Vascular endothelial growth factor (VEGF) is a potent mitogen for vascular endothelial cells. It has been associated with angiogenesis, growth, dissemination, metastasis, and poor outcome in solid tumors. To assess cellular VEGF levels and their prognostic significance in newly diagnosed acute myeloid leukemia (AML), we used a radioimmunoassay (RIA) to quantify VEGF levels in stored samples obtained before treatment from 99 patients with newly diagnosed AML treated at the MD Anderson Cancer Center from 1996 to 1998. Outcome in the 99 patients was representative of that observed in all patients seen at this institution with this diagnosis during these years, but the 99 patients had higher white blood cell (WBC) and blast counts than the other patients. Results of the RIA were confirmed by Western blot. There was a relationship between increasing VEGF levels and shorter survival (P = .01), as well as shorter disease-free survival, both from start of treatment and from complete response (CR) date. In contrast, there was no relationship between VEGF level and WBC or blast count, or between VEGF level and such established prognostic factors as age, cytogenetics, performance status, or presence of an antecedent hematologic disorder, and multivariate analysis indicated that VEGF was still prognostic for the above outcomes after accounting for these factors, as well as treatment. Our results suggest that at least in AML patients with higher WBC and blast counts, cellular VEGF level is an independent predictor of outcome.

VASCULAR ENDOTHELIAL growth factor (VEGF) is a 34- to 42-kD dimeric multifunctional glycoprotein with a 15% to 25% homology with platelet-derived growth factor (PDGF).1,2 The gene for human VEGF is located on chromosome 6p21.3.3 It contains 8 exons and 7 introns.4Several isoforms of VEGF have been described including 121-, 165-, 189-, and 206-amino acid forms.4 The 165-amino acid isoform appears to be the most common.5 VEGF is a potent mitogen for endothelial cells isolated from arteries, veins, and lymphatics. Overexpression of VEGF is associated with increased angiogenesis,6-11 growth, invasion, and metastasis in solid tumors.12-17 Furthermore, correlations have been reported between higher VEGF levels and poor prognosis in solid tumors.18-21 

VEGF was originally cloned from the leukemic cell line HL60,22 but the role of VEGF and the role of angiogenesis in leukemia have received only limited attention. Perez-Atayde et al23 observed heightened angiogenesis in the marrow in pediatric acute lymphoid leukemia and increased concentrations of basic fibroblast growth factor (bFGF), a mediator of angiogenesis, in patients with this disease. Fiedler et al24 found VEGF transcription in 23 of 33 (69%) patients with acute myeloid leukemia (AML). Leukemic cell cultures from 24 of these patients produced significantly higher VEGF levels than CD34-enriched cell cultures obtained from normal volunteer donors.24 

The purpose of this study was to examine the prognostic significance of cellular VEGF levels in newly diagnosed AML.

Patients and controls.

Pretreatment cellular VEGF concentrations were measured in peripheral blood and marrow samples obtained from 99 patients with newly diagnosed AML treated at the MD Anderson Cancer Center between 1996 and 1998. Samples were obtained at presentation and stored at −70°C until analysis. Samples were used if blasts comprised more than 70% of the mononuclear cells. VEGF was measured in blasts from 34 peripheral blood samples and 65 bone marrow samples. In 9 patients, VEGF was measured in both bone marrow and peripheral blood blasts. Table 1 compares the 99 patients with the other 167 patients treated here for newly diagnosed AML in the 1996 to 1998 period in whom samples were not stored. The study group had higher WBC and blast cell counts, leading to the decision to store the samples. Reflecting these higher counts, the study group more frequently had inv(16), but the 2 groups were similar with regard to other standard prognostic factors such as age, performance status, history of abnormal blood counts (AHD) (hemoglobin <12 g/dL, platelet count <150 × 109/L, neutrophil count <1.5 × 109/L, or WBC >20 × 109/L) for ≥1 month before diagnosis of AML, and presence of chromosome 5 and/or 7 abnormalities. Most importantly, the patients who were studied and those who were not had similar outcomes.

Table 1.

Characteristics of AML Patients at MD Anderson Cancer Center

VEGF Measured (%) VEGF not Measured (%)P Value
Number  99  167  
Median age 
Zurbrod >2  14 (14)  23 (14)  .94  
APL  9 (9) 12 (7)  .58  
t(8;21), or inv(16), or t(15;17)  17 (17) 15 (9)  .05  
Normal karyotype  34 (35)  59 (35) .99  
−5/−7  23 (24)  33 (20)  .50  
Other karyotype  25 (26)  60 (36)  .07  
AHD  34 (34) 71 (43)  .25  
WBC (×109/L)  14.10 6.0  <.0001  
% Circulating blasts  31  <.0001  
Treatment  
 Idarubicin + Ara-C  30 34  
 Fludarabine + Idarubicin + Ara-C  53  109 .24  
 Topotecan + Ara-C  7  12  
 APL treatment 9  12  
% CR  65 (66)  102 (61)  .453  
Deaths days 1-35  14 (14)  35 (21)  .165  
Median CR duration (wk)  46  51  .38  
Median survival (wk)  45  33 .33  
Median follow-up time censored patients  89 wk 81 wk 
VEGF Measured (%) VEGF not Measured (%)P Value
Number  99  167  
Median age 
Zurbrod >2  14 (14)  23 (14)  .94  
APL  9 (9) 12 (7)  .58  
t(8;21), or inv(16), or t(15;17)  17 (17) 15 (9)  .05  
Normal karyotype  34 (35)  59 (35) .99  
−5/−7  23 (24)  33 (20)  .50  
Other karyotype  25 (26)  60 (36)  .07  
AHD  34 (34) 71 (43)  .25  
WBC (×109/L)  14.10 6.0  <.0001  
% Circulating blasts  31  <.0001  
Treatment  
 Idarubicin + Ara-C  30 34  
 Fludarabine + Idarubicin + Ara-C  53  109 .24  
 Topotecan + Ara-C  7  12  
 APL treatment 9  12  
% CR  65 (66)  102 (61)  .453  
Deaths days 1-35  14 (14)  35 (21)  .165  
Median CR duration (wk)  46  51  .38  
Median survival (wk)  45  33 .33  
Median follow-up time censored patients  89 wk 81 wk 

To show the presence of VEGF, we used Western blot. Quantification of VEGF was performed with solid-phase radioimmunoassay (RIA). We describe these methods below.

Protein extraction.

Protein extraction was performed as previously described.25Cell pellets were lysed for 30 minutes on ice in TENN buffer (50 mmol/L Tris-HCL [pH 7.4], 5 mmol/L EDTA, 0.5% Nonidet P-40, and 150 mmol/L NaCl supplemented with 1 mmol/L phenylmethylsulfonyl fluoride, 2 μg/mL leupeptin, and 2 μg/mL pepstatin). Frequent vortexing was performed, and samples were then left on ice for 1 hour. Lysates were clarified by microcentrifugation for 1 hour at 14,000 rpm. Protein concentration was determined by the Bradford method, and 200 μg of cell extract was run on a 9.5% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), and stained with coomassie blue R-250 to check the protein profile and amount of protein loaded.

Western blot analysis of VEGF protein.

Two hundred micrograms of cell extract from AML patients and from normal individuals was electrophoretically separated on 12.5% SDS-PAGE gels and were transferred to nitrocellulose membrane papers. The nitrocellulose membranes were blocked for 6 to 8 hours at room temperature with 5% nonfat milk in phosphate-buffered saline (PBS) containing 0.1% Tween 20 and 0.01% sodium azide. The blots were then incubated over night at 4°C with goat anti-VEGF polyclonal antibody (R&D Systems, Minneapolis, MN), at a concentration of 1 μg/mL in PBS containing 2.5% nonfat milk, 2.5% bovine serum albumin (BSA), and 0.1% Tween 20. The membranes were washed with PBS containing 0.1% Tween 20. The blots then were incubated with 1:2,000 diluted antigoat immunoglobulin linked to horseradish peroxidase (Sigma, St Louis, MO) in PBS containing 1% nonfat milk and 0.1% Tween 20. Immunoreactive bands were developed using the ECL detection system (Amersham, Arlington Heights, IL). After ECL detection, the membranes were stripped off from primary and secondary antibodies under conditions recommended by Amersham Inc, and the stripped membranes were then blocked and probed with antiactin monoclonal antibody IgM (Amersham) to check for equal loading of protein in each lane. The Western blot bands were scanned and intensity analyzed using Scan Analysis software from Biosoft (Cambridge, UK). There was no visible band in samples with VEGF less than 2 as determined by RIA (see below). However, considering cases with visible bands, we found good correlation between Western blot and RIA with R = 0.72.

Solid-phase RIA.

VEGF protein levels were measured using solid-phase RIA as previously detailed.25 Microtiter plates were coated overnight at 4°C with 5 μg of protein extracted from AML patients and normal individuals in 50 μL of PBS. The RIA plates were then washed with PBS and blocked with 100 μL of 1% BSA (Amersham) in PBS for 1 hour at 37°C. The plates were incubated overnight at 4°C with 50 μL of rabbit anti-VEGF antibody (Santa Cruz Biotech, Santa Cruz, CA) diluted 1:1,000 in PBS containing 1% BSA. The plates were then washed with PBS and amplified with goat anti-rabbit IgG antisera (Sigma) diluted 1:1,000 in 0.1% BSA in PBS for 2 hours at 37°C. After washing, the plates were developed for 2 hours at room temperature with excess 125I-labeled protein G in 0.1% BSA in PBS per well. The specific activity in each well varied between 150,000 to 200,000 cpm dependent on the activity of the 125I. They were then washed with PBS, separated into individual wells, and the counts in each well were recorded with a gamma counter (LKB Biotechnology, Uppsala, Sweden). The assays were performed in triplicate, and the results were corrected for the nonspecific binding (1% to 2%) detected in control wells, which were not coated with a test antigen, but were blocked with BSA. A second set of plates was incubated with antiactin antibodies to confirm the use of equal amount of total cellular protein from each sample. Each sample was tested in triplicate and repeated twice in 2 different experiments. No significant difference was observed between measurements. Normal samples were repeated with each experiment and each plate contained 2 or more normal samples. The linear range of the RIA was determined by using mixing studies and purified VEGF from a commercially available kit (R&D System). The levels of VEGF detected using cell lysates (5 μg of total cell lysate) is extremely low and below the linear range of the commercially available ELISA assay. However, using diluted purified VEGF obtained from R&D, we established the linear range for our RIA as between 0.5 and 62 pg. All analyzed samples were in the linear range (1 to 10 pg). The median cpm detected in 31 normal bone marrow samples was assigned a score of 1 and the levels of the VEGF in AML samples were normalized to the median of the normal bone marrow. However, some VEGF protein expression was detected in normal individuals by solid phase RIA. The range of the VEGF in normal bone marrow samples was between 0.84 and 1.16.

Statistical methods.

Associations between patient characteristics (covariates) were assessed for pairs of numerical variables by Spearman correlation, for categorical and continuous variables by Wilcoxon-Mann-Whitney and Kruskal-Wallis test statistics, and for pairs of categorical variables by the Fisher exact test and its generalizations. Patients with acute progranulocytic leukemia (APL) were excluded from analysis of the effect of VEGF on treatment outcome. The outcomes studied were achievement of complete response (CR), survival, and disease-free survival from start of treatment and from CR date (events = relapse, death in CR). We examined the following covariates as assessed before treatment as potential predictors of outcome: age, WBC count, platelet count, performance status (Zubrod 0 to 2 v 3 to 4), treatment in the protected environment (PE), cytogenetics (normal karyotype, including patients with insufficient metaphases analysis vinv[16] or t[8;21] v −5, 5q−, −7 or 7q− [−5/−7] v other abnormalities), presence of an AHD, VEGF level, and whether the source of VEGF was marrow or peripheral blood blasts. We also examined the effect of treatment (Idarubicin + Ara-C v Idarubicin + Ara-C + Fludarabine). Logistic regression was used to assess the ability of the patients’ characteristics to predict the probability of CR, with goodness-of-fit assessed by residual and partial residual scatter plots and likelihood ratio (LR) statistics. Unadjusted time-to-event analyses were performed using Kaplan-Meier plots. The Cox proportional hazards model26 and its generalizations27 were used to assess the ability of treatment groups and patient characteristics to predict survival, with goodness-of-fit assessed by the Grambsch-Therneau test,27 Schoenfeld residual plots, martingale residual plots,28 and LR statistics. All scatterplots were smoothed using the lowess methods of Cleveland,29 with variables transformed as appropriate based on these plots. Multivariate logistic and Cox models were obtained by performing a backward elimination with P value cut-off .05, then allowing any variable previously deleted to enter the final model if its P value was <.05. All computations were performed on a DEC Alpha 2100 5/250 system computer (Digital Electronics Corp, Nashua, NH) in StatXact (Cytel Software Corp, Cambridge, MA) and S plus, using both standard S plus functions and the survival analysis package of Therneau (gift of Dr T.M. Therneau, Mayo Clinic, Rochester, MN).

The amount of VEGF detected using Western blot was in good agreement with the amount detected using RIA (Fig 1). The RIA values of AML samples were normalized to the RIA values of normal bone marrow, which were assigned a value of 1. There was considerable variability in the patients’ RIA values (median, 2.8; mean, 2.9; average, 0.5 to 8.1). While we did not asses the effect of length of storage on VEGF levels by comparing the levels in a given sample as measured in 1996 and as measured again in 1997 or 1998, those VEGF samples obtained in 1996 had similar values as those obtained in 1997 and 1998. However, VEGF levels as measured in blood were higher than those measures in marrow (mean, 3.1 v 2.8; median, 3.6v 2.9), although the difference was only marginally significant (P = .08). There was a strong correlation between VEGF as quantified by RIA in blood and quantified in marrow in 9 patients in whom we have both samples available (P = < .01).

Fig. 1.

Western blot. In lane C is a positive control, the next 2 lanes show control from 2 normal bone marrows. Lanes 1 through 13 show a considerable variability in expression of VEGF that was confirmed with RIA.

Fig. 1.

Western blot. In lane C is a positive control, the next 2 lanes show control from 2 normal bone marrows. Lanes 1 through 13 show a considerable variability in expression of VEGF that was confirmed with RIA.

Close modal

There was no relationship between VEGF concentration and such standard prognostic factors as age (P = .57), performance status (P = .12), karyotype (P = .40) (Table 2), or presence of antecedent hematologic disease probably signifying a prior myelodysplastic syndrome. There was also no correlation between VEGF concentration and pretreatment blood count. After excluding the 9 of our 99 patients who had APL, we compared CR rate and survival according to whether VEGF was above or below the median value (2.9). The CR rate was 64% in both groups and survival was also the same in both. However, it was possible that the relationship between VEGF and outcome was more complicated, and to examine the specific nature of this relationship, we used Martingale residual plots. These suggested that as VEGF continued to increase to a value of about 1.7, CR rate continued to decrease, and survival continued to shorten. At VEGF levels >1.7, there was no further effect. Hence, we compared CR rates and survival in patients who had VEGF levels above and below 1.7. Table 2 illustrates that CR rates were 59% and 84% in patients with VEGF levels above and below this value, respectively. The difference in CR rates was a result of a higher incidence of “resistance,” ie, failure to achieve CR despite living at least 35 days from the start of induction therapy.

Table 2.

CR Rates by VEGF Level

VEGF <1.7 (%) VEGF >1.7 (%) P Value
Patients  19  71  
CR  16 (84)  42 (59) .04  
Died before day 35  2 (11)  10 (14)  .69 
Lived more than 35 days, but no CR  1 (5)  18 (27) .05 
VEGF <1.7 (%) VEGF >1.7 (%) P Value
Patients  19  71  
CR  16 (84)  42 (59) .04  
Died before day 35  2 (11)  10 (14)  .69 
Lived more than 35 days, but no CR  1 (5)  18 (27) .05 

The lack of an association between VEGF levels and conventional prognostic factors noted above suggested that VEGF was an independent predictor factor at least for CR. We tested this suggestion by fitting a logistic model for CR and Cox model for survival and disease-free survival from start of therapy and from CR date (Table 3). In these models, we examined VEGF as a continuos variable as suggested by the Martingale residual plots. Table 3 shows that VEGF was a predictor of each outcome even after accounting for the other predictors. Of note, the source of the cells used to measure VEGF (marrow v peripheral blood) was not a predictor of outcome, or was the year when the sample was obtained. This suggests that results were not biased by the length of sample storage, eg, systematically lower levels in samples obtained in 1996 than in samples obtained in 1998. Within the limits of small numbers, there was no interaction between VEGF and cytogenetic groups, ie, the effect of VEGF on outcome was not greater in any cytogenetic group. Treatment with fludarabine-containing regimens was associated with poor outcome as previously noted,30,31 but the effect of VEGF on outcome remained after accounting for treatment.

Table 3.

Multivariate Analysis of Effects of VEGF on Outcome

Outcome Independent Predictors of Unfavorable OutcomeP Value
CR  VEGF  .048  
 Increasing age .049  
 Zubrod performance status 3-4  .042 
 Treatment with Fludarabine  .002  
DFS  VEGF  .010 
 −5/−7  .003  
 Increasing age  .017 
 Treatment with Fludarabine  .011  
 Treatment in laminar air flow room  .027  
DFS from CR  VEGF  .002 
 −5/−7  <.0001  
Survival  VEGF  .019 
 Increasing age  <.0001  
 Treatment with Fludarabine .04  
 Treatment in laminar air flow room  .002 
Outcome Independent Predictors of Unfavorable OutcomeP Value
CR  VEGF  .048  
 Increasing age .049  
 Zubrod performance status 3-4  .042 
 Treatment with Fludarabine  .002  
DFS  VEGF  .010 
 −5/−7  .003  
 Increasing age  .017 
 Treatment with Fludarabine  .011  
 Treatment in laminar air flow room  .027  
DFS from CR  VEGF  .002 
 −5/−7  <.0001  
Survival  VEGF  .019 
 Increasing age  <.0001  
 Treatment with Fludarabine .04  
 Treatment in laminar air flow room  .002 

Our data suggest that VEGF levels are, on average, higher in AML than in normal bone marrow. Normal marrow contains relatively few blasts, and high expression in a small number of normal blasts could easily have been missed. However, Fiedler et al24 have reported that normal CD34+ marrow cells have low VEGF levels. The principal purpose of this report was to examine the possible prognostic significance of cellular VEGF in AML. Our results suggest that cellular VEGF levels are prognostic in newly-diagnosed AML (Tables 3) (APL patients excluded from the analysis). Although we did not study consecutive patients, the population we analyzed was representative of our newly-diagnosed AML population (Table 1) with the principal exception of higher white blood cell and blast counts and a slightly higher incidence of inv(16). The higher white blood cell counts reflect the selective use of samples containing a high number of blasts so as to facilitate VEGF measurements. Because of the selected nature of our population, our conclusions about the prognostic significance of VEGF cannot be generalized to the usual AML population, which of course has a lower white blood cell count (Table 1). Three other possibly confounding factors are the various treatments the patients received (Table 1), the possible effect of storage on VEGF levels, and the source of the cells used to measure VEGF. However, multivariate analysis found that VEGF was still prognostic after accounting for the unfavorable effect of fludarabine, and found that outcome was unaffected by the year (1996, 1997, 1998) the sample was obtained or whether the cells were obtained from blood or bone marrow.

The mechanism by which high cellular VEGF levels affect prognosis is not clear. We are currently attempting to correlate VEGF levels with marrow microvascular density and with plasma VEGF levels. Such levels were unavailable in the 99 patients studied here. Regardless of the mechanism, VEGF appears to be prognostic in at least the subset of newly diagnosed AML patients presenting with relatively high WBC and high blast count.

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

1
Thomas
KA
Vascular endothelial growth factor, a potent and selective angiogenic agent.
J Biol Chem
271
1996
603
2
Fox
SB
Tumour angiogenesis and prognosis.
Histopathology
30
1997
294
3
Vincenti
V
Cassano
C
Rocchi
M
Persico
G
Assignment of the vascular endothelial growth factor gene to human chromosome 6p21.3.
Circulation
93
1996
1493
4
Tischer
E
Mitchell
R
Hartman
T
Silva
M
Gospodarowicz
D
Fiddes
JC
Abraham
JA
The human gene for vascular endothelial growth factor. Multiple protein forms are encoded through alternative exon splicing.
J Biol Chem
266
1991
11947
5
Ferrara
N
Vascular endothelial growth factor.
Eur J Cancer
32A
1996
2413
6
Toi
M
Taniguchi
T
Yamamoto
Y
Kurisaki
T
Suzuki
H
Tominaga
T
Clinical significance of the determination of angiogenic factors.
Eur J Cancer
32A
1996
2513
7
Ferrara
N
Henzel
WJ
Pituitary follicular cells secrete a novel heparin-binding growth factor specific for vascular endothelial cells.
Biochem Biophys Res Commun
161
1989
851
8
Plouet
J
Schilling
J
Gospodarowicz
D
Isolation and characterization of a newly identified endothelial cell mitogen produced by AtT-20 cells.
EMBO J
8
1989
3801
9
Pepper
MS
Wasi
S
Ferrara
N
Orci
L
Montesano
R
In vitro angiogenic and proteolytic properties of bovine lymphatic endothelial cells.
Exp Cell Res
210
1994
298
10
Jakeman
LB
Winer
J
Bennett
GL
Altar
CA
Ferrara
N
Binding sites for vascular endothelial growth factor are localized on endothelial cells in adult rat tissues.
J Clin Invest
89
1992
244
11
Jakeman
LB
Armanini
M
Phillips
HS
Ferrara
N
Developmental expression of binding sites and messenger ribonucleic acid for vascular endothelial growth factor suggests a role for this protein in vasculogenesis and angiogenesis.
Endocrinology
133
1993
848
12
Folkman
J
Tumor angiogenesis
The Molecular Basis of Cancer.
Mendelsohon
J
Howley
PM
Israel
MA
Liotta
LA
1995
206
Saunders
Philadelphia, PA
13
Folkman
J
Tumor angiogenesis: Therapeutic implications.
N Engl J Med
285
1971
1182
14
Folkman
J
Angiogenesis in cancer, vascular, rheumatoid and other disease.
Nat Med
1
1995
27
15
Folkman
J
Tumor Angiogenesis
Cancer Medicine, vol 1.
Holland
JF
Frei
E
III
Bast
RC
Jr
Kufe
DW
Morton
DL
Weichselbaum
RR
1993
153
Lea & Febiger
Philadelphia, PA
16
Folkman
J
Seminars in medicine of the Beth Israel Hospital, Boston. Clinical applications of research on angiogenesis.
N Engl J Med
333
1995
1757
17
Folkman
J
What is the evidence that tumors are angiogenesis dependent?.
J Natl Cancer Inst
82
1990
4
18
O’Brien
T
Cranston
D
Fuggle
S
Bicknell
R
Harris
AL
Different angiogenic pathways characterize superficial and invasive bladder cancer.
Cancer Res
55
1995
510
19
Toi
M
Hoshina
S
Takayanagi
T
Tominaga
T
Association of vascular endothelial growth factor expression with tumor angiogenesis and with early relapse in primary breast cancer.
Jpn J Cancer Res
85
1994
1045
20
Takahashi
Y
Kitadai
Y
Bucana
CD
Cleary
KR
Ellis
LM
Expression of vascular endothelial growth factor and its receptor, KDR, correlates with vascularity, metastasis, and proliferation of human colon cancer.
Cancer Res
55
1995
3964
21
Salven
P
Ruotsalainen
T
Mattson
K
Joensuu
H
High pre-treatment serum level of vascular endothelial growth factor (VEGF) is associated with poor outcome in small-cell lung cancer.
Int J Cancer
79
1998
144
22
Leung
DW
Cachianes
G
Kuang
WJ
Goeddel
DV
Ferrara
N
Vascular endothelial growth factor is a secreted angiogenic mitogen.
Science
246
1989
1306
23
Perez-Atayde
AR
Sallan
SE
Tedrow
U
Connors
S
Allred
E
Folkman
J
Spectrum of tumor angiogenesis in the bone marrow of children with acute lymphoblastic leukemia.
Am J Pathol
150
1997
815
24
Fiedler
W
Graeven
U
Ergun
S
Verago
S
Kilic
N
Stockschlader
M
Hossfeld
DK
Vascular endothelial growth factor, a possible paracrine growth factor in human acute myeloid leukemia.
Blood
89
1997
1870
25
Starostik
P
Manshouri
T
O’Brien
S
Freireich
E
Kantarjian
H
Haidar
M
Lerner
S
Keating
M
Albitar
M
Deficiency of the ATM protein expression defines an aggressive subgroup of B-cell chronic lymphocytic leukemia.
Cancer Res
58
1998
4552
26
Fleming
TR
Harrington
DP
Counting Processes and Survival Analysis.
1991
Wiley
New York, NY
27
Grambsch
PM
Therneau
TM
Proportional hazards tests and diagnostics based on weighted residuals.
Biometrika
81
1994
515
28
Cleveland
WS
Robust locally-weighted regression and smoothing scatterplots.
J Am Stat Assoc
74
1979
829
29
Becker
RA
Chambers
JM
Wilks
AR
The New S Language.
1988
Wadsworth
Pacific Grove, CA
30
Estey
E
Thall
P
Beran
M
Kantarjian
H
Pierce
S
Keating
M
Effect of diagnosis (RAEB, RAEB-t, or AML) on outcome of AML-type chemotherapy.
Blood
90
1997
2969
31
Estey
EH
Thall
PF
Pierce
S
Cortes
J
Beran
M
Kantarjian
H
Keating
MJ
Andreeff
M
Freireich
E
Randomized phase II study of fludarabine + cytosine arabinoside + idarubicin ± all-trans retinoic acid ± granulocyte-colony stimulating factor in poor prognosis newly-diagnosed non-APL AML and MDS.
Blood
93
1999
2478

Author notes

Address reprint requests to Maher Albitar, MD, Section of Hematopathology, Division of Laboratory Medicine, The University of Texas, MD Anderson Cancer Center, 1515 Holcombe Blvd, Box 72, Houston, TX 77030-4095; e-mail: malbitar@mdacc.tmc.edu.

Sign in via your Institution