Skip to main content
Proceedings of the National Academy of Sciences of the United States of America logoLink to Proceedings of the National Academy of Sciences of the United States of America
. 2011 Mar 7;108(12):5009–5014. doi: 10.1073/pnas.1100551108

Prospective separation of normal and leukemic stem cells based on differential expression of TIM3, a human acute myeloid leukemia stem cell marker

Max Jan a, Mark P Chao a, Adriel C Cha a, Ash A Alizadeh a,b,c,d, Andrew J Gentles e, Irving L Weissman a,1, Ravindra Majeti a,b,c,1
PMCID: PMC3064328  PMID: 21383193

Abstract

Hematopoietic tissues in acute myeloid leukemia (AML) patients contain both leukemia stem cells (LSC) and residual normal hematopoietic stem cells (HSC). The ability to prospectively separate residual HSC from LSC would enable important scientific and clinical investigation including the possibility of purged autologous hematopoietic cell transplants. We report here the identification of TIM3 as an AML stem cell surface marker more highly expressed on multiple specimens of AML LSC than on normal bone marrow HSC. TIM3 expression was detected in all cytogenetic subgroups of AML, but was significantly higher in AML-associated with core binding factor translocations or mutations in CEBPA. By assessing engraftment in NOD/SCID/IL2Rγ-null mice, we determined that HSC function resides predominantly in the TIM3-negative fraction of normal bone marrow, whereas LSC function from multiple AML specimens resides predominantly in the TIM3-positive compartment. Significantly, differential TIM3 expression enabled the prospective separation of HSC from LSC in the majority of AML specimens with detectable residual HSC function.


Acute myeloid leukemia (AML) is an aggressive malignancy of the bone marrow with a 5-y overall survival between 30% and 40%, and much poorer outcomes for patients over age 65 (1, 2). Cytogenetic abnormalities are prognostic in AML (3, 4), although the majority of cases have a normal karyotype (NKAML). Among NKAML patients, numerous prognostic molecular mutations have been identified (5), most commonly internal tandem duplications in the FLT3 tyrosine kinase (FLT3-ITD), which confer a poor prognosis, and cytoplasmic mislocalizing mutations in NPM1 (NPM1c), which confer a better prognosis (6).

According to the cancer stem cell model, tumors are organized as cellular hierarchies initiated and maintained by a small pool of self-renewing cancer stem cells (CSC) (7, 8). The first in vivo demonstration of this model was achieved in human AML, where leukemia-initiating potential was identified exclusively in the CD34+CD38 leukemic subpopulation (9). Notably, recent experiments suggest that leukemia stem cell (LSC) activity may exist in additional subpopulations in some cases (10, 11). Normal hematopoietic stem cells (HSC) also reside within the LinCD34+CD38 compartment and are contained in the subpopulation expressing CD90 (12, 13).

Hematopoietic tissues in AML patients contain both LSC and residual HSC, as indicated by the recovery of normal hematopoietic cells in most patients treated with induction chemotherapy (1). Experimental evidence of the presence of both of these stem cell populations is demonstrated by the observation that normal lymphoid-myeloid hematopoiesis can occasionally be detected upon transplantation of AML samples into immunodeficient mice (10).

The ability to prospectively separate residual HSC from AML LSC would facilitate important clinical and scientific investigation. Clinically, prospective separation raises the possibility of purged autologous hematopoietic cell transplants as part of AML therapy. Scientifically, we have demonstrated that preleukemic mutations occur in self-renewing HSC (1416). Thus, it is possible that residual functionally normal HSC in leukemia patients are preleukemic, and the ability to prospectively isolate these cells provides a unique opportunity to investigate early events in leukemogenesis.

Recently, a number of investigators have reported the identification of cell surface proteins preferentially expressed on AML LSC compared with normal HSC, including CD123 (17), CD44 (18), CLL-1 (19), CD25 (20), CD32 (20), CD96 (21), and CD47 (22, 23). Monoclonal antibodies targeting several of these antigens have shown promise in preclinical models and are in active clinical development (18, 2325). LSC surface markers may be used to monitor minimal residual disease by flow cytometry. Indeed, increased expression of CLL-1 within the LinCD34+CD38 Lin compartment can predict relapse in AML patients in remission (19). Thus far, prospective separation of HSC from LSC has been reported by using LSC marker expression (23, 26) and in CD34 AML (27), but not from a diverse collection of patient samples.

Here, we report the identification of a unique AML stem cell surface marker, T-cell Ig mucin-3 (TIM3). We determined that most HSC function resides in the TIM3-negative fraction of normal bone marrow (NBM), whereas most LSC function from multiple AML specimens was observed in the TIM3-positive compartment. Significantly, differential expression of TIM3 enabled the prospective separation of HSC from LSC in multiple primary human AML samples.

Results

TIM3 Is More Highly Expressed on AML LSC Than on NBM HSC.

Using our previously published gene expression data, we identified increased TIM3 expression in primary human AML LSC compared with NBM HSC (28). Cell surface expression of TIM3 protein in AML was investigated by flow cytometry. TIM3 was more highly expressed on multiple specimens of LinCD34+CD38CD90 AML LSC compared with LinCD34+CD38CD90+ NBM HSC; however, TIM3 expression was not significantly different between bulk AML cells and the LSC-enriched fraction (Fig. 1 A and B). Moreover, a greater percentage of LinCD34+CD38 cells expressed TIM3 in multiple specimens of AML than in NBM (Fig. 1C). TIM3 was expressed on 20 of 22 patient specimens examined, which consisted of diverse clinical and molecular subtypes (Fig. 1D).

Fig. 1.

Fig. 1.

TIM3 is more highly expressed on AML LSC than on NBM HSC. (A) Representative flow cytometry histograms indicating TIM3 expression on NBM HSC (LinCD34+CD38CD90+) and AML LSC (LinCD34+CD38CD90). (B) TIM3 protein expression was assessed by flow cytometry for multiple specimens of NBM HSC, primary AML LSC, and bulk AML. Mean fluorescence intensity was normalized for cell size and against lineage-positive cells for comparison between measurements conducted on different days. Normalized mean expression (and range) for each population were as follows: NBM HSC 8.8 (2.3–18.9), AML LSC 130.0 (3.0–488.5), bulk AML 148.1 (3.5–443.7). Using a two-sided Student's t test, differences in mean expression between NBM HSC and AML LSC (P = 0.01) and between NBM HSC and bulk AML (P = 0.01) were statistically significant, whereas the difference in mean expression between AML LSC and bulk AML (P = 0.67) was not statistically significant. (C) The percentage of cells positive for TIM3 expression by flow cytometry within the LinCD34+CD38 compartment of AML and normal bone marrow samples was determined by comparison with isotype control. Mean (and median) percentage of cells positive for TIM3 expression were as follows: NBM 30% (30%), AML 86% (98%). The difference in mean percentage of cells positive for TIM3 expression between NBM and AML (P < 0.0001) was statistically significant using a two-tailed Student's t test. (D) Clinical features of AML patients whose samples were studied here.

Because TIM3 expression was similar on bulk AML cells and the LSC-enriched fraction, bulk AML gene expression data from a previously described cohort of 526 adult AML patients (29) was used to examine TIM3 expression across cytogenetic and molecularly defined subgroups of AML. Across cytogenetic subgroups, TIM3 was more highly expressed in AML associated with core binding factor translocations, t(8;21)(q22;q22) and idt(16), but was also detected in samples from other subtypes (Fig. S1A). In NKAML, no significant associations were identified between TIM3 expression and either FLT3-ITD or NPM1c mutations; however, higher TIM3 expression was detected in the presence of mutations in one or both copies of CEBPA (Fig. S1B).

Most Functional NBM HSC Do Not Express TIM3.

To investigate the self-renewal and differentiation potential of TIM3+ and TIM3 cells in the HSC-enriched LinCD34+CD38 fraction of NBM, TIM3+ and TIM3 cells from four independent NBM specimens were purified by fluorescence-activated cell sorting (FACS) and transplanted into newborn NOD/SCID/IL2Rγnull (NSG) mice (Fig. 2A). In vivo HSC activity was indicated by the presence of both human CD45+CD19+ lymphoid cells and human CD45+CD33+ myeloid cells in the bone marrow 12 wk after transplant (Fig. 2B). When transplanted at equal cell doses, HSC engraftment occurred more frequently in TIM3 (8/12 mice) than TIM3+ subpopulations (2/12 mice) (P = 0.04, Fisher's exact test) (Fig. 2C). In summary, although TIM3+ cells from normal bone marrow have engraftment potential, HSC reside predominantly within the TIM3 compartment.

Fig. 2.

Fig. 2.

Functional NBM HSC and LSC differ in TIM3 expression. (A) TIM3+ and TIM3 fractions of the LinCD34+CD38 compartment from normal human bone marrow were identified by flow cytometry. (B) These cells were purified by two rounds of FACS and transplanted into NSG pups. Twelve weeks later, bone marrow cells were harvested and analyzed by flow cytometry for the presence of human CD45+ leukocyte engraftment (Left) whose lineage was further defined by expression of CD19 on lymphoid cells and CD33 on myeloid cells (Right). (C) Summary of long-term engraftment in NSG mice from normal bone marrow populations. (D) TIM3+ and TIM3 subpopulations were observed in LinCD34+ (SU018) or both LinCD34+CD38+ and LinCD34+CD38 (SU046) AML cells by flow cytometry. (E) Summary of bone marrow engraftment 12 wk after transplantation in NSG mice from multiple AML cases.

Most AML LSC Express TIM3.

LinCD34+ cells from seven AML cases were tested for LSC function, as defined by the ability to transplant long-term leukemic engraftment in NSG mice. In two of these cases, TIM3 cells were not detected, and leukemic engraftment was observed from the single TIM3+ fraction (Fig. 2E). In the remaining five cases, TIM3+ and TIM3 cells were tested for LSC function. When transplanted at equal cell doses, leukemic engraftment was observed more frequently from TIM3+ (10/11 mice) than TIM3 (4/10 mice) cells (P = 0.02, Fisher's exact test) (Table S1). Whereas only the TIM3+ subpopulation transplanted leukemic engraftment from AML cases SU018, SU047, and SU030, both TIM3+ and TIM3 cells transplanted leukemic engraftment from SU028 and the LinCD34+CD38+ fraction of SU046 by transplantation of >80,000 cells. The leukemic engraftment potential of TIM3+ and TIM3 fractions of LinCD34+CD38+ SU046 cells was investigated in further detail. Limiting dilution analysis for leukemic engraftment revealed similar LSC frequencies within TIM3+ and TIM3 subpopulations of SU046 (Table S2). To formally demonstrate LSC function, secondary transplantation experiments were conducted for AML cases SU018 and SU028. All cell populations capable of engrafting primary recipients also initiated secondary xenografts (Table S3). In summary, LSC function was restricted to the TIM3+ fraction of the LinCD34+compartment in five of seven AML cases.

Prospective Separation of Normal HSC From LSC in a Series of AML Patients.

We identified eight primary AML specimens with a high percentage of leukemic cells (mean and median blast counts of 80% and 86%, respectively) that exhibited evidence of residual normal HSC function as assessed by long-term engraftment of lymphoid-myeloid hematopoiesis in NSG mice (n = 6) or HPC function as assessed by morphologically normal erythro-myeloid colony formation in complete methylcellulose (n = 2). Because TIM3 expression differed between functional NBM HSC and AML LSC, we investigated the ability of TIM3 expression to discriminate residual HSC from LSC in the same patient sample.

Within the HSC- and LSC-enriched LinCD34+ population, rare TIM3 and majority TIM3+ populations were detected and FACS-separated in 6 of these 8 AML cases (Figs. 3A and 4A). All HSC engraftment in vivo and most HPC colony formation in vitro was restricted to each TIM3 subpopulation (Figs. 3 B and D and 4B). In AML cases SU043, SU031, and SU014, neither leukemic in vitro colony formation nor in vivo leukemic engraftment was observed, consistent with the previously reported inability of some primary human AML specimens to engraft in immunodeficient mice (30) (Fig. 3 B and D). To further investigate the relationship between these functionally normal hematopoietic cells and their patient-matched leukemias, FACS-purified TIM3 cells and their progeny from in vivo or in vitro assays were assessed for molecular mutations known to be present in each leukemia sample [FLT3-ITD or inv(16)]. In each case, the molecular mutation was detected in the TIM3+ cells but not in the rare TIM3 cells or their progeny (Fig. 3C). In summary, these data are consistent with prospective separation of functionally normal HSC and HPC from leukemic cells in three AML patient samples based on differential expression of TIM3.

Fig. 3.

Fig. 3.

Prospective separation of residual normal HSC from leukemic cells in three AML cases. (A) TIM3 expression was determined on the LinCD34+ or LinCD34+CD38 fraction of each AML sample by flow cytometry. (B) TIM3 and TIM3+ cells were double-sorted to >99% purity by FACS. For AML SU043, these sorted cells were plated in duplicate into complete methylcellulose media and, 14 d later, myeloid colony formation was determined by microscopy. No leukemic blast colonies were observed. For AML SU014 and SU031, sorted cells were transplanted into NSG pups and, 12 wk later, human engraftment in mouse bone marrow was analyzed by flow cytometry as in Fig. 2B. (C) Sorted cell populations, methylcellulose colonies from TIM3 cells, and/or bone marrow cells from mice engrafted with TIM3 cells were assessed by PCR for the presence of the FLT3-ITD mutation for AML SU043 and SU014. For AML SU031, sorted cells or human CD45+ cells purified from engrafted mice were assessed by RT-PCR for the presence of the CBFB-MYH11 fusion transcript produced by inv(16) and human β-actin. (D) Summary of long-term engraftment in NSG mice of subpopulations from multiple AML cases.

Fig. 4.

Fig. 4.

Prospective separation of functional HSC and functional LSC from three AML cases. (A) TIM3 expression was determined on the LinCD34+ or LinCD34+CD38 fractions of each AML sample by flow cytometry. (B) TIM3+ and TIM3 cells were double-sorted to >99% purity by FACS. These sorted cells were transplanted into newborn NSG mice. Twelve weeks after transplantation, bone marrow cells were harvested and analyzed by flow cytometry for the presence of human CD45+ leukocyte engraftment (Left) whose lineage was further defined by expression of CD19 on lymphoid cells and CD33 on myeloid cells (Right). (C) Sorted cell populations and/or bone marrow cells from engrafted mice were assessed by PCR for the presence of the FLT3-ITD mutation for AML SU030 and SU047. For AML SU046, FISH analysis was used to assess the presence of the MLL translocation produced by t(6;11) in FACS-sorted cell populations and human CD45+ cells purified from engrafted mice. Representative normal and abnormal MLL FISH results using a break-apart probe are shown.

In the additional three cases in which LinCD34+TIM3 populations were detected, engraftment studies in vivo identified functional HSC in the TIM3 fraction and LSC activity in the TIM3+ fraction (Fig. 4 A and B). In one of these cases, AML SU030, PCR analysis confirmed that FACS-purified TIM3 cells or their progeny did not contain the FLT3-ITD mutation, unlike FACS-sorted TIM3+ cells (Fig. 4C, Left). In the second of these cases, AML SU047, human CD45+ cells from one mouse engrafted with TIM3+ cells were homozygous for the FLT3-ITD mutation, whereas human CD45+ cells from one mouse engrafted with TIM3 cells were heterozygous for the FLT3-ITD and FLT3-WT alleles (Fig. 4C, Center). These data can be explained either by mixed engraftment of FLT3-WT normal HSC and FLT3-ITD-homozygous LSC or by engraftment of heterozygous FLT3-ITD preleukemic HSC in the TIM3 compartment. In the third case, AML SU046, FISH analysis demonstrated MLL rearragement in 38/40 sorted LinCD34+CD38TIM3+ cells, 13/40 sorted LinCD34+CD38TIM3 cells, and 1/40 human CD45+ cells from one mouse engrafted with TIM3 cells (Fig. 4C, Right). MLL rearrangement-positive cells within the LinCD34+CD38TIM3 compartment may be either leukemic or preleukemic. Therefore, differential TIM3 expression discriminated functional normal HSC from functional LSC in these 3 AML patient samples, permitting the prospective separation of these critical stem cell populations. In total, we identified LinCD34+TIM3 subpopulations mostly lacking patient matched leukemic molecular markers with HSC (n = 5) or HPC (n = 1) function in eight AML cases.

Discussion

We report here the identification of TIM3 as a unique AML stem cell surface marker that is highly expressed on multiple specimens of AML LSC, but not on NBM HSC. TIM3 is a negative regulator of Th1–T-cell immunity (31, 32). TIM3 is also expressed on innate immune cells including dendritic cells, monocytes, and macrophages, where it can synergize with Toll-like receptor signaling via crosslinking with Galectin-9 (33) and mediate phagocytosis of apoptotic cells by binding phosphatidylserine (34). TIM3 expression was detected in all cytogenetic subgroups of AML, but was significantly higher in AML-associated with core binding factor translocations or mutations in CEBPA. By assessing engraftment in NSG mice, we determined that most NBM HSC do not express TIM3, whereas LSC from multiple AML specimens express high levels of TIM3. Finally, TIM3 expression enabled the prospective separation of HSC from leukemic cells in the majority of AML samples with residual normal HSC function.

It is not known why TIM3 is highly expressed in AML. Elevated TIM3 expression may be a direct consequence of the molecular mutations present in AML. Indeed, TIM3 expression was detected in all cytogenetic subgroups of AML, but was significantly higher in AML-associated with core binding factor translocations or mutations in CEBPA (Fig. S1). Mutations in these transcription factors that regulate myeloid differentiation (35) may directly regulate TIM3 transcription or may arrest leukemic cells in a stage of differentiation normally associated with high TIM3 expression.

TIM3 is a candidate for AML LSC-targeted therapeutic monoclonal antibodies, as recently reported (36). The absence of TIM3 expression on functional NBM HSC and residual HSC in AML samples, in contrast to high expression on multiple samples of AML LSC, provides the rationale for developing such antibodies. Recently, we demonstrated that a blocking monoclonal antibody directed against CD47, a protein overexpressed on LSC, eliminated AML LSC by stimulating phagocytosis through Fc receptor-independent mechanisms (23). In this context, anti-TIM3 antibodies able to bind and activate Fc receptors might synergize with anti-CD47 antibodies to induce more effective phagocytic elimination of AML LSC (37).

The ability to prospectively separate HSC from AML LSC is an unmet and important goal. Previous reports of prospective separation of HSC and LSC show promise but have not been successfully demonstrated on a diverse cohort of AML specimens. Aberrant expression of mature hematolymphoid cell markers including CD7, CD19, and CD56 within the LinCD34+CD38 fraction of AML specimens can distinguish malignant marker-positive cells from nonmalignant marker-negative cells (26). However, cell populations with malignant and normal stem cell function have not yet been separated by this scheme. In NPM-mutated AML, which is associated with low CD34 expression, CD34 and CD38 expression can prospectively separate functional NPM–wild-type HSC and functional NPM-mutated LSC in ≈50% of AML cases considered (27). This strategy is specific to CD34-negative AML, which represents a minority of all AML cases. CD47 has been shown to separate functional HSC from LSC in a single AML case (23). In summary, TIM3 is a significant addition to current markers used for HSC and LSC discrimination.

Ultimately, prospective separation may have several clinical applications. First, autologous hematopoietic cell transplantation (AHCT) has been used in the treatment of AML, including investigation of protocols using ex vivo chemotherapy to eradicate residual leukemic cells in the autograft (3840). AHCT has fallen out of mainstream clinical practice because of equivalence to conventional chemotherapy (39, 41). However, purification of LSC-depleted, functionally normal HSC may improve AHCT outcomes and potentially broaden the use of AHCT to patients who do not achieve complete remission. Second, the ability to discriminate HSC and LSC by flow cytometry may enable evaluation of LSC-targeted therapeutics and prediction of relapse based on minimal residual disease monitoring at the level of the LSC (42). This possibility is supported by the demonstration that increased expression of the LSC marker CLL-1 in the CD34+CD38 bone marrow fraction from two AML patients in remission correlated with relapse (19).

Finally, prospective separation of residual functionally normal HSC from LSC based on TIM3 expression provides an opportunity to map the accumulation of mutations leading to AML. Because leukemogenesis involves the multistep accumulation of rare mutations (43), preleukemic mutations likely accumulate in self-renewing HSC (16). The ability to prospectively isolate functionally normal HSC from AML patients could enable the direct identification of preleukemic mutations.

Materials and Methods

Human Samples.

Normal human bone marrow mononuclear cells were purchased from AllCells. Human AML samples were obtained from patients at the Stanford Medical Center with informed consent, according to Institutional Review Board (IRB)-approved protocols (Stanford IRB no. 76935 and 6453).

Animal Care.

All mouse experiments were conducted according to an Institutional Animal Care and Use Committee-approved protocol and in adherence to the National Institutes of Health Guide for the Care and Use of Laboratory Animals.

Flow Cytometry Analysis and Cell Sorting.

A panel of antibodies was used for analysis and sorting of AML LSC (LinCD34+CD38) and HSC (LinCD34+CD38CD90+) as described (23, 28). For AML samples, the lineage consisted of CD3, CD19, and CD20. TIM3 antibody clone 344823 (R&D Systems) was used. Human CD34-positive cells were enriched from NBM by magnetic selection (StemCell Technologies).

Methylcellulose Colony Assay.

Erythro-myeloid colony formation was assayed by culturing hematopoietic cells in complete methylcellulose (Methocult GF+ H4435; Stem Cell Technologies) as described (13). Colony formation was assayed after 14 d in culture by microscopy. Colony types scored were as follows: CFU-GEMM, colony forming unit–granulocyte, erythrocyte, monocyte, megakaryocyte; BFU-E, blast forming unit–erythrocyte; CFU-E, colony forming unit–erythrocyte; and CFU-GM–colony forming unit–granulocyte, monocyte.

NSG Xenotransplantation Assay.

FACS-purified cell populations were transplanted into newborn NSG mice conditioned with 100 rads of irradiation as described (13, 23, 44). After 12 wk, mice were killed and peripheral blood and bone marrow were analyzed for human myeloid engraftment (hCD45+CD33+) and human lymphoid engraftment (hCD45+CD19+) as described (13, 23, 44).

PCR Analysis for inv(16) and FLT3-ITD.

Total RNA was isolated by Rneasy Micro Kit (Qiagen). cDNA was reverse-transcribed with SuperScript III First-Strand Synthesis Kit (Invitrogen). inv(16) was detected by PCR with the following primers: CBFB-C (F) 5′-GGGCTGTCTGGAGTTTGATG-3′ and MYH11-B2 (R) 5′- TCCTCTTCTCCTCATTCTGCTC-3′ as described (45). Genomic DNA was isolated by Gentra Puregene Cell Kit (Qiagen). FLT3-ITD was detected by PCR with the following primers: FLT3 11F (F) 5′-gcaatttaggtatgaaagccagc-3′ and FLT3 12R (R) 5′-CTTTCAGCATTTTGACGGCAACC-3′ as described (23).

FISH Analysis for MLL Translocation.

Slide preparations were analyzed by FISH by using the MLL breakapart probe (Abbott Molecular) specific for the MLL gene locus at chromosomal band 11q23. Briefly, dried, dehydrated specimens, pretreated per the manufacturer's instructions, were denatured (75 °C, 5 min) and hybridized (37 °C, 24 h) by using a VysisHYBrite instrument. Slides were washed with 2× SSC/0.3% Nonidet P-40 at 73 °C for 2 min, counterstained with DAPI, and analyzed with an Olympus BX51 microscope equipped with an 100× oil immersion objective, appropriate fluorescent filters, and CytoVision imaging software (Genetix).

Supplementary Material

Supporting Information

Acknowledgments

We acknowledge Theresa Storm for laboratory management, Feifei Zhao for assistance, and Dana Bangs for FISH expertise. M.J. is supported by the Lucille P. Markey Biomedical Research Fellowship and the National Science Foundation Graduate Research Fellowship. M.C. is supported by The Howard Hughes Medical Institute and the Stanford Cancer Biology Program. R.M. holds a Career Award for Medical Scientists from the Burroughs Wellcome Fund. This research was supported in part by grants from the Ellison Medical Foundation (to I.L.W.) and the Leukemia and Lymphoma Society (to I.L.W.).

Footnotes

Conflict of interest statement: I.L.W. is a Director of Stem Cells, Inc., and is a cofounder of Cellerant, Inc. and Stem Cells, Inc. I.L.W. and R.M. have filed International Patent Application No. PCT/US2009/000224 entitled “Markers of Acute Myeloid Leukemia Stem Cells.”

This article is a PNAS Direct Submission.

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1100551108/-/DCSupplemental.

References

  • 1.Estey E, Döhner H. Acute myeloid leukaemia. Lancet. 2006;368:1894–1907. doi: 10.1016/S0140-6736(06)69780-8. [DOI] [PubMed] [Google Scholar]
  • 2.Löwenberg B, Downing JR, Burnett A. Acute myeloid leukemia. N Engl J Med. 1999;341:1051–1062. doi: 10.1056/NEJM199909303411407. [DOI] [PubMed] [Google Scholar]
  • 3.Byrd JC, et al. Cancer and Leukemia Group B (CALGB 8461) Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse, and overall survival in adult patients with de novo acute myeloid leukemia: Results from Cancer and Leukemia Group B (CALGB 8461) Blood. 2002;100:4325–4336. doi: 10.1182/blood-2002-03-0772. [DOI] [PubMed] [Google Scholar]
  • 4.Grimwade D, et al. The Medical Research Council Adult and Children's Leukaemia Working Parties The importance of diagnostic cytogenetics on outcome in AML: Analysis of 1,612 patients entered into the MRC AML 10 trial. Blood. 1998;92:2322–2333. [PubMed] [Google Scholar]
  • 5.Mrózek K, Marcucci G, Paschka P, Whitman SP, Bloomfield CD. Clinical relevance of mutations and gene-expression changes in adult acute myeloid leukemia with normal cytogenetics: Are we ready for a prognostically prioritized molecular classification? Blood. 2007;109:431–448. doi: 10.1182/blood-2006-06-001149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Schlenk RF, et al. German-Austrian Acute Myeloid Leukemia Study Group Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. N Engl J Med. 2008;358:1909–1918. doi: 10.1056/NEJMoa074306. [DOI] [PubMed] [Google Scholar]
  • 7.Dick JE. Stem cell concepts renew cancer research. Blood. 2008;112:4793–4807. doi: 10.1182/blood-2008-08-077941. [DOI] [PubMed] [Google Scholar]
  • 8.Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105–111. doi: 10.1038/35102167. [DOI] [PubMed] [Google Scholar]
  • 9.Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997;3:730–737. doi: 10.1038/nm0797-730. [DOI] [PubMed] [Google Scholar]
  • 10.Taussig DC, et al. Anti-CD38 antibody-mediated clearance of human repopulating cells masks the heterogeneity of leukemia-initiating cells. Blood. 2008;112:568–575. doi: 10.1182/blood-2007-10-118331. [DOI] [PubMed] [Google Scholar]
  • 11.Taussig DC, et al. Leukemia initiating cells from some acute myeloid leukemia patients with mutated nucleophosmin reside in the CD34- fraction. Blood. 2010;115:1976–1984. doi: 10.1182/blood-2009-02-206565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.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;89:2804–2808. doi: 10.1073/pnas.89.7.2804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Majeti R, Park CY, Weissman IL. Identification of a hierarchy of multipotent hematopoietic progenitors in human cord blood. Cell Stem Cell. 2007;1:635–645. doi: 10.1016/j.stem.2007.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Jamieson CH, et al. Granulocyte-macrophage progenitors as candidate leukemic stem cells in blast-crisis CML. N Engl J Med. 2004;351:657–667. doi: 10.1056/NEJMoa040258. [DOI] [PubMed] [Google Scholar]
  • 15.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 USA. 2000;97:7521–7526. doi: 10.1073/pnas.97.13.7521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Weissman I. Stem cell research: Paths to cancer therapies and regenerative medicine. JAMA. 2005;294:1359–1366. doi: 10.1001/jama.294.11.1359. [DOI] [PubMed] [Google Scholar]
  • 17.Jordan CT, et al. The interleukin-3 receptor alpha chain is a unique marker for human acute myelogenous leukemia stem cells. Leukemia. 2000;14:1777–1784. doi: 10.1038/sj.leu.2401903. [DOI] [PubMed] [Google Scholar]
  • 18.Jin L, Hope KJ, Zhai Q, Smadja-Joffe F, Dick JE. Targeting of CD44 eradicates human acute myeloid leukemic stem cells. Nat Med. 2006;12:1167–1174. doi: 10.1038/nm1483. [DOI] [PubMed] [Google Scholar]
  • 19.van Rhenen A, et al. The novel AML stem cell associated antigen CLL-1 aids in discrimination between normal and leukemic stem cells. Blood. 2007;110:2659–2666. doi: 10.1182/blood-2007-03-083048. [DOI] [PubMed] [Google Scholar]
  • 20.Saito Y, et al. Identification of therapeutic targets for quiescent, chemotherapy-resistant human leukemia stem cells. Sci Transl Med. 2010;2:17ra19. doi: 10.1126/scitranslmed.3000349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hosen N, et al. CD96 is a leukemic stem cell-specific marker in human acute myeloid leukemia. Proc Natl Acad Sci USA. 2007;104:11008–11013. doi: 10.1073/pnas.0704271104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Jaiswal S, et al. CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell. 2009;138:271–285. doi: 10.1016/j.cell.2009.05.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Majeti R, et al. CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell. 2009;138:286–299. doi: 10.1016/j.cell.2009.05.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Jin L, et al. Monoclonal antibody-mediated targeting of CD123, IL-3 receptor alpha chain, eliminates human acute myeloid leukemic stem cells. Cell Stem Cell. 2009;5:31–42. doi: 10.1016/j.stem.2009.04.018. [DOI] [PubMed] [Google Scholar]
  • 25.Zhao X, et al. Targeting C-type lectin-like molecule-1 for antibody-mediated immunotherapy in acute myeloid leukemia. Haematologica. 2010;95:71–78. doi: 10.3324/haematol.2009.009811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.van Rhenen A, et al. Aberrant marker expression patterns on the CD34+CD38- stem cell compartment in acute myeloid leukemia allows to distinguish the malignant from the normal stem cell compartment both at diagnosis and in remission. Leukemia. 2007;21:1700–1707. doi: 10.1038/sj.leu.2404754. [DOI] [PubMed] [Google Scholar]
  • 27.Taussig DC, et al. Leukemia-initiating cells from some acute myeloid leukemia patients with mutated nucleophosmin reside in the CD34(-) fraction. Blood. 2010;115:1976–1984. doi: 10.1182/blood-2009-02-206565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Majeti R, et al. Dysregulated gene expression networks in human acute myelogenous leukemia stem cells. Proc Natl Acad Sci USA. 2009;106:3396–3401. doi: 10.1073/pnas.0900089106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Wouters BJ, et al. Double CEBPA mutations, but not single CEBPA mutations, define a subgroup of acute myeloid leukemia with a distinctive gene expression profile that is uniquely associated with a favorable outcome. Blood. 2009;113:3088–3091. doi: 10.1182/blood-2008-09-179895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Ailles LE, Gerhard B, Hogge DE. Detection and characterization of primitive malignant and normal progenitors in patients with acute myelogenous leukemia using long-term coculture with supportive feeder layers and cytokines. Blood. 1997;90:2555–2564. [PubMed] [Google Scholar]
  • 31.Zhu C, et al. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol. 2005;6:1245–1252. doi: 10.1038/ni1271. [DOI] [PubMed] [Google Scholar]
  • 32.Monney L, et al. Th1-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature. 2002;415:536–541. doi: 10.1038/415536a. [DOI] [PubMed] [Google Scholar]
  • 33.Anderson AC, et al. Promotion of tissue inflammation by the immune receptor Tim-3 expressed on innate immune cells. Science. 2007;318:1141–1143. doi: 10.1126/science.1148536. [DOI] [PubMed] [Google Scholar]
  • 34.Nakayama M, et al. Tim-3 mediates phagocytosis of apoptotic cells and cross-presentation. Blood. 2009;113:3821–3830. doi: 10.1182/blood-2008-10-185884. [DOI] [PubMed] [Google Scholar]
  • 35.Rosenbauer F, Tenen DG. Transcription factors in myeloid development: Balancing differentiation with transformation. Nat Rev Immunol. 2007;7:105–117. doi: 10.1038/nri2024. [DOI] [PubMed] [Google Scholar]
  • 36.Kikushige Y, et al. TIM-3 is a promising target to selectively kill acute myeloid leukemia stem cells. Cell Stem Cell. 2010;7:708–717. doi: 10.1016/j.stem.2010.11.014. [DOI] [PubMed] [Google Scholar]
  • 37.Chao MP, et al. Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma. Cell. 2010;142:699–713. doi: 10.1016/j.cell.2010.07.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Yeager AM, et al. Autologous bone marrow transplantation in patients with acute nonlymphocytic leukemia, using ex vivo marrow treatment with 4-hydroperoxycyclophosphamide. N Engl J Med. 1986;315:141–147. doi: 10.1056/NEJM198607173150301. [DOI] [PubMed] [Google Scholar]
  • 39.Cassileth PA, et al. Chemotherapy compared with autologous or allogeneic bone marrow transplantation in the management of acute myeloid leukemia in first remission. N Engl J Med. 1998;339:1649–1656. doi: 10.1056/NEJM199812033392301. [DOI] [PubMed] [Google Scholar]
  • 40.Gorin NC, et al. European Co-operative Group for Bone Marrow Transplantation (EBMT) Autologous bone marrow transplantation for acute myeloblastic leukemia in Europe: Further evidence of the role of marrow purging by mafosfamide. Leukemia. 1991;5:896–904. [PubMed] [Google Scholar]
  • 41.Breems DA, Löwenberg B. Acute myeloid leukemia and the position of autologous stem cell transplantation. Semin Hematol. 2007;44:259–266. doi: 10.1053/j.seminhematol.2007.08.002. [DOI] [PubMed] [Google Scholar]
  • 42.Freeman SD, Jovanovic JV, Grimwade D. Development of minimal residual disease-directed therapy in acute myeloid leukemia. Semin Oncol. 2008;35:388–400. doi: 10.1053/j.seminoncol.2008.04.009. [DOI] [PubMed] [Google Scholar]
  • 43.Gilliland DG, Jordan CT, Felix CA. The molecular basis of leukemia. Hematology Am Soc Hematol Educ Program. 2004:80–97. doi: 10.1182/asheducation-2004.1.80. [DOI] [PubMed] [Google Scholar]
  • 44.Park CY, Majeti R, Weissman IL. In vivo evaluation of human hematopoiesis through xenotransplantation of purified hematopoietic stem cells from umbilical cord blood. Nat Protoc. 2008;3:1932–1940. doi: 10.1038/nprot.2008.194. [DOI] [PubMed] [Google Scholar]
  • 45.van Dongen JJ, et al. Standardized RT-PCR analysis of fusion gene transcripts from chromosome aberrations in acute leukemia for detection of minimal residual disease. Report of the BIOMED-1 Concerted Action: Investigation of minimal residual disease in acute leukemia. Leukemia. 1999;13:1901–1928. doi: 10.1038/sj.leu.2401592. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supporting Information

Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

RESOURCES