Signal transducer and activator of transcription (STAT) proteins are involved in hematopoietic cytokine receptor signaling pathways that regulate cell proliferation, differentiation, and survival. STATs are dysregulated in acute myeloid leukemia (AML); mechanisms of dysregulation include constitutive activation and truncation of the C-terminal transactivation domain; the latter results in a β isoform that has a trans-dominant negative effect on gene induction mediated by the full-length STATα form. It was hypothesized that constitutive STAT activity might correlate with unfavorable treatment outcome in AML. Pretreatment bone marrow samples from 63 adult patients with AML were analyzed by electrophoretic mobility shift assay for the presence of STAT DNA-binding activity. Isoforms and relative levels of STAT proteins were determined by immunoblotting. Constitutive STAT3 activity was detected in samples from 28 (44%) patients. Pretreatment clinical characteristics, expression of STATα/β isoforms, and treatment regimens did not differ significantly between patients with and without constitutive STAT3 activity. Disease-free survival (DFS) was significantly shorter in patients with than in patients without constitutive STAT3 activity (median 8.7 vs 20.6 months;P = .01). Overall survival did not differ significantly. The subgroup of patients with constitutive STAT3 activity and the STAT3β isoform had the shortest DFS (P = .006) and shorter overall survival (P = .049) than all other patients. Whether adverse treatment outcome is attributable to constitutive STAT activity itself or to a process that leads to constitutive STAT activity remains to be determined. This is the first demonstration of a prognostic significance for STAT proteins in a malignancy.

Acute myeloid leukemia (AML) is a clonal disease characterized by maturation arrest of a malignant clone of myeloid cells. Because most AML cells are incapable of proliferating ex vivo without growth factor support, growth factors and response to growth factors are likely to determine the growth and differentiation state of leukemic blasts in vivo.1 

Signal transducer and activator of transcription (STAT) proteins are latent cytoplasmic transcription factors involved in hematopoietic growth factor signal transduction.2-5 STATs are activated by tyrosine phosphorylation through the action of receptor-associated Janus family tyrosine kinases (JAKs). Receptor-regulated signaling proteins appear to demonstrate specificity: interleukin-6 (IL-6),6 granulocyte (G) colony-stimulating factor (CSF),7 and thrombopoietin (TPO)8 activate STAT3, whereas granulocyte macrophage (GM)–CSF, TPO, and IL-3 primarily activate STAT5.9,10 Receptors for growth factor signaling through STAT3 and STAT5 are present on AML blasts, and these growth factors stimulate AML blast proliferation in vitro.1 Because multipotential, nonleukemic hematopoietic cells undergo differentiation, whereas leukemic cells maintain proliferation rather than differentiation in response to these growth factors, aberration of signaling pathways may contribute to leukemogenesis.

Dysregulation of the growth factor-STAT pathway can occur by 2 mechanisms. One is constitutive activation of the STATs9,10 and the other is the production of C-terminally truncated isoforms called STATβ.11-14 C-terminal transactivation domain truncation does not prevent tyrosine phosphorylation of STATβ, dimerization, or binding to DNA; rather, it either competitively inhibits STATα function or prevents transcription of STAT target genes in a dominant-negative fashion.11-14 Constitutive activation of STAT3 may also result from the introduction of a cysteine residue into the C-terminal domain of the molecule,15 and these C-terminally mutated STAT3 proteins may be oncogenic when constitutively activated. We hypothesized that constitutive STAT3 activity or production of the truncated isoform might be associated with drug resistance and might, therefore, correlate with adverse treatment outcome in AML.

Patient population

The patient population consisted of 63 adult patients with AML who had at least 5 × 107 bone marrow cells cryopreserved at the time of diagnosis and were treated in a time frame that allowed for 4 years of follow-up. Pretreatment characteristics are shown in Table 1; the patients were representative of a typical AML population. Thirty-two patients were men and 31 were women. Median age was 64 years (range, 18-85 years). Fifty patients had de novo AML, and 13 had secondary AML; 9 had antecedent hematologic disorders and 4 received chemotherapy for prior malignancy. Peripheral blood mononuclear cells from 3 healthy blood donors and CD34+ cells separated from peripheral stem cell collection samples from 3 patients with breast cancer without bone marrow involvement served as controls. Prior approval was obtained from the Roswell Park Cancer Institute Institutional Review Board. All patients and volunteers signed informed consent.

Table 1.

Pretreatment patient characteristics

CharacteristicsTotal
(n = 63)
STAT3+(n = 28)STAT3 (n = 35)P
Age, y    .20 
 60 or younger 27 18 
 Older than 60 36 19 17  
Sex (M/F) 32/31 15/13 17/18 .80 
Disease status    1.00 
 de novo 50 22 28 
 Secondary 13 
WBC count (median, ×109/L) 20.25 10.31 29.86 .09 
FAB    NA 
 M0 
 M1 15 
 M2 25 14 11 
 M3 
 M4 
 M5 
 M6 
Cytogenetics    .73 
 Favorable 
 Intermediate 30 18 12 
 Unfavorable* 26 12 14 
STAT subtype    .29 
 α 23 14 
 β 33 18 15  
CharacteristicsTotal
(n = 63)
STAT3+(n = 28)STAT3 (n = 35)P
Age, y    .20 
 60 or younger 27 18 
 Older than 60 36 19 17  
Sex (M/F) 32/31 15/13 17/18 .80 
Disease status    1.00 
 de novo 50 22 28 
 Secondary 13 
WBC count (median, ×109/L) 20.25 10.31 29.86 .09 
FAB    NA 
 M0 
 M1 15 
 M2 25 14 11 
 M3 
 M4 
 M5 
 M6 
Cytogenetics    .73 
 Favorable 
 Intermediate 30 18 12 
 Unfavorable* 26 12 14 
STAT subtype    .29 
 α 23 14 
 β 33 18 15  
*

Twelve patients had one aberration each—r(7), t(6;11), t(5;8), t(9;22), add(1), +8, +mar, +13, add(20), der(13;14), +2mar, or del(X)q23—and 14 had 2 or more cytogenetic aberrations.

FAB indicates French-American-British; NA, not analyzed.

Morphologic studies

Morphologic categorization of pretreatment bone marrow was made according to the French-American-British classification.16,17 Five patients had M0, 15 had M1, 25 had M2, 3 had M3, 6 had M4, 6 had M5, and 3 had M6 subtype disease.

Cytogenetic analysis

Cytogenetic analysis was performed on pretreatment bone marrow cells from all patients. Bone marrow samples were processed using short-term unstimulated cultures (24-72 hours). Clonality criteria and descriptions of chromosomal aberrations were according to the International System for Human Cytogenetic Nomenclature.18Patients were divided into 3 prognostic groups based on karyotype, as previously described19; prognostic groups were favorable [t(8;21), inv(16) or t(15;17)], intermediate (normal cytogenetics), and unfavorable (all others).

Treatment

Fifty-one patients received high-dose cytarabine and idarubicin induction therapy20 and 12 received other induction regimens.21-23 Of the 46 patients who achieved complete remission (CR), 44 received postremission therapy as outlined in Table2, and 2 patients did not receive any additional therapy. Of note, postremission therapy included autologous peripheral blood stem cell transplantation in 8 patients and allogeneic transplantation in 2. Additionally, 11 patients, including 5 with and 6 without constitutive STAT3 activity, underwent allogeneic bone marrow transplantation because they had refractory or relapsed disease or they were in second remission.

Table 2.

Treatment outcome

Total
(n = 63)
STAT3+ (n = 28)STAT3(n = 35)P
Induction treatment    .75 
 HIDAC/IDA* 51 22 29 
 Other 12 
Consolidation 46 21 25 .37 
 HIDAC/IDA group 12 
  HIDAC/IDA alone 
  HIDAC/IDA + VP/CY 
  HIDAC/IDA + HIDAC/DNR2-153 
 VP/CY 16 10 
 Other 16 10 
  AlloBMT in first remission 
  AutoPBSCT 
  ADE2-155 
  HIDAC alone 
  7 and 32-154 + HIDAC/DNR 
 None 
AlloBMT relapse/refractory AML/2nd remission 11 NA 
DFS (median, mo) 10.6 8.7 20.6 .01  
Overall survival (median, mo) 14.7 14.0 16.8 .1  
Attainment of CR: 
 Yes 46 21 25 .78 
 No 17 10  
Total
(n = 63)
STAT3+ (n = 28)STAT3(n = 35)P
Induction treatment    .75 
 HIDAC/IDA* 51 22 29 
 Other 12 
Consolidation 46 21 25 .37 
 HIDAC/IDA group 12 
  HIDAC/IDA alone 
  HIDAC/IDA + VP/CY 
  HIDAC/IDA + HIDAC/DNR2-153 
 VP/CY 16 10 
 Other 16 10 
  AlloBMT in first remission 
  AutoPBSCT 
  ADE2-155 
  HIDAC alone 
  7 and 32-154 + HIDAC/DNR 
 None 
AlloBMT relapse/refractory AML/2nd remission 11 NA 
DFS (median, mo) 10.6 8.7 20.6 .01  
Overall survival (median, mo) 14.7 14.0 16.8 .1  
Attainment of CR: 
 Yes 46 21 25 .78 
 No 17 10  

AlloBMT indicates allogeneic bone marrow transplantation; AutoPBSCT, autologous peripheral blood stem cell transplantation; NA, not analyzed; and STAT, signal transducer and activator of transcription proteins.

*

High-dose cytarabine 3 g/m2 every 12 hours for a total of 12 doses (1.5 g/m2 for patients older than 50) and idarubicin 12 mg/m2 daily for 3 consecutive days.

Five patients treated with cytarabine 100 mg/m2 continuous infusion over 7 days, daunorubicin 60 mg/m2 daily for 3 days, etoposide 100 mg/m2 for 3 days; three patients treated with cytarabine 100 mg/m2continuous infusion over 7 days, daunorubicin 45 mg/m2daily for 3 days, etoposide 100 mg/m2 for 3 days, and PSC 833 10 mg/m2 for 3 days; one patient treated with cytarabine 200 mg/m2 continuous infusion over 7 days and daunorubicin 45 mg/m2 daily for 3 days; one patient treated with all-trans retinoic acid 40 mg/m2 every 12 hours and idarubicin 12 mg/m2 every other day for 4 doses; one patient treated with cytarabine 1.5 g/m2 every 12 hours for a total of 12 doses; one patient treated with cytarabine 100 mg/m2 continuous infusion over 7 days.

Etoposide 3.6 g/m2 by continuous infusion and cyclophosphamide 50 mg/kg for 4 days.

F2-153

High-dose cytarabine 2 g/m2 every 12 hours for a total of 8 doses and daunorubicin 45 mg/m2 for 2 days.

F2-155

Cytarabine 100 mg/m2 continuous infusion over 5 days, daunorubicin 60 mg/m2 daily for 2 days, and etoposide 100 mg/m2 for 2 days.

F2-154

Cytarabine 100 mg/m2 continuous infusion over 7 days and daunorubicin 45 mg/m2 daily for 3 days.

Response criteria

CR was defined as the normalization of blood counts and bone marrow morphology and the disappearance of all signs of leukemia, lasting for 4 weeks or longer, in accordance with the recommendations of the National Cancer Institute–sponsored workshop.24Relapse was defined as the reappearance and persistence of blasts in the blood or the appearance of 5% or more blasts in the bone marrow not attributable to another cause.

Materials

All chemicals were purchased from Sigma Immunochemicals (St Louis, MO) unless otherwise specified. TPO and G-CSF were kindly provided by Amgen (Thousand Oaks, CA).

Cell collection

Bone marrow samples were collected at diagnosis. Light-density bone marrow cells were isolated by 1.077 g/μL Ficoll-Hypaque density gradient centrifugation and were cryopreserved using standard techniques. Cells were also thawed by standard techniques, and cell viability was verified by the Trypan blue dye exclusion test. Three samples were studied fresh (on the day of collection) and after cryopreservation and thawing to determine whether cryopreservation and thawing affected STAT activity.

Controls consisted of monocytes and granulocytes separated from samples of 3 healthy blood donors. Mononuclear cells were obtained by density centrifugation. Cells were resuspended in RPMI 1640 medium containing 2% fetal calf serum and were incubated in culture dishes for 2 hours at 37°C at 2 × 106 cells/mL. The supernatant containing the nonadherent cells was discarded, and the adherent cells were collected. To obtain granulocytes, the cell pellet was collected after density centrifugation. Red blood cells were lysed with 1.22% ammonium oxalate, allowing the isolation of granulocytes. Additional non-AML controls included CD34+ cells separated from peripheral stem cell collections from 3 patients with breast cancer without bone marrow involvement using the MACS progenitor cell isolation kit (Miltenvy Biotec, Auburn, CA).

Electrophoretic mobility shift assay

The DNA-binding activity of STAT3 and STAT5 in the absence of cytokine or growth factor treatment was assessed by electrophoretic mobility shift assay (EMSA). Whole-cell extracts prepared from AML cells as described13 were incubated with32P-labeled oligomers corresponding to the high-affinity binding element for STAT3, SIE,25 and STAT5 (TB2).26 Complexes were analyzed by 5% polyacrylamide gel electrophoresis and autoradiography. Extracts of TPO-treated MO7E cells displaying activated STAT3α, STAT5α (both A and B27), and CD34+ cells expressing inactive STAT3α, STAT5Aα, and STAT5Bα served as controls for these analyses. Relative DNA-binding activity was determined by densitometry of the autoradiographs; activated MO7E standard was defined as 100%. The detection limit of the EMSA system was set at 2% or less. Binding activity with values greater than 2% was defined as constitutive. Identity of the STAT-containing complexes was determined by antibody supershift with C-terminal–specific anti-STAT3 (C-20) or anti-STAT5 (C-17) monoclonal antibodies (Santa Cruz Biotechnology, Santa Cruz, CA).

Western blotting

Tyrosine-phosphorylated and unphosphorylated STAT3, STAT5A, and STAT5B proteins were quantitated by Western blot analysis as previously described.13 In brief, whole-cell extracts were separated on 7.5% polyacrylamide sodium dodecyl sulfate gels, and proteins were transferred to nitrocellulose membranes. Membranes were incubated with either antiphospho-STAT3 (Y705) or antiphospho-STAT5A/B (Y694/Y699) antibodies (Upstate Biotechnology, Lake Placid, NY) or with N-terminal–specific anti-STAT3 and anti-STAT5 (Transduction Laboratories, Lexington, KY) to quantitate unphosphorylated proteins. C-terminal–specific anti-STAT3 (C-20) and anti-STAT5 (C17) (Santa Cruz Biotechnology) were used to verify the identity of the C-terminal truncation of the STAT protein forms. Immune complexes were detected by the enhanced chemiluminescence reaction (Amersham Life Science, Arlington Heights, IL).

Statistical methods

The following data were analyzed: constitutive STAT3 activity (present vs absent), patient age (60 years or younger vs older than 60 years), sex, initial white blood cell count (median), karyotype subgroups (favorable vs intermediate vs unfavorable), AML type (de novo vs secondary), STAT isoform (α vs β), treatment (induction treatment with high-dose cytarabine and idarubicin vs other regimens; consolidation treatment with high-dose cytarabine and idarubicin vs high-dose etoposide and cyclophosphamide vs other regimens), attainment of CR (yes vs no), disease-free survival (DFS), and overall survival.

DFS was defined as the time from achievement of CR to relapse, death, or last follow-up visit. Patients alive and still in remission at last follow-up examination were censored in the analysis. Overall survival was calculated from the date of diagnosis to the date of death or the date of last follow-up for living patients. Follow-up time was defined as the time from the date of diagnosis to the date of last contact among patients known to be alive. Time to achieving CR was defined as the interval from date of diagnosis to the date of clinically documented remission. Estimates of DFS and overall survival probabilities were calculated using the Kaplan-Meier method.28 The log-rank statistic29 was used to test for differences in survival times between the 2 STAT3 groups. The proportionality assumption in the Cox proportional hazards regression model was tested for constitutive STAT3 activation for the endpoint of overall survival, and it was found that a Cox model that incorporated a time-dependent covariate30 was a better fit for the data. This model allowed the hazard function for STAT3 expression to vary over time. Patient age, AML type, cytogenetic status, and WBC count were analyzed in univariate Cox proportional hazards models. Variables significant at the 10% level were then analyzed in conjunction with STAT3 and the time-dependent covariate to determine the effect of each of these variables on the prognostic significance of STAT3 expression. Sparseness in the data set did not allow for multivariate analysis of these factors simultaneously or for an appropriate statistical model to be derived for disease-free survival. Associations between both STAT3 groups and categorical variables were evaluated using the Fisher exact test.31The Kruskal-Wallis test32 was used to test for an association between STAT3 and karyotype subgroups. WBC count and time to CR were compared between the 2 STAT3 groups using the Mann-Whitney rank sum test.33 All statistical tests were 2-sided, with statistical significance defined as P < .05.

Constitutive STAT activity

Constitutive STAT3 activity was present in leukemic blasts from 28 of the 63 (44%) patients but in none of the control samples. Constitutive STAT5 activity was detected in blasts from 11 of the 50 (22%) patients. Cryopreservation had no effect on constitutive STAT activity (data not shown), and the results obtained by EMSA and by Western blot analysis were identical. Figure1 depicts EMSA and Western blot analyses for STAT3 from 6 representative patients.

Fig. 1.

Constitutive STAT3 activity in AML blasts.

(A) The top panel shows EMSA, demonstrating the SIE-binding activity of STAT3, and the lower 2 panels show Western blotting. The middle panel shows hybridization with antibodies against phosphorylated STAT3, and the lower panel shows hybridization with anti-STAT3 antibodies to demonstrate equal loading. The positions of STAT3α and STAT3β are indicated. Lanes 1, 2, 3, 5, and 6 show patient samples with constitutive STAT3 activity. Lane 4 represents a patient sample without constitutive STAT3 activity. Note that the bands in lanes 1, 2, 3, and 6 demonstrate faster migration—that is, STAT3α—whereas the band in lane 5 shows slower migration—that is, STAT3β. (B) Supershift analysis of the SIE band complex formed with C-terminally directed anti-STAT3 antibodies in cells from a patient expressing predominantly STAT3β and MO7E cells expressing predominantly STAT3α. There is no supershift in the patient sample, whereas STAT3α is supershifted in MO7E cells. (C) Western blot analysis of STAT3 hybridized with antibodies directed against the N-terminal (top panel) and the C-terminal (lower panel) domains. When using antibodies directed against the C-terminal domain of STAT3, STAT3β was not detected. To verify that no change occurred after cytokine exposure, samples were exposed to human G-CSF (10 ng/mL) for 10 minutes as previously described.13 

Fig. 1.

Constitutive STAT3 activity in AML blasts.

(A) The top panel shows EMSA, demonstrating the SIE-binding activity of STAT3, and the lower 2 panels show Western blotting. The middle panel shows hybridization with antibodies against phosphorylated STAT3, and the lower panel shows hybridization with anti-STAT3 antibodies to demonstrate equal loading. The positions of STAT3α and STAT3β are indicated. Lanes 1, 2, 3, 5, and 6 show patient samples with constitutive STAT3 activity. Lane 4 represents a patient sample without constitutive STAT3 activity. Note that the bands in lanes 1, 2, 3, and 6 demonstrate faster migration—that is, STAT3α—whereas the band in lane 5 shows slower migration—that is, STAT3β. (B) Supershift analysis of the SIE band complex formed with C-terminally directed anti-STAT3 antibodies in cells from a patient expressing predominantly STAT3β and MO7E cells expressing predominantly STAT3α. There is no supershift in the patient sample, whereas STAT3α is supershifted in MO7E cells. (C) Western blot analysis of STAT3 hybridized with antibodies directed against the N-terminal (top panel) and the C-terminal (lower panel) domains. When using antibodies directed against the C-terminal domain of STAT3, STAT3β was not detected. To verify that no change occurred after cytokine exposure, samples were exposed to human G-CSF (10 ng/mL) for 10 minutes as previously described.13 

Close modal

Constitutive STAT activity and pretreatment clinical characteristics

Differences in patient age, sex, frequency of de novo compared with secondary AML, initial WBC counts, prognostic karyotype subgroups, and expression of STAT3α/β isoforms were not statistically significant between patients with and without constitutive STAT3 activity (Table 1). Patients with and without constitutive STAT5 activity were not compared because of the small proportion of patients with constitutive STAT5 activity.

Constitutive STAT activity and treatment outcome

There was no difference in the distribution of induction (P = .75) and consolidation (P = .37) treatment regimens between the patients with and without constitutive STAT3 activity (Table 2). The CR rate was 75% for patients with constitutive STAT3 activity and 71% for those without. Median follow-up duration was 40 months (range, 19 to 64 months). Fourteen patients are still alive in CR, 4 died in CR of unrelated conditions, and 28 patients have had relapses. The median time to achievement of CR was 42 days (range, 31-78 days) for patients with constitutive STAT3 activity and 39 days (range, 24-92 days) for those without it (P = .37).

In univariate analysis, DFS was significantly shorter in patients with constitutive STAT3 activity than in those without it (median, 8.7 vs 20.6 months; P = .01) (Figure2). The probability of DFS at 12 months was 0.24 in patients with constitutive STAT3 activity and 0.56 in those without. The difference in overall survival was not statistically significant between the 2 groups (median, 14.0 vs 16.8 months;P = .1) (Figure 3), probably because of the effect of salvage therapy after relapse. However, only 18% of patients with constitutive STAT3 activity were alive at 25 months compared with 37% of those with no constitutive activity.

Fig. 2.

Kaplan-Meier curve for DFS by constitutive STAT3 activity for 46 patients with AML.

Circles indicate censored patients. (●), patients whose blasts had constitutive STAT3 activity. (○), patients whose blasts did not have constitutive STAT3 activity. Of note, one patient whose blasts did not have constitutive STAT3 activity survived in remission for more than 5 years.

Fig. 2.

Kaplan-Meier curve for DFS by constitutive STAT3 activity for 46 patients with AML.

Circles indicate censored patients. (●), patients whose blasts had constitutive STAT3 activity. (○), patients whose blasts did not have constitutive STAT3 activity. Of note, one patient whose blasts did not have constitutive STAT3 activity survived in remission for more than 5 years.

Close modal
Fig. 3.

Kaplan-Meier overall survival curve by constitutive STAT3 activity for 63 AML patients.

Circles indicate censored patients. (●), patients whose blasts had constitutive STAT3 activity. (○), patients whose blasts did not have constitutive STAT3 activity. Of note, 2 patients whose blasts did not have constitutive STAT3 activity survived for more than 5 years.

Fig. 3.

Kaplan-Meier overall survival curve by constitutive STAT3 activity for 63 AML patients.

Circles indicate censored patients. (●), patients whose blasts had constitutive STAT3 activity. (○), patients whose blasts did not have constitutive STAT3 activity. Of note, 2 patients whose blasts did not have constitutive STAT3 activity survived for more than 5 years.

Close modal

Because age, de novo versus secondary AML, and karyotype are known to affect treatment outcome,34 we analyzed the effect of constitutive STAT3 activity on overall survival in association with each of these factors. Table 3 shows the results of the Cox proportional hazards and time-dependent regression analyses. On univariate analysis, age, AML type, and cytogenetic subgroups, but not initial WBC count, were significantly related to overall patient survival. Patients younger than 60 had a 66% reduced risk for death. Patients with de novo AML had a 59% lower risk for death than those with secondary AML. Those with favorable cytogenetics had a 91% lower risk for death than the remaining patients. In the time-dependent model, patients without constitutive STAT3 activity had a 75% lower risk for death (P = .03). However, the differences in risk between patients with constitutive STAT3 activity and those without changed slightly (20%) over time. Constitutive STAT3 activity retained its statistical significance when adjusted for in models with age, AML type, and cytogenetic subgroups in time-dependent covariate analysis. Treatment outcome for patients with and without constitutive STAT5 activity was not analyzed because of the small proportion of patients with constitutive STAT5 activity.

Table 3.

Cox proportional hazard and time-dependent regression analysis for overall survival

VariablenParameter estimateRelative risk95% CIP
Univariate analysis      
Age, y      
 60 or younger vs older than 60 63 −1.09 0.34 0.17-0.66 .002 
AML type      
 de novo vs secondary 63 −0.88 0.41 0.21-0.80 .009  
Cytogenetic status      
 favorable vs intermediate or unfavorable 62 −2.41 0.09 0.01-0.66 .018  
WBC count 60 0.001 1.00 1.00-1.01 .615  
Time-dependent covariate models      
 STAT3 (no activity vs activity) 63 −1.40 0.25 0.07-0.88 .031  
 STAT3 × time-dependent covariate  0.18 1.20 1.07-1.34 .002 
 STAT3 63 −1.57 0.21 0.06-0.75 .016 
 STAT3 × time-dependent covariate  0.17 1.19 1.06-1.33 .003 
 Age  −0.92 0.40 0.20-0.79 .009 
 STAT3 62 −1.42 0.24 0.06-0.75 .029 
 STAT3 × time-dependent covariate  0.17 1.19 1.06-1.33 .003  
 AML type  −0.61 0.55 0.28-1.07 .077 
 STAT3 62 −1.56 0.21 0.06-0.75 .016 
 STAT3 × time-dependent covariate  0.16 1.18 1.05-1.32 .005  
 Cytogenetic status  −1.97 0.14 0.02-1.03 .054 
VariablenParameter estimateRelative risk95% CIP
Univariate analysis      
Age, y      
 60 or younger vs older than 60 63 −1.09 0.34 0.17-0.66 .002 
AML type      
 de novo vs secondary 63 −0.88 0.41 0.21-0.80 .009  
Cytogenetic status      
 favorable vs intermediate or unfavorable 62 −2.41 0.09 0.01-0.66 .018  
WBC count 60 0.001 1.00 1.00-1.01 .615  
Time-dependent covariate models      
 STAT3 (no activity vs activity) 63 −1.40 0.25 0.07-0.88 .031  
 STAT3 × time-dependent covariate  0.18 1.20 1.07-1.34 .002 
 STAT3 63 −1.57 0.21 0.06-0.75 .016 
 STAT3 × time-dependent covariate  0.17 1.19 1.06-1.33 .003 
 Age  −0.92 0.40 0.20-0.79 .009 
 STAT3 62 −1.42 0.24 0.06-0.75 .029 
 STAT3 × time-dependent covariate  0.17 1.19 1.06-1.33 .003  
 AML type  −0.61 0.55 0.28-1.07 .077 
 STAT3 62 −1.56 0.21 0.06-0.75 .016 
 STAT3 × time-dependent covariate  0.16 1.18 1.05-1.32 .005  
 Cytogenetic status  −1.97 0.14 0.02-1.03 .054 

Time-dependent covariate shows how the relative risk associated with constitutive STAT3 activity varies over time.

Expression of truncated STAT3β form in leukemic cells with constitutive STAT3 activity identifies a group of patients with short DFS and overall survival

There was a statistically significant difference in DFS when comparing patients with constitutive STAT3 activity and expression of the truncated isoform versus the remaining patients (P = .006). The median DFS for those with constitutive STAT3β expression was 7.4 months, compared with a median time of 18.8 months for the others. The difference in overall patient survival between these 2 groups of patients was also significant (P = .049); patients with constitutive STAT3β expression had a median survival time of 12.9 months compared with 18.9 months for the others.

We have demonstrated that constitutive STAT3 activity is associated with short DFS. The difference in DFS persisted when controlling for age, de novo versus secondary AML, and karyotype. Additionally, the subgroup of patients with constitutive STAT3β activity had particularly short DFS and shorter overall survival. To our knowledge, the relationship between constitutive STAT3 activity and short DFS has not been reported. Moreover, this is the first demonstration of a relationship between STAT expression and disease outcome in any malignancy.

Constitutive STAT3 activity has been reported in 30% to 100% of patients with AML.13,35-37 Several mechanisms have been proposed to explain constitutive STAT3 activity in AML. One proposed mechanism is activation of the signaling pathway by an autocrine loop mediated by a hematopoietic growth factor. Schuringa et al38 showed that constitutive STAT3 activity in AML blasts resulted from autocrine secretion of IL-6. However, IL-6 is known to be inhibitory for AML proliferation.39 Therefore, the role of IL-6–induced STAT3 activation in leukemogenesis remains unclear. Other growth factors, TPO, and G-CSF, are also known to activate STAT3.7,8 We and others19,40-42 have previously demonstrated that the TPO receptor is expressed on AML blasts in 45% to 70% of patients with AML. Moreover, we found that DFS was significantly shorter in the subgroup of AML patients whose blasts expressed TPO receptor mRNA.19 Nine of the same patient samples were analyzed in that study and in the current study. Two patient samples that expressed c-mpl mRNA had constitutive STAT3 activity; among the 7 samples that did not express c-mpl mRNA, 6 did not demonstrate constitutive STAT3 activity. Although the numbers are small, the correlation is intriguing in that it suggests that constitutive STAT3 activity might be induced by TPO. Additional work is needed to address this possible mechanism. Similarly, G-CSF has been shown to induce the transformation and proliferation of AML blasts through STAT3α activation.7Finally, cellular transformation triggered by various tyrosine kinase oncoproteins, such as v-Src and Lck, has also been associated with the constitutive activation of STAT3,43-46 but involvement of these oncoproteins has not been demonstrated in AML and cannot explain the high proportion of patients with constitutive STAT3 activity in this disease. These studies strongly suggest that constitutive STAT3 activity plays an important role in malignant transformation. It is unclear whether constitutive STAT3 activity is a cause or a result of the transforming event.

The targets of STAT3 have not yet been identified. One possible target may be the antiapoptotic pathway of Bcl-2. Expression of antiapoptotic proteins Bcl-XL and Mcl-1, members of the Bcl-2 family, was shown to be increased in multiple myeloma cells with IL-6–induced constitutive STAT3 activity.47,48 Abrogation of STAT3 signaling has been demonstrated to block Bcl-XL expression, with subsequent induction of apoptosis.47 These data suggest that constitutive STAT3 activity may confer resistance to apoptosis in multiple myeloma cells. No similar data are available yet for AML.

Introducing a cysteine at the C-terminal loop of the SH2 domain of STAT3 causes the molecule to dimerize, promote transcription, and induce cell transformation,15 suggesting that altering the C-terminal domain of STAT3 induces constitutive activation, resulting in a transformed phenotype. C-terminally truncated STAT3β isoforms were shown to have a trans-dominant negative effect on gene induction in the STAT signal transduction pathway.11-14 In our assays, C-terminal mutation has not been evident; rather, we have found truncation and loss of the transactivating domain with preservation of the tyrosine phosphorylation capability.13,14 In our patient cohort, the expression of C-terminally truncated STAT3β isoform was associated with an even worse outcome when detected in AML blasts with constitutive STAT3 activity.

In summary, we have demonstrated that constitutive STAT3 activity is associated with short DFS in AML. Moreover, a subgroup of patients with constitutive STAT3 activity who displayed STAT3β isoform experienced a particularly poor outcome. The correlation between STAT expression and outcome remains to be explained. The long-term implication of our data is that the development of novel therapies targeting the signal transduction pathways in AML cells may hold promise for improving treatment outcome in this disease.

Supported in part by grants from the Tower Foundation (Buffalo, NY) and by National Cancer Institute grants CA16056 and CA26122. M.B. is a recipient of The Cancer and Leukemia Group B Clinical Research Award supported by Ortho Biotech, Inc.

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
Lowenberg
B
Touw
IP
Hematopoietic growth factors and their receptors in acute leukemia.
Blood.
81
1993
281
292
2
Ihle
JN
STATs: signal transducers and activators of transcription.
Cell.
84
1996
331
334
3
Ward
AC
Touw
I
Yoshimura
A
The Jak-Stat pathway in normal and perturbed hematopoiesis.
Blood.
95
2000
19
29
4
Imada
K
Leonard
WJ
The Jak-STAT pathway.
Mol Immunol.
37
2000
1
11
5
Takeda
K
Akira
S
STAT family of transcription factors in cytokine-mediated biological responses.
Cytokine Growth Factor Rev.
11
2000
199
207
6
Zhong
Z
Wen
Z
Darnell
JE
STAT3: a STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6.
Science.
264
1994
95
98
7
Chakraborty
A
Tweardy
DJ
Stat3 and G-CSF–induced myeloid differentiation.
Leuk Lymphoma.
30
1998
433
442
8
Miyakawa
Y
Oda
A
Druker
BJ
et al
Thrombopoietin induces tyrosine phosphorylation of Stat3 and Stat5 in human blood platelets.
Blood.
87
1997
439
446
9
Coffer
PJ
Koenderman
L
de Groot
RP
The role of STATs in myeloid differentiation and leukemia.
Oncogene.
19
2000
2511
2522
10
Lin
TS
Mahajan
S
Frank
DA
STAT signaling in the pathogenesis and treatment of leukemias.
Oncogene.
19
2000
2496
2504
11
Tweardy
DJ
Wright
TM
Ziegler
SF
et al
Granulocyte colony-stimulating factor rapidly activates a district STAT-like protein in normal myeloid cells.
Blood.
86
1995
4409
4416
12
Caldenhoven
E
van Dijk
TB
Solari
R
et al
STAT3β, a splice variant of transcription factor STAT3, is a dominant negative regulator of transcription.
J Biol Chem.
271
1996
13221
13227
13
Xia
Z
Baer
MR
Block
AW
Baumann
H
Wetzler
M
Expression of signal transducers and activators of transcription proteins in acute myeloid leukemia blasts.
Cancer Res.
58
1998
3173
3180
14
Xia
Z
Salzler
R
Kunz
DP
et al
A novel serine-dependent proteolytic activity is responsible for truncated signal transducer and activator of transcription proteins in acute myeloid leukemia blasts.
Cancer Res.
61
2001
1747
1753
15
Bromberg
JF
Wrzeszczynska
MH
Devgan
G
et al
Stat3 as an oncogene.
Cell.
98
1999
295
303
16
Bennett
JM
Catovsky
D
Daniel
MT
et al
Proposed revised criteria for the classification of acute myeloid leukemia: a report of the French-American-British Cooperative group.
Ann Intern Med.
103
1985
620
625
17
Bennett
JM
Catovsky
D
Daniel
MT
et al
Proposal for the recognition of minimally differentiated acute myeloid leukaemia (AML-M0).
Br J Haematol.
78
1991
325
329
18
Mitelman
F
ISCN: An International System for Human Cytogenetic Nomenclature.
1995
Karger
Basel, Switzerland
19
Wetzler
M
Baer
MR
Bernstein
SH
et al
Expression of c-mpl mRNA, the receptor for thrombopoietin, in acute myeloid leukemia blasts identifies a group of patients with poor response to intensive chemotherapy.
J Clin Oncol.
15
1997
2262
2268
20
Baer
MR
Pixley
LA
Ford
LA
et al
High-dose cytarabine and idarubicin induction produces a high complete remission rate in previously untreated de novo acute myeloid leukemia patients [abstract].
Blood.
96(suppl 1)
2000
322
21
Baer
MR
George
SL
Dodge
RK
et al
Phase III study of PSC-833 modulation of multidrug resistance in previously untreated acute myeloid leukemia patients ≥ 60 years (CALGB 9720) [abstract].
Blood.
94(suppl 1)
1999
383
22
Kolitz
JE
George
SL
Hurd
D
et al
Parallel phase I trials of multidrug resistance modulation with PSC-833 in untreated patients with acute myeloid leukemia < 60 years old: preliminary results of CALGB 9621 [abstract].
Blood.
94(suppl 1)
1999
384
23
Kolitz
JE
George
SL
Hurd
D
et al
Cytogenetic risk-adapted intensification followed by immunotherapy with recombinant interleukin-2 (rIL-2) in patients (PTS) < 60 years old with acute myeloid leukemia (AML) in first complete remission (CR): preliminary results of CALGB 9621 [abstract].
Blood.
94(suppl 1)
1999
579
24
Cheson
BD
Cassileth
PA
Head
DR
et al
Report of the National Cancer Institute-sponsored workshop on definitions of diagnosis and response in acute myeloid leukemia.
J Clin Oncol.
8
1990
813
819
25
Sadowski
HB
Shuai
K
Darnell
JE
Jr
Gilman
MZ
A common nuclear signal transduction pathway activated by growth factor and cytokine receptors.
Science.
261
1993
1739
1744
26
Brechner
T
Hocke
G
Goel
A
Fey
GH
Interleukin-6 response factor binds cooperatively at two adjacent sites in the promoter upstream region of the rat alpha 2-macroglobulin gene.
Mol Biol Med.
8
1991
267
285
27
Mui
AL
Wakao
H
O'Farrell
AM
Harada
N
Miyajima
A
Interleukin-3, granulocyte-macrophage colony stimulating factor and interleukin-5 transduce signals through two STAT5 homologs.
EMBO J.
14
1995
1166
1175
28
Kaplan
EL
Meier
P
Nonparametric estimation from incomplete observations.
J Am Stat Assoc.
53
1958
457
481
29
Mantel
N
Evaluation of survival data and two new rank order statistics arising in its consideration.
Cancer Chemother Rep.
50
1966
163
170
30
Norusis
MJ
SPSS for Windows Advanced Statistics User Manual, Release 6.0.
1993
275
320
IL
Chicago
31
Fisher
RA
The Design of Experiments.
1935
Oliver and Boyd
Edinburg, Scotland
32
Kruskal
WH
Wallis
WA
Use of ranks in one-criterion variance analysis.
J Am Stat Assoc.
47
1952
583
621
33
Mann
HB
Whitney
DR
Annals of Mathematical Statistics.
18
1947
50
54
34
Lowenberg
B
Downing
JR
Burnett
A
Acute myeloid leukemia.
N Engl J Med.
341
1999
1051
1062
35
Weber-Nordt
RM
Egen
C
Wehinger
J
et al
Constitutive activation of STAT proteins in primary lymphoid and myeloid leukemia cells and in Epstein-Barr virus (EBV)-related lymphoma cell lines.
Blood.
88
1996
809
816
36
Hayakawa
F
Towatari
M
Iida
H
et al
Differential constitutive activation between STAT-related proteins and MAP kinase in primary acute myelogenous leukaemia.
Br J Haematol.
101
1998
521
528
37
Gouileux-Gruart
V
Gouileux
F
Desaint
C
et al
STAT-related transcription factors are constitutively activated in peripheral blood cells from acute leukemia patients.
Blood.
87
1996
1692
1697
38
Schuringa
JJ
Wierenga
AT
Kruijer
W
Vellenga
E
Constitutive Stat3, Tyr705, and Ser727 phosphorylation in acute myeloid leukemia cells caused by the autocrine secretion of interleukin-6.
Blood.
95
2000
3765
3770
39
Koistinen
P
Saily
M
Poromaa
N
Savolainen
ER
Complex effects of interleukin 6 on clonogenic blast cell growth in acute myeloblastic leukemia.
Acta Haematol.
98
1997
14
21
40
Wetzler
M
Bernstein
SH
Baumann
H
et al
Expression and function of the megakaryocyte growth and development factor receptor in acute myeloid leukemia blasts.
Leuk Lymphoma.
30
1998
415
431
41
Bouscary
D
Prudhomme
C
Quesnel
B
Melle
J
Picard
F
Dreyfus
F
c-mpl expression in hematologic malignancies.
Leuk Lymphoma.
17
1995
19
26
42
Quentmeier
H
Zaborski
M
Graf
G
Ludwig
WD
Drexler
HG
Expression of the receptor MPL and proliferative effects of its ligand thrombopoietin on human leukemia cells.
Leukemia.
10
1996
297
310
43
Yu
CL
Meyer
DJ
Campbell
GS
et al
Enhanced DNA-binding activity of a Stat3-related protein in cells transformed by the Src oncoprotein.
Science.
269
1995
81
83
44
Bromberg
JF
Horvath
CM
Besser
D
Lathem
WW
Darnell
JE
Jr
Stat3 activation is required for cellular transformation by v-src.
Mol Cell Biol.
18
1998
2553
2558
45
Turkson
J
Bowman
T
Garcia
R
Caldenhoven
E
De Groot
RP
Jove
R
Stat3 activation by Src induces specific gene regulation and is required for cell transformation.
Mol Cell Biol.
18
1998
2545
2552
46
Yu
CL
Jove
R
Burakoff
SJ
Constitutive activation of the Janus kinase-STAT pathway in T lymphoma overexpressing the Lck protein tyrosine kinase.
J Immunol.
159
1997
5206
5210
47
Catlett-Falcone
R
Landowski
TH
Oshiro
MM
et al
Constitutive activation of Stat3 signaling confers resistance to apoptosis in human U266 myeloma cells.
Immunity.
10
1999
105
115
48
Puthier
D
Bataille
R
Amiot
M
IL-6 up-regulates mcl-1 in human myeloma cells through JAK/STAT rather than ras/MAP kinase pathway.
Eur J Immunol.
29
1999
3945
3950

Author notes

Meir Wetzler, Leukemia Section, Dept of Medicine, Roswell Park Cancer Institute, Elm and Carlton Sts, Buffalo, NY 14263; e-mail: meir.wetzler@roswellpark.org.

Sign in via your Institution