The model systems available for studying human hematopoiesis, malignant hematopoiesis, and hematopoietic stem cell (HSC) function in vivo have improved dramatically over the last decade, primarily due to improvements in xenograft mouse strains. Several recent reviews have focused on the historic development of immunodeficient mice over the last 2 decades, as well as their use in understanding human HSC and leukemia stem cell (LSC) biology and function in the context of a humanized mouse. However, in the intervening time since these reviews, a number of new mouse models, technical approaches, and scientific advances have been made. In this review, we update the reader on the newest and best models and approaches available for studying human malignant and normal HSCs in immunodeficient mice, including newly developed mice for use in chemotherapy testing and improved techniques for humanizing mice without laborious purification of HSC. We also review some relevant scientific findings from xenograft studies and highlight the continued limitations that confront researchers working with human HSC and LSC in vivo.

Since immunodeficient mice were first used in biomedical research, there has been a continual effort to improve their utility and expand their applicability to more areas of research. Over the last decade, with the advent of widespread transgenic approaches and the understanding by researchers of the potential for these models in moving our understanding of biology forward, a number of genetically engineered animals have become available. For a comprehensive overview of the history and use of xenografting, the reader is directed to a number of recent reviews.1-5  In this review, we will focus on the models and approaches of most relevance to researchers studying normal and malignant hematopoiesis and discuss future avenues to take that will address current limitations.

Of the different strains available to the research community, the most popular strains are the NOD/SCID-IL2RG−/− (NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ: NSG and NOD.Cg-Prkdcscid Il2rgtm1Sug/Jic: NOG) mice.6,7  Variations of this strain are becoming available through breeding strategies and genomic engineering techniques. It is clear that different strains will be optimal for different types of studies. For example, the humanized mouse, in which components of human bone marrow (BM), liver, and thymus (BLT) are grafted in immunodeficient mice, is most popular with investigators studying infectious disease due to the faithful development of mature, properly educated human T cells and a more complete, functional human immune system.5,8  The BLT approach is powerful but requires specialized surgical expertise and additional time, as well as tissue from human fetuses, raising feasibility issues. For studying hematologic malignancies, many researchers use the NSG mouse expressing cytokines that support human myelopoiesis (NSG-hSCF, hGM-CSF, hIL3, all driven from the cytomegalovirus promoter; NSGS), for their superiority in promoting robust engraftment of a wide range of patient samples.9,10  Designer mice are now becoming available through the efforts of a number of laboratories around the world, with Drs Mamoru Ito (NOG), Leonard Shultz (NSG), and Richard Flavell and Markus Manz (Rag2−/−IL2RG−/−) leading the way on improvements in these mouse strains. In a relatively short period of time, numerous genetically modified substrains will be available that seek to address the cross-species incompatibility of growth factors, receptors, adhesion molecules, and histocompatibility antigens. Engraftment levels in these 3 mouse strains are relatively equal now that the Rag2−/−IL2RG−/− mice express the human SIRPα molecule11,12  (Figure 1).

Figure 1

Development of immunodeficient mice for xenograft studies. Discovery of nude and SCID mice, in which T cells or T/B cells are deficient, contributed to the early-phase development of xenograft models. NOD/SCID mice, which harbor defects in T, B, and macrophage activity, supported higher levels of human engraftment and promoted the further development of xenomodels. Currently, mice with almost no murine immunity, such as NOG and NSG mice, are widely in xenograft studies. Variations of these strains including the BLT model and human cytokine mice are becoming available to provide optimal xenomodels for specific studies.

Figure 1

Development of immunodeficient mice for xenograft studies. Discovery of nude and SCID mice, in which T cells or T/B cells are deficient, contributed to the early-phase development of xenograft models. NOD/SCID mice, which harbor defects in T, B, and macrophage activity, supported higher levels of human engraftment and promoted the further development of xenomodels. Currently, mice with almost no murine immunity, such as NOG and NSG mice, are widely in xenograft studies. Variations of these strains including the BLT model and human cytokine mice are becoming available to provide optimal xenomodels for specific studies.

Close modal

For studies focused on human HSC self-renewal in vivo, the NSG/NOG mice are preferred due to their widespread availability and excellent engraftment rate. Although high levels of sustained engraftment can be obtained on transplant of human CD34+ cells into primary mice, secondary transplants are rarely performed, and for those reported, most show low levels of engrafted human cells. In 1 study, the authors demonstrated that the primitive stem/progenitor cells remain in active cycle for up to 8 weeks after engraftment, possibly implicating a lack of quiescence as the driving factor in the limited self-renewal.13  This could also explain the unusually high frequency of human CD34+ cells detected in the BM of xenografted mice (10-20% of the human graft) compared with a normal human BM containing 1% to 2% CD34+ cells at steady state.14,15  It is possible the signals that normally induce transplanted HSCs to enter quiescence are absent and/or non-cross-reactive in the immunodeficient mouse. To address this, Flavell’s group used Rag2−/−IL2RG−/− mice expressing human thrombopoietin (TPO) in place of murine. However, only a slight enhancement in serial transplantation was observed.16  It is likely that multiple signals are aberrant in the murine microenvironment, and a complex array of genetic changes will be needed to mimic the human BM niche. It is also possible that the presence of facilitating cells during transplant is critical for proper function of the HSCs. We recently described a simple procedure using total BM or cord blood samples in xenotransplants. The addition of the OKT3 monoclonal antibody to a cell mixture readily addressed the graft-versus-host disease that ensues on transplanting samples containing mature T cells into immunodeficient mice. This process also more closely mimics the stem cell transplant procedure as it is performed in humans.17  Limiting dilution analysis showed the stem cell frequency was as good or better than approaches using purified human CD34+ cells. Secondary transplant was very robust in a small pilot experiment, ranging between 10% and 50% multilineage human engraftment in the BM at 16 weeks, which is dramatically better than found in most published studies. More work is needed to determine whether this approach will contribute to solving the problems associated with analysis of human HSC self-renewal in xenotransplants.

Unfortunately, there is currently no standard in the field regarding which mice, approaches, and time points are best for measuring human HSC activity in vivo. A wide range of engraftment levels and durations have been used as experimental end points, and few researchers use secondary transplant as a readout due to poor engraftment. It is important that researchers adopt an accepted standard for measuring human HSC and hematopoietic stem and progenitor cell (HSPC) function as has been done for murine HSPC studies. This will allow a straightforward comparison of different approaches and models using a standard methodology.

Xenograft models have been used to determine immunophenotypes of human HSCs. In contrast to murine HSCs that are enriched in the CD34-negative fraction, CD34 has long been used as a positive marker of human HSCs not only in xenograft repopulation assays but also in clinical HSC transplantation.18,19  Additional HSC markers have been identified, and it is now possible to isolate human HSCs at the single cell level.20-28  Some primitive human HSCs may reside in a CD34-negative fraction similar to murine HSCs, but the frequency of CD34-negative SCID-repopulating cells (SRCs) is very low even with the use of additional HSC markers29-31  (Table 1). Interestingly, current evidence suggests that human HSCs have little correspondence with murine HSCs in terms of surface marker expression. Stem cell antigen-1 (Sca-1) is used extensively to enrich for murine HSCs, but it does not have a human homolog.32  Human HSCs express FLT3, whereas mouse HSCs do not.33  CD38 is not expressed on human HSCs but is expressed on murine HSCs.34-36  CD150, a member of the SLAM family receptors, has been widely used to isolate murine HSCs.37  However, human HSCs cannot be purified based only on SLAM markers.38  In addition, human HSCs expressing high levels of KIT (KIT-hi) contain more potent repopulating activity compared with HSCs expressing intermediate levels (KIT-int),39  whereas Kit-int murine HSCs are the best at repopulation and Kit-hi defines murine HSCs initiating the process of differentiation.40  These discrepancies likely indicate species differences but may partially be explained by different experimental conditions (syngeneic transplantation vs xenotransplantation). It should also be noted that many previous xenograft studies assessed engraftment of human HSCs at a relatively early time point (6-12 weeks after transplant; Table 1). Given that long-term HSCs show a delayed engraftment pattern,41  a longer period (or serial transplant) may be needed in future studies.

Table 1

Markers of human HSCs

MarkersSRC frequencyMouseTime point to assess HSC function (weeks)Reference
CD34+ 
 LinCD34+CD38 1 in 617 NS 6-9 22  
 LinCD34+CD38ρ(low) 1 in 30 NS 7-10 27  
 LinCD34+CD38CD90+CD45RA 1 in 10 NOG 12 26  
 LinCD34+CD38CD45RAThy1+ρ(low)CD49f+ 1 in a few NSG 20 + serial transplant 28  
CD34 
 LinCD34CD38CD93+ 1 in 7500 NS 8-10 29  
 LinCD34CD38CD93+ 1 in 6100 NSG ND 30  
 18LinCD34CD133+ 1 in 142 NOG 12 31  
MarkersSRC frequencyMouseTime point to assess HSC function (weeks)Reference
CD34+ 
 LinCD34+CD38 1 in 617 NS 6-9 22  
 LinCD34+CD38ρ(low) 1 in 30 NS 7-10 27  
 LinCD34+CD38CD90+CD45RA 1 in 10 NOG 12 26  
 LinCD34+CD38CD45RAThy1+ρ(low)CD49f+ 1 in a few NSG 20 + serial transplant 28  
CD34 
 LinCD34CD38CD93+ 1 in 7500 NS 8-10 29  
 LinCD34CD38CD93+ 1 in 6100 NSG ND 30  
 18LinCD34CD133+ 1 in 142 NOG 12 31  

18Lin, 18 lineage marker-negative; ND, not described; NOG, NOD/SCID IL2 receptor γ null; NS, NOD/SCID; NSG, NOD/SCID IL2 receptor γ−/−; SRC, SCID repopulating cells.

Studies for ex vivo HSC expansion combined with xenograft assays have revealed mechanisms governing self-renewal of human HSCs. Many studies have suggested that Notch signaling plays important roles in the regulation of human HSCs. Activation of Notch receptors by ligands (JAG1, DLL1, or DLL4) has been shown to promote ex vivo expansion of SRCs.42-45  In line with this, enforced expressions of HES1 (a Notch target gene) and NOV (an extracellular activator of Notch signaling) in human CB cells confer enhanced in vivo reconstitution ability in NOD/SCID mice.46,47  Notch signaling may also regulate the quiescent state of CD34 HSCs together with transforming growth factor β to repress the Wnt pathway.30  The effect of Notch on human HSCs appears to be dosage dependent, because low doses of DLL1 expand CD34+ cells, whereas higher doses induce apoptosis.48  Early studies also identified WNT and hedgehog pathways to promote HSC expansion,49-51  and several inhibitory pathways, including transforming growth factor β, tumor necrosis factor α, and chemokines, to suppress HSC proliferation.52  Recent studies have identified new pathways involved in human HSC regulation. Angptl5, a member of angiopoietin-like proteins, promotes 20-fold expansion of SRCs when used in serum-free culture media containing stem cell factor (SCF), TPO, fibroblast growth factor 1, and insulin-like growth factor binding protein 2 (IGFBP2).53  A neurite outgrowth factor Pleiotrophin also increases SRC counts in culture, and Notch and phosphatidylinositol 3-kinase pathways mediate the response to Pleiotrophin.54  A zebrafish screen and subsequent reports have shown that prostaglandin E2 (PGE2) enhances emergence and repopulating ability of HSCs through activation of WNT signaling and upregulation of CXCR4 and Survivin.55-58  Conversely, inhibition of endogenous PGE2 by nonsteroidal anti-inflammatory drug treatment promotes HSC egress from the BM to the circulating blood not only in mice but also in healthy human volunteers.59  More recently, an unbiased drug screening identified an aryl hydrocarbon receptor (AhR) antagonist SR1 that promotes ex vivo expansion of human CD34+ cells that retain the ability to engraft NSG mice.60  AhR requires ARNT hypoxia inducible factor (HIF1B) to regulate gene expression, and interestingly, ARNT is also required by HIF1A to enhance gene expression in response to hypoxia,61  suggesting the involvement of the HIF pathway in the regulation of human HSCs. Indeed, a study showed that knockdown of HIF2A, and to a lesser extent HIF1A, impedes the long-term repopulating ability of human cord blood (CB) CD34+ cells through increased reactive oxygen species (ROS) production and endoplasmic reticulum stress.62  In addition, treatment of recipient mice with the ROS scavenger N-acetyl-l-cysteine significantly improves engraftment of human HSCs in NOD/SCID and NSG mice.63  Another recent screening confirmed that multiple compounds suppressing the AhR pathway can promote HSC expansion ex vivo, although it appears that SR1 mainly expands short-term HSCs with limited regenerative potential. Interestingly, the study also identified a compound UM171 that efficiently promotes a robust ex vivo expansion of long-term human HSCs through AhR-independent mechanisms.64  As for the inhibitory signaling for human HSCs, short hairpin RNA library screening identified mitogen-activated protein kinase 14 (p38α) as a negative HSC regulator. Pharmacologic inhibition of p38 dramatically enhances the multilineage repopulation of human CB cells in NSG mice presumably by reducing levels of ROS.65 

Several transcription factors possess the ability to promote HSC expansion ex vivo. HOX proteins, HOXB4 and HOXC4, moderately (2- to 6-fold) improve the level of in vivo engraftment of human CD34+ cells.66,67  Similarly, enforced HLF expression confers increased repopulation potential to human HSCs.46  RUNX1 has isoform-specific activity to regulate self-renewal and differentiation of HSCs. Forced expression of RUNX1a, a short isoform of RUNX1, increases SRC activity of human HSCs and facilitates emergence of definitive human HSCs from human embryonic stem cells.68,69  In contrast, long isoforms of RUNX1 (RUNX1b and RUNX1c) inhibit repopulation of human/mouse HSCs in recipient mice by promoting myeloid differentiation68,70  and/or increasing quiescence of HSCs.71  GATA2 is highly expressed in quiescent HSC fractions, and enforcing GATA2 expression increases quiescence of human CB cells.72  DNA damage response is essential for maintaining HSC function, and p53 plays a key role in this process. In contrast to mouse HSCs with decreased sensitivity to cytotoxic agents, human HSCs exhibit enhanced p53-dependent apoptosis after irradiation. Inactivation of p53 reduces apoptosis and partially rescues the repopulating ability of the irradiated HSCs in primary recipient mice. However, p53-depleted HSCs show diminished SRC activity in secondary recipients probably due to accumulated DNA damage, suggesting that intact p53 is important to maintain human HSCs in the serial transplantation assay.73  In addition to these transcription factors, epigenetic modifiers also regulate human HSC function. The polycomb complex protein BMI1 was shown to promote the repopulation potential of human HSCs.74,75  Furthermore, addition of epigenetic drugs can increase the self-renewal of human HSCs ex vivo. Human CB cells treated with decitabine (DNA methylation inhibitor) and trichostatin A (histone deacetylase inhibitor) in the presence of hematopoietic cytokines resulted in significant expansion of CD34+CD90+ cells, including transplantable HSCs.76,77  Another histone deacetylase inhibitor, valproic acid, also stimulated proliferation and self-renewal of human HSCs accompanied by p21cip1/waf1 downregulation, WNT pathway activation, and HOXB4 upregulation.78  Finally, increasing evidence suggests the importance of miRNAs for human HSC regulation.79,80  Among the miRNAs highly expressed in human HSCs, miR-125 was shown to regulate HSC function positively,81  whereas miR-126 was shown to be a negative regulator of HSC proliferation.82 

Although these studies have revealed key players to maintain/expand human HSCs (Figure 2), it should be noted that signals needed for HSC expansion ex vivo may be different from those to maintain HSCs in human bodies. Indeed, a recent study showed that Notch signaling might be dispensable for self-renewal of human HSCs in vivo. Inhibition of Notch activity using a dominant-negative inhibitor of Mastermind-like (dnMAML) did not change SRCs when the dnMAML-transduced HSCs were directly transplanted to NSG mice, whereas dnMAML-transduced HSCs were not maintained ex vivo.83  Developing more humanized mice will be necessary to decipher actual mechanisms regulating human HSCs in vivo.

Figure 2

Regulators of human HSCs. Genes and pathways regulating human HSC function have been identified using xenograft models. Self-renewal, proliferation, differentiation, and apoptosis of human HSCs are regulated by the interplay of these stimulatory and inhibitory modulators. AhR, aryl hydrocarbon receptor; SR1, StemRegenin; Angptls, angiopoietin-like proteins; Hh, hedgehog; PGE2, prostaglandin E2.

Figure 2

Regulators of human HSCs. Genes and pathways regulating human HSC function have been identified using xenograft models. Self-renewal, proliferation, differentiation, and apoptosis of human HSCs are regulated by the interplay of these stimulatory and inhibitory modulators. AhR, aryl hydrocarbon receptor; SR1, StemRegenin; Angptls, angiopoietin-like proteins; Hh, hedgehog; PGE2, prostaglandin E2.

Close modal

Xenotransplant has proven highly successful for acute leukemia and to a lesser degree for the nonacute hematologic malignancies such as myelodysplastic syndrome (MDS), myeloproliferative neoplasms, and chronic lymphocytic leukemia (CLL).4  This indicates that the xenograft environment is suitable for the self-renewal of engrafting leukemia cells (SCID-leukemia-initiating cells [SL-IC]84 ). SL-IC is a functional description of those leukemia cells that possess engrafting potential, whereas LSC is a conceptual description of the self-renewing leukemia cell that can propagate the tumor. The connection between SL-IC and LSC is complex and beyond the scope of this review, and we will use the term LSCs throughout most of this review. Even in the context of the transgenic cytokine mice (eg, NSGS mice), SL-ICs do not exhaust and are readily transferred to secondary and tertiary mice.9  In contrast, HSCs are mobilized from the BM in NSGS mice, the human grafts are less durable than those in nontransgenic NSG mice, and HSCs do not efficiently transplant to secondary mice85  (and our unpublished data, January 2012). These data demonstrate a differential requirement for microenvironmental cues between HSCs and SL-ICs (and possibly LSCs). This may be due to the block in differentiation associated with leukemia, allowing unlimited self-renewal divisions in the absence of the normal niche signals needed by HSCs. These differences may also reflect discrepancies in the intrinsic nature of leukemia vs normal stem cells such that LSCs do not exhaust their self-renewal ability when mobilized into cycle, whereas HSC do (Figure 3).

Figure 3

The mouse niche may not support human HSCs in xenograft models. Mouse HSCs interact with the niche, which promotes quiescence. Several observations suggest that the mouse niche does not support human HSCs to maintain quiescence and stemness. As a result, human HSCs are proliferative in xenograft models, which leads to HSC differentiation. Human LSCs from aggressive types of leukemia do not appear to require the support from the mouse niche to maintain leukemic growth.

Figure 3

The mouse niche may not support human HSCs in xenograft models. Mouse HSCs interact with the niche, which promotes quiescence. Several observations suggest that the mouse niche does not support human HSCs to maintain quiescence and stemness. As a result, human HSCs are proliferative in xenograft models, which leads to HSC differentiation. Human LSCs from aggressive types of leukemia do not appear to require the support from the mouse niche to maintain leukemic growth.

Close modal

Although transfer of leukemia samples to immune deficient hosts has greatly improved, a substantial number of samples still fail to engraft even the most optimized host mice.9,86  Together, these data indicate that significant interspecies barriers to engraftment and expansion of certain LSCs remain, particularly for less aggressive hematologic malignancies. Specific subtypes of leukemia are significantly more problematic to engraft, for example, samples from patients with t(8;21). It may be that some subtypes of AML have low progenitor cell frequency, or, some samples may be particularly sensitive to the lack of a factor or cell type from the mouse BM. In the case of t(8;21), we and others have shown that signaling through TPO/mpl is particularly important for the growth of cells expressing AML1-ETO.87,88  Thus, hTPO knockin mice may improve engraftment of t(8;21) AML, as appears to be the case according to a presentation at the 2014 American Society of Hematology annual meeting.16,89,90  It should also be noted that engraftment itself does not indicate the detection of LSCs because residual normal HSCs in patients can repopulate mice. It is therefore important to confirm genetic and phenotypic abnormalities of the engrafted human cells in studies for human LSC. Interestingly, a recent xenograft study demonstrated the existence of preleukemic HSCs with a DNMT3A mutation. The preleukemic HSCs did not show leukemic engraftment characterized by a dominant myeloid graft but showed a multilineage repopulation advantage over nonmutated HSCs.91  Xenograft models combined with genetic analyses will enable the identification of preleukemic and leukemic stem cells in patients.

There is a growing list of studies that examine the effects of individual leukemia-associated oncogenes on human hematopoietic stem and progenitor cells (HSPCs), in effect building human leukemia using primary human HSPCs (Table 2). Although most of these studies report only partial phenotypes, consistent with a model of stepwise progression to transformation, there are now several examples of ex vivo generation of human leukemia from primary human cells as assayed by xenografts using defined genetic elements.92-95  These successes are limited to a select few recurrent translocations, likely indicating that most oncogenes require cooperating genetic mutations to elicit full-blown transformation. These studies also reiterate the finding that human cells are more refractory to transformation than are murine cells, as shown experimentally.96  It is also likely that BM microenvironmental cues are critical for preleukemic HSPC initiation and leukemia progression, similar to the situation for normal human HSCs, in contrast to a fully transformed LSCs.

Table 2

Leukemia development efforts using human HSPC and in vivo analyses

Gene(s)ReferenceKey findingsCell sourceMice used
TLS-ERG 143  Increased myeloid progenitors with serial replating and increased proliferation, decreased erythroid progenitors, increased lifespan of myeloid cells in culture. No engraftment in mice. UCB CD34+ lin NS 
HoxA10 144  Increased CFCs from in vitro cultures, block of erythroid differentiation. Engraftment skewed to myeloid in vivo, enriched for transduced cells. UCB lin, FL NS 
AML1-ETO 145  Clonal long-term culture of myeloid cells that retain multilineage potential, CFU-Cs, CAFC activity. Very low levels of human engraftment in mice. UCB/PBPC CD34+ NS 
STAT5A(1*6) 146  Increased proliferation of mostly erythroid progenitors, long-term CAFC cultures. Low levels of mostly erythroid lineage engraftment in mice. UCB CD34+ NS 
NRAS(G12D) 147  Increased proliferation, cell cycle, increased myeloid differentiation with reduced B and erythroid differentiation in vitro. Improved myeloid skewed multilineage engraftment in mice. UCB CD34+ NS 
TLS-ERG 148  Increased proliferation and lifespan, expansion of myeloid progenitors, incomplete myeloid differentiation. One immortalized line with high CD34+ and multilineage potential. Transient engraftment in mice. UCB lin NS 
BCR-ABL(p210) 149  Reduced myeloid colonies and increased erythroid colonies in vitro. A portion of engrafted mice showed myeloproliferation with increasing glyA positivity over time. UCB lin NS, NS-B2M 
CBFB-MYH11 150  Clonal expansion of myelomonocytic cells with eosinophilia and increased lifespan in vitro with reduced erythroid and B cell potential. Long-term cultures retain modest myeloid restricted engraftment of mice. UCB CD34+ NS-B2M 
TEL-JAK2 151  Increased proliferation and expansion of myeloid and erythroid cells in vitro. Mice engrafted similar to controls, but grafts skewed toward myeloid and erythroid lineages. Myelofibrosis induced in TEL-JAK2 mice. UCB lin NS 
NUP98-HoxA9 152  Increased expansion, decreased erythroid colonies, increased secondary myeloid colonies, more CAFCs. Proliferative advantage of transduced cells in engrafted mice. UCB CD34+ NS, NS-B2M, NSG 
MLL-AF9 94  Increased lifespan in vitro. pre-B ALL in 8/16 mice, AML in 2/16, mixed lineage in 1/16. UCB lin NS 
MLL-ENL 94  Increased lifespan in vitro. pre-B ALL in vivo. UCB lin NS 
MLL-AF9 93  Immortal myeloid and lymphoid cultures. B ALL in NS and NS-B2M mice and AML in NSS mice. UCB CD34+ NS, NS-B2M, NSS 
BMI1 74  Enhanced self-renewal (CAFC, LTC-IC) in vitro. Improved engraftment in primary and secondary (very low) mice. UCB CD34+ NS 
TEL-AML1 120  Serial engraftment of an abnormal CD34+CD38-CD19+ cell population, potentially containing pre-LSCs. UCB CD34+ NS 
BCR-ABL+BMI1 95  Increased proliferation, self-renewal (CAFC) in vitro. B-ALL generated in 4/8 mice, transplantable. UCB CD34+ NS 
MLL-AF9+NRAS(G12D) 9  Cytokine independent growth in vitro. Faster development of AML in vivo (relative to MLL-AF9 alone), engrafts non-conditioned hosts. UCB CD34+ NS, NSG, NSS, NSGS 
AML1-ETO+NRAS(G12D) 153  Increased culture lifespan, serial methylcell replating with erythroid to myeloid shift, cytokine independent growth in vitro. Improved engraftment over AE alone, subcutaneous tumors. UCB CD34+ NSG, NSGS 
MLL-AF9+FLT3-ITD 103  Cytokine independent growth in vitro. Faster development of AML in vivo (relative to MLL-AF9 alone), engrafts non-conditioned hosts. UCB CD34+ NSG, NSGS 
BCR-ABL+Ikaros 92  Proliferative advantage in vitro. Slight lifespan increase, skewed to erythroid, loss of B cell potential. Expansion of myeloid and erythroid cells in mice. Aggressive AML with disseminated myeloid sarcomas within four weeks; not transplantable. UCB lin NSG, NSGS 
ERG 154  Improved engraftment of transduced cells in thymus of mice, no difference otherwise. UCB CD34+ NSG 
AML1-ETO+c-Kit 155  Increased expansion over AE alone, c-kit attenuates AE-induced DNA damage, transient and low levels of human engraftment in mice. PBPC CD34+ NSG 
NUP98-HOXD13+MN1 156  Transient, non-transplantable myeloproliferation with MN1 alone. Aggressive and transplantable AML with NUP98-HoxD13+MN1 only in NSGS mice. UCB CD34+ NSG, NSGS 
PML-RARA 157  Reduced myeloid and erythroid colonies in vitro. Sorted CD34+CD38+, but not CD34+CD38- UCB cells led to myeloid skewed engraftment in NOG mice with several abnormal characteristics of APL cells. Low secondary transplantation from unsorted or CD34- cells. UCB CD34+ NOG 
Gene(s)ReferenceKey findingsCell sourceMice used
TLS-ERG 143  Increased myeloid progenitors with serial replating and increased proliferation, decreased erythroid progenitors, increased lifespan of myeloid cells in culture. No engraftment in mice. UCB CD34+ lin NS 
HoxA10 144  Increased CFCs from in vitro cultures, block of erythroid differentiation. Engraftment skewed to myeloid in vivo, enriched for transduced cells. UCB lin, FL NS 
AML1-ETO 145  Clonal long-term culture of myeloid cells that retain multilineage potential, CFU-Cs, CAFC activity. Very low levels of human engraftment in mice. UCB/PBPC CD34+ NS 
STAT5A(1*6) 146  Increased proliferation of mostly erythroid progenitors, long-term CAFC cultures. Low levels of mostly erythroid lineage engraftment in mice. UCB CD34+ NS 
NRAS(G12D) 147  Increased proliferation, cell cycle, increased myeloid differentiation with reduced B and erythroid differentiation in vitro. Improved myeloid skewed multilineage engraftment in mice. UCB CD34+ NS 
TLS-ERG 148  Increased proliferation and lifespan, expansion of myeloid progenitors, incomplete myeloid differentiation. One immortalized line with high CD34+ and multilineage potential. Transient engraftment in mice. UCB lin NS 
BCR-ABL(p210) 149  Reduced myeloid colonies and increased erythroid colonies in vitro. A portion of engrafted mice showed myeloproliferation with increasing glyA positivity over time. UCB lin NS, NS-B2M 
CBFB-MYH11 150  Clonal expansion of myelomonocytic cells with eosinophilia and increased lifespan in vitro with reduced erythroid and B cell potential. Long-term cultures retain modest myeloid restricted engraftment of mice. UCB CD34+ NS-B2M 
TEL-JAK2 151  Increased proliferation and expansion of myeloid and erythroid cells in vitro. Mice engrafted similar to controls, but grafts skewed toward myeloid and erythroid lineages. Myelofibrosis induced in TEL-JAK2 mice. UCB lin NS 
NUP98-HoxA9 152  Increased expansion, decreased erythroid colonies, increased secondary myeloid colonies, more CAFCs. Proliferative advantage of transduced cells in engrafted mice. UCB CD34+ NS, NS-B2M, NSG 
MLL-AF9 94  Increased lifespan in vitro. pre-B ALL in 8/16 mice, AML in 2/16, mixed lineage in 1/16. UCB lin NS 
MLL-ENL 94  Increased lifespan in vitro. pre-B ALL in vivo. UCB lin NS 
MLL-AF9 93  Immortal myeloid and lymphoid cultures. B ALL in NS and NS-B2M mice and AML in NSS mice. UCB CD34+ NS, NS-B2M, NSS 
BMI1 74  Enhanced self-renewal (CAFC, LTC-IC) in vitro. Improved engraftment in primary and secondary (very low) mice. UCB CD34+ NS 
TEL-AML1 120  Serial engraftment of an abnormal CD34+CD38-CD19+ cell population, potentially containing pre-LSCs. UCB CD34+ NS 
BCR-ABL+BMI1 95  Increased proliferation, self-renewal (CAFC) in vitro. B-ALL generated in 4/8 mice, transplantable. UCB CD34+ NS 
MLL-AF9+NRAS(G12D) 9  Cytokine independent growth in vitro. Faster development of AML in vivo (relative to MLL-AF9 alone), engrafts non-conditioned hosts. UCB CD34+ NS, NSG, NSS, NSGS 
AML1-ETO+NRAS(G12D) 153  Increased culture lifespan, serial methylcell replating with erythroid to myeloid shift, cytokine independent growth in vitro. Improved engraftment over AE alone, subcutaneous tumors. UCB CD34+ NSG, NSGS 
MLL-AF9+FLT3-ITD 103  Cytokine independent growth in vitro. Faster development of AML in vivo (relative to MLL-AF9 alone), engrafts non-conditioned hosts. UCB CD34+ NSG, NSGS 
BCR-ABL+Ikaros 92  Proliferative advantage in vitro. Slight lifespan increase, skewed to erythroid, loss of B cell potential. Expansion of myeloid and erythroid cells in mice. Aggressive AML with disseminated myeloid sarcomas within four weeks; not transplantable. UCB lin NSG, NSGS 
ERG 154  Improved engraftment of transduced cells in thymus of mice, no difference otherwise. UCB CD34+ NSG 
AML1-ETO+c-Kit 155  Increased expansion over AE alone, c-kit attenuates AE-induced DNA damage, transient and low levels of human engraftment in mice. PBPC CD34+ NSG 
NUP98-HOXD13+MN1 156  Transient, non-transplantable myeloproliferation with MN1 alone. Aggressive and transplantable AML with NUP98-HoxD13+MN1 only in NSGS mice. UCB CD34+ NSG, NSGS 
PML-RARA 157  Reduced myeloid and erythroid colonies in vitro. Sorted CD34+CD38+, but not CD34+CD38- UCB cells led to myeloid skewed engraftment in NOG mice with several abnormal characteristics of APL cells. Low secondary transplantation from unsorted or CD34- cells. UCB CD34+ NOG 

FL, fetal liver; NOG, NOD/SCID IL2 receptor γ null; NS, NOD/SCID; NS-B2M, NOD/SCID β-2 microglobulin; NSG, NOD/SCID IL2 receptor γ−/−; NSGS, NOD/SCID IL2RG−/− SCF GM-CSF IL-3; NSS, NOD/SCID SCF GM-CSF IL-3; PBPC, mobilized peripheral blood progenitor cell; UCB, umbilical cord blood.

Whether xenograft models can be used to predict response to leukemia therapy is an open question. Early studies showed a correlation between engraftment of AML samples in NOD/SCID mice and poor clinical outcome, and this finding has been replicated in NSG mice.97,98  However, whether the phenotypically defined LSC frequency (simplistically characterized as CD34+CD38) in AML is prognostic and whether this correlates with xenograft potential is controversial.98-100  Interestingly, it was shown that poor clinical outcome correlated with the degree to which the LSCs matched normal HSC gene expression.101  Whether this means that the transformative event(s) occurs initially in true HSCs or rather it results from transformed progenitors acquiring stem cell activity and gene expression remains unclear. More work remains to be done before strong conclusions can be drawn, but it may be possible to use data derived from xenografted AML samples to identify biomarkers that may predict clinical outcome.

Xenograft models also provide powerful tools to perform preclinical testing of candidate drugs for hematologic malignancies. However, we should keep it in mind that the dominant clone present at relapse in a patient is not necessarily the clone emerging on xenotransplantation.102  This may be due to different selective pressures in xenotransplant relative to what the AML clones encounter in patients undergoing induction and consolidation therapy. Xenomodels combined with therapeutic treatments may better mimic the situation of patients. We recently showed that NSGS mice can be used to mimic induction therapy during xenograft.103  One drawback to this approach is the frailty of SCID-based mice, making it impossible to simultaneously use conditioning to ensure engraftment of AML samples and subsequently treating with induction chemotherapy to force outgrowth of a relapse clone. We have crossed the NSGS mice with the NRG mice and found these mice (NRGS) are robust enough to permit conditioning, induction, intensified induction, and consolidation without death of the host (unpublished observations). This is consistent with the demonstration of radioresistance of the NRG strain.104  We expect the NRGS host combined with drug treatments will allow an examination of clonal evolution in leukemia under similar pressure to that in patients receiving therapies. These mice will be commercially available in 2015.

Xenograft studies have identified many useful markers to enrich LSCs, but it has become increasingly evident that LSC phenotype varies between individual patients. In addition, technical variation of xenograft assays also affects the results, with a trend that more immunodeficient strains allow the engraftment of variable LSC populations10,84,101,105-126  (Table 3). Early pioneering studies using NOD/SCID mice showed that AML is a hierarchical disease that is initiated by a rare subset of immature CD34+CD38 LSCs.84  However, LSCs were found in other compartments including the CD34+CD38+ fraction in recent studies using NSG mice or NOD/SCID mice treated with anti-CD122 to inhibit natural killer cell function.101,109,114  Furthermore, AMLs with NPM1 mutations were shown to contain LSCs in the CD34 fraction,110  indicating the subtype-specific features of AML LSCs. Although human LSCs and HSCs share similar immunophenotypes, some markers such as CD123 (IL3-Rα), CD96, CD44, CD47, CD32, CD25, CLL-1, IL1RAP, and TIM3 appear to be expressed higher in LSCs than normal HSCs, providing potential therapeutic targets to selectively eradicate LSCs.105,107,108,111,112,127-132  As another strategy to target LSCs in AML, a recent study showed that BCL2 inhibition reduced oxidative phosphorylation and selectively eradicated quiescent human LSCs.133  For chronic myeloid leukemia (CML), DPPIV (CD26) was shown to be specifically expressed in CML LSCs that can be a therapeutic target.116 

Table 3

Markers of human LSCs

LSC markerLeukemia samplesMice usedReferences
AML 
 CD34+CD38 FAB M1(1), M4(5), M5(1) NS 84  
 CD34+CD123+ FAB M1(5), M2(2), M4(7), MDS/AML (2), others (2) NS 105  
 CD34+CD38CD33+CD13+ CN-AML (7), CBF-AML (2), MLL-ENL (1), others (2) NS, NS-B2M 106  
 CD34+CLL1+ AMLs with FLT3-ITD (3) NS 107  
 CD34+CD38CD96+ CK-AML(1), CBFB-MYH11(1), PML-RARA(1), AML1-ETO(1), FAB M4(1) Rag2−/− IL2RG−/− 108  
 CD34+CD38+/− CN-AML (6), MLL-ENL(1), others(2) NS ± IVIG or anti-CD122, NS-B2M, NSG 109  
 CD34CD38+ AMLs with NPM1 mutation (16) NS-B2M, NSG + IVIG 110  
 TIM3+ FAB M1(2), M2(1), M4(1) NRG 111  
 CD34+CD38CD123+ AMLs from Fanconi Anemia patients (5) NSGS 112  
 CD34+CD38CD45RA+ (LMPP-like), CD34+CD38+CD45RA+ (GMP-like) AMLs spanning FAB subtypes, cytogenetic and molecular categories (100) NS + anti-CD122, NSG + anti-CD122 or IVIG 113  
 variable fractions CN-AML (4), CBFB-MYH11(2), MLL-AF6(1), others (4) NSG 114  
 variable fractions CN-AML(8), CBFB-MYH11 (1), CK-AML (2), others (5) NS + anti-CD122 101  
CML 
 CD34+CD38+/− chronic phase BCR-ABL+ (9) NS, NS-B2M 115  
 CD34+CD38CD26+ chronic phase BCR-ABL+ NSG 116  
MDS 
 CD34+CD38CD90+CD45RA Monosomy 7 (2), low-risk MDS (4) NSG 125  
 CD34+CD38 MDS 5q (7), MDS RCMD (14), MDS RAEB I (6), MDS-U (2), MDS RARS (2) NSG, NSGS + patient-derived MSCs 10  
 CD34+CD38CD90+CD45RA MDS 5q (2) NSG 126  
B-ALL 
 CD34+CD38 BCR-ABL[p190](7) NS 117  
 CD34+CD10, CD34+CD19 ETV6-RUNX1(3), 11q23(1), others(8) NS 118  
 CD19+ ETV6-RUNX1(3), BCR-ABL[p210](5), BCR-ABL[p190](5) NS 119  
 CD34+CD38/low CD19+ ETV-RUNX1(4) NS 120  
 variable fractions Childhood ALL(13) including MLL-AF4 and ETV6-RUNX1 NS+ anti-CD122, NSG 121  
T-ALL 
 CD34+CD4, CD34+CD7 T-ALL (7) NS 122  
 CD7+CD1a Notch1 mutation (3) and others (8) NS + anti-CD122 123  
 CD7+CD1a (major), CD7+CD1a+ (minor) NSG 
CLL 
 CD34+CD38CD90+ del13q14 (7), others (9) NSG, NRG 124  
LSC markerLeukemia samplesMice usedReferences
AML 
 CD34+CD38 FAB M1(1), M4(5), M5(1) NS 84  
 CD34+CD123+ FAB M1(5), M2(2), M4(7), MDS/AML (2), others (2) NS 105  
 CD34+CD38CD33+CD13+ CN-AML (7), CBF-AML (2), MLL-ENL (1), others (2) NS, NS-B2M 106  
 CD34+CLL1+ AMLs with FLT3-ITD (3) NS 107  
 CD34+CD38CD96+ CK-AML(1), CBFB-MYH11(1), PML-RARA(1), AML1-ETO(1), FAB M4(1) Rag2−/− IL2RG−/− 108  
 CD34+CD38+/− CN-AML (6), MLL-ENL(1), others(2) NS ± IVIG or anti-CD122, NS-B2M, NSG 109  
 CD34CD38+ AMLs with NPM1 mutation (16) NS-B2M, NSG + IVIG 110  
 TIM3+ FAB M1(2), M2(1), M4(1) NRG 111  
 CD34+CD38CD123+ AMLs from Fanconi Anemia patients (5) NSGS 112  
 CD34+CD38CD45RA+ (LMPP-like), CD34+CD38+CD45RA+ (GMP-like) AMLs spanning FAB subtypes, cytogenetic and molecular categories (100) NS + anti-CD122, NSG + anti-CD122 or IVIG 113  
 variable fractions CN-AML (4), CBFB-MYH11(2), MLL-AF6(1), others (4) NSG 114  
 variable fractions CN-AML(8), CBFB-MYH11 (1), CK-AML (2), others (5) NS + anti-CD122 101  
CML 
 CD34+CD38+/− chronic phase BCR-ABL+ (9) NS, NS-B2M 115  
 CD34+CD38CD26+ chronic phase BCR-ABL+ NSG 116  
MDS 
 CD34+CD38CD90+CD45RA Monosomy 7 (2), low-risk MDS (4) NSG 125  
 CD34+CD38 MDS 5q (7), MDS RCMD (14), MDS RAEB I (6), MDS-U (2), MDS RARS (2) NSG, NSGS + patient-derived MSCs 10  
 CD34+CD38CD90+CD45RA MDS 5q (2) NSG 126  
B-ALL 
 CD34+CD38 BCR-ABL[p190](7) NS 117  
 CD34+CD10, CD34+CD19 ETV6-RUNX1(3), 11q23(1), others(8) NS 118  
 CD19+ ETV6-RUNX1(3), BCR-ABL[p210](5), BCR-ABL[p190](5) NS 119  
 CD34+CD38/low CD19+ ETV-RUNX1(4) NS 120  
 variable fractions Childhood ALL(13) including MLL-AF4 and ETV6-RUNX1 NS+ anti-CD122, NSG 121  
T-ALL 
 CD34+CD4, CD34+CD7 T-ALL (7) NS 122  
 CD7+CD1a Notch1 mutation (3) and others (8) NS + anti-CD122 123  
 CD7+CD1a (major), CD7+CD1a+ (minor) NSG 
CLL 
 CD34+CD38CD90+ del13q14 (7), others (9) NSG, NRG 124  

CK, complex karyotype; CN, cytogenetically normal; FAB, French-American-British classification system; GMP, granulocyte-monocyte progenitor; LMPP, lymphoid-primed multipotent progenitor; MSCs, mesenchymal stromal cells; NRG, NOD/Rag1−/− IL2RG−/−; NS, NOD/SCID; NS-B2M, NOD/SCID β-2 microglobulin; NSG, NOD/SCID IL2 receptor γ−/−; NSGS, NOD/SCID IL2RG−/− SCF GM-CSF IL-3.

Studies using purified MDS patient samples have shown that MDS is a clonal disease of the HSCs.134  MDS has been an extremely challenging disease to model in the immunodeficient mouse. The difficulty encountered in xenografting MDS samples may relate to special requirements of MDS cells for specific signaling events in the niche.135  Recently, transplantation of matched mesenchymal stroma cells were shown to facilitate xenotransplant of MDS stem cells.10  This study showed that the MDS stem cell may be dependent on several differentially expressed genes from MDS stroma cells, some of which are induced by the MDS cells, including LIF, and may represent new targets for therapy. Additional xenograft studies have confirmed that only highly purified stem cells from multiple del(5q) MDS patients were able to engraft mice, showing definitively that MDS is driven by distinct identifiable malignant stem cells.126  These MDS stem cells generated myeloid progenitor cells in xenografts but had no lymphoid potential. Another study engrafting del(5q) MDS stem cells also found a myeloid-restricted output with a reduction of granulocyte-monocyte progenitors, similar to what is observed in low risk MDS patients.125  It is apparent the MDS stem cell has a skewed lineage potential in xenografts, similar to what is reported in patients.136  These MDS xenograft models may prove particularly useful for testing new therapies for this difficult disease, as shown recently in a xenograft model of MDS using a lenalidomide-responsive MDS cell line.137 

Hierarchical organization is less strict in acute lymphoid leukemia (ALL). Some early studies showed the exclusive engraftment of primitive ALL LSCs (CD34+CD19/CD10 for B-ALL and CD34+CD4/CD7 for T-ALL) in NOD/SCID mice,117,118,122  but many later studies demonstrated LSC potential of cells with phenotypic characteristics of differentiated progenitors (CD19+ cells for B-ALL and CD7+ cells for T-ALL).119-121,123  Interestingly, a recent study suggests that CLL may be derived from primitive HSPCs. Purified HSCs from CLL patients could initiate xenografts that gave clonal expansion of B cells, whereas other isolated fractions did not have this ability.124  Interestingly, leukemic clones derived from the same patient but expanded in different mice exhibited distinct VDJ recombination events, clearly suggesting a primitive preleukemic stem cell is driving a disease that continues to evolve in the xenografted mice. As immunodeficient models and techniques improve, more such studies will be possible using primary patient samples from a variety of hematologic malignancies.

Advances in the available mouse strains and in the techniques used to humanize the murine immune system and to model human hematologic malignancies in vivo have been dramatic over the last few years. Numerous important findings have been made with regard to the nature and function of human HSCs and LSCs. Nevertheless, some major hurdles remain to be overcome to broaden the use of this approach to studying human LT-HSCs and to model hematologic malignancies that are not currently amenable to xenograft.

The arm of the immune system that has proven most difficult to model in immunodeficient mice is the megakaryocytic and erythrocytic lineages. Numerous reasons have been proposed for the problems associated with development of these lineages, and it is likely that the problems are multifaceted and will not be solved with a single genetic fix.4  Phagocytosis of red blood cells and platelets by murine macrophages has been shown to play a role in the lack of these cell types in the periphery of humanized mice.138,139  However, humanized mice also have a dramatic defect in the generation of the megakaryocytic/erythroid progenitor (MEP), and this deficiency is not corrected in Rag2−/−IL2RG−/− mice expressing human thrombopoietin.16  The lack of human megakaryocytes in the BM of humanized mice could play a role in the quiescence and self-renewal problems of the human HSCs, given the recent findings that mature megakaryocytes are essential niche cells for HSCs.140,141  One possible contributing factor to both the HSC and MEP defects is the increased ROS levels found in NS and NSG mice.63  ROS levels have been shown to be particularly low in MEP, and low levels of ROS in the common myeloid progenitor correlate with gene expression signatures that favor the MEP fate.142  It is possible that correction of the high ROS levels in these immunodeficient mice may allow for development of a more complete immune system and simultaneously promote the quiescence and self-renewal of the human HSC through multiple mechanisms.

The development of genetically modified immunodeficient mice will continue, likely at an increased pace. These modifications will include not only substitutions of growth factors, receptors, and adhesion molecules but also major histocompatibility molecules to promote proper education and development of cells dependent on these molecules for function. In the near future, it is likely that an improved xenograft mouse will be available for most blood researchers, including specialists focused on human HSCs and LSCs. The in vivo approaches that are critical to a better understanding of stem cell function will be even more powerful than the excellent models that currently exist. In time, our scientific understanding of human stem cell function will rival that of the murine stem cell.

Contribution: S.G., M.W., and J.C.M. wrote the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: James C. Mulloy, Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45226; e-mail: james.mulloy@cchmc.org.

1
Laurenti
 
E
Dick
 
JE
Molecular and functional characterization of early human hematopoiesis.
Ann N Y Acad Sci
2012
, vol. 
1266
 (pg. 
68
-
71
)
2
Doulatov
 
S
Notta
 
F
Laurenti
 
E
Dick
 
JE
Hematopoiesis: a human perspective.
Cell Stem Cell
2012
, vol. 
10
 
2
(pg. 
120
-
136
)
3
Miller
 
PH
Knapp
 
DJ
Eaves
 
CJ
Heterogeneity in hematopoietic stem cell populations: implications for transplantation.
Curr Opin Hematol
2013
, vol. 
20
 
4
(pg. 
257
-
264
)
4
Rongvaux
 
A
Takizawa
 
H
Strowig
 
T
, et al. 
Human hemato-lymphoid system mice: current use and future potential for medicine.
Annu Rev Immunol
2013
, vol. 
31
 (pg. 
635
-
674
)
5
Akkina
 
R
New generation humanized mice for virus research: comparative aspects and future prospects.
Virology
2013
, vol. 
435
 
1
(pg. 
14
-
28
)
6
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
 
9
(pg. 
3175
-
3182
)
7
Shultz
 
LD
Lyons
 
BL
Burzenski
 
LM
, et al. 
Human lymphoid and myeloid cell development in NOD/LtSz-scid IL2R gamma null mice engrafted with mobilized human hemopoietic stem cells.
J Immunol
2005
, vol. 
174
 
10
(pg. 
6477
-
6489
)
8
Melkus
 
MW
Estes
 
JD
Padgett-Thomas
 
A
, et al. 
Humanized mice mount specific adaptive and innate immune responses to EBV and TSST-1.
Nat Med
2006
, vol. 
12
 
11
(pg. 
1316
-
1322
)
9
Wunderlich
 
M
Chou
 
FS
Link
 
KA
, et al. 
AML xenograft efficiency is significantly improved in NOD/SCID-IL2RG mice constitutively expressing human SCF, GM-CSF and IL-3.
Leukemia
2010
, vol. 
24
 
10
(pg. 
1785
-
1788
)
10
Medyouf
 
H
Mossner
 
M
Jann
 
JC
, et al. 
Myelodysplastic cells in patients reprogram mesenchymal stromal cells to establish a transplantable stem cell niche disease unit.
Cell Stem Cell
2014
, vol. 
14
 
6
(pg. 
824
-
837
)
11
Strowig
 
T
Rongvaux
 
A
Rathinam
 
C
, et al. 
Transgenic expression of human signal regulatory protein alpha in Rag2-/-gamma(c)-/- mice improves engraftment of human hematopoietic cells in humanized mice.
Proc Natl Acad Sci USA
2011
, vol. 
108
 
32
(pg. 
13218
-
13223
)
12
Takenaka
 
K
Prasolava
 
TK
Wang
 
JC
, et al. 
Polymorphism in Sirpa modulates engraftment of human hematopoietic stem cells.
Nat Immunol
2007
, vol. 
8
 
12
(pg. 
1313
-
1323
)
13
Shima
 
H
Takubo
 
K
Tago
 
N
, et al. 
Acquisition of G₀ state by CD34-positive cord blood cells after bone marrow transplantation.
Exp Hematol
2010
, vol. 
38
 
12
(pg. 
1231
-
1240
)
14
Ema
 
H
Suda
 
T
Miura
 
Y
Nakauchi
 
H
Colony formation of clone-sorted human hematopoietic progenitors.
Blood
1990
, vol. 
75
 
10
(pg. 
1941
-
1946
)
15
Bender
 
JG
Unverzagt
 
KL
Walker
 
DE
, et al. 
Identification and comparison of CD34-positive cells and their subpopulations from normal peripheral blood and bone marrow using multicolor flow cytometry.
Blood
1991
, vol. 
77
 
12
(pg. 
2591
-
2596
)
16
Rongvaux
 
A
Willinger
 
T
Takizawa
 
H
, et al. 
Human thrombopoietin knockin mice efficiently support human hematopoiesis in vivo.
Proc Natl Acad Sci USA
2011
, vol. 
108
 
6
(pg. 
2378
-
2383
)
17
Wunderlich
 
M
Brooks
 
RA
Panchal
 
R
Rhyasen
 
GW
Danet-Desnoyers
 
G
Mulloy
 
JC
OKT3 prevents xenogeneic GVHD and allows reliable xenograft initiation from unfractionated human hematopoietic tissues.
Blood
2014
, vol. 
123
 
24
(pg. 
e134
-
e144
)
18
Krause
 
DS
Fackler
 
MJ
Civin
 
CI
May
 
WS
CD34: structure, biology, and clinical utility.
Blood
1996
, vol. 
87
 
1
(pg. 
1
-
13
)
19
Dick
 
JE
Stem cell concepts renew cancer research.
Blood
2008
, vol. 
112
 
13
(pg. 
4793
-
4807
)
20
Hao
 
QL
Shah
 
AJ
Thiemann
 
FT
Smogorzewska
 
EM
Crooks
 
GM
A functional comparison of CD34 + CD38- cells in cord blood and bone marrow.
Blood
1995
, vol. 
86
 
10
(pg. 
3745
-
3753
)
21
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 USA
2002
, vol. 
99
 
1
(pg. 
413
-
418
)
22
Bhatia
 
M
Wang
 
JC
Kapp
 
U
Bonnet
 
D
Dick
 
JE
Purification of primitive human hematopoietic cells capable of repopulating immune-deficient mice.
Proc Natl Acad Sci USA
1997
, vol. 
94
 
10
(pg. 
5320
-
5325
)
23
Murray
 
L
Chen
 
B
Galy
 
A
, et al. 
Enrichment of human hematopoietic stem cell activity in the CD34+Thy-1+Lin- subpopulation from mobilized peripheral blood.
Blood
1995
, vol. 
85
 
2
(pg. 
368
-
378
)
24
Mayani
 
H
Dragowska
 
W
Lansdorp
 
PM
Characterization of functionally distinct subpopulations of CD34+ cord blood cells in serum-free long-term cultures supplemented with hematopoietic cytokines.
Blood
1993
, vol. 
82
 
9
(pg. 
2664
-
2672
)
25
Baum
 
CM
Weissman
 
IL
Tsukamoto
 
AS
Buckle
 
AM
Peault
 
B
Isolation of a candidate human hematopoietic stem-cell population.
Proc Natl Acad Sci USA
1992
, vol. 
89
 
7
(pg. 
2804
-
2808
)
26
Majeti
 
R
Park
 
CY
Weissman
 
IL
Identification of a hierarchy of multipotent hematopoietic progenitors in human cord blood.
Cell Stem Cell
2007
, vol. 
1
 
6
(pg. 
635
-
645
)
27
McKenzie
 
JL
Takenaka
 
K
Gan
 
OI
Doedens
 
M
Dick
 
JE
Low rhodamine 123 retention identifies long-term human hematopoietic stem cells within the Lin-CD34+CD38- population.
Blood
2007
, vol. 
109
 
2
(pg. 
543
-
545
)
28
Notta
 
F
Doulatov
 
S
Laurenti
 
E
Poeppl
 
A
Jurisica
 
I
Dick
 
JE
 
Isolation of single human hematopoietic stem cells capable of long-term multilineage engraftment. Science 2011;333(6039):218-221
29
Danet
 
GH
Luongo
 
JL
Butler
 
G
, et al. 
C1qRp defines a new human stem cell population with hematopoietic and hepatic potential.
Proc Natl Acad Sci USA
2002
, vol. 
99
 
16
(pg. 
10441
-
10445
)
30
Anjos-Afonso
 
F
Currie
 
E
Palmer
 
HG
Foster
 
KE
Taussig
 
DC
Bonnet
 
D
CD34(-) cells at the apex of the human hematopoietic stem cell hierarchy have distinctive cellular and molecular signatures.
Cell Stem Cell
2013
, vol. 
13
 
2
(pg. 
161
-
174
)
31
Takahashi
 
M
Matsuoka
 
Y
Sumide
 
K
, et al. 
CD133 is a positive marker for a distinct class of primitive human cord blood-derived CD34-negative hematopoietic stem cells.
Leukemia
2014
, vol. 
28
 
6
(pg. 
1308
-
1315
)
32
Holmes
 
C
Stanford
 
WL
Concise review: stem cell antigen-1: expression, function, and enigma.
Stem Cells
2007
, vol. 
25
 
6
(pg. 
1339
-
1347
)
33
Sitnicka
 
E
Buza-Vidas
 
N
Larsson
 
S
Nygren
 
JM
Liuba
 
K
Jacobsen
 
SE
Human CD34+ hematopoietic stem cells capable of multilineage engrafting NOD/SCID mice express flt3: distinct flt3 and c-kit expression and response patterns on mouse and candidate human hematopoietic stem cells.
Blood
2003
, vol. 
102
 
3
(pg. 
881
-
886
)
34
Tajima
 
F
Deguchi
 
T
Laver
 
JH
Zeng
 
H
Ogawa
 
M
Reciprocal expression of CD38 and CD34 by adult murine hematopoietic stem cells.
Blood
2001
, vol. 
97
 
9
(pg. 
2618
-
2624
)
35
Randall
 
TD
Lund
 
FE
Howard
 
MC
Weissman
 
IL
Expression of murine CD38 defines a population of long-term reconstituting hematopoietic stem cells.
Blood
1996
, vol. 
87
 
10
(pg. 
4057
-
4067
)
36
Zhao
 
Y
Lin
 
Y
Zhan
 
Y
, et al. 
Murine hematopoietic stem cell characterization and its regulation in BM transplantation.
Blood
2000
, vol. 
96
 
9
(pg. 
3016
-
3022
)
37
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
 
7
(pg. 
1109
-
1121
)
38
Larochelle
 
A
Savona
 
M
Wiggins
 
M
, et al. 
Human and rhesus macaque hematopoietic stem cells cannot be purified based only on SLAM family markers.
Blood
2011
, vol. 
117
 
5
(pg. 
1550
-
1554
)
39
Cosgun
 
KN
Rahmig
 
S
Mende
 
N
, et al. 
Kit regulates HSC engraftment across the human-mouse species barrier.
Cell Stem Cell
2014
, vol. 
15
 
2
(pg. 
227
-
238
)
40
Shin
 
JY
Hu
 
W
Naramura
 
M
Park
 
CY
High c-Kit expression identifies hematopoietic stem cells with impaired self-renewal and megakaryocytic bias.
J Exp Med
2014
, vol. 
211
 
2
(pg. 
217
-
231
)
41
Cheung
 
AM
Nguyen
 
LV
Carles
 
A
, et al. 
Analysis of the clonal growth and differentiation dynamics of primitive barcoded human cord blood cells in NSG mice.
Blood
2013
, vol. 
122
 
18
(pg. 
3129
-
3137
)
42
Karanu
 
FN
Murdoch
 
B
Gallacher
 
L
, et al. 
The notch ligand jagged-1 represents a novel growth factor of human hematopoietic stem cells.
J Exp Med
2000
, vol. 
192
 
9
(pg. 
1365
-
1372
)
43
Karanu
 
FN
Murdoch
 
B
Miyabayashi
 
T
, et al. 
Human homologues of Delta-1 and Delta-4 function as mitogenic regulators of primitive human hematopoietic cells.
Blood
2001
, vol. 
97
 
7
(pg. 
1960
-
1967
)
44
Ohishi
 
K
Varnum-Finney
 
B
Bernstein
 
ID
Delta-1 enhances marrow and thymus repopulating ability of human CD34(+)CD38(-) cord blood cells.
J Clin Invest
2002
, vol. 
110
 
8
(pg. 
1165
-
1174
)
45
Suzuki
 
T
Yokoyama
 
Y
Kumano
 
K
, et al. 
Highly efficient ex vivo expansion of human hematopoietic stem cells using Delta1-Fc chimeric protein.
Stem Cells
2006
, vol. 
24
 
11
(pg. 
2456
-
2465
)
46
Shojaei
 
F
Trowbridge
 
J
Gallacher
 
L
, et al. 
Hierarchical and ontogenic positions serve to define the molecular basis of human hematopoietic stem cell behavior.
Dev Cell
2005
, vol. 
8
 
5
(pg. 
651
-
663
)
47
Gupta
 
R
Hong
 
D
Iborra
 
F
Sarno
 
S
Enver
 
T
 
NOV (CCN3) functions as a regulator of human hematopoietic stem or progenitor cells. Science 2007;316(5824):590-593
48
Delaney
 
C
Varnum-Finney
 
B
Aoyama
 
K
Brashem-Stein
 
C
Bernstein
 
ID
Dose-dependent effects of the Notch ligand Delta1 on ex vivo differentiation and in vivo marrow repopulating ability of cord blood cells.
Blood
2005
, vol. 
106
 
8
(pg. 
2693
-
2699
)
49
Murdoch
 
B
Chadwick
 
K
Martin
 
M
, et al. 
Wnt-5A augments repopulating capacity and primitive hematopoietic development of human blood stem cells in vivo.
Proc Natl Acad Sci USA
2003
, vol. 
100
 
6
(pg. 
3422
-
3427
)
50
Trowbridge
 
JJ
Xenocostas
 
A
Moon
 
RT
Bhatia
 
M
Glycogen synthase kinase-3 is an in vivo regulator of hematopoietic stem cell repopulation.
Nat Med
2006
, vol. 
12
 
1
(pg. 
89
-
98
)
51
Bhardwaj
 
G
Murdoch
 
B
Wu
 
D
, et al. 
Sonic hedgehog induces the proliferation of primitive human hematopoietic cells via BMP regulation.
Nat Immunol
2001
, vol. 
2
 
2
(pg. 
172
-
180
)
52
Biancotti
 
JC
Town
 
T
 
Increasing hematopoietic stem cell yield to develop mice with human immune systems. BioMed Res Int 2013;2013:740892
53
Zhang
 
CC
Kaba
 
M
Iizuka
 
S
Huynh
 
H
Lodish
 
HF
Angiopoietin-like 5 and IGFBP2 stimulate ex vivo expansion of human cord blood hematopoietic stem cells as assayed by NOD/SCID transplantation.
Blood
2008
, vol. 
111
 
7
(pg. 
3415
-
3423
)
54
Himburg
 
HA
Muramoto
 
GG
Daher
 
P
, et al. 
Pleiotrophin regulates the expansion and regeneration of hematopoietic stem cells.
Nat Med
2010
, vol. 
16
 
4
(pg. 
475
-
482
)
55
North
 
TE
Goessling
 
W
Walkley
 
CR
, et al. 
Prostaglandin E2 regulates vertebrate haematopoietic stem cell homeostasis.
Nature
2007
, vol. 
447
 
7147
(pg. 
1007
-
1011
)
56
Goessling
 
W
Allen
 
RS
Guan
 
X
, et al. 
Prostaglandin E2 enhances human cord blood stem cell xenotransplants and shows long-term safety in preclinical nonhuman primate transplant models.
Cell Stem Cell
2011
, vol. 
8
 
4
(pg. 
445
-
458
)
57
Goessling
 
W
North
 
TE
Loewer
 
S
, et al. 
Genetic interaction of PGE2 and Wnt signaling regulates developmental specification of stem cells and regeneration.
Cell
2009
, vol. 
136
 
6
(pg. 
1136
-
1147
)
58
Hoggatt
 
J
Singh
 
P
Sampath
 
J
Pelus
 
LM
Prostaglandin E2 enhances hematopoietic stem cell homing, survival, and proliferation.
Blood
2009
, vol. 
113
 
22
(pg. 
5444
-
5455
)
59
Hoggatt
 
J
Mohammad
 
KS
Singh
 
P
, et al. 
Differential stem- and progenitor-cell trafficking by prostaglandin E2.
Nature
2013
, vol. 
495
 
7441
(pg. 
365
-
369
)
60
Boitano
 
AE
Wang
 
J
Romeo
 
R
, et al. 
 
Aryl hydrocarbon receptor antagonists promote the expansion of human hematopoietic stem cells. Science 2010;329(5997):1345-1348
61
Vorrink
 
SU
Domann
 
FE
Regulatory crosstalk and interference between the xenobiotic and hypoxia sensing pathways at the AhR-ARNT-HIF1α signaling node.
Chem Biol Interact
2014
, vol. 
218
 (pg. 
82
-
88
)
62
Rouault-Pierre
 
K
Lopez-Onieva
 
L
Foster
 
K
, et al. 
HIF-2α protects human hematopoietic stem/progenitors and acute myeloid leukemic cells from apoptosis induced by endoplasmic reticulum stress.
Cell Stem Cell
2013
, vol. 
13
 
5
(pg. 
549
-
563
)
63
Hu
 
L
Cheng
 
H
Gao
 
Y
, et al. 
Antioxidant N-acetyl-L-cysteine increases engraftment of human hematopoietic stem cells in immune-deficient mice.
Blood
2014
, vol. 
124
 
20
(pg. 
e45
-
e48
)
64
Fares
 
I
Chagraoui
 
J
Gareau
 
Y
, et al. 
Cord blood expansion. Pyrimidoindole derivatives are agonists of human hematopoietic stem cell self-renewal.
Science
2014
, vol. 
345
 
6203
(pg. 
1509
-
1512
)
65
Baudet
 
A
Karlsson
 
C
Safaee Talkhoncheh
 
M
Galeev
 
R
Magnusson
 
M
Larsson
 
J
RNAi screen identifies MAPK14 as a druggable suppressor of human hematopoietic stem cell expansion.
Blood
2012
, vol. 
119
 
26
(pg. 
6255
-
6258
)
66
Amsellem
 
S
Pflumio
 
F
Bardinet
 
D
, et al. 
Ex vivo expansion of human hematopoietic stem cells by direct delivery of the HOXB4 homeoprotein.
Nat Med
2003
, vol. 
9
 
11
(pg. 
1423
-
1427
)
67
Auvray
 
C
Delahaye
 
A
Pflumio
 
F
, et al. 
HOXC4 homeoprotein efficiently expands human hematopoietic stem cells and triggers similar molecular alterations as HOXB4.
Haematologica
2012
, vol. 
97
 
2
(pg. 
168
-
178
)
68
Tsuzuki
 
S
Hong
 
D
Gupta
 
R
Matsuo
 
K
Seto
 
M
Enver
 
T
Isoform-specific potentiation of stem and progenitor cell engraftment by AML1/RUNX1.
PLoS Med
2007
, vol. 
4
 
5
pg. 
e172
 
69
Ran
 
D
Shia
 
WJ
Lo
 
MC
, et al. 
RUNX1a enhances hematopoietic lineage commitment from human embryonic stem cells and inducible pluripotent stem cells.
Blood
2013
, vol. 
121
 
15
(pg. 
2882
-
2890
)
70
Goyama
 
S
Schibler
 
J
Cunningham
 
L
, et al. 
Transcription factor RUNX1 promotes survival of acute myeloid leukemia cells.
J Clin Invest
2013
, vol. 
123
 
9
(pg. 
3876
-
3888
)
71
Challen
 
GA
Goodell
 
MA
Runx1 isoforms show differential expression patterns during hematopoietic development but have similar functional effects in adult hematopoietic stem cells.
Exp Hematol
2010
, vol. 
38
 
5
(pg. 
403
-
416
)
72
Tipping
 
AJ
Pina
 
C
Castor
 
A
, et al. 
High GATA-2 expression inhibits human hematopoietic stem and progenitor cell function by effects on cell cycle.
Blood
2009
, vol. 
113
 
12
(pg. 
2661
-
2672
)
73
Milyavsky
 
M
Gan
 
OI
Trottier
 
M
, et al. 
A distinctive DNA damage response in human hematopoietic stem cells reveals an apoptosis-independent role for p53 in self-renewal.
Cell Stem Cell
2010
, vol. 
7
 
2
(pg. 
186
-
197
)
74
Rizo
 
A
Dontje
 
B
Vellenga
 
E
de Haan
 
G
Schuringa
 
JJ
Long-term maintenance of human hematopoietic stem/progenitor cells by expression of BMI1.
Blood
2008
, vol. 
111
 
5
(pg. 
2621
-
2630
)
75
Rizo
 
A
Olthof
 
S
Han
 
L
Vellenga
 
E
de Haan
 
G
Schuringa
 
JJ
Repression of BMI1 in normal and leukemic human CD34(+) cells impairs self-renewal and induces apoptosis.
Blood
2009
, vol. 
114
 
8
(pg. 
1498
-
1505
)
76
Milhem
 
M
Mahmud
 
N
Lavelle
 
D
, et al. 
Modification of hematopoietic stem cell fate by 5aza 2’deoxycytidine and trichostatin A.
Blood
2004
, vol. 
103
 
11
(pg. 
4102
-
4110
)
77
Araki
 
H
Yoshinaga
 
K
Boccuni
 
P
Zhao
 
Y
Hoffman
 
R
Mahmud
 
N
Chromatin-modifying agents permit human hematopoietic stem cells to undergo multiple cell divisions while retaining their repopulating potential.
Blood
2007
, vol. 
109
 
8
(pg. 
3570
-
3578
)
78
Bug
 
G
Gül
 
H
Schwarz
 
K
, et al. 
Valproic acid stimulates proliferation and self-renewal of hematopoietic stem cells.
Cancer Res
2005
, vol. 
65
 
7
(pg. 
2537
-
2541
)
79
Bissels
 
U
Bosio
 
A
Wagner
 
W
MicroRNAs are shaping the hematopoietic landscape.
Haematologica
2012
, vol. 
97
 
2
(pg. 
160
-
167
)
80
Khalaj
 
M
Tavakkoli
 
M
Stranahan
 
AW
Park
 
CY
Pathogenic microRNA’s in myeloid malignancies.
Front Genet
2014
, vol. 
5
 pg. 
361
 
81
O’Connell
 
RM
Chaudhuri
 
AA
Rao
 
DS
Gibson
 
WS
Balazs
 
AB
Baltimore
 
D
MicroRNAs enriched in hematopoietic stem cells differentially regulate long-term hematopoietic output.
Proc Natl Acad Sci USA
2010
, vol. 
107
 
32
(pg. 
14235
-
14240
)
82
Lechman
 
ER
Gentner
 
B
van Galen
 
P
, et al. 
Attenuation of miR-126 activity expands HSC in vivo without exhaustion.
Cell Stem Cell
2012
, vol. 
11
 
6
(pg. 
799
-
811
)
83
Benveniste
 
P
Serra
 
P
Dervovic
 
D
, et al. 
Notch signals are required for in vitro but not in vivo maintenance of human hematopoietic stem cells and delay the appearance of multipotent progenitors.
Blood
2014
, vol. 
123
 
8
(pg. 
1167
-
1177
)
84
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
 
7
(pg. 
730
-
737
)
85
Nicolini
 
FE
Cashman
 
JD
Hogge
 
DE
Humphries
 
RK
Eaves
 
CJ
NOD/SCID mice engineered to express human IL-3, GM-CSF and Steel factor constitutively mobilize engrafted human progenitors and compromise human stem cell regeneration.
Leukemia
2004
, vol. 
18
 
2
(pg. 
341
-
347
)
86
Sanchez
 
PV
Perry
 
RL
Sarry
 
JE
, et al. 
A robust xenotransplantation model for acute myeloid leukemia.
Leukemia
2009
, vol. 
23
 
11
(pg. 
2109
-
2117
)
87
Chou
 
FS
Griesinger
 
A
Wunderlich
 
M
, et al. 
The thrombopoietin/MPL/Bcl-xL pathway is essential for survival and self-renewal in human preleukemia induced by AML1-ETO.
Blood
2012
, vol. 
120
 
4
(pg. 
709
-
719
)
88
Pulikkan
 
JA
Madera
 
D
Xue
 
L
, et al. 
Thrombopoietin/MPL participates in initiating and maintaining RUNX1-ETO acute myeloid leukemia via PI3K/AKT signaling.
Blood
2012
, vol. 
120
 
4
(pg. 
868
-
879
)
89
Rongvaux
 
A
Willinger
 
T
Martinek
 
J
, et al. 
Development and function of human innate immune cells in a humanized mouse model.
Nat Biotechnol
2014
, vol. 
32
 
4
(pg. 
364
-
372
)
90
Ellegast
 
JM
Saito
 
Y
Flavell
 
RA
, et al. 
 
MISTRG mice support good-risk AML engraftment [abstract]. Blood. 2014;124(21). Abstract 3808
91
Shlush
 
LI
Zandi
 
S
Mitchell
 
A
, et al. 
HALT Pan-Leukemia Gene Panel Consortium
Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia.
Nature
2014
, vol. 
506
 
7488
(pg. 
328
-
333
)
92
Theocharides
 
AP
Dobson
 
SM
Laurenti
 
E
, et al. 
Dominant-negative Ikaros cooperates with BCR-ABL1 to induce human acute myeloid leukemia in xenografts.
Leukemia
2015
 
29(1):177-187
93
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
 
6
(pg. 
483
-
495
)
94
Barabe
 
F
Kennedy
 
JA
Hope
 
KJ
Dick
 
JE
 
Modeling the initiation and progression of human acute leukemia in mice. Science 2007;316(5824):600-604
95
Rizo
 
A
Horton
 
SJ
Olthof
 
S
, et al. 
BMI1 collaborates with BCR-ABL in leukemic transformation of human CD34+ cells.
Blood
2010
, vol. 
116
 
22
(pg. 
4621
-
4630
)
96
Rangarajan
 
A
Hong
 
SJ
Gifford
 
A
Weinberg
 
RA
Species- and cell type-specific requirements for cellular transformation.
Cancer Cell
2004
, vol. 
6
 
2
(pg. 
171
-
183
)
97
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
 
3
(pg. 
1166
-
1173
)
98
Vargaftig
 
J
Taussig
 
DC
Griessinger
 
E
, et al. 
Frequency of leukemic initiating cells does not depend on the xenotransplantation model used.
Leukemia
2012
, vol. 
26
 
4
(pg. 
858
-
860
)
99
Terwijn
 
M
Zeijlemaker
 
W
Kelder
 
A
, et al. 
Leukemic stem cell frequency: a strong biomarker for clinical outcome in acute myeloid leukemia.
PLoS ONE
2014
, vol. 
9
 
9
pg. 
e107587
 
100
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;11(18):6520-6527
101
Eppert
 
K
Takenaka
 
K
Lechman
 
ER
, et al. 
Stem cell gene expression programs influence clinical outcome in human leukemia.
Nat Med
2011
, vol. 
17
 
9
(pg. 
1086
-
1093
)
102
Klco
 
JM
Spencer
 
DH
Miller
 
CA
, et al. 
Functional heterogeneity of genetically defined subclones in acute myeloid leukemia.
Cancer Cell
2014
, vol. 
25
 
3
(pg. 
379
-
392
)
103
Wunderlich
 
M
Mizukawa
 
B
Chou
 
FS
, et al. 
AML cells are differentially sensitive to chemotherapy treatment in a human xenograft model.
Blood
2013
, vol. 
121
 
12
(pg. 
e90
-
e97
)
104
Pearson
 
T
Shultz
 
LD
Miller
 
D
, et al. 
Non-obese diabetic-recombination activating gene-1 (NOD-Rag1 null) interleukin (IL)-2 receptor common gamma chain (IL2r gamma null) null mice: a radioresistant model for human lymphohaematopoietic engraftment.
Clin Exp Immunol
2008
, vol. 
154
 
2
(pg. 
270
-
284
)
105
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
 
10
(pg. 
1777
-
1784
)
106
Taussig
 
DC
Pearce
 
DJ
Simpson
 
C
, et al. 
Hematopoietic stem cells express multiple myeloid markers: implications for the origin and targeted therapy of acute myeloid leukemia.
Blood
2005
, vol. 
106
 
13
(pg. 
4086
-
4092
)
107
van Rhenen
 
A
van Dongen
 
GA
Kelder
 
A
, et al. 
The novel AML stem cell associated antigen CLL-1 aids in discrimination between normal and leukemic stem cells.
Blood
2007
, vol. 
110
 
7
(pg. 
2659
-
2666
)
108
Hosen
 
N
Park
 
CY
Tatsumi
 
N
, et al. 
CD96 is a leukemic stem cell-specific marker in human acute myeloid leukemia.
Proc Natl Acad Sci USA
2007
, vol. 
104
 
26
(pg. 
11008
-
11013
)
109
Taussig
 
DC
Miraki-Moud
 
F
Anjos-Afonso
 
F
, et al. 
Anti-CD38 antibody-mediated clearance of human repopulating cells masks the heterogeneity of leukemia-initiating cells.
Blood
2008
, vol. 
112
 
3
(pg. 
568
-
575
)
110
Taussig
 
DC
Vargaftig
 
J
Miraki-Moud
 
F
, et al. 
Leukemia-initiating cells from some acute myeloid leukemia patients with mutated nucleophosmin reside in the CD34(-) fraction.
Blood
2010
, vol. 
115
 
10
(pg. 
1976
-
1984
)
111
Kikushige
 
Y
Shima
 
T
Takayanagi
 
S
, et al. 
TIM-3 is a promising target to selectively kill acute myeloid leukemia stem cells.
Cell Stem Cell
2010
, vol. 
7
 
6
(pg. 
708
-
717
)
112
Du
 
W
Li
 
XE
Sipple
 
J
Pang
 
Q
Overexpression of IL-3Rα on CD34+CD38- stem cells defines leukemia-initiating cells in Fanconi anemia AML.
Blood
2011
, vol. 
117
 
16
(pg. 
4243
-
4252
)
113
Goardon
 
N
Marchi
 
E
Atzberger
 
A
, et al. 
Coexistence of LMPP-like and GMP-like leukemia stem cells in acute myeloid leukemia.
Cancer Cell
2011
, vol. 
19
 
1
(pg. 
138
-
152
)
114
Sarry
 
JE
Murphy
 
K
Perry
 
R
, et al. 
Human acute myelogenous leukemia stem cells are rare and heterogeneous when assayed in NOD/SCID/IL2Rγc-deficient mice.
J Clin Invest
2011
, vol. 
121
 
1
(pg. 
384
-
395
)
115
Eisterer
 
W
Jiang
 
X
Christ
 
O
, et al. 
Different subsets of primary chronic myeloid leukemia stem cells engraft immunodeficient mice and produce a model of the human disease.
Leukemia
2005
, vol. 
19
 
3
(pg. 
435
-
441
)
116
Herrmann
 
H
Sadovnik
 
I
Cerny-Reiterer
 
S
, et al. 
Dipeptidylpeptidase IV (CD26) defines leukemic stem cells (LSC) in chronic myeloid leukemia.
Blood
2014
, vol. 
123
 
25
(pg. 
3951
-
3962
)
117
Cobaleda
 
C
Gutiérrez-Cianca
 
N
Pérez-Losada
 
J
, et al. 
A primitive hematopoietic cell is the target for the leukemic transformation in human philadelphia-positive acute lymphoblastic leukemia.
Blood
2000
, vol. 
95
 
3
(pg. 
1007
-
1013
)
118
Cox
 
CV
Evely
 
RS
Oakhill
 
A
Pamphilon
 
DH
Goulden
 
NJ
Blair
 
A
Characterization of acute lymphoblastic leukemia progenitor cells.
Blood
2004
, vol. 
104
 
9
(pg. 
2919
-
2925
)
119
Castor
 
A
Nilsson
 
L
Astrand-Grundström
 
I
, et al. 
Distinct patterns of hematopoietic stem cell involvement in acute lymphoblastic leukemia.
Nat Med
2005
, vol. 
11
 
6
(pg. 
630
-
637
)
120
Hong
 
D
Gupta
 
R
Ancliff
 
P
, et al. 
 
Initiating and cancer-propagating cells in TEL-AML1-associated childhood leukemia. Science 2008;319(5861):336-339
121
le Viseur
 
C
Hotfilder
 
M
Bomken
 
S
, et al. 
In childhood acute lymphoblastic leukemia, blasts at different stages of immunophenotypic maturation have stem cell properties.
Cancer Cell
2008
, vol. 
14
 
1
(pg. 
47
-
58
)
122
Cox
 
CV
Martin
 
HM
Kearns
 
PR
Virgo
 
P
Evely
 
RS
Blair
 
A
Characterization of a progenitor cell population in childhood T-cell acute lymphoblastic leukemia.
Blood
2007
, vol. 
109
 
2
(pg. 
674
-
682
)
123
Chiu
 
PP
Jiang
 
H
Dick
 
JE
Leukemia-initiating cells in human T-lymphoblastic leukemia exhibit glucocorticoid resistance.
Blood
2010
, vol. 
116
 
24
(pg. 
5268
-
5279
)
124
Kikushige
 
Y
Ishikawa
 
F
Miyamoto
 
T
, et al. 
Self-renewing hematopoietic stem cell is the primary target in pathogenesis of human chronic lymphocytic leukemia.
Cancer Cell
2011
, vol. 
20
 
2
(pg. 
246
-
259
)
125
Pang
 
WW
Pluvinage
 
JV
Price
 
EA
, et al. 
Hematopoietic stem cell and progenitor cell mechanisms in myelodysplastic syndromes.
Proc Natl Acad Sci USA
2013
, vol. 
110
 
8
(pg. 
3011
-
3016
)
126
Woll
 
PS
Kjällquist
 
U
Chowdhury
 
O
, et al. 
Myelodysplastic syndromes are propagated by rare and distinct human cancer stem cells in vivo.
Cancer Cell
2014
, vol. 
25
 
6
(pg. 
794
-
808
)
127
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
 
10
(pg. 
1167
-
1174
)
128
Majeti
 
R
Chao
 
MP
Alizadeh
 
AA
, et al. 
CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells.
Cell
2009
, vol. 
138
 
2
(pg. 
286
-
299
)
129
Saito
 
Y
Kitamura
 
H
Hijikata
 
A
, et al. 
Identification of therapeutic targets for quiescent, chemotherapy-resistant human leukemia stem cells.
Sci Transl Med
2010
, vol. 
2
 
17
pg. 
ra9
 
130
Jin
 
L
Lee
 
EM
Ramshaw
 
HS
, et al. 
Monoclonal antibody-mediated targeting of CD123, IL-3 receptor alpha chain, eliminates human acute myeloid leukemic stem cells.
Cell Stem Cell
2009
, vol. 
5
 
1
(pg. 
31
-
42
)
131
Askmyr
 
M
Ågerstam
 
H
Hansen
 
N
, et al. 
Selective killing of candidate AML stem cells by antibody targeting of IL1RAP.
Blood
2013
, vol. 
121
 
18
(pg. 
3709
-
3713
)
132
Barreyro
 
L
Will
 
B
Bartholdy
 
B
, et al. 
Overexpression of IL-1 receptor accessory protein in stem and progenitor cells and outcome correlation in AML and MDS.
Blood
2012
, vol. 
120
 
6
(pg. 
1290
-
1298
)
133
Lagadinou
 
ED
Sach
 
A
Callahan
 
K
, et al. 
BCL-2 inhibition targets oxidative phosphorylation and selectively eradicates quiescent human leukemia stem cells.
Cell Stem Cell
2013
, vol. 
12
 
3
(pg. 
329
-
341
)
134
Nilsson
 
L
Astrand-Grundström
 
I
Arvidsson
 
I
, et al. 
Isolation and characterization of hematopoietic progenitor/stem cells in 5q-deleted myelodysplastic syndromes: evidence for involvement at the hematopoietic stem cell level.
Blood
2000
, vol. 
96
 
6
(pg. 
2012
-
2021
)
135
Raaijmakers
 
MH
Myelodysplastic syndromes: revisiting the role of the bone marrow microenvironment in disease pathogenesis.
Int J Hematol
2012
, vol. 
95
 
1
(pg. 
17
-
25
)
136
van Kamp
 
H
Fibbe
 
WE
Jansen
 
RP
, et al. 
Clonal involvement of granulocytes and monocytes, but not of T and B lymphocytes and natural killer cells in patients with myelodysplasia: analysis by X-linked restriction fragment length polymorphisms and polymerase chain reaction of the phosphoglycerate kinase gene.
Blood
1992
, vol. 
80
 
7
(pg. 
1774
-
1780
)
137
Rhyasen
 
GW
Wunderlich
 
M
Tohyama
 
K
Garcia-Manero
 
G
Mulloy
 
JC
Starczynowski
 
DT
An MDS xenograft model utilizing a patient-derived cell line.
Leukemia
2014
, vol. 
28
 
5
(pg. 
1142
-
1145
)
138
Hu
 
Z
Van Rooijen
 
N
Yang
 
YG
Macrophages prevent human red blood cell reconstitution in immunodeficient mice.
Blood
2011
, vol. 
118
 
22
(pg. 
5938
-
5946
)
139
Hu
 
Z
Yang
 
YG
Full reconstitution of human platelets in humanized mice after macrophage depletion.
Blood
2012
, vol. 
120
 
8
(pg. 
1713
-
1716
)
140
Zhao
 
M
Perry
 
JM
Marshall
 
H
, et al. 
Megakaryocytes maintain homeostatic quiescence and promote post-injury regeneration of hematopoietic stem cells.
Nat Med
2014
, vol. 
20
 
11
(pg. 
1321
-
1326
)
141
Bruns
 
I
Lucas
 
D
Pinho
 
S
, et al. 
Megakaryocytes regulate hematopoietic stem cell quiescence through CXCL4 secretion.
Nat Med
2014
, vol. 
20
 
11
(pg. 
1315
-
1320
)
142
Shinohara
 
A
Imai
 
Y
Nakagawa
 
M
Takahashi
 
T
Ichikawa
 
M
Kurokawa
 
M
Intracellular reactive oxygen species mark and influence the megakaryocyte-erythrocyte progenitor fate of common myeloid progenitors.
Stem Cells
2014
, vol. 
32
 
2
(pg. 
548
-
557
)
143
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 USA
1998
, vol. 
95
 
14
(pg. 
8239
-
8244
)
144
Buske
 
C
Feuring-Buske
 
M
Antonchuk
 
J
, et al. 
Overexpression of HOXA10 perturbs human lymphomyelopoiesis in vitro and in vivo.
Blood
2001
, vol. 
97
 
8
(pg. 
2286
-
2292
)
145
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
 
13
(pg. 
4369
-
4376
)
146
Schuringa
 
JJ
Chung
 
KY
Morrone
 
G
Moore
 
MA
Constitutive activation of STAT5A promotes human hematopoietic stem cell self-renewal and erythroid differentiation.
J Exp Med
2004
, vol. 
200
 
5
(pg. 
623
-
635
)
147
Shen
 
SW
Dolnikov
 
A
Passioura
 
T
, et al. 
Mutant N-ras preferentially drives human CD34+ hematopoietic progenitor cells into myeloid differentiation and proliferation both in vitro and in the NOD/SCID mouse.
Exp Hematol
2004
, vol. 
32
 
9
(pg. 
852
-
860
)
148
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
 
10
(pg. 
1794
-
1805
)
149
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
 
3
(pg. 
442
-
448
)
150
Wunderlich
 
M
Krejci
 
O
Wei
 
J
Mulloy
 
JC
Human CD34+ cells expressing the inv(16) fusion protein exhibit a myelomonocytic phenotype with greatly enhanced proliferative ability.
Blood
2006
, vol. 
108
 
5
(pg. 
1690
-
1697
)
151
Kennedy
 
JA
Barabé
 
F
Patterson
 
BJ
, et al. 
Expression of TEL-JAK2 in primary human hematopoietic cells drives erythropoietin-independent erythropoiesis and induces myelofibrosis in vivo.
Proc Natl Acad Sci USA
2006
, vol. 
103
 
45
(pg. 
16930
-
16935
)
152
Chung
 
KY
Morrone
 
G
Schuringa
 
JJ
, et al. 
Enforced expression of NUP98-HOXA9 in human CD34(+) cells enhances stem cell proliferation.
Cancer Res
2006
, vol. 
66
 
24
(pg. 
11781
-
11791
)
153
Chou
 
FS
Wunderlich
 
M
Griesinger
 
A
Mulloy
 
JC
N-Ras(G12D) induces features of stepwise transformation in preleukemic human umbilical cord blood cultures expressing the AML1-ETO fusion gene.
Blood
2011
, vol. 
117
 
7
(pg. 
2237
-
2240
)
154
Tursky
 
ML
Beck
 
D
Thoms
 
JA
, et al. 
Overexpression of ERG in cord blood progenitors promotes expansion and recapitulates molecular signatures of high ERG leukemias [published online ahead of print October 13, 2014].
Leukemia
155
Wichmann
 
C
Quagliano-Lo Coco
 
I
Yildiz
 
O
, et al. 
Activating c-KIT mutations confer oncogenic cooperativity and rescue RUNX1/ETO-induced DNA damage and apoptosis in human primary CD34+ hematopoietic progenitors.
Leukemia
 
2015;29(2):279-289
156
Imren
 
S
Heuser
 
M
Gasparetto
 
M
, et al. 
Modeling de novo leukemogenesis from human cord blood with MN1 and NUP98HOXD13.
Blood
2014
, vol. 
124
 
24
(pg. 
3608
-
3612
)
157
Matsushita
 
H
Yahata
 
T
Sheng
 
Y
, et al. 
Establishment of a humanized APL model via the transplantation of PML-RARA-transduced human common myeloid progenitors into immunodeficient mice.
PLoS ONE
2014
, vol. 
9
 
11
pg. 
e111082
 
Sign in via your Institution