Interferon-α (IFN-α) is an established treatment for chronic myelogenous leukemia (CML) in chronic phase, but the mechanism of its antileukemic activity is not clear. One possible mechanism of action might include the induction of apoptosis, and especially Fas-mediated cell killing may play an important role in the elimination of malignant cells. We investigated Fas receptor (Fas-R) expression and the consequences of Fas-R triggering in CML patients. Using two-color flow cytometry, we found a significantly higher number of Fas-R–expressing CD34+ cells in the bone marrow (BM) of CML patients compared with normal subjects. We have previously shown that IFN-γ induces Fas-R expression on CD34+ cells; in this study, we investigated whether IFN-α induces Fas-R expression on CML progenitor cells. Dose-dependent induction of Fas-R expression was observed after IFN-α stimulation of CD34+ cells from CML BM. In methylcellulose culture, IFN-α alone at a therapeutic concentration showed only marginal antiproliferative effects on both normal and CML BM progenitors. In contrast, a Fas-R agonist, the anti-CD95 monoclonal antibody CH11, inhibited colony formation from normal progenitors, and the inhibition was even stronger on CML progenitors. When CML BM cells were cultured in the presence of IFN-α, Fas-R–mediated inhibition of colony growth was potentiated in a dose-dependent fashion, consistent with IFN-α induction of Fas-R expression. This functional effect did not require the presence of accessory cells, since similar results were obtained with purified CD34+ cells. In suspension cultures, we demonstrated that suppression of CML hematopoiesis by IFN-α and Fas-R agonist was exerted through Fas-R–mediated induction of apoptosis. Our findings suggest that the Fas-R/Fas-ligand system might be involved in the immunologic regulation of CML progenitor growth and that its effect can be amplified by IFN-α.

CHRONIC MYELOGENOUS leukemia (CML) arises from the clonal expansion of an altered stem cell capable of differentiation into mature myeloid cells.1-3 The Philadelphia chromosome (Ph) is the cytogenetic hallmark of the disease.4,5 The reciprocal translocation t(9,22) results in the creation of a chimeric bcr/abl gene6 believed to play a central role in leukemogenesis; mice transgenic for the fusion gene develop leukemia.7 Induction of clonal expansion by bcr/abl may be due to increased tyrosine kinase activity.8 Although the bcr/abl gene product may play a role in the inhibition of the apoptotic machinery,9-12 in some experimental systems a normal reaction of CML cells to death-inducing stimuli has been observed.13 CML bone marrow (BM) progenitor cells respond to colony-stimulating factors, but their adhesion to stroma is impaired, resulting in a loss of sensitivity to stromal inhibitory signals.14-17 

Several clinical trials have demonstrated that administration of interferon-α (IFN-α) is an effective treatment for CML in chronic phase: cytogenetic remission can be induced in 20% of patients treated with IFN-α, and long-term survival is improved compared with regimens including hydroxyurea and busulfan.18-21 A number of in vitro effects of IFN-α on CML cells has been described, such as direct inhibition of CML BM progenitor growth22,23 and restoration of the adherence to stroma.24-26 It has also been reported that IFN-α regulates the paracrine release of growth factors by human BM stroma by inhibiting the production of stimulatory cytokines such as granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-1β (IL-1β), and granulocyte colony-stimulating factor (G-CSF), as well as by increasing the production of inhibitory cytokines such as IL-1 receptor antagonist, transforming growth factor-β, and macrophage inflammatory protein-1α.26-28 However, the exact mechanism and especially the selectivity of IFN-α effects on CML cells remains a subject of controversy.29 

Inhibitory cytokines and cellular immune surveillance may play an important role in the control of expansion of the leukemic clone. In normal hematopoiesis, both direct T-cell–mediated hematopoietic suppression30 and inhibitory cytokine pathways have been described.31,32 IFN-γ and tumor necrosis factors (TNFs), alone or in combination with Fas receptor (Fas-R) agonists, inhibit the proliferation of hematopoietic progenitor cells, at least partially via induction of apoptosis.33-35 The inhibitory effects of IFN-γ seem to be mediated through induction of IFN regulatory factor-1 (IRF-1).36 Fas-R is expressed on activated lymphoid cells,37-41 in several hematologic malignancies,42-48 and also on normal CD34+ cells.35,36 Since the Fas-R/Fas ligand (Fas-L) system may be important in the elimination of virus-infected49,50 or malignant cells42-48 and since the Fas-R has been reported to be upregulated by IFN-γ and TNF-α on CD34+ cells34-36 and by IFN-α on Daudi cells,51 we hypothesized that the Fas-R/Fas-L system might be involved in the mechanisms responsible for the antileukemic effects of IFN-α in CML. Therefore, we investigated the expression and induction pathway of Fas and the consequences of Fas triggering in BM progenitor cells derived from CML patients.

Specimen collection.BM samples were obtained from 41 normal subjects, 45 patients with CML, and five patients with acute myeloid leukemia. Informed consent was obtained according to the protocol approved by the Institutional Review Board of the National Heart, Lung, and Blood Institute (Bethesda, MD) and the Hematology Division, Federico II University (Naples, Italy). The diagnosis of CML was confirmed by the cytogenetic finding of the Ph chromosome. Of the patients with CML, 40 were in chronic phase and five in blastic crisis (and untreated at the time of specimen collection). None of the patients were receiving IFN-α at the time of BM sampling or at least within 2 months before BM sampling.

BM cell separation.BM was aspirated from the posterior iliac crest into syringes containing Iscove's modified Dulbecco's medium (IMDM) supplemented 1:10 with heparin (O'Neil & Feldman, St Louis, MO). Mononuclear cells (BM-MNC) were isolated by density-gradient centrifugation using lymphocyte separation medium (Organon, Durham, NC). After washing with Hanks' balanced salt solution (HBSS), cells were resuspended in IMDM supplemented with 5% fetal calf serum (FCS). HBSS, IMDM, and FCS were purchased from Life Technologies (Gaithersburg, MD).

Flow cytometric analysis.A fluorescein isothiocyanate (FITC)-conjugated (Fab′) fragment of a murine anti-human CD95 (clone UB2; Amac, Westbrook, ME) was used to determine the expression of Fas-R on BM cells. For two-color analysis, phycoerythrin (PE)-conjugated monoclonal antibody (MoAb) to CD34 (Becton Dickinson, Mountain View, CA) was used in combination with FITC-conjugated CD95 MoAb. Proper isotypic controls were used in all experiments. BM-MNC were resuspended in 100 μL phosphate-buffered saline ([PBS] Life Technologies) containing 2% FCS, incubated with 20 μL MoAb, washed three times with PBS, and analyzed by flow cytometry (Epics; Coulter, Hialeah, FL). Fas expression on CD34+ cells was analyzed in a uniformly set blast gate.

Separation of CD34+ cells.In some experiments, purified CD34+ cells were used for analysis of Fas-R expression or for colony assay. CD34+ cells were separated using affinity columns (Cellpro, Bothel, WA). Briefly, nonadherent BM cells were incubated at room temperature with streptavidin-conjugated murine anti-human CD34 IgM, washed with PBS, and applied to an affinity column containing biotin-coated beads; the CD34+ fraction was eluted with PBS. An aliquot of eluted cells was stained with PE-conjugated anti-CD34 HPCA-2 MoAb (Becton Dickinson) for purity assessment. Usually, 70% to 90% of separated cells were CD34+.

Macrophage and T-cell depletion.Macrophages and monocytes were depleted from BM samples using adherence to plastic. For T-cell depletion, BM-MNC were incubated on ice with CD3 MoAb (50 μL for 2 × 106 cells) for 30 minutes, washed with PBS, and then incubated with anti-mouse IgG–conjugated magnetic beads (Dynal, Oslo, Finland) on ice for 10 minutes before separation using a magnetic separator. This method yielded a virtually macrophage- and T-cell–free mononuclear BM cell population. The degree of residual contamination was evaluated using flow cytometry after staining cell aliquots with CD14 and CD3 MoAb. BM cell fractions used for further experiments usually contained less than 2% CD3+ cells and 1% CD14+ cells.

Hematopoietic colony assay and suspension cultures.Proliferation of hematopoietic progenitors was measured in methylcellulose cultures. Fresh total BM or isolated CD34+ cells were plated in methylcellulose (Stem Cell Technologies, Vancouver, CA) at a concentration of 1 × 105 or 1 × 103/mL medium (35-mm dishes, 1 mL medium per dish), respectively. The growth factor cocktail consisted of 10 ng/mL IL-3, 50 ng/mL G-CSF, 50 ng/mL GM-CSF, 20 ng/mL stem cell factor, and 2 U/mL erythropoietin (all factors kindly donated by Amgen, Thousand Oaks, CA). Recombinant IFN-α (Hoffmann La Roche, Basel, Switzerland) and anti-Fas MoAb (clone CH11; Amac) were used at a concentration range of 20 to 1,000 U/mL and 0.5 to 1.0 μg/mL, respectively. For control experiments, nonrelevant isotypic MoAbs (Dako, Carpinteria, CA) were used. All cultures were performed in duplicate. Suspension cultures were performed in 24-well plates in IMDM containing 20% FCS and growth factors at the concentrations already described. All experimental procedures were performed in endotoxin-free plasticware.

Apoptosis assay.DNA fragmentation was measured after extraction of low–molecular-weight DNA from a constant number of cells. Cells (2 × 106) were resuspended in 900 μL 1× Tris-EDTA buffer and lysed with 25 μL 20% sodium dodecyl sulfate. The high–molecular-weight DNA fraction was precipitated for 6 hours in the presence of 5 mol/L NaCl and pelleted by high-speed centrifugation. The fragmented DNA was then extracted from the aqueous phase with phenol and chloroform and then precipitated with ethanol. After resuspension in water, DNA was electrophoresed using 1.5% agarose gel and visualized by ethidium bromide staining.

Reverse transcriptase–polymerase chain reaction (RT-PCR) for human Fas RNA.Total RNA was extracted from constant numbers of purified CD34+ cells using RNAsol (Cina/Biotecx, Friendswood, TX). Contaminating DNA was digested using RNAse-free DNAse I (Boehringer, Indianapolis, IN). RNA was re-extracted with phenol/chloroform, precipitated with ethanol, and diluted in RNAse-free water. After reverse transcription using an oligo d(T)16 primer, Fas-R cDNA was amplified using the primer pairs 5′-GGACATGGCTTAGAAGTGGA-3′ and 5′-CTGCTGTG TCTTGGACATTG-3′ specific for human Fas-R simultaneously with primers specific for human β-actin (5′-CAATTGTGATGGACTCCGGAGACGG-3′ as an upstream primer and 5′-CATCTGCTG CTCGAAGTCTAGAGC-3′ as a downstream primer). For the amplification reaction, reagents supplied in the Amplimer kit (Perkin Elmer, Foster City, CA) were used. For reverse transcription and amplification, the following conditions were used: 45 minutes at 37°C and 5 minutes at 96°C for the RT reaction, and 30 cycles times 2 minutes at 96°C, 1.5 minutes at 55°C, and 2 minutes at 72°C for amplification. PCR products were electrophoresed in 1.2% agarose gels. The bands were visualized after staining with ethidium bromide and UV light exposure.

Expression of Fas antigen on BM cells from CML patients.We analyzed the expression of Fas-R (CD95) on normal BM cells derived from 41 donors and from 45 patients with CML. A significantly higher percentage of cells expressing Fas-R was found in CML patients (31.3 ± 24 v 10.7 ± 9, P < .05; Table 1). Using two-color flow cytometry with FITC-conjugated anti-CD95 and PE-conjugated anti-CD34 (Fig 1), we found in normal BM a mean of 8.4% ± 6% CD34+ cells expressing Fas-R, and in CML BM, 25.8% ± 22% (P < .05; Table 1). When five patients in blastic crisis were compared with those in chronic phase, a significantly increased Fas-R expression was found (Fig 2).

Fig. 1.

Flow cytometric analysis of Fas-R expression on CD34+ cells derived from BM of CML patients. Total BM cells were stained with PE-conjugated CD34 and FITC-conjugated CD95 MoAb. Quadrants were set on the basis of cells treated with PE- and FITC-labeled isotypic controls. N.V., BM sample from a healthy volunteer; CML cp-1 to cp-5, BM samples from 5 patients with CML in chronic phase; CML bc, BM sample from a patient with CML in blastic crisis; AML, BM sample from a patient with acute myeloid leukemia. BM CD34+ cells were analyzed in the blast gate for CD95 expression.

Fig. 1.

Flow cytometric analysis of Fas-R expression on CD34+ cells derived from BM of CML patients. Total BM cells were stained with PE-conjugated CD34 and FITC-conjugated CD95 MoAb. Quadrants were set on the basis of cells treated with PE- and FITC-labeled isotypic controls. N.V., BM sample from a healthy volunteer; CML cp-1 to cp-5, BM samples from 5 patients with CML in chronic phase; CML bc, BM sample from a patient with CML in blastic crisis; AML, BM sample from a patient with acute myeloid leukemia. BM CD34+ cells were analyzed in the blast gate for CD95 expression.

Close modal
Fig. 2.

Expression of Fas-R on BM CD34+ cells from healthy subjects and CML patients. Values correspond to the percentage of CD95+CD34+ within total CD34+ cells (CD34+CD95+ + CD34+CD95). Statistical analysis (nonparametric Kruskal-Wallis test): control v CML, P < .05. Horizontal bars are the mean values, and vertical bars are the SD.

Fig. 2.

Expression of Fas-R on BM CD34+ cells from healthy subjects and CML patients. Values correspond to the percentage of CD95+CD34+ within total CD34+ cells (CD34+CD95+ + CD34+CD95). Statistical analysis (nonparametric Kruskal-Wallis test): control v CML, P < .05. Horizontal bars are the mean values, and vertical bars are the SD.

Close modal

Effects of IFN-α on Fas-R expression on CD34+ cells.IFNs, specifically IFN-γ, have been associated with induction of Fas-R expression,34,35 possibly via the IRF-1 pathway.36 Fas triggering leads to hematopoietic suppression and apoptosis. We investigated whether IFN-α was capable of inducing Fas-R expression on CD34+ cells derived from CML patients. When purified CD34+ cells from CML BM were cultured in the presence of IFN-α levels comparable to those found after administration of IFN-α to patients in vivo, an increased expression of Fas-R mRNA was detected using RT-PCR (Fig 3). IFN-α–induced Fas-R expression was also quantitatively documented by two-color flow cytometric analysis: IFN-α enhanced Fas-R expression on CD34+ cells from CML patients in a dose-dependent manner (Figs 4 and 5). CD34+ cells derived from normal volunteers were less responsive to the stimulatory effect of IFN-α (CD34+CD95+, 7.8 ± 6 and 12.6 ± 10 without and with 1,000 U/mL IFN-α, respectively; P > .05).

Fig. 3.

Expression of Fas-R mRNA in CD34+ cells from two CML patients after stimulation with IFN-α. Bands represent the product of the RT-PCR for Fas (upper gel) and β-actin (lower gel) from purified CD34+ cells of two patients with CML in chronic phase.

Fig. 3.

Expression of Fas-R mRNA in CD34+ cells from two CML patients after stimulation with IFN-α. Bands represent the product of the RT-PCR for Fas (upper gel) and β-actin (lower gel) from purified CD34+ cells of two patients with CML in chronic phase.

Close modal
Fig. 4.

Flow cytometric analysis of Fas-R expression on CD34+ cells from a patient with CML stimulated with various concentrations of IFN-α. Total BM cells were cultured with or without IFN-α for 48 hours, stained with PE-conjugated CD34 and FITC-conjugated CD95 MoAb, and analyzed in the blast gate. (A) Without IFN-α; (B) IFN-α 500 U/mL; (C) IFN-α 1,000 U/mL. Gates were set based on isotypic controls.

Fig. 4.

Flow cytometric analysis of Fas-R expression on CD34+ cells from a patient with CML stimulated with various concentrations of IFN-α. Total BM cells were cultured with or without IFN-α for 48 hours, stained with PE-conjugated CD34 and FITC-conjugated CD95 MoAb, and analyzed in the blast gate. (A) Without IFN-α; (B) IFN-α 500 U/mL; (C) IFN-α 1,000 U/mL. Gates were set based on isotypic controls.

Close modal
Fig. 5.

Dose-dependent induction of CD95 expression on CD34+ cells derived from BM of CML patients. Total BM cells were cultured in the presence of various concentrations of IFN-α for 48 hours, and stained with PE-conjugated CD34 and FITC-conjugated CD95 MoAb. Statistical analysis (paired t test): control v 500 and 1,000 U/mL IFN-α; P < .05 (summary of 7 experiments).

Fig. 5.

Dose-dependent induction of CD95 expression on CD34+ cells derived from BM of CML patients. Total BM cells were cultured in the presence of various concentrations of IFN-α for 48 hours, and stained with PE-conjugated CD34 and FITC-conjugated CD95 MoAb. Statistical analysis (paired t test): control v 500 and 1,000 U/mL IFN-α; P < .05 (summary of 7 experiments).

Close modal

Effects of IFN and Fas-R agonist MoAb CH-11 on CML progenitor growth.We tested the influence of IFN-α on colony formation by both total BM cells and purified CD34+ cells from normal subjects and CML patients. Generally, there was more variability in the number of colonies (higher standard deviations) among patients with CML (Table 2). IFN-α at concentrations comparable to those achieved after in vivo administration (15 to 50 U/mL52 ) marginally inhibited colony growth from normal and CML BM cells (Table 2). Higher concentrations of IFN-α consistently inhibited both erythroid and myeloid colony formation from normal and CML BM cells in a dose-dependent manner (Table 2). The ID50 of IFN-α (50% inhibition of colony formation) in normal BM progenitor cells was 770 U/mL and 14.5 U/mL without and with Fas MoAb, respectively. However, when the inhibitory capacity of IFN-α was tested in CML BM progenitor, the ID50 of IFN-α was 1,050 U/mL and 2.5 U/mL without and with Fas mAb, respectively. Using highly purified BM CD34+ cells, we showed that this effect did not require accessory cells, since similar results were generated with CD34+ cells and total BM cells in methylcellulose assays (Table 2). To exclude the possibility of involvement of other known inducers of Fas-R expression, two experiments were performed with neutralizing antibodies to IFN-γ and TNF-α. In the presence of anti–IFN-γ and anti–TNF-α antibodies added to the methycellulose cultures, similar results with regard to the synergistic effects of IFN-α and CH11 on colony inhibition were observed (data not shown).

In previous studies, we have demonstrated that an anti-Fas MoAb (CH11) acts synergistically with IFN-γ and TNF-α to inhibit hematopoietic colony formation from normal hematopoietic progenitors.34 Anti-Fas MoAb (CH11) mimicks the biologic activity of Fas-L on Fas-R–expressing cells, including hematopoietic progenitors. Using this MoAb as a Fas-R agonist, we investigated whether CH11-treated CD34+ cells from CML patients were more sensitive to Fas-R–mediated hematopoietic suppression. There was a correlation between the IFN-α–mediated enhancement of Fas expression and the degree of hematopoietic inhibition seen in the colony cultures. Since normal progenitor cells also express low levels of Fas-R, there was some inhibition of colony formation seen in cultures of normal cells treated with CH11 (without priming with IFN-α). In samples derived from patients, the effect of CH11 was much stronger in comparison to normal BM, since CML progenitors express higher levels of Fas-R (Table 2). In total BM cultures, IFN-α potentiated the effect of anti-Fas MoAb in a dose-dependent manner (Table 2). Similar results were obtained in a 3H-thymidine proliferation assay performed with total BM cells from CML patients (data not shown). To determine whether this effect requires the presence of accessory cells, we performed colony assays with purified CD34+ cells. IFN-α significantly potentiated Fas-R–mediated inhibition of colony formation derived from both CML and normal progenitors (Table 2).

Potentiation of Fas-R–mediated apoptosis by IFN.In experiments with purified CD34+ cells, we have previously demonstrated that Fas-R–mediated inhibition of hematopoiesis is associated with BM progenitor cell apoptosis,34 and this effect is potentiated by cytokines including IFN-γ and TNF-α.33 When BM cells from CML patients and normal subjects were treated with anti–Fas-R MoAb in combination with IFN-α, the Fas-R agonist potentiated IFN-α–mediated induction of apoptosis, as demonstrated by a DNA fragmentation assay (Fig 6). Normal BM cells treated with the same concentration of anti–Fas-R34 or IFN-α (Fig 6) showed no visible apoptosis in agarose gel electrophoresis. A sufficient number of purified CD34+ cells for apoptosis assay could not be obtained from CML patients to test whether apoptosis was a complex phenomenon involving other cell subset. However, similar results were obtained with macrophage- and T-cell–depleted BM, suggesting that under our experimental conditions the induction of apoptosis did not require the presence of accessory cells.

Fig. 6.

Induction of apoptosis in culture of total and T-cell–depleted BM cells from a normal subject and from a patient with CML stimulated by anti-Fas MoAb. (A) and (B) Ethidium bromide staining of agarose gel after electrophoresis of low–molecular-weight DNA. DNA was extracted from 1 × 106 total BM cells (A) and T-cell–depleted BM cells (B) after culture for 48 hours in the presence of the indicated concentrations of IFN-α. Anti-Fas MoAb was used at a concentration of 1 μg/mL.

Fig. 6.

Induction of apoptosis in culture of total and T-cell–depleted BM cells from a normal subject and from a patient with CML stimulated by anti-Fas MoAb. (A) and (B) Ethidium bromide staining of agarose gel after electrophoresis of low–molecular-weight DNA. DNA was extracted from 1 × 106 total BM cells (A) and T-cell–depleted BM cells (B) after culture for 48 hours in the presence of the indicated concentrations of IFN-α. Anti-Fas MoAb was used at a concentration of 1 μg/mL.

Close modal

We have investigated the role of Fas-R expression and triggering in the regulation of proliferation of hematopoietic progenitor cells in CML. Our results demonstrate that CML CD34+ cells in the chronic phase of the disease show significantly higher expression of Fas-R and increased sensitivity to Fas-R–mediated inhibition of colony growth. Furthermore, IFN-α enhanced the expression of Fas-R on CML progenitor cells and rendered them more susceptible to apoptosis induced by the Fas-R agonist anti–Fas MoAb CH11. IFN-α at high concentrations directly inhibited CML progenitor cells,22,23 whereas low concentrations (in the range achieved in blood after in vivo administration57 ) showed only a marginal suppressive effect in colony assay. In the presence of a Fas-R agonist, the same low IFN-α concentrations were capable of inducing a profound decrease in CML progenitor cell proliferation. Some selectivity of the Fas agonist–mediated effects on CML versus normal progenitor cells is inferred from the lower IFN-α concentrations required for 50% inhibition of colony formation in the cultures supplemented with Fas MoAb. The mechanism for such selectivity in Fas-R–mediated killing remains to be elucidated.

As inferred from studies on allogeneic BM transplants in CML, the cellular immune response may play an important role in inhibiting Ph+ hematopoiesis (graft versus leukemia [GvL] effect). Although the existence and significance of an autologous GvL effect is a subject of controversy,53 the presence of leukemia-specific T-cell clones has been demonstrated in CML.54 55 Fas expression may render CML progenitors more vulnerable to Fas-R–mediated killing, triggered by membrane-bound Fas-L on leukemia-specific T cells. Secretory Fas-L might also induce apoptosis of CML cells without the need for recognition of targets by specific T cells or direct cell-cell contact.

The effects of IFN-α in CML may be at least partially explained by Fas-R upregulation and the subsequent increased death rate of Fas+ CML progenitor cells. These cells, upon expression of Fas-R, showed low clonogenicity even in the absence of Fas agonist, as demonstrated in the sorting of CD34+CD95+ and CD34+CD95 cells (J.P. Maciejewski, unpublished results). It is possible that Fas-R expression is an event associated with preparation of the progenitor cell for apoptosis. Expression and subsequent triggering of Fas-R could represent the “final hit” in this process. Because increased Fas-R expression has been observed on several leukemic cell lines of both lymphoid and nonlymphoid origin,37,38,43-49 Fas-R induction pathways may not be tissue-specific. Recently, we have documented that IFN-γ and TNF-α induce Fas-R expression on CD34+ cells.34,35 The effects of these cytokines are mediated by intracellular induction of IRF-1.36 Involvement of the Fas-R pathway in IFN-α–induced apoptosis in the Daudi lymphoma cell line suggested that the Fas gene may belong to the IFN-inducible gene family,56 and its transcription may be transactivated through IRF-1 or other IFN-mediated signal transcription factors.57 However, since the promotor region of the Fas gene does not contain any IFN-responsive elements,58 further molecular analyses will be required to clarify the mechanisms of Fas-R induction by IFN-α.

The hybrid bcr/abl gene has been implicated in the suppression of programmed cell death.9-12 Recently, intrinsic kinase activity of bcr/abl was shown to be responsible for activation (phosphorylation) of STAT1 and STAT5, also involved in the signal transduction of several growth factor receptors.59 Fas effects could be due to several mechanisms, eg, the downmodulation of p210 that blocks the physiologic apoptotic pathway in CML progenitor cells. However, a recent report60 has shown that bcr/abl antiapoptotic activity is ineffective against apoptosis induced by natural killer (NK) or lymphokine-activated killer (Lak) cells. The NK-effector mechanism involves the perforin/granzyme pathway, but Lak cells may also use a Fas-R–mediated killing mechanism. In some reports, the sensitivity to apoptosis in CML cells did not differ from that of normal progenitor cells, a result that argues against a role for inhibition of apoptosis in clonal expansion.12 

Enhanced Fas-R expression of CML progenitor cells may be intrinsic to the genetic defect in CML and caused by bcr/abl. The absence of Fas-R on, eg, K562 cells carrying bcr/abl does not support this hypothesis (J.P. Maciejewski, unpublished data). Alternatively, increased expression of Fas-R on CML CD34+ cells may also be a result of hypersensitivity to endogenously produced IFN-α. Although the IFN-α effect on Fas-R expression was observed even in a colony assay with purified CD34+ cells, we cannot rigorously exclude the possibility that in vivo Fas-R upmodulation is a result of the action of several other cytokines released as part of the natural response to malignancy. These cytokines may include IFN-γ and TNF-α, which have been previously shown to enhance Fas-R expression on normal CD34+ cells.34 In vitro experiments with highly purified CD34+ cells lacking accessory IFN-γ and TNF-α producers and IFN-γ – and TNF-α–neutralizing experiments did not support this possibility.

The finding of increased CD95 expression on CD34+ cells from CML patients may be restricted to a discrete, phenotypically defined subpopulation of CD34+ cells more abundant in CML patients. Because most immature progenitor and stem cells can only be characterized by functional tests (hematopoietic colony and long-term culture–initiating cell assays), a selective analysis of Fas-R expression in these cells is not possible. In our experiments, elevated Fas-R levels were also observed on phenotypically more mature cells from CML (CD33+, CD13+, and CD3+ cells; data not shown). Since the induction of Fas-R may be part of the normal myeloid maturation process, the comparison of Fas-R expression on these cells in CML patients and normal subjects was less conclusive.

Although the number of CML patients in blastic crisis we studied is small, our findings suggest that during transformation Fas-R expression is elevated. One possible interpretation is that a selective pressure in vivo may confer an advantage on clones with acquired genetic resistance to apoptotic stimuli. Additional investigations now in progress will clarify whether IFN-α–induced Fas-R expression occurs in vivo and is specific for CML cells, as well as whether the increased Fas-R expression in the blastic phase of CML is associated with a loss of sensitivity to Fas-mediated apoptosis.

In the future, a proper understanding of the Fas-R/Fas-L system in the regulation of normal and leukemic hematopoiesis might lead to introduction of novel immunotherapeutic principles for the treatment of CML.

Address reprint requests to Jaroslaw P. Maciejewski, MD, PhD, Department of Internal Medicine, University of Nevada, Reno, Howard Medical Bldg 320, Reno, NV 89557-0046.

1
Fialkow
PJ
Jacobson
RJ
Papayannopoulou
T
Chronic myelocytic leukemia: Clonal origin in a stem cell common to the granulocyte, erythrocyte, platelet and monocyte/macrophage.
Am J Med
63
1972
125
2
Martin
PJ
Najfield
V
Hansen
JA
Penfold
JK
Fialkow
PJ
Involvement of the B-lymphoid system in chronic myelogenous leukemia.
Nature
287
1980
49
3
Nitta
M
Kato
Y
Strife
A
Wachter
M
Fried
J
Perez
A
Jhanwar
S
Duigou-Ostendorf
R
Chaganti
RSK
Clarkson
B
Incidence of involvement of the B and T lymphocyte lineages in chronic myelogenous leukemia.
Blood
66
1985
1053
4
Rowley
JD
A new consistent chromosomal abnormality in chronic myelogenous leukemia identified by quinacrine fluorescence and Giemsa staining.
Nature
243
1973
290
5
Nowell
PC
Hungerford
DA
A minute chromosome in human chronic granulocytic leukemia.
Science
132
1960
1497
6
Kurzrock
R
Gutterman
JU
Talpaz
M
The molecular genetics of Philadelphia chromosome–positive leukemias.
N Engl J Med
319
1988
990
7
Heisterkamp
N
Jenster
G
ten-Hoeve
J
Zovich
D
Pattengale
PK
Groffen
J
Acute leukemia in bcr/abl transgenic mice.
Nature
344
1990
251
8
McWhirter
JR
Wang
JYJ
Activation of tyrosine kinase and microfilament-binding functions of c-abl by bcr sequences in bcr/abl fusion protein.
Mol Cell Biol
11
1991
1553
9
Cotter
TG
bcr-abl: An anti-apoptosis gene in chronic myelogenous leukemia.
Leuk Lymphoma
18
1995
231
10
Bedi
A
Zehnbauer
BA
Barber
JP
Sharkis
SJ
Jones
RJ
Inhibition of apoptosis by bcr-abl in chronic myeloid leukemia.
Blood
83
1994
2038
11
Bedi
A
Barber
JP
Bedi
GC
El-Deiry
S
Sidranski
D
Vala
MS
Akhtar
AJ
Hilton
J
Jones
RJ
bcr-abl–mediated inhibition of apoptosis with delay of G2/M transition after DNA damage: A mechanism of resistance to multiple anticancer agents.
Blood
86
1995
1148
12
McGahon
A
Bissonette
R
Schmitt
M
Cotter
KM
Green
DR
Cotter
TG
BCR-ABL maintains resistance of chronic myelogenous leukemia cells to apoptotic cell death.
Blood
83
1994
1179
13
Amos
TAS
Lewis
JL
Grand
FH
Gooding
RP
Goldman
JM
Gordon
MY
Apoptosis in chronic myeloid leukemia: Normal responses by progenitor cells to growth factor deprivation, x-irradiation and glucocorticoids.
Br J Haematol
91
1995
387
14
Gordon
MY
Dowding
CR
Riley
GP
Goldman
JM
Altered adhesive interactions with marrow stroma of haematopoietic cells in chronic myeloid leukemia.
Nature
328
1987
342
15
Verfaillie
CM
McCarthy
JB
McGlave
PB
Mechanisms underlying abnormal trafficking of malignant progenitors in chronic myelogenous leukemia.
J Clin Invest
90
1992
1232
16
Wetzler
M
Kurzrock
R
Lowe
DG
Kantarjian
H
Gutterman
JU
Talpaz
M
Alteration in bone marrow adherent layer growth factor expression: A novel mechanism of chronic myelogenous leukemia progression.
Blood
78
1991
2400
17
Bathia
R
McGlave
PB
Dewald
GW
Blazar
BR
Verfaillie
CM
Abnormal function of the bone marrow microenvironment in chronic myelogenous leukemia: Role of malignant stromal macrophages.
Blood
85
1995
3636
18
Ozer
H
George
SL
Schiffer
CA
Rao
K
Rao
PN
Wurster-Hill
DH
Arthur
DD
Powell
B
Gottlieb
A
Peterson
BA
Rai
K
Testa
JR
LeBeau
M
Tantravahi
R
Bloomfield
CD
Prolonged subcutaneous administration of recombinant alpha-2b interferon in patients with previously untreated Philadelphia chromosome–positive chronic phase myelogenous leukemia: Effect on remission duration and survival: Cancer and Leukemia Group B Study 8583.
Blood
82
1993
2975
19
Italian
Cooperative Study Group on Chronic Myeloid Leukemia
Interferon alfa-2a as compared with conventional chemotherapy for the treatment of chronic myeloid leukemia.
N Engl J Med
30
1994
820
20
Talpaz
M
Kantarijian
H
Kurzrock
R
Trujillo
JM
Guttermann
JU
Interferon-alpha produces sustained cytogenetic response in chronic myeloid leukemia.
Ann Intern Med
114
1991
532
21
Lee
MS
Kantarijian
H
Talpaz
M
Freireich
EJ
Deisseroth
A
Trujillo
JM
Stass
SA
Detection of minimal residual disease by polymerase chain reaction in Philadelphia chromosome–positive chronic myelogenous leukemia following interferon therapy.
Blood
79
1992
1920
22
Broxmeyer
HE
Lu
E
Platzer
E
Feit
C
Juliano
L
Rubin
BY
Comparative analyses of the influences of human gamma, alpha and beta interferons on human multipotential (CFU-GEMM), erythroid (BFU-E) and granulocyte macrophage (CFU-GM) progenitor cells.
J Immunol
131
1983
1330
23
Geissler
D
Gastl
G
Aulitzky
W
Tilg
H
Gaggl
S
Konwalinka
G
Huber
C
Recombinant interferon-alpha-2C in chronic myelogenous leukaemia: Relationship of sensitivity of committed haematopoietic precursor cells in vitro (BFU-E, CFU-GM, CFU-Meg) and clinical response.
Leuk Res
14
1990
629
24
Upadhyaya
G
Guba
SC
Sih
SA
Feinberg
AP
Talpaz
M
Kantarjian
HM
Deisseroth
AB
Emerson
SG
Interferon-alpha restores the deficient expression of the cytoadhesion molecule lymphocyte function antigen-3 by chronic myelogenous leukemia progenitor cells.
J Clin Invest
88
1991
2131
25
Dowding
C
Guo
AP
Osterholz
J
Sickowski
M
Goldman
J
Gordon
M
Interferon-alpha overrides the deficient adhesion of chronic myelogenous leukemia primitive progenitor cells to bone marrow stromal cells.
Blood
78
1991
499
26
Bathia
R
McGlave
P
Verfaillie
CM
Treatment of marrow stroma with interferon-α restores normal β1 integrin–dependent adhesion of chronic myelogenous leukemia hematopoietic progenitors. Role of MIP-1α.
J Clin Invest
96
1995
931
27
Aman
JM
Keller
U
Derigs
G
Mohammadzadeh
M
Huber
C
Peschel
C
Regulation of cytokine expression by interpheron alpha in human bone marrow stroma cells: Inhibition of hematopoietic growth factors and induction of IL-1 receptor antagonist.
Blood
84
1994
4142
28
Bathia R, McGlave PB, Verfaillie CM: Interpheron-α treatment of marrow stroma results in enhanced adhesion of chronic myelogenous leukemia progenitors via mechanism involving MIP-1α and TGF-β. Exp Hematol 22:797a, 1994 (abstr)
29
Dowding
C
Guo
AP
Maisin
D
Gordon
MY
Goldman
JM
The effects of the proliferation of CML progenitor cells in vitro are not related to the precise position of the M-BCR breakpoint.
Br J Haematol
77
1991
165
30
Zoumbos
N
Gascon
P
Young
NS
The induction of lymphocytes in normal and suppressed hematopoiesis.
Blut
48
1984
1
31
Emerson
SB
Antin
JH
Bone marrow progenitor cells induce a regulatory autologous proliferative T lymphocyte response.
J Immunol
142
1989
766
32
Cashmann
JD
Eaves
AC
Raines
EW
Ross
R
Eaves
CJ
Mechanisms that regulate the cell cycle status of very primitive hematopoietic cells in long-term human marrow cultures. II. Analysis of positive and negative regulators produced by stromal cell within the adherent layer.
Blood
78
1991
110
33
Selleri
C
Sato
T
Anderson
S
Young
NS
Maciejewski
JP
Interferon-γ and tumor necrosis factor-α suppress both early and late stages of hematopoiesis and induce programmed cell death.
J Cell Physiol
165
1995
538
34
Maciejewski
JP
Selleri
C
Anderson
S
Young
NS
Fas antigen expression on CD34+ human bone marrow cells is induced by interferon-γ and tumor necrosis factor-α and potentiates cytokine-mediated hematopoietic suppression in vitro.
Blood
85
1995
3183
35
Sato T, Selleri C, Anderson S, Young NS, Maciejewski JP: Expression and modulation of cellular receptors for interferon-γ, tumor necrosis factor, and Fas on human bone marrow CD34+ cells. Blood 36:443a, 1995 (abstr, suppl 1)
36
Sato
T
Selleri
C
Young
NS
Maciejewski
J
Hematopoietic inhibition by interferon-γ is partially mediated through interferon regulatory factor-1.
Blood
86
1995
3373
37
Nagafuji
K
Shibuya
T
Harada
M
Mizuno
S
Takenaka
K
Miyamoto
T
Okamura
T
Gondo
T
Niho
Y
Functional expression of Fas antigen (CD95) on hematopoietic progenitor cells.
Blood
86
1995
883
38
Itoh
N
Yonehara
S
Ishii
A
Yonehara
M
Mizushima
SI
Sameshima
M
Hase
A
Seto
Y
Nagata
S
The polypeptide sequence encoded by the cDNA for human cell surface antigen Fas can mediate apoptosis.
Cell
66
1991
233
39
Oehm
A
Behrman
I
Falk
W
Pawlita
M
Purification and molecular cloning of the Apo-1 cell surface antigen, a member of the tumor necrosis factor/nerve growth factor receptor superfamily. Sequence identity with the Fas antigen.
J Biol Chem
267
1992
10709
40
Kagi
D
Vignaux
F
Ledermenn
B
Burki
K
Depraetere
V
Nagata
S
Hengartner
H
Golstein
P
Fas and perforin pathways as major mechanisms of T-cell mediated cytotoxicity.
Science
265
1994
528
41
Rouvier
E
Luciani
MF
Golstein
P
Fas involvement in Ca2+-independent T cell–mediated cytotoxicity.
J Exp Med
177
1993
195
42
Hanabuchi
S
Koyanagi
M
Kawasaki
A
Shinohara
N
Matsuzawa
A
Nishimura
Y
Kobayashi
Y
Yonehara
Y
Yagita
H
Okumura
K
Fas and its ligand is a general mechanism of T-cell–mediated cytotoxicity.
Proc Natl Acad Sci USA
91
1994
4930
43
Owen-Schaub
LB
Meterissian
S
Ford
RJ
Fas/APO-1 expression and function on malignant cells of hematologic and non-hematologic origin.
J Immunother
14
1993
234
44
Debatin
KM
Goldmann
CK
Waldmann
TA
Krammer
PH
APO-1–induced apoptosis of leukemia cells from patients with adult T-cell leukemia.
Blood
81
1993
2972
45
Mapara
MY
Bargou
R
Zugks
C
Dohner
H
Ustaoglu
F
Jonker
RR
Krammer
PH
Dorkenn
B
Apo-1 mediates apoptosis or proliferation in human chronic B lymphocytic leukemia: Correlation with bcl-2 oncogene expression.
Eur J Immunol
23
1993
702
46
Robertson
MJ
Manley
TJ
Pichert
G
Cameron
C
Cochran
KJ
Levine
H
Ritz
J
Functional consequences of APO-1/Fas (CD95) antigen expression by normal and neoplastic hematopoietic cells.
Leuk Lymphoma
17
1994
51
47
Munker
R
Lubbert
M
Yonehara
S
Tuchnitz
A
Mertelsmann
R
Wilmanns
W
Expression of the Fas antigen on primary human leukemia cells.
Ann Hematol
70
1995
15
48
Westendorf
JJ
Lammert
JM
Jelinek
DF
Expression and function of Fas (APO-1/CD95) in patient myeloma cells and myeloma cell lines.
Blood
85
1995
3566
49
Massaia
M
Borrione
P
Attisano
C
Barral
P
Beggiato
E
Montacchini
L
Bianchi
A
Boccadoro
M
Pileri
A
Dysregulated Fas and Bcl-2 expression leading to enhanced apoptosis in T cells of multiple myeloma patients.
Blood
85
1995
3679
50
Takizawa
T
Matsukawa
S
Higuchi
Y
Nakamura
S
Nakanishi
Y
Fukuda
R
Induction of programmed cell death (apoptosis) by influenza virus infection in tissue culture cells.
J Gen Virol
74
1993
2347
51
Gissingler H, Kurzrock K, Jang S, Andreeff M, Talpaz M: FAS/APO1, another interferon inducible gene. Blood 86:541a, 1995 (abstr, suppl 1)
52
Gutterman
JU
Fine
S
Quesada
J
Hornong
SJ
Levine
JF
Alexanian
R
Bernhardt
L
Kramer
M
Spiegel
H
Colburn
W
Trown
P
Merigan
T
Dziewanoski
Z
Recombinant leucocyte α interferon: Pharmacokinetics, single-dose tolerance, and biological effects in cancer patients.
Ann Intern Med
96
1982
549
53
Champlin
R
Separation of graft-vs.-host disease and graft-vs.-leukemia effect against chronic myelogenous leukemia.
Exp Hematol
23
1995
1148
54
Faber
LM
van Luxemburg-Heijs
SAP
Veenhof
FJ
Willemze
R
Falkemburg
JHF
Generation of CD4+ cytotoxic T-lymphocyte clones from a patient with severe graft-versus-host disease after allogeneic bone marrow transplantation: Implication for graft-versus-leukemia reactivity.
Blood
86
1995
2821
55
Jiang
Y-Z
Barrett
J
Cellular and cytokine-mediated effects of CD4-positive lymphocyte lines generated in vitro against chronic myelogenous leukemia.
Exp Hematol
23
1995
1167
56
Hiramatsu
N
Hayashi
N
Katayama
K
Mochizuki
K
Kawanishi
Y
Kasahara
A
Fusamoto
H
Kamada
T
Immunohistochemical detection of Fas antigen in liver tissue of patients with chronic hepatitis C.
Hepatology
19
1994
1354
57
Fisher
T
Aman
J
van der Kuip
H
Peschel
C
Aulitzky
WE
Huber
C
Induction of interferon regulatory factors, 2′-5′ oligoadenylate synthetase, P68 kinase and Rnase L in chronic myelogenous leukaemia cells and its relationship to clinical responsiveness.
Br J Haematol
92
1996
595
58
Cheng
J
Liu
C
Koopman
WJ
Mountz
JD
Characterization of human Fas gene. Exon/intron organization and promoter region.
J Immunol
154
1995
1239
59
Carlesso
N
Frank
DA
Griffin
JD
Tyrosyl phosphorylation and DNA binding activity of signal transducers and activators of transcription (STAT) proteins in hematopoietic cell lines transformed by Bcr/Abl.
J Exp Med
183
1996
811
60
Roger
R
Isaad
C
Pallardy
M
Leglise
M-C
Turhan
AG
Bertoglio
J
Breard
J
BCR-ABL does not prevent apoptotic death induced by human natural killer or lymphokine-activated killer cells.
Blood
87
1996
1113
Sign in via your Institution