Acute myeloid leukaemia (AML) is thought to be maintained by a small population of leukemic progenitor cells. To define the phenotype of such cells with long-term proliferative capacity in vitro and in vivo, we have used the production of leukemic clonogenic cells (CFU) after 2 to 8 weeks in suspension culture as a measure of these cells in vitro and compared their phenotype with that of cells capable of engrafting nonobese diabetic severe combined immune deficient (NOD/SCID) mice. Leukemic blast peripheral blood cells were evaluated for expression of CD34 and Thy-1 (CD90) antigens. The majority of AML blast cells at diagnosis lacked expression of Thy-1. Most primary CFU-blast and the CFU detected at up to 8 weeks from suspension cultures were CD34+/Thy-1. AML cells that were capable of engrafting NOD/SCID mice were also found to have the CD34+/Thy-1 phenotype. However, significant engraftment was achieved using both CD34+/Thy-1 and CD34 subfractions from one AML M5 patient. These results suggest that while heterogeneity exists between individual patients, the leukemic progenitor cells that are capable of maintaining the disease in vitro and in vivo differ from normal hematopoietic progenitor cells in their lack of expression of Thy-1.

ACUTE MYELOID LEUKEMIA (AML) is thought to arise as the result of an initial genetic change in a primitive hematopoietic progenitor cell which produces a competitive growth advantage for the resulting malignant clone.1,2 Subsequent to transformation some evidence suggests that cells from the malignant clone undergo further differentiation, creating a hierarchy of cells that differ in their expression of cell-surface antigens and may also differ in their proliferative potential.3-5 It has been possible to phenotypically characterize normal primitive hematopoietic cells using cell sorting combined with functional assays and to show that the expression or lack of expression of specific cell-surface antigens correlates with the maturation state of cells within the normal hematopoietic hierarchy.6-11 In this study we have used a fluorescence-activated cell sorting (FACS) strategy to characterize expression of CD34 and Thy-1 on AML cells with long-term proliferative ability in vitro and have compared this phenotype to that of the AML cells which will engraft and proliferate in immunodeficient mice.

In the investigation of AML progenitors, most studies have focused on the ability of cells to produce small “blast” colonies in methylcellulose (CFU-blast), usually comprised of 10 to 100 cells per colony. The phenotype of the clonogenic cell population in AML has been shown to differ in some cases from the majority of AML cells.3-5 This assay and other short-term in vitro assays are likely to detect progenitors with limited self-renewal and proliferative capacities.12,13 For this reason we have used the long-term (4- to 8-week) production of CFU-blast in a serum-free recombinant growth factor–containing suspension culture as a quantitative assay for a cell that is less frequent but has a more extensive proliferative ability than CFU-blast.14 AML progenitors capable of long-term proliferation for ≥4 weeks in this suspension culture assay have a phenotype associated with primitive normal progenitors as they are enriched in fractions selected for CD34+/CD38 and CD34+/CD71, whereas progenitors detected in short-term assays express higher levels of CD38 and CD71.14 

In recent years immune deficient mice have been found to be suitable hosts for the evaluation of both normal and leukemic human hematopoietic cells in vivo.15,16 Engraftment of human AML cells has been achieved in human growth factor–supplemented severe combined immune deficient (SCID) mice with the frequency of the SCID-leukemia initiating cells (SL-IC) estimated to range from 0.2 to 100 per 106 unsorted leukemic cells.16 In some of these patients, the SL-IC activity was exclusively found in the sorted CD34++/38 subfraction.17 Although SCID mice are defective in T- and B-cell function, they display normal macrophage and natural killer (NK) cell activity.18 The nonobese diabetic (NOD)/SCID strain is characterized by a functional deficit in NK cells, absence of circulating complement and defective antigen-presenting cells.19 We have found engraftment of human AML cells to be more reproducible and extensive in NOD/SCID mice as compared with SCID animals.20 Others have compared SCID and NOD/SCID mice as hosts for normal or chronic myeloid leukemia (CML) donor cells, with similar results.21 Therefore, we have chosen detection of engraftment of AML cells in NOD/SCID mice 14 weeks postinjection as evidence for the presence, in CD34 and Thy-1 sorted subpopulations, of AML cells with long-term in vivo proliferative ability.

In the present investigation we have evaluated the expression of CD34 and Thy-1 antigens on the earliest detectable AML progenitor cells. Thy-1 (CD90) is a cell-surface glycoprotein which was originally described as a cell-surface differentiation marker expressed predominately in murine brain and thymus.22-24 In humans Thy-1 is expressed on some early T and B lymphocytes, fibroblasts, and neural cells.25,26 Although still unclear, the postulated functions of Thy-1 include cell-cell recognition events and a role in neural differentiation.26,27 Treatment with antibody against Thy-1 has been reported to decrease the numbers of colony-forming cells (CFCs), particularly those with high proliferative potential (HPP-CFCs), suggesting that Thy-1 can mediate a negative signal which inhibits proliferation of primitive progenitor cells.9 Cross-linking of Thy-1 on murine T lymphocytes results in their activation and cell proliferation.28 In the hematopoietic system Thy-1 is expressed on ∼25% of CD34+ cells29 and on that subfraction which is capable of reconstituting SCID mice.30,31 We have previously shown that when CD34+ cells were sorted on the basis of Thy-1 expression, the majority of cells capable of producing colonies after 5 to 8 weeks in stromal cultures (long-term culture-initiating cells) were derived from the CD34+/Thy-1+ subfraction.29 When used in autologous bone marrow transplants, CD34+/Thy-1+ cells, isolated using high-gradient magnetic separation, are capable of engrafting and differentiating in rhesus macaque hosts.32 Moreover, highly FACS purified CD34+/Thy-1+/Lin peripheral blood cells have been shown to engraft in multiple myeloma patients with orderly maturation of erythroid, myeloid, and megakaryocyte lineages and kinetics only slightly delayed from that achieved with unpurified blood cells.33 Together these results suggest that normal CD34+/Thy-1+ cells have stem cell properties.

To determine whether AML cells have the same pattern of expression of CD34 and Thy-1 as that described for normal hematopoietic progenitors,29 30 we have evaluated the coexpression of CD34 and Thy-1 on AML cells with long-term proliferative ability both in vitro, using the suspension culture assay, and in vivo using sublethally irradiated NOD/SCID mice.

Patient cells.Peripheral blood cells were obtained from patients at diagnosis of AML and in a few cases acute lymphoblastic leukaemia (ALL) after informed consent. Blood cells were Ficoll (Pharmacia Biotech, Uppsala, Sweden) separated to obtain a mononuclear cell population, then frozen in Dulbecco's modified Eagle's medium (DMEM) (StemCell Technologies Inc, Vancouver, BC, Canada) with 30% fetal calf serum (FCS) (StemCell Technologies Inc) and 10% dimethylsulfoxide (DMSO), and stored at −135°C.

AML cell phenotyping and sorting.Thawed AML cells were suspended in HFN (Hanks' medium + 2% FCS and 0.1% sodium azide) at 107 cells/mL. Cells were stained for 30 minutes on ice with monoclonal antibodies 8G12 (anti-CD34)34 that had been directly coupled to the fluorochrome cyanine5-succinimidylester (Cy5) and 5E10 (anti-Thy-1)29 coupled to phycoerythrin (PE) or OKT9 (anti-CD71)35 labeled with fluorescein isothiocyanate (FITC) or CD38 labeled with PE (Becton Dickinson, San Jose, CA). All antibodies were used at 1 μg/mL with the exception of 8G12-Cy5, which was used at 4 μg/mL. Cells were then washed twice in HFN at 4°C, propridium iodide (PI) at 2 μg/mL was added to the cells before the second wash, and the cells were maintained on ice before phenotyping and sorting.

Cells were sorted on a dual laser FACStarplus (Becton Dickinson) on the basis of fluorescence intensity after gating out nonviable cells based on PI uptake. Fractions were sorted into Iscove's Modified Dulbecco's Medium (IMDM) (StemCell Technologies Inc) with 50% FCS in microcentrifuge tubes at 4°C. Sorted fractions were washed, resuspended at known cell concentrations, and used to initiate CFU-blast and suspension culture assays or injected into NOD/SCID mice. Phenotypic analysis was performed on a FACScan (Becton Dickinson) with gating out of nonviable cells based on PI uptake.

Blast colony assays (CFU-blast).FACS-sorted peripheral blood cells, immediately following sorting, or cells removed during media changes after at least 2 weeks in suspension culture and every other week thereafter, were plated in α-methylcellulose culture medium (Methocult M3230, StemCell Technologies Inc) containing the following growth factors: 3 U/mL recombinant human (rh) erythropoietin (StemCell Technologies Inc), 20 ng/mL rh interleukin-3 (rhIL-3; Sandoz, Basel, Switzerland), 20 ng/mL rhIL-6 (Terry Fox Laboratory), 20 ng/mL rh granulocyte colony-stimulating factor (rhG-CSF; Amgen Canada Inc, Mississauga, Ontario, Canada), 20 ng/mL rh granulocyte-macrophage CSF (rhGM-CSF; Sandoz), and 50 ng/mL rhSteel Factor (Terry Fox Laboratory). After 14 days of incubation at 37°C in a 5% CO2 humidified incubator, AML blast clusters (10 to 20 cells) or colonies (>20 cells) were counted under an inverted microscope and the numbers pooled to obtain CFU-blast counts. AML progenitors produced small dispersed colonies compared with colonies derived from normal progenitor cells. There was very little erythroid differentiation and uniform colony morphology was observed within individual patients.

Suspension culture assays.Suspension cultures were initiated with unsorted AML cells or with FACS-sorted AML peripheral blood cells at up to 106 cells/mL in serum-free medium (SFM) consisting of IMDM containing 10 μg/mL insulin, 200 μg/mL transferrin, 2% bovine serum albumin, 0.9% NaHCO3 (StemCell Technologies Inc), and the recombinant growth factors in the concentrations described above for the CFU-blast assay. Cultures were established in 1 mL Corning multiwells (Corning CostarCorp, Cambridge, MA) or Nunclon 4 multiwells (Canadian Life Technologies, Burlington, Ontario) and maintained at 37°C in a 5% CO2 humidified incubator. Cultures were demi-depopulated weekly by removal of half the cells plus media and replacing it with fresh media. Every second week the cells that were removed were washed twice in DMEM and cultured in α-methylcellulose to determine the CFU content of the suspension culture. Suspension cultures were maintained for 8 weeks, or until CFU could not be detected at sampling, then the entire contents of the wells were assessed for CFU content. To allow comparisons between experiments, the proportion of progenitors derived from each sorted fraction at each time point was determined by comparison to a progenitor recovery of 100% from all fractions at that time point.

Transplantation of leukemic cells in to NOD/SCID mice.NOD/SCID mice (Jackson Laboratory, Bar Harbor, ME) were bred and maintained in sterile microisolater cages. Twenty-four hours before transplantation, mice were irradiated with 4 Gy γ irradiation from a 137Cs source at a dose rate of 1.29 cGy/min. Unsorted and AML cells sorted for coexpression of CD34 and Thy-1 antigens were suspended in 0.2 mL Alpha Minimal Essential Medium (StemCell Technologies Inc) with 5% FCS and injected intravenously into the lateral tail vein of NOD/SCID mice. In the two final experiments, 106 irradiated normal human marrow cells, which by themselves never produced detectable CD45+ cells in the mice, were coinjected with sorted subfractions. Six micrograms of human IL-3 and 10 μg human Steel factor were injected subcutaneously 3 times per week and animals were killed at 14 weeks postinjection or when they exhibited clinical signs of the disease (ruffled fur, weight loss, lethargy, limb paralysis). Five animals had to be killed in these conditions at 4 to 9 weeks postinjection. The gross anatomy of each mouse was inspected and tissues were removed for flow cytometry, fluorescence in situ hybridization (FISH), and histologic analysis.

Flow cytometry analysis of murine tissues.Cell suspensions from blood, hind bone marrow, and spleen were incubated on ice with ammonium chloride for 20 minutes, to lyse the red blood cells, and then washed in HFN with 5% human serum. Cells from all three tissue samples were stained with a human pan leukocyte antibody, 9.4 (anti CD45),36 directly coupled to FITC for 30 minutes on ice. Separate aliquots were stained with an irrelevant IgG1 antibody labeled with FITC (Becton Dickinson) as an isotype control. Aliquots of normal human peripheral blood cells and bone marrow cells from a nonirradiated NOD/SCID mouse were also stained with CD45-FITC or the isotype control to serve as positive and negative controls for antibody specificity using methods as described.37 After staining, the cells were washed in HFN containing PI (2 μg/mL) and resuspended in 300 μL HFN. Samples were analyzed using a FACScan (Becton Dickinson), nonviable cells were gated out based on PI uptake, and isotype controls were run for each tissue sample in every experiment and used to set the gates. Cells were defined as positive using gate settings which excluded ≥99% of the cells in the matched isotype control (Fig 1). For further accuracy, the percentage of positive cells in the isotype control (ie, ≤1%) was then subtracted from the percentage positive in samples stained with CD45-FITC. Whenever possible the CD45+ cells derived from murine tissue were sorted and set up in short-term suspension cultures (≤3 days) or plated into methylcellulose with recombinant growth factors. The derived colonies were plucked onto slides and cells from the suspension cultures were transferred directly onto slides for subsequent cytogenetic analysis.

Fig. 1.

Analysis of human cell engraftment in NOD/SCID mice. Murine bone marrow cells (A) and human peripheral blood cells (B) were stained with CD45 as negative and positive controls for antibody specificity. Cell samples were stained with IgG1 as an isotype control (C) and gates were set to exclude ≥99% of cells in the matched isotype control. This gate was then used to determine the proportion of positive cells in the CD45-stained murine samples (D). FSC, forward light scatter.

Fig. 1.

Analysis of human cell engraftment in NOD/SCID mice. Murine bone marrow cells (A) and human peripheral blood cells (B) were stained with CD45 as negative and positive controls for antibody specificity. Cell samples were stained with IgG1 as an isotype control (C) and gates were set to exclude ≥99% of cells in the matched isotype control. This gate was then used to determine the proportion of positive cells in the CD45-stained murine samples (D). FSC, forward light scatter.

Close modal

Cytogenetic analysis.Directly sorted cells, colonies from CFU-blast, or CFU-blast derived from subculture of long-term suspension cultures were evaluated for the leukemia-specific cytogenetic change by either standard cytogenetics or by FISH, whenever possible, in the in vitro studies. Likewise, sorted CD45+ cells from tissue removed from NOD/SCID mice after a brief (≤3 days) culture and/or CFU-blast derived from directly sorted cells were evaluated for the leukemic karyotype. For analysis of CFU-blast, pools of five colonies were plucked and plated onto slides after synchronization of dividing cells in metaphase as previously described.38 In some cases multiple five-colony pools from a plated subfraction were evaluated and the mean presented. Whole chromosome 9 painting probe directly labeled with spectrum green (BRL Life Technologies, Gaithersburg, MD) was used to detect the t(9; 11) in one patient. The inversion (inv) 16 probe directly labeled with digoxigenin (Oncor, Gaithersburg, MD) was used to detect inv 16 in two other patients. The centromere specific chromosome 8 probe directly labeled to digoxigenin (Oncor) was used to identify at least one additional chromosome 8 in six other patients. In one patient the Y probe39 was used to detect a −Y, which was uniformly associated with t(8; 21) in the diagnostic cytogenetics in this patient. Standard G-banded cytogenetics40 was used to detect +13 and del 11q23 in two other patients. The +8 probe and inv 16 probes were hybridized in Hybrisol VI (Oncor) with amplification of the signal by rabbit-antisheep FITC (Jackson Immuno Research Laboratories, West Grove, PA) and detection by sheep-antidigoxigenin-FITC (Boehringer Mannheim, Quebec, Canada). Counterstaining was in PI at 200 ng/mL in Vectashield (Vector Laboratories, Inc, Burlingame, CA). The chromosome 9 probe was hybridized according to the manufacturer's instructions with counterstaining as above. Whenever both metaphase and interphase cells could be scored a mean of 200 cells were counted per 5-colony pool.

Coexpression of CD34 and Thy-1 on circulating acute leukemia cells.Ficoll-separated blood mononuclear cells from 52 AML patients and 11 ALL patients at diagnosis or relapse were analyzed by flow cytometry for coexpression of CD34 and Thy-1, CD71, or CD38 (Table 1). A large proportion of patients in both groups had high expression of CD34 (15% to 100% of cells positive). The mean ± SEM white blood cell count of the AML patients was 88 ± 13 × 109/L with 65% ± 4% of circulating cells being leukemic (blasts by morphology plus monocytes in 8 cases of M5b). Most circulating cells were leukemic in all of the CD34+ groups: CD34 high (15% to 100% CD34+, 63% ± 6% leukemic), CD34 intermediate (5% to 15% CD34+, 64% ± 10% leukemic), CD34 low (1% to 5% CD34+, 52% ± 13% leukemic), and undetectable CD34 (<1% CD34+, 80% ± 4% leukemic). The mean ± SEM white blood cell count of the ALL patients was 72% ± 18 × 109/L with 71% ± 10% being blasts.

Table 1.

Proportion of Acute Leukemia Patients With Cells in CD34 Subset Fractions

% of CellsCD34+CD34+/71CD34+/38CD34+/Thy-1+
in Subfraction(%)(%)(%)(%)
AML patients 
15-100 25/52 (48) 9/48 (19) 3/50 (6) 0/52 (0) 
5-15 7/52 (13) 10/48 (21) 7/50 (14) 1/52 (2) 
1-5 9/52 (17) 8/48 (17) 12/50 (24) 2/52 (4) 
<1 11/52 (21) 21/48 (44) 28/50 (56) 49/52 (94) 
ALL patients 
15-100 8/11 (73) 5/10 (50) 0/11 (0) 2/11 (18) 
5-15 1/11 (9) 1/10 (10) 1/11 (9) 0/11 (0) 
1-5 2/11 (18) 3/10 (30) 9/11 (82) 3/11 (27) 
<1 0 (0) 1/10 (10) 1/11 (9) 6/11 (54) 
% of CellsCD34+CD34+/71CD34+/38CD34+/Thy-1+
in Subfraction(%)(%)(%)(%)
AML patients 
15-100 25/52 (48) 9/48 (19) 3/50 (6) 0/52 (0) 
5-15 7/52 (13) 10/48 (21) 7/50 (14) 1/52 (2) 
1-5 9/52 (17) 8/48 (17) 12/50 (24) 2/52 (4) 
<1 11/52 (21) 21/48 (44) 28/50 (56) 49/52 (94) 
ALL patients 
15-100 8/11 (73) 5/10 (50) 0/11 (0) 2/11 (18) 
5-15 1/11 (9) 1/10 (10) 1/11 (9) 0/11 (0) 
1-5 2/11 (18) 3/10 (30) 9/11 (82) 3/11 (27) 
<1 0 (0) 1/10 (10) 1/11 (9) 6/11 (54) 

The AML patients were French-American-British (FAB) M1 (n = 8), M2 (n = 3), M3 (n = 2), M4 (n = 20), M5 (n = 13), or other (n = 6). Of these patients, although expression of CD34 was high (mean ± SEM, 33% ± 5% CD34+), almost all patients had less than 1% of cells coexpressing CD34 and Thy-1 (mean ± SEM, 0.48% ± 0.24%, CD34+/Thy-1+). This was significantly lower than the number of cells expressing CD34 and lacking expression of CD71 (10.9% ± 2.6%, CD34+/71) or CD38 (3.0% ± 0.8%, CD34+/38) (paired t-test, P = .0002 and .001, respectively). The proportion of CD34+/38 cells was also significantly lower than the CD34+/71 fraction (paired t-test, P = .002). Two ALL patients in this small group of 11 tested had high coexpression of CD34 and Thy-1.

Expression of CD34 and Thy-1 on AML cells with long-term in vitro proliferative ability.Cells from 15 patients were sorted for coexpression of CD34 and Thy-1. The clinical characteristics of these patients are shown in Table 2. Eleven of these patients had a significant number of CD34+ nucleated cells (range, 29% to 92%), while in four patients only a very small proportion of cells were CD34+ (0.1% to 4.2%). Nucleated cells that coexpressed CD34 and Thy-1 represented a very small fraction of cells in the blood at diagnosis in all 15 patients (mean ± SEM, 0.2% ± 0.1%, n = 15) (Fig 2). Similarly, a low fraction of CFU-blast (9% ± 9%, n = 9) and a small proportion of CFU derived from suspension culture at week 2 (0.6% ± 0.5%, n = 13), week 4 (2% ± 2%, n = 10), and weeks 6-8 (1.4% ± 1.4%, n = 7) were CD34+/Thy-1+. The higher proportion of CFU-blast from this fraction compared with other endpoints is the result of the growth of cells from a single patient with 83% of CFU-blast in this fraction. In this patient, the proportion of CFU derived from the CD34+/Thy-1+ fraction in suspension culture decreased with the length of time in culture to 19% at week 6 and 0% by week 8. Unfortunately, the few colonies produced by this CD34+/Thy-1+ fraction could not be analyzed cytogenetically because the patient had no karyotypic abnormality. Excluding this patient, the fraction of CFU-blast with this phenotype was 0.08% ± 0.04%. CD34+/Thy-1 cells represented half of all nucleated cells at diagnosis (50% ± 9%) and the majority of CFU-blast (60% ± 14%) were CD34+/Thy-1. Likewise, the majority of CFU detected in suspension culture at week 2 (62% ± 12%), week 4 (75% ± 11%), and weeks 6-8 (78% ± 10%) were derived from the CD34+/Thy-1 subfraction. The remainder of all nucleated cells, CFU-blast (31% ± 14%), and CFU detected in suspension culture at week 2 (38% ± 12%), week 4 (23% ± 12%), and weeks 6-8 (21% ± 10%) were derived from the CD34 subfraction.

Table 2.

Clinical Characteristics of AML Patients and Cell Growth in Baseline Cultures

Patient No.FABWBC × 109/L% Blasts in BloodCytogenetic ChangeCFU*/105CFU in SC wk 2 (per 105)
ND 43 25 +8 1,800 1,600 
M4 29 83 −Y, t(8; 21) 28 
M5 129 95 +8, del 11q23 ND 15 
M5 47 23 del 11q23 390 860 
M4 71 76 +8 550 75 
M4 143 90 Normal 20 420 
M4 410 35 +13 290 410 
M4e 130 70 inv 16 84 
M5 155 39 t(9; 11) 120 1,060 
10 M5 50 92 +8, +8, t(9; 11) 2,170 530 
11 M5 430 98 ND 14 14 
12 M4 103 42 del 16q 73 10 
13 M4e 65 80 inv 16 14 
14 M4e 19 34 inv 16 770 
15 M2 57 51 +8, t(12; 22), del 20q 2,920 740 
Patient No.FABWBC × 109/L% Blasts in BloodCytogenetic ChangeCFU*/105CFU in SC wk 2 (per 105)
ND 43 25 +8 1,800 1,600 
M4 29 83 −Y, t(8; 21) 28 
M5 129 95 +8, del 11q23 ND 15 
M5 47 23 del 11q23 390 860 
M4 71 76 +8 550 75 
M4 143 90 Normal 20 420 
M4 410 35 +13 290 410 
M4e 130 70 inv 16 84 
M5 155 39 t(9; 11) 120 1,060 
10 M5 50 92 +8, +8, t(9; 11) 2,170 530 
11 M5 430 98 ND 14 14 
12 M4 103 42 del 16q 73 10 
13 M4e 65 80 inv 16 14 
14 M4e 19 34 inv 16 770 
15 M2 57 51 +8, t(12; 22), del 20q 2,920 740 

Abbreviations: ND, not determined; WBC, white blood cells.

CFU*, leukemic clusters (10-20 cells) plus colonies (>20 cells) counted after 14 days in methylcellulose culture per 105 cells plated.

SC wk 2†, CFU-blast in 2-week-old suspension cultures initiated with 105 cells. CFU numbers obtained after 2 weeks were doubled to correct for demi-depopulation at week 1.

Fig. 2.

Proportion of AML peripheral blood cells and progenitors expressing CD34 and Thy-1 antigens from 15 patients.

Fig. 2.

Proportion of AML peripheral blood cells and progenitors expressing CD34 and Thy-1 antigens from 15 patients.

Close modal

In two of the four patients (nos. 11 and 12) with very low numbers of CD34+ cells in their blast population, the majority of CFU after 2 and 4 weeks in culture were derived from the CD34+/Thy-1 subfraction (100% and 62%, respectively), with the proportion of cells with this phenotype increasing almost 300-fold from 0.2% of nucleated cells to 57% of CFU derived from suspension culture at weeks 6-8 in one patient. However, in the two remaining patients (nos. 9 and 10), the majority of CFU-blast (97%) and CFU derived from suspension culture at week 2 (99.6% ± 0.4%) and week 4 (100%) were CD34.

Blood mononuclear cells from 3 of these 15 patients and cells from 5 other patients were sorted on the basis of Thy-1 alone. Sorted cells were used to establish suspension cultures and the proportion of progenitor cells in each fraction was determined. The majority of nucleated cells (99.7% ± 0.1%) and CFU-blast (99.7% ± 0.2%) in all 8 patients lacked expression of Thy-1. The suspension cultures were maintained for up to 4 weeks then assessed for CFU content. As with uncultured cells, the majority of cells that were capable of producing CFU after this culture period (98% ± 1.2%) lacked expression of Thy-1.

Cytogenetic analysis of cultured cells.In experiments with nine patient samples, standard cytogenetic analysis or FISH was performed on unsorted or sorted AML cells or on plucked CFU-blast colonies, established initially or derived from suspension cultures initiated with these cell populations (Table 3). At least 70% (range, 70% to 100%) of unsorted nucleated cells analyzed had the expected chromosomal abnormality, as did Thy-1+ or CD34+/Thy-1+ expressing cells which were sorted directly onto slides (69% to 99% of cells with abnormal karyotype by FISH). Similarly, primary CFU-blast and CFU from suspension culture from both unsorted or CD34+/Thy-1 sorted cells were derived from the leukemic clone (75% ± 10%, and 74% ± 4%, respectively). In contrast, while in five experiments (four patients) most cells from primary CFU-blast derived from Thy-1+ or CD34+/Thy-1+ fractions were also leukemic (83% ± 7%), most cells from CFU originating from suspension culture initiated with these fractions did not have the leukemic karyotype (28% ± 17% positive).

Table 3.

Percentage of Cells Directly Sorted or After In Vitro Assays With the Leukemic Change by FISH or Cytogenetics (no. of colonies analyzed)

Patient No.% Positive% Positive Cells% Positive Cells
Nucleated CellsFrom CFU-blastFrom CFU in SC
Control 
70 83 (20) 96 (30) 
100 ND ND 
82 76 (10) 69 (20) 
ND ND 63 (14) 
73 ND 76 (5) 
ND ND 60 (15) 
10 ND 65 (5) 76 (5) 
14 ND 81 (10) ND 
15 ND 81 (10) 76 (20) 
Thy-1+ cells 
89 ND 6 (20) 
81 ND ND 
CD34+/Thy-1+ 
69 91 (5) ND 
99 ND ND 
72 78 (10) 22 (10) 
ND ND 28 (10) 
75 ND 29 (5) 
15 ND 81 (5) 53 (5) 
CD34+/Thy-1 
ND 74 (10) ND 
ND ND 70 (10) 
ND ND 77 (30) 
10 ND ND 68 (5) 
15 ND 80 (15) 75 (20) 
Patient No.% Positive% Positive Cells% Positive Cells
Nucleated CellsFrom CFU-blastFrom CFU in SC
Control 
70 83 (20) 96 (30) 
100 ND ND 
82 76 (10) 69 (20) 
ND ND 63 (14) 
73 ND 76 (5) 
ND ND 60 (15) 
10 ND 65 (5) 76 (5) 
14 ND 81 (10) ND 
15 ND 81 (10) 76 (20) 
Thy-1+ cells 
89 ND 6 (20) 
81 ND ND 
CD34+/Thy-1+ 
69 91 (5) ND 
99 ND ND 
72 78 (10) 22 (10) 
ND ND 28 (10) 
75 ND 29 (5) 
15 ND 81 (5) 53 (5) 
CD34+/Thy-1 
ND 74 (10) ND 
ND ND 70 (10) 
ND ND 77 (30) 
10 ND ND 68 (5) 
15 ND 80 (15) 75 (20) 

Abbreviations: CFU-blast, leukemic clusters (10-20 cells) plus colonies (>20 cells) plucked from methylcellulose; CFU in SC, CFU-blast derived from suspension cultures; ND, not determined.

In vivo NOD/SCID assay.Unsorted AML cells and cells sorted for coexpression of CD34 and Thy-1 from 7 of the 15 patients analyzed in vitro were also evaluated for their ability to engraft sublethally irradiated NOD/SCID mice (Table 4). Engraftment of human cells in the blood (mean percent engraftment ± SEM, 21 ± 14), bone marrow (24 ± 11), and spleen (23 ± 14) as detected by CD45 antigen expression was achieved using 1 to 5 × 106 unsorted cells from every patient. CD34+/Thy-1+ cells represented a very small fraction of the total nucleated cell population (0.14% ± 0.02%) in these experiments. The number of these cells injected ranged from 400 to 4 × 104, which represented all cells with this phenotype from ∼107 unsorted cells. No significant engraftment was detected using these sorted cells in blood (0.1% ± 0.15%), marrow (0.3% ± 0.2%), or in spleen (0%) of recipient mice. CD34+/Thy-1 cells represented the majority (61% ± 13%) of the total nucleated cell population in five patients (≥106 cells injected). Substantial engraftment (3% to 25%) was detected from one or more tissues in all recipients of the CD34+/Thy-1 fraction with mean engraftment in blood (15% ± 9%), bone marrow (19% ± 11%), and spleen (13% ± 10%), which was not significantly different from that achieved with unsorted cells (paired t-test, blood P = .7, marrow P = .14, spleen P = .24). In two experiments CD34+/Thy-1 cells represented less than 0.5% of the nucleated cells and only 104 and 3 × 104 cells with this phenotype were available for injection into mice. Nevertheless, 80% and 16% engraftment in the mouse marrow was achieved with CD34+/Thy-1 cells from these two patients, respectively. CD34 cells (106 cells injected) from five patients, including one patient (no. 9) where CD34 cells represented 99.7% of the total nucleated cells, failed to engraft significantly in blood (0.6% ± 0.6%), bone marrow (1% ± 1%), or spleen (1% ± 1%). However, greater than 80% engraftment in all these tissues was achieved using 106 CD34 cells from another patient (no. 10), where this fraction represented 99.5% of total peripheral blood nucleated cells.

Table 4.

Percentage of NOD/SCID Mouse Tissue Cells Expressing Human CD45 Antigen

PatientPhenotypeNo. CellsBlood4-150BoneSpleen4-150
No.InjectedMarrow4-150
Unsorted 5 × 106 22 21 
 CD34+/Thy-1+ 6 × 103 
 CD34+/Thy-1 106 25 
 CD34 106 
Unsorted 5 × 106 0.02 0.1 
 CD34+/Thy-1+ 4 × 104 
 CD34+/Thy-1 5 × 106 20 0.2 
 CD34 106 ND ND ND 
Unsorted 5 × 106 0.3 
 Unsorted 106 0.5 
 CD34+/Thy-1+ 104 0.3 
 CD34+/Thy-1 106 0.3 
 CD34 106 
Unsorted 106 3.3 49 
 CD34+/Thy-1+ 2 × 104 0.5 0.2 
 CD34+/Thy-1 106 
 CD34 106 
Unsorted 5 × 106 0.3 
 Unsorted 106 
 CD34+/Thy-1+ 104 
 CD34+/Thy-1 106 0.3 
 CD34 106 ND 
Unsorted 5 × 106 89 92 85 
 Unsorted 106 67 55 64 
 CD34+/Thy-1+ 4 × 102 0.1 
 CD34+/Thy-1 3 × 104 16 
 CD34 106 
10 Unsorted 5 × 106 96 95 94 
 Unsorted 106 44 97 ND 
 CD34+/Thy-1+ 3 × 103 
 CD34+/Thy-1 104 68 80 72 
 CD34 106 84 98 87 
PatientPhenotypeNo. CellsBlood4-150BoneSpleen4-150
No.InjectedMarrow4-150
Unsorted 5 × 106 22 21 
 CD34+/Thy-1+ 6 × 103 
 CD34+/Thy-1 106 25 
 CD34 106 
Unsorted 5 × 106 0.02 0.1 
 CD34+/Thy-1+ 4 × 104 
 CD34+/Thy-1 5 × 106 20 0.2 
 CD34 106 ND ND ND 
Unsorted 5 × 106 0.3 
 Unsorted 106 0.5 
 CD34+/Thy-1+ 104 0.3 
 CD34+/Thy-1 106 0.3 
 CD34 106 
Unsorted 106 3.3 49 
 CD34+/Thy-1+ 2 × 104 0.5 0.2 
 CD34+/Thy-1 106 
 CD34 106 
Unsorted 5 × 106 0.3 
 Unsorted 106 
 CD34+/Thy-1+ 104 
 CD34+/Thy-1 106 0.3 
 CD34 106 ND 
Unsorted 5 × 106 89 92 85 
 Unsorted 106 67 55 64 
 CD34+/Thy-1+ 4 × 102 0.1 
 CD34+/Thy-1 3 × 104 16 
 CD34 106 
10 Unsorted 5 × 106 96 95 94 
 Unsorted 106 44 97 ND 
 CD34+/Thy-1+ 3 × 103 
 CD34+/Thy-1 104 68 80 72 
 CD34 106 84 98 87 
F4-150

The remainder of cells to 100% were CD45.

Cytogenetic analysis of transplanted cells.CD45+ cells were sorted from tissues of NOD/SCID recipients injected with AML cells from five patients (Table 5). In all cases the expected leukemic changes were detected in cells derived from recipients of unsorted and CD34+/Thy-1 fractions. More than 95% of the cells and CFU-blast derived from the blood, bone marrow, and spleen of the recipient of the CD34 subfraction from patient no. 10, who showed a high level of engraftment, were confirmed to have the leukemic karyotype. However, leukemic metaphases could not be detected in the tissues from mice injected with CD34 cells from two other patients (nos. 7 and 9) who had low levels (3% to 5%) of CD45+ cells detected in some tissues.

Table 5.

Percentage of Cells From NOD/SCID Mice With Leukemic Change as Detected by FISH or Cytogenetics (no. of colonies analyzed)

Patient No.Cytogenetic Change% Positive% Positive% Positive
in Bloodin Marrowin Spleen
Control 
del 11q23 ND 435-150 ND 
+13 ND 21 (5) ND 
inv 16 86 (10) 100 (10) ND 
t(9; 11) 60 71 60 
10 +8+8, t(9; 11) 80 (5) 85 (10) ND 
CD34+/Thy-1 
del 11q23 67 100 ND 
+13 22 (5) 40 ND 
inv 16 ND ND 83 (10) 
t(9; 11) 67 73 60 
10 +8+8, t(9; 11) 78 (5) 99 (6) ND 
CD34 
+13 ND ND 
t(9; 11) ND 
10 +8+8, t(9; 11) 96 (20) 97 (20) 99 (20) 
Patient No.Cytogenetic Change% Positive% Positive% Positive
in Bloodin Marrowin Spleen
Control 
del 11q23 ND 435-150 ND 
+13 ND 21 (5) ND 
inv 16 86 (10) 100 (10) ND 
t(9; 11) 60 71 60 
10 +8+8, t(9; 11) 80 (5) 85 (10) ND 
CD34+/Thy-1 
del 11q23 67 100 ND 
+13 22 (5) 40 ND 
inv 16 ND ND 83 (10) 
t(9; 11) 67 73 60 
10 +8+8, t(9; 11) 78 (5) 99 (6) ND 
CD34 
+13 ND ND 
t(9; 11) ND 
10 +8+8, t(9; 11) 96 (20) 97 (20) 99 (20) 
F5-150

Where no number of colonies is indicated, the cells for cytogenetics were derived from CD45+ cells after short-term (≤3 days) suspension culture.

The phenotype of the most primitive normal human hematopoietic progenitors has been difficult to determine because of the lack of available assays to measure cells with long-term multilineage marrow reconstitutive ability. As a surrogate assay for such cells we have used the long-term culture initiating cell (LTC-IC) assay, which measures a cell that produces clonogenic progenitors after more than 5 weeks in coculture with a supportive feeder layer.41 More recently, transplantation of human cells into immunodeficient mice has been used as an in vivo assay for normal human progenitors.15,21 Both the LTC-IC and the mouse assays predict that human cells responsible for both short- and long-term marrow reconstitution will express CD34 and Thy-1 and will lack expression of differentiation antigens.6,29-31,42 Furthermore, transplantation of CD34+ cells into both autologous and a few allogeneic hosts has resulted in prompt and sustained engraftment,43,44 whereas transplantation of CD34 cells into baboons did not produce engraftment.45 Autologous transplantation of further purified CD34+/Thy-1+ cells in humans33 and in rhesus macaques32 has also produced engraftment, suggesting that at least some stem cells are in this subfraction. CD34+ cells are highly heterogeneous both in their function and in their expression of other cell-surface antigens. To compare the phenotype of normal progenitors with marrow repopulating ability with that of malignant progenitors in AML which may maintain the malignant clone in patients, we have investigated the coexpression of CD34 and Thy-1 on leukemic cells with long-term proliferative ability both in vitro and in vivo.

Our findings here indicate that very few nucleated AML cells coexpress both CD34 and Thy-1 antigens, whereas a much higher proportion are CD34+/CD38 or CD34+/CD71. This finding is in agreement with that of Holden et al,42 who found that in 19 of 21 AML patients the CD34+ cells lacked expression of Thy-1 antigen. When leukemic cells from 15 patients were sorted for coexpression of CD34 and Thy-1, the CD34+/Thy-1+ subfraction contributed ≤2% of cells which were capable of producing colonies after 4 to 8 weeks in suspension culture. Similar results were obtained when cells from 8 patients were stained with Thy-1 alone, suggesting that cells with long-term proliferative capacity were not present in CD34/Thy-1+ subfraction either. Of the small number of CFU which were derived from suspension cultures initiated with Thy-1+ or CD34+/Thy-1+ cells, only the minority (28% ± 8%) were found to have the genetic change. In contrast, the majority of cells that were capable of proliferation for at least 4 weeks in suspension culture were derived from the CD34+/Thy-1 subfraction, and the majority of these were cytogenetically abnormal. Although a small number of AML progenitors may be present in the Thy-1+ or CD34+/Thy-1+ subfractions, such a sorting strategy does not markedly enrich for AML progenitors while it does enrich for normal primitive progenitors.33,46 In the present investigation the majority of nucleated cells in 4 patients were CD34. In 2 of these patients greater than 99% of CFU detected for up to 4 weeks in suspension culture were derived from the CD34 subfraction, which is consistent with other reports demonstrating the presence of CFU exclusively in the CD34 subfraction in some patients with AML.47 

The use of immunodeficient mice as an assay for malignant human hematopoietic cells with in vivo repopulating ability has been described by several groups of investigators. Although AML cells from many patients will engraft in SCID animals with a pattern of dissemination that resembles the human disease, the variability of success with which cells from different patients proliferate in these mice and the lack of detectable engraftment in some cases suggests that residual host immunity may be reducing the sensitivity with which malignant progenitors can be detected. In the current investigation, in an attempt to overcome this potential barrier, we have used the NOD/SCID mouse which has, in addition to the SCID defect, a reduction in NK cell activity and a relative deficiency in circulating complement and antigen-presenting cells. Using these animals we have found more consistent and extensive engraftment of unsorted AML cells as compared to that achieved with SCID mice.21 Because previous investigators had suggested that a minimum dose of 106 AML cells were required for engraftment, at least in SCID animals,48 in the latter two experiments we cotransplanted 106 irradiated human marrow cells when the number of cells available from sorted subpopulation was small. Using these strategies, engraftment of 1 to 5 × 106 unsorted AML cells was achieved for all seven patient samples. However, there was still considerable heterogeneity between patients in the degree of engraftment achieved. Nevertheless, high levels of engraftment were achieved using 1 to 3 × 104 CD34+/Thy-1 sorted cells from two patients in blood, bone marrow, and spleen, showing that small numbers of cells with this phenotype can engraft in NOD/SCID animals, at least in some cases. In addition, among sorted AML cells from the remaining five patients, cells that were capable of engrafting NOD/SCID mice as detected 14 weeks after injection were found exclusively in the CD34+/Thy-1 subfraction. The leukemic nature of cells obtained from mice injected with cells from this subfraction and unsorted cells was confirmed by FISH or standard cytogenetics in five patients. In contrast, a mean of 1.3 × 104 CD34+/Thy-1+ cells derived from sorting ∼107 AML cells, achieving an average 860-fold enrichment of cells with this phenotype, did not engraft.

In one patient the CD34 fraction also produced high engraftment and in two others a few (3% to 5%) CD45+ cells were present in some tissues but they lacked the leukemic cytogenetic marker. Engraftment of CD34 cells in SCID mice in one case of AML has been reported previously.47 These findings, together with our results, suggest that some AML patients' cells which do not express CD34 have the ability to divide and repopulate SCID or NOD/SCID mice, unlike normal CD34 cells. Although it is possible that leukemic transformation for these patients occurred in a mature cell that no longer expresses CD34 antigen, in the present study, engraftment was also achieved with the CD34+/Thy-1 subfraction from the same patient. Thus, it is possible that transformation occurred in a CD34+ cell with subsequent clonal evolution resulting in loss of CD34 expression in a subset of cells without loss of the ability to engraft.

Previous reports of engraftment of immune deficient mice with normal mobilized peripheral blood or bone marrow cells have documented that although both Thy-1+ and Thy-1 cells could engraft the thymus, only CD34+/Thy-1+/Lin cells had marrow repopulating ability.46 As few as 100 murine Thy-1lo/Lin/Sca+ marrow cells will engraft and repopulate the marrow of lethally irradiated mice.49 To our knowledge this is the first report of the characterization of Thy-1 antigen expression on primitive progenitors from patients with AML. The data we present here suggest that in most cases AML progenitor cells which have long-term proliferative capacity in vitro and the ability to repopulate sublethally irradiated NOD/SCID mice have the phenotype CD34+/Thy-1. These cells differ from many primitive normal hematopoietic cells in that they do not express the Thy-1 antigen. Some primitive AML cells have also been found to lack CD38 and CD71 antigens, a property they share with primitive normal cells.14,17 Whether AML cells lose Thy-1 subsequent to or upon transformation, a possibility that may have functional significance in view of its postulated role in the inhibition of proliferation of primitive cells,9 or whether transformation occurs in a subset of normal stem cells that are CD34+/CD38/CD71/Thy-1 is unknown. The latter possibility would suggest the existence of such a population among normal stem cell populations which may have unique properties.

To gain further understanding of cells that are responsible for maintenance of the AML clone, we have focused on cells capable of long-term proliferation in vivo or in vitro. The recognition that leukemic cells may be organized in a hierarchy similar to that which exists in normal hematopoiesis has important clinical implications. To treat these diseases effectively it may be necessary to ensure that strategies successful for the elimination of the bulk of AML cells also target the cells responsible for maintaining the leukemic clone. Differences between primitive normal and malignant progenitors, such as the expression of Thy-1 reported here, may allow the development of therapeutic strategies to specifically target AML stem cells.

We thank R. Zapf, B. Gerhard, G. Thornbury, and G. Shaw for excellent technical assistance.

Supported by grants from the National Cancer Institute of Canada with funds from the Terry Fox Run, and from the British Columbia Health Research Foundation. A.B. is a fellow of the Leukaemia Research Fund of Canada.

Address reprint requests to H.J. Sutherland, MD, PhD, Terry Fox Laboratory, 601 W 10th Ave, Vancouver, BC, Canada V5Z 1L3.

1
Fialkow
PJ
Singer
JW
Adamson
JW
Vaidya
K
Dow
LW
Ochs
J
Moohr
JW
Acute nonlymphocytic leukemia: Heterogeneity of stem cell origin.
Blood
57
1981
1068
2
Fialkow
PJ
Singer
JW
Raskind
WH
Adamson
JW
Jacobson
RJ
Bernstein
ID
Dow
LW
Najfeld
V
Veith
R
Clonal development, stem-cell differentiation, and clinical remissions in acute nonlymphocytic leukemia.
N Engl J Med
317
1987
468
3
Kabral
A
Bradstock
KF
Grimsley
P
Favaloro
EJ
Immunophenotype of clonogenic cells in myeloid leukaemia.
Leuk Res
12
1988
51
4
Griffin
JD
Larcom
P
Schlossman
SF
Use of surface markers to identify a subset of acute myelomonocytic leukemia cells with progenitor cell properties.
Blood
62
1983
1300
5
Sabbath
KD
Ball
ED
Larcom
P
Davis
RB
Griffin
JD
Heterogeneity of clonogenic cells in acute myeloblastic leukemia.
J Clin Invest
75
1985
746
6
Sutherland
HJ
Eaves
CJ
Eaves
AC
Dragowska
W
Lansdorp
PM
Characterization and partial purification of human marrow cells capable of initiating long-term hematopoiesis in vitro.
Blood
74
1989
1563
7
Sutherland HJ, Eaves CJ, Eaves AC, Lansdorp PM: Differential expression of antigens on cells that initiate haemopoiesis in long-term human marrow culture, in Knapp W, Dorken B, Gilks WR, Rieber EP, Schmidt RE, Stein H, von dem Borne AEG (eds): Leucocyte Typing IV. White Cell Differentiation Antigens. Oxford, UK, Oxford, 1989, p 910
8
Terstappen
LWMM
Huang
S
Safford
M
Lansdorp
PM
Loken
MR
Sequential generations of hematopoietic colonies derived from single nonlineage-committed CD34+CD38− progenitor cells.
Blood
77
1991
1218
9
Mayani
H
Lansdorp
PM
Thy-1 expression is linked to functional properties of primitive hematopoietic progenitor cells from human umbilical cord blood.
Blood
83
1994
2410
10
Andrews
RG
Singer
JW
Bernstein
ID
Precursors of colony-forming cells in humans can be distinguished from colony-forming cells by expression of the CD33 and CD34 antigens and light scatter properties.
J Exp Med
169
1989
1721
11
Lansdorp
PM
Sutherland
HJ
Eaves
CJ
Selective expression of CD45 isoforms on functional subpopulations of CD34+ hemopoietic cells from human bone marrow.
J Exp Med
172
1990
363
12
Griffin
JD
Lowenberg
B
Clonogenic cells in acute myeloblastic leukemia.
Blood
68
1986
1185
13
Lowenberg
B
Touw
IP
Hematopoietic growth factors and their receptors in acute leukemia.
Blood
81
1993
281
14
Sutherland
HJ
Blair
A
Zapf
RW
Characterization of a hierarchy in human acute myeloid leukemia progenitor cells.
Blood
87
1996
4754
15
Lapidot
T
Pflumio
F
Doedens
M
Murdoch
B
Williams
DE
Dick
JE
Cytokine stimulation of multilineage hematopoiesis from immature human cells engrafted in SCID mice.
Science
255
1992
1137
16
Lapidot
T
Sirard
C
Vormoor
J
Murdoch
B
Hoang
T
Caceres-Cortes
J
Minden
M
Paterson
B
Caligiuri
MA
Dick
JE
A cell initiating human acute myeloid leukaemia after transplantation into SCID mice.
Nature
367
1994
645
17
Bonnet D, Dick JE: The CD34++ CD38− stem cell subfraction is responsible for the initiation of human acute myeloid leukemia in NOD-SCID mice. Exp Hematol 24:1126, 1996 (abstr)
18
Dick
JE
Sirard
C
Pflumio
F
Lapidot
T
Murine models of normal and neoplastic human haematopoiesis.
Cancer Surv
15
1992
161
19
Shultz
LD
Schweitzer
PA
Christianson
SW
Gott
B
Schweitzer
IB
Tennent
B
McKenna
S
Mobraaten
L
Rajan
TV
Greiner
DL
Leiter
EH
Multiple defects in innate and adaptive immunologic function in NOD/LtSz-scid mice.
Immunology
154
1995
180
20
Ailles LE, Moore C, Hogge DE: Retroviral marking and engraftment of primary human AML cells in immunodeficient mice. J Hematother 4:227, 1995 (abstr)
21
Wang JCY, Lapidot T, Cashman JD, Sirard C, Doedens M, Larava P, Addy L, Minden M, Keating A, Eaves AC, Eaves CJ, Dick JE: Engraftment of primitive normal and leukemic hematopoietic cells from chronic phase CML patients in immunodeficient mice. Blood 86:601a, 1995 (abstr, suppl 1)
22
Reif
AE
Allen
JMV
The AKR thymic antigen and its distribution in leukemias and nervous tissues.
J Exp Med
120
1964
413
23
Reif
AE
Allen
JM
Mouse thymic iso-antigens.
Nature
209
1966
521
24
Lansdorp PM: CDw90 cluster workshop report, in Schlossman SF, Boumsell L, Gilks W, Harlan JM, Kishimoto T, Morimoto C, Ritz J, Shaw S, Silverstein R, Springer T, Tedder TF, Todd RF (eds): Leucocyte Typing V. Oxford, UK, Oxford, 1995, p 967
25
Ritter
MA
Sauvage
CA
Delio
D
Human Thy-1 antigen: Cell surface expression on early T and B lymphocytes.
Immunology
49
1983
555
26
Seeger
RC
Danon
YL
Rayner
SA
Hoover
H
Detection of a Thy-1 determinant on human neuroblastoma, glioma, sarcoma, and teratoma cells with a monoclonal antibody.
J Immunol
128
1982
983
27
Tiveron
M-C
Barboni
E
Rivero
FBP
Gormley
AM
Seeley
PJ
Grosveld
F
Morris
R
Selective inhibition of neurite outgrowth on mature astrocytes by Thy-1 glycoprotein.
Nature
355
1992
745
28
Kroczek
RA
Gunter
KC
Seligmann
B
Shevach
EM
Induction of T cell activation by monoclonal anti-Thy-1 antibodies.
J Immunol
136
1986
4379
29
Craig
W
Kay
R
Cutler
RL
Lansdorp
PM
Expression of Thy-1 on human hematopoietic progenitor cells.
J Exp Med
177
1993
1331
30
Baum
CM
Weissman
IL
Tsukamoto
AS
Buckle
AM
Peault
B
Isolation of a candidate human hematopoietic stem-cell population.
Proc Natl Acad Sci USA
89
1992
2804
31
Chen BP, Fraser C, Reading C, Murray L, Uchida N, Galy A, Sasaki D, Tricot G, Jagannath S, Barlogie B, Tsukamoto A, Hoffman R: Cytokine-mobilized peripheral blood CD34+Thy-1+Lin− human hematopoietic stem cells as target cells for transplantation-based gene therapy. Leukemia 9:S17, 1995 (suppl)
32
Donahue
RE
Byrne
ER
Thomas
TE
Kirby
MR
Agricola
BA
Sellers
SE
Gaudernack
G
Karlsson
S
Lansdorp
PM
Transplantation and gene transfer of the human glucocerebrosidase gene into immunoselected primate CD34+Thy-1+ cells.
Blood
88
1996
4166
33
Tricot G, Gazitt Y, Jagannath S, Vesole D, Reading C, Juttner C, Hoffman R, Barlogie B: CD34+Thy+Lin− peripheral blood stem cells (PBSC) effect timely trilineage engraftment in multiple myeloma (MM). Blood 86:293a, 1995 (abstr, suppl 1)
34
Lansdorp PM, Dougherty GJ, Humphries RK: CD34 epitopes, in Knapp W, Dorken B, Gilks WR, Rieber EP, Schmidt RE, Stein H, von dem Borne AEG (eds): Leucocyte Typing IV. White Cell Differentiation Antigens. Oxford, UK, Oxford, 1989, p 826
35
Sutherland
R
Delia
D
Schneider
C
Newman
R
Kemshead
J
Greaves
M
Ubiquitous cell-surface glycoprotein on tumor cells is proliferation-associated receptor for transferrin.
Proc Natl Acad Sci USA
78
1981
4515
36
Cobbold S, Hale G, Waldmann H: Non-lineage, LFA-1 family, and leucocyte common antigens: New and previously defined clusters, in McMichael AJ, Beverley PCL, Cobbold S, Crumpton MJ, Gilks W, Gotch FM, Hogg N, Horton M, Ling N, MacLennan ICM, Mason DY, Milstein C, Spiegelhalter D, Waldmann H (eds): Leucocyte Typing III. White Cell Differentiation Antigens. Oxford, UK, Oxford, 1987, p 788
37
Sirard
C
Lapidot
T
Vormoor
J
Cashman
JD
Doedens
M
Murdoch
B
Jamal
N
Messner
H
Addey
L
Minden
M
Laraya
P
Keating
A
Eaves
A
Lansdorp
PM
Eaves
CJ
Dick
JE
Normal and leukemic SCID-repopulating cells (SRC) co-exist in the bone marrow and peripheral blood from CML patients in chronic phase while leukemic SRC are detected in blast crisis.
Blood
87
1996
1539
38
Fraser
C
Eaves
CJ
Kalousek
DK
Fluorodeoxyuridine synchronization of hemopoietic colonies.
Cancer Genet Cytogenet
24
1987
1
39
Lau
YF
Huang
JC
Dozy
AM
Kan
YW
A rapid screening test for antenatal sex determination.
Lancet
1
1984
14
40
Seabright
M
A rapid banding technique for human chromosomes.
Lancet
2
1971
971
41
Sutherland
HJ
Lansdorp
PM
Henkelman
DH
Eaves
AC
Eaves
CJ
Functional characterization of individual human hematopoietic stem cells cultured at limiting dilution on supportive marrow stromal layers.
Proc Natl Acad Sci USA
87
1990
3584
42
Holden
JT
Geller
RB
Farhi
DC
Holland
HK
Stempora
LL
Phillips
CN
Bray
RA
Characterization of thy-1 (CDw90) expression in CD34+ acute leukemia.
Blood
86
1995
60
43
Shpall
EJ
Jones
RB
Bearman
SI
Franklin
WA
Archer
PG
Curiel
T
Bitter
M
Claman
HN
Stemmer
SM
Purdy
M
Myers
SE
Hami
L
Taffs
S
Heimfeld
S
Hallagan
J
Berenson
RJ
Transplantation of enriched CD34-positive autologous marrow into breast cancer patients following high-dose chemotherapy: Influence of CD34-positive peripheral blood progenitors and growth factors on engraftment.
J Clin Oncol
12
1994
28
44
Link
H
Arseniev
L
Bähre
O
Kadar
JG
Diedrich
H
Poliwoda
H
Transplantation of allogeneic CD34+ blood cells.
Blood
87
1996
4903
45
Berenson
RJ
Andrews
RG
Bensinger
WI
Kalamasz
D
Knitter
G
Buckner
CD
Bernstein
ID
Antigen CD34+ marrow cells engraft lethally irradiated baboons.
J Clin Invest
81
1988
951
46
Murray
L
Chen
B
Galy
A
Chen
S
Tushinski
R
Uchida
N
Negrin
R
Tricot
G
Jagannath
S
Vesole
D
Barlogie
B
Hoffman
R
Tsukamoto
A
Enrichment of human hematopoietic stem cell activity in the CD34+Thy-1+Lin− subpopulation from mobilized peripheral blood.
Blood
85
1995
368
47
Terpstra
W
Prins
A
Ploemacher
RE
Wognum
BW
Wagemaker
G
Lowenberg
B
Wielenga
JJ
Long-term leukemia-initiating capacity of a CD34− subpopulation of acute myeloid leukemia.
Blood
87
1996
2187
48
Terpstra
W
Prins
A
Visser
T
Wognum
B
Wagemaker
G
Lowenberg
B
Wielenga
J
Conditions for engraftment of human acute myeloid leukemia (AML) in SCID mice.
Leukemia
9
1995
1573
49
Uchida
N
Aguila
HL
Fleming
WH
Jerabek
L
Weissman
IL
Rapid and sustained hematopoietic recovery in lethally irradiated mice transplanted with purified Thy-1.1loLin−Sca-1+ hematopoietic stem cells.
Blood
83
1994
3758
Sign in via your Institution