Skip to main content
Erschienen in: Annals of Hematology 7/2015

Open Access 01.07.2015 | Original Article

Big data analysis of treatment patterns and outcomes among elderly acute myeloid leukemia patients in the United States

verfasst von: Bruno C. Medeiros, Sacha Satram-Hoang, Deborah Hurst, Khang Q. Hoang, Faiyaz Momin, Carolina Reyes

Erschienen in: Annals of Hematology | Ausgabe 7/2015

Abstract

Over half of patients diagnosed with acute myeloid leukemia (AML) are 65 years or older. We examined patient characteristics, treatment patterns, and survival among elderly patients in routine clinical practice. We utilized a retrospective cohort analysis of first primary AML patients in the linked Surveillance, Epidemiology, and End Results (SEER)-Medicare database. Patients were diagnosed between January 1, 2000 and December 31, 2009, >66 years, and continuously enrolled in Medicare Part A and B in the year prior to diagnosis. Kaplan-Meier curves and Cox proportional hazards regression assessed overall survival by treatment. There were 3327 (40 %) patients who received chemotherapy within 3 months of diagnosis. Treated patients were more likely younger, male, and married, and less likely to have secondary AML and poor performance indicators and comorbidity score compared to untreated patients. In multivariate survival analysis, treated patients exhibited a significant 33 % lower risk of death compared to untreated patients. Significant survival benefits were noted with receipt of intensive and hypomethylating agent (HMA) therapies compared to no therapy. A survival benefit with allogeneic hematopoietic stem cell transplantation was seen in younger Medicare patients. This real-world study showed that about 60 % of elderly AML patients remain untreated following diagnosis. Use of anti-leukemic therapy was associated with a significant survival benefit in this elderly cohort.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1007/​s00277-015-2351-x) contains supplementary material, which is available to authorized users.
Bruno C. Medeiros and Sacha Satram-Hoang contributed equally to this work.

Introduction

A disproportionate number of newly diagnosed acute myeloid leukemia (AML) occurs in elderly patients, and it is also the leading cause of mortality from leukemia in the USA [1, 2]. The median age at diagnosis is 66 years, and incidence increases with age with over half of the patients diagnosed at age 65 or older[3]. The use of chemotherapy has increased over time but fewer than half of elderly patients receive anti-leukemic therapy and their outcomes remain dismal [46]. After successful induction of remission, disease relapse is inevitable in the majority of cases without and despite additional post-remission therapy [7]. Without treatment, patients succumb to their illness within weeks to months of diagnosis [8].
Treatment efficacy and tolerability have been shown to deteriorate markedly with age [4]. Conventional chemotherapy treatments for AML may be highly toxic, usually requiring prolonged inpatient treatment [6]. For this reason, the NCCN guidelines describe separate treatment recommendations for patients older than 60 years based on performance status, cytogenetic or molecular mutation, and comorbid conditions rather than relying on chronologic age alone [7]. For medically fit older adults, treatment with a combination of an anthracycline and standard dose cytarabine is recommended rather than other chemotherapy regimens or supportive care alone. For older adults with poor physical function and/or unfavorable risk disease, supportive care alone or less intensive chemotherapy with DNA hypomethylating agents or low-dose cytarabine is recommended. The use of allogeneic hematopoietic stem cell transplantation (HSCT) is considered a potential cure for AML, but its use is limited in older patients because of significant baseline comorbidities and increased transplant-related morbidity and mortality [9, 10]. Nonetheless, data from the Swedish Acute Leukemia Registry show that most patients up to 80 years actually tolerate and benefit from intensive treatment, despite deteriorating organ function [11, 12].
Although patients 65 years or older represent the majority of patients with cancer in the USA, a minority of them are enrolled in randomized clinical trials (RCTs). In fact, several studies have shown that only 1– % of elderly cancer patients are participating in clinical trials, thus providing a limited evidence base in which to evaluate treatment efficacy and safety in this population [1315]. The most frequently cited factors for clinical trial ineligibility were advanced age or the presence of significant comorbidity [16]. In order to address the need for additional data in this population, given the limited clinical trial participation, the increased incidence of AML due to the aging population, and the limited treatment options, we used population-based data to examine Medicare beneficiaries following their diagnosis of AML. The information obtained provides an important context for identifying opportunities to improve the quality of treatment strategies and evaluate the benefits of new treatments under investigation.

Methods

Data sources

Patients were identified from the linkage of two data sources, the Surveillance, Epidemiology, and End Results (SEER) program database from the National Cancer Institute and the Medicare enrollment and claims files from the Centers for Medicare and Medicaid Services. Details of the linked SEER-Medicare database have been published elsewhere [17]. Briefly, the database combines clinical, demographic, cancer diagnosis, and cause of death information with claims data for adults age 65 and older enrolled in Medicare Parts A and B. SEER is a nationally representative collection of 18 population-based registries of all incident cancers from diverse geographic areas covering approximately 26 % of the US population. All incident cancer patients reported to the SEER registries are cross-matched with a master file of Medicare enrollment [18]. All Medicare beneficiaries receive Part A coverage (inpatient care, skilled nursing, home health care, and hospice care). Approximately 95 % of beneficiaries subscribe to Part B, which covers physician services and outpatient care. The SEER-Medicare linkage included all Medicare eligible persons appearing in the SEER data through 2009 and their Medicare claims for Part A (inpatient) and Part B (outpatient and physician services) through 2010. Institutional review board approval was waived because the SEER-Medicare data lack personal identifiers.

Study population

Patients were eligible for inclusion in the study if diagnosed with first primary AML between January 1, 2000 and December 31, 2009, at least 66 years of age, and continuously enrolled in Medicare Parts A and B in the 12 months prior to diagnosis. Patients were excluded if their date of death was recorded prior to or the same month as diagnosis, if they were enrolled in a health maintenance organization (HMO) at any time during the 12 months prior to diagnosis (because complete claims data were unavailable for these patients), and if they had two or more claims for chemotherapy prior to diagnosis (to ensure that the cases were previously untreated).

Study variables

SEER program registries routinely collect data on patient demographics (age, race/ethnicity, residence, and socioeconomic status [income and education per census tract]); primary tumor site, tumor morphology, and stage at diagnosis; first course of treatment; and follow-up for vital status. AML diagnosis was based on the International Classification of Disease for Oncology (3rd edition, ICD-O-3) histology codes in the SEER data. Median annual household income at the census tract level, and percentage of adults aged 25 or older with at least some college education at the ZIP code level in the SEER data were used as a proxy for socioeconomic status.
Risk status in AML is based on cytogenetics and molecular abnormalities, which were not available in the SEER data. Prior myelodysplastic syndrome (MDS) or myeloproliferative neoplasm (MPN) that transforms into AML has also poor prognostic features and occurs more commonly among elderly patients [19]. In the absence of cytogenetic data, prior MDS or MPN was used as a proxy for high-risk patients and was identified using diagnosis codes in Medicare Parts A and B claims files prior to AML diagnosis. SEER also does not include measures of performance status, such as Eastern Cooperative Oncology Group. Instead, we used Medicare claims to identify several indictors of poor performance status (PPI) [20], including the use of oxygen and related respiratory therapy supplies, wheelchair and supplies, home health agency services, and skilled nursing facility services that occurred 12 months prior to AML cancer diagnosis.
To assess baseline comorbidity burden, we utilized the National Cancer Institute (NCI) comorbidity index [21] to identify the 15 non-cancer comorbidities from the Charlson Comorbidity Index [22]. The index accounts for the number and seriousness of the conditions and a higher score indicates a greater burden of comorbid disease. Diagnosis and procedure codes were identified from Medicare claims 1 year prior to diagnosis and must appear on at least two different claims that are more than 30 days apart to ensure that “rule out” diagnoses are not counted as comorbid conditions.
Chemotherapy administration was identified using International Classification of Disease (9th revision), Clinical Modification (ICD-9-CM) diagnosis codes and procedural codes, and Healthcare Common Procedural Coding System (HCPCS) “J” codes were used to identify the specific drug administered [23]. The absence of these claims indicated lack of treatment. The first chemotherapy claim within 3 months from diagnosis indicated the start of therapy. Patients were classified into treatment groups based on all chemotherapy administered during the first 60 days after treatment initiation. Chemotherapy agent definition was not possible in approximately 70 % of patients who received therapy because chemotherapy was administered during inpatient stays which are paid based on ICD-9 diagnosis or procedure codes only and not chemotherapy codes. Medicare claims files were also searched for ICD-9-CM and HCPCS codes to identify patients undergoing allogeneic HSCT anytime during follow-up.
Overall survival was measured from date of diagnosis to date of death. The date of death was assigned by using the Medicare date or SEER date of death if Medicare date was missing. All other patients were assumed to be alive at the end of the follow-up period (December 31, 2010), although they may have been censored earlier for other reasons such as the development of a second primary cancer or Medicare claims no longer available.

Statistical analysis

All statistical analyses were performed using SAS software, version 9.1.3 (SAS Institute Inc., Cary, NC). Demographic and clinical characteristics were summarized descriptively by treatment status (treated vs. not treated) and treatment type. Chi-square test for categorical variables and ANOVA or t test for continuous variables determined differences between groups. We considered a p value < .05 to be statistically significant.
In the overall survival analyses, we made comparisons between the treated and not treated patients, between treated patients receiving HSCT and those who did not, and between those receiving low-dose therapy with a DNA methyltransferase (DNMT) inhibitor (azacitidine or decitabine (HMA therapy)), those receiving aggressive induction therapy (cytarabine + anthracycline (intensive therapy)), and those not receiving treatment. Kaplan-Meier survival curves and corresponding log rank tests examined unadjusted overall survival by treatment group. Since timing of treatment initiation differed between patients, the relationship between treatment and survival was evaluated using a Cox regression model with treatment as a time-dependent factor. In the time-varying Cox model, all patients belong to the “not treated” group and only switched to the “treated” group at the time of treatment receipt. Other confounders included in the Cox model were selected a priori from baseline demographic and clinical characteristics..
To assess the risk of early death (30-day mortality and 60-day mortality) after diagnosis, we also used a Cox regression model with treatment as a time-dependent factor. The treated group was limited to patients who received treatment within 30 days after diagnosis to minimize the introduction of immortal time bias in the analysis (period of follow-up time during which death cannot occur) [24].
As a sensitivity exercise for the comparison between HMA therapy, intensive therapy, and no treatment, we also conducted a propensity score-matched survival analysis. Multinomial logistic regression was used to calculate a propensity score—the conditional probability that each patient would be assigned to a specific treatment group given the patient’s pretreatment variables [25, 26]. Pairwise matching was conducted where each patient receiving HMA therapy was matched to one untreated patient and each patient receiving intensive therapy was matched to one untreated patient. Matching variables were age, sex, race, marital status, education, geographic region, year diagnosed, prior MDS, poor performance indicators, and comorbidity score. Matched survival analysis was completed using the Cox proportional hazards regression model, stratifying on the matched pair. Factors that were still found to be significantly different after matching (age, geographic region, and year diagnosed) were included as covariates in the Cox proportional hazards models.
In the survival models, follow-up was calculated beginning on the date of diagnosis up until the first occurrence of a censoring event: date of death, development of a second primary tumor, the last date for which Medicare claims are available, or the end of the follow-up period (December 31, 2010).

Results

Of the 8336 patients who met all study criteria, 3327 (40 %) received treatment with chemotherapy within 3 months of diagnosis and 5009 (60 %) did not (Table 1). Treatment rates increased over the study time period from 35 % in 2000 to 50 % in 2009 (Fig. 1). Treated patients were younger at diagnosis with mean age of 75 compared to those not receiving treatment (81 years; p < .0001). Fifty-two percent of untreated patients were over the age of 80 compared to 20 % in the treated group (p < .0001). Treated patients were also more likely to be male (55 vs. 50 %), married (61 vs. 47 %), and have lower incidence of secondary AML (15 vs. 19 % prior MDS or MPN), were less likely to have PPI (7 vs. 17 %), and had lower comorbidity burden (p < .0001) than untreated patients.
Table 1
Baseline patient characteristics by treatment status
Characteristic
Total (N = 8336)
Treated (N = 3327)
Not treated (N = 5009)
p value
n
%
n
%
n
%
Age at diagnosis
 66–70
1322
15.9
881
26.5
441
8.8
<0.0001
 71–75
1774
21.3
976
29.3
798
15.9
 76–80
1971
23.6
803
24.1
1168
23.3
  > 80
3269
39.2
667
20.0
2602
51.9
Sex
 Male
4331
52.0
1832
55.1
2499
49.9
<0.0001
 Female
4005
48.0
1495
44.9
2510
50.1
Race/ethnicity
 White
7285
87.4
2918
87.7
4367
87.2
0.4807
 Non-white
1051
12.6
409
12.3
642
12.8
Prior MDSa
 No
6896
82.7
2839
85.3
4057
81.0
<0.0001
 Yes
1440
17.3
488
14.7
952
19.0
PPIb
 No
7280
87.3
3111
93.5
4169
83.2
<0.0001
 Yes
1056
12.7
216
6.5
840
16.8
NCI co-morbidity score
 0
4266
51.2
1899
57.1
2367
47.3
<0.0001
 1
2104
25.2
842
25.3
1262
25.2
 2
1018
12.2
325
9.8
693
13.8
  ≥ 3
948
11.4
261
7.8
687
13.7
Marital status
 Married
4373
52.5
2028
61.0
2345
46.8
<0.0001
 Widowed
2492
29.9
726
21.8
1766
35.3
 Separated/divorced
543
6.5
218
6.6
325
6.5
 Single
535
6.4
216
6.5
319
6.4
 Unknown
393
4.7
139
4.2
254
5.1
% of adults with some college education
 0–50
3514
42.2
1370
41.2
2144
42.8
0.3260
 51–100
4439
53.3
1799
54.1
2640
52.7
 Unknown
383
4.6
158
4.7
225
4.5
Median income quartiles
 1–Low
2080
25.0
766
23.0
1314
26.2
0.0003
 2
2080
25.0
819
24.6
1261
25.2
 3
2081
25.0
834
25.1
1247
24.9
 4–High
2079
24.9
902
27.1
1177
23.5
Geographic region
 Midwest
856
10.3
377
11.3
479
9.6
0.0268
 Northeast
517
6.2
216
6.5
301
6.0
 South
3136
37.6
1253
37.7
1883
37.6
 West
3827
45.9
1481
44.5
2346
46.8
NCI National Cancer Institute, MDS prior myelodysplastic syndrome, PPI poor performance indicators
aPatients with a prior myelodysplastic syndrome (MDS) or myeloproliferative disease was identified from Medicare claims and was used as a proxy for high-risk patients in the absence of disease stage
bPoor performance indicators (PPI) were identified from Medicare claims and include the use of oxygen and related respiratory therapy supplies, wheelchair and supplies, home health agency services, and skilled nursing facility services that occurred 12 months prior to AML diagnosis
The median unadjusted overall survival was 2.5 months for the overall population and was longer for treated patients (5.0 months) compared to that for untreated patients (1.5 months; log rank p < .0001; Fig. 2a). As the observed overall survival was not significantly different between patients initiating therapy between 0–30 days, 30–60 days, and 60–90 days from diagnosis, all treated patients were analyzed as a single cohort. In multivariate survival analysis (Table 2), treated patients exhibited a 33 % lower risk of death compared to untreated patients (hazard ratio (HR) = 0.67; 95 % confidence interval (CI) = 0.64–0.71). Increasing age, increasing comorbidity score, and PPI were significantly associated with higher mortality risks. Prognosis also improved over time with a 7–12 % reduction in mortality during the years 2002–2004 (HR = 0.93; 95 % CI = 0.87–0.99), 2005–2007 (HR = 0.88; 95 % CI = 0.82–0.93), and 2008–2009 (HR = 0.90; 95 % CI = 0.84–0.97) compared to that during 2000–2001 (data not shown).
Table 2
Adjusted overall survival and risk of early death by treatment status
Covariates
All AMLa
N = 8320
30-day mortalitya,b
N = 7287
60-day mortalitya,b
N = 7287
N
HR
95 % CI
HR
95 % CI
HR
95 % CI
Treatment status
 Not-treated (ref)
4999
      
 Treated
3321
0.67
0.64–0.71
0.34
0.29–0.39
0.56
0.51–0.61
Age at diagnosis
 66–70 (ref)
1320
      
 71–75
1771
1.29
1.19–1.39
1.19
0.98–1.45
1.37
1.19–1.56
 76–80
1968
1.48
1.37–1.60
1.17
0.97–1.41
1.44
1.26–1.64
  > 80
3261
1.79
1.66–1.93
1.38
1.16–1.65
1.62
1.43–1.84
Sex
 Male (ref)
4323
      
 Female
3997
0.96
0.91–1.01
1.02
0.92–1.13
1.02
0.95–1.10
Marital status
 Married (ref)
4369
      
 Widowed
2486
1.15
1.08–1.21
1.16
1.03–1.31
1.15
1.06–1.25
 Separated/divorced
543
1.15
1.05–1.26
1.28
1.06–1.55
1.16
1.01–1.34
 Single
532
1.16
1.06–1.27
1.20
0.99–1.47
1.25
1.09–1.43
 Unknown
390
1.01
0.91–1.12
1.13
0.90–1.40
1.04
0.88–1.22
Prior MDSc
 No (ref)
6882
      
 Yes
1438
1.03
0.97–1.09
0.99
0.88–1.12
0.96
0.88–1.05
PPId
 No (ref)
7270
      
 Yes
1050
1.28
1.20–1.38
1.32
1.16–1.50
1.25
1.13–1.37
NCI co-morbidity score
 0 (ref)
4257
      
 1
2103
1.15
1.09–1.22
1.09
0.96–1.22
1.13
1.04–1.23
 2
1014
1.26
1.17–1.35
1.18
1.02–1.36
1.32
1.19–1.46
  ≥ 3
946
1.39
1.29–1.50
1.32
1.13–1.53
1.34
1.20–1.50
NCI National Cancer Institute, MDS prior myelodysplastic syndrome, PPI poor performance indicators, HR hazard ratio, CI confidence interval
aModel also includes race, geographic region, income, and year of diagnosis
bTreated group restricted to patients who received treatment within 30 days after diagnosis
cPatients with a prior myelodysplastic syndrome (MDS) or myeloproliferative disease was identified from Medicare claims and was used as a proxy for high-risk patients in the absence of disease stage
dPoor performance indicators(PPI) were identified from Medicare claims and include the use of oxygen and related respiratory therapy supplies, wheelchair and supplies, home health agency services, and skilled nursing facility services that occurred 12 months prior to AML diagnosis
We conducted exploratory subgroup analyses on the impact of treatment versus no treatment to examine whether specific prognostic subgroups benefited more or less from treatment (Supplementary Table S1). Receipt of treatment had a larger impact on mortality risk in the subgroup without a prior MDS (35 % reduction in mortality) compared to the subgroup with a prior MDS (20 % reduction in mortality). Treated patients in the younger age cohort, female gender, with presence of PPI, and NCI comorbidity score 1+ exhibited greater reductions in mortality compared to their subgroup counterparts. Marital status subgroups showed similar benefits with receipt of treatment.
The multivariate analysis of factors predicting early death is shown in Table 2. The median time to treatment initiation was 17 days. There were 1747 (24 %) of patients who died within 30 days of diagnosis and 3429 (47 %) that died within 60 days of diagnosis. Stratifying by treatment status, 210 (9 %) treated patients and 1537 (31 %) untreated patients died within 30 days of diagnosis. Treated patients had a 66 % lower likelihood of early death within 30 days of diagnosis and a 44 % lower likelihood of early death within 60 days of diagnosis compared to the untreated cohort. Other factors associated with increased risk of early death include older age, unmarried, higher comorbidity burden, and presence of poor performance indicators.

Outcomes according to treatment modality

Patients receiving intensive therapy were younger (mean age 73 vs. 78 and 81), were more likely male (62 vs. 59 and 50 %), were married (71 vs. 61 and 47 %), had less secondary AML (7 vs. 21 and 19 % with prior MDS), were less likely to have PPIs (2 vs. 9 and 17 %), and had lower comorbidity score compared to those receiving HMA therapy and not treated, respectively (Table 3). Similarities in age, comorbidity burden, and proportion with high-risk disease were noted in HMA therapy and not treated patients. The median unadjusted overall survival (Fig. 2b) was longer for patients treated with intensive therapy (18.9 months) compared to that with HMA therapy (6.6 months) and not treated (1.5 months; log rank p < .0001). After adjusting for all covariates in the survival model, a 67 % reduction in mortality was observed among patients treated with intensive therapy and a 50 % reduction in mortality was observed among patients treated with HMA therapy, compared to not treated (Table 4). The propensity score-matched survival analysis demonstrated similar risk reductions for both intensive (62 % reduction in mortality) and HMA (59 % reduction in mortality) regimens compared to patients who were not treated. Increasing age, increasing comorbidity score, and presence of PPIs were associated with significant increases in mortality. In a subset analysis stratified by age, similar mortality risk reductions with receipt of intensive and HMA therapies were maintained in the younger (≤75) and older (>75) cohorts (data not shown).
Table 3
Baseline patient characteristics by type of treatment received
Characteristic
Not Treated
N = 5009
HMA Therapy
N = 345
Intensive Therapy
N = 124
p
HSCT
N = 276
No HSCT
N = 3051
p
 
%
%
%
%
%
%
 
Age at diagnosis
 66–70
8.8
13.6
39.5
<0.0001
44.6
24.8
<0.0001
 71–75
15.9
24.1
31.5
 
25.4
29.7
 
 76–80
23.3
25.5
16.1
 
14.5
25.0
 
  > 80
51.9
36.8
12.9
 
15.6
20.5
 
Sex
 Male
49.9
59.1
62.1
0.0002
61.6
54.5
0.0228
 Female
50.1
40.9
37.9
 
38.4
45.5
 
Race/ethnicity
 White
87.2
90.4
87.9
0.2092
88.4
87.6
0.7118
 Non-white
6.7
9.6
12.1
 
11.6
12.4
 
Prior MDSa
 No
81.0
79.1
100c
0.0026
88.8
85.0
0.0920
 Yes
19.0
20.9
 
11.2
15.0
 
PPIb
 No
83.2
91.3
100c
<0.0001
94.2
93.4
0.6245
 Yes
16.8
8.7
 
5.8
6.6
 
NCI co-morbidity score
 0
47.3
50.7
55.6
0.1113
55.8
57.2
0.2711
 1
25.2
25.8
25.8
 
22.8
25.5
 
 2
13.8
11.6
18.5c
 
10.9
9.7
 
  ≥ 3
13.7
11.9
 
10.5
7.6
 
Marital status
 Married
46.8
61.2
71.0
<0.0001
59.4
61.1
0.0851
 Widowed
35.3
21.4
15.3
 
18.5
22.1
 
 Separated/divorced
6.5
5.5
13.6c
 
10.1
6.2
 
 Single
6.4
6.7
 
7.6
6.4
 
 Unknown
5.1
5.2
 
4.3
4.2
 
% of adults with some college education
 0–50
42.8
35.7
45.2
0.0744
35.9
41.7
0.1610
 51–100
52.7
58.0
54.8c
 
59.4
53.6
 
 Unknown
4.5
6.4
 
4.7
4.8
 
Median income quartiles
 1–Low
26.2
21.2
24.2
0.0359
17.0
23.6
0.0419
 2
25.2
21.2
30.6
 
25.7
24.5
 
 3
24.9
28.7
21.8
 
24.6
25.1
 
 4–High
23.5
29.0
23.4
 
32.6
26.6
 
Geographic region
 Midwest
9.6
9.3
11.3
0.0305
12.7
11.2
0.1858
 Northeast
6.0
7.2
10.5
 
4.0
6.7
 
 South
37.6
29.9
33.9
 
35.1
37.9
 
 West
46.8
53.6
44.4
 
48.2
44.2
 
HMA hypomethylating agents, HSCT allogeneic hematopoietic stem cell transplantation, NCI National Cancer Institute, MDS prior myelodysplastic syndrome, PPI poor performance indicators
aPatients with a prior myelodysplastic syndrome (MDS) or myeloproliferative disease was identified from Medicare claims and was used as a proxy for high-risk patients in the absence of disease stage
bPoor performance indicators(PPI) were identified from Medicare claims and include the use of oxygen and related respiratory therapy supplies, wheelchair and supplies, home health agency services, and skilled nursing facility services that occurred 12 months prior to AML diagnosis
cCells with counts of less than 11 are combined in compliance with the National Cancer Institute data use agreement for small cell sizes
Table 4
Adjusted overall survival by treatment type
Covariates
N
Multivariate cox regressiona
Propensity score matched cox regressionb
Treatment
HR
95 % CI
HR
95 % CI
Not treated (ref)
5009
    
HMA therapy
345
0.50
0.45–0.57
0.41
0.32–0.53
Intensive therapy
124
0.33
0.27–0.41
0.38
0.21–0.70
Age at diagnosis
 66–70 (ref)
537
    
 71–75
920
1.30
1.16–1.46
  
 76–80
1276
1.42
1.28–1.58
  
  > 80
2745
1.67
1.50–1.84
  
Sex
 Male (ref)
2780
    
 Female
2698
1.01
0.95–1.07
  
Marital status
 Married (ref)
2644
    
 Widowed
1859
1.13
1.05–1.20
  
 Separated/divorced
349
1.10
0.98–1.24
  
 Single
349
1.19
1.06–1.33
  
 Unknown
277
1.00
0.88–1.14
  
Prior MDSc
 No (ref)
4445
    
 Yes
1033
0.98
0.91–1.05
  
PPId
 No (ref)
4605
    
 Yes
873
1.28
1.18–1.39
  
NCI co-morbidity score
 0 (ref)
2611
    
 1
1383
1.17
1.10–1.25
  
 2
749
1.28
1.18–1.40
  
  ≥ 3
735
1.37
1.26–1.50
  
HMA hypomethylating agents, NCI National Cancer Institute, MDS prior myelodysplastic syndrome, PPI poor performance indicators, HR hazard ratio, CI confidence interval
aModel also includes race, geographic region, income, and year of diagnosis
bPropensity score matched for age, sex, race, marital status, education, geographic region, year of diagnosis, prior MDS, poor performance indicators, and comorbidity score
cPatients with a prior myelodysplastic syndrome (MDS) or myeloproliferative disease was identified from Medicare claims and was used as a proxy for high-risk patients in the absence of disease stage
dPoor performance indicators(PPI) were identified from Medicare claims and include the use of oxygen and related respiratory therapy supplies, wheelchair and supplies, home health agency services, and skilled nursing facility services that occurred 12 months prior to AML diagnosis

Effect of allogeneic stem cell transplantation on survival

Among treated patients, there were 276 (8 %) who underwent HSCT therapy and 3051 (92 %) who did not (Table 3). HSCT patients were younger at diagnosis with mean age of 73 compared to the non-HSCT group (75 years; p < .0001). Seventy percent of HSCT patients compared to 55 % of non-HSCT patients were under the age of 75 at diagnosis. HSCT patients were also more likely to be male (62 vs. 55; p = 0.0228). There were no statistical differences in comorbidity burden, PPI, or prior MDS between both groups. Figure 2c shows that the unadjusted median overall survival was higher for HSCT (9.7 months) compared to the non-HSCT group (4.7 months; log rank p ≤ 0.0001). In multivariate survival analysis (Table 5), treated patients who underwent HSCT had a significant 21 % lower risk of death compared to those who did not receive HSCT. Increasing age, male gender, unmarried, prior MDS, PPI, and increasing comorbidity score were significantly associated with higher risks of post-treatment mortality. In an exploratory subset analysis stratified by age, the survival benefit with HSCT was only demonstrated in the younger age cohort ≤75 years old, and no difference in mortality risks was noted in the older age cohort >75 years (Table 5).
Table 5
Adjusted overall survival among treated patients with and without HSCT
Covariates
Treateda
N = 3321
≤75 yearsa
N = 1854
>75 yearsa
N = 1467
Treatment
N
HR
95 % CI
HR
95 % CI
HR
95 % CI
No HSCT (ref)
3045
      
HSCT
276
0.79
0.69–0.91
0.63
0.53–0.75
1.20
0.95–1.52
Age at diagnosis
 66–70 (ref)
880
      
 71–75
974
1.23
1.11–1.36
    
 76–80
802
1.50
1.35–1.66
    
  > 80
665
2.01
1.79–2.25
    
Sex
 Male (ref)
1829
      
 Female
1492
0.88
0.82–0.95
0.83
0.75–0.93
0.96
0.85–1.08
Marital status
 Married (ref)
2025
      
 Widowed
724
1.16
1.05–1.28
1.23
1.06–1.43
1.16
1.02–1.31
 Separated/divorced
218
1.26
1.08–1.46
1.20
0.99–1.44
1.27
0.98–1.65
 Single
216
1.13
0.97–1.31
1.14
0.94–1.39
1.00
0.78–1.28
 Unknown
138
1.03
0.86–1.24
1.04
0.81–1.33
1.10
0.84–1.44
Prior MDSb
 No (ref)
2834
      
 Yes
487
1.19
1.08–1.32
1.22
1.06–1.41
1.20
1.03–1.38
PPIc
 No (ref)
3107
      
 Yes
214
1.28
1.10–1.49
1.30
1.03–1.65
1.36
1.11–1.67
NCI co-morbidity score
 0 (ref)
1896
      
 1
841
1.14
1.05–1.25
1.17
1.04–1.32
1.13
1.00–1.29
 2
323
1.22
1.07–1.38
1.18
0.98–1.42
1.22
1.01–1.46
  ≥ 3
261
1.42
1.24–1.64
1.42
1.17–1.73
1.40
1.14–1.73
NCI National Cancer Institute, HSCT allogeneic hematopoietic stem cell transplantation, MDS prior myelodysplastic syndrome, PPI poor performance indicators, HR hazard ratio, CI confidence interval
aModel also includes race, geographic region, income, and year of diagnosis
bPatients with a prior myelodysplastic syndrome (MDS) or myeloproliferative disease was identified from Medicare claims and was used as a proxy for high-risk patients in the absence of disease stage
cPoor performance indicators(PPI) were identified from Medicare claims and include the use of oxygen and related respiratory therapy supplies, wheelchair and supplies, home health agency services, and skilled nursing facility services that occurred 12 months prior to AML diagnosis

Discussion

Although therapy use has increased over time, this large observational study of Medicare beneficiaries showed that currently, about 50 % of elderly AML patients remain untreated following diagnosis, which represents an unmet need. We observed a significant survival benefit with receiving anti-leukemic therapy, even among the HMA therapy group who had similar characteristics to the untreated patients. Further, improved survival after receiving intensive therapy compared to HMA therapy was noted after adjustment for confounding variables. However, when patients were matched on sex, race, marital status, education, prior MDS, PPI, and comorbidity score, we found mortality risk reductions of a similar magnitude with receiving both regimens. Overall, these real-world results provide further support that age alone should not deter the use of guideline-recommended therapies particularly because of the high disparities in outcomes between treatment receipt and palliative care.
Results from our observational study have been supported in prior RCTs involving elderly patients. Over 20 years ago, the European Organization for Research and Treatment of Cancer (EORTC) Leukemia Group demonstrated an improvement in complete remission rate and overall survival for AML patients aged 65 years or older immediately treated with induction chemotherapy compared to supportive measures only [27]. Significant clinical improvements in outcomes have also been demonstrated in elderly patients following HMA therapy. When compared to best supportive care (BSC) or low-dose cytarabine (LDAC), treatment with decitabine was associated with a significantly higher CR rate plus CRp rate, a trend toward improvement in median overall survival and a 20 % reduction in the risk of death [28]. In comparison, two separate studies compared the effects of azacitidine against conventional care regimens (CCRs) including BSC, LDAC, and conventional induction chemotherapy. In oligoblastic AML (<30 % blasts), azacitidine treatment was associated with significant improvements in median OS and 2-year survival, albeit no improvement in complete remission rate was observed [29]. In patients with >30 % blasts, preliminary reports showed a trend toward improvement in median OS, a 15 % reduction in the risk of death, improved 1-year survival, and no differences in the CR rate plus CRp rate [30]. No significant safety concerns were raised in these studies following HMA therapy.
Our results confirm data from other registry-based analyses that showed that elderly AML patients who received treatment exhibited a lower early death rate compared to untreated patients or palliation [11, 12, 31]. Although our multivariate analysis demonstrated a greater reduction in mortality in patients receiving aggressive induction chemotherapy compared to HMA therapy, both therapeutic options appeared to be equally better than supportive measures when the cohorts were properly matched for relevant cofounders.
Only 8 % of patients receiving chemotherapy underwent subsequent HSCT therapy. Chronologic age appears to be the driving factor in receiving HSCT. HSCT therapy was associated with a 20 % lower risk of death compared to patients receiving chemotherapy only, and the survival benefit was more pronounced among the younger cohort (≤75 years) with a 37 % reduction in mortality risk. Although myeloablative allogeneic HSCT is rarely recommended in older patients with significant comorbidities, reduced-intensity conditioning (RIC) allogeneic HSCT is encouraging when used as post-remission therapy [9, 10, 32]. The NCCN guidelines consider RIC allogeneic HSCT an additional option for patients 60 years or older as post-remission therapy in those who achieved complete response from induction therapy [7]. Although our observations are at best hypothesis generating, they raise the question of whether allogeneic HSCT provides therapeutic benefit to AML patients older than 75 years of age. Prospective and well-controlled clinical trials are needed to define the role of allogeneic HSCT as post-remission therapy in this cohort of patients.
In the current study, receipt of treatment varied by gender, marital status, income, and geographic region, similar to patterns observed in prior oncology research [3335].Our results also demonstrate that married AML patients were more likely to receive therapy and had higher survival compared to unmarried patients, even after adjusting for known confounders [35]. These results highlight the importance of marital status, likely as a surrogate of social-economic support in patients with AML, and confirm results from previous reports focusing on solid tumor malignancies. Further research is warranted to better quantify how nonclinical factors such as social support contribute to receipt of cancer therapy and outcomes.
The finding that patients receiving intensive therapy were younger, were more likely male, were married, had less secondary AML, were less likely to have PPIs, and had lower comorbidity score compared to those receiving HMA therapy and no treatment may reflect a belief among physicians that elderly patients are frailer and less able to tolerate aggressive or more toxic treatments [4, 3638]. These observations are in agreement with previously reported patterns of treatment selection. For example, in two recent randomized trials where pre-selection of CCR was performed prior to randomization, subjects assigned to aggressive treatment modalities were a median of 5–8 years younger than their counterparts assigned to less intensive regimens [29, 30]. Elderly patients also have diverse attitudes toward cancer treatment; some desire aggressive treatment modalities while others decline therapies offered by their oncologist [39, 40]. These age disparities in treatment patterns are associated with higher mortality [4, 5], and our results provide further support that demographic factors such as age should not discourage the use of guideline-recommended therapies.

Strengths and limitations

Use of the SEER-Medicare data for this type of analysis has several strengths, including the large sample size from a population-based registry and the diverse geographic representation of AML patients in the USA. The database includes longitudinal data with claims for covered services from the time a person is eligible for Medicare until the date of death regardless of residence or service area.
The results of the comparative effectiveness analysis should be interpreted with caution due to the large amount of missing data and resulting small sample size of treatment groups. Induction chemotherapy with curative intent in the outpatient setting is applied to very select elderly AML patients, and our findings may not be representative of the general patient population receiving intensive induction therapy. Conventional chemotherapy treatments for AML are highly toxic [8] and generally requires inpatient treatment. Inpatient stays are paid based on ICD-9 diagnosis or procedure codes only, and therefore, we were unable to define the type of chemotherapy received for 70 % of the treated cohort without the specific chemotherapy J code. Further, dose selection was at the discretion of the physician and dosing information could not be determined retrospectively from available data within the claims dataset.
The SEER registry does not collect baseline molecular and cytogenetic information for leukemia, and our surrogate for stage (including claims for prior MDS as a marker of disease severity) may not adequately assess stage in all patients in our study. The SEER-Medicare data did not contain remission status prior to HSCT, and type of prior anti-AML therapy was not known for the majority of patients receiving transplant. In addition, the SEER data does not include measures of performance status, and using Medicare claims to identify several indictors of poor performance may also be subject to bias. Performance status influences clinicians’ decisions to treat or the specific regimen to administer. Information regarding treatment patterns and characteristics of patients enrolled in health maintenance organizations (HMOs) or fee-for-service plans was not available since Medicare does not collect these data. Treatment patterns, prognosis, and complications may differ between these alternative health care plans and Medicare enrollees, and this would be a productive area for additional evaluation.

Conclusion

Overall, these real-world results provide further support that age alone should not deter the use of guideline-recommended therapies in AML. Our results highlight the benefit of treatment in contrast to palliative therapy in this underserved patient population of elderly AML patients and suggest that anti-leukemic regimens should be strongly considered in the majority of older patients. But, even with treatment, outcomes remain dismal, and given this important unmet medical need, many new agents are currently in development for older patients with AML [4144]. Our findings provide an important context for therapeutic selection that occurs in older patients with AML in the USA. Moving forward, it will be important to identify patients less likely to be treated at diagnosis and design clinical trials to address the therapeutic challenges that exist in this cohort of patients.

Acknowledgments

The authors would like to thank Dr. Michelle Byrtek for statistical consult and thoughtful review of the manuscript. This study used the linked SEER-Medicare database. We acknowledge the efforts of the Applied Research Program, NCI (Bethesda, MD), the Office of Information Services and the Office of Strategic Planning, Health Care Financing Administration (Baltimore, MD), Information Management Services, Inc. (Silver Spring, MD), and the Surveillance, Epidemiology, and End Results (SEER) Program tumor registries in the creation of the SEER-Medicare database. The interpretation and reporting of these data are the sole responsibility of the authors.

Conflict of interest

This study was funded by Genentech, Inc. through a contract with Q.D. Research, Inc. Dr. Medeiros received research funding from Roche-Genentech for clinical research activities and honorarium for advisory board participation. Dr. Reyes and Dr. Hurst are employees of Genentech and shareholders of Roche. Dr. Satram-Hoang, Dr. Hoang, and Mr. Momin work for Q.D. Research in a research and consulting capacity.
Open Access This article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Yamamoto JF, Goodman MT (2008) Patterns of leukemia incidence in the United States by subtype and demographic characteristics, 1997-2002. Cancer Causes Control 19:379–390CrossRefPubMed Yamamoto JF, Goodman MT (2008) Patterns of leukemia incidence in the United States by subtype and demographic characteristics, 1997-2002. Cancer Causes Control 19:379–390CrossRefPubMed
2.
5.
Zurück zum Zitat Kantarjian H, O’Brien S, Cortes J et al (2006) Results of intensive chemotherapy in 998 patients age 65 years or older with acute myeloid leukemia or high-risk myelodysplastic syndrome: predictive prognostic models for outcome. Cancer 106:1090–1098CrossRefPubMed Kantarjian H, O’Brien S, Cortes J et al (2006) Results of intensive chemotherapy in 998 patients age 65 years or older with acute myeloid leukemia or high-risk myelodysplastic syndrome: predictive prognostic models for outcome. Cancer 106:1090–1098CrossRefPubMed
6.
Zurück zum Zitat Lang K, Earle CC, Foster T et al (2005) Trends in the treatment of acute myeloid leukaemia in the elderly. Drugs Aging 22:943–955CrossRefPubMed Lang K, Earle CC, Foster T et al (2005) Trends in the treatment of acute myeloid leukaemia in the elderly. Drugs Aging 22:943–955CrossRefPubMed
8.
Zurück zum Zitat Williams JP, Handler HL (2000) Antibody-targeted chemotherapy for the treatment of relapsed acute myeloid leukemia. Am J Manag Care 6:S975–S985PubMed Williams JP, Handler HL (2000) Antibody-targeted chemotherapy for the treatment of relapsed acute myeloid leukemia. Am J Manag Care 6:S975–S985PubMed
9.
Zurück zum Zitat Herr AL, Labopin M, Blaise D et al (2007) HLA-identical sibling allogeneic peripheral blood stem cell transplantation with reduced intensity conditioning compared to autologous peripheral blood stem cell transplantation for elderly patients with de novo acute myeloid leukemia. Leukemia 21:129–135CrossRefPubMed Herr AL, Labopin M, Blaise D et al (2007) HLA-identical sibling allogeneic peripheral blood stem cell transplantation with reduced intensity conditioning compared to autologous peripheral blood stem cell transplantation for elderly patients with de novo acute myeloid leukemia. Leukemia 21:129–135CrossRefPubMed
10.
Zurück zum Zitat Storb R (2007) Can reduced-intensity allogeneic transplantation cure older adults with AML? Best Pract Res Clin Haematol 20:85–90CrossRefPubMed Storb R (2007) Can reduced-intensity allogeneic transplantation cure older adults with AML? Best Pract Res Clin Haematol 20:85–90CrossRefPubMed
11.
Zurück zum Zitat Juliusson G (2011) Older patients with acute myeloid leukemia benefit from intensive chemotherapy: an update from the Swedish Acute Leukemia Registry. Clin Lymphoma Myeloma Leuk 11(Suppl 1):S54–S59CrossRefPubMed Juliusson G (2011) Older patients with acute myeloid leukemia benefit from intensive chemotherapy: an update from the Swedish Acute Leukemia Registry. Clin Lymphoma Myeloma Leuk 11(Suppl 1):S54–S59CrossRefPubMed
12.
Zurück zum Zitat Juliusson G, Antunovic P, Derolf A et al (2009) Age and acute myeloid leukemia: real world data on decision to treat and outcomes from the Swedish Acute Leukemia Registry. Blood 113:4179–4187CrossRefPubMed Juliusson G, Antunovic P, Derolf A et al (2009) Age and acute myeloid leukemia: real world data on decision to treat and outcomes from the Swedish Acute Leukemia Registry. Blood 113:4179–4187CrossRefPubMed
13.
Zurück zum Zitat Murthy VH, Krumholz HM, Gross CP (2004) Participation in cancer clinical trials: race-, sex-, and age-based disparities. JAMA 291:2720–2726CrossRefPubMed Murthy VH, Krumholz HM, Gross CP (2004) Participation in cancer clinical trials: race-, sex-, and age-based disparities. JAMA 291:2720–2726CrossRefPubMed
14.
Zurück zum Zitat Hutchins LF, Unger JM, Crowley JJ et al (1999) Underrepresentation of patients 65 years of age or older in cancer-treatment trials. N Engl J Med 341:2061–2067CrossRefPubMed Hutchins LF, Unger JM, Crowley JJ et al (1999) Underrepresentation of patients 65 years of age or older in cancer-treatment trials. N Engl J Med 341:2061–2067CrossRefPubMed
15.
Zurück zum Zitat Gross CP, Murthy V, Li Y et al (2004) Cancer trial enrollment after state-mandated reimbursement. J Natl Cancer Inst 96:1063–1069CrossRefPubMed Gross CP, Murthy V, Li Y et al (2004) Cancer trial enrollment after state-mandated reimbursement. J Natl Cancer Inst 96:1063–1069CrossRefPubMed
16.
Zurück zum Zitat Mengis C, Aebi S, Tobler A et al (2003) Assessment of differences in patient populations selected for excluded from participation in clinical phase III acute myelogenous leukemia trials. J Clin Oncol 21:3933–3939CrossRefPubMed Mengis C, Aebi S, Tobler A et al (2003) Assessment of differences in patient populations selected for excluded from participation in clinical phase III acute myelogenous leukemia trials. J Clin Oncol 21:3933–3939CrossRefPubMed
17.
Zurück zum Zitat Warren JL, Klabunde CN, Schrag D et al (2002) Overview of the SEER-medicare data: content, research applications, and generalizability to the United States elderly population. Med Care 40:IV-3–IV-18 Warren JL, Klabunde CN, Schrag D et al (2002) Overview of the SEER-medicare data: content, research applications, and generalizability to the United States elderly population. Med Care 40:IV-3–IV-18
18.
Zurück zum Zitat Potosky AL, Riley GF, Lubitz JD et al (1993) Potential for cancer related health services research using a linked Medicare-tumor registry database. Med Care 31:732–748CrossRefPubMed Potosky AL, Riley GF, Lubitz JD et al (1993) Potential for cancer related health services research using a linked Medicare-tumor registry database. Med Care 31:732–748CrossRefPubMed
19.
Zurück zum Zitat Vardiman JW, Thiele J, Arber DA et al (2009) The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood 114:937–951CrossRefPubMed Vardiman JW, Thiele J, Arber DA et al (2009) The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood 114:937–951CrossRefPubMed
20.
Zurück zum Zitat Davidoff AJ, Tang M, Seal B et al (2010) Chemotherapy and survival benefit in elderly patients with advanced non-small-cell lung cancer. J Clin Oncol 28:2191–2197CrossRefPubMed Davidoff AJ, Tang M, Seal B et al (2010) Chemotherapy and survival benefit in elderly patients with advanced non-small-cell lung cancer. J Clin Oncol 28:2191–2197CrossRefPubMed
21.
Zurück zum Zitat Klabunde CN, Legler JM, Warren JL et al (2007) A refined comorbidity measurement algorithm for claims-based studies of breast, prostate, colorectal, and lung cancer patients. Ann Epidemiol 17:584–590CrossRefPubMed Klabunde CN, Legler JM, Warren JL et al (2007) A refined comorbidity measurement algorithm for claims-based studies of breast, prostate, colorectal, and lung cancer patients. Ann Epidemiol 17:584–590CrossRefPubMed
22.
Zurück zum Zitat Charlson ME, Pompei P, Ales KL et al (1987) A new method of classifying prognostic comorbidity in longitudinal studies: development and validation. J Chronic Dis 40:373–383CrossRefPubMed Charlson ME, Pompei P, Ales KL et al (1987) A new method of classifying prognostic comorbidity in longitudinal studies: development and validation. J Chronic Dis 40:373–383CrossRefPubMed
23.
Zurück zum Zitat Warren JL, Harlan LC, Fahey A et al (2002) Utility of the SEER-medicare data to identify chemotherapy use. Med Care 40:IV-55-–IV-61 Warren JL, Harlan LC, Fahey A et al (2002) Utility of the SEER-medicare data to identify chemotherapy use. Med Care 40:IV-55-–IV-61
24.
25.
Zurück zum Zitat Kurth T, Walker AM, Glynn RJ et al (2006) Results of multivariable logistic regression, propensity matching, propensity adjustment, and propensity-based weighting under conditions of nonuniform effect. Am J Epidemiol 163:262–270CrossRefPubMed Kurth T, Walker AM, Glynn RJ et al (2006) Results of multivariable logistic regression, propensity matching, propensity adjustment, and propensity-based weighting under conditions of nonuniform effect. Am J Epidemiol 163:262–270CrossRefPubMed
26.
Zurück zum Zitat Rosenbaum PR, Rubin DB (1983) The central rose of the propensity score in observational studies for causal effects. Biometrika 70:41–55CrossRef Rosenbaum PR, Rubin DB (1983) The central rose of the propensity score in observational studies for causal effects. Biometrika 70:41–55CrossRef
27.
Zurück zum Zitat Lowenberg B, Zittoun R, Kerkhofs H et al (1989) On the value of intensive remission-induction chemotherapy in elderly patients of 65+ years with acute myeloid leukemia: a randomized phase III study of the European Organization for Research and Treatment of Cancer Leukemia Group. J Clin Oncol 7:1268–1274PubMed Lowenberg B, Zittoun R, Kerkhofs H et al (1989) On the value of intensive remission-induction chemotherapy in elderly patients of 65+ years with acute myeloid leukemia: a randomized phase III study of the European Organization for Research and Treatment of Cancer Leukemia Group. J Clin Oncol 7:1268–1274PubMed
28.
Zurück zum Zitat Kantarjian HM, Thomas XG, Dmoszynska A et al (2012) Multicenter, randomized, open-label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytarabine for the treatment of older patients with newly diagnosed acute myeloid leukemia. J Clin Oncol 30:2670–2677CrossRefPubMed Kantarjian HM, Thomas XG, Dmoszynska A et al (2012) Multicenter, randomized, open-label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytarabine for the treatment of older patients with newly diagnosed acute myeloid leukemia. J Clin Oncol 30:2670–2677CrossRefPubMed
29.
Zurück zum Zitat Fenaux P, Mufti GJ, Hellstrom-Lindberg E et al (2010) Azacitidine prolongs overall survival compared with conventional care regimens in elderly patients with low bone marrow blast count acute myeloid leukemia. J Clin Oncol 28:562–569CrossRefPubMed Fenaux P, Mufti GJ, Hellstrom-Lindberg E et al (2010) Azacitidine prolongs overall survival compared with conventional care regimens in elderly patients with low bone marrow blast count acute myeloid leukemia. J Clin Oncol 28:562–569CrossRefPubMed
30.
Zurück zum Zitat Dombret H, Seymour JF, Butrym A et al (2014) Results of a Phase 3, Multicenter, Randomized, Open-Label Study of Azacitidine (AZA) vs Conventional Care Regimens (CCR). In: Older Patients with Newly Diagnosed Acute Myeloid Leukemia (AML). 19th Congress of the European Hematology Association Milan, Italy Dombret H, Seymour JF, Butrym A et al (2014) Results of a Phase 3, Multicenter, Randomized, Open-Label Study of Azacitidine (AZA) vs Conventional Care Regimens (CCR). In: Older Patients with Newly Diagnosed Acute Myeloid Leukemia (AML). 19th Congress of the European Hematology Association Milan, Italy
31.
32.
Zurück zum Zitat Estey E, de Lima M, Tibes R et al (2007) Prospective feasibility analysis of reduced-intensity conditioning (RIC) regimens for hematopoietic stem cell transplantation (HSCT) in elderly patients with acute myeloid leukemia (AML) and high-risk myelodysplastic syndrome (MDS). Blood 109:1395–1400CrossRefPubMed Estey E, de Lima M, Tibes R et al (2007) Prospective feasibility analysis of reduced-intensity conditioning (RIC) regimens for hematopoietic stem cell transplantation (HSCT) in elderly patients with acute myeloid leukemia (AML) and high-risk myelodysplastic syndrome (MDS). Blood 109:1395–1400CrossRefPubMed
33.
Zurück zum Zitat Wang M, Burau KD, Fang S et al (2008) Ethnic variations in diagnosis, treatment, socioeconomic status, and survival in a large population-based cohort of elderly patients with non-Hodgkin lymphoma. Cancer 113:3231–3241CrossRefPubMed Wang M, Burau KD, Fang S et al (2008) Ethnic variations in diagnosis, treatment, socioeconomic status, and survival in a large population-based cohort of elderly patients with non-Hodgkin lymphoma. Cancer 113:3231–3241CrossRefPubMed
34.
Zurück zum Zitat Shavers VL, Brown ML (2002) Racial and ethnic disparities in the receipt of cancer treatment. J Natl Cancer Inst 94:334–357CrossRefPubMed Shavers VL, Brown ML (2002) Racial and ethnic disparities in the receipt of cancer treatment. J Natl Cancer Inst 94:334–357CrossRefPubMed
35.
Zurück zum Zitat Aizer AA, Chen MH, McCarthy EP et al (2013) Marital status and survival in patients with cancer. J Clin Oncol 31:3869–3876CrossRefPubMed Aizer AA, Chen MH, McCarthy EP et al (2013) Marital status and survival in patients with cancer. J Clin Oncol 31:3869–3876CrossRefPubMed
36.
Zurück zum Zitat Juliusson G, Billstrom R, Gruber A et al (2006) Attitude towards remission induction for elderly patients with acute myeloid leukemia influences survival. Leukemia 20:42–47CrossRefPubMed Juliusson G, Billstrom R, Gruber A et al (2006) Attitude towards remission induction for elderly patients with acute myeloid leukemia influences survival. Leukemia 20:42–47CrossRefPubMed
37.
Zurück zum Zitat Buchner T, Hiddemann W, Berdel W et al (2002) Acute myeloid leukemia: treatment over 60. Rev Clin Exp Hematol 6:46–59, discussion 86-7CrossRefPubMed Buchner T, Hiddemann W, Berdel W et al (2002) Acute myeloid leukemia: treatment over 60. Rev Clin Exp Hematol 6:46–59, discussion 86-7CrossRefPubMed
38.
Zurück zum Zitat Nabhan C, Smith SM, Helenowski I et al (2012) Analysis of very elderly (>/=80 years) non-hodgkin lymphoma: impact of functional status and co-morbidities on outcome. Br J Haematol 156:196–204CrossRefPubMed Nabhan C, Smith SM, Helenowski I et al (2012) Analysis of very elderly (>/=80 years) non-hodgkin lymphoma: impact of functional status and co-morbidities on outcome. Br J Haematol 156:196–204CrossRefPubMed
39.
Zurück zum Zitat Oxnard GR, Fidias P, Muzikansky A et al (2007) Non-small cell lung cancer in octogenarians: treatment practices and preferences. J Thorac Oncol 2:1029–1035CrossRefPubMed Oxnard GR, Fidias P, Muzikansky A et al (2007) Non-small cell lung cancer in octogenarians: treatment practices and preferences. J Thorac Oncol 2:1029–1035CrossRefPubMed
40.
Zurück zum Zitat Dale DC (2003) Poor prognosis in elderly patients with cancer: the role of bias and undertreatment. J Support Oncol 1:11–17PubMed Dale DC (2003) Poor prognosis in elderly patients with cancer: the role of bias and undertreatment. J Support Oncol 1:11–17PubMed
41.
Zurück zum Zitat Dohner H, Lubbert M, Fiedler W et al (2014) Randomized, phase 2 trial of low-dose cytarabine with or without volasertib in AML patients not suitable for induction therapy. Blood 124:1426–1433CrossRefPubMedCentralPubMed Dohner H, Lubbert M, Fiedler W et al (2014) Randomized, phase 2 trial of low-dose cytarabine with or without volasertib in AML patients not suitable for induction therapy. Blood 124:1426–1433CrossRefPubMedCentralPubMed
42.
Zurück zum Zitat Lancet JE, Cortes JE, Hogge DE et al (2014) Phase 2 trial of CPX-351, a fixed 5:1 molar ratio of cytarabine/daunorubicin, vs cytarabine/daunorubicin in older adults with untreated AML. Blood 123:3239–3246CrossRefPubMed Lancet JE, Cortes JE, Hogge DE et al (2014) Phase 2 trial of CPX-351, a fixed 5:1 molar ratio of cytarabine/daunorubicin, vs cytarabine/daunorubicin in older adults with untreated AML. Blood 123:3239–3246CrossRefPubMed
43.
Zurück zum Zitat Burnett AK, Russell NH, Hills RK et al (2012) Addition of gemtuzumab ozogamicin to induction chemotherapy improves survival in older patients with acute myeloid leukemia. J Clin Oncol 30:3924–3931CrossRefPubMed Burnett AK, Russell NH, Hills RK et al (2012) Addition of gemtuzumab ozogamicin to induction chemotherapy improves survival in older patients with acute myeloid leukemia. J Clin Oncol 30:3924–3931CrossRefPubMed
44.
Zurück zum Zitat Castaigne S, Pautas C, Terre C et al (2012) Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study. Lancet 379:1508–1516CrossRefPubMed Castaigne S, Pautas C, Terre C et al (2012) Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study. Lancet 379:1508–1516CrossRefPubMed
Metadaten
Titel
Big data analysis of treatment patterns and outcomes among elderly acute myeloid leukemia patients in the United States
verfasst von
Bruno C. Medeiros
Sacha Satram-Hoang
Deborah Hurst
Khang Q. Hoang
Faiyaz Momin
Carolina Reyes
Publikationsdatum
01.07.2015
Verlag
Springer Berlin Heidelberg
Erschienen in
Annals of Hematology / Ausgabe 7/2015
Print ISSN: 0939-5555
Elektronische ISSN: 1432-0584
DOI
https://doi.org/10.1007/s00277-015-2351-x

Weitere Artikel der Ausgabe 7/2015

Annals of Hematology 7/2015 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

„Jeder Fall von plötzlichem Tod muss obduziert werden!“

17.05.2024 Plötzlicher Herztod Nachrichten

Ein signifikanter Anteil der Fälle von plötzlichem Herztod ist genetisch bedingt. Um ihre Verwandten vor diesem Schicksal zu bewahren, sollten jüngere Personen, die plötzlich unerwartet versterben, ausnahmslos einer Autopsie unterzogen werden.

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Schlechtere Vorhofflimmern-Prognose bei kleinem linken Ventrikel

17.05.2024 Vorhofflimmern Nachrichten

Nicht nur ein vergrößerter, sondern auch ein kleiner linker Ventrikel ist bei Vorhofflimmern mit einer erhöhten Komplikationsrate assoziiert. Der Zusammenhang besteht nach Daten aus China unabhängig von anderen Risikofaktoren.

Semaglutid bei Herzinsuffizienz: Wie erklärt sich die Wirksamkeit?

17.05.2024 Herzinsuffizienz Nachrichten

Bei adipösen Patienten mit Herzinsuffizienz des HFpEF-Phänotyps ist Semaglutid von symptomatischem Nutzen. Resultiert dieser Benefit allein aus der Gewichtsreduktion oder auch aus spezifischen Effekten auf die Herzinsuffizienz-Pathogenese? Eine neue Analyse gibt Aufschluss.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.