Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

from the Experts

How I Treat Older Patients With Acute Myeloid Leukemia

Martha Arellano, MD
INTRODUCTION treatment, there is clearly a need for risk- Department of Hematology and
Medical Oncology, Winship Cancer
adapted treatment strategies for elderly
Acute myeloid leukemia (AML) is a clonal Institute, Emory University,
patients with AML. We herein discuss 3 cases Atlanta, Georgia
malignant proliferation of myeloid blast cells
that illustrate some of the complexities in
affecting primarily older adults, with a median
treatment planning, review the literature, and
age of onset of 68 years. The National Cancer Jennifer Wilkinson
highlight new treatments for older patients
Institute’s Surveillance, Epidemiology, and Carlisle, MD
with AML. Hematology and Medical Oncology
End Results program estimates 21,380 new
Fellowship Training Program,
AML cases will have been diagnosed and Emory University, Atlanta, Georgia
10,590 AML-related deaths will have occurred
Clinical Cases
in the United States in 2017.1 The main risk Case 1
factor for the development of AML is increas- An 81-year-old avid tennis player developed
ing age, and although there rarely are other rapidly progressive dyspnea on exertion and
identifiable predisposing factors, there is an fatigue. His complete blood count revealed
increasing number of AML cases after pancytopenia prompting bone marrow biopsy
exposure to chemotherapy or radiotherapy and aspiration. His bone marrow was 90% cel-
(collectively referred to as “therapy-related lular with 40% myeloid blasts, dysplastic
AML” [t-AML]). There also is a distinct high- changes, and complex cytogenetics (5q-, 18,
risk category of secondary AML (s-AML) after 111q25, 122p11, and t[1;22]). His past medi-
antecedent hematological disorders, such as cal history was significant for prostate cancer
myelodysplastic syndromes (MDS) or myelo- diagnosed 6 years prior, which was treated
proliferative neoplasms.2 Outcomes of AML in with radioactive seed implants; at presentation,
older adults (aged >60 years) are dismal, with the patient had an undetectable prostate-
survival rates reported to be <10% at 3 years specific antigen level. He enrolled on a single-
and <5% at 5 years.3,4 Therapeutic challenges arm phase 2 trial of azacitidine and a histone
include both a more aggressive biology of the deacetylase inhibitor. After 3 cycles, the patient
disease as well as a patient that is more was taken off study after an episode of pneu-
susceptible to the toxic effects of chemother- monia and atrial fibrillation. He was switched to
apy. Disappointingly, only 40% of patients decitabine and achieved complete remission
with AML who are aged >65 years are offered (CR) after 2 months. At the time of last follow-
chemotherapy within 3 months of diagnosis. up, the patient had completed 34 cycles of dec-
Moreover, elderly patients with AML who itabine and continued to play tennis.
receive treatment have a 33% lower risk of
AML-related death compared with their Case 2
untreated counterparts.5 Compared with A 71-year-old man was referred with a new
untreated elderly patients with AML, those diagnosis of AML after presenting with pancy-
who are offered treatment tend to be younger, topenia. He had low-risk prostate cancer diag-
are less likely to have s-AML, and are more nosed 3 years previously that was treated with
likely to have a good performance status (PS). radiation. His bone marrow was 10% to 20%
Furthermore, allogeneic hematopoietic stem cellular with 30% blasts and dysplastic ery-
cell transplantation (HSCT) is feasible and can throid precursors. Karyotype was normal male
offer a survival benefit for selected Medicare- and fluorescence in situ hybridization demon-
age patients.5 Although it generally is accepted strated monosomy 20. His Eastern Coopera-
that older patients with AML can benefit from tive Oncology Group (ECOG) PS had recently

2472 Cancer June 15, 2018


INSIGHT from the Experts

declined to 2. He received 8 cycles of decitabine (at a dose of The favorable-risk group included patients with the core-binding
20 mg/m2/day for 5 days on 28-day cycles) on a clinical trial. factor leukemias and acute promyelocytic leukemia, with a 5-
Physical therapy was initiated to improve his PS with the goal year overall survival (OS) rate of 34%. Those in the
of allogeneic HSCT. He achieved CR and after improvement in intermediate-risk category had a 5-year OS rate of 13%,
his PS, the patient underwent allogeneic HSCT from a whereas the 5-year OS for patients with adverse cytogenetic
matched unrelated donor after reduced intensity conditioning. risk was 2%. In terms of disease response, AML with complex
He developed gastrointestinal graft-versus-host disease but at cytogenetics (defined as 5 unrelated abnormalities in the
the time of last follow-up, 3.5 years after transplantation, the United Kingdom and more recently as 3 unrelated abnormali-
patient was off all immunosuppression and continued to work. ties in the United States) has particularly dismal CR rates. In the
MRC AML11 trial, the CR rate was 26% for those with a com-
Case 3 plex karyotype and 45% for patients with an adverse but non-
A 73-year-old woman presented to the emergency department complex karyotype.8 A subsequent study of 635 adults aged
with a 3-week history of progressive dyspnea and was found 60 years demonstrated that 25% of patients with complex
to have a white blood cell count of 86,200/lL with 68% blasts. karyotypes (3 abnormalities) achieved a CR compared with
She underwent bone marrow biopsy with aspiration and initi- a CR rate of 56% in the other cytogenetic risk groups
ated treatment with hydroxyurea for cytoreduction. Her bone combined.13
marrow was 50% cellular with 63% myeloid blasts; AML fluo- Specific molecular abnormalities influence AML outcomes and
rescence in situ hybridization panel and cytogenetics were nor- must be included in the initial risk assessment. The updated
mal. Her ECOG PS had recently declined to 1. She received National Comprehensive Cancer Network and European Leu-
induction with cytarabine and idarubicin (7 1 3 regimen), which kemiaNet AML risk stratifications include cytogenetic and
was complicated by neutropenic fever and severe mucositis, molecular abnormalities and are compared in Table 1.14,15
precluding a bone marrow evaluation on day 14. Next- Examples of molecular abnormalities influencing cytogenetic
generation sequencing for myeloid malignancies revealed a risk include mutant c-KIT, which is associated with a shorter
biallelic mutation in CCAAT/enhancer binding protein a gene time to disease recurrence and decreased OS in patients with
(CEBPA). She achieved CR and went on to complete 4 cycles t(8,21) and to a lesser extent in those with inv(16).16-18 In
of outpatient consolidation with intermediate-dose cytarabine. patients with cytogenetically normal AML, internal tandem
At the time of last follow-up, 18 months after treatment, the duplication (ITD) in the fms-like tyrosine kinase 3 (FLT3) gene is
patient was doing well. equivalent to the poor-risk cytogenetic group in terms of prog-
These cases refute the sense of nihilism regarding therapy in nosis.19-21 Conversely, mutation of nucleophosmin 1 (NPM1)
elderly patients with AML that still exists in some areas outside in the absence of a FLT3 mutation confers a favorable progno-
of the academic community and reinforce that the choice is no sis20 as does a double allelic mutation in CEBPA in patients
longer induction versus no treatment, and that one treatment with cytogenetically normal AML.22,23 Not surprisingly, in the
does not fit all. Optimal outcomes depend on the careful selec- prior European LeukemiaNet categorization, older patients had
tion of patients to identify which elderly patients might benefit worse outcomes when compared with younger patients
the most from the growing list of available AML treatments. within each risk group.24,25 Moreover, older patients are more
Below, we review assessment of disease and patient-related likely to fall into the poor-risk molecular category, with higher
factors as well as treatment advances, and present an algo- rates of adverse runt-related transcription factor 1 (RUNX1),
rithm with which to guide the management of the older adult additional sex combs-like 1 (ASXL1), and TP53 mutations, and
with AML. lower rates of favorable mutations compared with younger
patients.26
LITERATURE REVIEW Broad panel-based mutational analyses recently have defined
distinct genetic profiles that correspond to clinical subgroups
Risk Stratification of AML, including t-AML, s-AML, and de novo AML. Among a
Disease-related factors cohort of 42 patients aged  60 years with de novo AML, 33%
Cytogenetic abnormalities are crucial to guiding treatment deci- had mutations commonly found in s-AML, such as ASXL1,
sions because they remain the most important prognostic fac- SRSF2, and STAG2. Within the same cohort, 21% had TP53
tor in AML. In fact, recurrent genetic abnormalities underlie the mutations, whereas the remainder had mutations associated
recently updated World Health Organization classification of with de novo AML (ie, RUNX1, NPM1, RAS, DNA methyltrans-
myeloid neoplasms and acute leukemia.6 More unfavorable kar- ferase 3 alpha [DNMT3A], and isocitrate dehydrogenase [IDH]
yotypes are noted among older patients (those aged 60 years) 1/IDH2).27 Of particular importance are the TP53 gene muta-
(40%-50%) compared with younger patients (approximately tions, which lead to loss of important tumor suppressor func-
30%).7,8 Furthermore, older adults with AML are more likely to tions including DNA repair programs and cell cycle arrest.28
express high levels of the multidrug resistance gene,9 making Most often, TP53 mutations are noted in patients with AML
elderly patients with AML more resistant to therapy. Grimwade with a complex karyotype or t-AML.29 In a study of patients
et al analyzed 1065 patients with a median age of 66 years newly diagnosed with AML, TP53-mutated AML was associ-
(range, 44-91 years) who were treated with intensive chemo- ated with a lower CR rate (41% vs 57%) and OS rate (9% vs
therapy in the UK Medical Research Council (MRC) AML11 trial, 24% at 2 years) irrespective of age or the type of treatment
and outcomes were classified based on 3 cytogenetic risk received (high-intensity vs low-intensity chemotherapy).30 The
groups, which were similar across cooperative groups.8,10-12 list of prognostically relevant molecular abnormalities

Cancer June 15, 2018 2473


INSIGHT from the Experts

TABLE 1. Updated AML Risk Classification From the NCCN and ELN

Risk Category NCCN Cytogenetics15 NCCN Molecular15 ELN Genetic Abnormality14

Favorable Core binding factor: Normal cytogenetics: NPM1 t(8;21); RUNX1-RUNX1T1


inv(16), t(16;16), mutation in the absence Inv(16) or t(16;16); CBFB-MYH11
t(8;21), or t(15;17) of FLT3-ITD or isolated biallelic Mutated NPM1 without FLT3-ITD
(double) CEBPA mutation or with FLT3-ITDlow*
Biallelic mutated CEBPA
Intermediate Normal cytogenetics Core-binding factor with KIT Mutated NPM1 and FLT3-ITDhigh*
18 alone, t(9;11), mutation Wild-type NPM1 without FLT3-ITD
other nondefined T(9;11); MLLT3-KMT2A
Cytogenetic abnormalities not classified
as favorable or adverse
Poor (NCCN) Complex (3 clonal Normal cytogenetics: with T(6;9); DEK-NUP214
Adverse (ELN) chromosomal FLT3-ITD mutation T(v;11q23.3); KMT2A rearranged
abnormalities) TP53 mutation T(9;22); BCR-ABL1
Monosomal karyotype: Inv(3) or t(3;3); GATA2,MECOM(EVI1)
-5, 5q-, -7, 7q-, -5 or del (5q); -7;-17/abn(17p)
11q23- non t(9;11) Complex karyotype, monosomal karyotype
inv(3), t(3;3) Wild-type NPM1 and FLT3-ITDhigh
t(6;9) Mutated RUNX1
t(9;22) Mutated ASXL1
Mutated TP53
Abbreviations: AML, acute myeloid leukemia; ASXL1, additional sex combs-like 1; CBFB-MYH11, core-binding factor beta subunit-smooth muscle myosin
heavy chain 11; CEBPA, CCAAT/enhancer binding protein alpha; DEK-NUP214, DEK-nucleoporin 214; ELN, European LeukemiaNet; FLT3-ITD, fms-like tyrosine
kinase 3 internal tandem duplication; GATA2, GATA-binding protein 2; MECOM(EVI1), MDS1 and EVI1 complex locus; MLLT3-KMT2A, MLLT3-lysine methyl-
transferase 2A; NCCN, National Comprehensive Cancer Network; NPM1, nucleophosmin 1; RUNX1, runt-related transcription factor 1; RUNX1T1, RUNX1
translocation partner 1.
*FLT3-ITD “low” indicates an allelic ratio <0.5, whereas “high” indicates an allelic ratio 0.5 using semiquantitative DNA fragment analysis by taking the
ratio of the area under the curve “FLT3-ITD” divided by area under the curve “FLT3-wild-type.”

continues to grow and supports the standard use of myeloid patients with AML.35,36 In a single-institution prospective
panel sequencing during the workup of AML. Well- cohort study of consecutive adults aged 60 years undergoing
documented genetic abnormalities also provide potential tar- induction, pretreatment impairments in cognition and objective
gets for therapy and may aid in the detection of minimal resid- physical function using the Short Physical Performance Battery
ual disease.31 Protocol and Score Sheet (timed 4-meter walk, chair stands,
and standing balance) were significantly associated with
worse OS.37 Klepin et al have championed comprehensive
Patient-related factors geriatric assessment in patients with AML as a way to identify
Clinical trials in elderly patients with AML have focused on vulnerable patients in order for the field at large to consider
patients aged 60 to 80 years who have an ECOG PS between 0 more supportive therapy and find additional ways to improve
and 2. Sadly, this leaves the patients who are aged >80 years quality of life.33 Polypharmacy is common in older adults and
and considered unfit unrepresented, and further complicates has been associated with reduced functional capacity38 and
the application of clinical trial data to the individual patient. In increased mortality in those undergoing induction chemother-
the early 1990s, high AML induction mortality in the elderly apy for AML.39 Given that some patient-related factors, such
became a focus and led to investigations into lower intensity as PS and polypharmacy, are modifiable, we recommend dedi-
therapies, recognizing that some older adults should be treated cated efforts to preemptively address these factors at the time
differently from fit younger adults.32 Furthermore, chronologi- of AML diagnosis.
cal age cannot adequately account for the variety of factors,
including physical fitness, medical comorbidities, and cognitive
function, that appear to significantly impact an individual’s abil- Treatment Options
ity to tolerate induction and consolidation therapy.33 Newly diagnosed AML in the fit elderly
The decision to treat and how to treat AML in the elderly Induction chemotherapy
should not rely solely on PS and chronological age but ideally Since the 1980s, the backbone of induction chemotherapy in
should include comprehensive assessments that define the United States has been the “7 1 3” regimen, consisting of
degrees of frailty and predict tolerance to treatment and the continuous infusion of cytarabine for 7 days and an anthracy-
likelihood of benefiting from intensive approaches. Comorbid- cline for 3 days.40,41 This regimen led to remissions in only 40%
ities can be assessed using the Hematopoietic Cell Transplan- to 60% of older adults compared with 60% to 80% of younger
tation–Specific Comorbidity Index, in which higher scores adults.42 Numerous subsequent studies tested modifications in
correlate with worse outcomes in transplantation patients34 the dose of cytarabine, the dose or type of anthracycline, or the
and are independent predictors of early death in elderly addition of novel agents.43-46 The Acute Leukemia French

2474 Cancer June 15, 2018


INSIGHT from the Experts

Association-9801 study evaluated remission induction in “no further therapy” arm had developed disease recurrence by
patients aged 50 to 70 years using either high doses of daunoru- a median of 4 months.61 The choice of consolidation therapy
bicin (at a dose of 80 mg/m2/day for 3 days), high-dose idarubi- should be guided by baseline risk assessment using cytoge-
cin (IDA4; at a dose of 12 mg/m2/day for 4 days), or standard- netic and molecular markers, along with reassessment of
dose idarubicin (IDA3; at a dose of 12 mg/m2/day for 3 days). patient fitness. In younger adult patients with favorable-risk
There were no significant differences noted with regard to the disease, consolidation chemotherapy may lead to lasting
OS, event-free survival, or rate of disease recurrence, but CR remissions, whereas for those with poor-risk disease, alloge-
rates were significantly higher for the groups treated with IDA3 neic stem cell transplantation may provide the only possibility
and IDA4 compared with the high-dose daunorubicin group of cure. However, to the best of our knowledge, there is no
(83%, 78%, and 70%, respectively, in the IDA3, IDA4, and dau- clear evidence to support postremission therapy, except for
norubicin arms).47 Trials in younger adults (aged 18-60 years) transplantation, among older adults with AML. Given the sig-
demonstrated that high-dose daunorubicin (90 mg/m2) led to nificant morbidity and mortality related to allogeneic transplan-
better OS compared with daunorubicin at a dose of 45 mg/m2, tation, further risk stratification of intermediate-risk patients
and no difference was noted between high-dose daunorubicin remains a very active area of research to avoid both overtreat-
and IDA.48,49 In patients aged >60 years, daunorubicin at a ment and undertreatment.
dose of 90 mg/m2 in the first induction cycle was tolerated as High-dose cytarabine (HiDAC) became a standard consolida-
well and led to higher response rates (CR rate of 64% vs 54%) tion regimen for patients aged 60 years with favorable-risk
when compared with conventional dose daunorubicin (45 mg/
AML in first complete remission based on the Cancer and Leu-
m2/dose), but did not improve OS.50 Subsequently, the UK
kemia Group B (CALGB) trial, which evaluated 3 cytarabine
NCRI AML17 trial51 demonstrated no difference in any out- doses (3 g/m2 twice daily on days 1, 3, and 5 [HiDAC] vs 400
come when comparing daunorubicin at a dose of 60 mg/m2 ver- mg/m2/day for 5 days vs 100 mg/m2/day for 5 days). Each regi-
sus 90 mg/m2 for induction in patients with AML who were men was intended for 4 consolidation cycles. There was a
aged 16 to 72 years. Outside of a clinical trial, we routinely use higher probability of continuous CR at 4 years in the HiDAC
idarubicin at a dose of 12 mg/m2 for induction in older adults.
group compared with the groups receiving lower doses of
Alternatively, daunorubicin at a dose of 60 mg/m2 is a reason-
cytarabine (44% vs 24% and 29%, respectively).62 The benefit
able anthracycline choice.
of HiDAC was more evident within the patients with core-
Until very recently, induction options were limited to regimens binding factor AML when compared with the other cytogenetic
that have been around since the 1980s. Fortunately, with the groups.63 Unfortunately, <50% of patients aged 60 years
recent approval by the US Food and Drug Administration (FDA) were able to receive the intended number of cycles of HiDAC
of CPX-351 injection for adults with newly diagnosed high-risk due to increased toxicity, and age subsequently was capped at
AML (t-AML or AML with myelodysplasia-related changes), 60 years after an interim analysis demonstrated no benefit for
we now have a new option.52 This medication repackages the HiDAC among patients aged >60 years.62 Lowering the cytara-
2 classic chemotherapy agents cytarabine and daunorubicin in bine dose (1.5-2 g/m2 daily for 6 days) may be better tolerated
a 5:1 molar ratio within a liposomal formulation and is given for in older patients.64 Subsequent data from MRC AML15 sug-
induction at a dose of 100 U/m2 on days 1, 3, and 5.53 The treat- gest that, for consolidation, intermediate-dose cytarabine at a
ment of older adults (aged 60-75 years) with newly diagnosed dose of 1.5 g/m2 may be equivalent to 3 g/m2 and 2 consolida-
high-risk AML revealed significantly improved OS (median OS tion cycles may be equivalent to 3. However, this was noted
of 9.56 months vs 5.95 months) when compared with standard among younger patients who received 2 induction cycles.65
7 1 3 therapy. Grade 3 to 5 adverse events were similar to Our group uses 4 intermediate-dose cytarabine consolidation
those noted with the standard 7 1 3 regimen.53 Landmark sur- cycles for older patients (aged >60 years) with favorable-risk
vival analyses at the time of allogeneic HSCT are encouraging AML. For fit older adults with intermediate-risk AML, we use
for the use of CPX-351 as a possible bridge to successful trans- up to 3 cycles of intermediate-dose cytarabine while the
plantation for a very poor-risk subgroup of patients with patients are undergoing transplantation workup. Donor source,
AML.54 Currently, we use CPX-351 for patients with t-AML or match, and comprehensive patient assessments are consid-
s-AML and continue to use the 7 1 3 regimen in those with de ered when deciding who should proceed to transplantation in
novo AML. As clinical experience expands with an ongoing trial first CR and for whom transplantation should be reserved in
in patients at high risk of induction mortality,55 CPX-351 may case of disease recurrence. We use alternative regimens (clini-
become more broadly used. The recent reapproval of gemtuzu- cal trial, hypomethylating agents [HMAs]) or fewer cycles of
mab ozogamicin (discussed below) adds the option of adding intermediate-dose cytarabine for patients with unfavorable-
gemtuzumab ozogamicin to the 7 1 3 regimen in older patients risk AML as a bridge to transplantation.
with non–high-risk, de novo AML. Table 2 shows recently
For patients with unfavorable/high-risk disease, allogeneic
approved medications in AML.52,56-58
HSCT leads to improved OS.10 A landmark analysis of patients
aged <60 years with AML and high-risk cytogenetics in first
Postremission therapy complete remission demonstrated that patients who received
The need for additional chemotherapy to sustain initial remis- allogeneic transplantation experienced superior 5-year OS
sion in patients with AML has been recognized since the compared with patients who received continued consolidation
1970s and 1980s.59,60 A trial comparing consolidation or pro- chemotherapy (48% vs 18%, respectively; P 5 .004).66 How-
longed maintenance with no further therapy was stopped pre- ever the morbidity and mortality ascribed to allogeneic trans-
maturely after interim analysis because all the patients in the plantation cannot be taken lightly in elderly patients with AML.

Cancer June 15, 2018 2475


INSIGHT from the Experts

TABLE 2. Medications Approved by the FDA for AML in 2017

Therapeutic Specific Common and/or


(Brand Name) Target Approved Indication Dose/Regimen Serious Side Effects

Midostaurin FLT3 Adults with newly diagnosed 50 mg orally twice daily on d 8 to 21 of Febrile neutropenia,
(Rydapt)56 AML and FLT3 mutationa each cycle of induction with device-related infection,
cytarabine and daunorubicin and on and mucositis
d 8 to 21 of each cycle of consolida-
tion with high-dose cytarabine
Enasidenib IDH2 Adults with recurrent or 100 mg orally daily until disease Hyperbilirubinemia,
(Idhifa)57 refractory AML and IDH2 progression differentiation syndrome
mutationa (fever, dyspnea, effusions,
and edema)
CPX-351 NA Adults with newly diagnosed Induction: daunorubicin 44 mg/m2 and Hemorrhage, cardiotoxicity,
(Vyxeos)52 therapy-related or cytarabine 100 mg/m2 on d 1, 3, and 5; copper overload, and
MDS-related AML consolidation: daunorubicin 29 mg/m2 hypersensitivity reactions
and cytarabine 65 mg/m2 liposome on
d 1 and 3
Gemtuzumab CD33 Adults with newly diagnosed Combination induction: 3 mg/m2 on d 1, 4, Neutropenia, hepatotoxicity,
ozogamicin CD33-positive AML; adults or and 7 with daunorubicin and veno-occlusive disease,
(Mylotarg)58 children aged  2 y with cytarabine; single induction: 6 mg/m2 and hemorrhage
recurrent/refractory on d 1 and 3 mg/m2 on d 8, continued
CD33-positive AML for up to 8 courses of 2 mg/m2 once
every 4 wk; recurrent/refractory
AML: 3 mg/m2 on d 1, 4, and 7
Abbreviations: AML, acute myeloid leukemia; FDA, US Food and Drug Administration; FLT3, fms-like tyrosine kinase 3; IDH2, isocitrate dehydrogenase 2;
MDS, myelodysplastic syndromes; NA, not applicable.
a
Mutation must be documented by an FDA-approved test.

The advent of intermediate-intensity and reduced-intensity and intermediate-intensity regimens may offer disease stabiliza-
conditioning regimens has allowed more older adults to suc- tion, prolonged survival, and improved quality of life. Low-dose
cessfully undergo transplantation, thereby attaining the benefi- cytarabine (LDAC) treatment was established after a random-
cial graft-versus-leukemia effect while minimizing toxicities ized study demonstrated higher rates of CR (18% vs 1%) and
related to transplantation.67,68 In fact, a large study by the Cen- OS (4 months vs 3 months) compared with hydroxyurea in a
ter for International Blood and Marrow Transplant Research population of unfit older adults.73 LDAC given at a dose of 20
demonstrated similar outcomes in younger and older adults mg subcutaneously twice daily for 10 days on a cycle of 4 to 6
with AML and MDS who underwent allogeneic transplantation weeks was found to be well tolerated; unfortunately, patients
after reduced intensity conditioning.68 The choice of graft with adverse cytogenetics derived no benefit in remission or
donor, graft source, and conditioning regimen for the older survival.73 Gemtuzumab ozogamicin, a humanized monoclonal
adult with AML recently was reviewed.69,70 A large meta- antibody targeting CD33 conjugated to calicheamicin, a cyto-
analysis including 749 patients aged >60 years with AML sup- toxic antibiotic, initially was approved in 2001 for patients aged
ports the use of reduced-intensity allogeneic HSCT for elderly 60 years who were diagnosed with AML74 and was with-
patients with AML, with a 3-year recurrence-free survival rate drawn in 2010 due to toxicity.75 In a phase 2 trial of adults aged
of 35%.71 The limited numbers of patients aged >70 years, >75 years with newly diagnosed AML, the median OS was 4.9
with the oldest patient reported as being 74 years, precluded a months in the group treated with gemtuzumab ozogamicin ver-
meaningful analysis of transplantation outcomes in the group sus 3.6 months among individuals treated with best supportive
of patients aged >70 years. These results argue against using care.76 Single-agent use in patients with recurrent disease dem-
age alone to exclude allogeneic transplantation as a potentially onstrated a CR rate of 26% after 1 cycle.77 Subsequent ran-
curative approach in elderly patients with AML. Outcomes domized phase 3 data showed the clinical benefit both when
may be improved by the careful selection of patients for this gemtuzumab ozogamicin was used as monotherapy78 or when
approach.72 it was added to induction chemotherapy at a lower dose of 3
mg/m2.79 Given the improved toxicity profile when used at
Newly diagnosed AML in the unfit elderly lower doses, gemtuzumab ozogamicin recently was reap-
proved by the FDA for adults with newly diagnosed and recur-
Previously, treatment options for older adults who were not
rent/refractory, CD33-positive AML (Table 2).52,56-58
considered fit for induction chemotherapy were limited to sup-
portive care with transfusions and hydroxyurea or referral to The HMAs azacitidine and decitabine, which initially were
hospice. Supportive care alone for older adults with AML was developed as antimetabolites, were shown to target leukemo-
associated with a median survival of 1 month during the genic epigenetic changes.80 Both currently are approved by
1990s.4 For patients desiring treatment, several low-intensity the FDA for MDS and commonly are used off label for less

2476 Cancer June 15, 2018


INSIGHT from the Experts

intensive therapy or refractory disease in patients with AML. higher rates of temporary mutation clearance. Response in
Trial experience with azacitidine in patients with AML began patients with unfavorable cytogenetic risk was 67% versus
with the AZA-MDS-001 phase 3 study, which included patients 34% in those with favorable or intermediate risk, and
with higher-risk MDS with up to 30% blasts, which by World responses among patients with TP53 mutations were 100%
Health Organization criteria includes AML with >20% blasts.81 (all 21 patients) versus 41% for patients with wild-type TP53.92
Subjects were randomized in a 1:1 ratio to azacitidine at a dose An international phase 3 trial of decitabine at a dose of 20 mg/
of 75 mg/m2 per day for 7 days per 28-day cycle or to 1 of 3 con- m2/day for 5 days in 4-week cycles versus physician choice of
ventional care options (supportive care, LDAC, or intensive either LDAC at a dose of 20 mg/m2 subcutaneously once daily
chemotherapy). Post hoc analysis of the patients with AML for 10 days in 4-week cycles or best supportive care was per-
demonstrated CR rates of 18% among 55 patients treated formed.93 The study included 485 patients with a median age
with azacitidine for a median of 8 cycles, compared with a CR of 73 years, 63% of whom had intermediate-risk and 36% of
rate of 16% among 58 patients treated with conventional ther- whom had poor-risk cytogenetics. The primary endpoint of OS
apy for a median of 5.5 cycles (LDAC in 20 patients), 2.5 cycles did not reach statistical significance; the median OS for
(intensive chemotherapy in 11 patients), or best supportive patients receiving decitabine was 7.7 months (95% CI, 6.2-9.2
care (27 patients). Despite similar CR rates, the azacitidine months) after a median of 4 cycles of decitabine compared
group had a significantly higher median survival of 24.5 months with 5.0 months (95% CI, 4.3–6.3 months) for patients treated
compared with 16 months for patients treated with conven- with LDAC or best supportive care (P 5 .108).93 Ad hoc analy-
tional therapy.81 Two studies from the Austrian Azacitidine sis of more mature data demonstrated a statistical survival
Registry showed that azacitidine-treated patients with AML benefit. However, concerns about the lower dose of LDAC and
had a median OS of approximately 9 to 10 months.82,83 A sub- not meeting prespecified endpoints may have precluded
sequent international phase 3 trial included 488 patients aged approval for AML in the United States.94 Despite lacking FDA
65 years with newly diagnosed AML. Subjects were random- approval for AML, HMAs remain commonly used and currently
ized 1:1 to azacitidine or a preselected conventional regimen are under active investigation, especially for combination ther-
(induction chemotherapy, LDAC, or best supportive care). The apy. To the best of our knowledge, the precise mechanism(s)
median OS was 10.4 months for the azacitidine group (95% of HMAs are not fully elucidated,95 but novel studies have sug-
confidence interval [CI], 8.0-12.7 months) versus 6.5 months gested they may be used at lower and longer doses as an
for the conventional care group (95% CI, 5.0-8.6 months).84 “epigenetic primer.”96,97 Reversing aberrant methylation of
Initial phase 1 studies of decitabine in patients with AML dem- tumor suppressor genes may sensitize leukemic cells to cyto-
onstrated promising CR/CR with incomplete hematologic toxic therapy.98 This is especially relevant for AML that is asso-
ciated with mutations in TET2, IDH1 and IDH2, ASXL1, and
recovery (CRi) rates of 18% to 32%.85-87 Three phase 2 studies
DNMT3A, which encode proteins involved in epigenetic
established the effectiveness of decitabine in older adults.
regulation of transcription.99
Drawing on MDS literature, Cashen et al used decitabine at a
dose of 20 mg/m2/day for 5 days every 4 weeks in adults with a The selection of lower intensity treatment options relies on the
median age of 74 years. The median OS was 7.7 months for all assessment of disease characteristics/prior therapy, PS, and
patients and 14 months for responders. The CR rate was 24% patient wishes. If patients have a very poor premorbid PS,
after a median of 4.5 cycles.88 A European study treated 227 remain fully dependent despite treatment of acute complica-
elderly patients with AML with decitabine at a dose of 15 mg/ tions and attempts at rehabilitation, or clearly do not want treat-
m2 every 8 hours for 3 days on a 6-week cycle and demon- ment, then either best supportive care or hospice is indicated.
strated a CR in 13% and a partial response/CRi in 12%.89 Blum For unfit patients, HMAs are offered; for those who received
et al treated 53 patients with a median age of 74 years with prior HMAs (for MDS), we often used LDAC. The reapproval of
decitabine at a dose of 20 mg/m2/day for 10 days, with the gemtuzumab ozogamicin adds complexity to this choice, with
cycle length reduced to 5 days if a response was observed.90 practice patterns still emerging; nevertheless, patient comorbid
The CR rate was 47% and the median survival was 12.7 conditions, side effect profiles, and logistic details can inform a
months after a median of 3 cycles of decitabine. The longer practical choice.
treatment cycles were found to lead to increased febrile neu-
tropenia but not mortality.90 However, interim results of an Salvage therapy for patients with
ongoing randomized phase 2 study comparing 10-day versus recurrent/refractory disease
5-day decitabine as frontline therapy in elderly patients with Unfortunately, the majority of older adults with AML will
AML who were ineligible for induction demonstrated no signifi- develop disease recurrence and die of their disease. The prog-
cant difference in remission or survival outcomes between the nosis for patients with recurrent disease is dismal, with <10%
2 regimens (CR/pathologic CR/CRi rate of 51% for 5-day vs of these individuals surviving long term.100 To our knowledge,
41% for 10-day; P value was not significant).91 Ten-day decita- there is no standard salvage regimen for patients with recur-
bine may be particularly beneficial for the treatment of patients rent AML, and much of the data regarding treatment options
with TP53-mutated AML based on a recent study of 116 are derived from small and/or single-institution studies, which
patients with either AML or MDS who were treated with deci- have inherent limitations. Enrollment in clinical trials is critical.
tabine at a dose of 20 mg/m2 for 10 days per monthly cycle. Outside of clinical trials, frequently used regimens combine an
Enhanced exome, gene panel, and serial sequencing were anthracycline/anthracenedione with other active agents. The
used to evaluate mutation clearance, and patients with classi- addition of granulocyte colony-stimulating factor (G-CSF) to
cally worse prognostic markers actually were found to have regimens can accelerate time to neutrophil recovery,101 but

Cancer June 15, 2018 2477


INSIGHT from the Experts

TABLE 3. Selected Salvage Regimens for Recurrent/Refractory AML

No. of Patients
(Recurrent/ Median Age, Complete Median OS,
Regimen Refractory AML) Years Response Months Study

Mitoxantrone and etoposide (ME) 61 (20/21) 47 43% NA Ho 1988102


36 (27/9) 58 34% NA Trifilio 2012103
Mitoxantrone, etoposide, and IDAC (MEC) 32 (18/14) 24 66% 9 Amadori 1991104
74 (30/28) 37 55% NA Spadea 1993105
29 (23/6) 54 59% NA Trifilio 2012103
Fludarabine, cytarabine, and G-CSF (FLAG) 69 (NA) 63 81% >16 Estey 1994101
65 (21 early/44 late)a 47 81%/30% 17/3 Jackson 2001106
41 (19/11) 53 56% 11 Carella 2001107
Fludarabine, cytarabine, G-CSF, and 32 (NA) 59 53% <12 de la Rubia 2002108
idarubicin (FLAG-IDA)
46 (36/10) 41 52% 11 Pastore 2003109
Cladribine, ARA-C, and G-CSF (CLAG) 58 (8/50) 45 50% 8.5 Wrzesien-Kus 2003110
Cladribine, ARA-C, G-CSF, and 114 (39/75) 45 53% 9 Wierzbowska 2008111
mitoxantrone (CLAG-M)
Abbreviations: AML, acute myeloid leukemia; ARA-C, cytosine arabinoside; G-CSF, granulocyte colony-stimulating factor; IDAC, intermediate-dose ARA-C;
NA, not available; OS, overall survival.
a
Patients in group 1 experienced late recurrence  6 months after first-line chemotherapy, whereas patients in group 2 either experienced an early disease
recurrence <6 months after stopping chemotherapy or were refractory to first-line chemotherapy.

does not improve outcomes. Table 3 shows a summary of were found to have an IDH2 mutation based on a companion
selected salvage regimens.102-111 assay.57 The 2 common IDH2 mutations, R140 and R172, both
lead to accumulation of the metabolite (R)-2-hydroxyglutarate,
Mutation-specific targeted therapies leading to hematopoietic differentiation block, and enasidenib
Despite years of intense study in the field, to our knowledge suppresses (R)-2-hydroxyglutarate in both mutation sub-
no therapies were approved by the FDA for AML between types.117 Approval was based on a multicenter, open-label
2001 and 2016.112 Fortunately, there have been 2 recent phase 1/2 trial that included 199 patients with recurrent/refrac-
approvals of targeted therapies for AML, namely FLT3 and tory AML, 62% of whom were aged 65 years. In the popula-
IDH2. A focus on the poor prognostic FLT3-ITD led to the tion of patients with recurrent/refractory AML, the overall
development of several FLT3 inhibitors.113-115 Midostaurin response rate was 40.3% and the median OS was 9.3 months.
was approved by the FDA in April 2017 for newly diagnosed Grade 3 and 4 toxicities included indirect hyperbilirubinemia
adults with FLT3-positive AML in combination with standard (12%) and IDH inhibitor-associated differentiation syndrome
7 1 3 induction and cytarabine consolidation.56 Approval was (7%).118 Differentiation syndrome can be deadly and patients
based on an international, randomized, placebo-controlled present with fever; dyspnea; pulmonary infiltrates; pleural or
phase 3 trial (CALGB 10603) published in June 2017.116 Of the pericardial effusions; rapid weight gain or peripheral edema;
717 patients randomized and stratified according to the sub- lymphadenopathy; bone pain; and hepatic, renal, or multiorgan
type of FLT3 mutation (point mutation in the tyrosine kinase dysfunction and should be treated with systemic corticoste-
domain or low- or high-ratio ITD), 360 were assigned to the roids (Table 2).52,56-58 Targeted agents provide an opportunity
midostaurin group. The primary endpoint was OS, which was to intervene based on disease biology and possibly eliminate
found to be significantly longer in the group treated with mid- minimal residual disease, and warrant further study in the older
ostaurin compared with the placebo group (75 months vs 26 patient with AML.
months; hazard ratio for death, 0.78 [1-sided P 5 .009]), with a
consistent benefit noted across the FLT3 mutation subgroups. Promising therapies and ongoing clinical trials
The most common grade 3/4 adverse reactions were febrile In an effort to fast-track new therapies desperately needed for
neutropenia, device-related infection, and mucositis.116 It is AML, specifically in the elderly population, the Leukemia and
interesting to note that the trial only included adults aged 18 to Lymphoma Society is leading the Beat AML Master Trial. Up to
59 years. Despite lacking data in the elderly, we offer this treat- 500 newly diagnosed patients aged 60 years will undergo
ment to patients aged >60 years with close monitoring for rapid genomic testing that will direct patients to a relevant tar-
toxicity. geted therapy arm with correlative studies.119 Information
In August 2017, the FDA granted approval for enasidenib, an regarding the numerous other therapeutic trials in elderly
IDH2 inhibitor for adults with recurrent or refractory AML who patients with AML can be obtained at ClinicalTrials.gov.

2478 Cancer June 15, 2018


INSIGHT from the Experts

Figure 1. Acute myeloid leukemia (AML) treatment algorithm for the older adult. ARA-C indicates cytosine arabinoside; APL, acute
promyelocytic leukemia; FLT3, fms-like tyrosine kinase 3; MRD, minimal residual disease; NextGen, next-generation; s-AML, second-
ary acute myeloid leukemia; t-AML, therapy-related acute myeloid leukemia.

DISCUSSION therapy in octogenarians regardless of PS, the paucity of


transplantation data in this age group, and the potential for
The landscape of AML treatment is changing. Although the
long-lasting remissions with reduced intensity therapies
recently approved induction and targeted therapies offer new
allowed this patient an option that improved his quality life
hope for patients who receive this life-threatening diagnosis,
and prolonged his survival.
they add additional considerations to an already complex dis-
ease. The classic (albeit simplified) algorithm of induction, fol- Case 2 highlights the importance of comprehensive patient
lowed by consolidation (with or without transplantation) assessment, not just to define prognosis but also to identify
versus hospice is no longer sufficient. Instead, significant opportunities to intervene with regard to modifiable factors,
advances including broad panel-based genetic assessment, such as physical deconditioning and managing polypharmacy.
more focus on modifiable patient-related factors, and The 71-year-old patient had poor-risk disease and was unfit for
improved supportive care are moving the field forward, espe- induction; therefore, hypomethylating therapy was initiated
cially for elderly patients with AML. along with physical therapy. It took a compelling explanation by
his physician to explain that although the process of improving
The unique circumstances of each patient (disease related
his physical fitness may be painful due to arthritis, it potentially
and patient related) need to be thoughtfully considered when
could make life-saving treatment possible down the road. At
developing a treatment plan (Fig. 1.). The diagnosis of AML
his age, complications related to induction therapy were likely
is, frankly, sudden and scary. Patients and their support sys-
to make the patient much weaker and possibly preclude sub-
tems often are very fearful and can face a wide knowledge
sequent consolidation with transplantation. Conversely, the
gap in understanding the disease and its many attendant
patient in case 3 had an excellent PS with no medical comor-
complications. Often the urge to feel proactive in the face of
bidities, which is fortunate because she had proliferative dis-
cancer can lead to an understandable but sometimes danger-
ease with a white blood cell count of 86,200/lL. She required
ous push to “do everything possible.” We must be especially
cytoreduction and was able to complete induction therapy; her
prudent when caring for the elderly patient with AML to
favorable-risk disease may be cured, although follow-up was
ensure that we use all available data when building a treat-
ment plan and that all safety and supportive care measures short.
are available to these patients as we embark on treatments The care of the older adult with AML requires a comprehensive
that may do more harm than good. For example, in the approach and a constant reevaluation of the risks and potential
patient in case 1, clinicians may consider induction therapy benefits of all available treatments. Along with pharmaceutical
when sitting across the clinic room from a very functional and hospital-based interventions, support from family and
and fit patient who wants a chance at cure. However, trial skilled providers (nursing, physical therapy, occupational ther-
experience demonstrating dismal outcomes for induction apy, home health aides, nutritionists, and transportation) is

Cancer June 15, 2018 2479


INSIGHT from the Experts

essential. Clinical trials for disease-specific and best supportive or older with acute myeloid leukemia: results from Cancer and Leuke-
therapy should be strongly encouraged to continue to improve mia Group B 8461. Blood. 2006;108:63-73.
the care of patients with AML. 14. Dohner H, Estey E, Grimwade D, et al. Diagnosis and management
of AML in adults: 2017 ELN recommendations from an international
expert panel. Blood. 2017;129:424-447.
FUNDING SUPPORT 15. National Comprehensive Cancer Network. Acute myeloid leukemia
No specific funding was disclosed. (Version 3.2017-June 6, 2017). https://www.nccn.org/professionals/
physician_gls/pdf/aml.pdf. Accessed July 1, 2017.
16. Boissel N, Leroy H, Brethon B, et al. Incidence and prognostic impact
CONFLICT OF INTEREST DISCLOSURES of c-Kit, FLT3, and Ras gene mutations in core binding factor acute
The authors made no disclosures. myeloid leukemia (CBF-AML). Leukemia. 2006;20:965-970.
17. Paschka P, Marcucci G, Ruppert AS, et al; Cancer and Leukemia
Group B. Adverse prognostic significance of KIT mutations in adult
REFERENCES acute myeloid leukemia with inv and t(8;21): a Cancer and Leukemia
1. National Cancer Institute. Cancer Stat Facts: acute myeloid leukemia Group B Study. J Clin Oncol. 2006;24:3904-3911.
(AML). https://seer.cancer.gov/statfacts/html/amyl.html. Accessed July 18. Park SH, Chi HS, Min SK, Park BG, Jang S, Park CJ. Prognostic
1, 2017. impact of c-KIT mutations in core binding factor acute myeloid leuke-
2. Boddu PC, Kantarjian HM, Ravandi F, et al. Characteristics and out- mia. Leuk Res. 2011;35:1376-1383.
comes of older patients with secondary acute myeloid leukemia accord- 19. Gale RE, Green C, Allen C, et al. The impact of FLT3 internal tan-
ing to treatment approach. Cancer. 2017;123:3050-3060. dem duplication mutant level, number, size, and interaction with
3. Alibhai SM, Leach M, Minden MD, Brandwein J. Outcomes and NPM1 mutations in a large cohort of young adult patients with acute
quality of care in acute myeloid leukemia over 40 years. Cancer. 2009; myeloid leukemia. Blood. 2008;111:2776-2784.
115:2903-2911. 20. Whitman SP, Ruppert AS, Radmacher MD, et al. FLT3 D835/I836
4. Menzin J, Lang K, Earle CC, Kerney D, Mallick R. The outcomes mutations are associated with poor disease-free survival and a distinct
and costs of acute myeloid leukemia among the elderly. Arch Intern gene-expression signature among younger adults with de novo cytoge-
Med. 2002;162:1597-1603. netically normal acute myeloid leukemia lacking FLT3 internal tandem
duplications. Blood. 2008;111:1552-1559.
5. Medeiros BC, Satram-Hoang S, Hurst D, Hoang KQ, Momin F,
Reyes C. Big data analysis of treatment patterns and outcomes among 21. Kayser S, Schlenk RF, Londono MC, et al. Insertion of FLT3 internal
elderly acute myeloid leukemia patients in the United States. Ann tandem duplication in the tyrosine kinase domain-1 is associated with
Hematol. 2015;94:1127-1138. resistance to chemotherapy and inferior outcome. Blood. 2009;114:
2386-2392.
6. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the
World Health Organization classification of myeloid neoplasms and 22. Dohner K, Schlenk RF, Habdank M, et al. Mutant nucleophosmin
acute leukemia. Blood. 2016;127:2391-2405. (NPM1) predicts favorable prognosis in younger adults with acute
myeloid leukemia and normal cytogenetics: interaction with other gene
7. Appelbaum FR, Gundacker H, Head DR, et al. Age and acute myeloid mutations. Blood. 2005;106:3740-3746.
leukemia. Blood. 2006;107:3481-3485.
23. Tenen DG. Abnormalities of the CEBP alpha transcription factor: a major
8. Grimwade D, Walker H, Harrison G, et al; Medical Research Council target in acute myeloid leukemia. Leukemia. 2001;15:688-689.
Adult Leukemia Working Party. The predictive value of hierarchical
cytogenetic classification in older adults with acute myeloid leukemia 24. Dohner H, Estey EH, Amadori S, et al; European LeukemiaNet. Diag-
(AML): analysis of 1065 patients entered into the United Kingdom nosis and management of acute myeloid leukemia in adults: recom-
Medical Research Council AML11 trial. Blood. 2001;98:1312-1320. mendations from an international expert panel, on behalf of the
European LeukemiaNet. Blood. 2010;115:453-474.
9. Leith CP, Kopecky KJ, Godwin J, et al. Acute myeloid leukemia in
the elderly: assessment of multidrug resistance (MDR1) and cytogenet- 25. Mrozek K, Marcucci G, Nicolet D, et al. Prognostic significance of the
ics distinguishes biologic subgroups with remarkably distinct responses European LeukemiaNet standardized system for reporting cytogenetic
to standard chemotherapy. A Southwest Oncology Group study. Blood. and molecular alterations in adults with acute myeloid leukemia. J Clin
1997;89:3323-3329. Oncol. 2012;30:4515-4523.
10. Slovak ML, Kopecky KJ, Cassileth PA, et al. Karyotypic analysis pre- 26. Tsai CH, Hou HA, Tang JL, et al. Genetic alterations and their clini-
dicts outcome of preremission and postremission therapy in adult acute cal implications in older patients with acute myeloid leukemia. Leuke-
myeloid leukemia: a Southwest Oncology Group/Eastern Cooperative mia. 2016;30:1485-1492.
Oncology Group Study. Blood. 2000;96:4075-4083.
27. Lindsley RC, Mar BG, Mazzola E, et al. Acute myeloid leukemia is
11. Byrd JC, Mrozek K, Dodge RK, et al; Cancer and Leukemia Group B defined by distinct somatic mutations. Blood. 2015;125:1367–1376.
(CALGB 8461). Pretreatment cytogenetic abnormalities are predictive
28. Kim MP, Zhang Y, Lozano G. Mutant p53: multiple mechanisms
of induction success, cumulative incidence of relapse, and overall sur-
define biologic activity in cancer. Front Oncol. 2015;5:249.
vival in adult patients with de novo acute myeloid leukemia: results
from Cancer and Leukemia Group B (CALGB 8461). Blood. 2002; 29. Rucker FG, Schlenk RF, Bullinger L, et al. TP53 alterations in acute
100:4325-4336. myeloid leukemia with complex karyotype correlate with specific copy
number alterations, monosomal karyotype, and dismal outcome. Blood.
12. Grimwade D, Hills RK, Moorman AV, et al; National Cancer
2012;119:2114-2121.
Research Institute Adult Leukaemia Working Group. Refinement of
cytogenetic classification in acute myeloid leukemia: determination of 30. Kadia TM, Jain P, Ravandi F, et al. TP53 mutations in newly diag-
prognostic significance of rare recurring chromosomal abnormalities nosed acute myeloid leukemia: clinicomolecular characteristics,
among 5876 younger adult patients treated in the United Kingdom response to therapy, and outcomes [published online ahead of print
Medical Research Council trials. Blood. 2010;116:354-365. July 26, 2016]. Cancer. doi: 10.1002/cncr.30203.
13. Cancer and Leukemia Group B 8461, Farag SS, Archer KJ, et al. 31. Hourigan CS, Gale RP, Gormley NJ, Ossenkoppele GJ, Walter RB.
Pretreatment cytogenetics add to other prognostic factors predicting Measurable residual disease testing in acute myeloid leukaemia. Leuke-
complete remission and long-term outcome in patients 60 years of age mia. 2017;31:1482-1490.

2480 Cancer June 15, 2018


INSIGHT from the Experts

32. El Rassi F, Arellano M. Update on optimal management of acute mye- 51. Burnett AK, Russell NH, Hills RK, et al; UK NCRI AML Study
loid leukemia. Clin Med Insights Oncol. 2013;7:181-197. Group. A randomized comparison of daunorubicin 90 mg/m2 vs
60 mg/m2 in AML induction: results from the UK NCRI AML17
33. Klepin HD, Rao AV, Pardee TS. Acute myeloid leukemia and myelo-
trial in 1206 patients. Blood. 2015;125:3878-3885.
dysplastic syndromes in older adults. J Clin Oncol. 2014;32:2541-
2552. 52. US Food and Drug Administration. VYXEOS (daunorubicin and
cytarabine) liposome for injection, for intravenous use. https://
34. Sorror ML, Maris MB, Storb R, et al. Hematopoietic cell transplanta-
www.accessdata.fda.gov/drugsatfda_docs/label/2017/209401s000lbl.pdf.
tion (HCT)-specific comorbidity index: a new tool for risk assessment
Accessed August 9, 2017.
before allogeneic HCT. Blood. 2005;106:2912-2919.
53. Lancet JE, Uy GL, Cortes JE, et al. Final results of a phase III ran-
35. Savic A, Kvrgic V, Rajic N, et al. The hematopoietic cell transplanta-
domized trial of CPX-351 versus 713 in older patients with newly
tion comorbidity index is a predictor of early death and survival in
diagnosed high risk (secondary) AML. J Clin Oncol. 2016;34:7000-
adult acute myeloid leukemia patients. Leuk Res. 2012;36:479-482.
7000.
36. Giles FJ, Borthakur G, Ravandi F, et al. The haematopoietic cell trans-
54. Lancet JE, Hoering A, Uy GL, et al. Survival following allogeneic
plantation comorbidity index score is predictive of early death and sur-
hematopoietic cell transplantation in older high-risk acute myeloid leu-
vival in patients over 60 years of age receiving induction therapy for
kemia patients initially treated with CPX-351 liposome injection versus
acute myeloid leukaemia. Br J Haematol. 2007;136:624-627.
standard cytarabine and daunorubicin: subgroup analysis of a large
37. Klepin HD, Geiger AM, Tooze JA, et al. Geriatric assessment predicts phase III trial. Blood. 2016;128:906-906.
survival for older adults receiving induction chemotherapy for acute
55. Assi R, Kantarjian H, Verstovsek S, et al. CPX-351 for the treat-
myelogenous leukemia. Blood. 2013;121:4287-4294.
ment of high-risk patients with acute myeloid leukemia. Blood.
38. Maher RL, Hanlon J, Hajjar ER. Clinical consequences of polyphar- 2016;128:4047.
macy in elderly. Expert Opin Drug Saf. 2014;13:57-65.
56. US Food and Drug Administration. RYDAPT (midostaurin) for
39. Elliot K, Tooze JA, Geller R, et al. The prognostic importance of poly- oral use. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/
pharmacy in older adults treated for acute myelogenous leukemia 207997s000lbl.pdf. Accessed July 16, 2016.
(AML). Leuk Res. 2014;38:1184-1190.
57. US Food and Drug Administration. IDHIFAV R (enasidenib) tablets,

40. Yates J, Glidewell O, Wiernik P, et al. Cytosine arabinoside with dau- for oral use. https://www.accessdata.fda.gov/drugsatfda_docs/label/
norubicin or adriamycin for therapy of acute myelocytic leukemia: a 2017/209606s000lbl.pdf. Accessed August 1, 2017.
CALGB study. Blood. 1982;60:454-462.
58. US Food and Drug Administration. MYLOTARG (gemtuzumab ozo-
41. Rai KR, Holland JF, Glidewell OJ, et al. Treatment of acute myelo- gamicin) for injection. https://www.fda.gov/NewsEvents/Newsroom/
cytic leukemia: a study by Cancer and Leukemia Group B. Blood. PressAnnouncements/ucm574507.htm. Accessed September 15, 2017.
1981;58:1203-1212.
59. Peterson BA, Bloomfield CD. Prolonged maintained remissions of
42. Estey E, Smith TL, Keating MJ, McCredie KB, Gehan EA, Freireich adult acute non-lymphocytic leukaemia. Lancet. 1977;2:158-160.
EJ. Prediction of survival during induction therapy in patients with
60. Gale RP. Advances in the treatment of acute myelogenous leukemia. N
newly diagnosed acute myeloblastic leukemia. Leukemia. 1989;3:257-
Engl J Med. 1979;300:1189-1199.
263.
61. Cassileth PA, Harrington DP, Hines JD, et al. Maintenance chemo-
43. Vogler WR, Velez-Garcia E, Weiner RS, et al. A phase III trial com-
therapy prolongs remission duration in adult acute nonlymphocytic
paring idarubicin and daunorubicin in combination with cytarabine in
leukemia. J Clin Oncol. 1988;6:583-587.
acute myelogenous leukemia: a Southeastern Cancer Study Group
Study. J Clin Oncol. 1992;10:1103-1111. 62. Mayer RJ, Davis RB, Schiffer CA, et al. Intensive postremission che-
motherapy in adults with acute myeloid leukemia. Cancer and Leuke-
44. Reiffers J, Huguet F, Stoppa AM, et al. A prospective randomized trial
mia Group B. N Engl J Med. 1994;331:896-903.
of idarubicin vs daunorubicin in combination chemotherapy for acute
myelogenous leukemia of the age group 55 to 75. Leukemia. 1996;10: 63. Bloomfield CD, Lawrence D, Byrd JC, et al. Frequency of prolonged
389-395. remission duration after high-dose cytarabine intensification in acute
myeloid leukemia varies by cytogenetic subtype. Cancer Res. 1998;58:
45. Estey EH, Thall PF, Cortes JE, et al. Comparison of idarubicin 1 ara-
4173-4179.
C-, fludarabine 1 ara-C-, and topotecan 1 ara-C-based regimens in
treatment of newly diagnosed acute myeloid leukemia, refractory ane- 64. Arellano M, Winton E, Pan L, et al. High-dose cytarabine induction is
mia with excess blasts in transformation, or refractory anemia with well tolerated and active in patients with de novo acute myeloid leuke-
excess blasts. Blood. 2001;98:3575-3583. mia older than 60 years. Cancer. 2012;118:428-433.
46. Castaigne S, Chevret S, Archimbaud E, et al. Randomized comparison 65. Burnett AK, Russell NH, Hills RK, et al. Optimization of chemother-
of double induction and timed-sequential induction to a “3 1 7” induc- apy for younger patients with acute myeloid leukemia: results of the
tion in adults with AML: long-term analysis of the Acute Leukemia medical research council AML15 trial. J Clin Oncol. 2013;31:3360-
French Association (ALFA) 9000 study. Blood. 2004;104:2467-2474. 3368.
47. Pautas C, Merabet F, Thomas X, et al. Randomized study of intensi- 66. Stelljes M, Beelen DW, Braess J, et al. Allogeneic transplantation as
fied anthracycline doses for induction and recombinant interleukin-2 post-remission therapy for cytogenetically high-risk acute myeloid leu-
for maintenance in patients with acute myeloid leukemia age 50 to 70 kemia: landmark analysis from a single prospective multicenter trial.
years: results of the ALFA-9801 study. J Clin Oncol. 2010;28:808-814. Haematologica. 2011;96:972-979.
48. Fernandez HF, Sun Z, Yao X, et al. Anthracycline dose intensification 67. Horowitz MM, Gale RP, Sondel PM, et al. Graft-versus-leukemia
in acute myeloid leukemia. N Engl J Med. 2009;361:1249-1259. reactions after bone marrow transplantation. Blood. 1990;75:555-562.
49. Lee JH, Kim H, Joo YD, et al; Cooperative Study Group A for Hema- 68. McClune BL, Weisdorf DJ, Pedersen TL, et al. Effect of age on
tology. Prospective randomized comparison of idarubicin and high- outcome of reduced-intensity hematopoietic cell transplantation for
dose daunorubicin in induction chemotherapy for newly diagnosed older patients with acute myeloid leukemia in first complete remis-
acute myeloid leukemia. J Clin Oncol. 2017;35:2754-2763. sion or with myelodysplastic syndrome. J Clin Oncol. 2010;28:
1878-1887.
50. Lowenberg B, Ossenkoppele GJ, van Putten W, et al. High-dose dau-
norubicin in older patients with acute myeloid leukemia. N Engl J 69. Reshef R, Porter DL. Reduced-intensity conditioned allogeneic SCT in
Med. 2009;361:1235-1248. adults with AML. Bone Marrow Transplant. 2015;50:759-769.

Cancer June 15, 2018 2481


INSIGHT from the Experts

70. Jethava YS, Sica S, Savani B, et al. Conditioning regimens for alloge- 87. Blum W, Klisovic RB, Hackanson B, et al. Phase I study of decitabine
neic hematopoietic stem cell transplants in acute myeloid leukemia. alone or in combination with valproic acid in acute myeloid leukemia.
Bone Marrow Transplant. 2017;52:1504-1511. J Clin Oncol. 2007;25:3884-3891.
71. Rashidi A, Ebadi M, Colditz GA, DiPersio JF. Outcomes of allogeneic 88. Cashen AF, Schiller GJ, O’Donnell MR, DiPersio JF. Multicenter, phase
stem cell transplantation in elderly patients with acute myeloid leuke- II study of decitabine for the first-line treatment of older patients with
mia: a systematic review and meta-analysis. Biol Blood Marrow Trans- acute myeloid leukemia. J Clin Oncol. 2010;28:556-561.
plant. 2016;22:651-657.
89. Lubbert M, Ruter BH, Claus R, et al. A multicenter phase II trial of
72. Sorror ML. How I assess comorbidities before hematopoietic cell trans- decitabine as first-line treatment for older patients with acute myeloid
plantation. Blood. 2013;121:2854-2863. leukemia judged unfit for induction chemotherapy. Haematologica.
2012;97:393-401.
73. Burnett AK, Milligan D, Prentice AG, et al. A comparison of low-dose
cytarabine and hydroxyurea with or without all-trans retinoic acid for 90. Blum W, Garzon R, Klisovic RB, et al. Clinical response and miR-29b
acute myeloid leukemia and high-risk myelodysplastic syndrome in predictive significance in older AML patients treated with a 10-day
patients not considered fit for intensive treatment. Cancer. 2007;109: schedule of decitabine. Proc Natl Acad Sci U S A. 2010;107:7473-
1114-1124. 7478.
74. Bross PF, Beitz J, Chen G, et al. Approval summary: gemtuzumab 91. Khan MKH, Garcia-Manero G, Borthakur G, et al. Randomized phase
ozogamicin in relapsed acute myeloid leukemia. Clin Cancer Res. 2001; II trial of two schedules of decitabine as frontline therapy in elderly
7:1490-1496. patients with acute myeloid leukemia ineligible for standard cytotoxic
induction regimens. Blood. 2016;128:1612.
75. Kharfan-Dabaja MA, Hamadani M, Reljic T, et al. Gemtuzumab ozoga-
micin for treatment of newly diagnosed acute myeloid leukaemia: a sys- 92. Welch JS, Petti AA, Miller CA, et al. TP53 and decitabine in acute
tematic review and meta-analysis. Br J Haematol. 2013;163:315-325. myeloid leukemia and myelodysplastic syndromes. N Engl J Med.
2016;375:2023-2036.
76. Amadori S, Suciu S, Selleslag D, et al. Randomized trial of two sched-
ules of low-dose gemtuzumab ozogamicin as induction monotherapy 93. Kantarjian HM, Thomas XG, Dmoszynska A, et al. Multicenter, ran-
for newly diagnosed acute myeloid leukaemia in older patients not domized, open-label, phase III trial of decitabine versus patient choice,
considered candidates for intensive chemotherapy. A phase II study of with physician advice, of either supportive care or low-dose cytarabine
the EORTC and GIMEMA leukaemia groups (AML-19). Br J Haema- for the treatment of older patients with newly diagnosed acute myeloid
tol. 2010;149:376-382. leukemia. J Clin Oncol. 2012;30:2670-2677.

77. Taksin AL, Legrand O, Raffoux E, et al. High efficacy and safety pro- 94. Malik P, Cashen AF. Decitabine in the treatment of acute myeloid
file of fractionated doses of Mylotarg as induction therapy in patients leukemia in elderly patients. Cancer Manag Res. 2014;6:53-61.
with relapsed acute myeloblastic leukemia: a prospective study of the 95. Klco JM, Spencer DH, Lamprecht TL, et al. Genomic impact of tran-
alfa group. Leukemia. 2007;21:66-71. sient low-dose decitabine treatment on primary AML cells. Blood.
78. Amadori S, Suciu S, Selleslag D, et al. Gemtuzumab ozogamicin versus 2013;121:1633-1643.
best supportive care in older patients with newly diagnosed acute mye- 96. Scandura JM, Roboz GJ, Moh M, et al. Phase 1 study of epigenetic
loid leukemia unsuitable for intensive chemotherapy: results of the ran- priming with decitabine prior to standard induction chemotherapy for
domized phase III EORTC-GIMEMA AML-19 Trial. J Clin Oncol. patients with AML. Blood. 2011;118:1472-1480.
2016;34:972-979.
97. Halpern AB, Othus M, Huebner EM, et al. Mitoxantrone, etoposide
79. Burnett AK, Russell NH, Hills RK, et al. Addition of gemtuzumab and cytarabine following epigenetic priming with decitabine in adults
ozogamicin to induction chemotherapy improves survival in older with relapsed/refractory acute myeloid leukemia or other high-grade
patients with acute myeloid leukemia. J Clin Oncol. 2012;30:3924- myeloid neoplasms: a phase 1/2 study. Leukemia. 2017;31:2560-2567.
3931.
98. Qin T, Youssef EM, Jelinek J, et al. Effect of cytarabine and decitabine
80. Christman JK. 5-Azacytidine and 5-aza-20 -deoxycytidine as inhibitors in combination in human leukemic cell lines. Clin Cancer Res. 2007;
of DNA methylation: mechanistic studies and their implications for 13:4225-4232.
cancer therapy. Oncogene. 2002;21:5483-5495.
99. Abdel-Wahab O, Levine RL. Mutations in epigenetic modifiers in the
81. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Azacitidine prolongs pathogenesis and therapy of acute myeloid leukemia. Blood. 2013;121:
overall survival compared with conventional care regimens in elderly 3563-3572.
patients with low bone marrow blast count acute myeloid leukemia.
J Clin Oncol. 2010;28:562-569. 100. Breems DA, Van Putten WL, Huijgens PC, et al. Prognostic index for
adult patients with acute myeloid leukemia in first relapse. J Clin
82. Pleyer L, Stauder R, Burgstaller S, et al. Azacitidine in patients with Oncol. 2005;23:1969-1978.
WHO-defined AML-results of 155 patients from the Austrian Azaciti-
dine Registry of the AGMT-Study Group. J Hematol Oncol. 2013;6: 101. Estey E, Thall P, Andreeff M, et al. Use of granulocyte colony-
32. stimulating factor before, during, and after fludarabine plus cytarabine
induction therapy of newly diagnosed acute myelogenous leukemia or
83. Pleyer L, Burgstaller S, Girschikofsky M, et al. Azacitidine in 302 myelodysplastic syndromes: comparison with fludarabine plus cytara-
patients with WHO-defined acute myeloid leukemia: results from the bine without granulocyte colony-stimulating factor. J Clin Oncol.
Austrian Azacitidine Registry of the AGMT-Study Group. Ann Hema- 1994;12:671-678.
tol. 2014;93:1825-1838.
102. Ho AD, Lipp T, Ehninger G, et al. Combination of mitoxantrone and
84. Dombret H, Seymour JF, Butrym A, et al. International phase 3 study etoposide in refractory acute myelogenous leukemia–an active and
of azacitidine vs conventional care regimens in older patients with well-tolerated regimen. J Clin Oncol. 1988;6:213-217.
newly diagnosed AML with >30% blasts. Blood. 2015;126:291-299.
103. Trifilio SM, Rademaker AW, Newman D, et al. Mitoxantrone and
85. Issa JP, Garcia-Manero G, Giles FJ, et al. Phase 1 study of low-dose etoposide with or without intermediate dose cytarabine for the treat-
prolonged exposure schedules of the hypomethylating agent 5-aza-20 - ment of primary induction failure or relapsed acute myeloid leukemia.
deoxycytidine (decitabine) in hematopoietic malignancies. Blood. 2004; Leuk Res. 2012;36:394-396.
103:1635-1640.
104. Amadori S, Arcese W, Isacchi G, et al. Mitoxantrone, etoposide, and
86. Garcia-Manero G, Kantarjian HM, Sanchez-Gonzalez B, et al. Phase intermediate-dose cytarabine: an effective and tolerable regimen for the
1/2 study of the combination of 5-aza-20 -deoxycytidine with valproic treatment of refractory acute myeloid leukemia. J Clin Oncol. 1991;9:
acid in patients with leukemia. Blood. 2006;108:3271-3279. 1210-1214.

2482 Cancer June 15, 2018


INSIGHT from the Experts

105. Spadea A, Petti MC, Fazi P, et al. Mitoxantrone, etoposide and 116. Stone RM, Mandrekar SJ, Sanford BL, et al. Midostaurin plus chemo-
intermediate-dose Ara-C (MEC): an effective regimen for poor risk therapy for acute myeloid leukemia with a FLT3 mutation. N Engl J
acute myeloid leukemia. Leukemia. 1993;7:549-552. Med. 2017;377:454-464.
106. Jackson G, Taylor P, Smith GM, et al. A multicentre, open, non- 117. Amatangelo MD, Quek L, Shih A, et al. Enasidenib induces acute
comparative phase II study of a combination of fludarabine phosphate, myeloid leukemia cell differentiation to promote clinical response.
cytarabine and granulocyte colony-stimulating factor in relapsed and Blood. 2017;130:732-741.
refractory acute myeloid leukaemia and de novo refractory anaemia
with excess of blasts in transformation. Br J Haematol. 2001;112:127- 118. Stein EM, DiNardo CD, Pollyea DA, et al. Enasidenib in mutant-IDH2
137. relapsed or refractory acute myeloid leukemia. Blood. 2017;130:722-731.

107. Carella AM, Cascavilla N, Greco MM, et al. Treatment of “poor risk” 119. Leukemia and Lymphoma Society. Beat AML Master Trial: informa-
acute myeloid leukemia with fludarabine, cytarabine and G-CSF (flag tion for healthcare professionals. http://www.lls.org/beat-aml/beat-aml-
regimen): a single center study. Leuk Lymphoma. 2001;40:295-303. for-healthcare-professionals. Accessed July 16, 2017.
108. de la Rubia J, Regadera A, Martin G, et al. FLAG-IDA regimen
(fludarabine, cytarabine, idarubicin and G-CSF) in the treatment of
patients with high-risk myeloid malignancies. Leuk Res. 2002;26:725- Author Bios
730. Martha Arellano, MD
109. Pastore D, Specchia G, Carluccio P, et al. FLAG-IDA in the treatment Martha Arellano is an associate
of refractory/relapsed acute myeloid leukemia: single-center experience. professor of hematology and medical
Ann Hematol. 2003;82:231-235. oncology at the Winship Cancer Institute
110. Wrzesien-Kus A, Robak T, Lech-Maranda E, et al; Polish Adult Leuke- of Emory University in Atlanta, Georgia,
mia Group. A multicenter, open, non-comparative, phase II study of where she is a member of the
the combination of cladribine (2-chlorodeoxyadenosine), cytarabine, Discovery and Developmental
and G-CSF as induction therapy in refractory acute myeloid leukemia– Therapeutics Research Program. She is
a report of the Polish Adult Leukemia Group (PALG). Eur J Haema- principal investigator and coinvestigator
tol. 2003;71:155-162. on clinical trials for patients with acute
111. Wierzbowska A, Robak T, Pluta A, et al; Polish Adult Leukemia leukemias and myelodysplastic
Group. Cladribine combined with high doses of arabinoside cytosine, syndromes. She also is program
mitoxantrone, and G-CSF (CLAG-M) is a highly effective salvage regi- director for the Hematology and Medical
men in patients with refractory and relapsed acute myeloid leukemia of Oncology Fellowship Training Program,
the poor risk: a final report of the Polish Adult Leukemia Group. Eur
J Haematol. 2008;80:115-126. and is serving as interim director of the
Division of Hematology.
112. Levis M. Midostaurin approved for FLT3-mutated AML. Blood. 2017;
129:3403-3406.
Jennifer Wilkinson Carlisle, MD
113. Stone RM, DeAngelo DJ, Klimek V, et al. Patients with acute myeloid
leukemia and an activating mutation in FLT3 respond to a small- Jennifer Wilkinson Carlisle is a fellow in
molecule FLT3 tyrosine kinase inhibitor, PKC412. Blood. 2005;105: the Hematology and Medical Oncology
54-60. Fellowship Training Program of Emory
University. She received her medical
114. Langdon WY. FLT3 signaling and the development of inhibitors that
target FLT3 kinase activity. Crit Rev Oncog. 2012;17:199-209. degree from the University of Florida
College of Medicine and her internal
115. Levis M, Ravandi F, Wang ES, et al. Results from a randomized medicine training at Washington
trial of salvage chemotherapy followed by lestaurtinib for patients
University in St. Louis in St. Louis,
with FLT3 mutant AML in first relapse. Blood. 2011;117:3294-
3301. Missouri.

Cancer June 15, 2018 2483

You might also like