Chronic myeloid leukemia (CML) is characterized by an increased proliferative activity of the leukemic progenitors that produce an elevated number of mature granulocytes. Nevertheless, cell cycle-active agents, even in very high doses, are alone unable to eradicate the leukemic clone, suggesting the presence of a rare subset of quiescent leukemic stem cells. To isolate such cells, we first used Hoechst 33342 and Pyronin Y staining to obtain viable G0 and G1/S/G2/M fractions of CD34+cells by fluorescence-activated cell sorting (FACS) from 6 chronic-phase CML patients’ samples and confirmed the quiescent and cycling status of the 2 fractions by demonstration of expected patterns of Ki-67 and D cyclin expression. Leukemic (Ph+/BCR-ABL+) cells with in vitro progenitor activity and capable of engrafting immunodeficient mice were identified in the directly isolated G0 cells. Single-cell reverse transcriptase-polymerase chain reaction (RT-PCR) analysis showed that many leukemic CD34+ G0cells also expressed BCR-ABL mRNA. CD34+ from 8 CML patients were also labeled with carboxyfluorescein diacetate succinimidyl diester (CFSE) before being cultured (with and without added growth factors) to allow viable cells that had remained quiescent (ie, CFSE+) after 4 days to be retrieved by FACS. Leukemic progenitors were again detected in all quiescent populations isolated by this second strategy, including those exposed to a combination of flt3-ligand, Steel factor, interleukin-3, interleukin-6, and granulocyte colony-stimulating factor. These findings provide the first direct and definitive evidence of a deeply but reversibly quiescent subpopulation of leukemic cells in patients with CML with both in vitro and in vivo stem cell properties.

CHRONIC MYELOID leukemia (CML) is a myeloproliferative disorder that appears after the deregulated clonal expansion and differentiation of a totipotent hematopoietic stem cell.1,2 The cytogenetic hallmark of the disease is the Philadelphia (Ph) chromosome, which forms as a result of a reciprocal translocation between the long arms of chromosomes 9 and 22.3 At the DNA level, the corresponding change is the creation of a novel BCR-ABL fusion gene.4,5 Despite overgrowth of the marrow in CML patients by the Ph+/BCR-ABL+ clone, a residual population of primitive normal (Ph/BCR-ABL) hematopoietic stem cells typically persists (reviewed in Eaves et al2).

In recent years, considerable interest has focused on the observation that Ph+/BCR-ABL+ cells can show a reduced or absent growth factor dependence for their survival.6,7 We have demonstrated that highly purified populations of phenotypically very primitive Ph+/BCR-ABL+ cells obtained from CML patients will, in fact, proliferate in vitro for several weeks in the absence of added growth factors8 and recently showed that this involves an autocrine interleukin-3 (IL-3) and granulocyte colony-stimulating factor (G-CSF) mechanism.9 These observations could explain both the increased proliferative activity exhibited by many types of Ph+/BCR-ABL+progenitors in vivo2,10,11 and the decreased self-renewal exhibited by very primitive Ph+/BCR-ABL+ cells identified as long-term culture-initiating cells (LTC-IC),12,13 because exposure of primitive normal hematopoietic cells to excess concentrations of IL-3 promotes their differentiation as well as stimulating their proliferation.14,15 Constitutive activation of the leukemic progenitors in CML is also consistent with the ability of intensive chemotherapy regimens to induce cytogenetic remissions in chronic-phase patients.16,17 However, such remissions have generally been short-lived and CML is considered incurable by conventional chemotherapy alone. Therefore, it seemed likely that CML patients might possess a rare subpopulation of quiescent Ph+/BCR-ABL+ stem cells that had not been identified by previous strategies used to purify primitive populations from CML samples. To try to isolate such cells, we pursued 2 approaches for obtaining quiescent CD34+ cells. The first of these exploited the dual staining procedure with Hoechst 33342 (Hst) and Pyronin Y (Py) that allows G0 and G1/S/G2/M cells to be separated.18-20 The second exploited the resolving power of 5- (and 6-) carboxyfluorescein diacetate succinimidyl diester (CFSE) staining21 to distinguish undivided and divided cells in short-term cultures of CD34+ cells.

Cells.

Heparinized blood and bone marrow cells were obtained from 8 patients with untreated BCR-ABL+ chronic-phase CML (see Table 1 for details). Samples of normal adult marrow were either from harvests taken for allogeneic marrow transplants or were from cadaveric marrow samples (Northwest Tissue Center, Seattle, WA). Before use, all samples were enriched for CD34+ cells by negative immunomagnetic depletion of lineage marker+ (lin+) cells using StemSep columns (StemCell Technologies, Vancouver, British Columbia, Canada). For the CML samples, antibodies to CD15 and IgE were added to the recommended cocktail to enhance the removal of mature cells.22 All human cell samples were obtained with informed consent according to institutional guidelines.

Table 1.

Clinical Data on CML Patients’ Samples

CML No. Time From DiagnosisWBC (109/L) Marrow Karyotype (at the time the sample used was taken)*Sample Data
Cells LTC-IC Frequency (per 105 cells) % Ph+ LTC-IC (Ph+/Total)
6 days  113  46XX, t(9;9), 25/25  Blood  38 <17%  (0/6) 
2  0 days  540  46XY, t(9;22), 25/25  Marrow  0.1  <14%  (0/7)  
3  16 days 52  46XX, t(9;22), 25/25  Blood  230  100%  (17/17) 
4  0 days  176  46XY, t(9;22), 25/25  Blood  0.4 100%  (4/4)  
5  0 days  652  46XX, t(9;22), 25/25 Blood  391-153 100%  (33/33)  
6  0 days 640  46XY, t(9;22), 30/30  Blood  0.2  100%  (2/2) 
7  0 days  99  46XY, t(9;22), 23/25  Blood 551-153 100%  (14/14)  
   46XY, 2/25 
8  6 years  84  46XX, t(9;22), 16/26 polyploid with Ph+, 4/26  Blood  81-153 100%  (22/22) 
   46XX, 6/26 
CML No. Time From DiagnosisWBC (109/L) Marrow Karyotype (at the time the sample used was taken)*Sample Data
Cells LTC-IC Frequency (per 105 cells) % Ph+ LTC-IC (Ph+/Total)
6 days  113  46XX, t(9;9), 25/25  Blood  38 <17%  (0/6) 
2  0 days  540  46XY, t(9;22), 25/25  Marrow  0.1  <14%  (0/7)  
3  16 days 52  46XX, t(9;22), 25/25  Blood  230  100%  (17/17) 
4  0 days  176  46XY, t(9;22), 25/25  Blood  0.4 100%  (4/4)  
5  0 days  652  46XX, t(9;22), 25/25 Blood  391-153 100%  (33/33)  
6  0 days 640  46XY, t(9;22), 30/30  Blood  0.2  100%  (2/2) 
7  0 days  99  46XY, t(9;22), 23/25  Blood 551-153 100%  (14/14)  
   46XY, 2/25 
8  6 years  84  46XX, t(9;22), 16/26 polyploid with Ph+, 4/26  Blood  81-153 100%  (22/22) 
   46XX, 6/26 
*

Numbers after the karyotype indicate the number of metaphases with that karyotype.

Expressed per 105 light density (<1.077 g/cm3) cells obtained by centrifugation of the sample on ficol-hypaque. In some instances, LTC-IC assays were performed on lin subsets and the values shown then calculated assuming 100% recovery of LTC-IC in the lin fraction.

Based on absence of t(9;9).

F1-153

LTC-IC assays in this case used a 5-week culture on parental M2-10B4 feeders (not engineered to produce human growth factors) before assessment of CFC numbers.

Flow cytometry.

CD34+ cells were fractionated into G0 and G1/S/G2/M fractions according to their staining with Hst and Py.18-20 Briefly, cryopreserved lin+-depleted (lin) cells were thawed and incubated overnight in a serum-free medium (SFM) consisting of Iscove’s medium (StemCell), a serum substitute (BIT; StemCell), 40 μg/mL low-density lipoproteins (Sigma Chemicals, St Louis, MO), 10−4 mol/L 2-mercaptoethanol, 300 ng/mL each of recombinant human Steel factor (SF; Amgen, Thousand Oaks, CA) and Flt3-ligand (FL; Immunex, Seattle, WA), and 60 ng/mL each of recombinant human IL-3 (Novartis, Basel, Switzerland), IL-6 (Cangene, Mississauga, Ontario, Canada), and G-CSF (StemCell) to allow reactivation of RNA synthesis. The following day, the cells were washed once in Hank’s balanced salt solution supplemented with 2% fetal calf serum (HF/2) and then resuspended in HF/2 containing 10 μmol/L Hst (Molecular Probes, Eugene, OR). After 45 minutes at 37°C, Py (Sigma) was added to give a final concentration of 2.5 μg/mL and cells were incubated for an additional 45 minutes. Anti-CD34-fluorescein isothiocyanate (FITC) (8G12-FITC; Dr P. Lansdorp, Terry Fox Laboratory, Vancouver, British Columbia, Canada) was then added at 10 μg/mL for a final 20 minutes. Cells were washed once in HF/2 containing Hst (10 μmol/L) and Py (2.5 μg/mL) and a second time in the same medium containing 1 μg/mL propidium iodide (PI; Sigma). Cells were finally resuspended in HF/2 without PI and kept on ice in the dark until being sorted on a 3 laser FACStar Plus (Becton Dickinson [BD], San Jose, CA). Gates were set to collect Hstlo Pylo (G0) and all remaining (G1/S/G2/M) cells as separate fractions within the PI CD34+ population.23 

Relatively homogeneously CFSE (Molecular Probes)-labeled fractions of CD34+ cells were also obtained and then cultured for 4 days in SFM, with or without the following growth factors: 50 ng/mL thrombopoietin (TPO; Genentech, San Francisco, CA), or FL, SF, IL-3, IL-6, and G-CSF at the concentrations described above and with or without 100 ng/mL colcemid (GIBCO BRL, Burlington, Ontario, Canada), as indicated. At the end of the 4 days, cells were harvested, washed once in HF/2, and washed a second time in HF/2/PI. Cultures containing colcemid were used to establish the range of fluorescence expected for all cells that had not divided during the 4-day interval.21For both sorting strategies, single cells (and defined numbers of cells where indicated) were collected using the automated cell deposition unit of the fluorescence-activated cell sorter (FACS).

Detection of Ki-67 and D cyclins.

To detect intracellular Ki-67, the staining procedure described by Jordan et al24 was followed. Cells were analyzed for expression of cyclins D1, D2, and D3 after fixation and staining with an FITC-conjugated antibody (Ab) (Pharmingen, Mississauga, Ontario, Canada) according to the manufacturer’s directions.

Analysis of BCR-ABL mRNA expression.

Cells were centrifuged and lyzed in guanidinium isothiocyanate solution (5 mol/L GIT, 20 mmol/L 1,4-diothioerythritol [DTT], 25 mmol/L sodium citrate, pH 7.0, 0.5% Sarcosyl) before the application of a 2-step (nested) reverse transcriptase-polymerase chain reaction (RT-PCR) procedure using an initial oligo (dT)-based primer and poly (A) tailing strategy.25,26 After electrophoresis of amplified products, BCR-ABL–specific and actin-specific fragments were detected by Southern blotting and hybridization with BCR-ABL and actin probes.25,26 Blots were exposed for 6 to 16 hours.

In vitro progenitor assays.

Assays for different types of colony-forming cells (CFC) capable of generating pure or mixed colonies of erythroid cells, granulocytes, and macrophages in fetal calf serum (FCS)-containing methylcellulose medium (H4330; StemCell) supplemented with 50 ng/mL SF and 20 ng/mL each of IL-3, IL-6, granulocyte-macrophage colony-stimulating factor (GM-CSF; Novartis), and G-CSF, and 3 U/mL of erythropoietin (StemCell) were performed as previously described.27 Cells were assayed for LTC-IC by measurement of the CFC content of 6-week cocultures containing pre-established, irradiated murine fibroblasts genetically engineered to produce human IL-3, G-CSF, and SF.27,28 

Transplantation and detection of human cells in immunodeficient mice.

Breeding pairs of NOD/LtSz-scid/scid mice with homozygous disruption of their β2 microglobulin genes (NOD/SCID-β2M−/−mice29; kindly provided by Dr L. Schultz, Jackson Laboratory, Bar Harbor, ME) were expanded and maintained in the animal facility of the British Columbia Cancer Research Centre (Vancouver, British Columbia, Canada) in microisolators under defined sterile conditions. Six- to 8-week-old mice were irradiated with 300 cGy of137Cs γ-rays not more than 24 hours before being injected intravenously with human cells. In each case, 106irradiated (1,500 cGy) normal human bone marrow cells (as carriers) were coinjected into each mouse. After 6 weeks, the cells from both femurs and tibias from each mouse were labeled with various antimouse and antihuman antibodies and depleted immunomagnetically of all murine cells using a StemSep column,22 and human CD34CD45RA+ and/or CD71+(total human) and CD34+ (primitive human) cells were specifically identified and sorted using the FACS.26Aliquots of the sorted human CD34 and CD34+ cells were analyzed by RT-PCR for BCR-ABL mRNA as described above.

Statistics.

Error estimates shown on mean values from greater than 2 samples in a group are the SEM.

Direct isolation of G0 leukemic cells from chronic-phase CML patients.

In a first series of experiments, CD34+ cells from normal marrow (n = 3) and CML samples (no. 1 through 6, Table 1) were separated into 2 fractions: 1 containing HstloPylo (G0) cells and 1 containing all other (HstloPy+ and all Hst+) (G1/S/G2/M) cells (Fig 1). In all 9 samples, a relatively discrete population of HstloPylo cells was identified. On average, this population was largest in the 3 normal samples (39% ± 18%), intermediate in the 2 CML samples with predominantly normal (Ph) LTC-IC (21% and 31%), and smallest in the 4 CML samples with predominantly leukemic LTC-IC (4% ± 2%).

Fig. 1.

Representative examples of Hst and Py staining of viable CD34+ cells from normal marrow (left) and from CML patients with predominantly Ph (middle, no. 2) or Ph+ (right, no. 3) LTC-IC.

Fig. 1.

Representative examples of Hst and Py staining of viable CD34+ cells from normal marrow (left) and from CML patients with predominantly Ph (middle, no. 2) or Ph+ (right, no. 3) LTC-IC.

Close modal

To further characterize the cycling status of these 2 subpopulations of CD34+ cells, particularly in the CML samples with no detectable normal elements, their expression of intracellular Ki-67 and D cyclins was investigated. Ki-67 is a nuclear antigen found exclusively in proliferating cells,24,30 and the D cyclins are induced upon mitogenic stimulation of quiescent cells.31 Less than 2% of the HstloPylo cells from all 3 CML samples examined (no. 4 through 6, Table 1) were found to contain detectable levels of Ki-67 or any of the D cyclins, whereas a high level of expression of both Ki-67 and at least 1 of the D cyclins was consistently evident in the majority of the remaining CD34+ cells (Fig 2).

Fig. 2.

Representative examples of G0 and G1/S/G2/M fractions of CD34+cells after staining with anti-Ki-67-FITC/7AAD and anti-cyclins D1, D2, and D3-FITC/PI (CML no. 4).

Fig. 2.

Representative examples of G0 and G1/S/G2/M fractions of CD34+cells after staining with anti-Ki-67-FITC/7AAD and anti-cyclins D1, D2, and D3-FITC/PI (CML no. 4).

Close modal

In vitro assays for progenitor activity showed that most of the CML CFC and many of the LTC-IC were cycling (Hst+ and/or Hstlo Py+). However, as summarized in Table 2, both CML progenitor types could also be detected in the quiescent (HstloPylo) fraction of the CD34+ cells, although proportionately less so than was the case for normal marrow. Cytogenetic analyses32 of the colonies generated in the assays of the cells isolated from the CML samples showed that leukemic (Ph+) progenitors were present in 4 of 5 cases in which analyzable colony metaphases were obtained (Table 2). To determine whether these quiescent cells also produce BCR-ABL transcripts, RT-PCR analyses were performed directly on the freshly isolated cells from 3 patients (all of whom had predominantly Ph+ LTC-IC). BCR-ABL mRNA was detected in aliquots of 10, 100, and 1,000 CD34+ cells from all 3 patients. Analyses of single HstloPylo cells showed 8 of 12 (67%), 6 of 12 (50%), and 6 of 12 (50%) of these (from each of the 3 patients, respectively) to be BCR-ABL+(Fig 3).

Table 2.

Proportion and Genotype of Progenitors in the G0 (HstloPylo) Subpopulation of CD34+ Cells in CML Samples by Comparison to Normal Marrow

Sample Analyzed CFCLTC-IC
% in G0Ph+/total % in G0Ph+/total (% Ph+)
1* <0.5 —  >98  0/13  (<8%)  
2  0.5 1/1   
3  <0.6 —  31  13/13 (100%)  
4  3.82-153 —  59  31/31  (100%)  
1.32-153 —   
6  4.8  1/1  11/11  (100%)  
Normal BM (n = 3) 46 ± 18   93 ± 3 
Sample Analyzed CFCLTC-IC
% in G0Ph+/total % in G0Ph+/total (% Ph+)
1* <0.5 —  >98  0/13  (<8%)  
2  0.5 1/1   
3  <0.6 —  31  13/13 (100%)  
4  3.82-153 —  59  31/31  (100%)  
1.32-153 —   
6  4.8  1/1  11/11  (100%)  
Normal BM (n = 3) 46 ± 18   93 ± 3 

For each sample, G0 cells were isolated and assayed for CFC and LTC-IC and the colonies obtained in these assays were then analyzed cytogenetically to determine the proportion of CFC and LTC-IC that were Ph+. The values shown indicate the percentage of CFC and LTC-IC present in the original CD34+ population that were detected in the G0 fraction. Ph+/total refers to the number of colonies typed as Ph+ and the total number of colonies analyzed in the assays of the G0 cells.

*

CML sample numbers as per Table 1.

No colonies detected. Value shown is the maximum number possible if 1 colony had been generated from the number of cells assayed.

No LTC-IC detected in the aliquots tested either before or after isolation of the G0 subpopulation.

F2-153

No analyzable metaphases obtained.

Fig. 3.

BCR-ABL expression in G0 and G1/S/G2/M CD34+ cells isolated from CML patient samples no. 3 through 5. Controls included 1,000 G1/S/G2/M cells without RT (CON 1) and H2O blank (CON 2).

Fig. 3.

BCR-ABL expression in G0 and G1/S/G2/M CD34+ cells isolated from CML patient samples no. 3 through 5. Controls included 1,000 G1/S/G2/M cells without RT (CON 1) and H2O blank (CON 2).

Close modal

To determine whether the quiescent cells isolated from CML patients could also generate leukemic populations in vivo, 30 sublethally irradiated immunodeficient NOD/SCID-β2M−/− mice were injected either with HstloPyloCD34+ cells (1 to 2 × 105/mouse) or with the remaining CD34+ cells (1 to 20 × 105/mouse) isolated from 3 CML samples (no. 4 through 6, Table 1). When assessed 6 weeks later, all of the mice showed evidence of engraftment with human cells, including CD34+ as well as CD34 phenotypes (see example shown in Fig 4). Both of these subpopulations were then isolated by FACS and examined for the presence of leukemic (BCR-ABL+) cells by RT-PCR. Most mice transplanted with either G0 cells (6/6) or G1/S/G2/M cells (14/15) from 2 of the CML samples were found to contain both CD34 and CD34+ leukemic cells (see example in Fig 5). In the third case (CML no. 5, Table 1), no leukemic cells were detected in the CD34 population of human cells present in 1 of the 2 mice transplanted with G0 cells or in any of the 7 mice transplanted with G1/S/G2/M cells (despite the consistent presence of detectable human actin transcripts). Thus, some normal transplantable human hematopoietic cells must have been present in both the quiescent and cycling populations isolated from this CML sample (no. 5). The known ability of transplantable stem cells from normal human marrow and cord blood to generate predominantly CD34 B-lineage progeny in NOD/SCID mice33-35 may explain the prevalence of normal human CD34 cells in mice containing BCR-ABL+CD34+ cells.

Fig. 4.

Example of human CD34 cells (expressing either CD45RA or CD71) and human CD34+ cells obtained from the marrow of a NOD/SCID-β2M−/−mouse transplanted 6 weeks previously with 105G0 cells from CML no. 5. Cells were first depleted of coexisting mouse cells by immunodepletion as described in Materials and Methods.

Fig. 4.

Example of human CD34 cells (expressing either CD45RA or CD71) and human CD34+ cells obtained from the marrow of a NOD/SCID-β2M−/−mouse transplanted 6 weeks previously with 105G0 cells from CML no. 5. Cells were first depleted of coexisting mouse cells by immunodepletion as described in Materials and Methods.

Close modal
Fig. 5.

Examples of BCR-ABL expression in CD34 and CD34+ cells isolated from the marrow of immunodeficient mice transplanted 6 weeks earlier with G0 or G1/S/G2/M fractions of CD34+ CML cells (CML no. 6 in Table 1). Controls included 1,000 K562 cells (K562+), 1,000 K562 cells without RT (K562), and H2O blank.

Fig. 5.

Examples of BCR-ABL expression in CD34 and CD34+ cells isolated from the marrow of immunodeficient mice transplanted 6 weeks earlier with G0 or G1/S/G2/M fractions of CD34+ CML cells (CML no. 6 in Table 1). Controls included 1,000 K562 cells (K562+), 1,000 K562 cells without RT (K562), and H2O blank.

Close modal
Ability of some leukemic progenitors from CML patients to remain quiescent in vitro for at least 4 days independent of exposure to exogenous growth factors.

In a second series of experiments, the ability of quiescent leukemic CD34+ cells to survive and resist mitogenic activation for at least 4 days under different conditions of growth factor stimulation was compared with that characteristic of normal CD34+cells. Cells were labeled with CFSE, incubated overnight, sorted to obtain a homogeneously CFSE-labeled CD34+ population, and then cultured for another 4 days in SFM containing either no other factors, TPO only, or a combination of FL, SF, IL-3, IL-6, and G-CSF. The cultured cells were then harvested and resorted into 2 fractions according to their proliferative histories, as indicated by their individual levels of persisting CFSE fluorescence (Fig 6). Cultures containing no growth factors were included to favor the selection of quiescent leukemic cells. TPO alone has been previously shown to maintain the viability of primitive normal cells with minimal stimulation of their proliferation36 and was thus expected to maximize the detection of quiescent cells of either genotype. The 5 growth factor combination was included to identify quiescent cells that would be maximally resistant to activation, because these conditions have been found to mitotically stimulate most primitive adult normal hematopoietic cells within this time frame.21,23 

Fig. 6.

Distribution of cells according to their CFSE fluorescence after being cultured for 4 days in the presence or absence of growth factors as described in the text. The unshaded areas indicate the fluorescence distribution of (still viable) cells that had been prevented from dividing by the addition of colcemid to the cultures. The black peaks indicate the cells that had undergone 0 to 7 divisions during the 4-day culture period.

Fig. 6.

Distribution of cells according to their CFSE fluorescence after being cultured for 4 days in the presence or absence of growth factors as described in the text. The unshaded areas indicate the fluorescence distribution of (still viable) cells that had been prevented from dividing by the addition of colcemid to the cultures. The black peaks indicate the cells that had undergone 0 to 7 divisions during the 4-day culture period.

Close modal

The various populations of cells obtained from these cultures were then assayed for the presence of CFC and LTC-IC. As shown in Table 3, some persisting quiescent cells could be identified in all 8 CML samples studied, regardless of the growth factors to which the cells were exposed during the period they were cultured before analysis. Moreover, the CD34+ cells that remained quiescent for 4 days also always included a subset with CFC activity. As expected, the relative proportion of the persisting quiescent cells retrieved from the cultures (initiated with either normal or CML cells) decreased with increasing growth factor supplementation of the medium. This proportion was consistently lower in the cultures initiated with cells from the CML samples. Interestingly, the quiescent cells also showed lower forward light scattering characteristics indicative of a reduced cell size, regardless of their genotype (data not shown).

Table 3.

A Proportion of CD34+ Normal Bone Marrow (NBM) and CML Cells Can Remain Viable and Quiescent for at Least 4 Days When Cultured in the Presence or Absence of Growth Factors

Growth Factors Added Cell Type Assessed % of Input Cells, or CFC, or LTC-IC Remaining Quiescent for 4 Days
NBM CML (% Ph+)
None  Total cells  93 ± 1.4 38 ± 15  (100 ± 0, n = 3)  
 CFC 100 ± 0  7 ± 6  (100, n = 2)  
 LTC-IC 100 ± 0  ND  
TPO  Total cells  42 ± 17 15 ± 6  (84 ± 32, n = 3)  
 CFC  43 ± 22 9 ± 11  (100 ± 0, n = 6)  
 LTC-IC 97 (n = 1)  ND  
FL, SF, IL-3, IL-6, G-CSF Total cells CFC LTC-IC  18 ± 19 1 ± 1 92 (n = 1)  2.0 ± 0.5 1 ± 1 ND    (100, n = 1) 
Growth Factors Added Cell Type Assessed % of Input Cells, or CFC, or LTC-IC Remaining Quiescent for 4 Days
NBM CML (% Ph+)
None  Total cells  93 ± 1.4 38 ± 15  (100 ± 0, n = 3)  
 CFC 100 ± 0  7 ± 6  (100, n = 2)  
 LTC-IC 100 ± 0  ND  
TPO  Total cells  42 ± 17 15 ± 6  (84 ± 32, n = 3)  
 CFC  43 ± 22 9 ± 11  (100 ± 0, n = 6)  
 LTC-IC 97 (n = 1)  ND  
FL, SF, IL-3, IL-6, G-CSF Total cells CFC LTC-IC  18 ± 19 1 ± 1 92 (n = 1)  2.0 ± 0.5 1 ± 1 ND    (100, n = 1) 

Three normal marrow samples (NBM) and different numbers of CML samples (as shown) were assessed (no growth factors, n = 3; +TPO, n = 8; +5 growth factors, n = 2) except as shown. Values shown are the mean ± SEM. Genotyping was not performed (or not successful) on all CML samples assayed.

Abbreviation: ND, no data.

In designing the present study, we first sought a method that would allow the direct isolation from a CD34+ cell population of a viable G0 fraction of cells distinct from those in G1 as well as S/G2/M. For this, we used a nontoxic double staining procedure with Hoechst 33342 and Pyronin Y and multiparameter flow cytometry. This allowed the viable (PI) CD34+ cells to be divided into 2 fractions: 1 containing cells with a 2n DNA content and a low RNA content (Hstlo/Pylo; ie, the G0cells) and 1 containing all of the remaining (G1/S/G2/M) cells.18-20 Analysis of quiescent (G0) cells in other systems has shown that they typically are smaller in size and have reduced numbers of mitochondria, a decreased protein content, and a higher degree of chromatin condensation.37 In addition, expression of certain genes (eg, growth-arrest–specific, gas, genes,38 and statin39) is turned on and expression of others (eg, proliferating cell nuclear antigen,40Ki-67,24,30 and the D cyclins31) may be silenced. Validation of the G0 status of the cells assigned a Hstlo/Pylo phenotype here was provided by demonstrating the specific absence of 2 of the above-noted markers (Ki-67 and the D cyclins) in these cells in addition to noting their reduced size.

Functional characterization of these directly isolated G0cells confirmed that a significant proportion of the CFC and LTC-IC in normal bone marrow are quiescent,10,20,41 indicative of multilevel regulatory mechanisms normally constraining the expansion of primitive hematopoietic cells. Parallel studies of CML cells showed most of the leukemic CFC and LTC-IC to be cycling, as expected.2,10 However, quiescent leukemic CFC and LTC-IC could also be consistently isolated. Both quiescent and cycling populations of leukemic cells were capable of generating leukemic progeny in irradiated immunodeficient mice. Thus, the quiescent status of Ph+ progenitors, like that of their normal counterparts, is reversible both in vitro and in vivo.

Recent studies suggest that growth factor activation may modulate the engraftment capacity of transplantable hematopoietic cells.42 Gothot et al43 have proposed that this may result from early growth factor-induced changes in the homing function(s) of these cells rather than in their differentiated state. If the majority of the leukemic stem cells in patients with CML are proliferating more rapidly than is typical of their normal counterparts, as suggested by LTC-IC analyses,2 then it might be anticipated that the ability of CML stem cells to be transplanted might also be compromised. This would be consistent with the widely encountered difficulty in rapidly obtaining high levels of leukemic (Ph+/BCR-ABL+) cells in NOD/SCID mice.44-46 In the present studies, we found that both G0 and, to a lesser extent, G1/S/G2/M cells were able to engraft mice with leukemic cells. However, a more immunodeficient strain of NOD/SCID mice (also lacking β2-microglobulin29) was used, and limiting dilution analyses of the frequency of engrafting cells were not feasible. Thus, the intriguing question of whether an engraftment endpoint may underestimate the leukemic stem cell population in CML by a bias in favor of those that are quiescent must await resolution from further studies. Similarly, the possibility that some cycling leukemic stem cells contaminating the G0fraction may have contributed to the engraftment seen here cannot be completely ruled out.

The presence in CML patients of a quiescent population of leukemic cells with progenitor activity was also demonstrated using a completely independent approach. In this second strategy, we used CFSE labeling to allow the specific isolation of cells that would remain viable but not divide when cultured under different conditions. These experiments confirmed that primitive CML cells can proliferate in the absence of added growth factors,6,8,9 although a small number will also remain quiescent for at least 4 days, even in the presence of a strong mitogenic stimulus. The fact that such cells subsequently formed colonies in vitro again indicates the transient nature of their arrest in G0.

The mechanisms underlying the control of normal human hematopoietic progenitor cell cycling are known to be complex and to involve the integration of signals from the environment that either promote or block cell cycle progression. Defects in a subset of these mechanisms have been shown to exist in CML progenitors.47-49 However, it is now clear that very primitive normal hematopoietic cells undergo early changes in the types of external stimuli they respond to as well as in the types of responses elicited by a given stimulus.15,50,51 It is thus interesting to note that the chemokines able to inhibit the proliferation of primitive normal but not CML clonogenic cells are not effective in inhibiting the proliferation of more primitive normal progenitors detected as LTC-IC.52 Thus, the increased turnover exhibited by the leukemic LTC-IC isolated from CML patients2 is likely to be explained by another mechanism. One possibility would be the autocrine production of IL-3 that we have recently described to occur in primitive CML cells.9 

However, these findings are not readily reconciled with the existence of a subpopulation of primitive quiescent leukemic cells in patients with CML. As a first approach to investigating an underlying mechanism, we used single-cell RT-PCR to determine whether these cells might not express BCR-ABL. However, in all 3 patients studied, ≥50% of the freshly isolated G0 cells contained BCR-ABL mRNA. Thus, BCR-ABL expression alone is not sufficient to force the rapid transit through G1 of primitive hematopoietic cells. Interestingly, preliminary studies indicate that such G0BCR-ABL+ cells may not contain IL-3 or G-CSF mRNA. At least some of these leukemic G0 cells can, nevertheless, survive and subsequently proliferate even in the absence of further growth factor stimulation (Holyoake et al, manuscript submitted). This raises the possibility that quiescence and a non–growth factor producing phenotype may be linked properties. Investigation of this possibility is the subject of ongoing studies and could provide important clues to potential therapeutic targets.

The authors acknowledge the help of Dr Michael Barnett and the staff in the Leukemia and Bone Marrow Transplant Program as well as the Stem Cell Assay Service for the provision, storage, and preparation of normal bone marrow and CML samples. We also thank Pamela Austin, Chris Laird, and Margaret Hale for technical assistance; Gloria Shaw for the cytogenetic data; Maya Sinclaire for assistance with the animal procedures; Gayle Thornbury and Giovanna Cameron for help with the flow cytometry; Dr Peter Lansdorp (Terry Fox Laboratory); Amgen, Cangene, Genentech, Immunex, Novartis, and StemCell for generous gifts of reagents; and Bernadine Fox for typing the manuscript.

Supported by grants from the National Cancer Institute of Canada (NCIC) with funds from the Canadian Cancer Society and the Terry Fox Run and a grant from Novartis Canada. T.H. holds a United Kingdom Leukaemia Research Fund Senior Lectureship and C.E. is a Terry Fox Cancer Research Scientist of the NCIC.

The publication costs of this article were defrayed in part by page charge payment. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. section 1734 solely to indicate this fact.

1
Raskind
WH
Fialkow
PJ
The use of cell markers in the study of human hematopoietic neoplasia.
Adv Cancer Res
49
1987
127
2
Eaves
AC
Barnett
MJ
Ponchio
L
Cashman
JD
Petzer
A
Eaves
CJ
Differences between normal and CML stem cells: Potential targets for clinical exploitation.
Stem Cells
16
1998
77
3
Rowley
JD
A new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining.
Nature
243
1973
290
4
Groffen
J
Stephenson
JR
Heisterkamp
N
De Klein
A
Bartram
CR
Grosveld
G
Philadelphia chromosomal breakpoints are clustered within a limited region, bcr, on chromosome 22.
Cell
36
1984
93
5
Shtivelman
E
Lifshitz
B
Gale
RP
Roe
BA
Canaani
E
Alternative splicing of RNAs transcribed from the human abl gene and from the bcr-abl fused gene.
Cell
47
1986
277
6
Strife
A
Lambek
C
Wisniewski
D
Wachter
M
Gulati
SC
Clarkson
BD
Discordant maturation as the primary biological defect in chronic myelogenous leukemia.
Cancer Res
48
1988
1035
7
Bedi
A
Zehnbauer
BA
Barber
J
Sharkis
S
Jones
R
Inhibition of apoptosis by BCR-ABL in chronic myeloid leukemia.
Blood
83
1994
2038
8
Maguer-Satta
V
Burl
S
Liu
L
Damen
J
Chahine
H
Krystal
G
Eaves
A
Eaves
C
BCR-ABL accelerates C2-ceramide-induced apoptosis.
Oncogene
16
1998
237
9
Jiang X, Lopez A, Holyoake T, Eaves A, Eaves C: Autocrine production and action of IL-3 and G-CSF in chronic myeloid leukemia. (submitted)
10
Eaves
CJ
Eaves
AC
Cell culture studies in CML
Bailliere’s Clinical Haematology.
Hinton
K
1987
931
Bailliere Tindall, Saunders
London, UK
11
Traycoff
CM
Halstead
B
Rice
S
McMahel
J
Srour
EF
Cornetta
K
Chronic myelogenous leukaemia CD34+ cells exit G0/G1 phases of cell cycle more rapidly than normal marrow CD34+ cells.
Br J Haematol
102
1998
759
12
Udomsakdi
C
Eaves
CJ
Swolin
B
Reid
DS
Barnett
MJ
Eaves
AC
Rapid decline of chronic myeloid leukemic cells in long-term culture due to a defect at the leukemic stem cell level.
Proc Natl Acad Sci USA
89
1992
6192
13
Petzer
AL
Eaves
CJ
Barnett
MJ
Eaves
AC
Selective expansion of primitive normal hematopoietic cells in cytokine-supplemented cultures of purified cells from patients with chronic myeloid leukemia.
Blood
90
1997
64
14
Ponchio
L
Eaves
CJ
Very primitive hematopoietic cells (LTC-IC) in normal adult human blood and marrow show differences in the regulation of their cycling state.
Blood
86
1995
493a
(abstr, suppl 1)
15
Zandstra
PW
Conneally
E
Petzer
AL
Piret
JM
Eaves
CJ
Cytokine manipulation of primitive human hematopoietic cell self-renewal.
Proc Natl Acad Sci USA
94
1997
4698
16
Kantarjian
HM
Vellekoop
L
McCredie
KB
Keating
MJ
Hester
J
Smith
T
Barlogie
B
Trujillo
J
Freireich
EJ
Intensive combination chemotherapy (ROAP 10) and splenectomy in the management of chronic myelogenous leukemia.
J Clin Oncol
3
1985
192
17
Goto
T
Nishikori
M
Arlin
Z
Gee
T
Kempin
S
Burchenal
J
Strife
A
Wisniewski
D
Lambek
C
Little
C
Jhanwar
S
Chaganti
R
Clarkson
B
Growth characteristics of leukemic and normal hematopoietic cells in Ph1+ chronic myelogenous leukemia and effects of intensive treatment.
Blood
59
1982
793
18
Shapiro
HM
Flow cytometric estimation of DNA and RNA content in intact cells stained with Hoechst 33342 and Pyronin Y.
Cytometry
2
1981
143
19
Ladd
AC
Pyatt
R
Gothot
A
Rice
S
McMahel
J
Traycoff
CM
Srour
EF
Orderly process of sequential cytokine stimulation is required for activation and maximal proliferation of primitive human bone marrow CD34+ hematopoietic progenitor cells residing in G0.
Blood
90
1997
658
20
Gothot
A
Pyatt
R
McMahel
J
Rice
S
Srour
EF
Functional heterogeneity of human CD34+ cells isolated in subcompartments of the G0/G1 phase of the cell cycle.
Blood
90
1997
4384
21
Nordon
RE
Ginsberg
SS
Eaves
CJ
High resolution cell division tracking demonstrates the Flt3-ligand-dependence of human marrow CD34+CD38− cell production in vitro.
Br J Haematol
98
1997
528
22
Holyoake
TL
Cameron
G
Horrocks
C
Thomas
T
Eaves
C
Eaves
A
Development of immunomagnetic negative depletion procedures for obtaining enriched populations of primitive cells from CML blood samples or their progeny regenerated in NOD/SCID mice.
Exp Hematol
26
1998
757
(abstr)
23
Petzer
AL
Hogge
DE
Lansdorp
PM
Reid
DS
Eaves
CJ
Self-renewal of primitive human hematopoietic cells (long-term-culture-initiating cells) in vitro and their expansion in defined medium.
Proc Natl Acad Sci USA
93
1996
1470
24
Jordan
CT
Yamasaki
G
Minamoto
D
High-resolution cell cycle analysis of defined phenotypic subsets within primitive human hematopoietic cell populations.
Exp Hematol
24
1996
1347
25
Sauvageau
G
Lansdorp
PM
Eaves
CJ
Hogge
DE
Dragowska
WH
Reid
DS
Largman
C
Lawrence
HJ
Humphries
RK
Differential expression of homeobox genes in functionally distinct CD34+ subpopulations of human bone marrow cells.
Proc Natl Acad Sci USA
91
1994
12223
26
Maguer-Satta
V
Petzer
AL
Eaves
AC
Eaves
CJ
BCR-ABL expression in different subpopulations of functionally characterized Ph+ CD34+ cells from patients with chronic myeloid leukemia.
Blood
88
1996
1796
27
Hogge
DE
Lansdorp
PM
Reid
D
Gerhard
B
Eaves
CJ
Enhanced detection, maintenance and differentiation of primitive human hematopoietic cells in cultures containing murine fibroblasts engineered to produce human Steel factor, interleukin-3 and granulocyte colony-stimulating factor.
Blood
88
1996
3765
28
Petzer
AL
Eaves
CJ
Lansdorp
PM
Ponchio
L
Barnett
MJ
Eaves
AC
Characterization of primitive subpopulations of normal and leukemic cells present in the blood of patients with newly diagnosed as well as established chronic myeloid leukemia.
Blood
88
1996
2162
29
Christianson
SW
Greiner
DL
Hesselton
R
Leif
JH
Wagar
EJ
Schweitzer
IB
Rajan
TV
Gott
B
Roopenian
DC
Shultz
LD
Enhanced human CD4+ T cell engraftment in β2-microglobulin-deficient NOD-scid mice.
J Immunol
158
1997
3578
30
Schluter
C
Duchrow
M
Wohlenberg
C
Becker
MHG
Key
G
Flad
H-D
Gerdes
J
The cell proliferation-associated antigen of antibody Ki-67: A very large, ubiquitous nuclear protein with numerous repeated elements, representing a new kind of cell cycle-maintaining protein.
J Cell Biol
123
1993
513
31
Sherr
CJ
Mammalian G1 cyclins.
Cell
73
1993
1059
32
Fraser
C
Eaves
CJ
Kalousek
DK
Fluorodeoxyuridine synchronization of hemopoietic colonies.
Cancer Genet Cytogenet
24
1987
1
33
Cashman
J
Bockhold
K
Hogge
DE
Eaves
AC
Eaves
CJ
Sustained proliferation, multi-lineage differentiation and maintenance of primitive human haematopoietic cells in NOD/SCID mice transplanted with human cord blood.
Br J Haematol
98
1997
1026
34
Conneally
E
Cashman
J
Petzer
A
Eaves
C
Expansion in vitro of transplantable human cord blood stem cells demonstrated using a quantitative assay of their lympho-myeloid repopulating activity in nonobese diabetic-scid/scid mice.
Proc Natl Acad Sci USA
94
1997
9836
35
Bhatia
M
Wang
JCY
Kapp
U
Bonnet
D
Dick
JE
Purification of primitive human hematopoietic cells capable of repopulating immune-deficient mice.
Proc Natl Acad Sci USA
94
1997
5320
36
Borge
OJ
Ramsfjell
V
Cui
L
Jacobsen
SEW
Ability of early acting cytokines to directly promote survival and suppress apoptosis of human primitive CD34+CD38− bone marrow cells with multilineage potential at the single-cell level: Key role of thrombopoietin.
Blood
90
1997
2282
37
Pellicciari
C
Mangiarotti
R
Bottone
MG
Danova
M
Wang
E
Identification of resting cells by dual-parameter flow cytometry of statin expression and DNA content.
Cytometry
21
1995
329
38
Schneider
C
King
RM
Philipson
L
Genes specifically expressed at growth arrest of mammalian cells.
Cell
54
1988
787
39
Wang
E
A 57,000-mol-wt protein uniquely present in nonproliferating cells and senescent human fibroblasts.
J Cell Biol
100
1985
545
40
Celis
JE
Bravo
R
Larsen
PM
Fey
SJ
Cyclin: A nuclear protein whose level correlates directly with the proliferative state of normal as well as transformed cells.
Leuk Res
8
1984
143
41
Ponchio
L
Conneally
E
Eaves
C
Quantitation of the quiescent fraction of longterm culture-initiating cells (LTC-IC) in normal human blood and marrow and the kinetics of their growth factor-stimulated entry into S-phase in vitro.
Blood
86
1995
3314
42
Habibian
HK
Peters
SO
Hsieh
CC
Wuu
J
Vergilis
K
Grimaldi
CI
Reilly
J
Carlson
JE
Frimberger
AE
Stewart
FM
Quesenberry
PJ
The fluctuating phenotype of the lymphohematopoietic stem cell with cell cycle transit.
J Exp Med
188
1998
393
43
Gothot
A
Van der Loo
JCM
Clapp
W
Srour
EF
Cell cycle-related changes in repopulating capacity of human mobilized peripheral blood CD34+ cells in non-obese diabetic/severe combined immune-deficient mice.
Blood
92
1998
2641
44
Wang
JCY
Lapidot
T
Cashman
JD
Doedens
M
Addy
L
Sutherland
DR
Nayar
R
Laraya
P
Minden
M
Keating
A
Eaves
AC
Eaves
CJ
Dick
JE
High level engraftment of NOD/SCID mice by primitive normal and leukemic hematopoietic cells from patients with chronic myeloid leukemia in chronic phase.
Blood
91
1998
2406
45
Lewis
ID
McDiarmid
LA
Samels
LM
Bik To
L
Hughes
TP
Establishment of a reproducible model of chronic-phase chronic myeloid leukemia in NOD/SCID mice using blood-derived mononuclear or CD34+ cells.
Blood
91
1998
630
46
Dazzi
F
Capelli
D
Hasserjian
R
Cotter
F
Corbo
M
Poletti
A
Chinswangwatanakul
W
Goldman
JM
Gordon
MY
The kinetics and extent of engraftment of chronic myelogenous leukemia cells in non-obese diabetic/severe combined immunodeficiency mice reflect the phase of the donor’s disease: An in vivo model of chronic myelogenous leukemia biology.
Blood
92
1998
1390
47
Cashman
JD
Eaves
AC
Eaves
CJ
Granulocyte-macrophage colony-stimulating factor modulation of the inhibitory effect of transforming growth factor-β on normal and leukemic human hematopoietic progenitor cells.
Leukemia
6
1992
886
48
Eaves
CJ
Cashman
JD
Wolpe
SD
Eaves
AC
Unresponsiveness of primitive chronic myeloid leukemia cells to macrophage inflammatory protein 1α, an inhibitor of primitive normal hematopoietic cells.
Proc Natl Acad Sci USA
90
1993
12015
49
Cashman
JD
Eaves
CJ
Sarris
AH
Eaves
AC
MCP-1, not MIP-1α is the endogenous chemokine that cooperates with TGF-β to inhibit the cycling of primitive normal but not leukemic (CML) progenitors in long-term human marrow cultures.
Blood
92
1998
2338
50
Ogawa
M
Differentiation and proliferation of hematopoietic stem cells.
Blood
81
1993
2844
51
Zandstra
PW
Conneally
E
Piret
JM
Eaves
CJ
Ontogeny-associated changes in the cytokine responses of primitive human haematopoietic cells.
Br J Haematol
101
1998
770
52
Cashman JD, Clark-Lewis I, Eaves AC, Eaves CJ: Differentiation stage-specific regulation of primitive human hematopoietic progenitor cycling by exogenous and endogenous inhibitors in an in vivo model. Blood (in press)

Author notes

Address reprint requests to Allen Eaves, MD, PhD, Terry Fox Laboratory, 601 W 10th Ave, Vancouver, British Columbia, Canada V5Z 1L3; e-mail: allen@terryfox.ubc.ca.

Sign in via your Institution