Although uncontrolled proliferation is a distinguishing property of a tumor as a whole, the individual cells that make up the tumor exhibit considerable variation in many properties, including morphology, proliferation kinetics, and the ability to initiate tumor growth in transplant assays. Understanding the molecular and cellular basis of this heterogeneity has important implications in the design of therapeutic strategies. The mechanistic basis of tumor heterogeneity has been uncertain; however, there is now strong evidence that cancer is a cellular hierarchy with cancer stem cells at the apex. This review provides a historical overview of the influence of hematology on the development of stem cell concepts and their linkage to cancer.

Tumors from different patients, whether leukemic or solid, exhibit significant heterogeneity in terms of morphology, cell surface markers, genetic lesions, cell proliferation kinetics, and response to therapy. Heterogeneity of all these features is also seen within an individual tumor that is clonal (ie, initiated from a single cell). Although individual cells of a tumor all share common genetic aberrations reflective of their clonal origin, single-cell analysis has shown the existence of variation in genetic and epigenetic abnormalities between different cells or locations within a tumor. The cellular and molecular basis for this heterogeneity represents a fundamental problem that has interested cancer researchers for many decades. One possible explanation is that all tumor cells 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 with (cancer) stem cells at the apex.1  This review will focus on 50 years of hematology research that played a role in garnering evidence to support the hierarchy model

There is mounting evidence from cell purification studies that a subset of cells, termed cancer stem cells (CSCs), or cancer-initiating cells, that are distinct from the bulk of the tumor are responsible for long-term maintenance of tumor growth in several cancers.2  Evidence is strongest for the acute leukemias, although recent studies have identified the existence of CSCs in an increasingly longer list of solid tumors, including brain, breast, and colon.3  This conceptual shift has important implications, not only for researchers seeking to understand mechanisms of tumor initiation and progression but also for developing and evaluating effective anticancer therapies. Although the clinical relevance of CSCs beyond experimental models is still lacking, the high frequency of relapse after conventional cytotoxic chemotherapies predicts that CSCs survive standard treatments. Many papers, reviews, and funding bodies are making strong predictions that targeting CSCs more effectively will lead to improved patient outcomes. Indeed, there has been almost breathless excitement that this “new” field of CSC biology will solve the stubbornly high relapse rates that continue to exist for many cancers. However, stem cell concepts and how they apply to cancer are decades old, and many obstacles to the development of effective anticancer therapies still exist. It is important to understand the historical underpinnings of the CSC field to recognize the challenges we still face. By understanding where we came from, we will have a clearer view of the way forward. The history of stem cell research, and cancer stem cell research in particular, is long and better suited for a book rather than a short review. Thus, I only touch on a few highlights that I find significant and that played a role in guiding our own work in this field; most of my comments revolve around studies in hematopoiesis and acute myeloid leukemia (AML) specifically. My apologies go to those whose work I was not able to include or could only be treated in a superficial fashion.

The notion that cancer is a disease of abnormal or uncontrolled proliferation has governed cancer research for more than 7 decades. Because normal tissues also undergo continuous regeneration, albeit at rates specific to each organ, the historical thrust of cancer research was to develop drugs that targeted the enhanced rate of neoplastic proliferation. Clinicians used doses and schedules that achieved many log orders of cell kill while maintaining acceptable levels of toxicity to normal tissues. Based on a deep understanding of the molecular basis of cancer, current therapeutic strategies now focus on inhibiting the molecular drivers of cancer (eg, trastuzumab-Her2/Neu, imatinib-bcr/abl, erlotonib, epidermal growth factor receptor). With an increased appreciation of the extrinsic microenvironmental factors that support tumor growth, such as the vasculature, approaches that interfere with these factors (ie, bevacizumab, vascular endothelial growth factor) are also being brought forward. What all these strategies, whether from the past or present, have in common is the tacit perspective of cancer as a homogeneous, abnormal entity. Therefore, drugs targeting molecular lesions should be equally effective against all tumor cells, barring the emergence of resistant subclones. The goal is to ensure that enough drug(s) is given to target the primary molecular pathway and then combined with drugs that target other relevant or compensatory pathways to enable killing of every tumor cell. Indeed, historically, even research to understand the basic biology of cancer uses strategies that treat an individual cancer as functionally homogeneous. Much cancer research, even to the present day, follows a paradigm where the entire tumor or an entire dish of tissue culture cells is used for genetic, proteomic, or biochemical studies, mostly because the technology used to interrogate biologic processes (ie, proteomics, biochemistry) cannot be done on single cells and requires large cell numbers. Clearly, this has been a productive approach as evidenced by the thorough understanding gained on the genetic and epigenetic basis of the neoplastic process. However, an inability to deal meaningfully with limiting cell numbers has forced the cancer research enterprise to operate within a dichotomy because it is also well known that tumors are not homogeneous but exhibit significant heterogeneity.

Since the time of the great experimental pathologists (Virchow, Maximov, and others), it has been recognized that tumors, be they liquid or solid, exhibit marked morphologic heterogeneity. With the advent of transplantation of murine cancer cell lines into mice, Furth and Kahn in 1937 were able to establish that a single cell, and not a transmissible agent, could initiate a cancer graft.4  As others applied transplantation assays to a variety of solid tumors and leukemias throughout the next several decades, it became clear that there was wide variation in the frequency of cells that initiate cancer growth.5-7  Remarkably, human studies have been carried out since the late 1800s (reviewed by Southam et al8 ) where tumors were excised, single-cell suspensions made and autotransplanted into the thigh at different cell doses. These studies showed that tumor reinitiation was variable and rare, often requiring more than 106 cells. Finally, in an elegant series of experiments, Pierce et al showed that malignant teratocarcinoma cells can spontaneously differentiate into mature benign cells.9  Thus, a tumor can be considered a hierarchy defined by a maturation process in the same way that normal tissue development occurs. In 1961, Pierce and Speers proposed the idea that tumors are caricatures of normal development and that this maturation process could be harnessed as a means of therapy.10 

The concept of cellular heterogeneity reached its zenith with the functional cell proliferation studies of the 1940s, 1950s, and 1960s. The advent of radiolabeling cells, combined with autoradiography by Belanger and Leblond in 1946, enabled precise measurements of the proliferation, lifespan, and hierarchical relationships in normal and neoplastic tissues.11  Clermont and Leblond showed, for example, that the testis contained proliferating cells that took up a pulse of label and then by tracking the label showed their differentiation to mature cells.12 ; they also noted that these cells maintained themselves. In the 1960s and 1970s, several prominent hematologists, including Clarkson et al,13,14  Killmann et al,15  Gavosto,16  and others, carried out pioneering studies that were (perhaps more than any other findings) the studies establishing that tumors exhibited functional heterogeneity. These were cytokinetic studies carried out first in cell lines and murine models of the acute leukemias and then, remarkably by in vivo examination of leukemia blast proliferation kinetics in human AML and acute lymphoblastic leukemia (ALL) patients. The in vivo data could not have been clearer; the majority of leukemic blasts were postmitotic and needed to be continuously replenished from a relatively small proliferative fraction17  (Figure 1). Only a small fraction (∼ 5%) of leukemic blasts was rapidly cycling in vivo. However, careful analysis demonstrated that there were 2 proliferative fractions in patients: a larger, fast cycling subset with a 24-hour cell cycle time and a smaller, slow cycling fraction with a dormancy estimated to last from weeks to months. In particular, Clarkson, Rubinow, Skipper and their colleagues inferred that the slow cycling fraction was actually generating the fast cycling fraction and suggested these represented a leukemic stem cell (LSC) population because they had similar cytokinetic properties to those observed for normal hematopoietic stem cells (HSCs).18  They noted that the dormant cells were much smaller in size and had less granularity than the rapidly proliferating fraction, which led to speculation that these cells could represent distinct cellular fractions. Of course, such studies could not prove whether the cycling fractions represented the same population, existing in either a proliferative or a nonproliferative state, or whether these observations point to a hierarchical relationship representing 2 functionally distinct cell types. Even so, based on these observations, reviews of this era predicted that the inability to eradicate the theoretical LSCs represented the cause of relapse and the ultimate failure for antiproliferative chemotherapies, which formed the main chemotherapeutic armament at the time.14,19  Investigators then combined in vivo cytokinetic studies with drug treatment and tailored new therapies based on these findings. They made the interesting observation that LSCs respond to depletion of the leukemia cell mass that occurs when antiproliferative drugs are administered to AML patients. LSCs begin to go into cycle and expand in the same way normal stem cells go into cycle after chemotherapy-induced cytopenias. Thus, they suggested that one way to eliminate dormant LSCs would be to find the window when they cycle to kill them when they are most vulnerable. Indeed, the cytokinetic findings represented the underpinning for pioneering clinical trials carried out by Clarkson and his colleagues at Memorial Sloan-Kettering Cancer Center that used multiple drugs.20  The fundamental problem investigators faced at this time was the inability to directly identify and assay LSCs, making it impossible to characterize them and harness the acquired knowledge for therapeutic approaches in the future. Pointed comments made at least as early as 1970 predicted that antiproliferative chemotherapy agents would not effectively eradicate these dormant LSCs and that future progress must focus on understanding their properties.17  These remarkably prescient observations retain their potency and freshness even today, nearly 40 years later.

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.

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.

Close modal

The cytokinetic studies drove interest away from the study of bulk leukemias because they were mostly composed of postmitotic blasts. Because there was little that could be done to assay the dormant LSC, attention focused on the development of a clonogenic assay for the rapidly proliferating leukemic cells (AML-colony-forming unit [CFU]) that the in vivo studies had uncovered. McCulloch et al,21,22  Moore and Metcalf et al23,24 , Griffin and Löwenberg,25  and Dicke et al26  pioneered these assays and made detailed insights into their properties. Parallel clonal assays were developed for solid tumor clonogenic progenitors.27  In a key series of experiments, Sabbath et al developed a series of monoclonal antibodies (mAb) directed against myeloid differentiation antigens and assessed the phenotype of the AML-CFU itself by complement mediated lysis followed by clonogenic assay.28  Parallel studies were done with normal cells. The AML-CFU from different patients could be classified into 3 groups: primitive (similar to multipotent normal CFU), early lineage committed (similar to normal day 14 CFU-GM), and late (similar to day 7 CFU-GM). This work established that the AML-CFU were distinct from most leukemic blasts and the leukemic cells within an individual colony could be more differentiated than the individual AML-CFU that originated the colony. These clonal data provided the first strong proof for a hierarchy in AML. Moreover, this work suggested that the different classes of AML-CFU might originate from normal cells at different stages of differentiation.25  McCulloch et al showed additional evidence for heterogeneity with the finding that the AML-CFU were heterogeneous in their serial replating ability, some unable to generate secondary colonies whereas others had high capacity.22,29,30  Because this is an indicator of self-renewal, they predicted that some AML-CFU were more primitive than others. Although the properties of AML-CFU (number, proliferation, drug sensitivity) on their own did not predict clinical disease properties or response to therapy, the secondary replating capacity had better predictive power. The AML-CFU assay was used intensively from the late 1960s to the early 1980s and resulted in many important insights into AML biology. However, the limitations of the assay and the lessons that had been learned from the study of normal hematopoiesis underscored the need for functional in vivo assays to determine whether an LSC, earlier than AML-CFU, existed in AML.

Collectively, the data described in “Tumor heterogeneity” firmly established that tumors are not only morphologically heterogeneous as observed more than 100 years ago, but they are functionally heterogeneous in terms of cell proliferation or even tumor initiation based on transplant assays. Two mutually exclusive models could explain tumor heterogeneity. The stochastic model predicts that a tumor is biologically homogeneous and the behavior of the cancer cells is influenced by intrinsic (eg, levels of transcription factors, signaling pathways) or extrinsic (eg, host factors, microenvironment, immune response) factors. These influences are unpredictable or random and result in heterogeneity in the expression of cell surface markers or other markers of maturation, in entry to cell cycle, or in tumor initiation capacity. A key tenet of this model is that all cells of the tumor are equally sensitive to such stochastic influences and also that tumor cells can revert from one state to another because these influences do not induce permanent changes.1  In contrast, the hierarchy model predicts that the tumor is a caricature of normal tissue development where stem cells maintain normal tissue hierarchies (skin, colon, blood). In this 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 the existence of “functional leukemic stem cells,” cells that behave as leukemia stem cells. The essential difference is that, in the stochastic model, LSCs arise randomly and every tumor cell has the potential to behave like an LSC given the right influences; whereas in the hierarchy model, there is only a distinct subset of cells that has the potential to behave like LSCs. The only means to determine which model is correct is prospective isolation of cell fractions followed by clonal analysis with a functional LSC assay. If the hierarchy model is correct, then it should be possible to sort tumors into fractions that have LSC activity and those that do not. In contrast, the stochastic model predicts that LSC activity may be observed in any fraction and cannot be prospectively isolated because all tumor cells have the same potential (even if it is low) for tumor initiation.

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.

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.

Close modal

The term “leukemia stem cell” had been in use for many decades, and a large part of the hematology community assumed that the hierarchy model was true and the leukemia clone existed in a hierarchy sustained by an LSC, with obvious parallels to the role that HSCs play in sustaining the normal hematopoietic hierarchy. However, formal proof that LSCs had stem cell attributes and were distinct from the rest of the leukemic cells that could never behave as LSCs was still lacking. Clearly, technologies that were not available in the 1970s were required to move the field forward. However, I do not think the absence of technology can fully explain why lags existed from the mid-1970s when all the theoretical principles pointing to the existence of LSCs and a description of their properties were well worked out, to the mid-1990s when direct proof for the existence of LSC was finally discovered, and then to the most recent years when CSC concepts have been 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 environment was primed with the groundbreaking studies of normal HSCs, eventually leading to HSC transplant as therapy and to the crossover of universal stem cell concepts into many areas of biology. This created a backdrop in which technologies were developing, including the use of flow cell sorters in combination with mAb that bind cell surface markers and the development of human xenograft assays for human HSCs and LSCs. Because these conditions set the stage for discovery of LSCs and eventually CSCs from solid tumors, it is instructive to review the key highlights that culminated to identify HSCs and the universal concepts these stem cell studies generated.

The modern era of stem cell research can be traced back to the seismic shift in the 1960s when Till and McCulloch moved HSC research from a descriptive science to a quantitative science. Although careful studies using microscopic analysis pointed to the idea of a common stem cell for all blood lineages,31  equally robust was the view that independent stem cells existed for each major blood lineage, which was promulgated by textbooks from the 1940s.32  The experimental finding by Till and McCulloch that the “regeneration nodules” on the spleen of lethally irradiated mice (termed CFU-spleen [CFU-S])33  transplanted with BM cells were clonal (based on X-ray induced chromosomal lineage tracking34 ) and contained multiple blood lineages proved that the hematopoietic system derives from a multipotent HSC rather than from a series of lineage-specific stem cells. With a clonal, quantitative, and functional assay in hand, in short order they established all of the major stem cell principles under which we continue to operate today: HSCs have a high capacity for self-renewal, proliferation, and multipotential differentiation. Their observations that the self-renewal capacity of individual CFU-S was highly variable35  led them to propose the concept that the control of HSC self-renewal was governed by stochastic principles,36  a hypothesis that is still being tested and still able to provoke strong opinions among stem cell biologists.37-41  Their focus on the need for quantitative functional assays prompted the development of in vitro clonal progenitor assays by Pluznik and Sachs42  and Bradley and Metcalf43  that were central to the later discovery of hematopoietic colony stimulating factors (as reviewed in the 50th Anniversary Review by Metcalf44 ). With the use of progenitor assays, it was possible to describe precisely the lineage relationships that occur during maturation. Despite widespread use of the spleen colony assay as a surrogate HSC assay, by the mid 1980s it was clear that a more primitive HSC must exist. First, CFU-S were heterogeneous 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 5-fluorouracil treatment, CFU-S were killed whereas other dormant cells capable of CFU-S replenishment survived, suggesting the existence of a pre-CFU-S stem cell.49,50  Harrison, in particular, drove the paradigm that cells with long-term repopulation capacity could be distinguished from those with short-term potential.51  Using chromosomal translocation as a means of cellular marking (first developed by Ford et al52 ), in 1968 the first clonal evidence established multilineage repopulation, including CFU-S and B cells.53  Using the same approach, Abramson et al showed that repopulation of myeloid and lymphoid lineages could derive from a long-term multipotential stem cell or from lineage-restricted stem cells.54  However, the efficiency of marking was low and the need to irradiate the bone marrow to induce chromosomal translocation meant that it was difficult to establish whether the identified HSC classes truly existed or were experimentally induced artifacts. With the development of more benign methods for HSC clonal tracking using retroviral insertion sites, it was possible to establish that long-term, multipotential HSCs existed.55-57  Collectively, these studies established that clonal assays are essential to uncover stem cell properties; and when individual HSCs are assessed, they exhibit wide variations in repopulation dynamics and self-renewal capacity.

While these concepts were solidifying, stem cell concepts began to percolate into other research fields through a series of culminating events and set the stage for further discovery. Perhaps the most influential factor that drove stem cell concepts beyond the domain of a small number of stem cell biologists into a wider sphere of biomedical research was the development of HSC transplantation for treatment of leukemia and other hematologic diseases. Through a series of elegant studies in the 1950s, proof was provided that cells (as opposed to a humoral factor) from the bone marrow of mice could protect against lethal doses of radiation by effectively regenerating all of the blood lineages in transplant recipients.52,58  In 1956, Thomas et al carried out the first human bone marrow transplantations,59  although initially a failure, they brought a decade of improvements in tissue typing and methods for controlling graft-vs-host disease. Finally, the pioneering clinical trials of Good, Thomas, and others showed that human BM could be used to cure immune deficiency by sibling donor transplants in 196860  and neoplastic disease in 1969,61  beginning the modern era of human allogeneic marrow grafting. During the same time period, the new field of molecular biology was established. This made it possible not only to use genetics to characterize biologic processes in a much more rapid fashion, but also to produce biologic regulators on an industrial scale. All of these research paths collided in the 1980s with the birth of the biotechnology industry, which was mostly focused on the commercialization of products to stimulate blood cells. Thus, the arcane fields of blood diseases, HSC transplantation, and stem cells in general had moved from a backwater to front stage. The final influence in this fertile period was the discovery of embryonic stem cells by Evans and Kaufman62  and Martin63  in 1981 with the attendant promise of regenerative medicine. Stem cell biology had finally become a hot topic, and it had begun to capture the attention of a broad swath of the research community.

It was within this milieu that 2 important technologic advances took place that directly led to the renewal of interest in the concepts of LSC from 20 years earlier. The first was the development of fluorescence-activated cell sorting (FACS), and the second was the development of xenotransplantation assays for normal and leukemic human stem cells. Although Miller and Phillips had developed the first large-scale methods for cell separation based on sedimentation,64  these methods lacked the precision needed for meticulous cell fractionation. The development of FACS by Herzenberg's group at Stanford in 1972,65  combined with technology to create specific mAbs against cell surface differentiation antigens, finally provided the means to fractionate cells with high purity. As applied to stem cell fractionation, Visser et al were the first to use flow sorting based on wheat germ agglutinin binding as a means to enrich (6-fold) CFU-S compared with unfractionated bone marrow cells.66  Later it was possible to sort on the basis of cell cycle status using Hoechst dyes, and in this way CFU-S were distinguished from the more actively dividing granulocyte-macrophage-CFU progenitor cells.67  Improved cell fractionation based on cell surface markers and longer-term functional assays culminated in an important paper by Spangrude et al who purified mouse multipotential HSCs (although mostly those capable of relatively short-term repopulation) by combining depletion of several markers of maturation with expression of the Sca1 and c-Kit proteins (LSK cells).68  Improvements in markers and the use of single-cell transplantations have now resulted in essentially pure populations of HSCs capable of multipotential repopulation.40,69-72  However, only 20% to 50% of these clonal HSCs are capable of long-term 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 from those with more limited capacity, or whether this remaining variability is because of stochastic influences on an otherwise pure HSC population.39,40  The intense efforts to purify murine HSCs and their downstream progeny established a pathway of discovery that could be applied to characterize human hematopoiesis.

Because studies of murine hematopoiesis showed that HSCs are distinct from CFU and that HSCs with short-term repopulation capacity are distinct from those with long-term capacity, it was clear that progress to understand human hematopoiesis would require a repopulation assay. The second key technologic development leading to the identification of LSC was the beginning of the modern era of the xenotransplantation system, which provided the key ability to functionally assay both normal stem cells and LSCs.73  Although there was a long history of transplanting human tumor cells into irradiated or immune-deficient nude mice, the strong host resistance in these mice allowed only the most aggressive tumors and cell lines to be engrafted. Primary human leukemia cells could not be transplanted and most cell lines only grew subcutaneously or as ascites, which was not a normal pattern for hematologic proliferation. In 1988, 3 groups using severe combined immunodeficiency (SCID) or bg/nu/xid (BNX) mice, which are more immune-deficient than nude mice, showed that normal human hematopoietic cells could repopulate these mice. Mosier et al transplanted human peripheral blood leukocytes by intraperitoneal injection into SCID mice to enable long-term generation of mature T cells that could then be infected with HIV.74  McCune et al were also interested in developing an HIV model. They implanted SCID mice with pieces of human fetal liver, lymph node, and thymus,75  and later they injected human cells into fetal bone.76  The rationale of their approach was to provide a human tissue microenvironment that would continuously generate human T cells for HIV studies because the fetal liver (or fetal bone) would provide a source of stem cells. Of course, the long-term production of human cells within the fetal bone could be used to assay and identify HSCs, a premise that came to fruition when they showed that purified CD34+Thy+ cells could initiate multilineage engraftment of this bone fragment.77  Our approach was somewhat different and guided by the principles of bone marrow transplantation in humans and mice: intravenous transplantation into irradiated recipients resulting in human cells repopulating the hematopoietic tissues of a mouse.78-80  With improvements in recipient mice,81  it was finally possible to quantify and identify the cell that was capable of initiating a multilineage human graft, which was termed the SCID-repopulating cell (SRC).82-85  The SRC measured in this system generated many orders of magnitude higher cell numbers than the fetal bone in the SCID-hu model, providing a more robust 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 in both NK and macrophage activity86,87  or that have been treated with a mAb to CD122 that depletes these components of the innate immune system.88-90  Recent studies that identified polymorphic alleles of Sirp-alpha governing human HSC engraftment in NOD/SCID mice establish the importance of controlling macrophages to achieve HSC engraftment.91  Finally, direct intrafemoral delivery of cells resulted in more sensitive SRC detection, as well as assessment of progenitors that possess transient repopulation potential, or are lineage restricted and would not read out in classic intravenous-based HSC transplantation assays.90,92-95 

The SRC xenotransplantation assay has emerged as the “gold standard” surrogate assay for human HSCs.96  Combined with clonogenic and stromal-based assays, it has been possible to identify many cell surface markers that define the developmental hierarchy of human hematopoiesis, setting the stage to apply these same markers to understanding leukemic hematopoiesis. A detailed understanding of the repopulation potential, cell-surface phenotype, differentiation, and proliferation properties of SRCs has been gained over the past 15 years.96,97  Most SRCs express CD34, and distinct SRC activities can be found within the lineage-negative (Lin) CD34+CD38 and LinCD34+CD38lo subfractions.88,92,98-100 

Personally, the culmination of our normal HSC studies was the use of lentivector-mediated clonal tracking101,102  to monitor greater than 600 clones in a cohort of primary and secondary mice for a total of 7 months.39  These studies touched on the longstanding debate on whether the control of cell fates is rigid and tightly organized or whether these outcomes are unpredictable and governed by stochastic processes. A single HSC can generate the entire blood system for a lifetime,40,69-72  a result that can only occur if at cell division the HSC self-renews to produce at least one daughter cell with the undifferentiated characteristics of the parental stem cell. Whether a hematopoietic clone is maintained or extinguished ultimately rests on whether an HSC makes an asymmetric (maintenance) or a symmetric self-renewing (expansion) or differentiating (extinction) division. These outcomes are known as the “birth and death” fates of stem cells.36  (see McKenzie et al39  for a more detailed discussion). We clonally analyzed hundreds of individual SRCs in primary and secondary mice; as well, we had the opportunity to analyze the repopulation properties of SRC daughter cells that became physically separated to different marrow locations within a single mouse.39  We found that the proliferation and self-renewal properties varied greatly. Even with increasing SRC purification, the variation in self-renewal capacity remained. Our results are reminiscent of the murine CFU-S data that were first reported approximately 45 years ago that formed the basis for the prediction that regulation of individual HSC outcomes is stochastic and governed by probabilistic elements.36  Our data provided experimental in vivo evidence (albeit with all the caveats of a xenotransplantation model) that is consistent with the hypothesis that human HSCs are also governed at least in part according to stochastic principles. Collectively, the identification and characterization of human HSCs set the stage for identification of their leukemic counterparts. The subsequent comparisons between normal and leukemic stem cells continue to provide clues to the cellular and molecular programs that drive their biology.

Because we were able to transplant normal human cells into mice, it was obvious that we should attempt to grow leukemic cells in mice. Our initial studies transplanting pediatric pre-B ALL into SCID mice showed the power of the system to propagate ALL with great similarity to the human disease103  and correlation to clinical outcome.104  Because we were interested in establishing whether normal HSCs could be assayed in SCID mice, this focus on thinking clonally impacted our leukemia studies. At this point, we turned our attention to AML because of the more extensive work on clonogenic assay systems and the application of stem cell concepts to understand this disease. I had been especially enamored by the clonogenic assay for AML progenitors and the studies from Buick and McCulloch, which showed that only a subset of AML-CFU had high serial replating efficiency, pointing to heterogeneity in self-renewal capacity.22,29,30  In parallel, the studies outlined in “Identification of normal HSCs in the mouse” clearly showed that normal clonogenic progenitors were distinct from repopulating HSCs, raising the question of whether the same would hold true for AML and that a more primitive LSC existed that was distinct from AML-CFU. The question was how to assay such a cell. As with any new line of investigation, collegial support was essential, especially from M. Minden and T. Hoang who provided primary AML samples and taught us how to properly thaw cells. We initiated experiments transplanting primary AML into SCID mice and obtained reliable engraftment from most subtypes of AML, except for APL. The subsequent critical experiments were performed with the prompting of my mentor R. A. Phillips who suggested we establish that the assay was quantitative by measuring the frequency of the initiating cell. This suggestion launched the application of stem cell principles to the study of the initiating cell in AML. Our first step was to establish the frequency of the initiating cell. Although we found it to vary by more than 1000-fold between donors, it was exceedingly rare: on the average of 1 per 106 cells. This was the first hint as to whether this initiating cell was actually distinct from the AML-CFU, which has a typical frequency of 1 in 100. Second, with the suggestion of my colleague N. Iscove, we carried out kinetic studies to show expansion of the AML-CFU pool during repopulation, as would be expected if they were being generated from an earlier precursor. Of course, it was possible that the initiating cell and the AML-CFU were the same cell if in vivo repopulation were simply an inefficient method to assay AML-CFU. Clearly, the only way to resolve this question was by fractionating the cells (ie, separating AML-CFU away from the initiating cells). CD34 was an obvious choice to fractionate with because expression on human HSCs had already been shown105  and adopted clinically for HSC enrichment. Prior work from Terstappen et al had pointed to CD34 and CD38 as being heterogeneously expressed in AML with the prediction that the various combinations of these 2 markers reflected a developmental hierarchy, with CD34+CD38 being the most primitive.106  The immediate obstacle was that I knew little about flow cytometry, and we did not have robust sorting capabilities in Toronto at that time. However, I had made the fortunate acquaintance of M. Caliguiri from Roswell Park who offered us access to their new highly modified beta test FACStar instrument run by C. Stewart. I have vivid memories of traveling with my postdoctoral fellow T. Lapidot (who deserves enormous 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 U.S. border agents had with his Hebrew Israeli passport. As others had seen, CD34 expression was highly variable in the AML samples we had evaluated and the one FAB-M2 sample we had used for sorting had greater than 40% CD34+ cells; however, CD38 was a smaller fraction of the CD34+ cells. AML-CFU activity was high in the CD38+ fraction compared with unsorted leukemia blasts but was also present in the CD38 fraction. Remarkably, despite the rarity of the CD34+CD38 fraction and the presence of high levels of AML-CFU in the CD34+CD38+ fraction, leukemic engraftment could only be initiated from CD34+CD38 fractions. In keeping with a tradition of naming cells based on function, we termed this the SCID leukemia-initiating cell (SL-IC). The CD34+CD38 SL-IC generated large numbers of CD34+CD38+cells, AML-CFU, and mature blasts. This was proof that the cell population that initiated AML in SCID mice was distinct from AML-CFU; we had identified a new stem cell in AML.107 

Now is probably the best time to confess that I had no idea at the time that this experiment settled an even larger issue about the stochastic/hierarchy controversy, at least for AML. Not having read carefully all the literature I have outlined in this review, I did not realize that such a controversy existed because the hierarchy model was so well entrenched in the normal and leukemic hematopoiesis field; it was simply a given. In my view, all we had provided was the long sought after formal proof for the concept of a hierarchy in AML. There was one issue that concerned us. We had only tested a single sample in this initial study and it had a primitive FAB-M1 subtype and a high CD34 content. Perhaps the principle, or at least the CD34+CD38 phenotype, would not hold for AML, which exhibited a higher degree of cellular maturation, particularly the myelomonocytic M4/M5 subtypes. By this time, the new NOD/SCID mouse strain was found to be a superior recipient for normal HSC engraftment, permitting, for the first time, quantitative assessment and purification of normal HSCs.82-85  With this more robust assay, we studied in detail another 7 samples, 6 being M4 or M5. Many of these samples exhibited a distinct pattern of engraftment different from the primitive samples. The myelomonocytic samples disseminated widely, whereas the primitive subtypes typically grew well only in the bone marrow and only disseminated after long periods of time. Interestingly, although the M4 and M5 samples disseminated widely into many organs, they never infiltrated the central nervous system. This pattern was distinct from the pre-B ALL samples that we had studied earlier, which also disseminated widely but often infiltrated the central nervous system and the testis. These disease patterns that we observed in the NOD/SCID mouse reflected the clinical features of the disease in humans, pointing to the reliability of these new xenotransplantation models (compared with the older subcutaneous implantations in nude mice) to mimic the human disease. The cell purification data from these newer experiments were clear: despite their myelomonocytic morphology and their low CD34+ and CD34+CD38 populations, the LSCs were all found in the CD34+CD38 fraction.108  Although some exceptions were reported in subsequent years, a high proportion of AML patients show LSC activity in the CD34+CD38 fraction.109-111  As well, recent studies have suggested that the ability to engraft NOD/SCID mice112  as well as increased frequencies of LSC in AML are prognostic for poor outcomes.113  Collectively, these studies provide conclusive evidence for the hierarchy model in AML.

When it came time to write the paper, D. Bonnet and I spent much effort reading the older literature, and we realized that our data could address the “cell of origin” question: what is the normal cell from which leukemia arises? Work from a number of directions, including the variation in cell surface marker expression on AML-CFU by Sabbath et al28  described earlier and the pioneering clonality studies in humans by Fialkow et al using X-linked G6PD polymorphisms114,115  followed by DNA-based restriction fragment length polymorphism analysis by Fearon et al,116  had been used to develop a theory that the maturation state of the leukemia blast reflected the level of maturation of the cell of origin.25  In vivo clonal studies in humans examined whether the normal nonleukemic lineages (lymphocyte, erythroid cell) were or were not part of the leukemic clone. Such an approach had definitively established that chronic myeloid leukemia (CML) arose in a multipotential HSC.117  If the nonleukemic cells were part of the clone, this reflected an early multipotential cell of origin, whereas if they were not part of the same clone, this reflected a more lineage-restricted cell of origin. This view was also proposed based on some of the earlier in vivo cytokinetic studies of Clarkson and others, albeit with less evidence than these clonal analyses could bring to bear. The alternative view, best articulated by McCulloch, was that observed lineage restriction was indeed only an apparent restriction and could not rule out the possibility that even the mature or lineage restricted AMLs arose from a multipotent HSC.22,29  He argued that transforming events could occur in an HSC, but one of the consequences of this event was an inability of this stem cell to generate other “normal” lineage cells, such as erythroid cells. As discussed in the following paragraph, we proposed that the similar cell surface phenotype of the LSC and the HSC supported the idea that primitive cells were the cell of origin for AML regardless of the maturation state of the leukemia blasts.108  Perhaps more than any other, this paper has caused confusion about the nomenclature of a “cancer stem cell,” especially when the CSC field exploded in 2003/2004 when solid tumor CSCs were identified.118,119  Our use of this nomenclature comes from the normal blood literature where cells are defined by function, in this case, the cancer cell that possesses stem cell properties. Our attempts to address the “cell of origin” question were taken by some to mean that a CSC was derived from a normal stem cell that had become cancerous. It finally took a blue-ribbon panel established by the American Association for Cancer Research to strongly declare that a CSC simply meant the cell in a cancer that has stem cell–like properties, especially the capacity for self-renewal.120 

Although we made the argument that the cell of origin is primitive based on the consistency of the CD34+CD38 cell surface phenotype of SL-IC among patients regardless of AML subtype and their similarity to the phenotype of normal SRC, our reasoning had serious limitations. Cell surface markers are antigens whose expression changes with differentiation, and we know differentiation is perturbed in cancer. Therefore, one must be especially cautious when using a defined set of markers that rigidly reflect the steps of lineage determination of normal cells and then inferring that the same set will apply for neoplastic cells. Ideally, one would like to make comparisons between normal HSCs and LSCs using functional criteria. The only true stem cell property is capacity for maturation combined with self-renewal. In normal hematopoiesis, cell tracking and purification studies performed clonally (in mice and humans) have documented that long-term repopulating HSCs with high self-renewal capacity generate short-term HSCs whose self-renewal capacity is more limited.101,121  Thus, we carried out lentivector-mediated clonal tracking of AML-LSC in a manner identical to that outlined in “Characterization of normal human HSCs” for normal HSCs. These studies established that individual LSCs were not homogeneous but had wide variation in repopulation potential and self-renewal capacity similar to that observed for normal HSCs122  (Figure 3). However, a key difference is that, during NOD/SCID repopulation, LSCs expand to a much greater extent than normal HSCs, probably because of increased symmetric self-renewal cell divisions.108  As more information is gathered about the molecular machinery that underlies the self-renewal process, it is becoming clearer that LSCs and HSCs share at least some characteristics. For example, Bmi1123  and PTEN124  govern self-renewal of both murine HSCs and LSCs. Together with the findings that LSC can also be quiescent like normal HSCs,122,125-127  these discoveries add additional support to the notion that some types of AML may arise from within the primitive cell compartment, before full lineage commitment has occurred. Alternatively, it is also possible that AML could arise from committed progenitors if stem cell self-renewal programs are established during the leukemogenic process. Nevertheless, this evidence is still indirect and requires a more direct experimental approach.

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 existence of distinct classes with differing self-renewal capacity, 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 recipients, whereas long-term (LT) SL-ICs can sustain leukemic 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.

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 existence of distinct classes with differing self-renewal capacity, 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 recipients, whereas long-term (LT) SL-ICs can sustain leukemic 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.

Close modal

The most direct experiment is to introduce the same genetic element into pure populations of HSCs or progenitor subsets to determine which population is competent to initiate disease. Attempts to identify the cell of origin for murine leukemia have yielded conflicting results, demonstrating that cellular and molecular context is critically important. The fusion oncogene MLL-GAS7 induces mixed-lineage leukemias when expressed in HSCs or multipotent progenitors, but not in lineage-restricted progenitors.128  In contrast, MOZ-TIF,129  MLL-AF9,130  and MLL-ENL131  lead to initiation of AML regardless of the target cell population used. However, much lower numbers of transformed HSCs compared with progenitors are often required for tumor initiation in vivo and tumors arising from progenitors are monoclonal or oligoclonal not polyclonal, suggesting that the progenitor populations were not functionally homogeneously, were not equally susceptible to transformation, or additional genetic alterations were required. Murine models of CML resulting from inactivation of JunB or expressing bcr/abl must occur in HSCs and not more restricted progenitors to induce a transplantable myeloproliferative disorder.129,132  These murine studies have become even more difficult to interpret with the recent findings of Chen et al who used a knockin model of MLL-AF9.133  This approach was taken because of concerns that, for some oncogenes, the level of expression from exogenous promoters, such as transgenes or retroviral vectors, may be nonphysiologic. They found that the same construct shown to initiate disease in HSCs and progenitors by retroviral expression only initiated leukemia from the HSC fraction when expressed from the endogenous MLL promoter. Nevertheless, collectively, 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 of an HSC or self-renewal program must be established in the non-HSC cell types.134 

No direct studies of the design used for the murine studies have been reported for human hematopoietic cells. However, the in vivo clonality studies114,115  described earlier had pointed to variation in the cell of origin. A more refined approach was to examine whether phenotypically defined populations contained specific translocations and whether these exhibited normal or leukemic proliferation. A study of patients with t(8;21) AML demonstrated that, despite the presence of leukemia-specific AML1-ETO chimeric transcripts in primitive CD34+CD90CD38 cells from leukemic bone marrow, these cells give rise to normally differentiating multilineage clonogenic progenitors, whereas more mature CD34+CD90CD38+ cells form exclusively leukemic blast colonies (AML-CFC).135  Recent studies have documented that normal HSCs are CD34+CD90+CD38, whereas the CD34+CD90CD38 fraction represents a normal multipotential progenitor with little self-renewal capacity.136  Thus, a conclusion of this work is that, whereas the initial t(8;21) translocation occurs in a primitive stem cell, subsequent events occur in the committed progenitor pool, giving rise to LSCs. However, in vivo assays are required for formal proof that these are LSCs. A similar process of leukemic progression recently has been proposed for CML.137  The discovery of a marker chromosome,138  the nature of the translocation,139  followed by lineage involvement of the Ph chromosome,117  established that CML arose from an HSC; however, analysis of blast crisis samples suggests that additional genetic events occur at the level of more committed progenitors, resulting in the generation of LSCs.137  These provocative studies point to the need to understand not only the cell of origin of the LSCs but also the steps leading to so-called “preleukemic stem cells.”

Our current understanding of the potential pathways leading to generating CSCs and a proposed model for tumor progression are depicted in Figure 4. Because the lifespan of non–stem cells may be transient, any genetic alteration in their makeup may not persist long enough to acquire additional genetic events that are needed for full transformation, unless the initial event endows the non-stem cell with self-renewal capacity. In addition, even when the stem cell sustains the first hit, subsequent events may occur in the stem cell or in the downstream progenitors. Another consideration is determining how the hierarchy model relates to leukemic progression or tumor evolution. Figure 4 proposes that tumor progression is linked to self-renewal capacity. For example, genetic instability and clonal evolution can occur in both CSCs and non-CSCs, keeping in mind that the rate of evolution could be different if CSCs proliferate more slowly than non-CSCs. Within the CSC fraction, additional genetic and epigenetic alterations could result in more abnormal CSCs; and in the case of solid tumors, these could become metastatic. The non-CSCs could also acquire genetic and epigenetic alterations; but if these do not endow self-renewal capacity, then these alterations in the non-CSCs are passengers and not drivers of tumor progression. Finally, if stem cell-like properties, including self-renewal, were endowed on non-CSCs, they could also drive tumor progression (Figure 4). A recent report has established in a breast cancer model that an epithelial-to-mesenchymal transition program is linked to stem cell function and experimental induction of epithelial-to-mesenchymal transition in a non-CSCs can reprogram them to a CSC state, providing evidence for this tumor progression model.140 

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.

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.

Close modal

A major limitation in understanding the leukemogenic process in humans is the absence of good genetic tools and experimental models that use primary human cells as their starting point. For example, the first step to directly test the cell of origin hypothesis requires an experimental model of human leukemogenesis where oncogenes are transduced into primary human hematopoietic cells. Several reports have demonstrated that the initial steps in the leukemogenic process could be induced experimentally by expression of several different oncogenes, such as Ras,141  TLS-ERG,142,143  AML-ETO,144  and bcr/abl,145  but none caused full-blown disease (see Kennedy and Barabé146  for detailed review of human leukemogenesis studies). Using MLL fusions, 2 recent studies have demonstrated successful induction of AML and pre-B ALL in NOD/SCID transplanted with transduced primary stem/progenitor cells.147,148  These mice developed B-ALL and AML that reflected the human disease. We found that the B-ALL in this model maintained a germ line configuration of the IgH locus early in disease evolution but progressed to full rearrangement on serial passage.147  These data suggest that the LSC can evolve and that the cell of origin was not a mature B cell, but a more primitive stem or progenitor cell. Direct elucidation of the cell of origin will require prospective cell sorting of pure, functionally defined HSCs, and progenitor subsets as targets for testing.

Although the NOD/SCID studies were compelling, direct evidence for LSC evolution in humans was lacking. In a seminal series of studies beginning in the 1990s, Greaves and Wiemels demonstrated that many forms of childhood leukemia arose in utero.149  They studied twins in which one twin has leukemia whereas the other does not; both were shown to contain the same chromosomal translocations (MLL, Tel-AML1), a result that could only occur if a single cell in one twin acquired the translocation, expanded, and migrated to the other twin. By examination of Ig rearrangements, they concluded that after birth additional genetic alterations occur independently in each twin, resulting in the appearance of leukemia at different times. This predicts that the twin who has not yet developed leukemia must contain a preleukemic stem cell that is poised to acquire additional alterations because this twin is at high risk of developing leukemia. In an elegant series of studies, Hong et al have identified a unique cell population, not observed in normal donors, that contains a preleukemic stem cell that exhibited enhanced self-renewal capacity when assessed in the NOD/SCID xenotransplantation system.150  This preleukemic population was clonally related to the LSC that maintained the leukemia in the affected twin. Thus, this study provides strong proof in humans for the concept of clonal evolution of LSC from a preleukemic to a leukemic state.

Recently, several reports were published that questioned the universality of the CSC or hierarchy model.151,152  Several experimentally induced murine leukemia models were found where almost every cancer cell had LSC activity and there was little evidence of functional heterogeneity. These and other reports153  speculated that the rarity of human CSCs observed using xenotransplantation models may be the result of host resistance factors as well as the absence of cross-species reactivity of cytokines and other microenvironmental factors. Certainly, such factors play a role because the use of more immune-deficient mice has improved human CSC detection; in addition, some solid tumor studies are now using coimplantation of human stromal cells. It is important to note that a number of other genetically induced murine leukemia models are heterogeneous and contain discrete and rare LSCs.154  Moreover, when human and murine leukemia models were both induced with the same MLL fusion protein, the LSC frequency was the same.154  Thus, it is evident that LSC frequencies can vary 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 the CSC model is not directly related to the absolute frequency of these cells but rather on functional evidence of tumor heterogeneity and a hierarchical organization. The CSC model is only invoked to explain tumor heterogeneity; however, if a tumor is functionally homogeneous, the CSC model is no longer relevant. It is clear that some murine cancer models do not follow the hierarchy model, and it will be important to survey a much broader cross section of human tumors to determine whether any also lack tumor heterogeneity.

Despite the recent explosion of interest in CSCs, experimental studies have not been translated into improved survival outcomes for cancer patients. This presents a major question to the field: do cancer stem cells have meaningful relevance beyond the experimental systems in which they have been defined? Increasing evidence is pointing to CSCs having unique biologic properties (dormancy, drug/radiation resistance) that could permit them to survive therapies leading to eventual relapse. To date, CSCs have been studied in a relatively small number of patient samples and cancer types, so it is not known whether the CSC model is universal to all human cancers.1  The biologic relevance of CSCs in human cancer will be established by concentrating on the following research endeavors: improving the assay and purification of CSC and non-CSC subsets, carrying out detailed genomic or proteomic analysis on these subsets to identify CSC-specific signatures, and obtaining such signatures from a large number and wide range of tumors. Such an approach would make it possible to determine whether the “omics” of CSCs provide more predictive or prognostic relevance compared with analysis of the bulk tumor. An approach like this has recently been taken by Liu et al who showed that a breast CSC signature has improved correlation to patient outcomes.155  Of course, the most direct proof for the relevance of CSCs to cancer would be to use the CSC-specific signatures to elucidate the key molecular pathways involved, develop the means to target them effectively, and establish in clinical trials whether targeting them enhances patient survival.

For many types of leukemia, the long-term survival has increased dramatically over the past 50 years; however, AML remains one of the most difficult diseases to cure. The majority of patients will die because of relapse, drug resistance, or from complications associated with chemotherapies. Because LSCs and HSCs have many similar properties, a first step to develop novel therapies has been to identify unique properties that LSCs possess. Through expression analysis, Guzman et al determined that the NF-κB pathway was active in LSCs and not in HSCs.126  They then went on to identify a natural product, parthenolide, which targets this pathway and showed that the LSCs could be specifically targeted using the NOD/SCID model.156,157  It should be possible to develop a strategy that focuses on the biology of LSC and develop assay methods that are compatible with high throughput screening of small molecule libraries followed by counterscreens with normal HSCs or progenitors.158  Other approaches focus on identification of differences in cell surface markers with concomitant development of immunoconjugates with toxic moieties. LSCs could be targeted with a diphtheria toxin conjugated to granulocyte colony-stimulating factor immunotoxin.159,160  However, not all AML samples responded to these molecules, and regrowth or unsuccessful eradication of the leukemic cells occurred after different treatment strategies, suggesting that not all classes of SL-IC were killed by diphtheria toxin conjugated to granulocyte colony-stimulating factor immunotoxin. Similarly a DT-IL3 conjugate was shown to target the LSC and progressed to clinical trials. Unfortunately, this therapy resulted in the development of antibodies as well as other toxicities.161  As well, LSCs could be targeted with a CD8+ cytotoxic T lymphocyte clone specific for minor histocompatibility antigens,162,163  an approach that might be useful in the 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 these mAbs were injected into NOD/SCID mice repopulated with AML, the LSCs were markedly depleted; and in some cases, the mice were cured. Induction of differentiation represented a partial mechanism of action. Even more noteworthy, both CXCR4 and CD44 resulted in impaired trafficking of the LSC, suggesting that LSCs remain dependent on niche interactions. Because many other factors are involved in trafficking and homing (see 50th Anniversary Review by Papayannopoulou and Scadden166 ), this finding opens the way for testing the anti-LSC functionality of any molecule that interferes with these processes. Little is known of the LSC niche; however, a recent study points to the endosteal region,167  an area that has been proposed as the niche for murine HSCs as well. Another LSC-specific target, which is either low or absent from normal HSCs, is CD123, the alpha chain of the IL3R.168  A mAb targeting CD123 impairing homing of LSC to bone marrow of NOD/SCID mice, and it activates the innate immunity of NOD/SCID mice resulting in eradication of LSCs.169  Collectively, these studies provide clear validation for therapeutic mAb targeting of AML-LSCs and for translation of the NOD/SCID in vivo preclinical research findings toward a clinical application. Finally, future therapeutic approaches will need to be developed that target the self-renewal machinery of LSCs, although it may be difficult to find a therapeutic window that will spare normal tissue stem cells. As predicted from the cytokinetic studies, perhaps the most attractive option is to induce the quiescent LSCs into cycle. Recent studies are pointing to specific molecular pathways that might be attractive, including PTEN, p21, and PML. Ito et al have found that degradation of PML induces cycling of LSC and, interestingly, that one of the mechanisms of action of arsenic is degradation of PML, already putting a potential drug strategy in hand.170 

Although this review is focused on the premise that improvements in the stubbornly high relapse rates for many cancers will come from more research on the CSCs that drive the tumors, attention is needed on how to move CSC therapies into clinical trials and into the clinic (a more thorough discussion is done by Wang171 ). The current approaches to preclinical drug development and clinical evaluation of drug efficacy focus on initial requirements for tumor response. Therefore, a potentially effective anti-CSC therapy might be discarded from further testing if it does not cause a response of the non-CSCs cells rapidly enough. Similarly, an effective anti-CSC might take time to show efficacy or be most effective in an adjuvant setting. Much research effort will be required to find the best algorithms for clinical evaluation of CSC-targeted therapies.

In conclusion, stem cell concepts lie at the heart of the first cancer therapies developed almost 40 years ago, which showed the first tangible success in curing patients with acute leukemia. One hopes that the renewal of interest in the linkage between stem cells and cancer will lead to a better understanding of how the cellular context of tumors affects cancer-specific genetic and epigenetic pathways and will once again drive new therapeutic advances.

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 creating the figures; and Melissa Cooper for insightful editing.

This work was supported by grants from the Canadian Institutes of Health Research (CIHR), the Ontario Institute for Cancer Research and a Summit Award both with funds from the Province of Ontario, Genome Canada through the Ontario Genomics Institute, the Leukemia & Lymphoma Society, and the National Cancer Institute of Canada with funds from the Canadian Cancer Society and the Terry Fox Foundation, research contracts with CSL and Arius, and a Canada Research Chair.

Contribution: J.E.D. wrote the paper.

Conflict-of-interest disclosure: The author declares research contracts with CSL and Arius.

Correspondence: John E. Dick, Toronto Medical Discovery Tower, 101 College Street, Room 8-301, Toronto, ON, Canada M5G 1L7; e-mail: jdick@uhnres.utoronto.ca.

1
Wang
 
JC
Dick
 
JE
Cancer stem cells: lessons from leukemia.
Trends Cell Biol
2005
, vol. 
15
 (pg. 
494
-
501
)
2
Dalerba
 
P
Cho
 
RW
Clarke
 
MF
Cancer stem cells: models and concepts.
Annu Rev Med
2007
, vol. 
58
 (pg. 
267
-
284
)
3
Ailles
 
LE
Weissman
 
IL
Cancer stem cells in solid tumors.
Curr Opin Biotechnol
2007
, vol. 
18
 (pg. 
460
-
466
)
4
Furth
 
J
Kahn
 
M
The transmission of leukemia of mice with a single cell.
Am J Cancer
1937
, vol. 
31
 (pg. 
276
-
282
)
5
Makino
 
S
Further evidence favoring the concept of the stem cell in ascites tumors of rats.
Ann N Y Acad Sci
1956
, vol. 
63
 (pg. 
818
-
830
)
6
Hewitt
 
HB
Studies of the dissemination and quantitative transplantation of a lymphocytic leukaemia of CBA m ice.
Br J Cancer
1958
, vol. 
12
 (pg. 
378
-
401
)
7
Bruce
 
WR
Van Der Gaag
 
H
A quantitative assay for the number of murine lymphoma cells capable of proliferation in vivo.
Nature
1963
, vol. 
199
 (pg. 
79
-
80
)
8
Southam
 
C
Brunschwig
 
A
Dizon
 
Q
Brennan
 
MJ
Simpson
 
WL
Autologous and homologous transplantation of human cancer.
Biological Interactions in Normal and Neoplastic Growth: A Contribution to the Tumor-Host Problem
1962
Boston, MA
Little, Brown
(pg. 
723
-
738
)
9
Pierce
 
GB
Dixon
 
FJ
Verney
 
EL
Teratocarcinogenic and tissue-forming potentials of the cell types comprising neoplastic embryoid bodies.
Lab Invest
1960
, vol. 
9
 (pg. 
583
-
602
)
10
Pierce
 
GB
Speers
 
WC
Tumors as caricatures of the process of tissue renewal: prospects for therapy by directing differentiation.
Cancer Res
1988
, vol. 
48
 (pg. 
1996
-
2004
)
11
Belanger
 
L
Leblond
 
C
A method for locating radioactive elements in tissues by covering histological sections with a photographic emulsion.
Endocrinology
1946
, vol. 
39
 (pg. 
386
-
400
)
12
Clermont
 
Y
Leblond
 
C
Renewal of spermatagonia in the rat testis.
Am J Anat
1953
, vol. 
93
 (pg. 
475
-
502
)
13
Clarkson
 
B
Fried
 
J
Strife
 
A
Sakai
 
Y
Ota
 
K
Okita
 
T
Studies of cellular proliferation in human leukemia: III. Behavior of leukemic cells in three adults with acute leukemia given continuous infusions of 3H-thymidine for 8 or 10 days.
Cancer
1970
, vol. 
25
 (pg. 
1237
-
1260
)
14
Clarkson
 
BD
Fried
 
J
Changing concepts of treatment in acute leukemia.
Med Clin North Am
1971
, vol. 
55
 (pg. 
561
-
600
)
15
Killmann
 
SA
Cronkite
 
EP
Robertson
 
JS
Fliedner
 
TM
Bond
 
VP
Estimation of phases of the life cycle of leukemic cells from labeling in human beings in vivo with tritiated thymidine.
Lab Invest
1963
, vol. 
12
 (pg. 
671
-
684
)
16
Gavosto
 
F
The proliferative kinetics of the acute leukaemias in relation to their treatment.
Rev Eur Etud Clin Biol
1970
, vol. 
15
 (pg. 
1042
-
1047
)
17
Clarkson
 
BD
Review of recent studies of cellular proliferation in acute leukemia.
Natl Cancer Inst Monogr
1969
, vol. 
30
 (pg. 
81
-
120
)
18
Clarkson
 
B
Clarkson
 
B
Baserga
 
R
The survival value of the dormant state in neoplastic and normal populations.
Control of Proliferation in Animal Cells
1974
New York, NY
Cold Spring Harbor Laboratory
(pg. 
945
-
972
)
19
Cronkite
 
EP
Acute leukemia: is there a relationship between cell growth kinetics and response to chemotherapy?
Proc Natl Cancer Conf
1970
, vol. 
6
 (pg. 
113
-
117
)
20
Clarkson
 
BD
Dowling
 
MD
Gee
 
TS
Cunningham
 
IB
Burchenal
 
JH
Treatment of acute leukemia in adults.
Cancer
1975
, vol. 
36
 (pg. 
775
-
795
)
21
Buick
 
RN
Till
 
JE
McCulloch
 
EA
Colony assay for proliferative blast cells circulating in myeloblastic leukaemia.
Lancet
1977
, vol. 
1
 (pg. 
862
-
863
)
22
McCulloch
 
E
Stem cells in normal and leukemic hemopoiesis (Henry Stratton Lecture).
Blood
1983
, vol. 
62
 (pg. 
1
-
13
)
23
Metcalf
 
D
Moore
 
MA
Warner
 
NL
Colony formation in vitro myelomonocytic leukemic cells.
J Natl Cancer Inst
1969
, vol. 
43
 (pg. 
983
-
1001
)
24
Moore
 
MA
Williams
 
N
Metcalf
 
D
In vitro colony formation by normal and leukemic human hematopoietic cells: characterization of the colony-forming cells.
J Natl Cancer Inst
1973
, vol. 
50
 (pg. 
603
-
623
)
25
Griffin
 
J
Löwenberg
 
B
Clonogenic cells in acute myeloblastic leukemia.
Blood
1986
, vol. 
68
 (pg. 
1185
-
1195
)
26
Dicke
 
KA
Spitzer
 
G
Ahearn
 
MJ
Colony formation in vitro by leukaemic cells in acute myelogenous leukaemia with phytohaemagglutinin as stimulating factor.
Nature
1976
, vol. 
259
 (pg. 
129
-
130
)
27
Hamburger
 
AW
Salmon
 
SE
Primary bioassay of human tumor stem cells.
Science
1977
, vol. 
197
 (pg. 
461
-
463
)
28
Sabbath
 
KD
Ball
 
ED
Larcom
 
P
Davis
 
RB
Griffin
 
JD
Heterogeneity of clonogenic cells in acute myeloblastic leukemia.
J Clin Invest
1985
, vol. 
75
 (pg. 
746
-
753
)
29
McCulloch
 
E
Till
 
J
Blast cells in acute myeloblastic leukemia: a model.
Blood Cells
1981
, vol. 
7
 (pg. 
63
-
77
)
30
Buick
 
RN
Minden
 
MD
McCulloch
 
EA
Self-renewal in culture of proliferative blast progenitor cells in acute myeloblastic leukemia.
Blood
1979
, vol. 
54
 (pg. 
95
-
104
)
31
Maximov
 
A
Der lymphozyt als gemeinsame Stammzelle der verschieden Blutelemente in der embryonalen Entwicklung und postfetalen Leben der Sugetiere.
Folia Haematologica (Leipzig)
1909
, vol. 
8
 (pg. 
125
-
141
)
32
Lichtman
 
MA
The stem cell in the pathogenesis and treatment of myelogenous leukemia: a perspective.
Leukemia
2001
, vol. 
15
 (pg. 
1489
-
1494
)
33
Till
 
JE
McCulloch
 
EA
A direct measurement of the radiation sensitivity of normal mouse bone marrow cells.
Radiat Res
1961
, vol. 
14
 (pg. 
213
-
222
)
34
Becker
 
AJ
McCulloch
 
EA
Till
 
JE
Cytological demonstration of the clonal nature of spleen colonies derived from transplanted mouse marrow cells.
Nature
1963
, vol. 
197
 (pg. 
452
-
454
)
35
Siminovitch
 
L
McCulloch
 
EA
Till
 
JE
The distribution of colony-forming cells among spleen colonies.
J Cell Comp Physiol
1963
, vol. 
62
 (pg. 
327
-
336
)
36
Till
 
JE
McCulloch
 
EA
Siminovitch
 
L
A stochastic model of stem cell proliferation based on the growth of spleen colony-forming cells.
Proc Natl Acad Sci U S A
1964
, vol. 
51
 (pg. 
29
-
36
)
37
Lemischka
 
IR
The haematopoietic stem cell and its clonal progeny: mechanisms regulating the hierarchy of primitive haematopoietic cells.
Cancer Surv
1992
, vol. 
15
 (pg. 
3
-
18
)
38
Morrison
 
S
Uchida
 
N
Weissman
 
I
The biology of hematopoietic stem cells.
Annu Rev Cell Dev Biol
1995
, vol. 
11
 (pg. 
35
-
71
)
39
McKenzie
 
JL
Gan
 
OI
Doedens
 
M
Wang
 
JC
Dick
 
JE
Individual stem cells with highly variable proliferation and self-renewal properties comprise the human hematopoietic stem cell compartment.
Nat Immunol
2006
, vol. 
7
 (pg. 
1225
-
1233
)
40
Sieburg
 
HB
Cho
 
RH
Dykstra
 
B
Uchida
 
N
Eaves
 
CJ
Muller-Sieburg
 
CE
The hematopoietic stem compartment consists of a limited number of discrete stem cell subsets.
Blood
2006
, vol. 
107
 (pg. 
2311
-
2316
)
41
Huang
 
S
Guo
 
YP
May
 
G
Enver
 
T
Bifurcation dynamics in lineage-commitment in bipotent progenitor cells.
Dev Biol
2007
, vol. 
305
 (pg. 
695
-
713
)
42
Pluznik
 
DH
Sachs
 
L
The cloning of normal “mast” cells in tissue culture.
J Cell Physiol
1965
, vol. 
66
 (pg. 
319
-
324
)
43
Bradley
 
TR
Metcalf
 
D
The growth of mouse bone marrow cells in vitro.
Aust J Exp Biol Med Sci
1966
, vol. 
44
 (pg. 
287
-
299
)
44
Metcalf
 
D
Hematopoietic cytokines.
Blood
2008
, vol. 
111
 (pg. 
485
-
491
)
45
Magli
 
MC
Iscove
 
NN
Odartchenko
 
N
Transient nature of early hematopoietic spleen colonies.
Nature
1982
, vol. 
295
 (pg. 
527
-
529
)
46
Boggs
 
DR
Boggs
 
SS
Saxe
 
DF
Gress
 
LA
Canfield
 
DR
Hemopoietic stem cells with high proliferative potential: assay of their concentration in marrow by the frequency and duration of cure of W/Wv mice.
J Clin Invest
1982
, vol. 
70
 (pg. 
242
-
253
)
47
Paige
 
CJ
Kincade
 
PW
Moore
 
MAS
Lee
 
G
The fate of fetal and adult B-cell progenitors grafted into immunodeficient CBA/N mice.
J Exp Med
1979
, vol. 
150
 (pg. 
548
-
563
)
48
Paige
 
CJ
Kincade
 
PW
Shinefeld
 
LA
Sato
 
VL
Precursors of murine B lymphocytes: physical and functional characterization, and distinctions from myeloid stem cells.
J Exp Med
1981
, vol. 
153
 (pg. 
154
-
165
)
49
Hodgson
 
GS
Bradley
 
TR
Properties of hematopoietic stem cells surviving 5-fluorouracil treatment: evidence for a pre-CFU-S cell?
Nature
1979
, vol. 
281
 (pg. 
381
-
382
)
50
Ploemacher
 
RE
Brons
 
RH
Separation of CFU-S from primitive cells responsible for reconstitution of the bone marrow hemopoietic stem cell compartment following irradiation: evidence for a pre-CFU-S cell.
Exp Hematol
1989
, vol. 
17
 (pg. 
263
-
266
)
51
Harrison
 
DE
Competitive repopulation: a new assay for long-term stem cell functional capacity.
Blood
1980
, vol. 
55
 (pg. 
77
-
81
)
52
Ford
 
C
Hamerton
 
J
Barns
 
W
Loutet
 
J
Cytological identification of radiation chimeras.
Nature
1956
, vol. 
177
 (pg. 
452
-
454
)
53
Wu
 
AM
Till
 
JE
Siminovitch
 
L
McCulloch
 
EA
Cytological evidence for a relationship between normal hemopoietic colony-forming cells and cells of the lymphoid system.
J Exp Med
1968
, vol. 
127
 (pg. 
455
-
463
)
54
Abramson
 
S
Miller
 
RG
Phillips
 
RA
The identification in adult bone marrow of pluripotent and restricted stem cells of the myeloid and lymphoid systems.
J Exp Med
1977
, vol. 
145
 (pg. 
1567
-
1579
)
55
Dick
 
JE
Magli
 
MC
Huszar
 
D
Phillips
 
RA
Bernstein
 
A
Introduction of a selectable gene into primitive stem cells capable of long-term reconstitution of the hemopoietic system of W/Wv mice.
Cell
1985
, vol. 
42
 (pg. 
71
-
79
)
56
Keller
 
G
Paige
 
C
Gilboa
 
E
Wagner
 
E
Expression of a foreign gene in myeloid and lymphoid cells derived from multipotent hemopoietic precursors.
Nature
1985
, vol. 
318
 (pg. 
149
-
154
)
57
Lemischka
 
IR
Raulet
 
DH
Mulligan
 
RC
Developmental potential and dynamic behavior of hematopoietic stem cells.
Cell
1986
, vol. 
45
 (pg. 
917
-
927
)
58
Lorenz
 
E
Uphoff
 
D
Reid
 
TR
Shelton
 
E
Modification of irradiation injury in mice and guinea pigs by bone marrow injections.
J Natl Cancer Inst
1951
, vol. 
12
 (pg. 
197
-
201
)
59
Thomas
 
ED
Lochte
 
HL
Lu
 
WC
Ferrebee
 
JW
Intravenous infusion of bone marrow in patients receiving radiation and chemotherapy.
N Engl J Med
1957
, vol. 
257
 (pg. 
491
-
496
)
60
Gatti
 
RA
Meuwissen
 
HJ
Allen
 
HD
Hong
 
R
Good
 
RA
Immunological reconstitution of sex-linked lymphopenic immunological deficiency.
Lancet
1968
, vol. 
2
 (pg. 
1366
-
1369
)
61
Buckner
 
CD
Epstein
 
RB
Rudolph
 
RH
Clift
 
RA
Storb
 
R
Thomas
 
ED
Allogeneic marrow engraftment following whole body irradiation in a patient with leukemia.
Blood
1970
, vol. 
35
 (pg. 
741
-
750
)
62
Evans
 
MJ
Kaufman
 
MH
Establishment in culture of pluripotential cells from mouse embryos.
Nature
1981
, vol. 
292
 (pg. 
154
-
156
)
63
Martin
 
GR
Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells.
Proc Natl Acad Sci U S A
1981
, vol. 
78
 (pg. 
7634
-
7638
)
64
Miller
 
RG
Phillips
 
RA
Separation of cells by velocity sedimentation.
J Cell Physiol
1969
, vol. 
73
 (pg. 
191
-
201
)
65
Bonner
 
WA
Hulett
 
HR
Sweet
 
RG
Herzenberg
 
LA
Fluorescence activated cell sorting.
Rev Sci Instrum
1972
, vol. 
43
 (pg. 
404
-
409
)
66
Visser
 
JW
Bol
 
SJ
van den Engh
 
G
Characterization and enrichment of murine hemopoietic stem cells by fluorescence activated cell sorting.
Exp Hematol
1981
, vol. 
9
 (pg. 
644
-
655
)
67
Baines
 
P
Visser
 
JW
Analysis and separation of murine bone marrow stem cells by H33342 fluorescence-activated cell sorting.
Exp Hematol
1983
, vol. 
11
 (pg. 
701
-
708
)
68
Spangrude
 
GJ
Heimfeld
 
S
Weissman
 
IL
Purification and characterization of mouse hematopoietic stem cells.
Science
1988
, vol. 
241
 (pg. 
58
-
62
)
69
Osawa
 
M
Hanada
 
K
Hamada
 
H
Nakauchi
 
H
Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell.
Science
1996
, vol. 
273
 (pg. 
242
-
245
)
70
Kiel
 
MJ
Yilmaz
 
OH
Iwashita
 
T
Yilmaz
 
OH
Terhorst
 
C
Morrison
 
SJ
SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells.
Cell
2005
, vol. 
121
 (pg. 
1109
-
1121
)
71
Benveniste
 
P
Cantin
 
C
Hyam
 
D
Iscove
 
NN
Hematopoietic stem cells engraft in mice with absolute efficiency.
Nat Immunol
2003
, vol. 
4
 (pg. 
708
-
713
)
72
Kent
 
DG
Dykstra
 
BJ
Cheyne
 
J
Ma
 
E
Eaves
 
CJ
Steel factor coordinately regulates the molecular signature and biologic function of hematopoietic stem cells.
Blood
2008
, vol. 
112
 (pg. 
560
-
567
)
73
Dick
 
JE
Normal and leukemic human stem cells assayed in SCID mice.
Semin Immunol
1996
, vol. 
8
 (pg. 
197
-
206
)
74
Mosier
 
DE
Gulizia
 
RJ
Baird
 
SM
Wilson
 
DB
Transfer of a functional human immune system to mice with severe combined immunodeficiency.
Nature
1988
, vol. 
335
 (pg. 
256
-
259
)
75
McCune
 
JM
Namikawa
 
R
Kaneshima
 
H
Shultz
 
LD
Lieberman
 
M
Weissman
 
IL
The SCID-hu mouse: murine model for the analysis of human hematolymphoid differentiation and function.
Science
1988
, vol. 
241
 (pg. 
1632
-
1639
)
76
Fraser
 
CC
Kaneshima
 
H
Hansteen
 
G
Kilpatrick
 
M
Hoffman
 
R
Chen
 
BP
Human allogeneic stem cell maintenance and differentiation in a long-term multilineage SCID-hu graft.
Blood
1995
, vol. 
86
 (pg. 
1680
-
1693
)
77
Baum
 
CM
Weissman
 
IL
Tsukamoto
 
AS
Buckle
 
A-S
Peault
 
B
Isolation of a candidate human hematopoietic stem-cell population.
Proc Natl Acad Sci U S A
1992
, vol. 
89
 (pg. 
2804
-
2808
)
78
Kamel-Reid
 
S
Dick
 
JE
Engraftment of immune-deficient mice with human hematopoietic stem cells.
Science
1988
, vol. 
242
 (pg. 
1706
-
1709
)
79
Lapidot
 
T
Pflumio
 
F
Doedens
 
M
Murdoch
 
B
Williams
 
DE
Dick
 
JE
Cytokine stimulation of multilineage hematopoiesis from immature human cells engrafted in scid mice.
Science
1992
, vol. 
255
 (pg. 
1137
-
1141
)
80
Vormoor
 
J
Lapidot
 
T
Pflumio
 
F
, et al. 
Immature human cord blood progenitors engraft and proliferate to high levels in severe combined immunodeficient mice.
Blood
1994
, vol. 
83
 (pg. 
2489
-
2497
)
81
Shultz
 
L
Schweitzer
 
P
Christianson
 
S
, et al. 
Multiple defects in innate and adaptive immunological function in NOD/LtSz-scid mice.
J Immunol
1995
, vol. 
154
 (pg. 
180
-
191
)
82
Larochelle
 
A
Vormoor
 
J
Hanenberg
 
H
, et al. 
Identification of primitive human hematopoietic cells capable of repopulating NOD/SCID mouse bone marrow: implications for gene therapy.
Nat Med
1996
, vol. 
2
 (pg. 
1329
-
1337
)
83
Cashman
 
JD
Lapidot
 
T
Wang
 
JC
, et al. 
Kinetic evidence of the regeneration of multilineage hematopoiesis from primitive cells in normal human bone marrow transplanted into immunodeficient mice.
Blood
1997
, vol. 
89
 (pg. 
4307
-
4316
)
84
Wang
 
JC
Doedens
 
M
Dick
 
JE
Primitive human hematopoietic cells are enriched in cord blood compared with adult bone marrow or mobilized peripheral blood as measured by the quantitative in vivo SCID-repopulating cell assay.
Blood
1997
, vol. 
89
 (pg. 
3919
-
3924
)
85
Conneally
 
E
Cashman
 
J
Petzer
 
A
Eaves
 
C
Expansion in vitro of transplantable human cord blood stem cells demonstrated using a quantitative assay of their lympho-myeloid repopulating activity in nonobese diabetic-scid/scid mice.
Proc Natl Acad Sci U S A
1997
, vol. 
94
 (pg. 
9836
-
9841
)
86
Ito
 
M
Hiramatsu
 
H
Kobayashi
 
K
, et al. 
NOD/SCID/gamma(c)(null) mouse: an excellent recipient mouse model for engraftment of human cells.
Blood
2002
, vol. 
100
 (pg. 
3175
-
3182
)
87
Ishikawa
 
F
Yasukawa
 
M
Lyons
 
B
, et al. 
Development of functional human blood and immune systems in NOD/SCID/IL2 receptor γ chain (null) mice.
Blood
2005
, vol. 
106
 (pg. 
1565
-
1573
)
88
Hogan
 
CJ
Shpall
 
EJ
Keller
 
G
Differential long-term and multilineage engraftment potential from subfractions of human CD34+ cord blood cells transplanted into NOD/SCID mice.
Proc Natl Acad Sci U S A
2002
, vol. 
99
 (pg. 
413
-
418
)
89
Kerre
 
TC
De Smet
 
G
De Smedt
 
M
, et al. 
Adapted NOD/SCID model supports development of phenotypically and functionally mature T cells from human umbilical cord blood CD34(+) cells.
Blood
2002
, vol. 
99
 (pg. 
1620
-
1626
)
90
McKenzie
 
JL
Gan
 
OI
Doedens
 
M
Dick
 
JE
Human short-term repopulating stem cells are efficiently detected following intrafemoral transplantation into NOD/SCID recipients depleted of CD122+ cells.
Blood
2005
, vol. 
106
 (pg. 
1259
-
1261
)
91
Takenaka
 
K
Prasolava
 
TK
Wang
 
JC
, et al. 
Polymorphism in Sirpa modulates engraftment of human hematopoietic stem cells.
Nat Immunol
2007
, vol. 
8
 (pg. 
1313
-
1323
)
92
Mazurier
 
F
Doedens
 
M
Gan
 
OI
Dick
 
JE
Rapid myeloerythroid repopulation after intrafemoral transplantation of NOD-SCID mice reveals a new class of human stem cells.
Nat Med
2003
, vol. 
9
 (pg. 
959
-
963
)
93
Yahata
 
T
Ando
 
K
Sato
 
T
, et al. 
A highly sensitive strategy for SCID-repopulating cell assay by direct injection of primitive human hematopoietic cells into NOD/SCID mice bone marrow.
Blood
2003
, vol. 
101
 (pg. 
2905
-
2913
)
94
Wang
 
J
Kimura
 
T
Asada
 
R
, et al. 
SCID-repopulating cell activity of human cord blood-derived CD34-negative cells assured by intra-bone marrow injection.
Blood
2003
, vol. 
101
 (pg. 
2924
-
2931
)
95
Wang
 
L
Menendez
 
P
Shojaei
 
F
, et al. 
Generation of hematopoietic repopulating cells from human embryonic stem cells independent of ectopic HOXB4 expression.
J Exp Med
2005
, vol. 
201
 (pg. 
1603
-
1614
)
96
Dick
 
JE
Lapidot
 
T
Biology of normal and acute myeloid leukemia stem cells.
Int J Hematol
2005
, vol. 
82
 (pg. 
389
-
396
)
97
Wang
 
JC
Dorrell
 
C
Ito
 
CY
, et al. 
Zon
 
LI
Normal and leukemic human stem cells assayed in immune-deficient mice.
Hematopoiesis: A Developmental Approach
2001
New York, NY
Oxford University Press
(pg. 
99
-
118
)
98
Pflumio
 
F
Izac
 
B
Katz
 
A
Shultz
 
LD
Vainchenker
 
W
Coulombel
 
L
Phenotype and function of human hematopoietic cells engrafting immune-deficient CB17-severe combined immunodeficiency mice and nonobese diabetic-severe combined immunodeficiency mice after transplantation of human cord blood mononuclear cells.
Blood
1996
, vol. 
88
 (pg. 
3731
-
3740
)
99
Glimm
 
H
Eisterer
 
W
Lee
 
K
, et al. 
Previously undetected human hematopoietic cell populations with short-term repopulating activity selectively engraft NOD/SCID-beta2 microglobulin-null mice.
J Clin Invest
2001
, vol. 
107
 (pg. 
199
-
206
)
100
Kerre
 
TC
De Smet
 
G
De Smedt
 
M
, et al. 
Both CD34+38+ and CD34+38- cells home specifically to the bone marrow of NOD/LtSZ scid/scid mice but show different kinetics in expansion.
J Immunol
2001
, vol. 
167
 (pg. 
3692
-
3698
)
101
Guenechea
 
G
Gan
 
OI
Dorrell
 
C
Dick
 
JE
Distinct classes of human stem cells that differ in proliferative and self-renewal potential.
Nat Immunol
2001
, vol. 
2
 (pg. 
75
-
82
)
102
Mazurier
 
F
Gan
 
O
McKenzie
 
J
Doedens
 
M
Dick
 
J
Lentivector-mediated clonal tracking reveals intrinsic heterogeneity in the human hematopoietic stem cell compartment and culture-induced stem cell impairment.
Blood
2004
, vol. 
103
 (pg. 
545
-
552
)
103
Kamel-Reid
 
S
Letarte
 
M
Sirard
 
C
, et al. 
A model of human acute lymphoblastic leukemia in immune-deficient SCID mice.
Science
1989
, vol. 
246
 (pg. 
1597
-
1600
)
104
Uckun
 
FM
Sather
 
H
Reaman
 
G
, et al. 
Leukemic cell growth in SCID mice as a predictor of relapse in high-risk B-lineage acute lymphoblastic leukemia.
Blood
1995
, vol. 
85
 (pg. 
873
-
878
)
105
Civin
 
C
Strauss
 
L
Brovall
 
C
Fackler
 
M
Schwartz
 
J
Shaper
 
J
Antigenic analysis of hematopoiesis: III. A hematopoietic progenitor cell surface antigen defined by a monoclonal antibody raised against KG-1a cells.
J Immunol
1984
, vol. 
133
 (pg. 
157
-
165
)
106
Terstappen
 
LW
Safford
 
M
Unterhalt
 
M
, et al. 
Flow cytometric characterization of acute myeloid leukemia: IV. Comparison to the differentiation pathway of normal hematopoietic progenitor cells.
Leukemia
1992
, vol. 
6
 (pg. 
993
-
1000
)
107
Lapidot
 
T
Sirard
 
C
Vormoor
 
J
, et al. 
A cell initiating human acute myeloid leukaemia after transplantation into SCID mice.
Nature
1994
, vol. 
367
 (pg. 
645
-
648
)
108
Bonnet
 
D
Dick
 
JE
Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell.
Nat Med
1997
, vol. 
3
 (pg. 
730
-
737
)
109
Ailles
 
LE
Gerhard
 
B
Kawagoe
 
H
Hogge
 
DE
Growth characteristics of acute myelogenous leukemia progenitors that initiate malignant hematopoiesis in nonobese diabetic/severe combined immunodeficient mice.
Blood
1999
, vol. 
94
 (pg. 
1761
-
1772
)
110
Terpstra
 
W
Prins
 
A
Visser
 
T
, et al. 
Conditions for engraftment of human acute myeloid leukemia (AML) in SCID mice.
Leukemia
1995
, vol. 
9
 (pg. 
1573
-
1577
)
111
Terpstra
 
W
Prins
 
A
Ploemacher
 
RE
, et al. 
Long-term leukemia-initiating capacity of a CD34-subpopulation of acute myeloid leukemia.
Blood
1996
, vol. 
87
 (pg. 
2187
-
2194
)
112
Pearce
 
DJ
Taussig
 
D
Zibara
 
K
, et al. 
AML engraftment in the NOD/SCID assay reflects the outcome of AML: implications for our understanding of the heterogeneity of AML.
Blood
2006
, vol. 
107
 (pg. 
1166
-
1173
)
113
van Rhenen
 
A
Feller
 
N
Kelder
 
A
, et al. 
High stem cell frequency in acute myeloid leukemia at diagnosis predicts high minimal residual disease and poor survival.
Clin Cancer Res
2005
, vol. 
11
 (pg. 
6520
-
6527
)
114
Fialkow
 
PJ
Singer
 
JW
Adamson
 
JW
, et al. 
Acute nonlymphocytic leukemia: heterogeneity of stem cell origin.
Blood
1981
, vol. 
57
 (pg. 
1068
-
1073
)
115
Fialkow
 
P
Singer
 
J
Raskind
 
W
, et al. 
Clonal development, stem-cell differentiation, and clinical remissions in acute nonlymphocytic leukemia.
N Engl J Med
1987
, vol. 
317
 (pg. 
468
-
473
)
116
Fearon
 
E
Burke
 
P
Schiffer
 
C
Zehnbauer
 
B
Vogelstein
 
B
Differentiation of leukemia cells to polymorphonuclear leukocytes in patients with acute nonlymphoblastic leukemia.
N Engl J Med
1986
, vol. 
315
 (pg. 
15
-
24
)
117
Fialkow
 
P
Denman
 
A
Jacobson
 
R
Lowenthal
 
M
Chronic myelogenous leukemia: origin of some lymphocytes from leukemic stem cells.
J Clin Invest
1978
, vol. 
62
 (pg. 
815
-
823
)
118
Al-Hajj
 
M
Wicha
 
M
Morrison
 
SJ
Clarke
 
MF
Prospective identification of tumorigenic breast cancer cells.
Proc Natl Acad Sci U S A
2003
, vol. 
100
 (pg. 
3983
-
3988
)
119
Singh
 
SK
Hawkins
 
C
Clarke
 
ID
, et al. 
Identification of human brain tumour initiating cells.
Nature
2004
, vol. 
432
 (pg. 
396
-
401
)
120
Clarke
 
MF
Dick
 
JE
Dirks
 
PB
, et al. 
Cancer stem cells—perspectives on current status and future directions: American Association for Cancer Research Workshop on Cancer Stem Cells.
Cancer Res
2006
, vol. 
66
 (pg. 
9339
-
9344
)
121
Jordan
 
CT
Lemischka
 
IR
Clonal and systemic analysis of long-term hematopoiesis in the mouse.
Genes Dev
1990
, vol. 
4
 (pg. 
220
-
232
)
122
Hope
 
K
Jin
 
L
Dick
 
JE
Acute myeloid leukemia originates from a hierarchy of leukemic stem cell classes that differ in self-renewal capacity.
Nat Immunol
2004
, vol. 
5
 (pg. 
738
-
743
)
123
Lessard
 
J
Sauvageau
 
G
Bmi-1 determines the proliferative capacity of normal and leukemic stem cells.
Nature
2003
, vol. 
423
 (pg. 
455
-
460
)
124
Yilmaz
 
OH
Valdez
 
R
Theisen
 
BK
, et al. 
Pten dependence distinguishes haematopoietic stem cells from leukaemia-initiating cells.
Nature
2006
, vol. 
441
 (pg. 
475
-
482
)
125
Guan
 
Y
Gerhard
 
B
Hogge
 
DE
Detection, isolation, and stimulation of quiescent primitive leukemic progenitor cells from patients with acute myeloid leukemia (AML).
Blood
2003
, vol. 
101
 (pg. 
3142
-
3149
)
126
Guzman
 
ML
Neering
 
SJ
Upchurch
 
D
, et al. 
Nuclear factor-kappaB is constitutively activated in primitive human acute myelogenous leukemia cells.
Blood
2001
, vol. 
98
 (pg. 
2301
-
2307
)
127
Terpstra
 
W
Ploemacher
 
RE
Prins
 
A
, et al. 
Fluorouracil selectively spares acute myeloid leukemia cells with long-term growth abilities in immunodeficient mice and in culture.
Blood
1996
, vol. 
88
 (pg. 
1944
-
1950
)
128
So
 
CW
Karsunky
 
H
Passegue
 
E
Cozzio
 
A
Weissman
 
IL
Cleary
 
ML
MLL-GAS7 transforms multipotent hematopoietic progenitors and induces mixed lineage leukemias in mice.
Cancer Cell
2003
, vol. 
3
 (pg. 
161
-
171
)
129
Huntly
 
BJ
Shigematsu
 
H
Deguchi
 
K
, et al. 
MOZ-TIF2, but not BCR-ABL, confers properties of leukemic stem cells to committed murine hematopoietic progenitors.
Cancer Cell
2004
, vol. 
6
 (pg. 
587
-
596
)
130
Krivtsov
 
AV
Twomey
 
D
Feng
 
Z
, et al. 
Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9.
Nature
2006
, vol. 
442
 (pg. 
818
-
822
)
131
Cozzio
 
A
Passegue
 
E
Ayton
 
PM
Karsunky
 
H
Cleary
 
ML
Weissman
 
IL
Similar MLL-associated leukemias arising from self-renewing stem cells and short-lived myeloid progenitors.
Genes Dev
2003
, vol. 
17
 (pg. 
3029
-
3035
)
132
Passegue
 
E
Wagner
 
EF
Weissman
 
IL
JunB deficiency leads to a myeloproliferative disorder arising from hematopoietic stem cells.
Cell
2004
, vol. 
119
 (pg. 
431
-
443
)
133
Chen
 
W
Kumar
 
AR
Hudson
 
WA
, et al. 
Malignant transformation initiated by Mll-AF9: gene dosage and critical target cells.
Cancer Cell
2008
, vol. 
13
 (pg. 
432
-
440
)
134
Wang
 
Y
Armstrong
 
SA
Cancer: inappropriate expression of stem cell programs?
Cell Stem Cell
2008
, vol. 
2
 (pg. 
297
-
299
)
135
Miyamoto
 
T
Weissman
 
IL
Akashi
 
K
AML1/ETO-expressing nonleukemic stem cells in acute myelogenous leukemia with 8;21 chromosomal translocation.
Proc Natl Acad Sci U S A
2000
, vol. 
97
 (pg. 
7521
-
7526
)
136
Majeti
 
R
Park
 
C
Weissman
 
I
Identification of a hierarchy of multipotent hematopoietic progenitors in human cord blood.
Cell Stem Cell
2007
, vol. 
1
 (pg. 
635
-
645
)
137
Jamieson
 
CH
Ailles
 
LE
Dylla
 
SJ
, et al. 
Granulocyte-macrophage progenitors as candidate leukemic stem cells in blast-crisis CML.
N Engl J Med
2004
, vol. 
351
 (pg. 
657
-
667
)
138
Nowell
 
P
Hungerford
 
D
A minute chromosome in human chronic granulocytic leukemia.
Science
1960
, vol. 
132
 pg. 
1497
 
139
Rowley
 
JD
A new consistent chromosomal abnormality in myelogenous leukemia identified by quinacrine fluorescence and giemsa staining.
Nature
1973
, vol. 
243
 (pg. 
290
-
293
)
140
Mani
 
SA
Guo
 
W
Liao
 
MJ
, et al. 
The epithelial-mesenchymal transition generates cells with properties of stem cells.
Cell
2008
, vol. 
133
 (pg. 
704
-
715
)
141
Darley
 
RL
Hoy
 
TG
Baines
 
P
Padua
 
RA
Burnett
 
AK
Mutant N-RAS induces erythroid lineage dysplasia in human CD34+ cells.
J Exp Med
1997
, vol. 
185
 (pg. 
1337
-
1347
)
142
Pereira
 
DS
Dorrell
 
C
Ito
 
CY
, et al. 
Retroviral transduction of TLS-ERG initiates a leukemogenic program in normal human hematopoietic cells.
Proc Natl Acad Sci U S A
1998
, vol. 
95
 (pg. 
8239
-
8244
)
143
Warner
 
JK
Wang
 
JC
Takenaka
 
K
, et al. 
Direct evidence for cooperating genetic events in the leukemic transformation of normal human hematopoietic cells.
Leukemia
2005
, vol. 
19
 (pg. 
1794
-
1805
)
144
Mulloy
 
JC
Cammenga
 
J
Berguido
 
FJ
, et al. 
Maintaining the self-renewal and differentiation potential of human CD34+ hematopoietic cells using a single genetic element.
Blood
2003
, vol. 
102
 (pg. 
4369
-
4376
)
145
Chalandon
 
Y
Jiang
 
X
Christ
 
O
, et al. 
BCR-ABL-transduced human cord blood cells produce abnormal populations in immunodeficient mice.
Leukemia
2005
, vol. 
19
 (pg. 
442
-
448
)
146
Kennedy
 
JA
Barabé
 
F
Investigating human leukemogenesis from cell lines to in vivo models of human leukemia.
Leukemia
2008
8
7
 
[Epub ahead of print]
147
Barabé
 
F
Kennedy
 
JA
Hope
 
KJ
Dick
 
JE
Modeling the initiation and progression of human acute leukemia in mice.
Science
2007
, vol. 
316
 (pg. 
600
-
604
)
148
Wei
 
J
Wunderlich
 
M
Fox
 
C
, et al. 
Microenvironment determines lineage fate in a human model of MLL-AF9 leukemia.
Cancer Cell
2008
, vol. 
13
 (pg. 
483
-
495
)
149
Greaves
 
MF
Wiemels
 
J
Origins of chromosome translocations in childhood leukaemia.
Nat Rev Cancer
2003
, vol. 
3
 (pg. 
639
-
649
)
150
Hong
 
D
Gupta
 
R
Ancliff
 
P
, et al. 
Initiating and cancer-propagating cells in TEL-AML1-associated childhood leukemia.
Science
2008
, vol. 
319
 (pg. 
336
-
339
)
151
Kelly
 
PN
Dakic
 
A
Adams
 
JM
Nutt
 
SL
Strasser
 
A
Tumor growth need not be driven by rare cancer stem cells.
Science
2007
, vol. 
317
 pg. 
337
 
152
Williams
 
RT
den Besten
 
W
Sherr
 
CJ
Cytokine-dependent imatinib resistance in mouse BCR-ABL+, Arf-null lymphoblastic leukemia.
Genes Dev
2007
, vol. 
21
 (pg. 
2283
-
2287
)
153
Somervaille
 
TC
Cleary
 
ML
Identification and characterization of leukemia stem cells in murine MLL-AF9 acute myeloid leukemia.
Cancer Cell
2006
, vol. 
10
 (pg. 
257
-
268
)
154
Kennedy
 
JA
Barabé
 
F
Poeppl
 
AG
Wang
 
JC
Dick
 
JE
Comment on “Tumor growth need not be driven by rare cancer stem cells.”
Science
2007
, vol. 
318
 pg. 
1722
  
author reply 1722
155
Liu
 
R
Wang
 
X
Chen
 
GY
, et al. 
The prognostic role of a gene signature from tumorigenic breast-cancer cells.
N Engl J Med
2007
, vol. 
356
 (pg. 
217
-
226
)
156
Guzman
 
ML
Rossi
 
RM
Neelakantan
 
S
, et al. 
An orally bioavailable parthenolide analog selectively eradicates acute myelogenous leukemia stem and progenitor cells.
Blood
2007
, vol. 
110
 (pg. 
4427
-
4435
)
157
Guzman
 
ML
Li
 
X
Corbett
 
CA
, et al. 
Rapid and selective death of leukemia stem and progenitor cells induced by the compound 4-benzyl, 2-methyl, 1,2,4-thiadiazolidine, 3,5 dione (TDZD-8).
Blood
2007
, vol. 
110
 (pg. 
4436
-
4444
)
158
Hassane
 
DC
Guzman
 
ML
Corbett
 
C
, et al. 
Discovery of agents that eradicate leukemia stem cells using an in silico screen of public gene expression data.
Blood
2008
, vol. 
111
 (pg. 
5654
-
5662
)
159
Feuring-Buske
 
M
Frankel
 
A
Gerhard
 
B
Hogge
 
D
Variable cytotoxicity of diphtheria toxin 388-granulocyte-macrophage colony-stimulating factor fusion protein for acute myelogenous leukemia stem cells.
Exp Hematol
2000
, vol. 
28
 (pg. 
1390
-
1400
)
160
Terpstra
 
W
Rozemuller
 
H
Breems
 
DA
, et al. 
Diphtheria toxin fused to granulocyte-macrophage colony-stimulating factor eliminates acute myeloid leukemia cells with the potential to initiate leukemia in immunodeficient mice, but spares normal hemopoietic stem cells.
Blood
1997
, vol. 
90
 (pg. 
3735
-
3742
)
161
Frankel
 
A
Liu
 
JS
Rizzieri
 
D
Hogge
 
D
Phase I clinical study of diphtheria toxin-interleukin 3 fusion protein in patients with acute myeloid leukemia and myelodysplasia.
Leuk Lymphoma
2008
, vol. 
49
 (pg. 
543
-
553
)
162
Bonnet
 
D
Warren
 
EH
Greenberg
 
PD
Dick
 
JE
Riddell
 
SR
CD8(+) minor histocompatibility antigen-specific cytotoxic T lymphocyte clones eliminate human acute myeloid leukemia stem cells.
Proc Natl Acad Sci U S A
1999
, vol. 
96
 (pg. 
8639
-
8644
)
163
Rosinski
 
KV
Fujii
 
N
Mito
 
JK
, et al. 
DDX3Y encodes a class I MHC-restricted H-Y antigen that is expressed in leukemic stem cells.
Blood
2008
, vol. 
111
 (pg. 
4817
-
4826
)
164
Jin
 
L
Hope
 
KJ
Zhai
 
Q
Smadja-Joffe
 
F
Dick
 
JE
Targeting of CD44 eradicates human acute myeloid leukemic stem cells.
Nat Med
2006
, vol. 
12
 (pg. 
1167
-
1174
)
165
Tavor
 
S
Petit
 
I
Porozov
 
S
, et al. 
CXCR4 regulates migration and development of human acute myelogenous leukemia stem cells in transplanted NOD/SCID mice.
Cancer Res
2004
, vol. 
64
 (pg. 
2817
-
2824
)
166
Papayannopoulou
 
T
Scadden
 
DT
Stem-cell ecology and stem cells in motion.
Blood
2008
, vol. 
111
 (pg. 
3923
-
3930
)
167
Ishikawa
 
F
Yoshida
 
S
Saito
 
Y
, et al. 
Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region.
Nat Biotechnol
2007
, vol. 
25
 (pg. 
1315
-
1321
)
168
Jordan
 
CT
Upchurch
 
D
Szilvassy
 
SJ
, et al. 
The interleukin-3 receptor alpha chain is a unique marker for human acute myelogenous leukemia stem cells.
Leukemia
2000
, vol. 
14
 (pg. 
1777
-
1784
)
169
Lock
 
R
Jin
 
L
Lee
 
E
, et al. 
CD123 (IL-3 receptor chain) neutralization by a monoclonal antibody selectively eliminates human acute myeloid leukemic stem cells.
Blood
2007
, vol. 
110
 pg. 
161
 
170
Ito
 
K
Bernardi
 
R
Morotti
 
A
, et al. 
PML targeting eradicates quiescent leukaemia-initiating cells.
Nature
2008
, vol. 
453
 (pg. 
1072
-
1078
)
171
Wang
 
JC
Evaluating therapeutic efficacy against cancer stem cells: new challenges posed by a new paradigm.
Cell Stem Cell
2007
, vol. 
1
 (pg. 
497
-
501
)
Sign in via your Institution