To assess the prognostic impact of cytogenetics in elderly patients with acute myeloid leukemia (AML) receiving intensive induction and consolidation treatment according to a single protocol specifically designed for patients above age 60, pretreatment samples from 361 patients registered for the AML HD98-B trial of the German-Austrian AML Study Group were analyzed by chromosome banding and fluorescence in situ hybridization, and cytogenetic findings were correlated with outcome. Using a proportional hazards model with backward selection, 3 prognostic subgroups were identified based on the influence of cytogenetic abnormalities on overall survival (OS): low-risk, t(15;17), and inv(16) in 25 of 361 patients (7%); standard-risk, normal karyotype, t(8;21), t(11q23), +8 within a noncomplex karyotype, and +11 within a noncomplex karyotype in 208 of 361 patients (58%); high-risk, all other aberrations in 128 of 361 patients (35%). On multivariate analysis, high-risk cytogenetics (hazard ratio [HR], 2.24) and age above 70 years (HR, 2.34) were independent prognostic factors affecting OS, and stratification according to these parameters demonstrated that a large subgroup of patients (55%), characterized by age 70 or older or high-risk cytogenetics, or both, had very unfavorable treatment results despite intensive chemotherapy. Thus, karyotype and age are major determinants of outcome in elderly patients with AML.

Pretreatment karyotype is the most important predictor of outcome in adult acute myeloid leukemia (AML).1  In younger patients with AML, analysis of chromosome abnormalities allows categorization into favorable, intermediate, and adverse risk groups; most stratification systems distinguish t(8;21), inv(16), and t(15;17) as conferring a significantly better prognosis than normal karyotype, whereas other aberrations, such as –5/del(5q), –7, abn(3q), and complex karyotype, are associated with a high risk for induction failure, relapse, and shortened survival.2-5 

AML occurring in patients older than 55 to 60 years of age is characterized by profound biologic differences, including the distribution, but not the spectrum, of cytogenetic abnormalities. In particular, favorable-risk aberrations are relatively uncommon in the elderly, as opposed to normal and complex karyotypes.6-10 

Data regarding the prognostic relevance of cytogenetic abnormalities in older patients with AML receiving intensive chemotherapy are limited, and only 2 studies have assessed the impact of specific chromosome aberrations in elderly patients treated according to a single protocol. Grimwade et al7  found that diagnostic karyotype was a prognostic factor for outcome in 1065 patients (median age, 66 years) from the United Kingdom Medical Research Council (MRC) AML11 trial designed for patients aged 56 years and older. Based on their results, they proposed a hierarchical cytogenetic classification that identified subgroups with significantly different outcomes: patients with t(8;21), inv(16), or t(15;17) had a favorable prognosis, whereas, in the absence of these changes, the presence of 5 or more abnormalities predicted an adverse prognosis; the remaining patients, by far the largest group and including the patients with normal karyotype, had an intermediate prognosis. Rowe et al11  assessed the prognostic value of the 3-subgroup cytogenetic risk classification for younger AML patients developed by the Southwest Oncology Group5  in 213 patients older than age 55 who were entered into a trial conducted by the Eastern Cooperative Oncology Group. This analysis showed that patients with unfavorable karyotypes (n = 88), defined by the presence of inv(3), –5/del(5q), t(6;9), –7/del(7q), t(9;22), abn(9q), abn(11q), abn(17p), abn(20q), abn(21q), or 3 or more chromosome aberrations, had significantly inferior complete remission (CR) rates and median overall survival (OS) than patients with intermediate-risk cytogenetics (n = 122), defined as normal karyotype or the presence of –Y, +6, +8, or del(12p), or favorable cytogenetics (n = 3), defined as the presence of t(8;21), inv(16), or t(15;17).

We analyzed the prognostic value of pretreatment cytogenetic abnormalities, as assessed by chromosome banding and fluorescence in situ hybridization (FISH), in 361 patients older than 60 years registered for the AML HD98-B trial of the German-Austrian AML Study Group (AMLSG).12,13 

Patients and treatment

The study included 361 patients older than 60 years with AML, de novo or secondary (after treatment for a primary malignancy or following myelodysplasia), as defined by the French-American-British (FAB) classification.

All patients were registered for the AML HD98-B trial of the AMLSG.12,13  Randomized induction therapy consisted of 2 courses of 12 mg/m2 idarubicin on days 1 and 3, 100 mg/m2 cytarabine continuously on days 1 through 5, and 100 mg/m2 etoposide on days 1 and 3 (ICE) with or without all-transretinoic acid (ATRA; 45 mg/m2 on days 3 through 5 and 15 mg/m2 on days 6 through 28), followed by first consolidation therapy with a course of 0.5 g/m2 cytarabine every 12 hours on days 1 through 3 and 10 mg/m2 mitoxantrone on days 2 and 3 (HAM) with or without ATRA (15 mg/m2 on days 3 through 28). For further postremission therapy, patients were randomized to intensive second consolidation therapy (12 mg/m2 idarubicin on days 1 and 3, 100 mg/m2 etoposide on days 1 through 5) or 12 monthly courses of outpatient maintenance therapy (5 mg idarubicin orally on days 1, 4, 7, 10, and 13 and 100 mg etoposide orally on days 1 and 13). Patients not responding to the first course of induction therapy were assigned to receive a course of 0.5 g/m2 cytarabine every 12 hours on days 1 through 3, 250 mg/m2 etoposide continuously on days 4 and 5, 45 mg/m2 ATRA on days 3 through 5, and 15 mg/m2 ATRA on days 6 through 28 (A-HAE). Patients with acute promyelocytic leukemia (APL) did not undergo randomization but were treated with a course of 12 mg/m2 idarubicin continuously on days 2, 4, and 6, 45 mg/m2 ATRA on days 1 through 6, and 15 mg/m2 ATRA on days 7 through 35 (AIDA), followed by a course of A-HAM and 6 monthly courses of outpatient maintenance therapy (5 mg idarubicin on days 1, 4, 7, 10, and 13 and 15 mg/m2 ATRA on days 1 through 28).

The study was approved by the institutional review boards of the participating centers. Informed consent was obtained from all patients according to the Declaration of Helsinki.

Cytogenetic and molecular cytogenetic analysis

All leukemia samples were studied centrally in the Laboratory for Cytogenetic and Molecular Diagnostics of the AMLSG. Chromosome banding was performed using standard techniques, and karyotypes were described according to the International System for Human Cytogenetic Nomenclature. Complex karyotype was defined as 3 or more cytogenetic abnormalities in the absence of t(8;21), t(11q23), t(15;17), or inv(16)/t(16; 16), unless otherwise stated.

To improve cytogenetic diagnostics, all specimens were also analyzed by FISH using a comprehensive DNA probe set for the detection of the following AML-associated genomic aberrations14 : inv(3)/t(3;3), t(8;21), t(9;22), t(11q23), t(15;17), inv(16)/t(16;16), +3q, +4q, del(5q), del(7q), +8q, +11q, abn(12p), del(13q)/+13q, del(17p), del(20q), +21q, +22q, and del(Xq).

Criteria for treatment outcomes

Response to induction therapy was assessed after 2 courses of chemotherapy. In accordance with standard criteria,15  CR was defined as less than 5% bone marrow (BM) blasts, an absolute neutrophil count of 1.0 × 109/L or more, a platelet count of 100 × 109/L or more, no blasts in the peripheral blood (PB), and no extramedullary leukemia. Therapeutic failures were classified as either refractory disease (RD) or early/hypoplastic death (ED/HD), which was death less than 7 days after completion of the first course of induction therapy/death during the remainder of double-induction therapy. Relapse was defined as more than 5% BM blasts unrelated to recovery from the preceding course of chemotherapy or new extramedullary leukemia in patients with previously documented CR. End points for OS, measured from the date of study entry, were death (failure) and alive at last follow-up (censored). Analyses of cumulative incidence of relapse (CIR) and cumulative incidence of death in CR (CID) included only patients attaining CR, with time calculated from the date of CR to the occurrence of an event (relapse or death).

Statistical analyses

The median duration of follow-up was calculated according to the method of Korn.16  The Kaplan-Meier method was used to estimate the distribution of OS.17  Confidence interval (CI) estimation was based on the cumulative hazard using the Greenwood formula for standard error (SE) estimation.18  Cumulative incidence of relapse, CID, their SEs, and differences between groups were estimated according to Gray.19  Survival distributions were compared using the log-rank test. A Cox model was used to evaluate prognostic variables.20  Missing data were estimated with a multipleimputation technique using predictive mean matching with n = 100 for multiple imputations.21  Backward selection applying a stopping rule based on the Akaike Information Criterion was used to exclude redundant or unnecessary variables.21  Testing and estimation of possible cut-off values for continuous variables were done by maximally selected log-rank statistics.22  To correct for overestimation, a shrinkage procedure and a bootstrap correction of the CIs were applied according to Holländer et al.23  CIs for binomial probabilities were estimated using the method proposed by Agresti and Coull.24  All statistical analyses were performed with R software, version 2.1.1, using R packages Design, version 2.0, and cmprsk, version 2.1 (all available at http://www.r-project.org).25 

Patient characteristics

The clinical features at presentation of all 361 patients are shown in Table 1. Of the 361 patients, 338 (94%) had adequate results on chromosome banding analysis. In the remaining 23 patients, FISH analyses revealed numerical aberrations in 5 cases and balanced rearrangements in 2 cases.

Table 1.

Clinical characteristics of 361 patients older than 60 with AML


Characteristic

Value
No. of female patients (%)   166 (46)  
Median age, y (range)   67 (61-84)  
Disease status, no. of patients (%)  
    De novo AML   231 (64)  
        M0   13 (6)  
        M1   40 (17)  
        M2   66 (29)  
        M3   13 (6)  
        M4   53 (23)  
        M5   22 (10)  
        M6   3 (1)  
        M7   1 (0)  
        Missing   20 (9)  
    Secondary AML   119 (33)  
        Preceding MDS   78 (22)  
        Preceding malignancy   41 (11)  
        Missing   11 (3)  
Median hemoglobin level, g/L  91 (38-172)  
    Missing, no. of patients (%)   17 (5)  
Median platelet count, × 109/L (range)  61.5 (4-848)  
    Missing, no. of patients (%)   17 (5)  
Median WBC count, × 109/L (range)  8.4 (0.2-303)  
    Missing, no. of patients (%)   16 (4)  
Median PB blasts, % (range)  31 (0-100)  
    Missing, no. of patients (%)   66 (18)  
Median BM blasts, % (range)  70 (0-100)  
    Missing, no. of patients (%)   55 (15)  
Median LDH level, U/L (range)  358 (57-6623)  
    Missing, no. of patients (%)   32 (9)  
Lymphadenopathy, no. of patients (%)  46 (13)  
    Missing   42 (12)  
Gum hypertrophy, no. of patients (%)  14 (4)  
    Missing   28 (8)  
Extramedullary involvement, no. of patients (%)  16 (4)  
    Missing
 
37 (10)
 

Characteristic

Value
No. of female patients (%)   166 (46)  
Median age, y (range)   67 (61-84)  
Disease status, no. of patients (%)  
    De novo AML   231 (64)  
        M0   13 (6)  
        M1   40 (17)  
        M2   66 (29)  
        M3   13 (6)  
        M4   53 (23)  
        M5   22 (10)  
        M6   3 (1)  
        M7   1 (0)  
        Missing   20 (9)  
    Secondary AML   119 (33)  
        Preceding MDS   78 (22)  
        Preceding malignancy   41 (11)  
        Missing   11 (3)  
Median hemoglobin level, g/L  91 (38-172)  
    Missing, no. of patients (%)   17 (5)  
Median platelet count, × 109/L (range)  61.5 (4-848)  
    Missing, no. of patients (%)   17 (5)  
Median WBC count, × 109/L (range)  8.4 (0.2-303)  
    Missing, no. of patients (%)   16 (4)  
Median PB blasts, % (range)  31 (0-100)  
    Missing, no. of patients (%)   66 (18)  
Median BM blasts, % (range)  70 (0-100)  
    Missing, no. of patients (%)   55 (15)  
Median LDH level, U/L (range)  358 (57-6623)  
    Missing, no. of patients (%)   32 (9)  
Lymphadenopathy, no. of patients (%)  46 (13)  
    Missing   42 (12)  
Gum hypertrophy, no. of patients (%)  14 (4)  
    Missing   28 (8)  
Extramedullary involvement, no. of patients (%)  16 (4)  
    Missing
 
37 (10)
 

Subheadings under “De novo AML” refer to FAB subtypes. Percentages may not add to 100 because of rounding. MDS indicates myelodysplastic syndrome.

Frequency of cytogenetic abnormalities

The frequencies of specific cytogenetic abnormalities that were detected in at least 5 patients are shown in Table 2. The distribution of specific cytogenetic abnormalities among different age groups is shown in Table 3.

Table 2.

Frequency and characteristics of specific cytogenetic abnormalities among 361 AML patients older than 60



No. of patients (%)
Cytogenetic abnormality
Total
1 abnormality
2 abnormalities
3 or more abnormalities
5 or more abnormalities
Normal karyotype   161 (45)   NA   NA   NA   NA  
-5/del(5q)  57 (16)   2 (4)   4 (7)   51 (89)   39 (68)  
    -5   16 (4)   0 (0)   0 (0)   16 (100)   15 (94)  
    del(5q)*  41 (11)   2 (5)   4 (10)   35 (85)   24 (59)  
    Balanced   3   0   0   3   2  
-7/del(7q)  52 (14)   9 (17)   11 (21)   32 (62)   25 (48)  
    -7   23 (6)   2 (9)   5 (22)   16 (70)   12 (52)  
    del(7q)  29 (8)   7 (24)   4 (14)   18 (62)   13 (45)  
    Balanced   3   0   1   0   2  
+8  47 (13)   18 (38)   4 (9)   25 (53)   18 (38)  
    Balanced   9   0   3   6   2  
-17/del(17p)  25 (7)   0 (0)   1 (4)   24 (96)   24 (96)  
    Balanced   1   0   1   0   0  
abn(12p)  25 (7)   3 (12)   2 (8)   20 (80)   15 (60)  
    Balanced   3   0   1   2   1  
-20/del(20q)  23 (6)   3 (13)   2 (9)   18 (78)   13 (57)  
    Balanced   4   0   0   4   1  
+11   20 (6)   4 (20)   2 (10)   14 (70)   13 (65)  
+13   17 (5)   8 (47)   0 (0)   9 (53)   8 (47)  
+21  17 (5)   2 (12)   2 (12)   13 (76)   8 (47)  
    Balanced   1   0   0   1   0  
+22  17 (5)   0 (0)   1 (6)   16 (94)   12 (71)  
    Balanced   3   0   1   2   1  
del(13q)  15 (4)   1 (7)   1 (7)   13 (87)   12 (80)  
    Balanced   1   0   0   0   1  
abn(3q)   14 (4)   1 (7)   1 (7)   12 (86)   10 (71)  
inv(16)   14 (4)   9 (64)   3 (21)   2 (14)   0 (0)  
t(8;21)   12 (3)   6 (50)   4 (33)   2 (17)   0 (0)  
t(11q23)  11 (3)   3 (27)   3 (27)   5 (45)   3 (27)  
    t(9;11)   5 (1)   2 (40)   2 (40)   1 (20)   0 (0)  
    t(11;various)   6 (2)   1 (17)   1 (17)   4 (67)   3 (50)  
t(15;17)   11 (3)   7 (64)   3 (27)   1 (9)   0 (0)  
+14   9 (2)   0 (0)   1 (11)   8 (89)   6 (67)  
-18   9 (2)   0 (0)   0 (0)   9 (100)   9 (100)  
-Y  8 (2)   1 (13)   2 (25)   5 (63)   2 (25)  
    Balanced   4   0   2   2   0  
+4   5 (1)   1 (20)   0 (0)   4 (80)   4 (80)  
del(11q)  5 (1)   0 (0)   1 (20)   4 (80)   3 (60)  
    Balanced
 
2
 
0
 
0
 
2
 
1
 


No. of patients (%)
Cytogenetic abnormality
Total
1 abnormality
2 abnormalities
3 or more abnormalities
5 or more abnormalities
Normal karyotype   161 (45)   NA   NA   NA   NA  
-5/del(5q)  57 (16)   2 (4)   4 (7)   51 (89)   39 (68)  
    -5   16 (4)   0 (0)   0 (0)   16 (100)   15 (94)  
    del(5q)*  41 (11)   2 (5)   4 (10)   35 (85)   24 (59)  
    Balanced   3   0   0   3   2  
-7/del(7q)  52 (14)   9 (17)   11 (21)   32 (62)   25 (48)  
    -7   23 (6)   2 (9)   5 (22)   16 (70)   12 (52)  
    del(7q)  29 (8)   7 (24)   4 (14)   18 (62)   13 (45)  
    Balanced   3   0   1   0   2  
+8  47 (13)   18 (38)   4 (9)   25 (53)   18 (38)  
    Balanced   9   0   3   6   2  
-17/del(17p)  25 (7)   0 (0)   1 (4)   24 (96)   24 (96)  
    Balanced   1   0   1   0   0  
abn(12p)  25 (7)   3 (12)   2 (8)   20 (80)   15 (60)  
    Balanced   3   0   1   2   1  
-20/del(20q)  23 (6)   3 (13)   2 (9)   18 (78)   13 (57)  
    Balanced   4   0   0   4   1  
+11   20 (6)   4 (20)   2 (10)   14 (70)   13 (65)  
+13   17 (5)   8 (47)   0 (0)   9 (53)   8 (47)  
+21  17 (5)   2 (12)   2 (12)   13 (76)   8 (47)  
    Balanced   1   0   0   1   0  
+22  17 (5)   0 (0)   1 (6)   16 (94)   12 (71)  
    Balanced   3   0   1   2   1  
del(13q)  15 (4)   1 (7)   1 (7)   13 (87)   12 (80)  
    Balanced   1   0   0   0   1  
abn(3q)   14 (4)   1 (7)   1 (7)   12 (86)   10 (71)  
inv(16)   14 (4)   9 (64)   3 (21)   2 (14)   0 (0)  
t(8;21)   12 (3)   6 (50)   4 (33)   2 (17)   0 (0)  
t(11q23)  11 (3)   3 (27)   3 (27)   5 (45)   3 (27)  
    t(9;11)   5 (1)   2 (40)   2 (40)   1 (20)   0 (0)  
    t(11;various)   6 (2)   1 (17)   1 (17)   4 (67)   3 (50)  
t(15;17)   11 (3)   7 (64)   3 (27)   1 (9)   0 (0)  
+14   9 (2)   0 (0)   1 (11)   8 (89)   6 (67)  
-18   9 (2)   0 (0)   0 (0)   9 (100)   9 (100)  
-Y  8 (2)   1 (13)   2 (25)   5 (63)   2 (25)  
    Balanced   4   0   2   2   0  
+4   5 (1)   1 (20)   0 (0)   4 (80)   4 (80)  
del(11q)  5 (1)   0 (0)   1 (20)   4 (80)   3 (60)  
    Balanced
 
2
 
0
 
0
 
2
 
1
 

Patients may be counted more than once due to the coexistence of more than one cytogenetic abnormality in the leukemic clone. Percentages may not add to 100 because of rounding.

Balanced indicates the number of patients exhibiting a given abnormality in combination with t(8;21), t(11q23), t(15;17), or inv(16)/t(16;16); NA, not applicable.

*

Four cases detected only by FISH.

Six cases detected only by FISH.

Table 3.

Frequencies of specific cytogenetic abnormalities in different age groups


Cytogenetic abnormality

60-69 y

70-79 y

80 y or older
Normal karyotype   109 (45)   50 (45)   2 (33)  
Unbalanced*    
    +4   4 (2)   1 (1)   0 (0)  
    -5/del(5q)   35 (14)   21 (19)   1 (17)  
        -5   10 (4)   6 (5)   0 (0)  
        del(5q)   25 (10)   15 (13)   1 (17)  
    -7/del(7q)   36 (15)   16 (14)   0 (0)  
        -7   12 (5)   11 (10)   0 (0)  
        del(7q)   24 (10)   5 (4)   0 (0)  
    +8   32 (13)   14 (13)   1 (17)  
        Noncomplex   17 (7)   4 (4)   1 (17)  
        Complex   15 (6)   10 (9)   0 (0)  
    +11   13 (5)   7 (6)   0 (0)  
        Noncomplex   5 (2)   1 (1)   0 (0)  
        Complex   8 (3)   6 (5)   0 (0)  
    +13   12 (5)   4 (4)   1 (17)  
    del(13q)   9 (4)   6 (5)   0 (0)  
    +14   5 (2)   4 (4)   0 (0)  
    -17/del(17p)   16 (7)   8 (7)   1 (17)  
        -17   8 (3)   5 (4)   1 (17)  
        del(17p)   8 (3)   3 (3)   0 (0)  
    -18   5 (2)   4 (4)   0 (0)  
    -20/del(20q)   17 (7)   6 (5)   0 (0)  
        -20   6 (2)   2 (2)   0 (0)  
        del(20q)   11 (5)   4 (4)   0 (0)  
    +21   12 (5)   5 (4)   0 (0)  
    +22   12 (5)   5 (4)   0 (0)  
Balanced    
    t(8;21)   7 (3)   5 (4)   0 (0)  
    t(11q23)   7 (3)   4 (4)   0 (0)  
    t(15;17)   7 (3)   4 (4)   0 (0)  
    inv(16)/t(16;16)   12 (5)   2 (2)   0 (0)  
Other    
    abn(3q)   9 (4)   5 (4)   0 (0)  
    abn(12p)   10 (4)   14 (13)   1 (17)  
Complex    
    3 or more unrelated   37 (15)   23 (21)   1 (17)  
    5 or more unrelated   28 (12)   15 (13)   1 (17)  
Total
 
243 (100)
 
112 (100)
 
6 (100)
 

Cytogenetic abnormality

60-69 y

70-79 y

80 y or older
Normal karyotype   109 (45)   50 (45)   2 (33)  
Unbalanced*    
    +4   4 (2)   1 (1)   0 (0)  
    -5/del(5q)   35 (14)   21 (19)   1 (17)  
        -5   10 (4)   6 (5)   0 (0)  
        del(5q)   25 (10)   15 (13)   1 (17)  
    -7/del(7q)   36 (15)   16 (14)   0 (0)  
        -7   12 (5)   11 (10)   0 (0)  
        del(7q)   24 (10)   5 (4)   0 (0)  
    +8   32 (13)   14 (13)   1 (17)  
        Noncomplex   17 (7)   4 (4)   1 (17)  
        Complex   15 (6)   10 (9)   0 (0)  
    +11   13 (5)   7 (6)   0 (0)  
        Noncomplex   5 (2)   1 (1)   0 (0)  
        Complex   8 (3)   6 (5)   0 (0)  
    +13   12 (5)   4 (4)   1 (17)  
    del(13q)   9 (4)   6 (5)   0 (0)  
    +14   5 (2)   4 (4)   0 (0)  
    -17/del(17p)   16 (7)   8 (7)   1 (17)  
        -17   8 (3)   5 (4)   1 (17)  
        del(17p)   8 (3)   3 (3)   0 (0)  
    -18   5 (2)   4 (4)   0 (0)  
    -20/del(20q)   17 (7)   6 (5)   0 (0)  
        -20   6 (2)   2 (2)   0 (0)  
        del(20q)   11 (5)   4 (4)   0 (0)  
    +21   12 (5)   5 (4)   0 (0)  
    +22   12 (5)   5 (4)   0 (0)  
Balanced    
    t(8;21)   7 (3)   5 (4)   0 (0)  
    t(11q23)   7 (3)   4 (4)   0 (0)  
    t(15;17)   7 (3)   4 (4)   0 (0)  
    inv(16)/t(16;16)   12 (5)   2 (2)   0 (0)  
Other    
    abn(3q)   9 (4)   5 (4)   0 (0)  
    abn(12p)   10 (4)   14 (13)   1 (17)  
Complex    
    3 or more unrelated   37 (15)   23 (21)   1 (17)  
    5 or more unrelated   28 (12)   15 (13)   1 (17)  
Total
 
243 (100)
 
112 (100)
 
6 (100)
 

Values indicates number of patients, with percentage in parentheses. Patients may be counted more than once due to the coexistence of more than 1 cytogenetic abnormality in the leukemic clone.

*

In the absence of t(8;21), t(11q23), t(15;17), or inv(16)/t(16;16).

Irrespective of karyotype complexity.

Including cases with unbalanced or balanced abnormalities.

Of the 361 patients, 161 (45%) had a normal karyotype, whereas 200 (55%) had at least one clonal aberration. The balanced rearrangements t(8;21), t(11q23), t(15;17), and inv(16) were present in 48 (13%) of 361 patients. In the absence of these balanced rearrangements, 73 (20%) patients exhibited a single aberration, 18 (5%) patients had 2 aberrations, and 61 (17%) patients had at least 3 cytogenetic abnormalities, including 44 (12%) patients with 5 or more aberrations. Abnormalities with a frequency above 5% were –5/del(5q) in 16%, –7/del(7q) in 14%, +8 in 13%, abn(12p) in 7%, –17/del(17p) in 7%, –20/del(20q) in 6%, and +11 in 6%.

The balanced rearrangements t(8;21), t(11q23), t(15;17), and inv(16) occurred predominantly as the sole aberration in 25 of 48 cases (52%) or in combination with a single additional abnormality in 13 of 48 cases (27%).

Among the numerical and structural chromosome abnormalities, 4 occurred frequently within a noncomplex karyotype in the absence of a balanced rearrangement: –7/del(7q) in19 of 49 cases (39%), +8 in 19 of 38 cases (50%), +11 in 6 of 20 cases (30%), and +13 in 8 of 17 cases (47%). All other chromosomal imbalances were seen as part of a complex karyotype, defined as 3 or more aberrations in the absence of a balanced rearrangement, in the majority of cases.

Response to induction therapy according to cytogenetic abnormalities

Of all 361 patients, 155 (43%) achieved CR. The rates of CR after 2 courses of induction therapy, RD, and ED/HD for patients with specific cytogenetic abnormalities are shown in Table 4.

Table 4.

Response to induction therapy according to cytogenetic abnormalities


Cytogenetic abnormality

No. of patients

CR, %

95% CI

ED/HD, %

RD, %
Normal karyotype   161   52   44-60   11   37  
Unbalanced*      
    +4   5   40   11-77   20   40  
    -5/del(5q)   54   7   3-18   26   67  
        -5   16   0   0-19   38   63  
        del(5q)   38   11   5-27   21   68  
        -7/del(7q)   49   6   2-17   18   76  
        -7   22   9   3-28   18   73  
        del(7q)   27   4   0-20   19   78  
    +8   38   32   19-47   26   42  
        Noncomplex   19   53   32-73   16   32  
        Complex   19   11   3-31   37   53  
    +11   20   30   12-49   30   40  
        Noncomplex   6   83   38-99   0   17  
        Complex   14   7   0-31   43   50  
    +13   17   18   6-41   24   59  
    del(13q)   14   0   0-22   21   79  
    +14   9   22   6-55   22   56  
    -17/del(17p)   24   4   0-20   29   67  
        -17   14   0   0-22   21   79  
        del(17p)   9   11   0-43   33   56  
    -18   9   0   0-30   33   67  
    -20/del(20q)   19   0   0-17   21   79  
        -20   7   0   0-35   29   71  
        del(20q)   12   0   0-24   17   83  
    +21   16   19   7-43   13   69  
    +22   14   14   4-40   14   71  
Balanced      
    t(8;21)   12   58   32-81   17   25  
    t(11q23)   11   55   28-79   18   27  
    t(15;17)   11   73   43-90   27   0  
    inv(16)/t(16;16)   14   86   60-96   14   0  
Other      
    abn(3q)   14   0   0-22   14   86  
    abn(12p)   22   18   7-39   23   59  
Complex      
    3 or more unrelated   61   10   5-20   28   62  
    5 or more unrelated   44   7   2-18   25   68  
All patients
 
861
 
43
 
38-48
 
14
 
43
 

Cytogenetic abnormality

No. of patients

CR, %

95% CI

ED/HD, %

RD, %
Normal karyotype   161   52   44-60   11   37  
Unbalanced*      
    +4   5   40   11-77   20   40  
    -5/del(5q)   54   7   3-18   26   67  
        -5   16   0   0-19   38   63  
        del(5q)   38   11   5-27   21   68  
        -7/del(7q)   49   6   2-17   18   76  
        -7   22   9   3-28   18   73  
        del(7q)   27   4   0-20   19   78  
    +8   38   32   19-47   26   42  
        Noncomplex   19   53   32-73   16   32  
        Complex   19   11   3-31   37   53  
    +11   20   30   12-49   30   40  
        Noncomplex   6   83   38-99   0   17  
        Complex   14   7   0-31   43   50  
    +13   17   18   6-41   24   59  
    del(13q)   14   0   0-22   21   79  
    +14   9   22   6-55   22   56  
    -17/del(17p)   24   4   0-20   29   67  
        -17   14   0   0-22   21   79  
        del(17p)   9   11   0-43   33   56  
    -18   9   0   0-30   33   67  
    -20/del(20q)   19   0   0-17   21   79  
        -20   7   0   0-35   29   71  
        del(20q)   12   0   0-24   17   83  
    +21   16   19   7-43   13   69  
    +22   14   14   4-40   14   71  
Balanced      
    t(8;21)   12   58   32-81   17   25  
    t(11q23)   11   55   28-79   18   27  
    t(15;17)   11   73   43-90   27   0  
    inv(16)/t(16;16)   14   86   60-96   14   0  
Other      
    abn(3q)   14   0   0-22   14   86  
    abn(12p)   22   18   7-39   23   59  
Complex      
    3 or more unrelated   61   10   5-20   28   62  
    5 or more unrelated   44   7   2-18   25   68  
All patients
 
861
 
43
 
38-48
 
14
 
43
 

Patients may be counted more than once due to the coexistence of more than 1 cytogenetic abnormality in the leukemic clone. Percentages may not add to 100 because of rounding.

*

In the absence of t(8;21), t(11q23), t(15;17), or inv(16)/t(16;16).

Irrespective of karyotype complexity.

Including cases with unbalanced or balanced abnormalities.

Compared with the normal karyotype group (CR rate, 52%), CR rates were similar or even higher for patients with the following aberrations: t(8;21), t(11q23), t(15;17), inv(16), +8 within a noncomplex karyotype, and +11 within a noncomplex karyotype. All other abnormalities were associated with lower CR rates than normal karyotype, with RD being the predominant cause of therapeutic failure as opposed to ED/HD (Table 4).

Long-term outcome according to cytogenetic abnormalities

OS and CIR for patients with specific cytogenetic abnormalities are summarized in Table 5. The estimated median follow-up duration for survival was 57 months. The median OS for the entire study population was 9.9 months.

Table 5.

Long-term outcome according to cytogenetic abnormalities


Cytogenetic abnormality

CR, no. of patients

2-y CIR (SE)*

P

No. of patients

Median OS, mo

3-y OS (95% CI)

P
Normal   83   0.72 (0.05)   NA   161   12.4   0.19 (0.13-0.26)   NA  
Unbalanced        
    +4   2   —   —   5   12.1   0.20 (0.03-1.0)   .95  
    -5/del(5q)   4   —   —   54   2.7   0   < .001  
    -7/del(7q)   3   —   —   49   5.2   0   < .001  
    +8   12   0.83 (0.12)   .94   38   7.5   0.16 (0.07-0.33)   .22  
        Noncomplex   10   0.80 (0.15)   .94   19   14.2   0.26 (0.12-0.56)   .40  
        Complex   2   —   —   19   2.7   0.05 (0.01-0.35)   < .001  
    +11   5   1.0   .05   20   5.0   0.07 (0.01-0.41)   .02  
        Noncomplex   5   1.0   .1   6   15.1   0.20 (0.03-1.0)   .76  
        Complex   1   —   —   14   2   0   < .001  
    +13   3   —   —   17   3.1   0.06 (0.01-0.39)   < .001  
    del(13q)   0   —   —   14   2.3   0   < .001  
    +14   2   —   —   9   5.9   0.11 (0.02-0.70)   .08  
    -17/del(17p)   1   —   —   24   2.1   0   < .001  
    -18   0   —   —   9   2.2   0   < .001  
    -20/del(20q)   0   —   —   19   5.7   0   .006  
    +21   3   —   —   16   5.6   0   < .001  
    +22   2   —   —   14   5.7   0.10 (0.02-0.59)   .05  
Balanced§        
    t(8;21)   7   1.0   .04   12   12.8   0.10 (0.02-0.65)   .67  
    t(11q23)   6   0.83 (0.2)   .92   11   11.7   0.18 (0.05-0.64)   .95  
    t(15;17)   8   0.29 (0.19)   .03   11   NR   0.55 (0.32-0.94)   .02  
    inv(16)   12   0.75 (0.14)   .83   14   21   0.26 (0.10-0.66)   .13  
Other        
    abn(3q)   0   —   —   14   4.9   0   < .001  
    abn(12p)   4   —   —   22   4.1   0.05 (0.01-0.31)   < .001  
Complex        
    3 or more unrelated   6   1.0   .009   61   3.1   0.04 (0.01-0.15)   < .001  
    5 or more unrelated   3   —   —   44   2.7   0.03 (0.01-0.20)   < .001  
All patients
 
155
 
0.74 (0.03)
 
NA
 
361
 
9.9
 
0.15 (0.11-0.19)
 
NA
 

Cytogenetic abnormality

CR, no. of patients

2-y CIR (SE)*

P

No. of patients

Median OS, mo

3-y OS (95% CI)

P
Normal   83   0.72 (0.05)   NA   161   12.4   0.19 (0.13-0.26)   NA  
Unbalanced        
    +4   2   —   —   5   12.1   0.20 (0.03-1.0)   .95  
    -5/del(5q)   4   —   —   54   2.7   0   < .001  
    -7/del(7q)   3   —   —   49   5.2   0   < .001  
    +8   12   0.83 (0.12)   .94   38   7.5   0.16 (0.07-0.33)   .22  
        Noncomplex   10   0.80 (0.15)   .94   19   14.2   0.26 (0.12-0.56)   .40  
        Complex   2   —   —   19   2.7   0.05 (0.01-0.35)   < .001  
    +11   5   1.0   .05   20   5.0   0.07 (0.01-0.41)   .02  
        Noncomplex   5   1.0   .1   6   15.1   0.20 (0.03-1.0)   .76  
        Complex   1   —   —   14   2   0   < .001  
    +13   3   —   —   17   3.1   0.06 (0.01-0.39)   < .001  
    del(13q)   0   —   —   14   2.3   0   < .001  
    +14   2   —   —   9   5.9   0.11 (0.02-0.70)   .08  
    -17/del(17p)   1   —   —   24   2.1   0   < .001  
    -18   0   —   —   9   2.2   0   < .001  
    -20/del(20q)   0   —   —   19   5.7   0   .006  
    +21   3   —   —   16   5.6   0   < .001  
    +22   2   —   —   14   5.7   0.10 (0.02-0.59)   .05  
Balanced§        
    t(8;21)   7   1.0   .04   12   12.8   0.10 (0.02-0.65)   .67  
    t(11q23)   6   0.83 (0.2)   .92   11   11.7   0.18 (0.05-0.64)   .95  
    t(15;17)   8   0.29 (0.19)   .03   11   NR   0.55 (0.32-0.94)   .02  
    inv(16)   12   0.75 (0.14)   .83   14   21   0.26 (0.10-0.66)   .13  
Other        
    abn(3q)   0   —   —   14   4.9   0   < .001  
    abn(12p)   4   —   —   22   4.1   0.05 (0.01-0.31)   < .001  
Complex        
    3 or more unrelated   6   1.0   .009   61   3.1   0.04 (0.01-0.15)   < .001  
    5 or more unrelated   3   —   —   44   2.7   0.03 (0.01-0.20)   < .001  
All patients
 
155
 
0.74 (0.03)
 
NA
 
361
 
9.9
 
0.15 (0.11-0.19)
 
NA
 

Patients may be counted more than once due to the coexistence of more than 1 cytogenetic abnormality in the leukemic clone.

NA indicates not applicable; —, not determined; NR, not reached.

*

Determined for abnormalities present in 5 or more patients.

For comparison of a given abnormality with normal karyotype.

In the absence of t(8;21), t(11q23), t(15;17), or inv(16).

§

Irrespective of karyotype complexity.

Including cases with unbalanced or balanced abnormalities.

When we analyzed the impact of specific chromosome aberrations on OS, we noted a pattern that was very similar to the one we had observed during assessment of the influence of karyotype on the response to induction therapy: t(8;21), t(11q23), t(15;17), inv(16), +8 within a noncomplex karyotype, and +11 within a noncomplex karyotype were associated with OS comparable to or higher than that of patients with normal karyotype. The highest 3-year OS rates were observed in patients with t(15;17) (55%, P = .02), followed by inv(16) (26%, P = .13), and +8 within a noncomplex karyotype (26%, P = .40). Low 3-year OS rates were seen in patients with noncomplex cytogenetic abnormalities, defined by fewer than 3 or fewer than 5 chromosome aberrations, in the absence of t(8;21), t(11q23), t(15;17), or inv(16) (6% and 6%, respectively).

Risk stratification according to cytogenetic abnormalities

We used a proportional hazards model to identify prognostic subgroups based on the influence of cytogenetic abnormalities on OS and to generate a cytogenetic stratification system for patients older than 60 years with AML. Cytogenetic categories were defined as follows: normal karyotype, t(8;21), t(11q23), t(15;17), inv(16), –7/del(7q), +8, +11, +13, and other. The category “other” included complex karyotype as well as the following individual chromosome aberrations in the absence of a balanced rearrangement that in the majority of cases were part of a complex karyotype: +4, –5/del(5q), abn(12p), del(13q), +14, –17/del(17p), –18, –20/del(20q), +21, and +22 (Table 2).

Using this approach, we identified 3 prognostic subgroups: a low-risk group including t(15;17) and inv(16) (25 of 361 patients, 7%), a standard-risk group including t(8;21), t(11q23), normal karyotype, +8 within a noncomplex karyotype, and +11 within a noncomplex karyotype (208 of 361 patients, 58%), and a high-risk group including all other aberrations (128 of 361 patients, 35%). The CR rates, median OS, and 3-year OS rates that resulted from the application of this stratification system are given in Table 6; the corresponding OS curves are shown in Figure 1.

Table 6.

CR rates, median OS, and 3-year OS rates resulting from cytogenetic risk stratification of 361 patients older than 60 with AML


Cytogenetic risk group

No. of patients

CR, %

Median OS, mo

3-y OS (95% CI)
Low   25   80   26.4   0.38 (0.19-0.57)  
Standard   208   53   12.5   0.18 (0.13-0.24)  
High
 
128
 
19
 
5.1
 
0.05 (0.02-0.10)
 

Cytogenetic risk group

No. of patients

CR, %

Median OS, mo

3-y OS (95% CI)
Low   25   80   26.4   0.38 (0.19-0.57)  
Standard   208   53   12.5   0.18 (0.13-0.24)  
High
 
128
 
19
 
5.1
 
0.05 (0.02-0.10)
 

Multivariate analysis

To develop a clinically meaningful system that can be used to identify patients who are likely to benefit from intensive chemotherapy as well as those who are better suited for investigational treatment or supportive care, assessment of prognostic factors was performed for OS.

The following variables were evaluated: sex, age, disease status (de novo AML versus secondary AML), hemoglobin level, platelet count, white blood cell (WBC) count, percentage of PB blasts, percentage of BM blasts, lactate dehydrogenase (LDH) level, extramedullary involvement, and cytogenetic risk group as defined (see “Risk stratification according to cytogenetic abnormalities”). Age at diagnosis was the only continuous variable for which a clear cut point was evident on maximally selected log-rank statistics (70 years; P < .001; Figure 2). Therefore, age was included as a dichotomized variable in the model.

Figure 1.

OS based on cytogenetic risk stratification. OS of 361 patients older than 60 years with AML according to the system for cytogenetic risk stratification generated in this study.

Figure 1.

OS based on cytogenetic risk stratification. OS of 361 patients older than 60 years with AML according to the system for cytogenetic risk stratification generated in this study.

Close modal

The multivariate model for OS was built using limited backward selection. To correct for overestimation of hazard ratios (HRs), a shrinkage procedure and a bootstrap correction of CI were applied. The final model included age above 70 years (HR, 2.34; 95% CI, 1.77-3.08), cytogenetic high-risk group (HR, 2.24; 95% CI, 1.74-2.88), and logarithm of LDH level (HR, 2.49; 95% CI, 1.66-3.74) as prognostic factors. Stratification of the patients according to the variables “cytogenetic high-risk group” and “dichotomized age” showed that younger patients (60-70 years) without high-risk cytogenetics had the best outcome (3-year OS, 26%), followed by younger patients with high-risk cytogenetics and patients older than 70 years without high-risk cytogenetics (3-year OS, 6%), and older patients with high-risk cytogenetics (3-year OS, 2%; Table 7 and Figure 3)

Table 7.

Outcome of 361 patients older than 60 with AML according to cytogenetic risk group and age


AMLSG score

No. of patients

CR, %

Median OS, mo

3-y OS (95% CI)
Younger than 70 y     
    Not high risk   161   62   17.5   0.26 (0.20-0.33)  
    High risk   82   21   7.2   0.06 (0.02-0.13)  
70 y or older     
    Not high risk   72   39   6.3   0.06 (0.02-0.13)  
    High risk
 
46
 
15
 
3.1
 
0.02 (0.00-0.10)
 

AMLSG score

No. of patients

CR, %

Median OS, mo

3-y OS (95% CI)
Younger than 70 y     
    Not high risk   161   62   17.5   0.26 (0.20-0.33)  
    High risk   82   21   7.2   0.06 (0.02-0.13)  
70 y or older     
    Not high risk   72   39   6.3   0.06 (0.02-0.13)  
    High risk
 
46
 
15
 
3.1
 
0.02 (0.00-0.10)
 

Cytogenetic risk stratification according to the system proposed in this study.

Figure 2.

Maximally selected log-rank statistics for age. Maximally selected log-rank statistics performed for age to test for a potential cut point separating 2 groups with different survival distributions. Age is shown on the x-axis, corresponding standardized log-rank statistics are shown on the y-axis. The estimated cut-off point was 70.42 years with an M statistics of 6.06 and a corresponding corrected P < .001. Vertical dashed line represents the age cut point evident on maximally selected log-rank statistics and the corresponding M statistics. Horizonal line represents the minimal level of significance based on M statistics.

Figure 2.

Maximally selected log-rank statistics for age. Maximally selected log-rank statistics performed for age to test for a potential cut point separating 2 groups with different survival distributions. Age is shown on the x-axis, corresponding standardized log-rank statistics are shown on the y-axis. The estimated cut-off point was 70.42 years with an M statistics of 6.06 and a corresponding corrected P < .001. Vertical dashed line represents the age cut point evident on maximally selected log-rank statistics and the corresponding M statistics. Horizonal line represents the minimal level of significance based on M statistics.

Close modal

To determine the value of cytogenetics for identifying older patients with AML who are likely to benefit from intensive chemotherapy, we assessed the prognostic impact of pretreatment chromosome aberrations in a prospective series of 361 elderly patients with de novo or secondary AML receiving induction and consolidation treatment according to a single protocol specifically designed for patients above the age of 60.12,13  Cytogenetic diagnostics was performed by chromosome banding and FISH using a comprehensive DNA probe set that was previously shown to enhance the sensitivity of conventional cytogenetics.14  The most important finding of our study was that a substantial proportion of patients (55%), characterized by high-risk cytogenetics or age 70 years or older, or both, had very poor outcomes with a median OS not exceeding 7.2 months and 3-year OS rates of 6% or less.

We identified 3 prognostic categories based on the influence of pretreatment cytogenetic parameters on OS. The cytogenetic high-risk group included abn(3q), abn(12p), and the vast majority of unbalanced aberrations. Whereas most chromosomal imbalances were almost invariably part of a complex karyotype, the 47 patients with +8 could be divided into 2 similarly sized and prognostically different subgroups: 22 (47%) patients had +8 as the sole abnormality or in combination with a single additional aberration, and their prognosis was comparable to that of patients with normal cytogenetics; in 25 (53%) patients, +8 was detected within a complex karyotype, and the prognosis of these patients was extremely poor. These findings extend previous data obtained in patient populations that were more heterogeneous with regard to age and treatment showing that +8 as the sole abnormality was associated with an intermediate prognosis similar to that of patients with normal karyotype.26,27  In contrast, Byrd et al found that in patients with de novo AML (median age, 52 years; proportion of patients older than 60, 36%) who were enrolled on 5 different Cancer and Leukemia Group B (CALGB) trials, sole +8 or +8 with one additional abnormality other than t(8;21), inv(16), or t(9;11) were also associated with significantly inferior OS compared with normal karyotype.2  Similar to our observations in patients with +8, the prognosis of patients with +11 as part of a complex karyotype was significantly worse than that of patients with +11 within a noncomplex karyotype or normal cytogenetics, although reliability was limited by small sample size.

Figure 3.

OS based on cytogenetic stratification system and age. OS of 361 patients older than 60 years with AML according to cytogenetic risk group, as assessed using the stratification system generated in this study, and age. Stratification of patients according to the 2 strongest prognostic factors for OS that were evident on multivariate analysis showed that younger patients without high-risk cytogenetics had the best outcome, followed by younger patients with high-risk cytogenetics, older patients without high-risk cytogenetics, and older patients with high-risk cytogenetics.

Figure 3.

OS based on cytogenetic stratification system and age. OS of 361 patients older than 60 years with AML according to cytogenetic risk group, as assessed using the stratification system generated in this study, and age. Stratification of patients according to the 2 strongest prognostic factors for OS that were evident on multivariate analysis showed that younger patients without high-risk cytogenetics had the best outcome, followed by younger patients with high-risk cytogenetics, older patients without high-risk cytogenetics, and older patients with high-risk cytogenetics.

Close modal

In addition to +8 and +11 within a noncomplex karyotype, and consistent with the study by Grimwade et al,7  OS of patients with t(11q23) was also not significantly different from that of patients with normal karyotype. However, the frequency of t(11q23) in our cohort of elderly patients was low (11 of 361 patients, 3%), which is in keeping with previous observations.7,9,28  As a consequence, we were not able to compare the impact of different types of t(11q23), such as t(6;11), t(9;11), t(10;11), and t(11;19).

Among the 2 low-risk aberrations that were identified in our study, t(15;17) and inv(16), the former was associated with particularly favorable outcome (median OS, not reached; 3-year OS, 55%). This observation reflects that (15;17)-positive APL is a distinct disease entity,29  and it confirms the high antileukemic efficacy, low toxicity, and high degree of compliance of protocols using ATRA in combination with chemotherapy for induction and consolidation therapy in elderly patients with APL.7,9,30 

In contrast to t(15;17) and inv(16), long-term outcome of patients with t(8;21) was not better than that of patients with normal karyotype. This result is in line with 2 recent reports. Marcucci et al found that older age was an independent negative prognostic factor for survival in patients with t(8;21), whereas this was not the case when the analysis was restricted to patients younger than 60 years31 ; Schoch et al observed that t(8;21) was not predictive of favorable OS in patients older than 59 years with de novo AML who were treated according to various protocols of the AML Cooperative Group (AMLCG).9  In the study by Grimwade et al,7  on the other hand, patients with t(8;21) had a relatively high OS rate (35% at 5 years) despite a 5-year relapse rate of 84%. Similar results were recently reported by Farag et al who demonstrated that t(8;21) and inv(16) were associated with comparable 5-year OS rates (20% and 19%, respectively) in patients 60 years of age or older entered into the CALGB cytogenetic study 8461.10  Considering that t(8;21)-positive AML in younger patients is highly sensitive to intensive chemotherapy, in particular high cumulative doses of cytarabine,32-35  it is possible that the worse outcome of our patients was related to the less aggressive treatment that was given in the AML HD98-B trial. Although previous studies have shown that the administration of intensive therapy to older patients was associated with excessive toxicity,36  improvements in supportive care have increased the tolerability of regimens such as repetitive courses of high-dose cytarabine. Therefore, studies using similar treatment approaches for younger and older patients with t(8;21)-positive AML might be warranted. Alternatively, novel approaches targeting leukemogenic mechanisms, such as aberrant recruitment of histone deacetylase activity, may prove therapeutically useful.37-40  Clearly, our results regarding the prognostic relevance of t(8;21) need to be interpreted with caution because they are based on a small cohort of patients. This limitation is related to the rarity of this aberration in older patients that has also been noted in previous studies.7,9,10 

In addition to individual chromosome aberrations, we were also interested in the prognostic impact of karyotype complexity. Although it is commonly accepted that the presence of a complex karyotype predicts an unfavorable clinical course, varying definitions are being used. In the MRC study, complex karyotype was defined by the presence of at least 5 chromosome aberrations in the absence of a prognostically favorable balanced rearrangement.7  Similarly, Farag et al reported that the long-term outcome of patients with 5 or more cytogenetic abnormalities was significantly worse than that of patients with 3 or 4 aberrations.10  In contrast, we found that the prognosis of patients with complex karyotype was extremely poor, regardless of definition, and we observed no clinically meaningful difference between patients with 3 or 4 aberrations and patients with 5 or more aberrations. Therefore, our results suggest that discriminating cases with 3 or 4 aberrations from cases with a more complex karyotype does not contribute to the identification of patients who benefit from chemotherapy regimens currently used to treat elderly patients with AML. The reasons that account for the disparity between our findings and previous observations are currently unknown.

When we compared our cytogenetic classification to the MRC stratification system for elderly patients with AML,7  we noted 2 important differences. First, our system did not identify t(8;21) as a favorable risk abnormality. Second, using our system, 35% of the patients were assigned to the unfavorable risk category, as opposed to 14% in the MRC study,7  because we identified a large number of patients who had significantly worse OS than patients with normal karyotype but would have been placed in the intermediate-risk category using MRC criteria. This result is in accordance with the most recent CALGB cytogenetic study,10  in which the majority of patients benefited minimally from chemotherapy.

Our system for outcome prediction using genetic parameters is based solely on the analysis of pretreatment cytogenetic abnormalities, as assessed by conventional chromosome banding and FISH. In younger patients with AML, a number of molecular genetic abnormalities, such as FLT3 internal tandem duplications, CEBPA mutations, or the recently discovered NPM1 mutations, have been demonstrated to be of prognostic relevance, especially in the large subgroup of patients with normal karyotype.41-46  Due to the lack of systematic studies in adequately sized patient populations receiving intensive therapy, it remains unclear whether the assessment of molecular disease markers can also improve prognostication and risk stratification in elderly patients with AML.

In many clinical trials, age 60 is arbitrarily used to define “older” patients although it is unknown whether this cut-off actually discriminates patient subgroups with different outcomes. In our cohort of elderly patients, we identified age 70 as a prognostically significant cut-off, and multivariate analysis revealed age above 70 as the other major negative prognostic factor for OS besides high-risk cytogenetics. Stratification of patients according to these 2 parameters demonstrated that a large subgroup of patients, characterized by age 70 or older or high-risk cytogenetics, or both, had very unfavorable long-term outcomes, with patients above age 70 with high-risk cytogenetics showing a particularly poor prognosis. We believe that these findings are of potential relevance for the design of future clinical trials as well as for individual treatment decisions. It is possible that a higher age cut-off point, for example, 80 years, is even more discriminating.10  However, the number of patients older than 80 years in our series was too small to draw definitive conclusions.

Collectively, our results demonstrate the clinical importance of cytogenetics and age in patients older than 60 years with AML. In addition, our observations also highlight the dilemma that more than half of the elderly patients who are treated according to current protocols have a very unfavorable prognosis despite intensive chemotherapy. Although it remains to be determined whether the overall prognosis of AML in older patients can be improved by risk-adapted treatment strategies, our findings underscore the need for trials of novel investigational therapies in patients with poor-risk disease.47 

We thank the members of the AML Study Group (AMLSG) for providing leukemia specimens: Universitätsklinikum Bonn, Germany, A. Glasmacher; Universitätsklinikum Düsseldorf, Germany, U. Germing; Universitätsklinikum Giessen, Germany, H. Pralle; Universitätsklinikum Göttingen, Germany, D. Haase; Allgemeines Krankenhaus Altona, Hamburg, Germany, H. Salwender; Universitätskliniken des Saarlandes, Homburg, Germany, F. Hartmann; Universitätsklinikum Innsbruck, Austria, A. Petzer; Städtisches Klinikum Karlsruhe, Germany, M. Bentz; Universitätsklinikum Kiel, Germany, M. Kneba; Klinikum rechts der Isar der Technischen Universität München, Germany, K. Götze; Städtisches Krankenhaus München-Schwabing, Germany, C. Nerl; Städtische Kliniken Oldenburg, Germany, F. del Valle; Caritasklinik St Theresia Saarbrücken, Germany, J. Preiβ; Klinikum Stuttgart, Germany, H. G. Mergenthaler; Krankenhaus der Barmherzigen Brüder, Trier, Germany, H. Kirchen; and Hanusch-Krankenhaus, Vienna, Austria, E. Koller.

Prepublished online as Blood First Edition Paper, July 13, 2006; DOI 10.1182/blood-2006-04-014324.

A complete list of the members of the German-Austrian AML Study Group appears in “Appendix.”

Supported by grant 01GI9981 from the Bundesministerium für Bildung und Forschung (Kompetenznetz “Akute und chronische Leukämien”), Germany.

The authors declare no competing financial interests.

S.F. and R.F.S. contributed equally to this work.

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 USC section 1734.

We thank the members of the AMLSG for providing leukemia specimens and clinical data. We are also grateful to Simone Miller and Christa Wieland for technical support and to Bettina Addamo for data management.

1
Mrozek K, Heerema NA, Bloomfield CD. Cytogenetics in acute leukemia.
Blood Rev
.
2004
;
18
:
115
-136.
2
Byrd JC, Mrozek K, Dodge RK, et al. Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse, and overall survival in adult patients with de novo acute myeloid leukemia: results from Cancer and Leukemia Group B (CALGB 8461).
Blood
.
2002
;
100
:
4325
-4336.
3
Grimwade D, Walker H, Oliver F, et al. The importance of diagnostic cytogenetics on outcome in AML: analysis of 1,612 patients entered into the MRC AML 10 trial. The Medical Research Council Adult and Children's Leukaemia Working Parties.
Blood
.
1998
;
92
:
2322
-2333.
4
Schlenk RF, Benner A, Hartmann F, et al. Risk-adapted postremission therapy in acute myeloid leukemia: results of the German multicenter AML HD93 treatment trial.
Leukemia
.
2003
;
17
:
1521
-1528.
5
Slovak ML, Kopecky KJ, Cassileth PA, et al. Karyotypic analysis predicts outcome of preremission and postremission therapy in adult acute myeloid leukemia: a Southwest Oncology Group/Eastern Cooperative Oncology Group Study.
Blood
.
2000
;
96
:
4075
-4083.
6
Dastugue N, Payen C, Lafage-Pochitaloff M, et al. Prognostic significance of karyotype in de novo adult acute myeloid leukemia. The BGMT group.
Leukemia
.
1995
;
9
:
1491
-1498.
7
Grimwade D, Walker H, Harrison G, et al. The predictive value of hierarchical cytogenetic classification in older adults with acute myeloid leukemia (AML): analysis of 1065 patients entered into the United Kingdom Medical Research Council AML11 trial.
Blood
.
2001
;
98
:
1312
-1320.
8
Leith CP, Kopecky KJ, Godwin J, et al. Acute myeloid leukemia in the elderly: assessment of multidrug resistance (MDR1) and cytogenetics distinguishes biologic subgroups with remarkably distinct responses to standard chemotherapy. A Southwest Oncology Group study.
Blood
.
1997
;
89
:
3323
-3329.
9
Schoch C, Kern W, Schnittger S, Buchner T, Hiddemann W, Haferlach T. The influence of age on prognosis of de novo acute myeloid leukemia differs according to cytogenetic subgroups.
Haematologica
.
2004
;
89
:
1082
-1090.
10
Farag SS, Archer KJ, Mrozek K, et al. Pretreatment cytogenetics add to other prognostic factors predicting complete remission and long-term outcome in patients 60 years of age or older with acute myeloid leukemia: results from Cancer and Leukemia Group B 8461.
Blood
.
2006
;
108
:
63
-73.
11
Rowe JM, Neuberg D, Friedenberg W, et al. A phase 3 study of three induction regimens and of priming with GM-CSF in older adults with acute myeloid leukemia: a trial by the Eastern Cooperative Oncology Group.
Blood
.
2004
;
103
:
479
-485.
12
Schlenk RF, Frohling S, Hartmann F, et al. Phase III study of all-trans retinoic acid in previously untreated patients 61 years or older with acute myeloid leukemia.
Leukemia
.
2004
;
18
:
1798
-1803.
13
Schlenk RF, Frohling S, Hartmann F, et al. Intensive consolidation versus oral maintenance therapy in patients 61 years or older with acute myeloid leukemia in first remission: results of second randomization of the AML HD98-B treatment trial [letter].
Leukemia
.
2006
;
20
:
748
-750.
14
Frohling S, Kayser S, Mayer C, et al. Diagnostic value of fluorescence in situ hybridization for the detection of genomic aberrations in older patients with acute myeloid leukemia.
Haematologica
.
2005
;
90
:
194
-199.
15
Cheson BD, Bennett JM, Kopecky KJ, et al. Revised recommendations of the International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia.
J Clin Oncol
.
2003
;
21
:
4642
-4649.
16
Korn EL. Censoring distributions as a measure of follow-up in survival analysis.
Stat Med
.
1986
;
5
:
255
-260.
17
Kaplan E, Meier P. Nonparametric estimation from incomplete observations.
J Am Stat Assoc
.
1958
;
53
:
457
-481.
18
Therneau TM, Grambsch PM.
Modelling Survival Data: Extending the Cox Model
. New York, NY: Springer;
2000
.
19
Gray RJ. A class of k-sample tests for comparing the cumulative incidence of a competing risk.
Ann Stat
.
1988
;
16
:
1141
-1154.
20
Cox DR. Regression models and life tables.
J R Stat Soc
.
1972
;
34
:
187
-220.
21
Harrell FE.
Regression Modeling Strategies: With Applications to Linear Models, Logistic Regression, and Survival Analysis
. New York, NY: Springer;
2001
.
22
Lausen B, Schumacher M. Maximally selected rank statistics.
Biometrics
.
1992
;
48
:
73
-85.
23
Hollander N, Sauerbrei W, Schumacher M. Confidence intervals for the effects of a prognostic factor after selection of an `optimal' cutpoint.
Statist Med
.
2004
;
23
:
1701
-1713.
24
Agresti A, Coull BA. Approximate is better than “exact” for interval estimation of binomial proportions.
Am Stat
.
1998
;
52
:
119
-126.
25
R Development Core Team.
R: A Language and Environment for Statistical Computing
. Vienna, Austria: R Foundation for Statistical Computing;
2005
.
26
Schoch C, Haase D, Fonatsch C, et al. The significance of trisomy 8 in de novo acute myeloid leukaemia: the accompanying chromosome aberrations determine the prognosis. German AML Cooperative Study Group.
Br J Haematol
.
1997
;
99
:
605
-611.
27
Wolman SR, Gundacker H, Appelbaum FR, Slovak ML. Impact of trisomy 8 (+8) on clinical presentation, treatment response, and survival in acute myeloid leukemia: a Southwest Oncology Group study.
Blood
.
2002
;
100
:
29
-35.
28
Schoch C, Schnittger S, Klaus M, Kern W, Hiddemann W, Haferlach T. AML with 11q23/MLL abnormalities as defined by the WHO classification: incidence, partner chromosomes, FAB subtype, age distribution, and prognostic impact in an unselected series of 1897 cytogenetically analyzed AML cases.
Blood
.
2003
;
102
:
2395
-2402.
29
Sanz MA, Tallman MS, Lo-Coco F. Tricks of the trade for the appropriate management of newly diagnosed acute promyelocytic leukemia.
Blood
.
2005
;
105
:
3019
-3025.
30
Sanz MA, Vellenga E, Rayon C, et al. All-trans retinoic acid and anthracycline monochemotherapy for the treatment of elderly patients with acute promyelocytic leukemia.
Blood
.
2004
;
104
:
3490
-3493.
31
Marcucci G, Mrozek K, Ruppert AS, et al. Prognostic factors and outcome of core binding factor acute myeloid leukemia patients with t(8;21) differ from those of patients with inv(16): a cancer and leukemia group B study.
J Clin Oncol
.
2005
;
23
:
5705
-5717.
32
Bloomfield CD, Lawrence D, Byrd JC, et al. Frequency of prolonged remission duration after high-dose cytarabine intensification in acute myeloid leukemia varies by cytogenetic subtype.
Cancer Res
.
1998
;
58
:
4173
-4179.
33
Byrd JC, Dodge RK, Carroll A, et al. Patients with t(8;21)(q22;q22) and acute myeloid leukemia have superior failure-free and overall survival when repetitive cycles of high-dose cytarabine are administered.
J Clin Oncol
.
1999
;
17
:
3767
-3775.
34
Byrd JC, Ruppert AS, Mrozek K, et al. Repetitive cycles of high-dose cytarabine benefit patients with acute myeloid leukemia and inv(16)(p13q22) or t(16;16)(p13;q22): results from CALGB 8461.
J Clin Oncol
.
2004
;
22
:
1087
-1094.
35
Schlenk RF, Benner A, Krauter J, et al. Individual patient data-based meta-analysis of patients aged 16 to 60 years with core binding factor acute myeloid leukemia: a survey of the German Acute Myeloid Leukemia Intergroup.
J Clin Oncol
.
2004
;
22
:
3741
-3750.
36
Mayer RJ, Davis RB, Schiffer CA, et al. Intensive postremission chemotherapy in adults with acute myeloid leukemia. Cancer and Leukemia Group B.
N Engl J Med
.
1994
;
331
:
896
-903.
37
Byrd JC, Marcucci G, Parthun MR, et al. A phase 1 and pharmacodynamic study of depsipeptide (FK228) in chronic lymphocytic leukemia and acute myeloid leukemia.
Blood
.
2005
;
105
:
959
-967.
38
Gozzini A, Rovida E, Dello Sbarba P, Galimberti S, Santini V. Butyrates, as a single drug, induce histone acetylation and granulocytic maturation: possible selectivity on core binding factor-acute myeloid leukemia blasts.
Cancer Res
.
2003
;
63
:
8955
-8961.
39
Klisovic MI, Maghraby EA, Parthun MR, et al. Depsipeptide (FR 901228) promotes histone acetylation, gene transcription, apoptosis and its activity is enhanced by DNA methyltransferase inhibitors in AML1/ETO-positive leukemic cells.
Leukemia
.
2003
;
17
:
350
-358.
40
Wang J, Saunthararajah Y, Redner RL, Liu JM. Inhibitors of histone deacetylase relieve ETO-mediated repression and induce differentiation of AML1-ETO leukemia cells.
Cancer Res
.
1999
;
59
:
2766
-2769.
41
Frohling S, Scholl C, Gilliland DG, Levine RL. Genetics of myeloid malignancies: pathogenetic and clinical implications.
J Clin Oncol
.
2005
;
23
:
6285
-6295.
42
Marcucci G, Mrozek K, Bloomfield CD. Molecular heterogeneity and prognostic biomarkers in adults with acute myeloid leukemia and normal cytogenetics.
Curr Opin Hematol
.
2005
;
12
:
68
-75.
43
Dohner K, Schlenk RF, Habdank M, et al. Mutant nucleophosmin (NPM1) predicts favorable prognosis in younger adults with acute myeloid leukemia and normal cytogenetics: interaction with other gene mutations.
Blood
.
2005
;
106
:
3740
-3746.
44
Schnittger S, Schoch C, Kern W, et al. Nucleophosmin gene mutations are predictors of favorable prognosis in acute myelogenous leukemia with a normal karyotype.
Blood
.
2005
;
106
:
3733
-3739.
45
Suzuki T, Kiyoi H, Ozeki K, et al. Clinical characteristics and prognostic implications of NPM1 mutations in acute myeloid leukemia.
Blood
.
2005
;
106
:
2854
-2861.
46
Verhaak RG, Goudswaard CS, van Putten W, et al. Mutations in nucleophosmin NPM1 in acute myeloid leukemia (AML): association with other gene abnormalities and previously established gene expression signatures and their favorable prognostic significance.
Blood
.
2005
;
106
:
3747
-3754.
47
Estey E. Clinical trials in AML of the elderly: should we change our methodology?
Leukemia
.
2004
;
18
:
1772
-1774.
Sign in via your Institution