Abstract
Purpose of review:
The acute myeloid leukemia (AML) treatment landscape has rapidly evolved over the past few years. These changes have several implications for the care of older adults (≥ 60 years), who have inferior clinical outcomes. We review decision-making in older adults, focusing on patient- and disease-related factors. We then summarize current treatment options, including multiple recently approved therapies, based on hypothetical clinical scenarios.
Recent findings:
In lieu of using chronological age to determine fitness, we highlight the importance of standardized fitness assessments using geriatric assessments. Next, we review intensive and lower-intensity treatment options in the upfront setting. We focus on multiple newly approved medications, including venetoclax, midostaurin, CPX-351, gemtuzumab, glasdegib, enasidenib, and ivosidenib, and their specific indications. Lastly, we briefly discuss supportive care of older adults with AML.
Summary:
Outcomes of older adults with AML remain poor; fortunately, there are many new promising treatment options. Personalized treatment plans based on patient- and disease-specific factors are essential to the care of older adults with AML.
Keywords: acute myeloid leukemia, treatment, older adults, geriatric hematology
Introduction
Acute myeloid leukemia (AML) is the second most common leukemia in adults. In 2019, an estimated 21,450 new cases and 10,920 deaths were reported in the United States.1 Older adults account for most cases, with a median age of 68 years at diagnosis. Incidence rates of AML increase with age from 1.3 per 100,000 in adults < 50 years, to 5.1 per 100,000 in adults 50–64 years, to 20.1 per 100,000 in adults ≥ 65 years. Five-year overall survival (OS) is poor and estimated to be 28.3% for all patients with AML and as low as 3–8% in patients ≥ 60 years.1,2
The National Comprehensive Cancer Care Network (NCCN) defines older patients as age ≥ 60 years,3 while the European Leukemia Network (ELN) defines older age less rigidly as ≥ 60–65 years.4 This aged-based distinction is relevant since AML in older adults is a distinct clinical entity compared to AML in younger patients.5 Both disease- and patient-specific differences exist.5 Unfavorable cytogenetics and expression of multidrug resistance are more common in older adults.5 Also, the incidence of secondary AML (sAML), due to underlying myelodysplastic syndrome (MDS) or therapy for other malignancies, increases with age.6 Each of these factors is an independent risk factor for worse prognosis and is associated with poorer response to chemotherapy and lower complete remission (CR) rates compared to younger patients. Comorbidities, as well as cognitive and functional impairments, are more common in older adults and contribute to poorer physiologic reserves and greater treatment-related toxicity.7
Between 2000 and 2010, only 40% of patients ≥ 65 years received AML-directed therapy.1 This treatment disparity was more pronounced in the oldest group of patients (> 80 years); in this group, only 20% received antileukemic treatment.1,8 Recent studies have shown a trend towards more frequent use of leukemia-directed therapy in adults 65–80 years, which has been associated with improved survival and quality of life.9,10 With the decreasing treatment disparity in older adults, and the emergence of multiple new therapies, the question of how best to treat AML in older or less fit adults becomes increasingly relevant.
In this article, we highlight various factors clinicians should consider when managing older patients with AML and review the current therapeutic landscape with a focus on novel therapies.
Initial Evaluation of an Older Adult with Newly Diagnosed AML
Treatment Decision-Making & Prognosis
Treatment decision-making is a complex yet critical component of care of all patients with AML and it is arguably more challenging in older patients. Treatment tolerability and benefits are important factors that patients and physicians consider during treatment decision-making.11,12 Predicting toxicity and treatment success is difficult in older adults. Not only are older adults underrepresented in cancer-related clinical trials, but those enrolled on trials may not be representative of patients seen in practice, thereby limiting generalizability of findings.13 Often clinicians use expertise or experience alongside evidence to guide decision-making.14 However, many currently approved and NCCN-recommended therapies for adults ≥ 60 years are novel, so providers may have limited experience with them.3 Older adults may also value maintenance of independence, time spent at home, cognition, emotional well-being, and health-related quality of life (HRQoL) over survival.8 Fortunately, clinical trials have begun to include patient-reported outcomes such as HRQoL measures in addition to survival outcomes.15
Patient-related factors:
Increasing age is associated with poor prognosis.5 This is likely at least partially due to the presence of unmeasured factors such as poor performance status and high comorbidity indexes.8,16 Performance status is frequently measured using the Eastern Cooperative Oncology Group Performance Status (ECOG PS) scale, which is based on clinicians’ general impression. In patients with poor performance status (ECOG ≥ 3), it may predict functional status and prognosis well; however, this is not consistent across scores.8,16,17 As such, reliance on clinical gestalt and ECOG performance status to evaluate an older patient is often insufficient. Older patients diagnosed with AML often develop rapid disability due to the acuity of the disease, which further complicates assessments of their health.
A geriatric assessment (GA) is a comprehensive method that can be used to identify vulnerabilities in older adults that may impact their cancer treatment. 17,18 Components evaluated in a GA include functional status, physical performance and falls, comorbid medical conditions, psychological health, social activity/support, medications, nutritional status, and cognition.19 Each of these domains is assessed with standardized questionnaires and/or objective measures. For example, functional status can be evaluated by assessing activities of daily living (ADLs) and instrumental activities of daily living (IADLS) and by using performance-based measures such as grip strength or the Short Physical Performance Battery.20 Multiple options for assessments by non-geriatrician have been developed, including screening tools and online geriatric assessment–based calculators, which have been reviewed previously.21
Studies have demonstrated that GA can predict prognosis in older patients with AML.17 The NCCN, the American Society of Clinical Oncology, and the International Society of Geriatric Oncology recommend geriatric assessment in patients ≥ 65 years, especially those receiving chemotherapy, to identify impairments that may be missed by routine oncologic assessment tools.19,22,23
Disease-related factors:
With advancing age, leukemia cell biology changes which contributes to poor prognosis. Unfavorable risk cytogenetics, such as monosomal karyotypes and complex cytogenetics (≥ 3 acquired chromosome aberrations), are more common with increasing age.5,24 Secondary AML is also more frequent with increasing age, as described above.24,25
Molecular genetic testing and conventional cytogenetic analysis are important in the diagnostic work-up for AML.26 Next generation sequencing (NGS) and gene expression analysis have restructured our understanding of AML.27 This is reflected by the incorporation of mutations in the 2016 updated World Health Organization (WHO) AML subtype classifications and the NCCN treatment guidelines.28
Important actionable mutations that may be identified include fms-like tyrosine kinase 3–internal tandem duplication (FLT3-ITD) and isocitrate dehydrogenase (IDH) 1 and 2.26 Additional mutations that provide prognostic information include FLT3 tyrosine kinase domain (FLT3-TKD), CCAAT/enhancer-binding protein a (CEBPA), DNA methyltransferase 3 alpha (DNMT3A), tumor protein p53 (TP53), tet methylcytosine dioxygenase 2 (TET2), Wilms tumor 1 (WT1), DNA methyltransferase 3 alpha (DNMT3A), Nucleophosmin (NPM1), and runt-related transcription factor 1 (RUNX1).26 Early molecular genetic studies included few, if any, older adults29 but more recent data has shown that molecular profiles differ by age.30,31 For example, in one report of patients with AML and normal karyotype, the frequencies of both NPM1 and FLT3-ITD mutations were shown to decrease with age (NPM1: 50–59 years, 60%; 60–69 years, 43%; ≥ 70 years 41%; FLT-ITD: 50–59 years, 31%; 60–69 years, 25%; ≥ 70 years 20%).32 On the other hand, the frequencies of different mutations including FLT3-TKD and CEBPA did not change with increasing age.32 IDH 1/2 mutations are also more frequent in older patients and often co-occur with NPM1 and FLT3-ITD.33 Understanding the implications of gene expression profiling is an area of active research. We summarize important genetic mutations in older adults with AML in Table 1.
Table 1:
Common genetic mutations in older adults with AML
Genetic mutations | Prognostic implications and genetic risk stratification group* | Comments | Frequencies |
---|---|---|---|
NPM1 | • Prognosis is complex and depends on gene-gene interactions4 • Genetic risk stratification group: ○ Favorable: Mutated NPM1 without FLT3-ITD (or with FLT3-ITDlow †) ○ Intermediate: Mutated with FLT3-ITDhigh † ○ Adverse: Wild-type NPM1 with FLT3-ITDhigh †4 |
• Screen in all patients at diagnosis, mutation defines risk category4 • Specific for de novo AML and most common in cytogenetically normal AML4 • Frequency of mutations decrease with age32 |
• Mutated in 25–30%92 • Frequency increases with age: 50–59 years, 60%; 60–69 years, 43%; ≥ 70 years 41%32 |
FLT3-ITD | • Depends on NPM1, see above | • Screen in all patients at diagnosis, mutations are prognostic, and may guide treatment decision-making related to targeted novel therapies (i.e., FLT3 inhibitors)4 • Frequency of FLT3-ITD mutations decrease with age (frequency of FLT3-TKD does not change with age)32 |
• Mutated in 20–25% of AML in adults32,49,92 • Frequency increases with age: 50–59 years, 31%; 60–69 years, 25%; ≥ 70 years 20%32 |
CEBPA†† | • Genetic risk stratification group: favorable (Biallelic mutated CEBPA)4 | • Screen in all patients at diagnosis, mutation defines risk category4 | • Biallelic mutations present in 6–10%92 • No change with age32 |
IDH 1/2 | • Genetic risk stratification group: not assigned due to insufficient evidence4 | • Screen in all patients at diagnosis, may guide treatment decision-making related to targeted novel therapies (i.e., IDH 1/2 inhibitors)4 • More frequent in older patients32 • Often co-occur with NPM1 and FLT3-ITD4,32,92 |
• IDH1: 6–14%72,92 • IDH2: 8–19%92 • Frequencies of IDH1/2 increase with age32,33,92 |
RUNX1 | • Poor prognosis; genetic risk stratification group: adverse 4 • Presence in MDS predicts increased risk of progression to AML4 • Associated with secondary AML and resistance to induction92 |
• Screen in all patients at diagnosis, mutation defines risk category4 | • 10% of patients with de novo AML93 • Frequency increases with age92 |
TP53 | • Poor prognosis; genetic risk stratification group: adverse 4 • Presence in MDS predicts increased risk of progression to AML4 • Associated with monosomal and complex karyotypes (combinations have the worst outcomes)4 |
• Screen in all patients at diagnosis, mutation defines risk category4 • Associated with chemoresistance and high relapse risk94 |
• TP53 is a tumor suppressor gene that is mutated in many cancers but is rarely mutated in de novo AML: 8% of de novo AML; 27% of therapy-related AML; in older patients with complex karyotypes incidence is up to 70%94 |
Abbreviations: AML, acute myeloid leukemia; NPM1, Nucleophosmin; FLT3-ITD, fms-like tyrosine kinase 3-internal tandem duplication; IDH, isocitrate dehydrogenase; FLT3-TKD, fms-like tyrosine kinase domain; CEBPA, CCAAT/enhancer-binding protein a; DNMT3A, DNA methyltransferase 3 alpha; TP53, tumor protein p53; WT1, Wilms tumor 1; RUNX1, runt-related transcription factor 1.
Genetic risk stratification groups are based on the 2017 European Leukemia Network Classification.4
Low, allelic ratio <0.5; High, allelic ratio ≥0.5
Only AML with biallelic mutated CEBPA is included in the clinicopathologic entity
Several targeted treatment options have recently received Food and Drug Administration (FDA) approval for use in AML with specific mutations, as discussed next.
Treatment
AML treatment is broadly divided into intensive and lower-intensity options. Select trials are shown in Table 2 and 3. Since 2017, eight new targeted therapies have received FDA approval for AML treatment.34 In the setting of these recent approvals, hematologists are faced with additional challenges when deciding on appropriate treatments. Early identification of actionable mutations and the ability to anticipate and manage drug complications are essential.3,35 As discussed above, information from fitness assessments rather than chronological age alone may assist in predicting outcomes and with treatment decision-making.13,36
Table 2:
Select clinical trials of intensive therapy in acute myeloid leukemia that are applicable to older adults
Intensive induction regimen | Indication in study | Median age (years) | N | CR (%) | OS | Toxicity | Study |
---|---|---|---|---|---|---|---|
Daunorubicin 90 mg/m2/day with cytarabine | Newly diagnosed AML or high-risk MDS | 67 | 402 | 64 | 31% at 2 years | • Longer interval between 1st and 2nd cycle in the higher-dose group (43 vs. 38 days, P=0.001) • No significant difference in the rate of moderate, severe, or life-threatening side effects between the two groups |
Lowenberg et al., 200940 |
vs. Daunorubicin 45 mg/m2/day with cytarabine | 411 | 54 (P=0.002) | 26% at 2 years (P=0.16) | ||||
Daunorubicin 90 mg/m2/day with cytarabine | 53 | 604 | 73 | 59% at 2 years | • More grade 3–4 GI toxicity in the higher-dose group | Burnett et al., 201539 | |
vs. Daunorubicin 60 mg/m2/day with cytarabine | 602 | 75 (P=0.60) | 60% at 2 years (P=0.15) | ||||
Midostaurin added to cytarabine and daunorubicin (50 mg twice daily for 14 days starting induction day 8) | Newly diagnosed AML with FLT3 mutation | 48 | 360 | 59 | Median: 74.7 months | • Anemia, grade ≥3 higher in the midostaurin group (92.7% vs. 87.8%, P=0.03) • Rash higher in the midostaurin group (14.1% vs. 7.6%, P=0.008) |
Stone et al., 201751 |
vs. Cytarabine and daunorubicin | 357 | 54 (P=0.15) | Median: 25.6 months (P=0.009) | • Nausea (9.6% vs. 5.6%, P=0.05) higher in the midostaurin group | |||
CPX-351 (100 mg/m2 cytarabine + 44 mg/m2 daunorubicin on induction days 1, 3, 5) |
Newly diagnosed secondary AML | 68 | 153 | 48 | Median: 9.6 months | • Febrile neutropenia higher in the CPX-351 group (68.0% v 70.9%) • Pneumonia higher in the CPX-351 group (19.6% v 14.6%) • Hypoxia lower in the CPX-351 group (13.1% v 15.2%) • Prolonged time to neutrophil (median 35 vs. 29 days) and platelet count recovery (median 37 vs. 29 days) in the CPX-351 group |
Lancet et al., 201855 |
vs. Cytarabine and daunorubicin | 156 | 33 (P=0.016) | Median: 6.0 months (P=0.003) | ||||
Gemtuzumab ozogomycin added to cytarabine and daunorubicin (3 mg/m2 on days 1, 4, 7) | Newly diagnosed AML | 62 | 139 | 75 | 41.9% at 2 years | • Persistent thrombocytopenia more common in the GO group (16% vs 3%, P<0.0001) • Veno-occlusive disease (occurred in 3/139 patients, 2 of the 3 patients died) in the GO group |
Castaigne et al., 201242 |
vs. Cytarabine and daunorubicin | 139 | 81 (P=0.25) | 53.2% at 2 years (P=0.03) | ||||
Gemtuzumab ozogomycin added to cytarabine and daunorubicin (3 mg/m2 on day 1) |
Newly diagnosed AML or high risk MDS | 67 | 556 | 70 | 25% at 3 years | • Nausea and vomiting, grade 3–4 higher in the GO group (9% vs. 4%, P=0.002) • Hyperbilirubinemia, grade 3–4 higher in the GO group (7% vs. 6%, P=0.001) • Increased number of platelet unit transfusions in the GO group (13.7 vs. 9.6, P<0.001) • Days of intravenous antibiotics longer in the group without GO (19.2 vs. 18.1, P=0.04) |
Burnett et al., 201244 |
vs. Cytarabine and daunorubicin | 559 | 68 (P=0.3) | 20% at 3 years (P=0.05) |
Abbreviations: AML, acute myeloid leukemia; N, number of patients enrolled; CR, complete remission; OS, overall survival; ‘7+3’, 7 days of cytarabine and 3 days of anthracycline; BSC best supportive care; LDAC, low dose cytarabine; HMA, hypomethylating agent; MDS, myelodysplastic syndrome; FLT3-ITD, fms-like tyrosine kinase 3-internal tandem duplication; HR, hazard ratio; GI, gastrointestinal
Table 3:
Select clinical trials of lower-intensity treatment options in acute myeloid leukemia that are applicable to older adults
Lower-intensity regimens | Indication in trial | Median age (years) | N | CR (%) | Median OS (months) | Toxicity | Study |
---|---|---|---|---|---|---|---|
Low dose cytarabine ± ATRA (20 mg twice daily, for 10 days, every 4 weeks until progression) | Patients not considered candidates for intensive chemotherapy | 74 | 102 | 18 | Not available, improved with LDAC | • No difference compared to BSC | Burnett et al., 200786 |
vs. Hydroxyurea ± ATRA | 99 | 1 (P<0.05) | |||||
Decitabine (20 mg/m2, for 5 days, every 4 weeks until progression) | 73 | 242 | 17.8 | 7.7 | • Rate of death attributed to AE did not differ between groups | Kantarjian et al., 201256 | |
vs. LDAC or supportive care | 243 | 7.8 (P<0.05) | 5.0 (P=0.20) |
||||
Azacitidine (75 mg/m2/day for 7 days, every 4 weeks for at least 6 cycles) | 70 | 18 | 24.5 | • No increased toxicity compared to CCR • Fewer hospital admissions compared to CCR (3.4 vs. 4.3 per patient-year) • Higher rate of fever requiring intravenous antibiotics in the CCR group (1.1 vs. 0.6 instances per patient-year) |
Fenaux et al., 201058 | ||
vs. CCR (IC, LDAC, BSC) | 16 (P>0.05) | 16 (P<0.05) | |||||
Venetoclax + HMA
(Venetoclax ramp-up* with decitabine 20 mg/m2/day for 5 days or azacitidine 75 mg/m2/day for 7 days) |
74 | 145 | 67 | 17.5 | • Common AEs (>30%): GI symptoms, febrile neutropenia and fatigue • AE leading to venetoclax dose interruption (47%) • No tumor lysis syndrome |
DiNardo et al., 201960 | |
Venetoclax + LDAC (Venetoclax ramp-up* with LDAC 20 mg/m2/day) |
74 | 82 | 54 | 10.1 | • Febrile neutropenia (42%) • Thrombocytopenia (38%) • WBC count decreased (34%) |
Wei et al., 201961 | |
Glasdegib with LDAC (Glasdegib 100 mg/day on days 1 to 28) |
76 | 88 | 17 | 8.8 | • Higher pneumonia, grade 3–4 in the combination group (16.7% vs. 14.6%) • Fatigue, grade 3–4 (14.3%) |
Cortes et al., 201967 | |
vs. LDAC | 44 | 2 (P<0.05) | 4.9 (P<0.05) | ||||
Gemtuzumab ozogomycin (6 mg/m2 on day 1, 3 mg/m2 on day 8) | Newly diagnosed CD33 mutated AML | 77 | 118 | 27 | 4.9 | • No difference in adverse events compared to BSC | Amadori et al., 201669 |
vs. BSC | 119 | 30 (P>0.05) | 3.6 (P<0.05) | ||||
Ivosidenib (Dose range, 200–1200 mg daily for 28 days) | Newly diagnosed IDH1 mutated AML | 77 | 34 | 30 | Not available yet | • Diarrhea (53%), nausea (38%) • Peripheral edema (26%) • Differentiation syndrome: leukocytosis and nonspecific symptoms (18%) |
Roboz et al., 201987 |
Enasidenib (Total doses of 50–650 mg daily for 28days) | Newly diagnosed IDH2 mutated AML | 77 | 39 | 18 | 11.3 (data for responders not yet available) | • Grade 3–4 cytopenias (21%) • Indirect hyperbilirubinemia (31%) • GI complaints (23%) • Fatigue (18%) |
Pollyea et al., 201973 |
Venetoclax ramp-up, daily dose: day 1, 100 mg; day 2, 200 mg; day 3, 300 mg; day 4, 400 mg when combined with HMA or 600 mg when combined with LDAC. Dose adjustments are recommended in specific scenarios.
Abbreviations: AML, acute myeloid leukemia; N, number of patients enrolled; CR, complete remission; OS, overall survival; BSC, best supportive care; CCR, conventional care regimen; IC, intensive chemotherapy; LDAC, low dose cytarabine; HMA, hypomethylating agent; IDH, isocitrate dehydrogenase; FLT3-ITD, fms-like tyrosine kinase 3-internal tandem duplication; AE, adverse events; GI, gastrointestinal; WBC, white blood cell
Any patient diagnosed with AML who is interested in pursuing treatment should be considered for a clinical trial. We will review several potential clinical scenarios of older patients with newly diagnosed AML, summarizing current treatment options and relevant key clinical trials. Figure 1 shows our proposed treatment algorithm for these clinical scenarios.
Figure 1: Treatment algorithm.
A proposed approach for older adults with newly diagnosed acute myeloid leukemia91
The dotted line represents a controversial treatment option: in fit older adults with poor-risk disease, options include intensive induction therapy with ‘7+3’, CPX-351, or lower-intensity options.
Abbreviations: AML, acute myeloid leukemia; GA, geriatric assessment; “7+3”, 7 days of cytarabine and 3 days of anthracycline; HMA, hypomethylating agent; LDAC, low-dose cytarabine; IDH, isocitrate dehydrogenase; FLT3-ITD, fms-like tyrosine kinase 3-internal tandem duplication
Clinical Scenario 1A: De novo AML in an adult ≥60 years considered a good candidate for induction, without unfavorable cytogenetics, and without CD33-positivity, FLT-3 mutation
Intensive therapy
A traditional induction chemotherapy regimen consists of 7 days of cytarabine and 3 days of anthracycline, often referred to as “7+3”. The goal is to achieve CR, which is successful in 40–60% of adults ≥ 60 years compared to 60–80% in younger adults.4
Anthracyclines and cytarabine
Daunorubicin is the most commonly used anthracycline for induction therapy in AML (60–90 mg/m2/day).3,37 In 2009, daunorubicin 45 mg/m2/day was compared to 90 mg/m2/day for induction in older adults with AML (median age 67 years).38 Higher CR was seen in adults treated with 90 mg/m2/day compared to 45 mg/m2/day (64% vs. 54%, P=0.002), without higher toxicity.38 However, due to concerns about toxicity with the higher dose, daunorubicin 60 mg/m2/day was frequently used thereafter.37 In 2015, daunorubicin 60 mg/m2/day was compared to 90 mg/m2/day (median age 53 years) for induction. The higher dose was associated with increased 60-day mortality (10% vs. 5%, P=0.001), without improved OS at 2 years (60% vs. 59%, P=0.15).37 Idarubicin is another commonly used anthracycline in AML induction. In older adults, idarubicin 12 mg/m2 for 3 days has been shown to have similar efficacy and tolerability to daunorubicin 80 mg/m2 for 3 days.39 Standard dose cytarabine (Ara-c) is 100–200 mg/m2/day for 7 days.3
Based on these studies, “7+3” induction with cytarabine 100–200 mg/m2/day for 7 days and daunorubicin 60–90 mg/m2/day or idarubicin 12 mg/m2/day for 3 days can be considered in patients ≥ 60 years who are candidates for intensive induction and do not have unfavorable-risk cytogenetics.
Clinical Scenario 1B: De novo AML in an adult ≥60 years considered a good candidate for induction, without unfavorable cytogenetics and with CD33 positivity
Gemtuzumab ozogamicin (GO)
Gemtuzumab ozogamicin (GO), an anti-CD33 conjugate, has been studied as an addition to frontline intensive therapy in patients with de novo AML. A randomized phase 3 trial evaluated the addition of GO (3 mg/m2/day on days 1, 4, and 7 during induction) to intensive chemotherapy in patients with AML aged 50–70 years (median age 62 years).40 Addition of GO was associated with improved event-free survival (EFS) [Hazard Ratio (HR) 0.58, P=0.0003], OS (HR 0.69, P=0.036), and relapse-free survival (RFS) (HR 0.52, P=0.0003). GO was associated with higher hematologic toxicity (mainly persistent thrombocytopenia) but not increased mortality.40,41 Another randomized trial compared adding GO (a single 3 mg/m2 dose) to induction chemotherapy in older adults with AML or high-risk MDS (median age 67 years). Compared to induction chemotherapy without GO, 3-year OS was improved in the group who received GO (20% vs. 25%, P=0.05), without additional toxicity.42 A third randomized controlled trial (RCT) evaluated the addition of GO (6 mg/m2/day on days 1 and 15) to standard induction therapy in older patients with AML (median age 67 years).43 This trial failed to demonstrate OS benefit. In a subgroup of patients 70–75 years, increased early mortality risk was noted. The aforementioned trials, along with a few other phase III trials evaluating the addition of GO to induction therapy44–47 showed that GO’s efficacy was observed mainly in patients with favorable risk disease and sometimes those with intermediate risk disease.48 Based on these data, in patients with favorable-risk AML and CD33-positivity, the addition of GO to induction can be considered.
Clinical Scenario 1C: De novo AML in an adult ≥60 years considered a good candidate for induction, without unfavorable cytogenetics, with a FLT3-mutation
FLT3 inhibitor
Midostaurin, an oral multitargeted kinase inhibitor, was evaluated in patients with AML (median age 48 years) and a FLT3 mutation.49 Midostaurin was added to induction (daunorubin 60 mg/m2/day for 3 days and cytarabine 200 mg/m2/day for 7 days) and consolidation chemotherapy. Although the midostaurin group had similar CR (58.9% vs. 53.5%, P=0.15), OS was longer (74.7 months vs. 25.6 months, P=0.009).49 The average patient age at trial entry was < 50 years, limiting the generalizability of these results to older patients. Nonetheless, midostaurin received FDA approval without an upper age-limit.50 The NCCN recommends the addition of midostaurin to induction with 7+3 in fit older adults with FLT3 mutations.3
Clinical Scenario 2: Secondary AML or AML with MDS-related changes in an adult ≥60 years considered a good candidate for induction
CPX-351
CPX-351, a liposomal encapsulation of cytarabine 100 mg/m2 and daunorubicin 44 mg/m2 at a synergistic 5:1 ratio, received FDA approval for the treatment of newly diagnosed sAML in 2017.51 CPX-351 was studied in the frontline setting in a randomized phase II trial of older patients (median age 68 years) with newly diagnosed de novo and secondary AML.52 CPX-351 showed a non-significant trend towards higher remission rates (defined as complete + incomplete remission) when compared to conventional induction therapy (66.7% vs 51.2%, P=0.07). In a subgroup analysis of patients with sAML, CPX-351 was associated with higher remission rates (57.6% vs 31.6%, P=0.06).52 A subsequent phase III randomized trial in older patients (median age 68 years) with newly diagnosed high-risk/secondary AML compared induction with 7+3 to CPX-351.53 CPX-351 led to an improved CR rate (47.7% vs. 33.3%, P=0.016) and OS (HR 0.69, P=0.003) compared to conventional 7+3; it was, however, associated with a longer treatment phase and prolonged cytopenia.53
In summary, CPX-351 is recommended for patients ≥ 60 years with sAML, including those with therapy-related disease, an antecedent hematologic disorder, or AML with myelodysplasia-related changes.
Lower-intensity treatment
Clinical Scenario 3: Adult ≥60 years with good functional status but unfavorable-risk cytogenetic or who is not a candidate for (or declines) intensive treatment
In older adults considered fit for intensive therapy with poor-risk cytogenetics or unfit for intensive therapy, lower-intensity treatment options are considered. It is important to note that assigning treatment based on cytogenetics is controversial (i.e., among older adults fit for intensive therapy with poor-risk cytogenetics, intensive therapy is often considered). Lower-intensity treatments are generally given as outpatient therapies with palliative intent and patients are generally not considered candidates for HSCT. Lower-intensity treatments may be administered as monotherapy or combinations, as discussed next.
Hypomethylating agents: Decitabine & Azacitidine
Decitabine (20 mg/m2/day for 5 days every 28 days) was compared with provider’s treatment choice [low-dose cytarabine (LDAC) or best supportive care] in a randomized phase III trial of older patients with AML (median age 73 years).54 In initial analyses, decitabine showed a nonsignificant but favorable trend towards increased median OS. Mature survival data showed that the trend eventually reached statistical significance (P=0.037).54
Azacitidine (AZA) was studied in older patients (median age 75 years) with AML with > 30% blasts in a randomized phase III study.55 Azacitidine was associated with a trend towards better OS compared to conventional care regimens (CCR, standard induction chemotherapy, LDAC, or supportive care only) (10.4 months vs. 6.5 months, P=0.10).55 AZA was compared to CCR in another phase III trial, in patients with low bone marrow blast counts of 20–30% (median age 70 years).56 In this study, azacitidine was associated with improved OS (24.5 months vs. 16 months, P=0.05), as well as shorter fewer hospitalizations and number of days spent in the hospital.
Both decitabine and azacitidine modestly improve OS compared to BSC. Hypomethylating agents (HMAs) are also more tolerable than intensive induction.
Venetoclax-containing regimens
Based on two key trials, venetoclax, an oral B-cell lymphoma-2 (BCL-2) protein inhibitor, in combination with HMA or LDAC, received FDA approval in 2018 for treatment of older adults with de novo AML considered unfit for intensive chemotherapy.57 The trials that led to venetoclax’s FDA approval58,59 included patients 60–74 years with ECOG PS 0–3, ≥ 75 years with ECOG PS 0–2, or ≥ 60 years with predefined comorbidities (or comorbidities judged to be incompatible with intensive chemotherapy by the treating physician).
Venetoclax + HMA:
A phase Ib escalation trial for older patients (median age 74 years) ineligible for intensive chemotherapy evaluated venetoclax with a HMA (decitabine 20 mg/m2/day for 5 days or azacitidine 75 mg/m2/day for 7 days).58 Venetoclax combined with HMA was associated with 73% CR + CRi. CR + CRi rates were 60% in patients with poor-risk cytogenetics and 65% for those ≥ 75 years.58 An ongoing confirmatory phase III trial evaluating venetoclax with azacitidine compared to azacitidine alone in patients with AML who were ineligible for IC has recently reported having met the primary endpoints of improvement in OS and composite CR (complete and incomplete). 60
Venetoclax + LDAC:
A phase Ib/II study of venetoclax (600 mg daily) with LDAC in older adults (median age 74 years) demonstrated 54% CR + CRi, with a manageable safety profile.59 Venetoclax (600 mg daily) with LDAC was compared to LDAC alone in a phase III randomized, multicenter trial of patients with AML who were ineligible for IC (median age 76 years).61 At the pre-planned median follow-up time of 12 months, median OS was 7.2 months in the venetoclax/LDAC group and 4.1 months in LDAC alone, with a non-statistically significant difference (HR 0.75, P=0.11). The lack of significant difference in OS may be in part due to the higher rates of secondary AML and poorer cytogenetic risks noted in the group treated with venetoclax.61 Secondary endpoints included CR/CRi which was higher in the venetoclax arm compared to LDAC alone (48% and 13%, P<0.01).61
Prior to studies on venetoclax, HMA or LDAC alone had been the standard of care of patients not considered fit for intensive therapy.62 Venetoclax combination therapies have shown promising results as described above. 58,59 We will await the final results of the phase III venetoclax/azacitidine trial as well as the final analysis of the venetoclax/LDAC trial with longer follow-up. 63,64 There are several challenges when using venetoclax-based regimens include mitigation of tumor lysis syndrome, management of severe bone marrow suppression, and optimization of venetoclax dose when other CYP34-inhibiting drugs are utilized.65 Data from real-world experience are emerging66 and practical guidance are available.35
In conclusion, venetoclax with an HMA or LDAC can be considered for patients with AML who are not candidates for or decline intensive treatment.
Clinical Scenario 3 - Alternative treatment options:
Glasdegib + LDAC
Glasdegib, a hedgehog pathway inhibitor, was studied in a phase II randomized trial for older patients with AML or high-risk MDS (median age 76 years) unfit for intensive chemotherapy.67 Glasdegib combined with LDAC was associated with higher CR (17% vs. 2.3%, P<0.05) and improved OS (8.8 months vs. 4.9 months, P=0.0004) compared to LDAC alone, without significant toxicities.67 Based on these results, glasdegib received FDA approval for treatment of AML in older or unfit patients.68
GO
In patients with CD33-positive AML, GO monotherapy can be considered.3 A randomized phase III study in patients with AML not considered candidates for intensive therapy compared GO to BSC (median age 77 years).69 Patients who received GO (6 mg/m2 on day 1 and 3 mg/m2 on day 8) had an improved median OS of 4.9 months, compared to 3.6 months in the BSC group (HR 0.69, P=0.005) without additional toxicity.69
Ivosidenib and Enasidenib
In patients with IDH1/2 mutant AML, ivosidenib and enasidenib are being evaluated.33 Ivosidenib and enasidenib target IDH1 and IDH2 mutations respectively and were initially approved in the relapsed/refractory (R/R) setting.70,71
An ongoing phase 1 dose escalation study of ivosidenib has enrolled 34 older patients who were not candidates for intensive treatment (median age 77 years).72 ORR was defined as CR, CRi, partial response or morphologic remission. Patients who received ivosidenib had a CR of 30% and ORR 55%. Treatment was well-tolerated: grade 1–2 toxicities were most common (~25% of patients) and were predominantly gastrointestinal (GI) symptoms, fatigue and cytopenias. IDH differentiation syndrome occurred in 16% of patients.72 IDH differentiation syndrome is a rare syndrome theorized to occur after rapid neutrophil recovery; it presents with significant neutrophil-predominant leukocytosis and nonspecific symptoms such as fever, hypotension, and intra-thoracic effusions.72
Enasidenib was evaluated in a phase I/II trial of older patients (median age 77 years) with AML unfit for intensive treatment.73 ORR was 30.8% and CR was 18%. Toxicity included indirect hyperbilirubinemia, GI complaints and fatigue. Grade 3–4 cytopenias occurred in 21% of participants. Overall, enasidenib was found to be safe and well-tolerated.73
The NCCN recommends considering ivosidenib and enasidenib, respectively, for newly diagnosed patients with IDH1 and IDH2 mutations.3 Ivosidenib has received FDA approval in the upfront setting for patients with IDH1 mutant AML who are not suitable candidates for intensive therapy.74
In summary, in the absence of actionable mutations, options include LDAC (monotherapy, with glasdegib, or with venetoclax), HMA (monotherapy or with venetoclax), or best supportive care. In the presence of IDH1 mutation, IDH2 mutation, and CD33-positivitity, alternative options include ivosidenib, enasidenib and GO, respectively.
Post-remission therapy
Relapse rates for older adults with AML who achieve CR are high which contributes to poor overall survival.75 More aggressive disease biology contribute to higher relapse rates in older compared to younger patients.5,76 Post-remission therapy options include allogeneic hematopoietic stem cell transplant (HSCT) and/or conventional chemotherapy (e.g., high dose cytarabine); both were more commonly utilized after intensive induction therapies. Patient- and disease-related factors must be re-evaluated at the time of decision-making for consolidation regimens. Some patients who receive intensive induction may experience significant treatment toxicity precluding further intensive treatment.
Allogeneic HSCT is recommended for older adults with poor-risk AML in first remission who are deemed appropriate candidates for HSCT.3 Historically, treatment toxicity has been considered prohibitive to the use of allogeneic HSCT in older patients.77 However, advances in HSCT such as the use of reduced-intensity conditioning regimens have been developed to decrease toxicities. Devine et al conducted a study of 114 patients (median age 65 years) who received transplantation following reduced-intensity conditioning. Survival was compared to historical cohorts treated without HSCT. OS was improved (48%, P<0.05) and complications including non-relapse mortality (15% at 2 years) were lower than expected.77
For patients who are not candidates for allogeneic HSCT, chemotherapy should be considered. Various regimens are used in clinical practice and are recommended by the NCCN which may include intermediate dose-cytarabine (1.0–1.5 g/m2/day for 4–6 days for 1–2 cycles). Addition of targeted therapies such as GO and midostaurin is recommended for CD-33 positivity and FLT-3 mutations, respectively. For those with secondary and therapy-related AML, consolidation with CPX-351 should be considered if used for induction.3
Health-Related Quality of Life and Hospitalization
Historically, overall survival has been the most measured outcome in AML trials. More recently, interest in patient-centered outcomes such as HRQoL, has increased markedly. HRQoL incorporates physical, mental, emotional, and social functioning domains; however, it is often difficult to quantify since there are no universally accepted measures of HRQoL. We recently provided a summary of different HRQoL tools that have been incorporated in trials of patients with AML.15 HRQoL assessments provide critical information that can influence decision-making. In the context of the multiple newly approved treatments, many with otherwise comparable efficacy and tolerability, HRQoL may be the differentiating factor.
Patients with AML have significant morbidity, often requiring inpatient admissions.78 Common causes for admission include urgent evaluation following a new diagnosis, managing acute illness, induction chemotherapy, complications from outpatient therapies, and supportive care. Based on a national inpatient sample database, patients ≥ 60 years with AML often have shorter lengths of stay (LOS) than those < 60 years (5.4 vs. 6.8 days). Factors associated with shorter LOS in patients ≥ 60 years include older age, higher income, and admission to nonteaching hospitals.78 The lower length of stay in older adults may be due to lower utilization of AML-directed therapy and higher in-hospital mortality.78 Understanding factors associated with hospitalization and length of stay can help with treatment decision-making as the ability to stay at home and spend time with family may be important to older adults.79
Supportive care: Growth Factors
Neutropenia is a common cause of significant morbidity in patients with AML. It is multifactorial, due to the disease itself and the myelosuppressive treatments, particularly traditional “7+3” induction and venetoclax.65 Myeloid growth factors such as granulocyte-colony stimulating factor (G-CSF) have been extensively studied in patients with hematologic malignancies.7 Their benefits include accelerated neutrophil count recovery, decreased duration of fever, and decreased length of hospital stay. However, when studied in older patients with AML, G-CSF did not portend survival benefits.7,80,81 There are also theoretical concerns about stimulation of leukemic cell growth and cost-effectiveness.7 The routine use of G-CSF is not recommended; however, G-CSF is sometimes used during consolidation therapy when remission has been achieved.
Supportive care: Exercise, Cognitive Behavioral Therapy, and Palliative Care
Patients with cancer are at high risk of functional and cognitive declines due to the disease itself and its treatment.82 Older patients are most at risk for these consequences, and lower levels of physical activity have been associated with more deconditioning in this patient population. Exercise helps with symptom control, fatigue, and quality of life during and after treatment.83,84 In a systematic review of exercise in adults with hematologic malignancies, aerobic exercise was associated with positive effects on mood and fatigue.85 A recent trial of a supervised inpatient exercise program was conducted in patients with AML admitted for induction (median age 53 years).84 This 4-week mixed-modality program resulted in improved patient-reported fatigue and improved physical function compared to placebo. Despite the small sample size (n=17), results of this study are consistent with previously reported studies in patients with leukemia.86,87 Increased awareness about and promotion of the benefits of exercise among providers are important as patients are more likely to exercise if recommended by their oncologist.83 Randomized controlled trials are needed to determine the appropriate type, timing and “dose” of exercise.
Symptoms of cancer-related cognitive decline have been shown to improve with cognitive behavioral therapy (CBT).82 An RCT is currently being conducted in adults with AML and lymphoma to evaluate the role of CBT in mitigating cancer-related fatigue, an almost universally reported symptom.88
Given the poor outcomes associated with AML in the older adult population, incorporation of palliative care services should be considered.89 The integration of palliative care with typical oncology care has recently been shown to be beneficial in a multi-site randomized control trial of patients > 60 years with AML undergoing IC.90 The palliative care intervention group included at least twice weekly visits by a palliative care provider during each hospitalization. Patients were found to have improved QOL as well as psychological distress. Intervention effects were significant at 2 weeks and sustained up to week 24.90 The integration of palliative care may help patients mitigate the many challenges they face related to symptoms and psychological distress throughout their disease.
Conclusion
In an aging population, the prevalence of AML in older adults is rising. Although treatment disparities are decreasing in patients 60–80 years, studies are still lacking in patients > 80 years. Treatment of AML in older adults is complex. Whenever possible, patients should be included in clinical trials,91 which will improve our understanding of AML therapies. The advent of multiple novel agents is promising, yet oncologists/hematologists now face new challenges. These include how best to assess fitness and anticipate risks and benefits of treatment. Comprehensive assessments such as GA predict functional reserve and assess overall health more accurately than chronological age alone. Providers and patients are also discussing and considering broader definitions of “treatment benefit” with a focus on measures beyond longevity, such as HRQoL and length of hospitalization. Personalizing both therapeutic and non-therapeutic treatments is critical given the heterogeneity of AML and older patients.
Supplementary Material
Key References | Brief Description |
---|---|
** Ref 8: Klepin et al.8 | Discusses the changing treatment landscape of AML in older adults and associated issues |
* Ref 16: Klepin et al16 | Predictive value of GA in older adults with AML |
** Ref 18: Loh and Klepin18 | Role of GA in personalized care of older adults with AML and discussion of future directions |
* Ref 35: DiNardo et al35 | Review of treating AML in the era of novel agents, not specific to older adults |
** Ref 53: Lancet et al53 | Trial demonstrating efficacy of CPX in sAML |
** Ref 58 and 59: DiNardo et al58, Wei et al59 | Trials that led to Venetoclax-based therapy becoming approved for older patients and those who are not candidates for intensive induction |
*Ref 72 and 73: Roboz et al72, Pollyea et al73 | Key trials of ivosedinib and enasidenib for use in newly diagnosed IDH1/2 mutant AML |
Acknowledgement
We would like to acknowledge Dr. Susan Rosenthal, MD for her editorial assistance.
Funding
Dr. Loh is supported by the National Cancer Institute at the National Institute of Health (K99CA237744) and Wilmot Research Fellowship Award.
Footnotes
Conflicts of interest/Competing interests
Dr. Loh serves as a consultant to Pfizer and Seattle Genetics.
References
- 1.PDF Version - SEER Cancer Statistics Review (CSR), 1975–2016. SEER Accessed January 18, 2020. https://seer.cancer.gov/csr/1975_2016/sections.html
- 2.Oran B, Weisdorf DJ. Survival for older patients with acute myeloid leukemia: a population-based study. Haematologica. 2012;97(12):1916–1924. doi: 10.3324/haematol.2012.066100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Tallman MS, Wang ES, Altman JK, et al. Acute Myeloid Leukemia, Version 3.2019, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Cancer Netw JNCCN. 2019;17(6):721–749. doi: 10.6004/jnccn.2019.0028 [DOI] [PubMed] [Google Scholar]
- 4.Döhner H, Estey E, Grimwade D, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129(4):424–447. doi: 10.1182/blood-2016-08-733196 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Appelbaum FR, Gundacker H, Head DR, et al. Age and acute myeloid leukemia. Blood. 2006;107(9):3481–3485. doi: 10.1182/blood-2005-09-3724 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Oliai C, Schiller G. How to address second and therapy-related acute myelogenous leukaemia. Br J Haematol. 2020;188(1):116–128. doi: 10.1111/bjh.16354 [DOI] [PubMed] [Google Scholar]
- 7.Döhner H, Estey EH, Amadori S, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115(3):453–474. doi: 10.1182/blood-2009-07-235358 [DOI] [PubMed] [Google Scholar]
- 8.Klepin HD, Estey E, Kadia T. More Versus Less Therapy for Older Adults With Acute Myeloid Leukemia: New Perspectives on an Old Debate. Am Soc Clin Oncol Educ Book Am Soc Clin Oncol Annu Meet. 2019;39:421–432. doi: 10.1200/EDBK_239097 [DOI] [PubMed] [Google Scholar]
- 9.Juliusson G, Antunovic P, Derolf A, et al. Age and acute myeloid leukemia: real world data on decision to treat and outcomes from the Swedish Acute Leukemia Registry. Blood. 2009;113(18):4179–4187. doi: 10.1182/blood-2008-07-172007 [DOI] [PubMed] [Google Scholar]
- 10.Löwenberg B, Zittoun R, Kerkhofs H, et al. 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 Off J Am Soc Clin Oncol. 1989;7(9):1268–1274. doi: 10.1200/JCO.1989.7.9.1268 [DOI] [PubMed] [Google Scholar]
- 11.Tariman JD, Doorenbos A, Schepp KG, Becker PS, Berry DL. Patient, Physician and Contextual Factors Are Influential in the Treatment Decision Making of Older Adults Newly Diagnosed with Symptomatic Myeloma. Cancer Treat Commun. 2014;2(2–3):34–47. doi: 10.1016/j.ctrc.2014.08.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Mohile SG, Magnuson A, Pandya C, et al. Community Oncologists’ Decision-Making for Treatment of Older Patients With Cancer. J Natl Compr Cancer Netw JNCCN. 2018;16(3):301–309. doi: 10.6004/jnccn.2017.7047 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Abel GA, Klepin HD. Frailty and the management of hematologic malignancies. Blood. 2018;131(5):515–524. doi: 10.1182/blood-2017-09-746420 [DOI] [PubMed] [Google Scholar]
- 14.The Paez A. “architect analogy” of evidence-based practice: Reconsidering the role of clinical expertise and clinician experience in evidence-based health care. J Evid-Based Med. 2018;11(4):219–226. doi: 10.1111/jebm.12321 [DOI] [PubMed] [Google Scholar]
- 15.Loh KP, Abdallah M, Kumar AJ, Neuendorff NR, Dahiya S, Klepin HD. Health-Related Quality of Life and Treatment of Older Adults with Acute Myeloid Leukemia: a Young International Society of Geriatric Oncology Review Paper. Curr Hematol Malig Rep. 2019;14(6):523–535. doi: 10.1007/s11899-019-00552-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Klepin HD, Geiger AM, Tooze JA, et al. Geriatric assessment predicts survival for older adults receiving induction chemotherapy for acute myelogenous leukemia. Blood. 2013;121(21):4287–4294. doi: 10.1182/blood-2012-12-471680 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Sherman AE, Motyckova G, Fega KR, et al. Geriatric assessment in older patients with acute myeloid leukemia: a retrospective study of associated treatment and outcomes. Leuk Res. 2013;37(9):998–1003. doi: 10.1016/j.leukres.2013.05.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Loh KP, Klepin HD. Geriatric assessment in acute myeloid leukemia: current and future landscape. Blood Adv. 2018;2(18):2418. doi: 10.1182/bloodadvances.2018016477 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Mohile SG, Dale W, Somerfield MR, et al. Practical Assessment and Management of Vulnerabilities in Older Patients Receiving Chemotherapy: ASCO Guideline for Geriatric Oncology. J Clin Oncol Off J Am Soc Clin Oncol. 2018;36(22):2326–2347. doi: 10.1200/JCO.2018.78.8687 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Extermann M, Hurria A. Comprehensive geriatric assessment for older patients with cancer. J Clin Oncol Off J Am Soc Clin Oncol. 2007;25(14):1824–1831. doi: 10.1200/JCO.2007.10.6559 [DOI] [PubMed] [Google Scholar]
- 21.Loh KP, Soto-Perez-de-Celis E, Hsu T, et al. What Every Oncologist Should Know About Geriatric Assessment for Older Patients With Cancer: Young International Society of Geriatric Oncology Position Paper. J Oncol Pract. 2018;14(2):85–94. doi: 10.1200/JOP.2017.026435 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Wildiers H, Heeren P, Puts M, et al. International Society of Geriatric Oncology consensus on geriatric assessment in older patients with cancer. J Clin Oncol Off J Am Soc Clin Oncol. 2014;32(24):2595–2603. doi: 10.1200/JCO.2013.54.8347 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Mohile SG, Velarde C, Hurria A, et al. Geriatric Assessment-Guided Care Processes for Older Adults: A Delphi Consensus of Geriatric Oncology Experts. J Natl Compr Cancer Netw JNCCN. 2015;13(9):1120–1130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Gbadamosi B, Ezekwudo D, Bastola S, Jaiyesimi I. Predictive and Prognostic Markers in Adults With Acute Myeloid Leukemia: A Single-Institution Experience. Clin Lymphoma Myeloma Leuk. 2018;18(7):e287–e294. doi: 10.1016/j.clml.2018.05.005 [DOI] [PubMed] [Google Scholar]
- 25.Kumar CC. Genetic Abnormalities and Challenges in the Treatment of Acute Myeloid Leukemia. Genes Cancer. 2011;2(2):95–107. doi: 10.1177/1947601911408076 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Arber DA, Borowitz MJ, Cessna M, et al. Initial Diagnostic Workup of Acute Leukemia: Guideline From the College of American Pathologists and the American Society of Hematology. Arch Pathol Lab Med. 2017;141(10):1342–1393. doi: 10.5858/arpa.2016-0504-CP [DOI] [PubMed] [Google Scholar]
- 27.Leisch M, Jansko B, Zaborsky N, Greil R, Pleyer L. Next Generation Sequencing in AML—On the Way to Becoming a New Standard for Treatment Initiation and/or Modulation? Cancers. 2019;11(2). doi: 10.3390/cancers11020252 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127(20):2391–2405. doi: 10.1182/blood-2016-03-643544 [DOI] [PubMed] [Google Scholar]
- 29.Prassek VV, Rothenberg-Thurley M, Sauerland MC, et al. Genetics of acute myeloid leukemia in the elderly: mutation spectrum and clinical impact in intensively treated patients aged 75 years or older. Haematologica. 2018;103(11):1853–1861. doi: 10.3324/haematol.2018.191536 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Mrózek K, Marcucci G, Nicolet D, et al. Prognostic significance of the European LeukemiaNet standardized system for reporting cytogenetic and molecular alterations in adults with acute myeloid leukemia. J Clin Oncol Off J Am Soc Clin Oncol. 2012;30(36):4515–4523. doi: 10.1200/JCO.2012.43.4738 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Metzeler KH, Herold T, Rothenberg-Thurley M, et al. Spectrum and prognostic relevance of driver gene mutations in acute myeloid leukemia. Blood. 2016;128(5):686–698. doi: 10.1182/blood-2016-01-693879 [DOI] [PubMed] [Google Scholar]
- 32.Schneider F, Hoster E, Schneider S, et al. Age-dependent frequencies of NPM1 mutations and FLT3-ITD in patients with normal karyotype AML (NK-AML). Ann Hematol. 2012;91(1):9–18. doi: 10.1007/s00277-011-1280-6 [DOI] [PubMed] [Google Scholar]
- 33.DiNardo CD, Ravandi F, Agresta S, et al. Characteristics, clinical outcome, and prognostic significance of IDH mutations in AML. Am J Hematol. 2015;90(8):732–736. doi: 10.1002/ajh.24072 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Lai C, Doucette K, Norsworthy K. Recent drug approvals for acute myeloid leukemia. J Hematol OncolJ Hematol Oncol. 2019;12. doi: 10.1186/s13045-019-0774-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.DiNardo CD, Wei AH. How I treat acute myeloid leukemia in the era of new drugs. Blood. 2020;135(2):85–96. doi: 10.1182/blood.2019001239 [DOI] [PubMed] [Google Scholar]
- 36.Soto-Perez-de-Celis E, Li D, Yuan Y, Lau YM, Hurria A. Functional versus chronological age: geriatric assessments to guide decision making in older patients with cancer. Lancet Oncol. 2018;19(6):e305–e316. doi: 10.1016/S1470-2045(18)30348-6 [DOI] [PubMed] [Google Scholar]
- 37.Burnett AK, Russell NH, Hills RK, et al. A randomized comparison of daunorubicin 90 mg/m2 vs 60 mg/m2 in AML induction: results from the UK NCRI AML17 trial in 1206 patients. Blood. 2015;125(25):3878–3885. doi: 10.1182/blood-2015-01-623447 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Löwenberg B, Ossenkoppele GJ, van Putten W, et al. High-dose daunorubicin in older patients with acute myeloid leukemia. N Engl J Med. 2009;361(13):1235–1248. doi: 10.1056/NEJMoa0901409 [DOI] [PubMed] [Google Scholar]
- 39.Pautas C, Merabet F, Thomas X, et al. Randomized study of intensified anthracycline doses for induction and recombinant interleukin-2 for maintenance in patients with acute myeloid leukemia age 50 to 70 years: results of the ALFA-9801 study. J Clin Oncol Off J Am Soc Clin Oncol. 2010;28(5):808–814. doi: 10.1200/JCO.2009.23.2652 [DOI] [PubMed] [Google Scholar]
- 40.Castaigne S, Pautas C, Terré C, et al. 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 Lond Engl. 2012;379(9825):1508–1516. doi: 10.1016/S0140-6736(12)60485-1 [DOI] [PubMed] [Google Scholar]
- 41.Lambert J, Pautas C, Terré C, et al. Gemtuzumab ozogamicin for de novo acute myeloid leukemia: final efficacy and safety updates from the open-label, phase III ALFA-0701 trial. Haematologica. 2019;104(1):113–119. doi: 10.3324/haematol.2018.188888 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Burnett AK, Russell NH, Hills RK, et al. Addition of Gemtuzumab Ozogamicin to Induction Chemotherapy Improves Survival in Older Patients With Acute Myeloid Leukemia. J Clin Oncol. 2012;30(32):3924–3931. doi: 10.1200/JCO.2012.42.2964 [DOI] [PubMed] [Google Scholar]
- 43.Amadori S, Suciu S, Stasi R, et al. Sequential combination of gemtuzumab ozogamicin and standard chemotherapy in older patients with newly diagnosed acute myeloid leukemia: results of a randomized phase III trial by the EORTC and GIMEMA consortium (AML-17). J Clin Oncol Off J Am Soc Clin Oncol. 2013;31(35):4424–4430. doi: 10.1200/JCO.2013.49.0771 [DOI] [PubMed] [Google Scholar]
- 44.Petersdorf S, Kopecky K, Stuart RK, et al. Preliminary Results of Southwest Oncology Group Study S0106: An International Intergroup Phase 3 Randomized Trial Comparing the Addition of Gemtuzumab Ozogamicin to Standard Induction Therapy Versus Standard Induction Therapy Followed by a Second Randomization to Post-Consolidation Gemtuzumab Ozogamicin Versus No Additional Therapy for Previously Untreated Acute Myeloid Leukemia. Blood. 2009;114(22):790–790. doi: 10.1182/blood.V114.22.790.790 [DOI] [Google Scholar]
- 45.Burnett AK, Hills RK, Milligan D, et al. Identification of patients with acute myeloblastic leukemia who benefit from the addition of gemtuzumab ozogamicin: results of the MRC AML15 trial. J Clin Oncol Off J Am Soc Clin Oncol. 2011;29(4):369–377. doi: 10.1200/JCO.2010.31.4310 [DOI] [PubMed] [Google Scholar]
- 46.Delaunay J, Recher C, Pigneux A, et al. Addition of Gemtuzumab Ozogamycin to Chemotherapy Improves Event-Free Survival but Not Overall Survival of AML Patients with Intermediate Cytogenetics Not Eligible for Allogeneic Transplantation. Results of the GOELAMS AML 2006 IR Study. Blood. 2011;118(21):79–79. doi: 10.1182/blood.V118.21.79.79 [DOI] [Google Scholar]
- 47.Burnett AK, Hills RK, Hunter AE, et al. The addition of gemtuzumab ozogamicin to low-dose Ara-C improves remission rate but does not significantly prolong survival in older patients with acute myeloid leukaemia: results from the LRF AML14 and NCRI AML16 pick-a-winner comparison. Leukemia. 2013;27(1):75–81. doi: 10.1038/leu.2012.229 [DOI] [PubMed] [Google Scholar]
- 48.Efficacy Takeshita A. and resistance of gemtuzumab ozogamicin for acute myeloid leukemia. Int J Hematol. 2013;97(6):703–716. doi: 10.1007/s12185-013-1365-1 [DOI] [PubMed] [Google Scholar]
- 49.Stone RM, Mandrekar SJ, Sanford BL, et al. Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a FLT3 Mutation. N Engl J Med. 2017;377(5):454–464. doi: 10.1056/NEJMoa1614359 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Research C for DE and. Midostaurin. FDA. Published online February 9, 2019. Accessed January 21, 2020. http://www.fda.gov/drugs/resources-information-approved-drugs/midostaurin
- 51.Commissioner O of the. FDA approves first treatment for certain types of poor-prognosis acute myeloid leukemia. FDA. Published September 10, 2019. Accessed January 21, 2020. http://www.fda.gov/news-events/press-announcements/fda-approves-first-treatment-certain-types-poor-prognosis-acute-myeloid-leukemia
- 52.Lancet JE, Cortes JE, Hogge DE, et al. 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. 2014;123(21):3239–3246. doi: 10.1182/blood-2013-12-540971 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Lancet JE, Uy GL, Cortes JE, et al. CPX-351 (cytarabine and daunorubicin) Liposome for Injection Versus Conventional Cytarabine Plus Daunorubicin in Older Patients With Newly Diagnosed Secondary Acute Myeloid Leukemia. J Clin Oncol Off J Am Soc Clin Oncol. 2018;36(26):2684–2692. doi: 10.1200/JCO.2017.77.6112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kantarjian HM, Thomas XG, Dmoszynska A, et al. 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 Off J Am Soc Clin Oncol. 2012;30(21):2670–2677. doi: 10.1200/JCO.2011.38.9429 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Dombret H, Seymour JF, Butrym A, et al. International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts. Blood. 2015;126(3):291–299. doi: 10.1182/blood-2015-01-621664 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Fenaux P, Mufti GJ, Hellström-Lindberg E, et al. Azacitidine prolongs overall survival compared with conventional care regimens in elderly patients with low bone marrow blast count acute myeloid leukemia. J Clin Oncol Off J Am Soc Clin Oncol. 2010;28(4):562–569. doi: 10.1200/JCO.2009.23.8329 [DOI] [PubMed] [Google Scholar]
- 57.Research C for DE and. FDA approves venetoclax in combination for AML in adults. FDA. Published online December 20, 2019. Accessed January 21, 2020. http://www.fda.gov/drugs/fda-approves-venetoclax-combination-aml-adults
- 58.DiNardo CD, Pratz K, Pullarkat V, et al. Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood. 2019;133(1):7–17. doi: 10.1182/blood-2018-08-868752 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Wei AH, Strickland SA, Hou J-Z, et al. Venetoclax Combined With Low-Dose Cytarabine for Previously Untreated Patients With Acute Myeloid Leukemia: Results From a Phase Ib/II Study. J Clin Oncol Off J Am Soc Clin Oncol. 2019;37(15):1277–1284. doi: 10.1200/JCO.18.01600 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.AbbVie Announces Positive Topline Results from Phase 3 Trial of VENCLEXTA® (venetoclax) in Combination with Azacitidine in Patients with Acute Myeloid Leukemia (AML) | AbbVie News Center. Accessed May 18, 2020. https://news.abbvie.com/news/press-releases/abbvie-announces-positive-topline-results-from-phase-3-trial-venclexta-venetoclax-in-combination-with-azacitidine-in-patients-with-acute-myeloid-leukemia-aml.htm
- 61.Wei AH, Montesinos P, Ivanov V, et al. Venetoclax plus LDAC for patients with untreated AML ineligible for intensive chemotherapy: phase 3 randomized placebo-controlled trial. Blood. doi: 10.1182/blood.2020004856 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Pollyea DA, Amaya M, Strati P, Konopleva MY. Venetoclax for AML: changing the treatment paradigm. Blood Adv. 2019;3(24):4326–4335. doi: 10.1182/bloodadvances.2019000937 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.A Study of Venetoclax in Combination With Low Dose Cytarabine Versus Low Dose Cytarabine Alone in Treatment Naive Patients With Acute Myeloid Leukemia Who Are Ineligible for Intensive Chemotherapy - Full Text View - ClinicalTrials.gov. Accessed February 12, 2020. https://clinicaltrials.gov/ct2/show/NCT03069352
- 64.A Study of Venetoclax in Combination With Azacitidine Versus Azacitidine in Treatment Naïve Subjects With Acute Myeloid Leukemia Who Are Ineligible for Standard Induction Therapy - Full Text View - ClinicalTrials.gov. Accessed February 12, 2020. https://clinicaltrials.gov/ct2/show/NCT02993523
- 65.Jonas BA, Pollyea DA. How we use venetoclax with hypomethylating agents for the treatment of newly diagnosed patients with acute myeloid leukemia. Leukemia. Published online October 18, 2019:1–10. doi: 10.1038/s41375-019-0612-8 [DOI] [PubMed] [Google Scholar]
- 66.Winters AC, Gutman JA, Purev E, et al. Real-world experience of venetoclax with azacitidine for untreated patients with acute myeloid leukemia. Blood Adv. 2019;3(20):2911–2919. doi: 10.1182/bloodadvances.2019000243 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Cortes JE, Heidel FH, Hellmann A, et al. Randomized comparison of low dose cytarabine with or without glasdegib in patients with newly diagnosed acute myeloid leukemia or high-risk myelodysplastic syndrome. Leukemia. 2019;33(2):379–389. doi: 10.1038/s41375-018-0312-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Norsworthy KJ, By K, Subramaniam S, et al. FDA Approval Summary: Glasdegib for Newly Diagnosed Acute Myeloid Leukemia. Clin Cancer Res. Published online May 7, 2019. doi: 10.1158/1078-0432.CCR-19-0365 [DOI] [PubMed] [Google Scholar]
- 69.Amadori S, Suciu S, Selleslag D, et al. Gemtuzumab Ozogamicin Versus Best Supportive Care in Older Patients With Newly Diagnosed Acute Myeloid Leukemia Unsuitable for Intensive Chemotherapy: Results of the Randomized Phase III EORTC-GIMEMA AML-19 Trial. J Clin Oncol Off J Am Soc Clin Oncol. 2016;34(9):972–979. doi: 10.1200/JCO.2015.64.0060 [DOI] [PubMed] [Google Scholar]
- 70.Research C for DE and. FDA approves ivosidenib for relapsed or refractory acute myeloid leukemia. FDA. Published online February 9, 2019. Accessed February 10, 2020. http://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-ivosidenib-relapsed-or-refractory-acute-myeloid-leukemia
- 71.Research C for DE and. FDA granted regular approval to enasidenib for the treatment of relapsed or refractory AML. FDA. Published online February 9, 2019. Accessed February 10, 2020. http://www.fda.gov/drugs/resources-information-approved-drugs/fda-granted-regular-approval-enasidenib-treatment-relapsed-or-refractory-aml
- 72.Roboz GJ, DiNardo CD, Stein EM, et al. Ivosidenib induces deep durable remissions in patients with newly diagnosed IDH1-mutant acute myeloid leukemia. Blood. Published online December 16, 2019. doi: 10.1182/blood.2019002140 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Pollyea DA, Tallman MS, de Botton S, et al. Enasidenib, an inhibitor of mutant IDH2 proteins, induces durable remissions in older patients with newly diagnosed acute myeloid leukemia. Leukemia. 2019;33(11):2575–2584. doi: 10.1038/s41375-019-0472-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Research C for DE and. FDA approves ivosidenib as first-line treatment for AML with IDH1 mutation. FDA. Published online December 20, 2019. Accessed February 10, 2020. http://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-ivosidenib-first-line-treatment-aml-idh1-mutation
- 75.Kantarjian H, O’brien S, Cortes J, et al. 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. 2006;106(5):1090–1098. doi: 10.1002/cncr.21723 [DOI] [PubMed] [Google Scholar]
- 76.Burnett A, Wetzler M, Löwenberg B. Therapeutic Advances in Acute Myeloid Leukemia. J Clin Oncol. 2011;29(5):487–494. doi: 10.1200/JCO.2010.30.1820 [DOI] [PubMed] [Google Scholar]
- 77.Devine SM, Owzar K, Blum W, et al. Phase II Study of Allogeneic Transplantation for Older Patients With Acute Myeloid Leukemia in First Complete Remission Using a Reduced-Intensity Conditioning Regimen: Results From Cancer and Leukemia Group B 100103 (Alliance for Clinical Trials in Oncology)/Blood and Marrow Transplant Clinical Trial Network 0502. J Clin Oncol Off J Am Soc Clin Oncol. 2015;33(35):4167–4175. doi: 10.1200/JCO.2015.62.7273 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Kumar AJ, Henzer T, Rodday AM, Parsons SK. Risk factors for length of stay and charge per day differ between older and younger hospitalized patients with AML. Cancer Med. 2018;7(6):2744–2752. doi: 10.1002/cam4.1492 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Crawford R, Sully K, Conroy R, et al. Patient-Centered Insights on Treatment Decision Making and Living with Acute Myeloid Leukemia and Other Hematologic Cancers. Patient - Patient-Centered Outcomes Res. 2020;13(1):83–102. doi: 10.1007/s40271-019-00384-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Wheatley K, Goldstone AH, Littlewood T, Hunter A, Burnett AK. Randomized placebo-controlled trial of granulocyte colony stimulating factor (G-CSF) as supportive care after induction chemotherapy in adult patients with acute myeloid leukaemia: a study of the United Kingdom Medical Research Council Adult Leukaemia Working Party. Br J Haematol. 2009;146(1):54–63. doi: 10.1111/j.1365-2141.2009.07710.x [DOI] [PubMed] [Google Scholar]
- 81.Amadori S, Suciu S, Jehn U, et al. Use of glycosylated recombinant human G-CSF (lenograstim) during and/or after induction chemotherapy in patients 61 years of age and older with acute myeloid leukemia: final results of AML-13, a randomized phase-3 study. Blood. 2005;106(1):27–34. doi: 10.1182/blood-2004-09-3728 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Pergolotti M, Battisti NML, Padgett L, et al. Embracing the complexity: Older adults with cancer-related cognitive decline-A Young International Society of Geriatric Oncology Position Paper. J Geriatr Oncol. Published online October 13, 2019. doi: 10.1016/j.jgo.2019.09.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Loh KP, Lin P-J, Uth J, Quist M, Klepin H, Mustian K. Exercise for managing cancer- and treatment-related side effects in older adults. J Geriatr Oncol. 2018;9(4):405–410. doi: 10.1016/j.jgo.2018.03.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Bryant AL, Deal AM, Battaglini CL, et al. The Effects of Exercise on Patient-Reported Outcomes and Performance-Based Physical Function in Adults With Acute Leukemia Undergoing Induction Therapy: Exercise and Quality of Life in Acute Leukemia (EQUAL). Integr Cancer Ther. 2018;17(2):263–270. doi: 10.1177/1534735417699881 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Knips L, Bergenthal N, Streckmann F, Monsef I, Elter T, Skoetz N. Aerobic physical exercise for adult patients with haematological malignancies. Cochrane Database Syst Rev. 2019;(1). doi: 10.1002/14651858.CD009075.pub3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Klepin HD, Danhauer SC, Tooze JA, et al. Exercise for older adult inpatients with acute myelogenous leukemia: A pilot study. J Geriatr Oncol. 2011;2(1):11–17. doi: 10.1016/j.jgo.2010.10.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Alibhai SMH, O’Neill S, Fisher-Schlombs K, et al. A clinical trial of supervised exercise for adult inpatients with acute myeloid leukemia (AML) undergoing induction chemotherapy. Leuk Res. 2012;36(10):1255–1261. doi: 10.1016/j.leukres.2012.05.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Cognitive Behavioral Therapy in Helping Patients With Acute Myeloid Leukemia or Lymphoma With Cancer-Related Fatigue - Full Text View - ClinicalTrials.gov. Accessed January 16, 2020. https://clinicaltrials.gov/ct2/show/NCT03747757
- 89.Boucher NA, Johnson KS, LeBlanc TW. Acute Leukemia Patients’ Needs: Qualitative Findings and Opportunities for Early Palliative Care. J Pain Symptom Manage. 2018;55(2):433–439. doi: 10.1016/j.jpainsymman.2017.09.014 [DOI] [PubMed] [Google Scholar]
- 90.Areej El-Jawahri TWL, Connor SML, Massachusetts General Hospital B, et al. Multisite randomized trial of integrated palliative and oncology care for patients with acute myeloid leukemia (AML). Accessed May 18, 2020. https://meetinglibrary.asco.org/record/186130/abstract
- 91.Ossenkoppele G, Löwenberg B. How I treat the older patient with acute myeloid leukemia. Blood. 2015;125(5):767–774. doi: 10.1182/blood-2014-08-551499 [DOI] [PubMed] [Google Scholar]
- 92.Döhner H, Weisdorf DJ, Bloomfield CD. Acute Myeloid Leukemia. N Engl J Med. 2015;373(12):1136–1152. doi: 10.1056/NEJMra1406184 [DOI] [PubMed] [Google Scholar]
- 93.Haferlach T, Stengel A, Eckstein S, et al. The new provisional WHO entity ‘ RUNX1 mutated AML’ shows specific genetics but no prognostic influence of dysplasia. Leukemia. 2016;30(10):2109–2112. doi: 10.1038/leu.2016.150 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Barbosa K, Li S, Adams PD, Deshpande AJ. The role of TP53 in acute myeloid leukemia: Challenges and opportunities. Genes Chromosomes Cancer. 2019;58(12):875–888. doi: 10.1002/gcc.22796 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Burnett AK, Milligan D, Prentice AG, et al. A comparison of low-dose cytarabine and hydroxyurea with or without all-trans retinoic acid for acute myeloid leukemia and high-risk myelodysplastic syndrome in patients not considered fit for intensive treatment. Cancer. 2007;109(6):1114–1124. doi: 10.1002/cncr.22496 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.