Tumor necrosis factor (TNF)–related apoptosis-inducing ligand (TRAIL) and Fas ligand (FasL) have been implicated in antitumor immunity and therapy. In the present study, we investigated the sensitivity of Philadelphia chromosome (Ph1)–positive leukemia cell lines to TRAIL- or FasL-induced cell death to explore the possible contribution of these molecules to immunotherapy against Ph1-positive leukemias. TRAIL, but not FasL, effectively induced apoptotic cell death in most of 5 chronic myelogenous leukemia–derived and 7 acute leukemia–derived Ph1-positive cell lines. The sensitivity to TRAIL was correlated with cell-surface expression of death-inducing receptors DR4 and/or DR5. The TRAIL-induced cell death was caspase-dependent and enhanced by nuclear factor κB inhibitors. Moreover, primary leukemia cells from Ph1-positive acute lymphoblastic leukemia patients were also sensitive to TRAIL, but not to FasL, depending on DR4/DR5 expression. Fas-associated death domain protein (FADD) and caspase-8, components of death-inducing signaling complex (DISC), as well as FLIP (FLICE [Fas-associating protein with death domain–like interleukin-1–converting enzyme]/caspase-8 inhibitory protein), a negative regulator of caspase-8, were expressed ubiquitously in Ph1-positive leukemia cell lines irrespective of their differential sensitivities to TRAIL and FasL. Notably, TRAIL could induce cell death in the Ph1-positive leukemia cell lines that were refractory to a BCR-ABL–specific tyrosine kinase inhibitor imatinib mesylate (STI571; Novartis Pharma, Basel, Switzerland). These results suggested the potential utility of recombinant TRAIL as a novel therapeutic agent and the possible contribution of endogenously expressed TRAIL to immunotherapy against Ph1-positive leukemias.

Tumor necrosis factor (TNF)–related apoptosis-inducing ligand (TRAIL) is a proapoptotic member of the TNF superfamily that also includes TNF-α and Fas ligand (FasL).1 TRAIL induces apoptosis in a variety of tumor cells2,3 by interacting with its cell-surface receptors DR4 (TRAIL-R1)4 and DR5 (TRAIL-R2).5 DR4 and DR5 contain a cytoplasmic region designated as the “death domain,” which is responsible for transducing the death signal. Ligation by TRAIL results in recruitment of a cytoplasmic adaptor molecule, Fas-associated death domain protein (FADD), to the death domain of DR4 and DR5,6 leading to activation of caspase-8, which subsequently initiates proteolytic activation of downstream effector caspases such as caspase-3, -6 and -7, and finally induces apoptosis.7 In contrast to the DR4 and DR5 receptors, 2 other cell-surface TRAIL receptors, DcR1 (TRAIL-R3)5,8 and DcR2 (TRAIL-R4),9 lack a functional death domain and cannot transduce a proapoptotic signal. These “decoy receptors” compete with DR4 and DR5 for TRAIL binding. Thus, the preferential expression of decoy receptors on normal cells is regarded as one of the mechanisms for the preferential proapoptotic action of TRAIL against tumor cells but not normal cells. In addition to the perforin- and FasL-mediated pathways,10-12 it has been recently suggested that the TRAIL-mediated pathway also plays an important role in antitumor immunity. TRAIL is expressed on human CD4+T-cell clones13 and murine activated natural killer (NK) cells,14 and is involved in their cytotoxic activities against tumor target cells. Interferon-alpha (IFN-α) enhances the TRAIL expression on anti-CD3–stimulated human peripheral blood T cells, and this is involved in their cytotoxic activities against renal cell carcinomas.15 Although there are a few reports that TRAIL selectively induced apoptosis in multiple myeloma cells16,17 and some myeloid leukemia cells,18it remains to be determined whether TRAIL also participates in antitumor immunity against hematologic malignancies.

Chronic myelogeneous leukemia (CML) is a clonal myeloproliferative expansion of transformed hematopoietic progenitor cells characterized by Philadelphia chromosome (Ph1) created from t(9;22) (q34; q11).19-21 Approximately 20% of adult22 and 5% of childhood23 acute lymphoblastic leukemia (ALL) and 2% of acute myeloblastic leukemia (AML)20 are also Ph1 positive. The Ph1 results in the juxtaposition ofbcr and abl genes, and this generates a chimeric protein termed BCR-ABL with a marked tyrosine kinase activity.19,20 Virtually all cases of CML express a 210-kDa form of BCR-ABL (p210), while half of adult and most of childhood Ph1-positive ALLs express a shorter form of BCR-ABL termed p190. IFN-α is the first-line therapy for patients with CML in chronic phase who are not eligible for allogeneic hematopoietic stem cell transplantation (allo-SCT).24-26 IFN-α can prolong survival, but complete elimination of Ph1-positive leukemia cells has been attained in only 5% to 10% of the patients. Several reports have suggested a direct antileukemic effect of IFN-α on CML cells27 and indirect effects modulating the immune system.28-30 Allo-SCT is presently an only curative therapy not only for chronic phase of CML, but also for blastic crisis of CML (CML-BC) and Ph1-positive ALL.31 A critical role for the immune system, termed graft-versus-leukemia (GVL) effect, in achieving a cure in patients with CML and Ph1-positive ALL has been well documented.20,21 

Although the clinical observations strongly suggest a pivotal role of cytotoxic T lymphocytes (CTLs) in suppressing Ph1-positive leukemias, the underlying effector mechanisms have not been well characterized. A role of FasL has been suggested, but its contribution to the elimination of Ph1-positive leukemia cells is still controversial. Selleri et al reported that Fas was expressed on CD34+cells from CML patients and up-regulated by IFN-α.28They also reported that CD34+ cells from CML patients who showed an optimal response to IFN-α therapy underwent apoptosis upon Fas triggering, whereas those derived from patients with a poor response were resistant to Fas-mediated killing.29 These results suggested a pivotal role of FasL in the elimination of CML clones by IFN-α therapy. However, Gora-Tybor et al reported that most of Ph1-positive leukemia cell lines did not express Fas and were resistant to Fas-mediated apoptosis.32 In addition, a large-scale clinical study has indicated that the higher Fas expression on CD34+ cells from CML patients was associated rather with poor cytogenetic response to IFN-α therapy.33 Moreover, using a retrovirally induced murine CML model, Shlomchik and Pear reported that GVL effect was not impaired in Fas-deficient mice, suggesting that the mechanism other than the Fas/FasL interaction may be sufficient for the immune-mediated eradication of CML clones.34 Given the IFN-α–inducible expression of TRAIL on human T cells,15 TRAIL may participate in the process of antileukemic effects against Ph1-positive leukemias. In the present study, by using Ph1-positive leukemia cell lines and primary leukemia cells, we showed that TRAIL, but not FasL, effectively induced apoptosis in Ph1-positive leukemia cells, expressing surface DR4 and/or DR5. Notably, TRAIL was also effective against leukemia cells that were refractory to the BCR-ABL kinase inhibitor imatinib mesylate (STI571; Novartis Pharma, Basel, Switzerland). These results suggest not only the potential utility of recombinant TRAIL for clinical treatment of Ph1-positive leukemias but also shed light on the possible involvement of the TRAIL/TRAIL receptor interaction in the IFN-α therapy for CML and/or the GVL effect after allo-SCT for CML and Ph1-positive ALL.

Leukemia cells

A total of 12 Ph1-positive leukemia cell lines were used in this study and are listed in Table 1. K56235 and Nalm136 were established from patients with erythroid and lymphoid crisis of CML, respectively, while other cell lines have been established in our laboratory and described previously.37 KOPM28, KOPM53, and KOPN55bi were established from CML-BC cases rearranged in major bcr, while KOPM30, KOPN30bi, KOPN57bi, KOPN66bi, KOPN72bi, and YAMN91 were established from Ph1-positive acute leukemia (AL) cases rearranged in minor bcr. YAMN73 was established from a patient with Ph1-positive ALL expressing the Abl exon 2–spliced BCR-ABL (203-kDa) protein.38 KOPM30 and KOPN30bi were established from the same patient at different stages of the disease.39 These cell lines were classified into 2 groups based on cytochemistry and immunophenotype as follows: “myeloid” type, positive for myeloperoxidase (POX) and/or expressing myeloid antigens (K562, KOPM28, KOPM30, and KOPM53); and “lymphoid” type, negative for POX and expressing immature B-lymphoid antigens (Nalm1, KOPN30bi, KOPN55bi, KOPN57bi, KOPN66bi, KOPN72bi, YAMN73, and YAMN91). T-leukemic cell lines (Jurkat and MOLT4F) were used as controls. All cell lines were maintained in RPMI1640 medium supplemented with 10% fetal calf serum (FCS) in a humidified atmosphere of 5% CO2 at 37°C. For the use of patient samples, the informed consents were obtained from the patients and/or their parents. Mononuclear cells (blasts > 95%) isolated from bone marrow aspirates or peripheral blood samples by Ficoll-Hypaque density centrifugation were stored in liquid nitrogen with 15% dimethyl sulfoxide in FCS. For experiments, each sample was thawed, washed with serum-free RPMI1640 medium, and resuspended in RPMI1640 with 10% FCS.

Table 1.

Characteristics and sensitivity to FasL in Ph1-positive leukemia cell lines

Cell lineBCR-ABLLinage3H-thymidine uptake, cpm*% inhibition by FasLExpression of Fas, RFI
ControlFasL
CML-BC-derived       
 KOPM28 p210 Myeloid 226 181 ± 2256 244 361 ± 1326 −8 3.0 
 KOPM53 p210 Myeloid 178 218 ± 1645 174 291 ± 1364 2.4 
 KOPN55bi p210 Lymphoid 8 184 ± 340 7 969 ± 493 1.0 
 Nalm1 p210 Lymphoid 40 925 ± 269 36 365 ± 1361 12 1.9 
 K562 p210 Myeloid 9 611 ± 110 9 906 ± 295 −3 1.0 
Ph1-positive AL-derived       
 KOPM30 p190 Myeloid 10 849 ± 141 7 511 ± 375 30 2.2 
 KOPN30bi p190 Lymphoid 186 993 ± 4663 180 406 ± 3438 1.1 
 KOPN57bi p190 Lymphoid 90 605 ± 2044 86 368 ± 5687 1.5 
 KOPN66bi p190 Lymphoid 117 272 ± 2663 114 914 ± 2197 1.3 
 KOPN72bi p190 Lymphoid 145 957 ± 3415 140 332 ± 4242 1.2 
 YAMN73 p2031-153 Lymphoid 60 397 ± 991 59 396 ± 1840 1.0 
 YAMN91 p190 Lymphoid 13 884 ± 246 14 143 ± 167 −2 1.3 
Cell lineBCR-ABLLinage3H-thymidine uptake, cpm*% inhibition by FasLExpression of Fas, RFI
ControlFasL
CML-BC-derived       
 KOPM28 p210 Myeloid 226 181 ± 2256 244 361 ± 1326 −8 3.0 
 KOPM53 p210 Myeloid 178 218 ± 1645 174 291 ± 1364 2.4 
 KOPN55bi p210 Lymphoid 8 184 ± 340 7 969 ± 493 1.0 
 Nalm1 p210 Lymphoid 40 925 ± 269 36 365 ± 1361 12 1.9 
 K562 p210 Myeloid 9 611 ± 110 9 906 ± 295 −3 1.0 
Ph1-positive AL-derived       
 KOPM30 p190 Myeloid 10 849 ± 141 7 511 ± 375 30 2.2 
 KOPN30bi p190 Lymphoid 186 993 ± 4663 180 406 ± 3438 1.1 
 KOPN57bi p190 Lymphoid 90 605 ± 2044 86 368 ± 5687 1.5 
 KOPN66bi p190 Lymphoid 117 272 ± 2663 114 914 ± 2197 1.3 
 KOPN72bi p190 Lymphoid 145 957 ± 3415 140 332 ± 4242 1.2 
 YAMN73 p2031-153 Lymphoid 60 397 ± 991 59 396 ± 1840 1.0 
 YAMN91 p190 Lymphoid 13 884 ± 246 14 143 ± 167 −2 1.3 
*

Data are shown as means ± SE of triplicate cultures.

% inhibition by FasL was determined by the 3H-thymidine uptake assay in the presence or absence of 100 ng/mL rhsFasL as described in “Materials and methods.”

Relative fluorescence intensity (RFI) was determined by the ratio of mean fluorescence intensity for specific staining to that for control staining.

F1-153

p203 corresponds to the Abl exon2-spliced BCR-ABL protein.

Reagents

Recombinant human soluble FasL (rhsFasL, SUPER FasL) and recombinant human soluble TRAIL (rhsTRAIL, Killer TRAIL), purchased from Alexis Biochemicals (San Diego, CA), are constituted from the extracellular domains of human FasL and human TRAIL, respectively; fused at the N-terminal to linker peptides and FRAG- and His-tags, respectively; and do not require a cross-linker for biologic activities. z-VAD-fmk, a caspase inhibitor with broad spectrum, was purchased from Enzyme Systems Products (Livermore, CA). A proteasome inhibitor N-acetyl-Leu-Leu-norLeu-al (LLnL), a cell-permeable nuclear factor κB (NF-κB) inhibitory peptide SN50,40 and its control peptide SN50M were purchased from Sigma-Aldrich (Tokyo, Japan) and BIOMOL (Plymouth Meeting, PA), respectively. Imatinib mesylate41 was kindly provided by Novartis Pharma (Basel, Switzerland).

3H-thymidine uptake assay

Leukemia cell lines (2-5 × 104 cells/well) were cultured in triplicate in 200 μL RPMI1640 medium supplemented with 10% FCS in a flat-bottomed 96-well plate (Costar, Cambridge, MA). The plates were incubated for the indicated periods, pulsed for the last 6 hours with 3H-thymidine (1 μCi/well [0.037 MBq/well]), and harvested onto glass-fiber filters. Radioactivity incorporated into DNA was measured by liquid scintillation counting.

The effects of rhsFasL and rhsTRAIL were determined by the last 6-hour pulse of the 42-hour culture in the absence or presence of 3-fold diluted concentrations (3.7, 11, 33, 100, and 300 ng/mL) of rhsFasL or rhsTRAIL. In some experiments, a neutralizing anti-TRAIL monoclonal antibody (mAb) (RIK-2; 10 μg/mL)13 or z-VAD-fmk (20 μM) was used to block the activity of rhsTRAIL and caspases, respectively. In other experiments, cell lines were preincubated for 3 hours with LLnL (2.5 μM), SN50, or SN50M (100 μg/mL) and then cultured in the absence or presence of rhsTRAIL. The effect of imatinib mesylate was also determined after 42-hour incubation in the absence or presence of imatinib mesylate (1.0 μM). The percent inhibition by TRAIL or imatinib mesylate was calculated as follows: {1 − [(cpm of treated well)/(cpm of untreated well)]} × 100. The percent3H-thymidine uptake was calculated as follows: [(cpm of treated well)/(cpm of untreated well)] × 100.

Viability and apoptosis assays

The cytotoxic effects of FasL and TRAIL were examined by the dye exclusion assay. Cell lines (1 × 105 cells/well) were cultured in the presence of rhsFasL or rhsTRAIL at 100 ng/mL, harvested at 12, 24, and 36 hours, and the viability was determined by staining with trypan blue. The early apoptotic event in leukemia cell lines was examined by binding of Annexinne-V to surface-exposed phosphatidylserine. Cell lines (4 × 105 cells/mL) were cultured in the absence or presence of rhsTRAIL (100 ng/mL), harvested at 12 hours, stained with fluorescein isothiocyanate (FITC)–conjugated Annexin-V (MBL, Nagoya, Japan), and analyzed by flow cytometry (EPICS PROFILE; Coulter, Miami, FL). In experiments with primary leukemias, cells (2 × 104 cells/well) were incubated in the absence or presence of rhsFasL or rhsTRAIL at 100 ng/mL with or without a neutralizing anti-TRAIL mAb RIK-2 (10 μg/mL) in triplicate in a 96-well plate for 24 hours, and the viability was determined by staining with trypan blue. In some patient samples, apoptosis was also examined by binding of FITC-conjugated Annexin-V after 12-hour culture.

Cell-surface expression of TRAIL receptors and Fas

mAbs specific for DR4 (DJR1, mouse immunoglobulin G 1 [IgG 1]), DR5 (DJR2, mouse IgG 1), DcR1 (DJR3, mouse IgG 1), and DcR2 (DJR4, mouse IgG 1) were raised against soluble human IgG1 Fc fusion proteins containing the extracellular domain of each TRAIL receptor and identified by their specific reactivity with the respective fusion protein in enzyme-linked immunosorbent assay (ELISA). Leukemia cell lines, primary leukemias, and baby hamster kidney (BHK) cell lines stably expressing DR4, DR5, DcR1, or DcR2 cDNA (1 × 106 cells) were incubated with 1 μg of biotinylated control mouse IgG 1 or mAb for 30 minutes on ice. After washing, the cells were incubated with phycoerythrin-conjugated streptavidin (Biomeda, Foster City, CA) for 30 minutes on ice, and then analyzed by flow cytometry. The relative fluorescence intensity (RFI) was determined by calculating the ratio of mean fluorescence intensity for specific staining to that for control staining. For the Fas expression, each cell line was incubated with mouse antihuman Fas (4A5; MBL) or irrelevant mouse IgG for 30 minutes on ice, and subsequently with FITC-conjugated anti–mouse IgG, and analyzed by flow cytometry.

Western blot analysis

The nonidet P-40 lysates of cell lines were separated on a sodium dodecyl sulfate–polyacrylamide gel under reducing conditions and then transferred to polyvinyl difluoride membranes as previously described.42 After blocking with 5% nonfat dry milk in 0.05% Tween-20 Tris (tris(hydroxymethyl)aminomethane)–buffered saline (TBS), membranes were incubated with mouse antihuman FADD (1:250 dilution; BD Transduction Laboratories, Lexington, KY), antihuman caspase-8 (1:1000 dilution; MBL), antihuman kinase domain of c-ABL (1:400 dilution; Pharmingen, San Diego, CA), or rat antihuman FLIP (FLICE [Fas-associating protein with death domain–like interleukin-1–converting enzyme]/caspase-8 inhibitory protein; 1:1000 dilution; Kamiya Biochemical, Seattle, WA) antibodies in 5% milk TBS at 4°C overnight. Membranes were incubated with horseradish peroxidase–conjugated goat antimouse or rat IgG (1:1000 dilution; MBL) at room temperature for 1 hour and were then developed using the enhanced chemiluminescence kit (Amersham Pharmacia Biotec, Buckinghamshire, United Kingdom).

Cytotoxic effect of FasL against Ph1-positive leukemia cell lines

We first examined the susceptibility of 12 Ph1-positive leukemia cell lines to rhsFasL by the 3H-thymidine uptake for the last 6 hours of the 42-hour culture with various concentrations of rhsFasL. As shown Figure 1A, a marked growth inhibition was observed in a dose-dependent manner against well-characterized FasL-sensitive T-leukemia cell lines (Jurkat and MOLT4F).43 This growth inhibition was due to loss of cell viability, rather than cytostasis, as estimated by the trypan blue exclusion assay (Figure 1B). Although 1 of 5 CML-BC–derived (Nalm1) and 1 of 7 AL-derived (KOPM30) cell lines were moderately susceptible, the other 10 Ph1-positive cell lines were highly resistant to FasL as estimated by either the 3H-thymidine uptake (Figure 1A and Table 1) or the trypan blue exclusion assay (Figure1B).

Fig. 1.

Fas expression and cytotoxic effect of FasL against Ph1-positive leukemia cell lines.

(A) Dose response of growth inhibition. CML-BC–derived (left panel) or Ph1-positive AL-derived (right panel) cell lines were cultured for 42 hours in the absence or presence of indicated concentrations of rhsFasL, and 3H-thymidine uptake was evaluated for the last 6 hours. FasL-sensitive T-cell leukemia cell lines (Jurkat and MOLT4F) were also included as positive controls and are shown by the dotted lines. Data with myeloid cell lines are shown as the closed symbols. All data are represented as the mean of triplicate samples. Standard errors were always less than 10% (not shown). (B) Time course of cell viability. CML-BC–derived (left panel) or Ph1-positive AL-derived (right panel) cell lines were cultured with or without 100 ng/mL rhsFasL for 12, 24, or 36 hours, and then the viability was determined by the trypan blue exclusion assay. Data with Jurkat and MOLT4F are also shown by dotted lines. Data with myeloid cell lines are shown as closed symbols. All data are represented as the mean of triplicate samples. Standard errors were always less than 10% (not shown). (C) Cell-surface staining of Ph1-positive cell lines with anti-Fas mAb. CML-BC–derived FasL-sensitive (Nalm1) or -resistant (KOPM53) cell lines and Ph1-positive AL-derived FasL-sensitive (KOPM30) or -resistant (KOPN30bi) cell lines were stained with control mouse IgG or antihuman Fas mAb and analyzed by flow cytometry. Shaded and unshaded peaks correspond to specific and control stainings, respectively. RFI is indicated in each panel.

Fig. 1.

Fas expression and cytotoxic effect of FasL against Ph1-positive leukemia cell lines.

(A) Dose response of growth inhibition. CML-BC–derived (left panel) or Ph1-positive AL-derived (right panel) cell lines were cultured for 42 hours in the absence or presence of indicated concentrations of rhsFasL, and 3H-thymidine uptake was evaluated for the last 6 hours. FasL-sensitive T-cell leukemia cell lines (Jurkat and MOLT4F) were also included as positive controls and are shown by the dotted lines. Data with myeloid cell lines are shown as the closed symbols. All data are represented as the mean of triplicate samples. Standard errors were always less than 10% (not shown). (B) Time course of cell viability. CML-BC–derived (left panel) or Ph1-positive AL-derived (right panel) cell lines were cultured with or without 100 ng/mL rhsFasL for 12, 24, or 36 hours, and then the viability was determined by the trypan blue exclusion assay. Data with Jurkat and MOLT4F are also shown by dotted lines. Data with myeloid cell lines are shown as closed symbols. All data are represented as the mean of triplicate samples. Standard errors were always less than 10% (not shown). (C) Cell-surface staining of Ph1-positive cell lines with anti-Fas mAb. CML-BC–derived FasL-sensitive (Nalm1) or -resistant (KOPM53) cell lines and Ph1-positive AL-derived FasL-sensitive (KOPM30) or -resistant (KOPN30bi) cell lines were stained with control mouse IgG or antihuman Fas mAb and analyzed by flow cytometry. Shaded and unshaded peaks correspond to specific and control stainings, respectively. RFI is indicated in each panel.

Close modal

These results suggested that Ph1-positive leukemia cell lines are generally resistant to the FasL-induced cell death. Next, we analyzed the cell-surface expression of Fas by flow cytometry. As indicated in Figure 1C and summarized in Table 1, Fas was detectable on myeloid cell lines except for K562 but rarely detectable on lymphoid cell lines except for Nalm1. Accordingly, the relatively low expression of Fas could explain the resistance to FasL in lymphoid cell lines, while some factor other than the Fas expression might contribute to the resistance to FasL in myeloid cell lines.

Cytotoxic effect of TRAIL against Ph1-positive leukemia cell lines

We next investigated the cytotoxic effect of rhsTRAIL against Ph1-positive leukemia cell lines. Among the CML-BC–derived cell lines (Figure 2A, left panel), 2 cell lines (Nalm1 and KOPM28) showed a marked growth inhibition in a dose-dependent manner, whereas the other 3 cell lines (KOPM53, KOPN55bi, and K562) were rather resistant. Among the Ph1-positive AL-derived cell lines (Figure 2A, right panel), 3 cell lines (YAMN91, KOPN66bi, and KOPM30) were highly sensitive, 2 cell lines (YAMN73 and KOPN72bi) were moderately sensitive, but 2 cell lines (KOPN57bi and KOPN30bi) were resistant. To demonstrate that these antileukemic effects were really mediated by TRAIL, we performed the blocking experiment using a neutralizing anti-TRAIL mAb (RIK-2).13 As shown in Figure 2B, the growth inhibition in Nalm1 was totally abolished by RIK-2, substantiating the specific activity of rhsTRAIL. As summarized in Table2, the sensitivity of Ph1-positive leukemia cell lines to TRAIL was not correlated with the type of disease (CML-BC or AL), the type of BCR-ABL fusion protein (p210, p203, or p190), or the type of lineage (myeloid or lymphoid).

Fig. 2.

Cytotoxic effect of TRAIL against Ph1-positive leukemia cell lines.

(A) Dose response of growth inhibition. CML-BC–derived (left panel) or Ph1-positive AL-derived (right panel) cell lines were cultured for 42 hours in the absence or presence of the indicated concentrations of rhsTRAIL, and the 3H-thymidine uptake was evaluated for the last 6 hours. T-cell leukemia cell lines (Jurkat and MOLT4F) were also included as controls and are shown by the dotted lines. Data with myeloid cell lines are shown as the closed symbols. All data are represented as the mean of triplicate samples. Standard errors were always less than 10% (not shown). (B) Neutralization by anti-TRAIL mAb. Nalm1 cells were cultured for 42 hours with rhsTRAIL (10 ng/mL) in the presence or absence of neutralizing anti-TRAIL mAb (10 μg/mL), and the 3H-thymidine uptake was evaluated for the last 6 hours. Data are represented as the mean ± SD of triplicate samples. (C) Time course of cell viability. CML-BC–derived (left panel) or Ph1-positive AL-derived (right panel) cell lines were cultured with 100 ng/mL of rhsTRAIL for 12, 24, or 36 hours, and then the viability was determined by the trypan blue exclusion assay. Data with Jurkat and MOLT4F are also shown by the dotted lines. Data with myeloid cell lines are shown as the closed symbols. All data are represented as the mean of triplicate samples. Standard errors were always less than 10% (not shown).

Fig. 2.

Cytotoxic effect of TRAIL against Ph1-positive leukemia cell lines.

(A) Dose response of growth inhibition. CML-BC–derived (left panel) or Ph1-positive AL-derived (right panel) cell lines were cultured for 42 hours in the absence or presence of the indicated concentrations of rhsTRAIL, and the 3H-thymidine uptake was evaluated for the last 6 hours. T-cell leukemia cell lines (Jurkat and MOLT4F) were also included as controls and are shown by the dotted lines. Data with myeloid cell lines are shown as the closed symbols. All data are represented as the mean of triplicate samples. Standard errors were always less than 10% (not shown). (B) Neutralization by anti-TRAIL mAb. Nalm1 cells were cultured for 42 hours with rhsTRAIL (10 ng/mL) in the presence or absence of neutralizing anti-TRAIL mAb (10 μg/mL), and the 3H-thymidine uptake was evaluated for the last 6 hours. Data are represented as the mean ± SD of triplicate samples. (C) Time course of cell viability. CML-BC–derived (left panel) or Ph1-positive AL-derived (right panel) cell lines were cultured with 100 ng/mL of rhsTRAIL for 12, 24, or 36 hours, and then the viability was determined by the trypan blue exclusion assay. Data with Jurkat and MOLT4F are also shown by the dotted lines. Data with myeloid cell lines are shown as the closed symbols. All data are represented as the mean of triplicate samples. Standard errors were always less than 10% (not shown).

Close modal
Table 2.

TRAIL-sensitivity and cell-surface expression of TRAIL receptors in Ph1-positive leukemia cell lines

Cell line3H-thymidine uptake, cpm*% inhibition by TRAILExpression of TRAIL receptors, RFI
ControlTRAILDR4DR5DcR1DcR2
CML-BC-derived        
 KOPM28 103 712 ± 1849 6 000 ± 54 95 1.0 3.4 1.1 1.0 
 KOPM53 200 509 ± 9527 165 725 ± 8340 18 1.1 1.0 1.0 1.0 
 KOPN55bi 11 705 ± 1730 9 954 ± 398 10 1.0 1.3 1.1 1.1 
 Nalm1 28 920 ± 621 167 ± 14 99 1.7 2.1 1.0 1.0 
 K562 12 099 ± 213 10 034 ± 61 20 1.5 5.0 1.0 1.0 
Ph1-positive AL-derived        
 KOPM30 29 182 ± 1796 7 055 ± 368 75 1.6 3.2 1.0 1.0 
 KOPN30bi 114 528 ± 4592 116 241 ± 2666 −1 1.0 1.7 1.1 1.1 
 KOPN57bi 40 975 ± 2021 35 681 ± 2001 1.0 1.3 1.0 1.1 
 KOPN66bi 75 603 ± 1175 9 031 ± 157 85 1.5 2.6 1.0 1.0 
 KOPN72bi 112 855 ± 5916 68 357 ± 4469 52 1.7 1.1 1.0 1.0 
 YAMN73 36 437 ± 716 20 553 ± 498 45 1.2 2.9 1.2 1.0 
 YAMN91 27 788 ± 1529 1 062 ± 79 96 1.0 3.4 1.1 1.0 
Cell line3H-thymidine uptake, cpm*% inhibition by TRAILExpression of TRAIL receptors, RFI
ControlTRAILDR4DR5DcR1DcR2
CML-BC-derived        
 KOPM28 103 712 ± 1849 6 000 ± 54 95 1.0 3.4 1.1 1.0 
 KOPM53 200 509 ± 9527 165 725 ± 8340 18 1.1 1.0 1.0 1.0 
 KOPN55bi 11 705 ± 1730 9 954 ± 398 10 1.0 1.3 1.1 1.1 
 Nalm1 28 920 ± 621 167 ± 14 99 1.7 2.1 1.0 1.0 
 K562 12 099 ± 213 10 034 ± 61 20 1.5 5.0 1.0 1.0 
Ph1-positive AL-derived        
 KOPM30 29 182 ± 1796 7 055 ± 368 75 1.6 3.2 1.0 1.0 
 KOPN30bi 114 528 ± 4592 116 241 ± 2666 −1 1.0 1.7 1.1 1.1 
 KOPN57bi 40 975 ± 2021 35 681 ± 2001 1.0 1.3 1.0 1.1 
 KOPN66bi 75 603 ± 1175 9 031 ± 157 85 1.5 2.6 1.0 1.0 
 KOPN72bi 112 855 ± 5916 68 357 ± 4469 52 1.7 1.1 1.0 1.0 
 YAMN73 36 437 ± 716 20 553 ± 498 45 1.2 2.9 1.2 1.0 
 YAMN91 27 788 ± 1529 1 062 ± 79 96 1.0 3.4 1.1 1.0 
*

Data are shown as means ± SE.

% inhibition by TRAIL was determined by the 3H-thymidine uptake assay in the presence or absence of 100 ng/mL rhsTRAIL as described in “Materials and methods.”

RFI was determined by the ratio of mean fluorescence intensity for specific staining to that for control staining.

To more directly evaluate the cytotoxic activity of TRAIL, we performed the dye exclusion assay (Figure 2C).

Consistent with the growth inhibition as estimated by the3H-thymidine uptake assay, 7 of 12 Ph1-positive leukemia cell lines were moderately or highly sensitive to the TRAIL-induced cell death.

We also examined the binding of Annexin-V by flow cytometry after 12-hour treatment with rhsTRAIL (100 ng/mL). As shown in Figure 3, the Annexin-V–positive population was increased to 95% in the highly sensitive cell line (KOPM28), 72% in the moderately sensitive cell line (YAMN73), but only 13% in the resistant cell line (KOPM53), indicating that the TRAIL-induced cell death in Ph1-positive leukemia cell lines was caused by apoptosis.

Fig. 3.

Flow cytometric analysis of TRAIL-induced apoptosis.

TRAIL-sensitive cell lines (KOPM28 and YAMN73) and TRAIL-resistant cell line (KOPM53) were cultured for 12 hours with or without rhsTRAIL (100 ng/mL) and then stained with FITC-conjugated Annexin-V. The percentages of positive cells are indicated.

Fig. 3.

Flow cytometric analysis of TRAIL-induced apoptosis.

TRAIL-sensitive cell lines (KOPM28 and YAMN73) and TRAIL-resistant cell line (KOPM53) were cultured for 12 hours with or without rhsTRAIL (100 ng/mL) and then stained with FITC-conjugated Annexin-V. The percentages of positive cells are indicated.

Close modal

To confirm that the apoptotic cell death induced by TRAIL was dependent on the activation of caspases, we performed the 3H-thymidine uptake assay in the presence of z-VAD-fmk, a broad caspase inhibitor. As shown in Figure4, the TRAIL-induced growth inhibition was abrogated by z-VAD-fmk partially in Nalm1 and almost completely in KOPM28, indicating that the activation of caspases was required for the TRAIL-induced cell death in Ph1-positive leukemia cells.

Fig. 4.

Effect of a caspase inhibitor on growth inhibition by TRAIL.

Ph1-positive cell lines sensitive to TRAIL (Nalm1 and KOPM28) were cultured for 30 hours in the absence or presence of rhsTRAIL (25 and 50 ng/mL, respectively) with or without z-VAD-fmk (20 μM) pretreatment, and the 3H-thymidine uptake was determined for the last 6 hours. Data are represented as the mean ± SD of triplicate samples.

Fig. 4.

Effect of a caspase inhibitor on growth inhibition by TRAIL.

Ph1-positive cell lines sensitive to TRAIL (Nalm1 and KOPM28) were cultured for 30 hours in the absence or presence of rhsTRAIL (25 and 50 ng/mL, respectively) with or without z-VAD-fmk (20 μM) pretreatment, and the 3H-thymidine uptake was determined for the last 6 hours. Data are represented as the mean ± SD of triplicate samples.

Close modal

Expression of TRAIL receptors on Ph1-positive leukemia cell lines

To investigate whether the sensitivity of Ph1-positive leukemia cells to TRAIL depends on the expression of TRAIL receptors, we analyzed the cell-surface expression of DR4, DR5, DcR1, and DcR2 by flow cytometry. The specificity of each mAb is shown against BHK cell lines transfected with respective TRAIL receptor cDNAs (Figure5A).

Fig. 5.

Cell-surface expression of TRAIL receptors on Ph1-positive leukemia cell lines.

(A) Specificity of mAbs against human TRAIL receptors. The parental BHK and the transfectants stably expressing human DR4, DR5, DcR1, or DcR2 were stained with control mouse IgG or murine antihuman DR4, DR5, DcR1, or DcR2 mAbs and analyzed by flow cytometry. Dotted and solid lines correspond to control and specific stainings, respectively. (B) Cell-surface staining of Ph1-positive cell lines with anti-TRAIL receptor mAbs. CML-BC–derived TRAIL-sensitive (KOPM28) or -resistant (KOPM53) cell lines and Ph1-positive AL-derived TRAIL-sensitive cell lines (KOPM30 and KOPN72bi) were stained with control mouse IgG or antihuman DR4, DR5, DcR1, and DcR2 mAbs, and analyzed by flow cytometry. Shaded and unshaded peaks correspond to specific and control stainings, respectively. RFI is indicated in each panel. (C) Correlation between the cell-surface DR4/DR5 expression and the growth inhibition by TRAIL. The horizontal axes represent the RFI of DR4 and DR5. The vertical axes represent the percent inhibition by TRAIL (100 ng/mL).

Fig. 5.

Cell-surface expression of TRAIL receptors on Ph1-positive leukemia cell lines.

(A) Specificity of mAbs against human TRAIL receptors. The parental BHK and the transfectants stably expressing human DR4, DR5, DcR1, or DcR2 were stained with control mouse IgG or murine antihuman DR4, DR5, DcR1, or DcR2 mAbs and analyzed by flow cytometry. Dotted and solid lines correspond to control and specific stainings, respectively. (B) Cell-surface staining of Ph1-positive cell lines with anti-TRAIL receptor mAbs. CML-BC–derived TRAIL-sensitive (KOPM28) or -resistant (KOPM53) cell lines and Ph1-positive AL-derived TRAIL-sensitive cell lines (KOPM30 and KOPN72bi) were stained with control mouse IgG or antihuman DR4, DR5, DcR1, and DcR2 mAbs, and analyzed by flow cytometry. Shaded and unshaded peaks correspond to specific and control stainings, respectively. RFI is indicated in each panel. (C) Correlation between the cell-surface DR4/DR5 expression and the growth inhibition by TRAIL. The horizontal axes represent the RFI of DR4 and DR5. The vertical axes represent the percent inhibition by TRAIL (100 ng/mL).

Close modal

Representative cytofluorographic data on highly sensitive (KOPM28, KOPM30), moderately sensitive (KOPN72bi), and resistant (KOPM53) cell lines are shown in Figure 5B, and the RFI in each cell line is summarized in Table 2. Among 4 TRAIL receptors, DR4 and DR5 were detectable (RFI ≤ 1.5) on 5 and 8, respectively, of 12 Ph1-positive leukemic cell lines. DcR1 and DcR2 were not detectable on any tested cell lines.

Correlation between the cell-surface DR4/DR5 expression and the percent inhibition by TRAIL (Figure 5C) revealed that all TRAIL-sensitive cell lines expressed DR4 and/or DR5 at significant levels. In contrast, the TRAIL-resistant cell lines except for K562 showed undetectable or low levels of DR4 and DR5. These results suggested that the sensitivity of Ph1-positive leukemia cells to TRAIL is mostly correlated with the cell-surface expression levels of DR4 and DR5.

Cytotoxic effects of TRAIL and FasL against primary Ph1-positive leukemia cells

To verify the antileukemic effects of TRAIL and FasL against primary leukemia cells, leukemic blasts from 10 Ph1-positive ALL cases and 2 CML-BC cases were tested. Each sample was cultured for 24 hours in the absence or presence of 100 ng/mL of rhsFasL or rhsTRAIL in combination with a neutralizing anti-TRAIL mAb, RIK-2, and the viability was determined by the trypan blue exclusion assay. As summarized in Table 3, viability of the cells was significantly reduced by the addition of TRAIL in 6 of 10 Ph1-positive ALL cases, while only 1 case was moderately sensitive to FasL. The specific activity of TRAIL was demonstrated by the significant recovery of viability with the RIK-2 treatment. Induction of apoptosis by TRAIL was also confirmed in leukemic blasts from patients 8 and 9 by Annexin-V binding on flow cytometry (Figure 6A). Importantly, primary leukemia cells from patient 3 were sensitive to TRAIL, while the cell line (KOPN57bi) established from this patient was resistant to TRAIL (Figure 2A and Table 2). Leukemic blasts from patients 4 and 8, the origin of TRAIL-sensitive YAMN91 and YAMN73, respectively, were sensitive to TRAIL, while those from patient 2, the origin of TRAIL-resistant KOPN30bi, were resistant. Regarding TRAIL sensitivity in leukemic blasts from CML-BC, only one case (patient 11) was evaluated and showed resistance.

Table 3.

Sensitivity to FasL/TRAIL and TRIL receptors expression in primary Ph1-positive leukemia cells

Patient no.SexAgeDerived cell lineOnset/RelapseSampleType of BCR-ABLViability after 24-h culture3-150Expression
of TRAIL receptors, RFI3-152
Control, %FasL, %TRAIL, %TRAIL + RIK2, %DR4DR5
Ph1-ALL             
  1 — Onset BM p190 89 ± 2 91 ± 1 90 ± 3 90 ± 1 1.0 1.2 
  2 KOPN30bi Onset PB p190 75 ± 4 71 ± 6 77 ± 4 78 ± 2 1.0 1.1 
  3 11 KOPN57bi Onset PB p190 84 ± 1 77 ± 2 34 ± 53-151 72 ± 33-151,3-153 ND ND 
  4 YAMN91 Onset PB p190 80 ± 4 76 ± 1 52 ± 33-151 80 ± 23-153 1.0 1.5 
  5 12 — Onset BM p190 85 ± 1 93 ± 2 90 ± 4 95 ± 2 1.0 1.1 
  6 16 — Relapse PB p210 85 ± 1 95 ± 3 90 ± 3 95 ± 2 ND ND 
  7 — Relapse BM p190 89 ± 1 90 ± 1 62 ± 53-151 86 ± 23-153 ND ND 
  8 YAMN73 Relapse PB p2033-155 87 ± 1 69 ± 23-151 46 ± 33-151 78 ± 33-151,3-153 1.2 1.6 
  9 — Relapse PB p190 92 ± 2 87 ± 3 46 ± 33-151 88 ± 13-153 1.5 2.0 
 10 15 — Relapse BM ND 93 ± 3 83 ± 3 66 ± 33-151 77 ± 23-151,3-153 ND ND 
CML-BC             
 11 — — PB p210 91 ± 4 91 ± 3 88 ± 4 92 ± 2 ND ND 
 12 11 — — PB p210 NA# NA NA NA 2.2 2.9 
Patient no.SexAgeDerived cell lineOnset/RelapseSampleType of BCR-ABLViability after 24-h culture3-150Expression
of TRAIL receptors, RFI3-152
Control, %FasL, %TRAIL, %TRAIL + RIK2, %DR4DR5
Ph1-ALL             
  1 — Onset BM p190 89 ± 2 91 ± 1 90 ± 3 90 ± 1 1.0 1.2 
  2 KOPN30bi Onset PB p190 75 ± 4 71 ± 6 77 ± 4 78 ± 2 1.0 1.1 
  3 11 KOPN57bi Onset PB p190 84 ± 1 77 ± 2 34 ± 53-151 72 ± 33-151,3-153 ND ND 
  4 YAMN91 Onset PB p190 80 ± 4 76 ± 1 52 ± 33-151 80 ± 23-153 1.0 1.5 
  5 12 — Onset BM p190 85 ± 1 93 ± 2 90 ± 4 95 ± 2 1.0 1.1 
  6 16 — Relapse PB p210 85 ± 1 95 ± 3 90 ± 3 95 ± 2 ND ND 
  7 — Relapse BM p190 89 ± 1 90 ± 1 62 ± 53-151 86 ± 23-153 ND ND 
  8 YAMN73 Relapse PB p2033-155 87 ± 1 69 ± 23-151 46 ± 33-151 78 ± 33-151,3-153 1.2 1.6 
  9 — Relapse PB p190 92 ± 2 87 ± 3 46 ± 33-151 88 ± 13-153 1.5 2.0 
 10 15 — Relapse BM ND 93 ± 3 83 ± 3 66 ± 33-151 77 ± 23-151,3-153 ND ND 
CML-BC             
 11 — — PB p210 91 ± 4 91 ± 3 88 ± 4 92 ± 2 ND ND 
 12 11 — — PB p210 NA# NA NA NA 2.2 2.9 

M indicates male; F, female; BM, bone marrow; PB, peripheral blood; ND, not determined; NA, not available; and —, not applicable.

F3-150

Data are shown as mean ± SE.

F3-151

Significant reduction in viability (P < .01,t test).

F3-152

RFI was determined by the ratio of mean fluorescence intensity for specific staining to that for control staining.

F3-153

Significant increase in viability (P < .01,t test) in comparison with TRAIL treatment alone.

F3-155

p203 corresponds to the Abl exon 2-spliced BCR-ABL protein.

#Sensitivity could not be evaluated because of complete spontaneous cell death in control culture.

Fig. 6.

Flow cytometric analysis of TRAIL-induced apoptosis and cell-surface expression of DR4 and DR5 in primary Ph1-positive ALL cells.

(A) Leukemic blasts from patients 8 and 9 listed in Table 3 were cultured for 12 hours with or without rhsTRAIL (100 ng/mL) in the presence or absence of neutralizing anti-TRAIL mAb RIK-2 (10 μg/mL), and then stained with FITC-conjugated Annexin-V. The percentages of positive cells are indicated in each panel. (B) Cell-surface staining of Ph1-positive primary leukemia cells with anti-DR4 and DR5 mAbs. Leukemic blasts from Ph1-positive ALL cases (patients 1, 2, 8, and 9 listed in Table 3) and a CML-BC case (patient 12) were stained with control mouse IgG or antihuman DR4 and DR5 mAbs, and analyzed by flow cytometry. Shaded and unshaded peaks correspond to specific and control stainings, respectively. Relative fluorescence intensity (RFI) is indicated in each panel.

Fig. 6.

Flow cytometric analysis of TRAIL-induced apoptosis and cell-surface expression of DR4 and DR5 in primary Ph1-positive ALL cells.

(A) Leukemic blasts from patients 8 and 9 listed in Table 3 were cultured for 12 hours with or without rhsTRAIL (100 ng/mL) in the presence or absence of neutralizing anti-TRAIL mAb RIK-2 (10 μg/mL), and then stained with FITC-conjugated Annexin-V. The percentages of positive cells are indicated in each panel. (B) Cell-surface staining of Ph1-positive primary leukemia cells with anti-DR4 and DR5 mAbs. Leukemic blasts from Ph1-positive ALL cases (patients 1, 2, 8, and 9 listed in Table 3) and a CML-BC case (patient 12) were stained with control mouse IgG or antihuman DR4 and DR5 mAbs, and analyzed by flow cytometry. Shaded and unshaded peaks correspond to specific and control stainings, respectively. Relative fluorescence intensity (RFI) is indicated in each panel.

Close modal

Next, the cell-surface expression of DR4 and DR5 was analyzed by flow cytometry as indicated in Figure 6B and Table 3. In Ph1-positive ALL cases, the expression of DR4/DR5 was detectable on TRAIL-sensitive leukemia cells from patients 4, 8, and 9, but almost undetectable on TRAIL-resistant leukemia cells from patients 1, 2, and 5. In the CML-BC case (patient 12), both DR4 and DR5 were clearly detectable, although its TRAIL-sensitivity could not be evaluated because of excessive spontaneous cell death in control culture.

Expression of molecules consisting of death-inducing signaling complex (DISC)

Ligation of death receptors by FasL44 and TRAIL45,46 triggers a series of protein-protein interactions that leads to assembly of a DISC. Thus, expression levels of the molecules consisting of DISC are critical determinants for sensitivity. It is known that Fas and DR4/DR5 recruit FADD47,48 and caspase-849,50 into DISC, and that FLIP51 acts as a negative regulator of caspase-8.

We therefore performed Western blot analysis of these molecules in Ph1-positive leukemia cell lines (Figure7). FADD, caspase-8, and FLIP were almost ubiquitously expressed in Ph1-positive leukemia cell lines irrespective of their differential sensitivities to FasL and TRAIL. These observations indicated that an absence of FADD and/or caspase-8 or an excessive expression of FLIP could not be a mechanism for resistance to TRAIL and FasL.

Fig. 7.

Western blot analysis of molecules consisting of DISC in Ph1-positive leukemia cell lines.

Lysates from 12 Ph1-positive leukemia cell lines were analyzed by Western blot using antibodies against c-ABL kinase domain, FADD, caspase-8, and FLIP as described in “Materials and methods.” Small arrows and arrowheads indicate BCR-ABL proteins (p210, p203, and p190) and c-ABL protein, respectively, in panel A. Large arrows indicate FADD in panel B, caspase-8 in panel C, and FLIP in panel D.

Fig. 7.

Western blot analysis of molecules consisting of DISC in Ph1-positive leukemia cell lines.

Lysates from 12 Ph1-positive leukemia cell lines were analyzed by Western blot using antibodies against c-ABL kinase domain, FADD, caspase-8, and FLIP as described in “Materials and methods.” Small arrows and arrowheads indicate BCR-ABL proteins (p210, p203, and p190) and c-ABL protein, respectively, in panel A. Large arrows indicate FADD in panel B, caspase-8 in panel C, and FLIP in panel D.

Close modal

Modulation of TRAIL sensitivity by NF-κB inhibitors

It has been reported that NF-κB is constitutively activated by BCR-ABL in Ph1-positive leukemias52 and that NF-κB could modulate TRAIL-induced cell death.6,9,53 We attempted to examine whether the NF-κB inhibitors could modulate the TRAIL sensitivity of Ph1-positive leukemia cells. For this purpose, 10 leukemia cell lines (6 sensitive and 4 resistant) were preincubated for 3 hours with 2.5 μM proteasome inhibitor LLnL,54 which is known to inhibit the activation of NF-κB by blocking the degradation of the IκB inhibitory protein, followed by 42-hour exposure to lower concentrations of rhsTRAIL (10 ng/mL for sensitive and 50 ng/mL for resistant cell lines), and then the3H-thymidine uptake was assessed for the last 6 hours (Figure 8A). As expected from a considerable role of NF-κB in the BCR-ABL–mediated transformation, the treatment with LLnL alone moderately repressed the3H-thymidine uptake in most of Ph1-positive leukemia cell lines. In the TRAIL-sensitive cell line KOPM28, for instance, the treatment with either TRAIL or LLnL reduced the3H-thymidine uptake to approximately 70% or 50%, respectively, but the treatment with both TRAIL and LLnL almost completely abrogated the 3H-thymidine uptake. Similar results were obtained in all TRAIL-sensitive cell lines examined. In contrast, LLnL did not enhance the proapoptotic activity of TRAIL in all TRAIL-resistant cell lines, including K562, which considerably expressed DR5. These results suggested that the inhibition of NF-κB activation by LLnL either augmented the TRAIL sensitivity or synergistically acted with TRAIL in the process of apoptosis induction, but could not convert the TRAIL-resistant cells to be TRAIL sensitive.

Fig. 8.

Modulation of TRAIL sensitivity by NF-κB inhibitors.

(A) Effect of LLnL. TRAIL-sensitive or resistant cell lines were cultured for 42 hours in the absence or presence of rhsTRAIL (10 ng/mL for sensitive cell lines, 50 ng/mL for resistant cell lines) with or without LLnL (2.5 μM) pretreatment, and the 3H-thymidine uptake was determined for the last 6 hours. Data are represented as the mean ± SD of triplicate sample. (B) Effect of SN50. The TRAIL-sensitive cell line KOPM28 was cultured for 42 hours in the absence or presence of rhsTRAIL (10 ng/mL) with or without SN50 or SN50M (100 μg/mL) pretreatment, and the 3H-thymidine uptake was determined for the last 6 hours. Data are represented as the mean ± SD of triplicate sample.

Fig. 8.

Modulation of TRAIL sensitivity by NF-κB inhibitors.

(A) Effect of LLnL. TRAIL-sensitive or resistant cell lines were cultured for 42 hours in the absence or presence of rhsTRAIL (10 ng/mL for sensitive cell lines, 50 ng/mL for resistant cell lines) with or without LLnL (2.5 μM) pretreatment, and the 3H-thymidine uptake was determined for the last 6 hours. Data are represented as the mean ± SD of triplicate sample. (B) Effect of SN50. The TRAIL-sensitive cell line KOPM28 was cultured for 42 hours in the absence or presence of rhsTRAIL (10 ng/mL) with or without SN50 or SN50M (100 μg/mL) pretreatment, and the 3H-thymidine uptake was determined for the last 6 hours. Data are represented as the mean ± SD of triplicate sample.

Close modal

A similar experiment was also performed in KOPM28 using another NF-κB–specific inhibitor, SN50, which blocks the nuclear translocation of NF-κB.40 As shown in Figure 8B, SN50 (100 μg/mL), but not its mutant control peptide SN50M, reduced the 3H-thymidine uptake to nearly 60% by itself and enhanced sensitivity to TRAIL, further substantiating the modulation of TRAIL sensitivity by NF-κB.

Comparison of antileukemic effects of TRAIL and imatinib mesylate

Imatinib mesylate is a specific inhibitor of BCR-ABL tyrosine kinase activity41 and shows a potent cytotoxic effect on Ph1-positive leukemias.55,56 In a clinical trial, imatinib mesylate was reported to be very effective in CML patients who were resistant to the IFN-α therapy.55 

Thus, we compared the cytotoxic effects of TRAIL and imatinib mesylate against 12 Ph1-positive cell lines (Figure9). All these cell lines were established from the patients who had not been treated with imatinib mesylate and were not selected by imatinib mesylate treatment in vitro. All myeloid cell lines (KOPM28, KOPM30, KOPM53, and K562; closed symbols) showed high sensitivity to imatinib mesylate (percent inhibition, > 60%). In contrast, only 1 of 8 lymphoid cell lines (KOPN57bi) was highly sensitive, and 4 lymphoid cell lines (Nalm1, KOPN66bi, YAMN73, and YAMN91) were naturally resistant to imatinib mesylate (percent inhibition, < 40%). Two myeloid cell lines (KOPM28 and KOPM30) showed high sensitivity to both TRAIL and imatinib mesylate. The 4 lymphoid cell lines resistant to imatinib mesylate were sensitive to TRAIL, while 5 cell lines (myeloid 2; lymphoid 3) were resistant to TRAIL but sensitive to imatinib mesylate. Most importantly, none of 12 cell lines were resistant to both TRAIL and imatinib mesylate. These results suggested that TRAIL and imatinib mesylate could be used complementarily to eliminate Ph1-positive leukemia cells.

Fig. 9.

Correlation between percent inhibition by TRAIL and percent inhibition by imatinib mesylate.

The indicated cell lines were cultured for 42 hours in the presence or absence of rhsTRAIL (100 ng/mL) or imatinib mesylate (1.0 μM), and the 3H-thymidine uptake was assessed for the last 6 hours. Data on myeloid cell lines are shown by closed symbols.

Fig. 9.

Correlation between percent inhibition by TRAIL and percent inhibition by imatinib mesylate.

The indicated cell lines were cultured for 42 hours in the presence or absence of rhsTRAIL (100 ng/mL) or imatinib mesylate (1.0 μM), and the 3H-thymidine uptake was assessed for the last 6 hours. Data on myeloid cell lines are shown by closed symbols.

Close modal

In the present study, to explore the possible contribution of FasL and TRAIL to the immune-mediated antileukemic effects against Ph1-positive leukemia cells, we first investigated whether FasL or TRAIL could induce cell death in 12 Ph1-positive leukemia cell lines. Consistent with a previous report,32 most of the Ph1-positive cell lines were resistant to FasL. In contrast, 2 of 5 CML-BC–derived and 5 of 7 Ph1-AL–derived cell lines were highly or moderately sensitive to TRAIL-induced apoptotic cell death. Similar results were obtained with primary Ph1-positive ALL cells. Consistent with our data, Plasilova et al recently reported that the growth of CML progenitors in chronic phase as well as in accelerated or blastic phases was significantly suppressed by recombinant TRAIL.57 

We next analyzed the cell-surface expression of TRAIL receptors to understand the differences in TRAIL sensitivity among the Ph1-positive leukemia cell lines. All TRAIL-sensitive cell lines and primary cells expressed the death-inducing receptors DR4 and/or DR5 on their surface, whereas the TRAIL-resistant cell lines, except for K562, and primary cells expressed neither DR4 nor DR5. In addition, none of these cell lines expressed DcR1 and DcR2, which are thought to act as decoy receptors. These results suggested that the sensitivity of Ph1-positive leukemia cells to TRAIL is primarily determined by the cell-surface expression of DR4 and/or DR5.

In addition to the receptor expression, regulators in the death-signaling pathway would be critical for the determination of sensitivity to FasL and TRAIL. We demonstrated that caspase-8, FADD, and FLIP were ubiquitously expressed in Ph1-positive leukemia cell lines irrespective of their differential sensitivities to FasL and TRAIL, suggesting that these molecules are not critically involved in the determination of sensitivity to FasL and TRAIL at least in their expression levels. We observed a marked discrepancy between sensitivity to TRAIL and FasL in several Ph1-positive cell lines despite expression of both death receptors. In particular, KOPM28 expressing both DR5 and Fas at highest levels showed a high sensitivity to TRAIL but not to FasL. Recently, a similar discrepancy has also been documented in some solid tumors.58,59 In addition, distinct intracellular signaling pathways in TRAIL- and FasL-mediated apoptosis have been reported.60 Thus, KOPM28 would serve as a useful subject to explore distinct intracellular signaling via TRAIL and FasL in further studies.

It has been demonstrated that the binding of TRAIL to DR4 and DR5 as well as DcR2 induced the NF-κB activation,6,9,53 and that the increased NF-κB activity protects tumor cells from various proapoptotic stimuli.62 Therefore, we examined whether the NF-κB activity could modulate the sensitivity to TRAIL in Ph1-positive leukemia cells. The pretreatment with NF-κB inhibitors, LLnL and SN50, enhanced sensitivity to TRAIL in the TRAIL-sensitive cell lines, suggesting that the DR4/DR5-mediated or BCR-ABL–mediated NF-κB activation plays a substantial role in protecting Ph1-positive leukemia cells from TRAIL-induced cell death. Of importance, in TRAIL-resistant cell lines not expressing DR4 or DR5, NF-κB inhibitors could not overcome their TRAIL resistance. Since a proteasome inhibitor that inhibits NF-κB activation (PS-341) has already been in phase 3 clinical trials against multiple myelomas,62,63 the combination with PS-341 would be a promising way to enhance the TRAIL-mediated elimination of Ph1-positive leukemia cells in a clinical setting.

We previously demonstrated that IFN-α up-regulated the cell-surface expression of TRAIL, on TCR/CD3-stimulated T cells in vitro, which mediated cytotoxicity against TRAIL-sensitive renal cell carcinomas.15 In addition, Wen et al reported that antileukemic agents including Ara-C increased the expression levels of DR5 on leukemia cell lines.64 Moreover, it has been reported that a combination of Ara-C and IFN-α increased the rate of remission and prolonged survival in CML patients.65Therefore, it may be feasible to expect that Ara-C enhances the DR5 expression in CML progenitors and IFN-α induces the TRAIL expression on CTLs in CML patients, leading to elimination of Ph1-positive leukemias through the TRAIL/DR5 interaction. To address this possibility, the correlation between the in vitro susceptibility of CML progenitors to TRAIL and the clinical responses to AraC and/or IFN-α therapy have to be determined.

Allo-SCT is a potentially curative therapy against CML and Ph1-positive ALL, and its effectiveness is thought to be achieved mainly by the immune-mediated GVL effect. We herein showed that TRAIL could efficiently induce apoptosis in Ph1-positive AL-derived cell lines as well as CML-BC–derived cell lines. In this regard, endogenously expressed TRAIL on CTLs or NK cells may be involved in the GVL effect. To address this hypothesis, the correlation between the in vitro TRAIL sensitivity of leukemia cells and the leukemia-free survival after allo-SCT in CML and Ph1-positive ALL patients has to be determined in future study.

A significant number of CML patients cannot tolerate the IFN-α therapy because of lethargy, malaise, anorexia, depression, and autoimmune-like syndromes. The GVL effect after allo-SCT is frequently associated with life-threatening graft-versus-host disease. In contrast, at least in mice and nonhuman primates, administration of rhsTRAIL did not exhibit a serious toxicity.66,67 Thus, rhsTRAIL could be a novel therapeuetic agent for CML and Ph1-positive ALL patients as a safer alternative.

Finally, we investigated the correlation between TRAIL sensitivity and imatinib mesylate sensitivity. imatinib mesylate exerted a potent antileukemic effect against Ph1-positive cells not only in patients with CML in chronic phase who failed to the IFN-α therapy,56 but also in patients with CML in advanced phase or Ph1-positive ALL.56,61 Importantly, we demonstrated that imatinib mesylate efficiently repressed most of the TRAIL-resistant cell lines, while TRAIL repressed most of the imatinib mesylate-resistant cell lines. This suggests a potential clinical utility of TRAIL particularly for patients with imatinib mesylate-resistant CML and Ph1-positive ALL.

Prepublished online as Blood First Edition Paper, December 27, 2002; DOI 10.1182/blood-2002-06-1770.

Supported in part by research grants from the Ministry of Education, Science, and Culture, Japan.

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

1
Griffith
TS
Lynch
DH
TRAIL: a molecule with multiple receptors and control mechanisms.
Curr Opin Immunol.
10
1998
559
563
2
Wiley
SR
Schooley
K
Smolak
PJ
et al
Identification and characterization of a new member of the TNF family that induces apoptosis.
Immunity.
3
1995
673
682
3
Thomas
WD
Hersey
P
TNF-related apoptosis-inducing ligand (TRAIL) induces apoptosis in Fas ligand-resistant melanoma cells and mediates CD4 T cell killing of target cells.
J Immunol.
161
1998
2195
2200
4
Pan
G
O'Rourke
K
Chinnaiyan
AM
et al
The receptor for the cytotoxic ligand TRAIL.
Science.
276
1997
111
113
5
Pan
G
Ni
J
Wei
YF
Yu
G
Gentz
R
Dixit
VM
An antagonist decoy receptor and death domain-containing receptor for TRAIL.
Science.
277
1997
815
821
6
Schneider
P
Thome
M
Burns
K
et al
TRAIL receptors 1 (DR4) and 2 (DR5) signal FADDdependent apoptosis and activate NF-κB.
Immunity.
7
1997
831
836
7
Ashkenazi
A
Dixit
VM
Death receptors: signaling and modulation.
Science.
281
1998
1305
1308
8
Sheridan
JP
Marsters
SA
Pitti
RM
et al
Control of TRAIL-induced apoptosis by a family of signaling and decoy receptors.
Science.
277
1997
818
821
9
Degli-Esposti
MA
Dougall
WC
Smolak
PJ
Waugh
JY
Smith
CA
Goodwin
RG
The novel receptor TRAIL-R4 induces NF-kappaB and protects against TRAIL-mediated apoptosis, yet retains an incomplete death domain.
Immunity.
7
1997
813
820
10
Kagi
D
Vignaux
F
Ledermann
B
et al
Fas and perforin pathways as major mechanisms of T cell-mediated cytotoxicity.
Science.
265
1994
528
530
11
Kojima
H
Shinohara
N
Hanaoka
S
et al
Two distinct pathways of specific killing revealed by perforin mutant cytotoxic T lymphocytes.
Immunity.
1
1994
357
364
12
Lowin
B
Hahne
M
Mattmann
C
Tschopp
J
Cytolytic T-cell cytotoxicity is mediated through perforin and Fas lytic pathways.
Nature.
370
1994
650
652
13
Kayagaki
N
Yamaguchi
N
Nakayama
M
et al
Involvement of TNF-related apoptosis-inducing ligand in human CD4+ T cell-mediated cytotoxicity.
J Immunol.
162
1999
2639
2647
14
Kayagaki
N
Yamaguchi
N
Nakayama
M
et al
Expression and function of TNF-related apoptosis-inducing ligand on murine activated NK cells.
J Immunol.
163
1999
1906
1913
15
Kayagaki
N
Yamaguchi
N
Nakayama
M
Eto
H
Okumura
K
Yagita
H
Type 1 interferons (IFNs) regulate tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) expression on human T cells: a novel mechanism for the antitumor effects of type 1 IFNs.
J Exp Med.
189
1999
1454
1460
16
Mitsiades
CS
Treon
SP
Mitsiades
N
et al
TRAIL/Apo2L ligand selectively induces apoptosis and overcomes drug resistance in multiple myeloma: therapeutic applications.
Blood.
98
2001
795
804
17
Chen
Q
Gong
B
Mahmoud-Ahmed
AS
et al
Apo2L/TRAIL and Bcl-2-related proteins regulate type I interferon-induced apoptosis in multiple myeloma.
Blood.
98
2001
2183
2192
18
Plasilova
M
Zivny
J
Jelinek
J
et al
TRAIL (Apo2L) suppresses growth of primary human leukemia and myelodysplasia progenitors.
Leukemia.
16
2002
67
73
19
Kurzrock
R
Gutterman
JU
Talpaz
M
The molecular genetics of Philadelphia chromosome-positive leukemias.
N Engl J Med.
319
1988
990
998
20
Faderl
S
Talpaz
M
Estrov
Z
O'Brien
S
Kurzrock
R
Kantarjian
HM
The biology of chronic myeloid leukemia.
N Engl J Med.
341
1999
164
172
21
Sawyers
CL
Chronic myeloid leukemia.
N Engl J Med.
340
1999
1330
1340
22
Preti
HA
O'Brien
S
Giralt
S
Beran
M
Pierce
S
Kantarjian
HM
Philadelphia-chromosome-positive adult acute lymphocytic leukemia: characteristics, treatment results, and prognosis in 41 patients.
Am J Med.
97
1994
60
65
23
Uckun
FM
Nachman
JB
Sather
HN
et al
Clinical significance of Philadelphia chromosome positive pediatric acute lymphoblastic leukemia in the context of contemporary intensive therapies: a report from the Children's Cancer Group.
Cancer.
83
1998
2030
2039
24
Talpaz
M
McCredie
KB
Mavligit
GM
Gutterman
JU
Leukocyte interferon-induced myeloid cytoreduction in chronic myelogenous leukemia.
Blood.
62
1983
689
692
25
Interferon alpha-2a as compared with conventional chemotherapy for the treatment of chronic myeloid leukemia: the Italian Cooperative Study Group on Chronic Myeloid Leukemia.
N Engl J Med.
330
1994
820
825
26
Silver
RT
Woolf
SH
Hehlmann
R
et al
An evidence-based analysis of the effect of busulfan, hydroxyurea, interferon, and allogenic bone marrow transplantation in treating the chronic phase of chronic myeloid leukemia: developed for the American Society of Hematology.
Blood.
94
1999
1517
1536
27
Cornelissen
JJ
Ploemacher
RE
Wognum
BW
et al
An in vitro model for cytogenetic conversion in CML interferon-α preferentially inhibits the outgrowth of malignant stem cells preserved in long-term culture.
J Clin Invest.
102
1998
976
983
28
Selleri
C
Sato
T
Del Vecchio
L
et al
Involvement of Fas-mediated apoptosis in the inhibitory effects of interferon-alpha in chronic myelogenous leukemia.
Blood.
89
1997
957
964
29
Selleri
C
Maciejewski
JP
Pane
F
et al
Fas-mediated modulation of Bcr/Abl in chronic myelogeneous leukemia results in differential effects on apoptosis.
Blood.
92
1998
981
989
30
Friedman
RL
Stark
GR
Alpha-interferon-induced transcription of HLA and metallothionein genes containing homologous upstream sequences.
Nature.
314
1985
637
639
31
Mori
T
Manabe
A
Tuchida
M
et al
Allogeneic bone marrow transplantation in first remission rescues children with Philadelphia chromosome-positive acute lymphoblastic leukemia: Tokyo Children's Cancer Study Group (TCCSG) studies L89–12 and L92–13.
Med Pediatr Oncol.
37
2001
426
431
32
Gora-Tybor
J
Deininger
MW
Goldman
JM
Melo
JV
The susceptibility of Philadelphia chromosome positive cells to FAS-mediated apoptosis is not linked to the tyrosine kinase activity of BCR-ABL.
Br J Haematol.
103
1998
716
720
33
Ravandi
F
Kantarjian
HM
Talpaz
M
et al
Expression of apoptosis proteins in chronic myelogenous leukemia: associations and significance.
Cancer.
91
2001
1964
1972
34
Shlomchik
W
Pear
WS
Graft-vs-leukemia in a retrovirally induced murine CML model: mechanisms of leukemia recognition [abstract].
Blood.
98
2001
812a
35
Andersson
LC
Jokinen
M
Gahmberg
CG
Induction of erythroid differentiation in the human leukaemia cell line K562.
Nature.
278
1979
364
365
36
LeBien
TW
Hozier
J
Minowada
J
Kersey
JH
Origin of chronic myelocytic leukemia in a precursor of pre-B lymphocytes.
N Engl J Med.
301
1979
144
147
37
Inukai
T
Sugita
K
Mitsui
K
et al
Participation of granulocyte colony-stimulating factor in the growth regulation of leukemia cells from Philadelphia chromosome-positive acute leukemia and blast crisis of chronic myeloid leukemia.
Leukemia.
14
2000
1386
1395
38
Inukai
T
Sugita
K
Suzuki
T
et al
A novel 203 kD aberrant BCR-ABL product in a girl with Philadelphia chromosome positive acute lymphoblastic leukaemia.
Br J Haematol.
85
1993
823
825
39
Nakazawa
S
Saito
M
Mori
T
et al
Establishment of two cell lines (biphenotypic and myelomonocytic) derived from a patient with Ph1 positive acute leukemia [abstract no. 1226].
Proc Am Asso Cancer Res.
31
1990
206
40
Lin
YZ
Yao
S
Veach
RA
Torgerson
TR
Hawiger
J
Inhibition of nuclear translocation of transcription factor NF-κB by a synthetic peptide containing a cell membrane-permeable motif and nuclear localization sequence.
J Biol Chem.
270
1995
14255
14258
41
Druker
BJ
Tamura
S
Buchdunger
E
et al
Effects of a selective inhibitor of the Abl tyrosine kinase on growth of Bcr-Abl positive cells.
Nat Med.
2
1996
561
566
42
Yamakawa
N
Sugita
K
Inukai
T
et al
Ligand activation of peroxisome proliferator-activated receptor γ induces apoptosis of leukemia cells by down-regulating the c-myc gene expression via blockade of the Tcf-4 activity.
Cell Death Differ.
9
2002
513
526
43
Trauth
BC
Klas
C
Peters
AM
et al
Monoclonal antibody-mediated tumor regression by induction of apoptosis.
Science.
245
1989
301
305
44
Kischkel
F
Hellbardt
S
Behrman
I
et al
Cytotoxicity-dependent APO-1 (Fas/CD95)-associated proteins from a death-inducing signaling complex (DISC) with the receptor.
EMBO J.
14
1995
579
5588
45
Sprick
M
Weigand
M
Rieser
E
et al
FADD/MORT1 and caspase-8 are recruited to TRAIL receptors 1 and 2 and are essential for apoptosis mediated by TRAIL receptor 2.
Immunity.
12
2000
599
609
46
Kischkel
FC
Lawrence
DA
Chuntharapai
A
et al
Apo2L/TRAIL-dependent recruitment of endogenous FADD and caspase-8 to death receptors 4 and 5.
Immunity.
12
2000
611
620
47
Chinnaiyan
A
O'Rourke
K
Tewari
M
Dixit
VM
FADD, a novel death-domain-containing protein, interacts with the death domain of Fas and initiates apoptosis.
Cell.
81
1995
505
512
48
Boldin
M
Varfolomeev
E
Pancer
Z
et al
A novel protein that interacts with the death domain of Fas/Apo1 contains a sequence motif related to the death domain.
J Biol Chem.
270
1995
7795
7798
49
Boldin
M
Goncharov T, Goltsev Y, et al. Involvement of MACH, a novel MORT1/FADD-interacting protease, in Fas/Apo-1- and TNF receptor-induced cell death.
Cell.
85
1996
803
815
50
Muzio
M
Chinnaiyan
A
Kischkel
F
et al
FLICE, a novel FADD-homologous ICE/CED-3-like protease, is recruited to the CD95 (Fas/APO-1) death-inducing signaling complex.
Cell.
85
1996
817
827
51
Irmler
M
Thome
M
Hahne
M
et al
Inhibition of death receptor signals by cellular FLIP.
Nature.
388
1997
190
195
52
Reuther
JY
Reuther
GW
Cortez
D
Pendergast
AM
Baldwin
AS
Jr
A requirement for NF-κB activation in Bcr-Abl-mediated transformation.
Genes Dev.
12
1998
968
981
53
Chaudhary
PM
Eby
M
Jasmin
A
Bookwalter
A
Murray
J
Hood
L
Death receptor 5, a new member of the TNFR family, and DR4 induce FADD-dependent apoptosis and activate the NF-kappaB pathway.
Immunity.
7
1997
821
830
54
Griscavage
JM
Wilk
S
Ignarro
LJ
Inhibitors of the proteasome pathway interfere with induction of nitric oxide synthase in macrophages by blocking activation of transcription factor NF-kappaB.
Proc Natl Acad Sci U S A.
93
1996
3308
3312
55
Druker
BJ
Talpaz
M
Resta
DJ
et al
Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia.
N Engl J Med.
344
2001
1031
1037
56
Druker
BJ
Sawyers
CL
Kantarjian
H
et al
Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome.
N Engl J Med.
344
2001
1038
1042
57
Plasilova
M
Zivny
J
Jelinek
J
et al
TRAIL (Apo2L) suppresses growth of primary human leukemia and myelodysplasia progenitors.
Leukemia.
16
2002
67
73
58
Petak
I
Douglas
L
Tillman
D
et al
Pediatric rhabdomyosarcoma cell lines are resistant to Fas-induced apoptosis and highly sensitive to TRAIL-induced apoptosis.
Clin Cancer Res.
6
2000
4119
4127
59
Knight
MJ
Riffkin
CD
Muscat
AM
Ashley
DM
Hawkins
CJ
Analysis of FasL and TRAIL induced apoptosis pathways in glioma cells.
Oncogene.
20
2001
5789
5798
60
Velthuis
J
Rouschop
K
de Bont
H
et al
Distinct intracellular signaling in tumor necrosis factor-related apoptosis-inducing ligand- and CD95 ligand-mediated apoptosis.
J Biol Chem.
277
2002
24631
24637
61
Mercurio
F
Manning
AM
Multiple signals converging on NF-κB.
Curr Opin Cell Biol.
11
1999
226
232
62
Hideshima
T
Richadson
P
Chauhan
D
et al
The proteasome inhibitor PS-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human multiple myeloma cells.
Cancer Res.
61
2001
3071
3076
63
Adams
J
Proteasome inhibition in cancer: development of PS-341.
Semin Oncol.
6
2001
613
619
64
Wen
J
Ramadevi
N
Nguyen
D
Perkins
C
Worthington
E
Bhalla
K
Antileukemic drugs increase death receptor 5 levels and enhance Apo-2L-induced apoptosis of human acute leukemia cells.
Blood.
96
2000
3900
3906
65
Guilhot
F
Chastang
C
Michallet
M
et al
Interferon alfa-2b combined with cytarabine versus interferon alone in chronic myelogenous leukemia: French Chronic Myeloid Leukemia Study Group.
N Engl J Med.
337
1997
223
229
66
Walczak
H
Miller
RE
Ariail
K
et al
Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo.
Nat Med.
5
1999
157
163
67
Ichikawa
K
Liu
W
Zhao
L
et al
Tumoricidal activity of a novel anti-human DR5 monoclonal antibody without hepatocyte cytotoxicity.
Nat Med.
7
2001
954
960

Author notes

Takeshi Inukai, Department of Pediatrics, School of Medicine, University of Yamanashi, Tamaho, Nakakoma, Yamanashi 409-3898, Japan; e-mail:tinukai@res.yamanashi-med.ac.jp.

Sign in via your Institution