, it has been possible to
applied to characterize human hematopoiesis. identify many cell surface markers that define the developmental
Because studies of murine hematopoiesis showed that HSCs are hierarchy of human hematopoiesis, setting the stage to apply these
distinct from CFU and that HSCs with short-term repopulation same markers to understanding leukemic hematopoiesis. A detailed
capacity are distinct from those with long-term capacity, it was understanding of the repopulation potential, cell-surface pheno-
clear that progress to understand human hematopoiesis would type, differentiation, and proliferation properties of SRCs has been
require a repopulation assay. The second key technologic develop- gained over the past 15 years.96,97 Most SRCs express CD34, and
ment leading to the identification of LSC was the beginning of the distinct SRC activities can be found within the lineage-negative
modern era of the xenotransplantation system, which provided the (Lin⫺) CD34⫹CD38⫺ and Lin⫺CD34⫹CD38lo subfractions.88,92,98-100
key ability to functionally assay both normal stem cells and Personally, the culmination of our normal HSC studies was the
LSCs.73 Although there was a long history of transplanting human use of lentivector-mediated clonal tracking101,102 to monitor greater
tumor cells into irradiated or immune-deficient nude mice, the than 600 clones in a cohort of primary and secondary mice for a
strong host resistance in these mice allowed only the most total of 7 months.39 These studies touched on the longstanding
aggressive tumors and cell lines to be engrafted. Primary human debate on whether the control of cell fates is rigid and tightly
leukemia cells could not be transplanted and most cell lines only organized or whether these outcomes are unpredictable and gov-
grew subcutaneously or as ascites, which was not a normal pattern erned by stochastic processes. A single HSC can generate the entire
for hematologic proliferation. In 1988, 3 groups using severe blood system for a lifetime,40,69-72 a result that can only occur if at
combined immunodeficiency (SCID) or bg/nu/xid (BNX) mice, cell division the HSC self-renews to produce at least one daughter
which are more immune-deficient than nude mice, showed that cell with the undifferentiated characteristics of the parental stem
normal human hematopoietic cells could repopulate these mice. cell. Whether a hematopoietic clone is maintained or extinguished
Mosier et al transplanted human peripheral blood leukocytes by ultimately rests on whether an HSC makes an asymmetric (mainte-
intraperitoneal injection into SCID mice to enable long-term nance) or a symmetric self-renewing (expansion) or differentiating
generation of mature T cells that could then be infected with HIV.74 (extinction) division. These outcomes are known as the “birth and
McCune et al were also interested in developing an HIV model. death” fates of stem cells.36 (see McKenzie et al39 for a more
They implanted SCID mice with pieces of human fetal liver, lymph detailed discussion). We clonally analyzed hundreds of individual
node, and thymus,75 and later they injected human cells into fetal SRCs in primary and secondary mice; as well, we had the
bone.76 The rationale of their approach was to provide a human opportunity to analyze the repopulation properties of SRC daughter
tissue microenvironment that would continuously generate human cells that became physically separated to different marrow loca-
T cells for HIV studies because the fetal liver (or fetal bone) would tions within a single mouse.39 We found that the proliferation and
provide a source of stem cells. Of course, the long-term production self-renewal properties varied greatly. Even with increasing SRC
of human cells within the fetal bone could be used to assay and purification, the variation in self-renewal capacity remained. Our
identify HSCs, a premise that came to fruition when they showed results are reminiscent of the murine CFU-S data that were first
that purified CD34⫹Thy⫹ cells could initiate multilineage engraft- reported approximately 45 years ago that formed the basis for the
ment of this bone fragment.77 Our approach was somewhat prediction that regulation of individual HSC outcomes is stochastic
different and guided by the principles of bone marrow transplanta- and governed by probabilistic elements.36 Our data provided
tion in humans and mice: intravenous transplantation into irradi- experimental in vivo evidence (albeit with all the caveats of a
ated recipients resulting in human cells repopulating the hematopoi- xenotransplantation model) that is consistent with the hypothesis
etic tissues of a mouse.78-80 With improvements in recipient mice,81 that human HSCs are also governed at least in part according to
it was finally possible to quantify and identify the cell that was stochastic principles. Collectively, the identification and character-
capable of initiating a multilineage human graft, which was termed ization of human HSCs set the stage for identification of their
the SCID-repopulating cell (SRC).82-85 The SRC measured in this leukemic counterparts. The subsequent comparisons between nor-
system generated many orders of magnitude higher cell numbers mal and leukemic stem cells continue to provide clues to the
than the fetal bone in the SCID-hu model, providing a more robust cellular and molecular programs that drive their biology.
method to detect only those cells with the highest repopulation
capacity. Many improvements to the NOD/SCID assay continue to
be made by using recipient mice that are engineered to be deficient Identification of leukemia stem cells in AML
in both NK and macrophage activity86,87 or that have been treated
with a mAb to CD122 that depletes these components of the innate Because we were able to transplant normal human cells into
immune system.88-90 Recent studies that identified polymorphic mice, it was obvious that we should attempt to grow leukemic
alleles of Sirp-alpha governing human HSC engraftment in NOD/ cells in mice. Our initial studies transplanting pediatric pre-B
BLOOD, 15 DECEMBER 2008 䡠 VOLUME 112, NUMBER 13 CANCER STEM CELLS 4799

ALL into SCID mice showed the power of the system to had used for sorting had greater than 40% CD34⫹ cells;
propagate ALL with great similarity to the human disease103 and however, CD38 was a smaller fraction of the CD34⫹ cells.
correlation to clinical outcome.104 Because we were interested in AML-CFU activity was high in the CD38⫹ fraction compared
establishing whether normal HSCs could be assayed in SCID with unsorted leukemia blasts but was also present in the CD38⫺
mice, this focus on thinking clonally impacted our leukemia fraction. Remarkably, despite the rarity of the CD34⫹CD38⫺
studies. At this point, we turned our attention to AML because of fraction and the presence of high levels of AML-CFU in the
the more extensive work on clonogenic assay systems and the CD34⫹CD38⫹ fraction, leukemic engraftment could only be
application of stem cell concepts to understand this disease. I initiated from CD34⫹CD38⫺ fractions. In keeping with a
had been especially enamored by the clonogenic assay for AML tradition of naming cells based on function, we termed this the
progenitors and the studies from Buick and McCulloch, which SCID leukemia-initiating cell (SL-IC). The CD34⫹CD38⫺ SL-IC
showed that only a subset of AML-CFU had high serial replating generated large numbers of CD34⫹CD38⫹cells, AML-CFU, and
efficiency, pointing to heterogeneity in self-renewal capac- mature blasts. This was proof that the cell population that
ity.22,29,30 In parallel, the studies outlined in “Identification of initiated AML in SCID mice was distinct from AML-CFU; we
normal HSCs in the mouse” clearly showed that normal had identified a new stem cell in AML.107
clonogenic progenitors were distinct from repopulating HSCs, Now is probably the best time to confess that I had no idea at the
raising the question of whether the same would hold true for time that this experiment settled an even larger issue about the
AML and that a more primitive LSC existed that was distinct stochastic/hierarchy controversy, at least for AML. Not having read
from AML-CFU. The question was how to assay such a cell. As carefully all the literature I have outlined in this review, I did not
with any new line of investigation, collegial support was realize that such a controversy existed because the hierarchy model
essential, especially from M. Minden and T. Hoang who was so well entrenched in the normal and leukemic hematopoiesis
provided primary AML samples and taught us how to properly field; it was simply a given. In my view, all we had provided was
thaw cells. We initiated experiments transplanting primary AML the long sought after formal proof for the concept of a hierarchy in
into SCID mice and obtained reliable engraftment from most AML. There was one issue that concerned us. We had only tested a
subtypes of AML, except for APL. The subsequent critical single sample in this initial study and it had a primitive FAB-M1
experiments were performed with the prompting of my mentor subtype and a high CD34 content. Perhaps the principle, or at least
R. A. Phillips who suggested we establish that the assay was the CD34⫹CD38⫺ phenotype, would not hold for AML, which
quantitative by measuring the frequency of the initiating cell. exhibited a higher degree of cellular maturation, particularly the
This suggestion launched the application of stem cell principles myelomonocytic M4/M5 subtypes. By this time, the new NOD/
to the study of the initiating cell in AML. Our first step was to SCID mouse strain was found to be a superior recipient for normal
establish the frequency of the initiating cell. Although we found HSC engraftment, permitting, for the first time, quantitative
it to vary by more than 1000-fold between donors, it was assessment and purification of normal HSCs.82-85 With this more
exceedingly rare: on the average of 1 per 106 cells. This was the robust assay, we studied in detail another 7 samples, 6 being M4 or
first hint as to whether this initiating cell was actually distinct M5. Many of these samples exhibited a distinct pattern of
from the AML-CFU, which has a typical frequency of 1 in 100. engraftment different from the primitive samples. The myelomono-
Second, with the suggestion of my colleague N. Iscove, we cytic samples disseminated widely, whereas the primitive subtypes
carried out kinetic studies to show expansion of the AML-CFU typically grew well only in the bone marrow and only disseminated
pool during repopulation, as would be expected if they were after long periods of time. Interestingly, although the M4 and M5
being generated from an earlier precursor. Of course, it was samples disseminated widely into many organs, they never infil-
possible that the initiating cell and the AML-CFU were the same trated the central nervous system. This pattern was distinct from the
cell if in vivo repopulation were simply an inefficient method to pre-B ALL samples that we had studied earlier, which also
assay AML-CFU. Clearly, the only way to resolve this question disseminated widely but often infiltrated the central nervous system
was by fractionating the cells (ie, separating AML-CFU away and the testis. These disease patterns that we observed in the
from the initiating cells). CD34 was an obvious choice to NOD/SCID mouse reflected the clinical features of the disease in
fractionate with because expression on human HSCs had already humans, pointing to the reliability of these new xenotransplantation
been shown105 and adopted clinically for HSC enrichment. Prior models (compared with the older subcutaneous implantations in
work from Terstappen et al had pointed to CD34 and CD38 as nude mice) to mimic the human disease. The cell purification data
being heterogeneously expressed in AML with the prediction from these newer experiments were clear: despite their myelomono-
that the various combinations of these 2 markers reflected a cytic morphology and their low CD34⫹ and CD34⫹CD38⫺ popula-
developmental hierarchy, with CD34⫹CD38⫺ being the most tions, the LSCs were all found in the CD34⫹CD38⫺ fraction.108
primitive.106 The immediate obstacle was that I knew little about Although some exceptions were reported in subsequent years, a
flow cytometry, and we did not have robust sorting capabilities high proportion of AML patients show LSC activity in the
in Toronto at that time. However, I had made the fortunate CD34⫹CD38⫺ fraction.109-111 As well, recent studies have sug-
acquaintance of M. Caliguiri from Roswell Park who offered us gested that the ability to engraft NOD/SCID mice112 as well as
access to their new highly modified beta test FACStar instru- increased frequencies of LSC in AML are prognostic for poor
ment run by C. Stewart. I have vivid memories of traveling with outcomes.113 Collectively, these studies provide conclusive evi-
my postdoctoral fellow T. Lapidot (who deserves enormous dence for the hierarchy model in AML.
credit for these studies) to Buffalo, sorting cells all day and
racing back to Toronto to transplant the precious sorted cells
into mice. I still recall with amusement the difficulties that the Cell of origin
U.S. border agents had with his Hebrew Israeli passport. As
others had seen, CD34 expression was highly variable in the When it came time to write the paper, D. Bonnet and I spent much
AML samples we had evaluated and the one FAB-M2 sample we effort reading the older literature, and we realized that our data
4800 DICK BLOOD, 15 DECEMBER 2008 䡠 VOLUME 112, NUMBER 13

Figure 3. Hierarchy of leukemia stem cells in AML.


Like the normal hematopoietic system, AML is organized
as a hierarchy of distinct cell classes that is sustained by
a subset of leukemia stem cells (or SCID-leukemia
initiating cells [SL-ICs], as assayed in immunodeficient
mice). Genetic tracking experiments have shown that
SL-ICs are heterogeneous in their ability to repopulate
secondary and tertiary recipients, pointing to the exis-
tence of distinct classes with differing self-renewal capac-
ity, similar to what is seen in the normal hematopoietic
stem cell compartment. Short-term (ST) SL-ICs are able
to initiate leukemia in primary but not secondary recipi-
ents, whereas long-term (LT) SL-ICs can sustain leuke-
mic growth for multiple passages. Quiescent LT SL-ICs
may not initiate a substantial graft in primary recipients
and may therefore only be detected on serial
transplantation.

could address the “cell of origin” question: what is the normal cell simply meant the cell in a cancer that has stem cell–like properties,
from which leukemia arises? Work from a number of directions, especially the capacity for self-renewal.120
including the variation in cell surface marker expression on Although we made the argument that the cell of origin is
AML-CFU by Sabbath et al28 described earlier and the pioneering primitive based on the consistency of the CD34⫹CD38⫺ cell
clonality studies in humans by Fialkow et al using X-linked G6PD surface phenotype of SL-IC among patients regardless of AML
polymorphisms114,115 followed by DNA-based restriction fragment subtype and their similarity to the phenotype of normal SRC, our
length polymorphism analysis by Fearon et al,116 had been used to reasoning had serious limitations. Cell surface markers are antigens
develop a theory that the maturation state of the leukemia blast whose expression changes with differentiation, and we know
reflected the level of maturation of the cell of origin.25 In vivo differentiation is perturbed in cancer. Therefore, one must be
clonal studies in humans examined whether the normal nonleuke- especially cautious when using a defined set of markers that rigidly
mic lineages (lymphocyte, erythroid cell) were or were not part of reflect the steps of lineage determination of normal cells and then
the leukemic clone. Such an approach had definitively established inferring that the same set will apply for neoplastic cells. Ideally,
that chronic myeloid leukemia (CML) arose in a multipotential one would like to make comparisons between normal HSCs and
HSC.117 If the nonleukemic cells were part of the clone, this LSCs using functional criteria. The only true stem cell property is
reflected an early multipotential cell of origin, whereas if they were capacity for maturation combined with self-renewal. In normal
not part of the same clone, this reflected a more lineage-restricted hematopoiesis, cell tracking and purification studies performed
cell of origin. This view was also proposed based on some of the clonally (in mice and humans) have documented that long-term
earlier in vivo cytokinetic studies of Clarkson and others, albeit repopulating HSCs with high self-renewal capacity generate short-
with less evidence than these clonal analyses could bring to bear. term HSCs whose self-renewal capacity is more limited.101,121
The alternative view, best articulated by McCulloch, was that Thus, we carried out lentivector-mediated clonal tracking of
observed lineage restriction was indeed only an apparent restriction AML-LSC in a manner identical to that outlined in “Characteriza-
and could not rule out the possibility that even the mature or lineage tion of normal human HSCs” for normal HSCs. These studies
restricted AMLs arose from a multipotent HSC.22,29 He argued that established that individual LSCs were not homogeneous but had
transforming events could occur in an HSC, but one of the wide variation in repopulation potential and self-renewal capacity
consequences of this event was an inability of this stem cell to similar to that observed for normal HSCs122 (Figure 3). However, a
generate other “normal” lineage cells, such as erythroid cells. As key difference is that, during NOD/SCID repopulation, LSCs
discussed in the following paragraph, we proposed that the similar expand to a much greater extent than normal HSCs, probably
cell surface phenotype of the LSC and the HSC supported the idea because of increased symmetric self-renewal cell divisions.108 As
that primitive cells were the cell of origin for AML regardless of the more information is gathered about the molecular machinery that
maturation state of the leukemia blasts.108 Perhaps more than any underlies the self-renewal process, it is becoming clearer that LSCs
other, this paper has caused confusion about the nomenclature of a and HSCs share at least some characteristics. For example, Bmi1123
“cancer stem cell,” especially when the CSC field exploded in and PTEN124 govern self-renewal of both murine HSCs and LSCs.
2003/2004 when solid tumor CSCs were identified.118,119 Our use of Together with the findings that LSC can also be quiescent like
this nomenclature comes from the normal blood literature where normal HSCs,122,125-127 these discoveries add additional support to
cells are defined by function, in this case, the cancer cell that the notion that some types of AML may arise from within the primitive
possesses stem cell properties. Our attempts to address the “cell of cell compartment, before full lineage commitment has occurred.Alterna-
origin” question were taken by some to mean that a CSC was tively, it is also possible that AML could arise from committed
derived from a normal stem cell that had become cancerous. It progenitors if stem cell self-renewal programs are established during the
finally took a blue-ribbon panel established by the American leukemogenic process. Nevertheless, this evidence is still indirect and
Association for Cancer Research to strongly declare that a CSC requires a more direct experimental approach.
BLOOD, 15 DECEMBER 2008 䡠 VOLUME 112, NUMBER 13 CANCER STEM CELLS 4801

The most direct experiment is to introduce the same genetic Our current understanding of the potential pathways leading to
element into pure populations of HSCs or progenitor subsets to generating CSCs and a proposed model for tumor progression are
determine which population is competent to initiate disease. depicted in Figure 4. Because the lifespan of non–stem cells may be
Attempts to identify the cell of origin for murine leukemia have transient, any genetic alteration in their makeup may not persist
yielded conflicting results, demonstrating that cellular and molecu- long enough to acquire additional genetic events that are needed for
lar context is critically important. The fusion oncogene MLL- full transformation, unless the initial event endows the non-stem
GAS7 induces mixed-lineage leukemias when expressed in HSCs cell with self-renewal capacity. In addition, even when the stem cell
or multipotent progenitors, but not in lineage-restricted progeni- sustains the first hit, subsequent events may occur in the stem cell
tors.128 In contrast, MOZ-TIF,129 MLL-AF9,130 and MLL-ENL131 or in the downstream progenitors. Another consideration is deter-
lead to initiation of AML regardless of the target cell population mining how the hierarchy model relates to leukemic progression or
used. However, much lower numbers of transformed HSCs com- tumor evolution. Figure 4 proposes that tumor progression is linked
pared with progenitors are often required for tumor initiation in to self-renewal capacity. For example, genetic instability and clonal
vivo and tumors arising from progenitors are monoclonal or evolution can occur in both CSCs and non-CSCs, keeping in mind
oligoclonal not polyclonal, suggesting that the progenitor popula- that the rate of evolution could be different if CSCs proliferate
tions were not functionally homogeneously, were not equally more slowly than non-CSCs. Within the CSC fraction, additional
susceptible to transformation, or additional genetic alterations were genetic and epigenetic alterations could result in more abnormal
required. Murine models of CML resulting from inactivation of CSCs; and in the case of solid tumors, these could become
JunB or expressing bcr/abl must occur in HSCs and not more metastatic. The non-CSCs could also acquire genetic and epige-
restricted progenitors to induce a transplantable myeloproliferative netic alterations; but if these do not endow self-renewal capacity,
disorder.129,132 These murine studies have become even more then these alterations in the non-CSCs are passengers and not
difficult to interpret with the recent findings of Chen et al who used drivers of tumor progression. Finally, if stem cell-like properties,
a knockin model of MLL-AF9.133 This approach was taken because including self-renewal, were endowed on non-CSCs, they could
of concerns that, for some oncogenes, the level of expression from also drive tumor progression (Figure 4). A recent report has
exogenous promoters, such as transgenes or retroviral vectors, may established in a breast cancer model that an epithelial-to-
be nonphysiologic. They found that the same construct shown to mesenchymal transition program is linked to stem cell function and
initiate disease in HSCs and progenitors by retroviral expression experimental induction of epithelial-to-mesenchymal transition in
only initiated leukemia from the HSC fraction when expressed a non-CSCs can reprogram them to a CSC state, providing
from the endogenous MLL promoter. Nevertheless, collectively, evidence for this tumor progression model.140
these studies demonstrate that, under the correct circumstances and
with the correct oncogene, it is possible to transform non-HSC in
mice. The critical feature of these studies is that some component Modeling the human leukemogenic process
of an HSC or self-renewal program must be established in the
non-HSC cell types.134 A major limitation in understanding the leukemogenic process in
No direct studies of the design used for the murine studies have humans is the absence of good genetic tools and experimental
been reported for human hematopoietic cells. However, the in vivo models that use primary human cells as their starting point. For
clonality studies114,115 described earlier had pointed to variation in example, the first step to directly test the cell of origin hypothesis
the cell of origin. A more refined approach was to examine whether requires an experimental model of human leukemogenesis where
phenotypically defined populations contained specific transloca- oncogenes are transduced into primary human hematopoietic cells.
tions and whether these exhibited normal or leukemic proliferation. Several reports have demonstrated that the initial steps in the
A study of patients with t(8;21) AML demonstrated that, despite the leukemogenic process could be induced experimentally by expres-
presence of leukemia-specific AML1-ETO chimeric transcripts in sion of several different oncogenes, such as Ras,141 TLS-
primitive CD34⫹CD90⫺CD38⫺ cells from leukemic bone marrow, ERG,142,143 AML-ETO,144 and bcr/abl,145 but none caused full-
these cells give rise to normally differentiating multilineage blown disease (see Kennedy and Barabé146 for detailed review of
clonogenic progenitors, whereas more mature CD34⫹CD90⫺CD38⫹ human leukemogenesis studies). Using MLL fusions, 2 recent
cells form exclusively leukemic blast colonies (AML-CFC).135 studies have demonstrated successful induction of AML and pre-B
Recent studies have documented that normal HSCs are ALL in NOD/SCID transplanted with transduced primary stem/
CD34⫹CD90⫹CD38⫺, whereas the CD34⫹CD90⫺CD38⫺ fraction progenitor cells.147,148 These mice developed B-ALL and AML that
represents a normal multipotential progenitor with little self- reflected the human disease. We found that the B-ALL in this
renewal capacity.136 Thus, a conclusion of this work is that, model maintained a germ line configuration of the IgH locus early
whereas the initial t(8;21) translocation occurs in a primitive stem in disease evolution but progressed to full rearrangement on serial
cell, subsequent events occur in the committed progenitor pool, passage.147 These data suggest that the LSC can evolve and that the
giving rise to LSCs. However, in vivo assays are required for cell of origin was not a mature B cell, but a more primitive stem or
formal proof that these are LSCs. A similar process of leukemic progenitor cell. Direct elucidation of the cell of origin will require
progression recently has been proposed for CML.137 The discovery prospective cell sorting of pure, functionally defined HSCs, and
of a marker chromosome,138 the nature of the translocation,139 progenitor subsets as targets for testing.
followed by lineage involvement of the Ph chromosome,117 estab- Although the NOD/SCID studies were compelling, direct
lished that CML arose from an HSC; however, analysis of blast evidence for LSC evolution in humans was lacking. In a seminal
crisis samples suggests that additional genetic events occur at the series of studies beginning in the 1990s, Greaves and Wiemels
level of more committed progenitors, resulting in the generation of demonstrated that many forms of childhood leukemia arose in
LSCs.137 These provocative studies point to the need to understand utero.149 They studied twins in which one twin has leukemia
not only the cell of origin of the LSCs but also the steps leading to whereas the other does not; both were shown to contain the same
so-called “preleukemic stem cells.” chromosomal translocations (MLL, Tel-AML1), a result that could
4802 DICK BLOOD, 15 DECEMBER 2008 䡠 VOLUME 112, NUMBER 13

Figure 4. Models of tumor initiation and progression. Cancer stem cells may arise through neoplastic changes initiated in normal self-renewing stem cells or downstream
progenitors, causing expansion of the stem cell and/or progenitor pool. Secondary events may occur in expanded pools of target cells. Oncogenic events acquired by
short-lived progenitors may not persist if self-renewal is not reactivated, as these cells will probably die or undergo terminal differentiation before enough mutations occur for full
neoplastic transformation. Tumor progression may be linked to ongoing genetic instability and acquisition of additional changes by cancer stem cells, or possibly by
nontumorigenic bulk cells if such changes endow self-renewal. In both cases, evolution of tumor phenotype (including genetic and epigenetic signatures) may be observed.

only occur if a single cell in one twin acquired the translocation, mentally induced murine leukemia models were found where
expanded, and migrated to the other twin. By examination of Ig almost every cancer cell had LSC activity and there was little
rearrangements, they concluded that after birth additional genetic evidence of functional heterogeneity. These and other reports153
alterations occur independently in each twin, resulting in the speculated that the rarity of human CSCs observed using xenotrans-
appearance of leukemia at different times. This predicts that the plantation models may be the result of host resistance factors as
twin who has not yet developed leukemia must contain a preleuke- well as the absence of cross-species reactivity of cytokines and
mic stem cell that is poised to acquire additional alterations because other microenvironmental factors. Certainly, such factors play a
this twin is at high risk of developing leukemia. In an elegant series role because the use of more immune-deficient mice has improved
of studies, Hong et al have identified a unique cell population, not human CSC detection; in addition, some solid tumor studies are
observed in normal donors, that contains a preleukemic stem cell now using coimplantation of human stromal cells. It is important to
that exhibited enhanced self-renewal capacity when assessed in the note that a number of other genetically induced murine leukemia
NOD/SCID xenotransplantation system.150 This preleukemic popu- models are heterogeneous and contain discrete and rare LSCs.154
lation was clonally related to the LSC that maintained the leukemia Moreover, when human and murine leukemia models were both
in the affected twin. Thus, this study provides strong proof in induced with the same MLL fusion protein, the LSC frequency was
humans for the concept of clonal evolution of LSC from a the same.154 Thus, it is evident that LSC frequencies can vary
preleukemic to a leukemic state. widely among different cancers regardless of whether they are
quantified using xeno- or syngeneic transplantation assays. It is
important to emphasize that the fundamental concept underlying
Controversies the CSC model is not directly related to the absolute frequency of
these cells but rather on functional evidence of tumor heterogeneity
Recently, several reports were published that questioned the and a hierarchical organization. The CSC model is only invoked to
universality of the CSC or hierarchy model.151,152 Several experi- explain tumor heterogeneity; however, if a tumor is functionally
BLOOD, 15 DECEMBER 2008 䡠 VOLUME 112, NUMBER 13 CANCER STEM CELLS 4803

homogeneous, the CSC model is no longer relevant. It is clear that resulted in the development of antibodies as well as other
some murine cancer models do not follow the hierarchy model, and toxicities.161 As well, LSCs could be targeted with a CD8⫹
it will be important to survey a much broader cross section of cytotoxic T lymphocyte clone specific for minor histocompatibil-
human tumors to determine whether any also lack tumor ity antigens,162,163 an approach that might be useful in the
heterogeneity. context of AML patients who undergo allogeneic transplant and
then relapse. More recently, several studies reported that LSCs
could be targeted with mAb to CD44164 or CXCR4.165 When
The future of CSC research these mAbs were injected into NOD/SCID mice repopulated
with AML, the LSCs were markedly depleted; and in some
Despite the recent explosion of interest in CSCs, experimental cases, the mice were cured. Induction of differentiation repre-
studies have not been translated into improved survival outcomes sented a partial mechanism of action. Even more noteworthy,
for cancer patients. This presents a major question to the field: do both CXCR4 and CD44 resulted in impaired trafficking of the
cancer stem cells have meaningful relevance beyond the experimen- LSC, suggesting that LSCs remain dependent on niche interac-
tal systems in which they have been defined? Increasing evidence is tions. Because many other factors are involved in trafficking and
pointing to CSCs having unique biologic properties (dormancy, homing (see 50th Anniversary Review by Papayannopoulou and
drug/radiation resistance) that could permit them to survive thera- Scadden166), this finding opens the way for testing the anti-LSC
pies leading to eventual relapse. To date, CSCs have been studied in functionality of any molecule that interferes with these pro-
a relatively small number of patient samples and cancer types, so it cesses. Little is known of the LSC niche; however, a recent
is not known whether the CSC model is universal to all human study points to the endosteal region,167 an area that has been
cancers.1 The biologic relevance of CSCs in human cancer will be proposed as the niche for murine HSCs as well. Another
established by concentrating on the following research endeavors:
LSC-specific target, which is either low or absent from normal
improving the assay and purification of CSC and non-CSC subsets,
HSCs, is CD123, the alpha chain of the IL3R.168 A mAb
carrying out detailed genomic or proteomic analysis on these
targeting CD123 impairing homing of LSC to bone marrow of
subsets to identify CSC-specific signatures, and obtaining such
NOD/SCID mice, and it activates the innate immunity of
signatures from a large number and wide range of tumors. Such an
NOD/SCID mice resulting in eradication of LSCs.169 Collec-
approach would make it possible to determine whether the “omics”
of CSCs provide more predictive or prognostic relevance compared tively, these studies provide clear validation for therapeutic
with analysis of the bulk tumor. An approach like this has recently mAb targeting of AML-LSCs and for translation of the NOD/
been taken by Liu et al who showed that a breast CSC signature has SCID in vivo preclinical research findings toward a clinical
improved correlation to patient outcomes.155 Of course, the most application. Finally, future therapeutic approaches will need to
direct proof for the relevance of CSCs to cancer would be to use the be developed that target the self-renewal machinery of LSCs,
CSC-specific signatures to elucidate the key molecular pathways although it may be difficult to find a therapeutic window that
involved, develop the means to target them effectively, and will spare normal tissue stem cells. As predicted from the
establish in clinical trials whether targeting them enhances patient cytokinetic studies, perhaps the most attractive option is to
survival. induce the quiescent LSCs into cycle. Recent studies are
For many types of leukemia, the long-term survival has pointing to specific molecular pathways that might be attractive,
increased dramatically over the past 50 years; however, AML including PTEN, p21, and PML. Ito et al have found that
remains one of the most difficult diseases to cure. The majority degradation of PML induces cycling of LSC and, interestingly,
of patients will die because of relapse, drug resistance, or from that one of the mechanisms of action of arsenic is degradation of
complications associated with chemotherapies. Because LSCs PML, already putting a potential drug strategy in hand.170
and HSCs have many similar properties, a first step to develop Although this review is focused on the premise that improve-
novel therapies has been to identify unique properties that LSCs ments in the stubbornly high relapse rates for many cancers will
possess. Through expression analysis, Guzman et al determined come from more research on the CSCs that drive the tumors,
that the NF-␬B pathway was active in LSCs and not in HSCs.126 attention is needed on how to move CSC therapies into clinical
They then went on to identify a natural product, parthenolide, trials and into the clinic (a more thorough discussion is done by
which targets this pathway and showed that the LSCs could be Wang171). The current approaches to preclinical drug development
specifically targeted using the NOD/SCID model.156,157 It should
and clinical evaluation of drug efficacy focus on initial require-
be possible to develop a strategy that focuses on the biology of
ments for tumor response. Therefore, a potentially effective
LSC and develop assay methods that are compatible with high
anti-CSC therapy might be discarded from further testing if it does
throughput screening of small molecule libraries followed by
not cause a response of the non-CSCs cells rapidly enough.
counterscreens with normal HSCs or progenitors.158 Other
approaches focus on identification of differences in cell surface Similarly, an effective anti-CSC might take time to show efficacy
markers with concomitant development of immunoconjugates or be most effective in an adjuvant setting. Much research effort
with toxic moieties. LSCs could be targeted with a diphtheria will be required to find the best algorithms for clinical evaluation of
toxin conjugated to granulocyte colony-stimulating factor immu- CSC-targeted therapies.
notoxin.159,160 However, not all AML samples responded to these In conclusion, stem cell concepts lie at the heart of the first
molecules, and regrowth or unsuccessful eradication of the cancer therapies developed almost 40 years ago, which showed the
leukemic cells occurred after different treatment strategies, first tangible success in curing patients with acute leukemia. One
suggesting that not all classes of SL-IC were killed by diphtheria hopes that the renewal of interest in the linkage between stem cells
toxin conjugated to granulocyte colony-stimulating factor immu- and cancer will lead to a better understanding of how the cellular
notoxin. Similarly a DT-IL3 conjugate was shown to target the context of tumors affects cancer-specific genetic and epigenetic
LSC and progressed to clinical trials. Unfortunately, this therapy pathways and will once again drive new therapeutic advances.
4804 DICK BLOOD, 15 DECEMBER 2008 䡠 VOLUME 112, NUMBER 13

Canadian Cancer Society and the Terry Fox Foundation,


Acknowledgments research contracts with CSL and Arius, and a Canada
Research Chair.
The author thanks the Dick laboratory members, Jean Wang, Bob
Phillips, Norman Iscove, Tsvee Lapidot, and Mark Minden for
advice and comments; Barney Clarkson for discussions and for
sharing a treasure trove of “hard-to-find” papers; Jean Wang for Authorship
creating the figures; and Melissa Cooper for insightful editing.
This work was supported by grants from the Canadian Contribution: J.E.D. wrote the paper.
Institutes of Health Research (CIHR), the Ontario Institute for Conflict-of-interest disclosure: The author declares research
Cancer Research and a Summit Award both with funds from the contracts with CSL and Arius.
Province of Ontario, Genome Canada through the Ontario Correspondence: John E. Dick, Toronto Medical Discovery
Genomics Institute, the Leukemia & Lymphoma Society, Tower, 101 College Street, Room 8-301, Toronto, ON, Canada
and the National Cancer Institute of Canada with funds from the M5G 1L7; e-mail: jdick@uhnres.utoronto.ca.

References
1. Wang JC, Dick JE. Cancer stem cells: lessons 17. Clarkson BD. Review of recent studies of cellular 33. Till JE, McCulloch EA. A direct measurement of
from leukemia. Trends Cell Biol. 2005;15:494- proliferation in acute leukemia. Natl Cancer Inst the radiation sensitivity of normal mouse bone
501. Monogr. 1969;30:81-120. marrow cells. Radiat Res. 1961;14:213-222.
2. Dalerba P, Cho RW, Clarke MF. Cancer stem 18. Clarkson B. The survival value of the dormant 34. Becker AJ, McCulloch EA, Till JE. Cytological
cells: models and concepts. Annu Rev Med. state in neoplastic and normal populations. In: demonstration of the clonal nature of spleen colo-
2007;58:267-284. Clarkson B, Baserga R, eds. Control of Prolifera- nies derived from transplanted mouse marrow
3. Ailles LE, Weissman IL. Cancer stem cells in tion in Animal Cells. New York, NY: Cold Spring cells. Nature. 1963;197:452-454.
solid tumors. Curr Opin Biotechnol. 2007;18:460- Harbor Laboratory; 1974;945-972. 35. Siminovitch L, McCulloch EA, Till JE. The distri-
466. 19. Cronkite EP. Acute leukemia: is there a relation- bution of colony-forming cells among spleen colo-
4. Furth J, Kahn M. The transmission of leukemia of ship between cell growth kinetics and response to nies. J Cell Comp Physiol. 1963;62:327-336.
mice with a single cell. Am J Cancer. 1937;31: chemotherapy? Proc Natl Cancer Conf. 1970;6: 36. Till JE, McCulloch EA, Siminovitch L. A stochastic
276-282. 113-117. model of stem cell proliferation based on the
5. Makino S. Further evidence favoring the concept 20. Clarkson BD, Dowling MD, Gee TS, Cunningham growth of spleen colony-forming cells. Proc Natl
of the stem cell in ascites tumors of rats. Ann N Y IB, Burchenal JH. Treatment of acute leukemia in Acad Sci U S A. 1964;51:29-36.
Acad Sci. 1956;63:818-830. adults. Cancer. 1975;36:775-795. 37. Lemischka IR. The haematopoietic stem cell and
6. Hewitt HB. Studies of the dissemination and 21. Buick RN, Till JE, McCulloch EA. Colony assay its clonal progeny: mechanisms regulating the
quantitative transplantation of a lymphocytic leu- for proliferative blast cells circulating in myelo- hierarchy of primitive haematopoietic cells. Can-
kaemia of CBA m ice. Br J Cancer. 1958;12:378- blastic leukaemia. Lancet. 1977;1:862-863. cer Surv. 1992;15:3-18.
401. 22. McCulloch E. Stem cells in normal and leukemic 38. Morrison S, Uchida N, Weissman I. The biology of
7. Bruce WR, Van Der Gaag H. A quantitative assay hemopoiesis (Henry Stratton Lecture). Blood. hematopoietic stem cells. Annu Rev Cell Dev
for the number of murine lymphoma cells capable 1983;62:1-13. Biol. 1995;11:35-71.
of proliferation in vivo. Nature. 1963;199:79-80. 23. Metcalf D, Moore MA, Warner NL. Colony forma- 39. McKenzie JL, Gan OI, Doedens M, Wang JC,
8. Southam C, Brunschwig A, Dizon Q. Autologous tion in vitro myelomonocytic leukemic cells. J Natl Dick JE. Individual stem cells with highly variable
and homologous transplantation of human can- Cancer Inst. 1969;43:983-1001. proliferation and self-renewal properties comprise
cer. In: Brennan MJ, Simpson WL, eds. Biological the human hematopoietic stem cell compartment.
24. Moore MA, Williams N, Metcalf D. In vitro colony
Interactions in Normal and Neoplastic Growth: A Nat Immunol. 2006;7:1225-1233.
formation by normal and leukemic human hema-
Contribution to the Tumor-Host Problem. Boston,
topoietic cells: characterization of the colony- 40. Sieburg HB, Cho RH, Dykstra B, Uchida N,
MA: Little, Brown; 1962:723-738.
forming cells. J Natl Cancer Inst. 1973;50:603- Eaves CJ, Muller-Sieburg CE. The hematopoietic
9. Pierce GB Jr, Dixon FJ Jr, Verney EL. Teratocar- 623. stem compartment consists of a limited number
cinogenic and tissue-forming potentials of the cell of discrete stem cell subsets. Blood. 2006;107:
25. Griffin J, Löwenberg B. Clonogenic cells in acute
types comprising neoplastic embryoid bodies. 2311-2316.
myeloblastic leukemia. Blood. 1986;68:1185-
Lab Invest. 1960;9:583-602.
1195. 41. Huang S, Guo YP, May G, Enver T. Bifurcation
10. Pierce GB, Speers WC. Tumors as caricatures of dynamics in lineage-commitment in bipotent pro-
the process of tissue renewal: prospects for 26. Dicke KA, Spitzer G, Ahearn MJ. Colony forma-
tion in vitro by leukaemic cells in acute myelog- genitor cells. Dev Biol. 2007;305:695-713.
therapy by directing differentiation. Cancer Res.
1988;48:1996-2004. enous leukaemia with phytohaemagglutinin as 42. Pluznik DH, Sachs L. The cloning of normal
stimulating factor. Nature. 1976;259:129-130. “mast” cells in tissue culture. J Cell Physiol. 1965;
11. Belanger L, Leblond C. A method for locating ra- 66:319-324.
dioactive elements in tissues by covering histo- 27. Hamburger AW, Salmon SE. Primary bioassay of
logical sections with a photographic emulsion. human tumor stem cells. Science. 1977;197:461- 43. Bradley TR, Metcalf D. The growth of mouse
Endocrinology. 1946;39:386-400. 463. bone marrow cells in vitro. Aust J Exp Biol Med
28. Sabbath KD, Ball ED, Larcom P, Davis RB, Griffin Sci. 1966;44:287-299.
12. Clermont Y, Leblond C. Renewal of spermatago-
nia in the rat testis. Am J Anat. 1953;93:475-502. JD. Heterogeneity of clonogenic cells in acute 44. Metcalf D. Hematopoietic cytokines. Blood. 2008;
myeloblastic leukemia. J Clin Invest. 1985;75: 111:485-491.
13. Clarkson B, Fried J, Strife A, Sakai Y, Ota K, Okita
746-753. 45. Magli MC, Iscove NN, Odartchenko N. Transient
T. Studies of cellular proliferation in human leuke-
mia: III. Behavior of leukemic cells in three adults 29. McCulloch E, Till J. Blast cells in acute myelo- nature of early hematopoietic spleen colonies.
with acute leukemia given continuous infusions of blastic leukemia: a model. Blood Cells. 1981;7: Nature. 1982;295:527-529.
3H-thymidine for 8 or 10 days. Cancer. 1970;25: 63-77. 46. Boggs DR, Boggs SS, Saxe DF, Gress LA,
1237-1260. 30. Buick RN, Minden MD, McCulloch EA. Self-re- Canfield DR. Hemopoietic stem cells with high
14. Clarkson BD, Fried J. Changing concepts of newal in culture of proliferative blast progenitor proliferative potential: assay of their concentra-
treatment in acute leukemia. Med Clin North Am. cells in acute myeloblastic leukemia. Blood. 1979; tion in marrow by the frequency and duration of
1971;55:561-600. 54:95-104. cure of W/Wv mice. J Clin Invest. 1982;70:242-
15. Killmann SA, Cronkite EP, Robertson JS, Fliedner 31. Maximov A. Der lymphozyt als gemeinsame 253.
TM, Bond VP. Estimation of phases of the life Stammzelle der verschieden Blutelemente in der 47. Paige CJ, Kincade PW, Moore MAS, Lee G. The
cycle of leukemic cells from labeling in human embryonalen Entwicklung und postfetalen Leben fate of fetal and adult B-cell progenitors grafted
beings in vivo with tritiated thymidine. Lab Invest. der Sugetiere. Folia Haematologica (Leipzig). into immunodeficient CBA/N mice. J Exp Med.
1963;12:671-684. 1909;8:125-141. 1979;150:548-563.
16. Gavosto F. The proliferative kinetics of the acute 32. Lichtman MA. The stem cell in the pathogenesis 48. Paige CJ, Kincade PW, Shinefeld LA, Sato VL.
leukaemias in relation to their treatment. Rev Eur and treatment of myelogenous leukemia: a per- Precursors of murine B lymphocytes: physical
Etud Clin Biol. 1970;15:1042-1047. spective. Leukemia. 2001;15:1489-1494. and functional characterization, and distinctions
BLOOD, 15 DECEMBER 2008 䡠 VOLUME 112, NUMBER 13 CANCER STEM CELLS 4805

from myeloid stem cells. J Exp Med. 1981;153: a single CD34-low/negative hematopoietic stem 88. Hogan CJ, Shpall EJ, Keller G. Differential long-
154-165. cell. Science. 1996;273:242-245. term and multilineage engraftment potential from
49. Hodgson GS, Bradley TR. Properties of hemato- 70. Kiel MJ, Yilmaz OH, Iwashita T, Yilmaz OH, subfractions of human CD34⫹ cord blood cells
poietic stem cells surviving 5-fluorouracil treat- Terhorst C, Morrison SJ. SLAM family receptors transplanted into NOD/SCID mice. Proc Natl
ment: evidence for a pre-CFU-S cell? Nature. distinguish hematopoietic stem and progenitor Acad Sci U S A. 2002;99:413-418.
1979;281:381-382. cells and reveal endothelial niches for stem cells. 89. Kerre TC, De Smet G, De Smedt M, et al.
50. Ploemacher RE, Brons RH. Separation of CFU-S Cell. 2005;121:1109-1121. Adapted NOD/SCID model supports develop-
from primitive cells responsible for reconstitution 71. Benveniste P, Cantin C, Hyam D, Iscove NN. He- ment of phenotypically and functionally mature T
of the bone marrow hemopoietic stem cell com- matopoietic stem cells engraft in mice with abso- cells from human umbilical cord blood CD34(⫹)
partment following irradiation: evidence for a pre- lute efficiency. Nat Immunol. 2003;4:708-713. cells. Blood. 2002;99:1620-1626.
CFU-S cell. Exp Hematol. 1989;17:263-266. 72. Kent DG, Dykstra BJ, Cheyne J, Ma E, Eaves CJ. 90. McKenzie JL, Gan OI, Doedens M, Dick JE. Hu-
51. Harrison DE. Competitive repopulation: a new Steel factor coordinately regulates the molecular man short-term repopulating stem cells are effi-
assay for long-term stem cell functional capacity. signature and biologic function of hematopoietic ciently detected following intrafemoral transplan-
Blood. 1980;55:77-81. stem cells. Blood. 2008;112:560-567. tation into NOD/SCID recipients depleted of
CD122⫹ cells. Blood. 2005;106:1259-1261.
52. Ford C, Hamerton J, Barns W, Loutet J. Cytologi- 73. Dick JE. Normal and leukemic human stem cells
cal identification of radiation chimeras. Nature. assayed in SCID mice. Semin Immunol. 1996;8: 91. Takenaka K, Prasolava TK, Wang JC, et al. Poly-
1956;177:452-454. 197-206. morphism in Sirpa modulates engraftment of hu-
man hematopoietic stem cells. Nat Immunol.
53. Wu AM, Till JE, Siminovitch L, McCulloch EA. Cy- 74. Mosier DE, Gulizia RJ, Baird SM, Wilson DB.
2007;8:1313-1323.
tological evidence for a relationship between nor- Transfer of a functional human immune system to
mal hemopoietic colony-forming cells and cells of mice with severe combined immunodeficiency. 92. Mazurier F, Doedens M, Gan OI, Dick JE. Rapid
the lymphoid system. J Exp Med. 1968;127:455- Nature. 1988;335:256-259. myeloerythroid repopulation after intrafemoral
463. 75. McCune JM, Namikawa R, Kaneshima H, Shultz transplantation of NOD-SCID mice reveals a new
LD, Lieberman M, Weissman IL. The SCID-hu class of human stem cells. Nat Med. 2003;9:959-
54. Abramson S, Miller RG, Phillips RA. The identifi-
mouse: murine model for the analysis of human 963.
cation in adult bone marrow of pluripotent and
restricted stem cells of the myeloid and lymphoid hematolymphoid differentiation and function. Sci- 93. Yahata T, Ando K, Sato T, et al. A highly sensitive
systems. J Exp Med. 1977;145:1567-1579. ence. 1988;241:1632-1639. strategy for SCID-repopulating cell assay by di-
76. Fraser CC, Kaneshima H, Hansteen G, Kilpatrick rect injection of primitive human hematopoietic
55. Dick JE, Magli MC, Huszar D, Phillips RA,
M, Hoffman R, Chen BP. Human allogeneic stem cells into NOD/SCID mice bone marrow. Blood.
Bernstein A. Introduction of a selectable gene into
cell maintenance and differentiation in a long- 2003;101:2905-2913.
primitive stem cells capable of long-term reconsti-
tution of the hemopoietic system of W/Wv mice. term multilineage SCID-hu graft. Blood. 1995;86: 94. Wang J, Kimura T, Asada R, et al. SCID-repopu-
Cell. 1985;42:71-79. 1680-1693. lating cell activity of human cord blood-derived
56. Keller G, Paige C, Gilboa E, Wagner E. Expres- 77. Baum CM, Weissman IL, Tsukamoto AS, Buckle CD34-negative cells assured by intra-bone mar-
sion of a foreign gene in myeloid and lymphoid A-S, Peault B. Isolation of a candidate human row injection. Blood. 2003;101:2924-2931.
cells derived from multipotent hemopoietic pre- hematopoietic stem-cell population. Proc Natl 95. Wang L, Menendez P, Shojaei F, et al. Generation
cursors. Nature. 1985;318:149-154. Acad Sci U S A. 1992;89:2804-2808. of hematopoietic repopulating cells from human
57. Lemischka IR, Raulet DH, Mulligan RC. Develop- 78. Kamel-Reid S, Dick JE. Engraftment of immune- embryonic stem cells independent of ectopic
mental potential and dynamic behavior of hema- deficient mice with human hematopoietic stem HOXB4 expression. J Exp Med. 2005;201:1603-
topoietic stem cells. Cell. 1986;45:917-927. cells. Science. 1988;242:1706-1709. 1614.

58. Lorenz E, Uphoff D, Reid TR, Shelton E. Modifi- 79. Lapidot T, Pflumio F, Doedens M, Murdoch B, 96. Dick JE, Lapidot T. Biology of normal and acute
cation of irradiation injury in mice and guinea pigs Williams DE, Dick JE. Cytokine stimulation of myeloid leukemia stem cells. Int J Hematol. 2005;
by bone marrow injections. J Natl Cancer Inst. multilineage hematopoiesis from immature hu- 82:389-396.
1951;12:197-201. man cells engrafted in scid mice. Science. 1992; 97. Wang JC, Dorrell C, Ito CY, et al. Normal and leu-
59. Thomas ED, Lochte HL Jr, Lu WC, Ferrebee JW. 255:1137-1141. kemic human stem cells assayed in immune-defi-
Intravenous infusion of bone marrow in patients 80. Vormoor J, Lapidot T, Pflumio F, et al. Immature cient mice. In: Zon LI, ed. Hematopoiesis: A De-
receiving radiation and chemotherapy. N Engl human cord blood progenitors engraft and prolif- velopmental Approach. New York, NY: Oxford
J Med. 1957;257:491-496. erate to high levels in severe combined immuno- University Press; 2001:99-118.
60. Gatti RA, Meuwissen HJ, Allen HD, Hong R, deficient mice. Blood. 1994;83:2489-2497. 98. Pflumio F, Izac B, Katz A, Shultz LD, Vainchenker
Good RA. Immunological reconstitution of sex- 81. Shultz L, Schweitzer P, Christianson S, et al. Mul- W, Coulombel L. Phenotype and function of hu-
linked lymphopenic immunological deficiency. tiple defects in innate and adaptive immunologi- man hematopoietic cells engrafting immune-defi-
Lancet. 1968;2:1366-1369. cal function in NOD/LtSz-scid mice. J Immunol. cient CB17-severe combined immunodeficiency
1995;154:180-191. mice and nonobese diabetic-severe combined
61. Buckner CD, Epstein RB, Rudolph RH, Clift RA,
immunodeficiency mice after transplantation of
Storb R, Thomas ED. Allogeneic marrow engraft- 82. Larochelle A, Vormoor J, Hanenberg H, et al.
human cord blood mononuclear cells. Blood.
ment following whole body irradiation in a patient Identification of primitive human hematopoietic
1996;88:3731-3740.
with leukemia. Blood. 1970;35:741-750. cells capable of repopulating NOD/SCID mouse
bone marrow: implications for gene therapy. Nat 99. Glimm H, Eisterer W, Lee K, et al. Previously un-
62. Evans MJ, Kaufman MH. Establishment in culture
Med. 1996;2:1329-1337. detected human hematopoietic cell populations
of pluripotential cells from mouse embryos. Na-
with short-term repopulating activity selectively
ture. 1981;292:154-156. 83. Cashman JD, Lapidot T, Wang JC, et al. Kinetic
engraft NOD/SCID-beta2 microglobulin-null mice.
evidence of the regeneration of multilineage he-
63. Martin GR. Isolation of a pluripotent cell line from J Clin Invest. 2001;107:199-206.
matopoiesis from primitive cells in normal human
early mouse embryos cultured in medium condi-
bone marrow transplanted into immunodeficient 100. Kerre TC, De Smet G, De Smedt M, et al. Both
tioned by teratocarcinoma stem cells. Proc Natl
mice. Blood. 1997;89:4307-4316. CD34⫹38⫹ and CD34⫹38- cells home specifi-
Acad Sci U S A. 1981;78:7634-7638.
84. Wang JC, Doedens M, Dick JE. Primitive human cally to the bone marrow of NOD/LtSZ scid/scid
64. Miller RG, Phillips RA. Separation of cells by ve- mice but show different kinetics in expansion.
hematopoietic cells are enriched in cord blood
locity sedimentation. J Cell Physiol. 1969;73:191- J Immunol. 2001;167:3692-3698.
compared with adult bone marrow or mobilized
201.
peripheral blood as measured by the quantitative 101. Guenechea G, Gan OI, Dorrell C, Dick JE. Dis-
65. Bonner WA, Hulett HR, Sweet RG, Herzenberg in vivo SCID-repopulating cell assay. Blood. tinct classes of human stem cells that differ in
LA. Fluorescence activated cell sorting. Rev Sci 1997;89:3919-3924. proliferative and self-renewal potential. Nat Im-
Instrum. 1972;43:404-409. munol. 2001;2:75-82.
85. Conneally E, Cashman J, Petzer A, Eaves C. Ex-
66. Visser JW, Bol SJ, van den Engh G. Character- pansion in vitro of transplantable human cord 102. Mazurier F, Gan O, McKenzie J, Doedens M, Dick
ization and enrichment of murine hemopoietic blood stem cells demonstrated using a quantita- J. Lentivector-mediated clonal tracking reveals
stem cells by fluorescence activated cell sorting. tive assay of their lympho-myeloid repopulating intrinsic heterogeneity in the human hematopoi-
Exp Hematol. 1981;9:644-655. activity in nonobese diabetic-scid/scid mice. Proc etic stem cell compartment and culture-induced
67. Baines P, Visser JW. Analysis and separation of Natl Acad Sci U S A. 1997;94:9836-9841. stem cell impairment. Blood. 2004;103:545-552.
murine bone marrow stem cells by H33342 fluo- 86. Ito M, Hiramatsu H, Kobayashi K, et al. NOD/ 103. Kamel-Reid S, Letarte M, Sirard C, et al. A model
rescence-activated cell sorting. Exp Hematol. SCID/gamma(c)(null) mouse: an excellent recipi- of human acute lymphoblastic leukemia in im-
1983;11:701-708. ent mouse model for engraftment of human cells. mune-deficient SCID mice. Science. 1989;246:
68. Spangrude GJ, Heimfeld S, Weissman IL. Purifi- Blood. 2002;100:3175-3182. 1597-1600.
cation and characterization of mouse hematopoi- 87. Ishikawa F, Yasukawa M, Lyons B, et al. Develop- 104. Uckun FM, Sather H, Reaman G, et al. Leukemic
etic stem cells. Science. 1988;241:58-62. ment of functional human blood and immune sys- cell growth in SCID mice as a predictor of relapse
69. Osawa M, Hanada K, Hamada H, Nakauchi H. tems in NOD/SCID/IL2 receptor ␥ chain (null) in high-risk B-lineage acute lymphoblastic leuke-
Long-term lymphohematopoietic reconstitution by mice. Blood. 2005;106:1565-1573. mia. Blood. 1995;85:873-878.
4806 DICK BLOOD, 15 DECEMBER 2008 䡠 VOLUME 112, NUMBER 13

105. Civin C, Strauss L, Brovall C, Fackler M, 124. Yilmaz OH, Valdez R, Theisen BK, et al. Pten de- evidence for cooperating genetic events in the
Schwartz J, Shaper J. Antigenic analysis of he- pendence distinguishes haematopoietic stem leukemic transformation of normal human hema-
matopoiesis: III. A hematopoietic progenitor cell cells from leukaemia-initiating cells. Nature. topoietic cells. Leukemia. 2005;19:1794-1805.
surface antigen defined by a monoclonal antibody 2006;441:475-482. 144. Mulloy JC, Cammenga J, Berguido FJ, et al.
raised against KG-1a cells. J Immunol. 1984;133: 125. Guan Y, Gerhard B, Hogge DE. Detection, isola- Maintaining the self-renewal and differentiation
157-165. tion, and stimulation of quiescent primitive leuke- potential of human CD34⫹ hematopoietic cells
106. Terstappen LW, Safford M, Unterhalt M, et al. mic progenitor cells from patients with acute my- using a single genetic element. Blood. 2003;102:
Flow cytometric characterization of acute myeloid eloid leukemia (AML). Blood. 2003;101:3142- 4369-4376.
leukemia: IV. Comparison to the differentiation 3149. 145. Chalandon Y, Jiang X, Christ O, et al. BCR-ABL-
pathway of normal hematopoietic progenitor 126. Guzman ML, Neering SJ, Upchurch D, et al. transduced human cord blood cells produce ab-
cells. Leukemia. 1992;6:993-1000. Nuclear factor-kappaB is constitutively activated normal populations in immunodeficient mice.
107. Lapidot T, Sirard C, Vormoor J, et al. A cell initiat- in primitive human acute myelogenous leukemia Leukemia. 2005;19:442-448.
ing human acute myeloid leukaemia after trans- cells. Blood. 2001;98:2301-2307. 146. Kennedy JA, Barabé F. Investigating human leu-
plantation into SCID mice. Nature. 1994;367:645- 127. Terpstra W, Ploemacher RE, Prins A, et al. Flu- kemogenesis from cell lines to in vivo models of
648. orouracil selectively spares acute myeloid leuke- human leukemia. Leukemia. 2008 Aug 7 [Epub
108. Bonnet D, Dick JE. Human acute myeloid leuke- mia cells with long-term growth abilities in immu- ahead of print].
mia is organized as a hierarchy that originates nodeficient mice and in culture. Blood. 1996;88: 147. Barabé F, Kennedy JA, Hope KJ, Dick JE. Model-
from a primitive hematopoietic cell. Nat Med. 1944-1950. ing the initiation and progression of human acute
1997;3:730-737.
128. So CW, Karsunky H, Passegue E, Cozzio A, leukemia in mice. Science. 2007;316:600-604.
109. Ailles LE, Gerhard B, Kawagoe H, Hogge DE. Weissman IL, Cleary ML. MLL-GAS7 transforms 148. Wei J, Wunderlich M, Fox C, et al. Microenviron-
Growth characteristics of acute myelogenous leu- multipotent hematopoietic progenitors and in- ment determines lineage fate in a human model
kemia progenitors that initiate malignant hemato- duces mixed lineage leukemias in mice. Cancer of MLL-AF9 leukemia. Cancer Cell. 2008;13:483-
poiesis in nonobese diabetic/severe combined Cell. 2003;3:161-171. 495.
immunodeficient mice. Blood. 1999;94:1761-
129. Huntly BJ, Shigematsu H, Deguchi K, et al. MOZ- 149. Greaves MF, Wiemels J. Origins of chromosome
1772.
TIF2, but not BCR-ABL, confers properties of leu- translocations in childhood leukaemia. Nat Rev
110. Terpstra W, Prins A, Visser T, et al. Conditions for kemic stem cells to committed murine hematopoi- Cancer. 2003;3:639-649.
engraftment of human acute myeloid leukemia etic progenitors. Cancer Cell. 2004;6:587-596.
(AML) in SCID mice. Leukemia. 1995;9:1573- 150. Hong D, Gupta R, Ancliff P, et al. Initiating and
130. Krivtsov AV, Twomey D, Feng Z, et al. Transfor- cancer-propagating cells in TEL-AML1-associ-
1577.
mation from committed progenitor to leukaemia ated childhood leukemia. Science. 2008;319:336-
111. Terpstra W, Prins A, Ploemacher RE, et al. Long- stem cell initiated by MLL-AF9. Nature. 2006;442: 339.
term leukemia-initiating capacity of a CD34-sub- 818-822.
population of acute myeloid leukemia. Blood. 151. Kelly PN, Dakic A, Adams JM, Nutt SL, Strasser
131. Cozzio A, Passegue E, Ayton PM, Karsunky H, A. Tumor growth need not be driven by rare can-
1996;87:2187-2194.
Cleary ML, Weissman IL. Similar MLL-associated cer stem cells. Science. 2007;317:337.
112. Pearce DJ, Taussig D, Zibara K, et al. AML en- leukemias arising from self-renewing stem cells
graftment in the NOD/SCID assay reflects the and short-lived myeloid progenitors. Genes Dev. 152. Williams RT, den Besten W, Sherr CJ. Cytokine-
outcome of AML: implications for our understand- 2003;17:3029-3035. dependent imatinib resistance in mouse BCR-
ing of the heterogeneity of AML. Blood. 2006;107: ABL⫹, Arf-null lymphoblastic leukemia. Genes
132. Passegue E, Wagner EF, Weissman IL. JunB de- Dev. 2007;21:2283-2287.
1166-1173.
ficiency leads to a myeloproliferative disorder
113. van Rhenen A, Feller N, Kelder A, et al. High arising from hematopoietic stem cells. Cell. 2004; 153. Somervaille TC, Cleary ML. Identification and
stem cell frequency in acute myeloid leukemia at 119:431-443. characterization of leukemia stem cells in murine
diagnosis predicts high minimal residual disease MLL-AF9 acute myeloid leukemia. Cancer Cell.
and poor survival. Clin Cancer Res. 2005;11: 133. Chen W, Kumar AR, Hudson WA, et al. Malignant 2006;10:257-268.
6520-6527. transformation initiated by Mll-AF9: gene dosage
and critical target cells. Cancer Cell. 2008;13: 154. Kennedy JA, Barabé F, Poeppl AG, Wang JC,
114. Fialkow PJ, Singer JW, Adamson JW, et al. Acute 432-440. Dick JE. Comment on “Tumor growth need not be
nonlymphocytic leukemia: heterogeneity of stem driven by rare cancer stem cells.” Science. 2007;
cell origin. Blood. 1981;57:1068-1073. 134. Wang Y, Armstrong SA. Cancer: inappropriate 318:1722; author reply 1722.
expression of stem cell programs? Cell Stem
115. Fialkow P, Singer J, Raskind W, et al. Clonal de- Cell. 2008;2:297-299. 155. Liu R, Wang X, Chen GY, et al. The prognostic
velopment, stem-cell differentiation, and clinical role of a gene signature from tumorigenic breast-
remissions in acute nonlymphocytic leukemia. 135. Miyamoto T, Weissman IL, Akashi K. AML1/ETO- cancer cells. N Engl J Med. 2007;356:217-226.
N Engl J Med. 1987;317:468-473. expressing nonleukemic stem cells in acute my-
elogenous leukemia with 8;21 chromosomal 156. Guzman ML, Rossi RM, Neelakantan S, et al. An
116. Fearon E, Burke P, Schiffer C, Zehnbauer B, orally bioavailable parthenolide analog selectively
translocation. Proc Natl Acad Sci U S A. 2000;97:
Vogelstein B. Differentiation of leukemia cells to eradicates acute myelogenous leukemia stem
7521-7526.
polymorphonuclear leukocytes in patients with and progenitor cells. Blood. 2007;110:4427-4435.
acute nonlymphoblastic leukemia. N Engl J Med. 136. Majeti R, Park C, Weissman I. Identification of a
hierarchy of multipotent hematopoietic progeni- 157. Guzman ML, Li X, Corbett CA, et al. Rapid and
1986;315:15-24. selective death of leukemia stem and progenitor
tors in human cord blood. Cell Stem Cell. 2007;1:
117. Fialkow P, Denman A, Jacobson R, Lowenthal M. cells induced by the compound 4-benzyl,
635-645.
Chronic myelogenous leukemia: origin of some 2-methyl, 1,2,4-thiadiazolidine, 3,5 dione (TDZD-
lymphocytes from leukemic stem cells. J Clin In- 137. Jamieson CH, Ailles LE, Dylla SJ, et al. Granulo- 8). Blood. 2007;110:4436-4444.
vest. 1978;62:815-823. cyte-macrophage progenitors as candidate leuke-
mic stem cells in blast-crisis CML. N Engl J Med. 158. Hassane DC, Guzman ML, Corbett C, et al. Dis-
118. Al-Hajj M, Wicha M, Morrison SJ, Clarke MF. Pro- covery of agents that eradicate leukemia stem
2004;351:657-667.
spective identification of tumorigenic breast can- cells using an in silico screen of public gene ex-
cer cells. Proc Natl Acad Sci U S A. 2003;100: 138. Nowell P, Hungerford D. A minute chromosome in pression data. Blood. 2008;111:5654-5662.
3983-3988. human chronic granulocytic leukemia. Science.
1960;132:1497. 159. Feuring-Buske M, Frankel A, Gerhard B, Hogge
119. Singh SK, Hawkins C, Clarke ID, et al. Identifica- D. Variable cytotoxicity of diphtheria toxin 388-
tion of human brain tumour initiating cells. Nature. 139. Rowley JD. A new consistent chromosomal ab- granulocyte-macrophage colony-stimulating fac-
2004;432:396-401. normality in myelogenous leukemia identified by tor fusion protein for acute myelogenous leuke-
quinacrine fluorescence and giemsa staining. Na- mia stem cells. Exp Hematol. 2000;28:1390-
120. Clarke MF, Dick JE, Dirks PB, et al. Cancer stem
ture. 1973;243:290-293. 1400.
cells—perspectives on current status and future
directions: American Association for Cancer Re- 140. Mani SA, Guo W, Liao MJ, et al. The epithelial- 160. Terpstra W, Rozemuller H, Breems DA, et al.
search Workshop on Cancer Stem Cells. Cancer mesenchymal transition generates cells with Diphtheria toxin fused to granulocyte-macro-
Res. 2006;66:9339-9344. properties of stem cells. Cell. 2008;133:704-715. phage colony-stimulating factor eliminates acute
121. Jordan CT, Lemischka IR. Clonal and systemic 141. Darley RL, Hoy TG, Baines P, Padua RA, Burnett myeloid leukemia cells with the potential to initiate
analysis of long-term hematopoiesis in the AK. Mutant N-RAS induces erythroid lineage dys- leukemia in immunodeficient mice, but spares
mouse. Genes Dev. 1990;4:220-232. plasia in human CD34⫹ cells. J Exp Med. 1997; normal hemopoietic stem cells. Blood. 1997;90:
122. Hope K, Jin L, Dick JE. Acute myeloid leukemia 185:1337-1347. 3735-3742.
originates from a hierarchy of leukemic stem cell 142. Pereira DS, Dorrell C, Ito CY, et al. Retroviral 161. Frankel A, Liu JS, Rizzieri D, Hogge D. Phase I
classes that differ in self-renewal capacity. Nat transduction of TLS-ERG initiates a leukemo- clinical study of diphtheria toxin-interleukin 3 fu-
Immunol. 2004;5:738-743. genic program in normal human hematopoietic sion protein in patients with acute myeloid leuke-
123. Lessard J, Sauvageau G. Bmi-1 determines the cells. Proc Natl Acad Sci U S A. 1998;95:8239- mia and myelodysplasia. Leuk Lymphoma. 2008;
proliferative capacity of normal and leukemic 8244. 49:543-553.
stem cells. Nature. 2003;423:455-460. 143. Warner JK, Wang JC, Takenaka K, et al. Direct 162. Bonnet D, Warren EH, Greenberg PD, Dick JE,
BLOOD, 15 DECEMBER 2008 䡠 VOLUME 112, NUMBER 13 CANCER STEM CELLS 4807

Riddell SR. CD8(⫹) minor histocompatibility anti- lates migration and development of human acute marker for human acute myelogenous leukemia
gen-specific cytotoxic T lymphocyte clones elimi- myelogenous leukemia stem cells in transplanted stem cells. Leukemia. 2000;14:1777-1784.
nate human acute myeloid leukemia stem cells. NOD/SCID mice. Cancer Res. 2004;64:2817-
169. Lock R, Jin L, Lee E, et al. CD123 (IL-3 receptor
Proc Natl Acad Sci U S A. 1999;96:8639-8644. 2824.
chain) neutralization by a monoclonal antibody
163. Rosinski KV, Fujii N, Mito JK, et al. DDX3Y en- 166. Papayannopoulou T, Scadden DT. Stem-cell ecol- selectively eliminates human acute myeloid leu-
codes a class I MHC-restricted H-Y antigen that ogy and stem cells in motion. Blood. 2008;111: kemic stem cells. Blood. 2007;110:161.
is expressed in leukemic stem cells. Blood. 2008; 3923-3930.
111:4817-4826. 167. Ishikawa F, Yoshida S, Saito Y, et al. Chemo- 170. Ito K, Bernardi R, Morotti A, et al. PML targeting
164. Jin L, Hope KJ, Zhai Q, Smadja-Joffe F, Dick JE. therapy-resistant human AML stem cells home to eradicates quiescent leukaemia-initiating cells.
Targeting of CD44 eradicates human acute my- and engraft within the bone-marrow endosteal Nature. 2008;453:1072-1078.
eloid leukemic stem cells. Nat Med. 2006;12: region. Nat Biotechnol. 2007;25:1315-1321. 171. Wang JC. Evaluating therapeutic efficacy against
1167-1174. 168. Jordan CT, Upchurch D, Szilvassy SJ, et al. The cancer stem cells: new challenges posed by a
165. Tavor S, Petit I, Porozov S, et al. CXCR4 regu- interleukin-3 receptor alpha chain is a unique new paradigm. Cell Stem Cell. 2007;1:497-501.

My introduction to hematology began during my postdoctoral fellowship with Alan Bernstein at


the storied Ontario Cancer Institute. This was in the mid-1980s during the early days of retrovirus-
mediated gene transfer and the breathless prediction that gene therapy was imminent and would
be the cure of many human diseases. Prior work from this group had already developed retroviral
vectors and provided the first evidence of transduction of primary murine clonogenic progeni-
tors; my project was to develop the means to transduce the hematopoietic stem cell. Although
there was intense competition to be the first group to show hematopoietic stem cell transduction,
the 2 most important lessons I learned in this period were: always look back to the past to guide
the directions of the future so as not to “reinvent the wheel,” and the drive to do research should
come from the intrinsic value and importance of the question being asked rather than from look-
ing over your shoulder to stay ahead of an imagined or real competitor. The project was a close
collaboration with Cristina Magli, a postdoctoral fellow in Dr Bob Phillips’ laboratory, who shared
a wealth of stem cell experience with me. Moreover, Bob was a leading member of the hematopoi-
esis and immunology research program at the Ontario Cancer Institute and had carried out a se-
ries of pioneering studies in stem cell biology, and clonal tracking of repopulating cells in par-
ticular. In response to some “new” idea or experimental design that Cristina and I would come up
with, our weekly laboratory meetings were often punctuated by critical observations from Bob
and Alan, including “well, in 1960-1970 Till and McCulloch, Metcalf, Dexter, we, etc, carried out
such-and-such experiments.” This prevented us from entering many a blind alley and deeply in-
stilled the principle that clonal long-term repopulation assays are the only ones that mattered.
Indeed, one could argue that my whole career has been spent repeating my postdoctoral project
over and over again, albeit in human cells. So the lesson for new trainees is that a judicious
choice of project and mentor can have benefits that last a lifetime!
Because this review already describes the pathway of discovery of our work once I established
John E. Dick my own laboratory, I will only make 2 additional points. First, time and place are essential. As the
review notes, I was fortunate to establish my laboratory at a time when key technologies were
sufficiently developed to enable our work. I was also fortunate to be surrounded by a remarkable group of colleagues in Toronto who set high standards
of excellence to aspire to but who were also committed to creating an unusually collaborative and collegial environment with few internal competitions.
Second, trainees drive research. I have been privileged to work with a wonderful group of trainees who drove all the work mentioned in the review. It was
their commitment to the projects and willingness to engage in a selfless fashion to the ferment of ideas from which research directions emerged and
evolved.

Das könnte Ihnen auch gefallen