You are on page 1of 24

Received: 18 June 2018 Revised: 26 June 2018 Accepted: 10 July 2018

DOI: 10.1002/ajh.25214

ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL


MALIGNANCIES: A CONTINUING MEDICAL EDUCATION SERIES

Acute myeloid leukemia: 2019 update on risk-stratification and


management
Elihu H. Estey

Division of Hematology, Clinical Research

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


Division, Fred Hutchinson Cancer Research Abstract
Center, University of Washington and Outcome in patients with acute myeloid leukemia (AML) ranges from death within a few days of
Member, Seattle, Washington
beginning treatment (treatment related mortality, TRM) to likely cure. The major reason patients
Correspondence
are not cured is resistance to treatment, often manifested as relapse from remission, rather than,
Elihu H. Estey, Division of Hematology, Clinical
Research Division, Fred Hutchinson Cancer even in older patients, TRM, whose incidence is decreasing. Knowledge of the pre-treatment
Research Center, University of Washington mutation status of various genes has improved our ability to assign initial treatment and, of par-
and Member, Seattle, Washington. ticular importance, knowledge of whether patients ostensibly in remission have measurable
Email: eestey@u.washington.edu
residual disease should influence subsequent management. Several new drugs have been
approved by the FDA and we discuss their role in treatment.

1 | DIAGNOSIS described below are more important in determining outcome than the
AML-MDS distinction,4 some centers treat patients with >10% blasts
Acute myeloid leukemia (AML) results from accumulation of abnormal as AML, even if they are considered as excess blasts 2 by the WHO.2
myeloblasts, most commonly in the bone marrow, leading to bone Biologically, patterns of mutations in AML arising after a preceding
marrow failure and death. Peripheral blood involvement is frequent, hematologic disorder such as myelodysplasia often bear more resem-
while infiltration of organs, most ominously the brain and/or lung is blance to those seen in MDS than in primary (de novo) AML. 5,6
rare and seen most often in patients with high blood blast counts (eg,
>50 000/μL). Granulocytic sarcoma (GS) refers to AML seemingly lim-
ited to sites outside marrow or blood. Because it progresses to mar- 2 | DISEASE OVERVIEW
row involvement usually within 1 year, GS should generally be
By age 70, about 10% of presumably normal individuals have clonal
managed like AML with marrow involvement.1
The 2016 World Health Organization's (WHO) criterion for AML mutations in hematopoietic cells.7 Such “age-related clonal hemato-

is at least 20% myeloblasts in marrow (or blood) with myeloid lineage poiesis” (ARCH) predisposes to acquisition of other mutations.6 Some

established by multiparameter flow cytometry (MFC).2 Exceptions to involved in RNA splicing (SRSF2), DNA methylation (DNMT3a, TET2,

the ≥20% criterion are cases of CBF AML (cytogenetic abnormalities t IDH 1/2), chromatin modification (ASXL1), or the cohesion complex

[8;21], inv [16], or t[16;16]), NPM1 mutated AML, or acute promyelo- (STAG2) are associated with development of MDS. Subsequent gain

cytic leukemia; in each the diagnosis of AML is independent of blast of mutations in genes encoding myeloid transcription factors (RUNX1,

%. Occasionally myeloid blasts may have T- or B-cell markers, or sepa- CEBPA) or signal transduction proteins (FLT3) leads to development
rate myeloid and lymphoid populations may exist. These cases are of AML “secondary” to MDS (s-AML).6 In contrast, patients with pri-
referred to as “mixed phenotype acute leukemia.”2 It is unclear mary (“de novo”) AML often have RUNX1, CEBPA, FLT3, or MLL
whether they should be managed as AML, ALL, or both and manage- mutations but not mutations associated with prior MDS.6 Patients
ment may depend on the degree to which the myeloid or lymphoid with AML after receipt of prior chemotherapy (t-AML) can show
component predominates. Cases with >20% blasts but which lack either the s-AML or the de novo pattern of mutations; those exhibit-
markers are referred to as acute undifferentiated leukemia and often ing the de novo pattern have the relatively favorable outcome seen in
managed like AML. de novo AML while those showing the secondary pattern fare as simi-
The 20% blast cut-off used to distinguish “AML” from “MDS” is larly poorly as s-AML. 7 Perhaps, 1/3 of patients believed to have de
arbitrary. Based on data suggesting the prognostic covariates novo AML based on history will exhibit genetic mutations that are

Am J Hematol. 2018;93:1267–1291. wileyonlinelibrary.com/journal/ajh © 2018 Wiley Periodicals, Inc. 1267

/
1268 ESTEY

95% specific for s-AML; despite a clinical diagnosis of de novo AML, 1 year if given azacytidine rather than BSC, 45% if given azacytidine
these patients appear to have a similar outcome as s-AML.7 rather than LDAC and 55% if given azacytidine rather than 7 + 3
Mutations present in hematopoietic cells at birth are germline The azacitidine13 and Swedish data 11,12 indicate there is less rea-
mutations (ie, present at birth in all cells).8,9 Germline mutations often son for the therapeutic nihilism previously exhibited toward at least
involve RUNX1, GATA2, and DDX41 and characterize hereditary some older patients,10 although the need for better therapy in these

myeloid malignancy syndromes (HHMS). These frequently culminate patients is undeniable. However, it is also undeniable that, as dis-
cussed below, despite our current emphasis on age as the principal
in MDS/AML, which, although usually presenting in childhood, can be
consideration in management of AML, age is not the most important
seen in adults usually age <40 years but occasionally older. Screening
determinant of either “treatment-related mortality” (TRM)
of families for germline mutations is advisable if a family member is a
or resistance to therapy, defined as failure to achieve complete remis-
potential donor for allogeneic hematopoietic cell transplant (allo-
sion (CR) despite not incurring TRM, or, more commonly, relapse.
HCT), or if a patient has a potentially germline mutation (eg, RUNX1,
GATA2, and DDX41), HHMS organ manifestations, or a suggestive
3.1 | Factors associated with TRM
family history 8,9

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


Distinguishing TRM from resistance seems useful because different
strategies might be used in patients at high risk of TRM vs those at
3 | PRETREATMENT RISK STRATIFICATION high risk of resistance. Because the time needed to attain CR may vary
according to the characteristics of a patient's AML and its treatment,
Patients with AML are primarily considered, for example in clinical tri- there will necessarily be overlap between TRM and resistance. None-
als or by practicing hematologists/oncologists, as “younger” or “older.” theless death within 28-30 days of beginning induction is a frequently
Age 60-65 years commonly divides the two groups, with 65-70 year used criterion for TRM in patients given 7 + 3. There is a sharp reduc-
being median age of patients at diagnosis. Medicare records from tion in weekly death rates once 4 weeks have elapsed from start of
2000 to 2009 indicated only 40% of patients age >65 years with AML such therapy or therapy containing higher doses of cytarabine and
received chemotherapy within 3 months of diagnosis. 10 Patients who purine analogs such as fludarabine, suggesting patients who die in this
were treated lived longer. 10 While this may have reflected treated time frame are qualitatively different from those who do not.14 Simi-
patients simply had better inherent prognoses, the Swedish Acute larly, suggestive are data that different factors are associated with
Leukemia Registry observed lower death rates in 864 patients aged TRM thus defined and resistance.
70-79 if they resided in regions of the country where a higher propor- Recent years have seen declines in TRM following the “intensive”
tion of patients received 7 + 3 rather than “best supportive care,”,11; therapy noted in the preceding paragraph. 15 These declines likely
there were no regional differences in performance status and 7 + 3 reflecting better supportive care, for example prophylactic use of
was superior regardless of performance status, although other poten- posaconazole during induction, may underlie the improvements in sur-
tially confounding covariates were not be accounted for (nor was vival noted in the Swedish Registry.16 TRM rates fell from 18% in
quality of life). About 25% of 1337 patients alive at least 1 year after SWOG and 16% at MD Anderson (MDA) in 1991-1995 to 3% at
being diagnosed between 1997 and 2013 were age >64 years and SWOG and 4% at MDA in 2006-2009. This decrease was indepen-
25% of these were age >68 in 2014.12 Examining the period dent of age, PS, and measurements of organ function. Although selec-
1973-2011, the largest improvements in survival relative to a compa- tion bias such that older patients who would have received intensive
rable group from the general population (relative survival ratio) therapy in the earlier period and often received less intense therapies
occurred in patients age <60 until 1997 but thereafter occurred in (eg, azacytidine) in the more recent period hence would not have been
patients aged 61-70 although a 65-year old still lost 2/3 s of her/his included in the analysis may have been responsible, this would not
projected life expectancy even if diagnosed in 2011, and improve- explain the decline in TRM in younger patients.
ments were not seen in individuals over age 70. 12 Because several factors, primarily PS but also creatinine, albumin,
There remains a tendency even in fitter older patients to offer and blood counts, determine risk of TRM multivariate models, includ-
less rather than more “intense” therapy, with the former represented ing some available online, have been developed to predict this out-
by the “hypomethylating agents' (HMAs) azacytidine or decitabine. come.3 Removing age from these models has only minimal effects on
Dombret at al. asked physicians whether they would choose “best their predictive ability suggesting age is a surrogate for other TRM
supportive care” only (BSC), low dose cytarabine (LDAC), or “intense covariates.14 Hence current European Leukemia-Net (ELN) and
00
chemotherapy” (IC), that is, conventional “7 + 3 for each of National Comprehensive Cancer Network guidelines recommend age
488 patients age >65 with >30% blasts. 13 Patients were then random- be considered only in the context of other covariates when deciding
ized to the preselected “conventional care regimen” (CCR) or azacyti- to whether to recommend more vs less intensive induction ther-
dine. Although 75% of patients had performance score (PS) 0-1 and apy. 17,18 In the case of conventional intense induction regimens, these
none PS 3-4, only 18% were preselected to receive IC vs 64% for low covariates would include those primarily associated with resistance
dose cytarabine and 18% for BSC, documenting a predilection for less (see below) as the more likely the risk of resistance the less willingness
intense therapy. Survival with azacytidine was superior to that with there would be to accept a given risk of TRM.
CCR, although statistically essentially due to the comparison between Since 2016 version of this update, it is becoming clearer comor-
azacytidine and BSC or LDAC and relatively modest (30% alive at bidities, site of treatment, cohabitation status, and educational level

/
ESTEY 1269

may influence survival, perhaps largely through TRM. In a validation with the possibility of TRM than of resistance. However even in
set of 367 patients Sorror et al. noted the importance of comorbidities patients in their 70s and 80s resistance is often the greater problem,
as described in the hematopoietic cell transplantation comorbidity at least in patients reported in the literature. Considering studies con-
index (HCT-CI, see below) in forecasting survival rates at 8 weeks ducted before 2000 Appelbaum et al. found that in patients aged
19
after treatment initiation. Examining 7007 patients in the California >75 years, rates of not entering CR despite surviving the first 30 days
Cancer Registry Ho et al. found 25% had been treated at a National of induction therapy were similar with those of death by day 3023; the
Cancer Institute designated Cancer Center (NCI-CC, defined as a cen- latter rates are likely to have fallen more than the former in more
ter which treated a median of 13 AML patients annually) and 75% at a recent years.15 Even in patients aged >70 years with performance sta-
non-NCI-CC that treated at least one person with AML annually tus 2-4 at time of CR, the risk of relapse is threefold that of death in
(median 2). Sixty-day mortality was 12% at an NCI-CC and 24% at a CR, 24 the distinction between TRM (death in remission) and resistance
non-NCI-CC. Age <65-years, higher socio-economic status, fewer perhaps being more objective in patients in remission than in patients
comorbidities, and public health insurance were associated with treat- undergoing induction.
ment in an NCI-CC. However, after accounting for these covariates, Genetics encompassing both classical cytogenetics and the muta-
treatment at an NCI-CC remained associated with lower 60-day mor-

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


tional status of various genes is the most important predictor of resis-
tality rates (odds ratio 0.46, 95% CI 0.38-0.57).20 Data from 3243 tance18 (Figure 1, Table 1), that is, failure to enter CR despite not
patients in the Danish National Acute Leukemia Registry (DNLR) sug-
incurring TRM or relapse from remission. Much of the prognostic
gest patients aged >60 years are more likely to receive intensive
import of “de novo” vs “secondary” AML is subsumed by genetics thus
induction and have better survival when cohabitating rather than liv-
defined.5 The ELN guidelines, updated in 2017, are probably the most
ing alone, independent of PS, comorbidities, and cytogenetic risk. 21
widely-used source for assessing risk of resistance and classify
Since median follow-up was only about 6 months, it is possible a fair
patients into “favorable,” “intermediate,” and “adverse” groups based
number of deaths reflected TRM, which is often most common during
on leukemia cell cytogenetics and mutations18 (Table 2). The single
initial induction. The same group reported an association between
most adverse factor is a TP53 mutation, commonly associated with
higher education levels and better survival in 1588 DNLR patients
complex cytogenetics, but adding to the negative effect of the latter.
(median age about 60 years) also independent of PS, comorbidities,
The favorable effect of NPM1 and (bi-allelic) CEBPA mutations are
and cytogenetic risk 22; the longer follow-up (median about 1 year)
considered unaffected by cytogenetic status.18 A FLT3 internal tan-
makes it more difficult to distinguish TRM from resistance.
dem duplication (ITD) is regarded as unfavorable only if the ratio of
mutated to normal alleles (allelic ratio, AR) is >0.5. 18
3.2 | Factors associated with resistance While Harada et al. found the ELN 2017 effectively stratified
Experience suggests patients, perhaps because of the negative conno- Japan Acute Leukemia Study Group patients (n = 197, 90% age < 60),
tations associated with “chemotherapy,” are often more concerned with the exception of the FLT3 AR cut-point25 Eisfeld et al. found

FIGURE 1 Distribution of genetic aberrations in AML (see reference 18)

/
1270 ESTEY

TABLE 1 Proposed genomic classification of AML (see reference 29) TABLE 2 2017 ELN risk stratification by genetics (see reference 18)

Frequency in the Risk


*
Study Cohort category Genetic abnormality
(N = 1540) Most Frequently Favorable t(8;21)(q22;q22.1); RUNX1-RUNX1T1
No. of Mutated Genes
Genomic Subgroup patients (%) Gene (%) inv(16)(p13.1q22) or t(16;16)(p13.1;q22); CBFB-MYH11
AML with NPM1 418 (27) NPM1 (100), DNMT3A Mutated NPM1 without FLT3-ITD or with
mutation (54), FLT3 ITD (39), FLT3-ITDlow = allelic r atio < 0.5
NRAS (19), Biallelic mutated CEBPA
TET2 (16), PTPN11 (15)
Intermediate Mutated NPM1 and FLT3 -ITD high = allelic ratio > 0.5
AML with mutated 275 (18) RUNX1 (39), MLLPTD
chromatin, (25), SRSF2 (22), Wild-type NPM1 without FLT3-ITD or with
RNA-splicing genes, DNMT3A (20), FLT3-ITDlow (without adverse-risk genetic lesions)
or both† ASXL1 (17), STAG2 (16), t(9;11)(p21.3;q23.3); MLLT3-KMT2A
NRAS (16), TET2 (15),
FLT3ITD (15) Cytogenetic abnormalities not classified as favorable or
adverse
AML with TP53 199 (13) Complex karyotype
Adverse t(6;9)(p23;q34.1); DEK-NUP214

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


mutations, (68), −5/5q (47),
chromosomal −7/7q (44), t(v;11q23.3); KMT2A rearranged
aneuploidy, or both‡ TP53 (44), −17/17p
t(9;22)(q34.1;q11.2); BCR-ABL1
(31), −12/12p (17),
+8/8q (16) inv(3)(q21.3q26.2) or t(3;3)(q21.3;q26.2);
81 (5) inv(16) (100), NRAS (53), GATA2,MECOM(EVI1)
AML with inv(16)
(p13.1q22) or t +8/8q (16), +22 (16), −5 or del(5q); − 7; −17/abn(17p)
(16;16)(p13.1;q22); KIT (15), FLT3TKD (15)
Complex karyotype monosomal karyotype
CBFB–MYH11
Wild-type NPM1 and FLT3 -ITD high†
AML with biallelic 66 (4) CEBPAbiallelic (100),
CEBPA mutations NRAS (30), WT1 (21), Mutated RUNX1
GATA2 (20) Mutated ASXL1
AML with t(15;17) 60 (4) t(15;17) (100), FLT3ITD Mutated TP53
(q22;q12); PML– (35), WT1 (17)
RARA
AML with t(8;21)(q22; 60 (4) t(8;21) (100), KIT (38), “next generation sequencing” (NGS) to assess mutations in 80 cancer
q22); RUNX1– −Y (33), −9q (18)
and/or leukemia related genes. 26 Patients with NPM1 mutation con-
RUNX1T1
stituted a “favorable” group but only if the NPM1 mutation was
AML with MLL fusion 44 (3) t(x;11q23) (100), NRAS
genes; t(x;11)(x; (23) accompanied by a mutation in a variety of other genes; this favorable
q23)§
group had an 81% CR rate, with 3-year survival and disease-free sur-
AML with inv(3) 20 (1) inv(3) (100), −7 (85),
vival rates of 45-46% (Figure 3). Patients in this favorable group were
(q21q26.2) or t(3;3) KRAS (30), NRAS (30),
(q21;q26.2); GATA2, PTPN11 (30), ETV6 (15), uniformly also classified as favorable by ELN 2017 and constituted
MECOM(EVI1) PHF6 (15), SF3B1 (15) about 1/3 of ELN 2017 favorable patients and about 10% of all the
AML with IDH2 R172 18 (1) IDH2R172 (100), authors' patients. Ostronoff et al. have likewise noted patients aged
mutations and no DNMT3A (67), +8/8q
other class-defining (17) >65 years in the ELN favorable group NPM1 mutated/FLT3 normal
lesions have much poorer outcomes than younger adults27 while Patel
ITD
AML with t(6;9)(p23; 15 (1) t(6;9) (100), FLT3 et al. have reported higher NPM1 mutation burden (higher proportion
q34); DEK–NUP214 (80), KRAS (20)
of mutated/normal alleles, that is, “variant allele frequency”) was asso-
AML with driver 166 (11) FLT3ITD (39), DNMT3A
mutations but no (16) ciated with shorter EFS and survival as were DNMT3a mutations in
detected 109 patients with de novo AML and mutated NPM1. 28
class-defining
lesions Although Eisfeld et al. ‘s26 and Patel et al.'s 28 findings await con-

AML with no detected 62 (4) firmation in an independent population, data from Papaemmanuil
driver mutations et al. also speak to the potential value of examining a broader range of
AML meeting criteria 56 (4) genes than indicated as prognostic in ELN 2017.29 In particular, after
for ≥2 genomic
examining survival in 1540 patients treated on one of three separate
subgroups
trials of intensive induction  allogeneic HCT and validating findings
using sophisticated statistical techniques, these authors stressed the
ELN 2017 was problematic in 423 patients age >60 years with de importance of gene-gene interactions, and similarly to Eisfeld et al.26
26
novo AML treated on several Alliance protocols ; no patients the mutation status of other genes in patients with NPM1 mutations.
received a transplant in CR1. ELN favorable risk patients had longer Thus, the negative effect of a FLT3 ITD in patients with an NPM1
survival and relapse-free survival than ELN intermediate and adverse mutation was much more pronounced in patients with than without a
risk patients, but the latter two groups were essentially indistinguish- DNMT3a mutation (Figure 4). Likewise, in patients with an NPM1
able (Figure 2). Moreover, at 3 years, disease(relapse)-free survival in mutation presence of a RAS mutation improved survival more in the
the “favorable” group was only 25% and survival 30% (Figure 2). To presence than the absence of a DNMT3a mutation (Figure 4). Analo-
identify patients who might have better outcomes the authors used gously, the adverse effect of an MLL aberration noted in ELN 2017

/
ESTEY 1285

The use of MRD assessment in patients ostensibly in CR has plau- principal factor most consistently associated with response to salvage
sibly made auto HCT a more viable option. Specifically, patients in the therapy has been duration of prior CR. Although various cut-points
ELN 2017 intermediate group pretreatment who become MRD nega- have been used to define lower vs shorter CR1 durations, it is likely
tive after induction therapy might be appropriate candidates for auto the shorter the CR1 duration, assigning primary refractory patients a
HCT even if fit for allo HCT in the expectation auto might avoid some CR1 duration of zero, the poorer the outcome.132 Several systems
of the long-term complications of allo. which combine CR1 duration with other factors are available for
assessing prognosis. 133 A convenient one, derived from 667 adults
aged <60 in first relapse, remains Breems, et al.134 (Table 6) Survival
6 | THERAPY OF RELAPSED AND probabilities at 1-year were 70% in the best group, (46% at 5 years),
R E F R A C T O R Y ( R / R ) A M L (“ S A L V A G E
which however constituted only 9% of patients, with a mean CR1
THERAPY”)
duration of 17 months), and were 46% in the intermediate group
(25% of patients, mean CR1 duration 16 months) and 16% in the
6.1 | Definitions and assessing prognosis with worst group (2/3 s of patients, mean CR1 duration 7 months)
standard therapy

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


(Supporting Information Figure 8). CR rates were 85%,60%, and 34%

Not infrequently AML is considered “primary refractory” if CR is not respectively among patients in the “favorable”, intermediate, and
observed after a first course of 7 + 3. Examining 1505 adults given adverse groups who received treatment. Although reasons underlying
7 + 3 on one of 4 SWOG protocols Othus et al.130 found 48% initial therapeutic choices were difficult to ascertain and therapy-
achieved CR on course 1 and 9% died, leaving 638 people alive but specific CR rates were accordingly not provided, these results suggest
not in CR. Although the protocols called for administration of a 2nd patients in the worst group (alternatively those with CR1 durations
7 + 3, this occurred in only 333 (52%). A similar proportion has been <1 year) are unlikely to benefit from standard intensive therapy (eg,
noted by ECOG. Examining various pretreatment factors, such as age, FLAG-ida), thus becoming candidates for “clinical trials.” In contrast
performance status, and cytogenetics, and post-treatment factors, standard therapy might be more appropriate in the best and interme-
such as results of a “day 14” marrow alone or compared to pretreat- diate groups. In general, however repetition at relapse of the same
ment, administration of a second course was associated only with induction and post-remission therapy used initially leads to worse
treatment at a “non-academic” SWOG center. The CR rate on a 2nd results. This is consistent with the notion that induction chemother-
7 + 3 was 43% with a TRM rate of 10%. None of the covariates noted apy exerts selective pressure such that new potentially resistant leu-
above nor the day a second course began influenced 2nd course CR kemic clones emerge as do non-leukemic clones harboring similar
rate. Although patients in CR only after two courses had shorter RFS mutations as seen in age-related clonal hematopoiesis. 135 This sug-
and survival, this reflected older age and cytogenetics, rather than gests that even should 2nd CR be attained with standard induction,
courses to CR. These data prompted ELN to define refractory AML as post-remission therapy should optimally include allo HCT, particularly
having failed two courses of 7 + 3,18 although the relation between if this has not been done before; indeed, regardless of prognostic
number of courses to CR and subsequent outcomes is not definitively group patients lived longer if in CR2 they received allo HCT, although,
established. Whether patients whose AML has failed to respond to a as usual, various selection biases may have affected the decision to
first 7 + 3 would be better served by a second 7 + 3 rather than perform allo HCT.134 The recommendation for investigational therapy
change to alternative therapy can only answered via a randomized
trial. However, the data suggest the minimum acceptable CR rate for TABLE 6 Prognostic scoring system for AML in first relapse
studies investigating new therapies in AML refractory to a first 7 + 3 “favorable” group 1-6 points, intermediate group 7-9 points, adverse
group 10-14 points (see reference 134)
should be 40%-45% rather than the typical 25%-30%; use of the latter
results in people continuing to receive the new therapy when it is Factor Points

already known likely to be worse than a second 7 + 3. Whether no CR1 duration

CR after two courses should also be the criterion for primary refrac- ≥18 mo 0

tory AML after therapy with high-dose cytarabine-containing regi- 7-18 mo 3

mens is less clear 131 ≤6 months 5

It can be difficult to distinguish relapse of the original AML from a Cytogenetics at diagnosis
t(16;16) or inv16 0
“new” AML brought on for example by chemotherapy, with the latter
t(8;21) 3
suggested by findings of a substantively new cytogenetic or muta-
tional profile. The proportion of relapsed AMLs that truly represent Other 5
Age at relapse
“new” disease is unknown and depends on what is meant by
≤35 y 0
“substantively.”
36-45 y 1
The fundamental decision to be made with salvage therapy, as
>45 y 2
with initial therapy, is the choice between standard (eg, FLAG or
SCT before first relapse
FLAG-ida) and investigational re-induction therapies. Again, since the
No 0
results with the latter are, by definition, unknown the decision must
Yes 2
rest on degree of dissatisfaction with the former. Probably the

/
ESTEY 1271

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


FIGURE 2 Assessment of ELN 2017 risk stratification system in patients aged 60 or above (see reference 26)

depended on the presence of a mutation in the tyrosine kinase suggests however the same factors that influence response to one
domain of the FLT3 gene and although ELN 2017 regarded mutations treatment do so with another. For example, the presence of complex
in IDH2 or in DNMT3a as having no prognostic effect, prognosis cytogenetics, TP53 mutations, or high AR FLT3 ITDs are associated
became considerably worse when both IDH2 and DNMT3a mutations with relapse with both allogeneic HCT and treatment without
were present (Figure 4). It cannot be overemphasized that the prog- allogeneic HCT.
nostic effect of a given mutation almost always depends on what Although many prognostic systems focus on survival as the out-
other mutations are present. come of interest, survival is likely confounded by treatment given
The ongoing replacement of single gene assays by NGS to assess after an initial treatment fails more so than shorter term outcomes
a panel of genes, and possibly in the future by exome sequencing and such as event-free survival.36 The relative limited prognostic ability of
genome wide assays30 will facilitate development of more sophisti- even sophisticated models cannot be overemphasized. For example,
cated prognostic systems than ELN 2017; for example, the seemingly Papaemmanuil et al developed a model incorporating cytogenetic,
straightforward entity of CBF AML is mutationally complex and this genomic, and clinical data (the latter including age, performance sta-
complexity has prognostic implications. 31,32
AML is conventionally tus, blood counts, treatment protocol, and year of treatment) and
regarded as an indication for immediate treatment, raising the ques- evaluated how often the model correctly predicted which one of a
tion of whether treatment can be delayed even the usually 2-7 days pair of patients had longer survival.29 Iterative repetitions over many
needed to receive NGS results and potentially using them to decide pairs indicated the model's concordance with survival was only 71%
on therapy. Both Sekeres et al. (in older patients) 33
and Bertoli et al. in with genomic features accounting for about 2/3 s of the explained
younger and older patients including those with WBC >50 000/μL) 34 variation and clinical, demographic, and treatment factors for 1/3.
concluded delay of therapy had no effect on outcome after account- Various other models perform similarly. 37 However, it is likely incor-
ing for other prognostic covariates; the deleterious effect on outcome poration of post-treatment data with the pretreatment data discussed
in younger patients in Sekeres et al. was relatively small and has to be above will improve prognostic accuracy; we will return to this topic

considered in context of potential benefit. Median time from diagnosis after a discussion of induction therapy.

to beginning therapy, that is, delay, was only 4-8 days and the effect
of potential selection bias cannot be quantified. Although a trial ran-
4 | INITIAL INDUCTION THERAPY
domizing patients to immediate treatment vs waiting until NGS results
are available is likely impossible, it seems any potential harm resulting
from delay might well be less than the harm in immediately beginning 4.1 | Goal of induction therapy
a therapy that is unlikely to be effective, for example, 7 + 3 in subjects A case can be made this should be CR without measurable residual
with TP53 mutations. disease (MRD). After accounting for de novo vs secondary disease,
It is useful to distinguish between a prognostic and a predictive cytogenetics, age, blood counts, and the longer time needed to
covariate. The former refers to a covariate such as PS 3-4 or a TP53 achieve CR than other responses (“survival by response bias”), Othus
mutation35 that likely applies regardless of specific treatment (eg, et al. noted a survival advantage for CR rather than CR with incom-
7 + 3 or azacitidine), the latter to a covariate that is therapy specific. plete count recovery (CRi) in 261 patents aged <60 years given 7 + 3
Many models regard treatment as a single entity and do not examine or more intense therapy and in 133 patients (median age 73) given
whether the effect of a covariate differs according to treatment, rec- azacytidine + gemtuzumab ozogamicin (GO) on SWOG studies. The
ognizing most of the prognostic effect of various mutations has been effect of CR was on survival was similar with azacitidine as with more
worked out in subjects given intensive therapy Experience to date intense regimens.38 Examining rates of death at 6 month intervals in

/
1272 ESTEY

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


FIGURE 3 Modification of ELN 2017 risk stratification in patients aged 60 or above (see reference 26)

patients treated with 7 + 3 or more intense regimens since 2005, without MRD as superior to CR because of the former's associations
Othus et al. noted these were constant during years 1-2 after treat- with better long-term outcome,18 as discussed below.
ment began but fell by half in year 3, leading them to define “poten-
tially cured” patients as those alive 2-3 years after treatment began, 4.2 | Candidates for clinical trials of new induction
although subsequent death rates in those alive at these times were
therapy
still considerably higher than expected for an age-matched normal
It should be stressed there are some patients in whom the risk of
population.39 The primary predictor of being alive at 3 years was
achievement of CR by day 100.39 Many clinicians also believe “quality TRM is not commensurate with the benefit of achievement of CR 

of life” is better in people who achieve CR because they are likely to MRD and some in whom the likelihood of achievement of CR  MRD

receive fewer transfusions and spend less time in hospital than people with conventional therapy is so low that even a relatively low risk of
who achieve CRi or hematologic improvement, although this seems an TRM would be difficult to justify. As indicated above, age alone should
important area for further investigation. ELN 2017 distinguished CR not be the sole or even principal means to identify such patients18 in

/
ESTEY 1273

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].

FIGURE 4 Interactions between different mutations affecting prognosis (see reference 29)

contrast to covariates such as PS 3-4 or comorbidities for high risk of cytogenetics, a monosomal karyotype, or a TP53 mutation, the proba-
TRM or such as ELN 2017 adverse risk for low probability of entering bility of CR with 7 + 3 is so slight that investigational therapies are
(and staying) in CR. Both groups are prime candidates for clinical trials, advisable once AML is diagnosed. Given the importance of CR with-
the former with “less intense” therapies, although patients at high risk out MRD in determining post induction outcome, it is likely that as
of TRM with conventional therapies are often excluded from such tri- rates of CR without MRD (rather than merely of CR) become more
als. 40
Patients with ELN 2017 adverse risk might plausibly receive widely known, other groups in whom this rate is <50% will become
conventional induction with 7 + 3 with the goal of allogeneic HCT in prima facie candidates for clinical trials of new induction therapies.
CR1. However, in some such patients, particularly those with complex Even today National Comprehensive Cancer Network guidelines

/
1274 ESTEY

recommend consideration of trials for all patients.17 Indeed, survival in receiving HCT in CR1 was similar (about 48% among arms), there
the great majority of patients with the exception of ELN favorable were no differences in EFS, DFS, or survival (Supporting Information
patients age <60-65 is such that clinical trials should spring to mind Figure 1A). The same was true considering NPM1, CEBPA, FLT3 ITD
for all but such patients once AML is diagnosed. status, or cytogenetic status with the exception that favorable-risk
patients appeared to have better EFS, RFS, and survival with 7 + 3
(Supporting Information Figure 1B). However, patients given IA or IAV
4.3 | Intensity of induction therapy
received less cytarabine once in CR than patients given 7 + 3, and it is
Several studies have noted superior survival, or at least similar survival plausible results might have been different had fludarabine + HDAC +
with less toxicity/inconvenience, with conventional lower intensity idarubicin (FLAG-ida) rather than 7 + 3 been used, given results from
regimens (eg, azacytidine or decitabine) than with higher intensity reg- a large randomized NCRI study in the UK that found reduced relapse
imens (eg, those containing “high-dose” cytarabine [HDAC], defined rates with FLAG-ida than with 10 days of standard cytarabine + dau-
as individual doses at least 1 g/m2).41,42 In contrast in a multicenter norubicin.49 However, there were no differences in survival possibly
study Sorror et al.43 reported longer 2 year survival rates in those of because of higher treatment-related mortality rate in CR in the FLAG-
1295 patients given more intense (typically 7 + 3 or HDAC- contain- ida arm. Further complicating the issue is the very realistic possibility

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


ing) rather than more intense (typically azacitdine) induction regimens, a given result may not be generally applicable. For example, although
even if patients were aged 70-79 years and regardless of AML com- a 1205 patient randomized NCRI trial found no difference, other than
19
posite model or treatment-related mortality model 14 scores43; as more TRM in the higher dose arm, between 7 + 3 using daunorubicin
expected the proportion of patients given more intense therapy varied doses of 90 mg/m 2 vs the usual 60 mg/m2 each daily X 3,50 patients
by age: 96% if 50-59 (n = 264), 81% if 60-69 (n = 421), 41% if age with FLT3 ITD (n = 200) had less relapse leading to longer survival if
70-79 (n = 242), and 20% if age 80-92 (n = 62).However, the differ- given 90 mg/m.51 Likewise, a HOVON trial randomizing 795 evaluable
ences in survival have been slight (eg, 7 vs 5 months median in Boddu adults age <65 between 7 + 3 as cycle 1 followed by amsacrine (simi-
et al.41 ) and even when larger (eg, 53% vs 32% and 37% vs 12% at lar to idarubicin or daunorubicin) + HDAC as cycle 2 each
2 years in those with AML- CM scores 4-6 and 7-9, respectively, in clofarabine (a purine analog similar to fludarabine except with
Sorror et al.43) have nonetheless been clearly unsatisfactory in the greater single agent activity at tolerable doses) found longer EFS and
worst prognosis patients (22% vs 11% if AML-CM score ≥10). All survival with addition of clofarabine but only in the ELN 2010
these studies have been historically controlled, problematic given limi- intermediate-1 group (EFS at 4-years 40% vs 26%, P = .002 and in
tations in evaluating the likelihood of response if patients given a cur- subjects who were NPM1 mutated but FLT3 wild type (EFS at 4-years
rent therapy had received a historical one instead.37 Although 40% vs 18%, P < .001).52
randomized, the Dombret at al. azacytidine vs CCR trial noted above Given these data, it is difficult to be dogmatic about intensity of
enrolled relatively few patients in the 7 + 3 vs azacytidine arm.13 An induction therapy, with the possible exception that regimens similar in
ongoing EORTC trial (NCT02172872, “inDACtion”) is randomizing intensity to 7 + 3 or FLAG-ida be used rather than azacytidine or deci-
patients aged ≥60 years with PS 0-2 and normal kidney/liver function tabine in people at low risk of TRM. At the author's center, we have
between standard 7 + 3 induction and 10-day decitabine followed by used G-CLAM (in which, because of its greater single agent activity)
allogeneic HCT in responding patients; estimated completion date is cladribine replaces the fludarabine of FLAG-ida) provided patients
December, 2019. While is seems clear TP53 mutations and a monoso- have a low TRM score regardless of age. However, although results
mal karyotype, if not adverse cytogenetics per se, predict poor out- with GCLAM seem, in the absence of randomization, at least similar to
come with both azacytidine and more intense therapies13,18,35 it is 7 + 3, the rate of CR without MRD is only 71%. 53 Hence, we are
less clear if other genetic features can distinguish patients more likely interested in combining GCLAM with potentially less toxic agents that
to do better with one of these than the other44; for example, it has might improve efficacy. Whether this becomes reality is probably
been reported patients with NPM1 mutations have shorter remissions more important than the intensity of induction therapy. Addition of
after therapy with azacitidine or decitabine.45 Suggestively, after some agents to 7 + 3 or FLAG-ida has already improved outcome,
accounting for prognostic covariates, failure to respond to azacytidine making these agents an integral part of induction therapy, as
remains highly associated with failure to respond to 7 + 3,46 suggest- described below and as reviewed in Wei and Tiong.54
ing these two therapies have much in common and more important
than assessing which is better may be efforts to improve either
4.4 | Midostaurin and FLT3 inhibition
or both.
Regarding 7 + 3 vs more intensive therapy, a German randomized FLT 3 ITDs occur in about 25% of patients aged <60 years and in
comparison involving 3375 adults found equivalent EFS and RFS with about 15% of older patients.55 Mutations in the tyrosine kinase
47
7 + 3 and high-dose cytarabine-containing therapy. More recently, domain of the gene occur in an additional 5%-10%. Midostaurin is an
Garcia-Manero et al. in SWOG1203 randomized 738 adults aged oral multitargeted tyrosine kinase inhibitor active in patients with a
<60 years among 7 + 3 (daunorubicin dose 90 mg/m2 daily X3) idaru- FLT3 mutation. Stone et. al. screened 3277 adults aged <60 for a
bicin + HDAC (cytarabine dose 1.5 g/m2 daily X 4, IA) and IA + vori- FLT3 mutation, found 896 to be FLT3 “positive,” of whom 717 were
nostat (IAV).48 CR rates were 75-80% with each arm. Although randomized to 7 + 3  midostaurin; results of testing were reported
remissions were achieved more often after course 1 with IA, and the to investigators within 48 hours of specimen receipt in a central labo-
proportion of individuals with intermediate and adverse cytogenetics ratory. 56 Midostaurin or placebo was given at 50 mg twice daily on

/
ESTEY 1275

active against FLT3 TKDs) improved EFS, although not survival, when
compared to 7 + 3 in a randomized trial in adults age <60, but had no
benefit in older adults. 58,59 The effect on EFS was the same in
patients with or without an ITD; patients with FLT3 TKDs were not
treated. Future comparisons of chemotherapy + either midostaurin or
more potent, specific FLT3 inhibitors in patients with FLT3 aberra-
tions may shed light on the importance of targeting multiple, rather
than a single, aberration.
Among these more potent, specific inhibitors are crenolanib, qui-
zartinib, and gilteritinib.60 These are likely to be combined with che-
motherapy and compared to chemotherapy + midostaurin. These
drugs also are relatively active vs the FLT3 D835 TKD mutation,
which often appears when resistance to earlier FLT3 inhibitors
develop, 61 perhaps suggesting combinations of newer inhibitors and

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


midostaurin. Treatment with FLT3 inhibitors also appears to unmask a
dependence on glutaminolysis, suggesting incorporation of glutamin-
ase inhibitors to overcome development of resistance.62

4.5 | Gemtuzumab ozogamicin and CD33 inhibition


The cell surface antigen CD33 is commonly expressed on the AML cell
surface (including possibly in some cases AML “stem cells”) 63 as well
as on normal myeloid cells but not on other normal cells, with the

FIGURE 5 Survival after 7 + 3 + either midostaurin or placebo (see exception of sinusoidal cells in the liver. GO consists of an antibody to
reference 56) CD33 combined with the toxin caleacheamicin. Used alone GO is
highly effective against acute promyelocytic leukemia (APL) presum-
days 8-21 during induction and “consolidation” (4 cycles of cytarabine ably because of the heavy expression of CD33 in APL.64 It is much
at the commonly used 3 g/m 2 BID on days 1, 3, and 5), and was used less effective as a single agent in other AML with CR and CRi rates of
alone as “maintenance” at the same dose for 1 year. Although CR only 13% each in relapsed disease.65 Hence, as with FLT3 inhibitors,
rates were similar in both groups (59% midostaurin and 54% placebo), attention has focused on combinations of GO with chemotherapy.
survival, the primary endpoint, was longer with midostaurin (median Hills et al. performed an individual-patient meta-analysis of five rele-
follow-up 59 months in the 359 patients still alive). This largely vant trials (UK NCRI 2, French Goelams group, French Alfa group and
reflected a lower relapse rate, as also evidenced by longer EFS with US SWOG group 1 each) combining GO with 7 + 3 or FLAG-ida and
midostaurin. Midostaurin was superior regardless of ITD allelic ratio using data from 3325 adults, median age 58.66 There was no intertrial
(<vs > 0.7) or whether the FLT3 mutation was an ITD or a TKD heterogeneity, considering various endpoints. Although CR rates were
(Figure 5). Similar proportions in each arm received HCT at any time similar GO, significant reductions in risk of relapse with GO contrib-
(55%-59%) or in CR1 (23%-28%) and results were unaffected if cen- uted to a survival benefit at 5 years confined to those with “favorable”
soring was done at HCT, although survival was superior with midos- cytogenetics 55% vs 76% (95% confidence interval 0.31-0.73 for
taurin if HCT was done in CR1 but not later. Although the reduction in risk of death) and with intermediate cytogenetics (34% vs
contribution of “maintenance” to the results has yet to be 39%, HR 0.75-0.95). Administration of 3 mg/m 2 GO on days 1, 4, and
57
demonstrated, and the greater effect of midostaurin on survival, but 7 of chemotherapy was associated with the least toxicity and equal
not EFS, in men than women remains unexplained, 56 the Stone benefit. Based on these data on September 1, 2017, the FDA
et al. study clearly indicate addition of midostaurin to 7 + 3 is now approved GO for adults with CD33 positive AML, and it should rou-
standard therapy for adults aged <60 with newly diagnosed AML and tinely be combined with 7 + 3 or FLAG-ida in those with ELN favor-
a FLT3 aberration. On April 28, 2017, the FDA approved midostaurin able or intermediate risk assuming an acceptable risk of TRM, as was
for all patients with a FLT3 aberration regardless of age, despite a lack the case in the studies noted above. Amadori et al compared GO used
of randomized data in older patients. NCRI data suggest in patients alone vs “best supportive care ‘only in patients over age 75, age 61-75
with a FLT3 ITD and a low probability of TRM daunorubicin doses of with performance status 3-4, or who declined’ intensive chemother-
90, rather than 60 mg/m2 daily × 3 be used in 7 + 3 together with apy”, finding a statistically significant (P = .005) but medically less sig-
51
midostaurin, although this remains speculative. nificant survival advantage for GO (medians of 5.9 vs 3.6 months) and
Midostaurin inhibits kinases other than FLT3. Hence it is plausible a CR + CRi rate of 27% (30/111).67
these drugs will also be effective in people with no FLT3 aberrations, In an effort to better stratify patients likely to respond to GO + che-
and studies investigating this possibility are in progress. As an exam- motherapy Olombel et al. reported in 200 patients EFS and RFS were lon-
ple, Röllig et al. have reported addition of sorafenib, often thought of ger with GO than chemotherapy without GO only in those with high CD
as a “FLT3 inhibitor” but actually a multikinase inhibitor (although not 33 expression (at least 70%) independent of cytogenetics and NPM1/

/
1276 ESTEY

FLT3 status.68 In contrast, Khan et al., examining 1583 younger and older identified such conventional covariates as poorer PS and adverse
patients, found no relation between CD33 expression and outcome fol- cytogenetics as associated with poorer rates of CR, EFS, and sur-
lowing GO in patients with “favorable (CBF)” cytogenetics, but a sugges- vival.77 Likewise, given the frequency with which people with second-
tion in other cytogenetic groups that those in the lowest quartile of ary AML have previously received azacitidine, it is noteworthy the
2 69
CD33 expression might benefit from 6, rather than 3, mg/m GO dose. benefit of CPX vs 7 + 3 appears limited to patients who have not
CD33 single nucleotide polymorphisms (SNPs) regulate the received azacitidine. Nonetheless the totality of evidence from the
expression of CD33 isoforms. Certain SNPs denoted as TT result in a phase 3 trial prompted the FDA on August 3, 2017 to approve CPX
lack of exon 2, resulting in absence of the CD33 IgV domain, which is 351 for patients with therapy-related AML (t-AML) or AML with
the antibody-binding site for GO, as well as diagnostic immunopheno- MDS-related changes2 regardless of age even though the trial leading
70
typic panels. Lamba et al. found only children with the CC SNP (51% to approval was conducted only in patients aged 60-75, and, as with
of 816 children) which encodes a full exon 2 benefitted from addition many trials, the proportion of eligible patients who participated in the
of GO to chemotherapy regardless of cytogenetics or CD33 expres- randomization (a convenient measure of selection bias) is unclear.40 It
71
sion. However, this finding has not been confirmed by the UK is important to note the adverse prognostic significance of t-AML is
NCRI72 : material for SNP determination was available in 536/2063 limited to patients who have received cytotoxic chemotherapy, rather

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


adults entered on their chemotherapy GO trials. The proportion of than only surgery  XRT for a prior malignancy.78 Future directions
patients with the CC SNP (47%) was similar to that in Lamba et al. and might include combinations with FLT3 inhibitors, or given lower
as in Lamba et al. CD33 expression was lowest with the TT. However, 60-day mortality rates in the randomized trial, use at full dose in
although the favorable cytogenetic group had the expected longer patients at high risk of TRM for example, those with PS 3-4 or organ
survival when given chemotherapy + GO, SNP had no influence on dysfunction, who were generally not included in the randomized
outcome. Similar results have been reported in adults by SWOG trial.75 Results of trials of doses of CPX 67% or 33% those used in the
(Othus personal communication). Currently we do not yet appear randomized trial in such patients were not better than those in histori-
ready to assign adults to receive GO on a basis other than cytogenet- cal controls.79 It is however clear CPX-351 is a viable option in fit
ics, especially considering likely differences in quantifying CD33 older patients with ELN intermediate risk18 “secondary AML” as
expression except if done in large specialized centers. defined above.
Development of improved anti-CD33 antibodies is being investi-
gated. For example, it might be advantageous to design an antibody in
4.7 | Venetoclax
which the CD33- anti CD33 binding site is closer to the cell surface as
experimental data suggest such proximity would enhance the efficacy Venetoclax is an oral inhibitor of the anti-apoptotic protein BCL-2
thought to mediate resistance to typical AML therapy. DiNardo
of CD3/CD33 bi-specific T-cell engagers (BiTEs), analogous to
et al. administered the drug at daily doses of 400 800, or 1200 mg
CD3/CD19 BiTEs in ALL, or CD33 directed chimeric antigen receptor
(CAR) T-cells.73 daily together with either decitabine or azacytidine to 145 people
(median age 74, range 65-86) considered unfit for standard therapy
and with intermediate or adverse cytogenetics and noted a 30-day
4.6 | CPX 351 (“Vyxeos”) death rate of 3%, CR rate of 35%, and a CR + CRi rate of 66%80
CPX 351 is a liposomal formulation of cytarabine and daunorubicin at (Supporting Information Figure 3A).The latter rates were higher with
a fixed 5:1 M ratio of cytarabine/daunorubicin that was determined intermediate cytogenetics CR 41% vs 30%, CR + CRi 75% vs 57%.
as optimum in preclinical testing. The liposomal encapsulation leads to Median survival was 18 months, although median time on study was
prolonged exposure to the two drugs.74 Lancet et al. randomized only 7 months. Similar results were reported with daily venetoclax
309 patients aged 60-75 and PS 0-2, with either (a) prior cytotoxic (600 mg) + low-dose cytarabine 20 m/m2 daily × 10 in 61 patients,
therapy, (b) antecedent MDS or CMML (receipt of azacytidine or median age 74, range 66-87) likewise judged unfit for intensive ther-
daunorubicin), or (c) WHO-defined MDS-like cytogenetic abnormali- apy: 30-day mortality rate 3%, CR rate 26%, CR + CRi rate 62%,
ties to either 7 + 3 (60 mg/m 2 daunorubicin daily × 3) or CPX-351. median survival about 1 year, with median follow-up about 1 year80
CPX was associated with higher CR + CRi rates (48% vs 33%) and, (Supporting Information Figure 3B)
with a minimum follow-up of 13 months, longer EFS (HR = 0.74, Based on these data the FDA granted “breakthrough” therapy des-
P = .02) and survival (HR = 0.69, P = .05, with medians of 10 vs ignation for venetoclax + either azacytidine or decitabine (on January
6 months). Sixty-day mortality was 21% with 7 + 3 and 13% with 28, 2016) or + low-dose cytarabine (on July 28, 2017) for patients who
75
CPX and toxicity was similar. About 34% of CPX and 25% of 7 + 3 are ineligible for intensive therapy. Several questions remain. First, how
patients received HCT; the transplanted CPX patients were older but to define “ineligible”? Of note, although the patients in the venetoclax
more often in CR than the transplanted 7 + 3 patients. A landmark studies described above were “older,” eligibility required PS 0-2 with
76
analysis suggested better post HCT survival in the CPX group about 75%-80% having PS 0-1, that is, minimally symptomatic. Normal
(Supporting Information Figure 2). Although this may have simply organ function was presumably also required, recalling ELN 2017 rec-
reflected the higher proportion of CPX than 7 + 3 patients in CR/CRi, ommendations that something other than older age is needed to con-
it is plausible a higher proportion of CPX patients had no MRD in CR sider someone inappropriate for intensive induction, for example
or CRi There are also suggestions CPX may be particularly effective vs performance status 3-4, comorbidities, or a high likelihood of resistance
7 + 3 in people with FLT3 ITD. Conversely, a multivariate analysis to standard intensive induction.18 Will responsiveness to venetoclax +

/
1286 ESTEY

in all patients in the intermediate and, particularly worst, Breems blasts. Enasidenib inhibits mutant IDH2 protein thus reducing ele-
et al. prognostic groups certainly extends to older patients, who all vated HG levels and promoting differentiation. Stein et al. reported an
else being equal would be expected to do even worse than the objective response rate of 39% (95% CI 28%-44%) among 109 adults
patients analyzed by Breems et al. and to all patients who are receiv- with R/R AML and IDH2 mutations who were given the 100 mg daily
ing a 2nd re-induction attempt after failing the first.136,137
dose used in the expansion cohort of the phase 2 study140 the CR and
Table 7 summarizes recommendations for management of R/R CRi rates were 20% and 7%, respectively. In distinct contrast with
AML. If feasible, allo HCT is favored in CR2 even in cases with long chemotherapy, the probability of CR or CRi on a given course
initial CR1s and in which allo HCT was performed in CR1. A possible appeared similar until at least the sixth (Supporting Information
exception may be cases in which allo HCT was done in CR1 but CR Figure 9). The most notable toxicity consistent with the drug's pro-
duration was <6 months. Here, unless some very different approach is posed mechanism of action was a differentiation syndrome entirely
taken to 2nd HCT the morbidities of the procedure may not be com- reminiscent of that following treatment of APL with all-trans retinoic
mensurate with the benefits. acid or arsenic trioxide and thus often accompanied by leukocytosis
and effectively treated with steroids141 ; the differentiation syndrome
tends to resolve, as does hyperbilirubinemia; whether it is associated

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


6.2 | Newer approaches
with response is not clear. With a median follow-up of 8 months,
Perusal of clinicaltrials.gov indicates many new therapies are available median survival was 9 months. As usual, it was longer in responders
for R/R AML. Although this is undoubtedly encouraging, such avail- but whether this was totally reflective of time spent in response,
ability suggests significant uncertainty as to which therapy is best. rather than an inherently better prognosis is not clear, and the relation
Given interest in venetoclax and CPX-351 in newly diagnosed AML, between various types of “response” and survival time, after account-
thoughts might turn to these in people not previously exposed. ing for guarantee time, also remains to be resolved. Based on these
DiNardo et al. treated 43 patients median age 68, 84% of whom were results, on August 1, 2017, the FDA approved enasidenib for treat-
receiving the drug as 2nd or later salvage, and 91% of whom received ment of R/R AML characterized by an IDH2 mutation.
the drug together with azacitidine, decitabine, or “low dose” cytara- Of note, the median survival following administration of chemo-
bine.138 Although the objective response rate (ORR: CR+ CRi + mar- therapies such as FLAG to R/R patients with an IDH2 mutations
row leukemia free-state) was 21%), the CR rate was only 5%, the CRi resembles that seen after enasidenib142 This raises the possibility of
rate 7% and median survival was 3 months. Four of the nine combining “intensive” chemotherapy with enasidenib to avoid devel-
responses occurred in the eight patients with RUNX1 mutations. Feld- opment of resistance. Examples such as GO or midostaurin suggest
man et al. noted CR rate of 5/20 at the CPX-351 dose subsequently the benefit of “targeted” therapies are likely to be much greater when
used in untreated patients; four of the five had initial CR1 durations combined with chemotherapy than when used alone.87 The same may
of at least 1 year.139 Despite the small numbers treated and the plau- apply with other targeted agents that show some, but limited, activity
sible existence of various selection biases, it is difficult to view results in R/R AML (eg, the DOT1L inhibitor pinometostat which affects the
with venetoclax or CPX351 in R/R AML as materially superior to his- MLL rearrangement characteristic of 11q23 translocations,143 or the
torical results. IDH1 inhibitor inasidenib, which has been approved by the patients
with IDH1 mutations.144,145 An example of such a trial combines
6.2.1 | Enasidenib, ivosidenib, and other targeted 7 + 3 with enasidenib (if IDH2 mutation) or ivosidenib (if IDH 1 muta-
therapies tion) in newly diagnosed AML.145 Analysis of resistance to enasidenib
Mutations in IDH2 occur in about 10% of AML patients and lead to indicate that while levels of HG typically remain suppressed at relapse
accumulation of R-2 hydroxyglutarate(HG), leading to DNA and his- mutant IDH clones which had persisted at relapse acquire additional
tone hypermethylation and consequent differentiation arrest of AML lesions (eg, 7q deletion or various mutations) which re-impose a

TABLE 7 Management of relapsed/refractory AML

Number of prior Breems score (alternatively prior


re-induction attempts CR1 duration) Re-induction therapy If CR is achieved
0 (1st salvage) 1-6 points Similar to initial induction therapy, for Allo HCT if feasible; if not, auto HCT if
example, 7 + 3, FLAG  idarubicin, no MRD, or clinical trial
(>18 months)
GCLAM (Halpern et al. phase 1/2
trial of GCLAM with dose-escalated
mitoxantrone for newly diagnosed
AML or other high-grade myeloid
neoplasms. Leukemia e-published
April 17,2018
0 (1st salvage) 7-9 points As above or clinical trial As above
(12-18 months)
0(1st salvage) 10-14 points Clinical trial (consider allo HCT as part As above
of induction)
(<12 months)
1 or more (2nd or higher N/A Clinical trial (consider allo HCT as part As above
salvage) of induction)

/
ESTEY 1277

azacytidine or + low dose cytarabine depend on the same factors asso- of post CR high-dose cytarabine, and thereafter alone at 100 mg daily
ciated with responsiveness to other therapies, noting the lower × 1 year as “maintenance.” 83 Median age was the expected 50 years
81
response rate in people with TP53 mutations in the Wei et al. study and the CR/CRi rate was expectedly high 94% (84/89), 92% in
(Supporting Information Figure 3B) and with adverse cytogenetics in 37 patients with t(8;21), and 96% in 52 with inv 16. However, with
81
the DiNardo et al. study? Might addition of venetoclax to 7 + 3 or median follow-up of 4 years, 4-year relapse rate is less 33% vs 43%
FLAG-ida improve outcomes in patients at low risk of TRM? Might ven- and 4-year EFS (but not survival) longer (58% vs 48%) with addition of
toclax be useful in a post-remission setting, for example in people with dasatinib. 83 Toxicity, including pleural effusions in about 8%, was not
MRD after induction and/or “consolidation”? Several of these questions undue. At relapse, KIT mutation was lost in 5/9 patients presenting
will be addressed in upcoming trials (see https://www.clinicaltrials.gov/ with these mutations, again speaking to the complexity of relapse.
ct2/results?term=venetoclax+%2B+AML&Search=Search), including Alliance has reported similar results, while also finding addition of
those comparing azacytidine (or decitabine)  venetoclax or low dose dasatinib abrogated the negative impact of KIT mutations.84 Both
cytarabine  venetoclax. Given the issue of ineligibility noted above a AMLSG and Alliance are following up with randomization to intensive
comparison of venetoclax + azacytidine (or low dose cytarabine) to chemotherapy  dasatinib.

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


7 + 3 might be of academic interest.

4.10 | “Beat AML” and targeted therapy


4.8 | Ten-day decitabine Numerous drugs have been designed to inhibit the “downstream” con-
Although used to treat AML for many years, decitabine has typically sequences of the many mutations observed in AML (although only a
2
been given for 5 days at a daily dose of 20 mg/m . However, a paper few mutations are usually observed in a given patient.85 These conse-
by Welch et al. reporting a rate of 100% in 21 patients with TP53 quences might for example be functionally abnormal proteins that ini-
mutations, who received 20 mg/m 2 daily × 10 days has received tiate/maintain the disease. Given the heterogeneity of AML numerous
much attention.82 The CR rate was 19%, the remainder of the such “targeted agents” are being investigated, midostaurin, and the
response being varying forms of CRi with <5% marrow blasts by mor- IDH2 inhibitor enasidenib, described further below, are prime exam-
phology. While reductions in TP53 mutation burden were universal, ples. The Leukemia and Lymphoma Society is sponsoring a “BEAT
they were invariably incomplete, no more frequent in people with AML” trial designed to examine a variety of these agents.86 The trial
than without blood count recovery, with remission duration similar in plans to accrue 500 patients aged 60 or above with newly diagnosed
these two groups. Remissions generally lasted less than 1 year and AML. The first aim is to assess the feasibility of enrolling patients,
median survival in TP53 mutated patients was 12.8 months, which based on mutation status, within 7 days of procurement of a marrow
however resembled survival in patients without TP53 mutations; such sample, which is sent to a central lab. Table 3 indicates the relation
similarity was, as the authors note, remarkable. between mutation status and assignment to therapy in BEAT AML.
Although these results have likely prompted considerable use of An issue is the use of the targeted agent alone for, in some cases,
the 10-day schedule, it not clear that the results can be generalized. several cycles (Table 3). It has been pointed out agents such as midos-
Efforts are in progress to address this question in a large single-arm taurin and GO have only modest activity when used alone but with
study. A randomized single-center comparison of 5- and 10-days considerable incremental benefit upon addition of chemotherapy.87
schedules of decitabine closed because the probability the 10-day However, given advances in therapy of infections it is likely rapid
schedule would be superior was low, although the statistical properties attainment of CR, and a concomitant neutrophil count >1000/μL is
of the design in TP53 mutated patients was not noted (Short N, pre- less important than previously. Indeed, it has been noted a majority of
sented at Acute Leukemia Forum, Newport Beach CA 2018). Although survival time is, currently, not uncommonly spent out of CR (Shaw C
there is no obvious harm in administering 10 rather than 5 days of deci- presented at Society of Hematology Oncology [SOHO] meeting,
tabine, perhaps the paper's principal value was demonstration of the September, 2017). Hence the risk in receiving a targeted agent alone
complexity of relapse, with at least in some cases, the dominant clone for a limited time, as in BEAT AML, is probably quite low.
at relapse, not the dominant one at diagnosis, suggesting in some cases
treatment can facilitate relapse. The dissociation of CR (vs CRi) from 4.11 | Recommendations for initial induction
survival suggests this relation may vary by treatment.
therapy
Table 4 considers standard therapy for both ELN favorable or interme-
4.9 | Dasatinib diate risk disease to encompass GO or midostaurin, the latter for
About 25%-35% of cases of CBF AML (ie, AML with t[8;21], inv [16] patients with a FLT3 ITD or TKD. Patients with CBF AML (ELN favor-
or t[16;16]) have mutations in C-KIT [Figure 1], which have been asso- able) rarely have FLT3 aberrations (Figure 1) and whether any such
ciated with shorter remissions. 83 Cases without mutations often have patients should receive both GO and midostaurin is hard to decipher
KIT overexpression, and overexpression is more common in CBF AML absent more data; the same is true for the larger group of ELN interme-
than in other types.83 Dasatinib inhibits not only the BCR-ABL kinase diate patients with a FLT3 aberration. A lack of data makes it difficult
of CML but also KIT kinase including that encoded by KIT mutations to know whether midostaurin should be administered to all patients, as
83
that are resistant to other kinases. The German AMLSG added dasa- suggested by the FDA, or only patients aged <60 as in the trial leading
tinib on days 8-21 after beginning 7 + 3, days 6-28 of each of 4 cycles to approval; the same is true for CPX-351 (approved for all but tested

/
1278 ESTEY

TABLE 3 Arms of “BEAT AML” (see reference 86)

Mutation “Targeted” agent Other therapies


None, or RUNX1/RUNX1T1[t(8;21)],or Samalizumab Smalizumab given with 7 + 3 for induction,
used alone for “maintenance”
CBFB/MYH11 (inv 16) (p13;q22)or t (antibody against CD200 which is
(16;16)(p13;q22) correlated with frequency
immunosuppressive T regulatory cells
(Leukemia 2012;26(9) 2146-48))
None, or TET 2, or IDH1, or WT1 BI 836858 BI 838568 given with azacitidine for
induction and “maintenance”
(anti CD33 antibody augments antibody
dependent cellular toxicity after
treatment with decitabine; [Vasu
et al. Blood 2016;127:2879-2889]
IDH2 Enasidenib Add azacitidine if neither CR nor CRi after
5 cycles enasidenib
(IDH2 inhibitor, see below)
MLL Entospletinib Add azacitidine after 1-4 cycles

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


entospletinib if no CR/CRi or
SYK (spleen tyrosine kinase) inhibitor
“progressive disease”
J Hematol Oncol 2017; Jul 28;10(1):145.
doi: 10.1186/s13045-017-0512-1.
TP53 Entospletinib Add 2 cycles decitabine after 1 cycle
entospletinib if subsequent induction
SYK inhibitor
needed; at least 1 of the 2 cycles must
give Decitabine × 10 days
NPM1 with no FLT3 ITD Entospletinib Entospletinib given with 7 + 3 if age <75
with PS 0-2 ; if age >75 or PS >2 add
SYK inhibitor
azacitidine if no CR/CRi after 1 cycle
entospletinib
TP53 Pevonedistat Pevonedistat given with azacitidine ×
4 cycles and then for “maintenance”
(inhibitor of the NEDD8-activating enzyme,
combined with azacitidine reported to
improve response rate of latter [ Swords
et al. Blood 2018;131:1415-24]

only in ages 60-75); here, however I have simply followed the FDA's ELN intermediate group (<50% long term survival) a case can be made
labeling instructions. Less intensive therapy seems intuitively prefera- that the first option even in such patients should be a clinical trial. This
43
ble in patients at higher risk of TRM, but some data question this and is certainly true in patients at higher risk of TRM, criteria for which
resolution awaits a randomized trial as does the role of CPX 351 vs should not be based exclusively on age. A problem of course is that
chemotherapy + GO or + midostaurin in secondary AML. It is plausible many patients at higher risk of TRM are currently excluded from trials
some less intense therapy might be not only less toxic but more effec- based on poor performance status or comorbidities, making it difficult
tive than today's more intensive therapy or that CPX351 might have a to generalize the results to people who are not only older but also less
role in de novo AML, for example if characterized by FLT3 ITD. These fit.40 Montalban-Bravo et al. have demonstrated the potentially favor-
and other questions can only be answered in clinical trials and given able benefit/risk ratio of a trial in cases where eligibility criteria are
the unsatisfactory outcomes of even patients at low risk of TRM in the essentially ineligibility for standard trials. 88

TABLE 4 Recommendations for initial induction therapy

ELN group TRM risk lowa TRM risk higherb


c
“Favorable” 7 + 3 + GO or FLAG-ida + GO Azacytidine (or decitabine) + GO c or clinical
trial
“Intermediate” 7 + 3 + GO or FLAG-ida + GO d,e Azacytidine (or decitabine) + GO d,e or
clinical trial, for example, combinations
Or clinical trial, for example, combinations
involving venetoclax
of 7 + 3 + venetoclax
Adverse Clinical trial, for example, “BEAT AML” or Clinical trial, for example, “BEAT AML” or
combinations involving venetoclax combinations involving venetoclax
+7 + 3 or FLAG-ida; add midostaurin if
add midostaurin if FLT3 ITD positivee
FLT3 ITD or TKD positivee
a
Usually age <70-75 with performance status <2 and no cardiac, hepatic, renal dysfunction, and albumin >3.5
b
Usually age >70 plus performance status 2-4 and/or 1 of above comorbidities
c
Consider midostaurin if FLT3 ITD positive (allelic ratio <0.5)
d
Midostaurin if FLT3 ITD or TKD
e
Consider CPX −351 if secondary AML as described in Ref. 75

/
ESTEY 1287

differentiation block146 again suggesting the importance of combining pre-HCT MRD and post-HCT relapse it seems natural to question the
several different targeted agents with each other or with chemother- value of bridge to HCT when underlaid by these responses.
apy to prevent emergence of resistance. In any event relapse is the principal cause of failure of HCT. Aza-
The successes of immunotherapy in various solid tumors has led citidine may be the most widely used treatment for post HCT relapse.
to an upsurge of interest in immunotherapy in AML.147 Because AML After administering the drug at the usual 75 mg/m2 daily × 5 or
is not notably immunogenic per se immune-modulatory drugs (ipilimu- 7 days to 181 patients with morphologic relapse (>5%) blasts after
mab, nivolumab, and pembrolizumb) are being combined, often with HCT, Craddock et al. devised a prognostic index based on time from
decitabine or azacitidine; several trials are in progress (reviewed in ref- HCT to relapse (2 points if <6 months, 1-2 if 6-12 months, 0 if
erence 148). Azacitidine and/or decitabine have effects that in princi- >12 months) and marrow blast % (1 point if > the median 20%, 0 oth-
ple may be favorable (eg, increased receptor expression on T cells or erwise). 2 year survival rates were 37%, 15% and 3% in people with
antigen presentation sites on AML blasts) or unfavorable (eg, scores of 0, 1-2, and 3 who constituted 18%, 28%, and 54% of the
increased PD-1 or CTLA-4 expression on T-cells leading to T-cell patients, respectively; survival rates were paralleled by CR (CR + PR)
exhaustion. Eventually this type therapy may have more success when rates of 34% (49%), 14%,(28%), and 8% (17%). 151 Addition of donor

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


tested in patients in remission  MRD (see below). lymphocytes infusions (DLI) had no effect, in keeping with older
BiTEs are described in the section on gemtuzumab ozogamicin reports of the typical inefficacy of DLI and withdrawal of immunosup-
above. pression. 152 As with relapse in general, standard approaches are of lit-
tle use except in people with “long” CR durations, for example,
6.2.2 | Allogeneic HCT >1 year, more than justifying referral of patients for clinical trials, for

The success of allo HCT is in considerable measure due to immune example, those that might augment the GVL effect.

effect exerted by donor T- and NK-cells. Allo HCT is the therapy often
viewed as most likely to cure R/R AML. However, because it is typi-
7 | NEWER SCENARIOS FOR TE STING NEW
cally not done until CR2 is observed only a small minority of R/R
AGENTS
patients receive it. For example, only 14% of patients in Breems
et al.’s worst prognostic group received HCT as initial salvage therapy Conventionally new agents are tested in relapsed or adverse-risk
for 1st relapse.134 This has led to interest in an “early transplant” newly diagnosed AML. This practice may predispose to discarding
approach in which patients receive intensive re-induction therapy, fol- these agents, which however might perform better in less advanced
lowed by “obligatory” HCT several weeks after administration of che- disease. One possibility is in the setting of MRD. However, enrollment
motherapy. Because it is well-known that HCT done in relapse is into many trials of new drugs is restricted to morphological relapse
much less effective than HCT in CR the idea is to reduce the amount (>5% blasts). It is plausible response rates to new drugs might be
of disease prior to HCT while still performing HCT without however higher if tested in cases where MRD is the only evidence of disease.
requiring achievement of CR. For example, Middeke et al.149 gave Justification would be the very high predictive value of a positive
CLARA (clofarabine + ara-C, 1 g/m2 daily × 5) to 84 patients, median MRD test for relapse, particularly in people with ELN 2017 adverse-
age 61, 43 relapsed after a median CR1 duration of 6 months, risk pretreatment, the usually short (<1 year) interval between appear-
41 refractory to initial therapy. 52% of the patients had <10% marrow ance of MRD and morphologic relapse, and the generally limited side
blasts on day 15 and 56 of the 84 patients received HCT. A total of effects of “targeted” therapies, resulting in likely favorable/benefit risk
39 of the 84 had a donor identified prior to enrollment; 32 of these ratios when used to treat MRD. Indeed, it is likely the next few years
39 received HCT a median of 29 days after enrollment, as did, at a will see more use of targeted therapies to treat MRD. These years
median of 37 days, 24 of the 45 without a donor identified at enroll- may also see the partial replacement of standard morphologic assess-
ment. The major factor associated with performance of HCT was pre- ment of marrow to detect relapse by MRD-based techniques; it
enrollment marrow blast % (median 22%, range 5%-92% HCT vs appears that when MFC testing is negative morphology will inevitably
median 60%, range 17%-90% no HCT. Conditioning was clofarabine show <5% blasts, below the threshold for morphologic relapse.153
2 2
(30 mg/m days minus 6 to minus 3) + melphalan (140 mg/m on day Treating patients with MRD only AML would allow comparison of
minus 2). With a median follow-up of 40 months 3-year survival rate outcomes when the same therapy is used to treat an MRD only
was 40% (95% CI 31-52%), including 36% for refractory disease, 44% relapse and morphologic relapse. It would also allow testing of the yet
for relapsed disease, and 45% for initial CR1 duration <6 months unproven, but eminently plausible, hypothesis that reduction in MRD
(n = 21). 149 While these results compare very favorably to those would lead to better outcomes. A further step would allow testing of
expected without early HCT it remains unclear whether they will new drugs in cases that are MRD negative but still at relatively high
prove generalizable, noting the results largely reflect outcomes in the risk of relapse based on pretreatment factors.
75% of patients with HCT CI scores <3. Finally, we may overestimate how much we know about the tar-
The possibility of a “bridge to HCT” is often invoked when noting gets of targeted therapy. We noted above the beneficial effect of sor-
responses such as CRi or marrow-leukemia free state following single- afenib, often thought of as a FLT 3 inhibitor, when added to 7 + 3,
agent targeted therapies. However, these responses seem more likely seem similar in people regardless of whether they have a FLT3 ITD.58
150
to be associated with MRD than CR Given the association between Hence once a targeted therapy demonstrates activity in patients with

/
ESTEY 1279

TABLE 5 Incorporation of MRD data with pretreatment ELN risk score to determine post-remission therapy

Risk of non-relapse mortality


(NRM) post-HCT
Risk of relapse at 1-2 years justifying allo. HCT (HCT-CI score Possible subsequent treatment if
Pretreatment MRD status after two without allogeneic HCT associated with that risk) excessive risk/benefit ratio with
ELN risk courses therapy ( 94,96–98) (see reference 111) allogeneic HCT
Favorable Positive 40-50% if <20-25% Clinical trial
CBF (≤2)
50-60% if NPM1+/FLT3
negative
Negative 10% (CBF) None Ara-C 1 g/m 2 BID days 1,2,3 ×
3 cycles + GO (or midostaurin if
15-20% if NPM+/FLT3
FLT3 positive)
negative
Intermediate Positive 60-75% <30% Clinical trial
(≤3-4)

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


Negative 20% < 10% Clofarabine + ara-C, autologous
HCT,
(<1)
Adverse Positive 90-100% <40-50% Clinical trial
(≤5)
Negative 60% <20-25% (≤2) Autologous HCT, clofarabine +
ara-clinical trial

FIGURE 6 Comparison of outcomes in patients in CR without MRD by MFC, in patients in CR but with MRD by MFC and in patients with
“active disease”(>5% blasts by morphology) (see reference 95)

/
1288 ESTEY

the target it may be potentially valuable to assess outcome in people 19. Sorror ML, Storer BE, Fathi AT, et al. Development and validation of
without the target. 87 a novel acute myeloid leukemia-composite model to estimate risks of
mortality. JAMA Oncol. 2017;3:1675-1682.
20. Ho G, Wun T, Muffly L, et al. Decreased early mortality associated
with the treatment of acute myeloid leukemia at National Cancer
CONFLICT OF INTEREST Institute-designated cancer centers in California. Cancer. 2018;124:
1938-1945.
Nothing to report.
21. stgård LSG, Norgaard M, Medeiros BC, et al. Associations between
cohabitation status, treatment, and outcome in AML patients: a
national population-based study. Blood. 2018 Jun 14;131(24):
REFERENCES 2730-2733.
1. Lazarevic V, Orsmark-Pietras C, Lilljebjorn H, et al. Isolated myelosar- 22. stgård LSG, Norgaard M, Medeiros BC, et al. Effects of education
coma is characterized by recurrent NFE2 mutations and concurrent and income on treatment and outcome in patients with acute mye-
preleukemic clones in the bone marrow. Blood. 2018;131:577-581. loid leukemia in a tax-supported health care system: a National
2. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the Population-Based Cohort Study. J Clin Oncol. 2017;35:3678-3687.
World Health Organization classification of myeloid neoplasms and 23. Appelbaum FR, Gundacker H, Head DR, et al. Age and acute myeloid
acute leukemia. Blood. 2016;127:2391-2405. leukemia. Blood. 2006;107:3481-3485.

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


3. Walter RB, Othus M, Borthakur G, et al. Prediction of early death fol- 24. Yanada M, Garcia-Manero G, Borthakur G, et al. Relapse and death
lowing induction therapy for newly diagnosed acute myeloid leuke- during first remission in acute myeloid leukemia. Haematologica.
mia with pretreatment risk scores: a novel paradigm for treatment 2008;93:633-634.
assignment. J Clin Oncol. 2011;29(33):4417-4424. 25. Harada Y, Nagata Y, Kihara R, et al. Prognostic analysis according to
4. Estey E, Thall P, Beran M, et al. Effect of diagnosis (refractory anemia the 2017 ELN risk stratification by genetics in adult acute myeloid
with excess blasts, refractory anemia with excess blasts in transfor- leukemia patients treated in the Japan Adult Leukemia Study Group
mation, or acute myeloid leukemia [AML]) on outcome of AML-type (JALSG) AML201 study. Leukemia Res. 2018;66:20-27.
chemotherapy. Blood. 1997;90:2969-2977. 26. Eisfeld AK, Kohlschmidt J, Mrozek K, et al. Mutation patterns identify
5. Lindsley RC, Mar BG, Mazzola E, et al. Acute myeloid leukemia adult patients with de novo acute myeloid leukemia aged 60 years or
ontogeny is defined by distinct somatic mutations. Blood. 2015;125: older who respond favorably to standard chemotherapy: an analysis
1367-1376. of alliance studies. Leukemia. 2018;32:1338-1348.
6. Walter MJ. What came first: MDS or AML? Blood. 2015;125: 27. Ostronoff F, Othus M, Lazenby M, et al. Prognostic significance of
1357-1358. NPM1 mutations in the absence of FLT3-internal tandem duplication
7. Shlush LI. Age-related clonal hematopoiesis. Blood. 2018;131: in older patients with acute myeloid leukemia: a SWOG and UK
496-504. National Cancer Research Institute/Medical Research Council report.
8. University of Chicago Hematopoietic Malignancies Cancer Risk J Clin Oncol. 2015;33:1157-1164.
28. Patel SS, Kuo FC, Gibson CJ, et al. High NPM1 mutant allele burden
Team. How I diagnose and manage individuals at risk for inherited
at diagnosis predicts unfavorable outcomes in de novo AML. Blood.
myeloid malignancies. Blood . 2016;128:1800-1813.
2018 Jun 21;131(25):2816-2825.
9. Godley LA, Shimamura A. Genetic predisposition to hematologic
29. Papaemmanuil E, Gerstung M, Bullinger L, et al. Genomic classifica-
malignancies: management and surveillance. Blood . 2017;130:
tion and prognosis in acute myeloid leukemia. N Engl J Med. 2016;
424-432.
374:2209-2221.
10. Medeiros BC, Satram-Hoang S, Hurst D, et al. Big data analysis of
30. Kuo FC, Mar BG, Lindsley RC, et al. The relative utilities of
treatment patterns and outcomes among elderly acute myeloid leu-
genome-wide, gene panel, and individual gene sequencing in clinical
kemia patients in the United States. Ann Hematol. 2015;94:
practice. Blood. 2017;130:433-439.
1127-1138.
31. Duployez N, Marceau-Renaut A, Boissel N, et al. Comprehensive
11. Juliusson G, Antunovic P, Derolf A, et al. Age and acute myeloid leu-
mutational profiling of core binding factor acute myeloid leukemia.
kemia: real world data on decision to treat and outcomes from the
Blood. 2016;127:2451-2459.
Swedish acute leukemia registry. Blood. 2009;113:4179-4187.
32. Itzykson R, Duployez N, Fasan A, et al. Clonal interference of signal-
12. Juliusson G, Abrahamsson J, Lazarevic V, et al. Prevalence and char-
ing mutations holds prognostic relevance in core binding factor acute
acteristics of survivors from acute myeloid leukemia in Sweden. Leu-
myeloid leukemia. Blood . 2018 Jul 12;132(2):187-196.
kemia. 2017;31:728-731. 33. Sekeres MA, Elson P, Kalaycio ME, et al. Time from diagnosis to
13. Dombret H, Seymour JF, Butrym A, et al. International phase 3 study treatment initiation predicts survival in younger, but not older, acute
of azacitidine vs conventional care regimens in older patients with
myeloid leukemia patients. Blood. 2009;113:28-36.
newly diagnosed AML with >30% blasts. Blood. 2015;126:291-299. 34. Bertoli S, Berard E, Huguet F, et al. Time from diagnosis to intensive
14. Walter RB, Othus M, Borthakur G, et al. Prediction of early death chemotherapy initiation does not adversely impact the outcome of
after induction therapy for newly diagnosed acute myeloid leukemia patients with acute myeloid leukemia. Blood. 2013;121:2618-2626.
with pretreatment risk scores: a novel paradigm for treatment assign- 35. Takahashi K, Patel K, Bueso-Ramos C, et al. Clinical implications of
ment. J Clin Oncol. 2011;29(33):4417-4423. TP53 mutations in myelodysplastic syndromes treated with hypo-
15. Othus M, Kantarjian H, Petersdorf S, et al. Declining rates of methylating agents. Oncotarget . 2016;7:14172-14187.
treatment-related mortality in patients with newly diagnosed AML 36. Estey E, Othus M, Lee SJ, et al. New drug approvals in acute myeloid
given 'intense' induction regimens: a report from SWOG and MD leukemia: what's the best end point? Leukemia. 2016;30:521-525.
Anderson. Leukemia. 2014;28:289-292. 37. Estey E, Gale RP. How good are we at predicting the fate of someone
16. Halpern AB, Lyman GH, Walsh TJ, et al. Primary antifungal prophy- with acute myeloid leukaemia? Leukemia. 2017;31:1255-1258.
laxis during curative-intent therapy for acute myeloid leukemia. 38. Othus M, Sekeres MA, Nand S, et al. Complete remissions (CRs) with
Blood. 2015;126:2790-2797. Azacitidine regimens compared to Crs with 7+3 induction chemo-
17. O'Donnell MR, Tallman MS, Abboud CN, et al. Acute myeloid leuke- therapy and the effect on overall survival. Blood. 2016;128:
mia, version 3.2017, NCCN clinical practice guidelines in oncology. 1613-1613.
Journal of the National Comprehensive Cancer Network : JNCCN. 2017; 39. Othus M, Garcia-Manero G, Godwin J, et al. Associations between
15:926-957. complete remissions (CRs) with 7+3 induction chemotherapy for
18. Döhner H, Estey E, Grimwade D, et al. Diagnosis and management of acute myeloid leukemia and 2-3 year survival ("potential cure") over
AML in adults: 2017 ELN recommendations from an international the past four decades: analysis of SWOG trial data. Blood. 2017;130:
expert panel. Blood. 2017;129:424-447. 1301-1301.

/
1280 ESTEY

5 | POST-REMISSION THERAPY from normal” methodology is applicable in almost all cases. Molecular
techniques can be divided into real-time PCR and next generation
sequencing (NGS) . PCR provides sensitivity of 10(−4) to 10(−6) but is
5.1 | Risk stratification: Importance of post-
limited to the 40%-50% of patients with the fusion transcripts
treatment MRD
RUNX1-RUNX1T1 (characteristic of t[8;21]), CBFB-MYH11 [inv 16]
Once remission (CR or CRi) is observed physicians must decide or t[16;16]) or with an NPM1 mutation.93 In contrast, NGS can, theo-
whether to proceed to hematopoietic cell transplant (HCT, typically retically, be applied to all leukemia specific genetic aberrations. How-
allogeneic in the U.S. and either allogeneic or autologous elsewhere) ever, some genes are not suitable for MRD detection either because,
to prevent relapse (up to Ref. 88, Table 5 and Figure 6). In general, the although positive at diagnosis, may be negative at morphologic relapse
risk of NRM is greater with HCT (particularly allogenic), while the risk (eg, FLT3) or because they are associated with age-related clonal
of relapse is less with HCT (particularly allogeneic). Quantifying each hematopoiesis7 and, as such, may have arisen before development of
risk with HCT vs non HCT approaches is critical in deciding between AML. At least with molecular methods, peripheral blood is usually
them. For example, a plausible >10% improvement in relapse-free sur- 10-fold less sensitive than marrow, although blood can be sampled
vival after allo HCT has been proposed to justify the potential long- more frequently, which in principle might increase its sensitivity.

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


term risks of allo HCT, for example, chronic graft-vs-host disease and MRD has been assessed upon achievement of CR (after 1 or
development of malignancies due to immunosuppressive therapy. 2 induction cycles), after completion of “consolidation” therapy, or
Essentially all patients in the ELN 2017 adverse risk category, most in subsequently.93 Which of these is optimal, or whether it is preferable
the intermediate risk, and almost none in the “favorable” risk catego- to assess MRD at different times, focusing on an increase or decrease
ries would meet this criterion. If it is decided the risk of relapse is rela- rather than a single result, is unsettled.93 Likewise, the criterion for a
tively low based on ELN 2017 without HCT, the most commonly used positive test (eg, >0% or >0.01% by MFC) is unclear, as is whether
post remission therapy is probably “high-dose” cytarabine, often given patients who are MRD negative but have only CRi (ie, incomplete
as 3 g/m2 every 12 hours on days 1, 3, and 5. However, a German- count recovery) have the same prognosis as patients who are in CR
Austrian AML Study Group found administration of this dose on days but are MRD positive. MRD testing lacks standardization (although
1, 2, and 3 to 392 patients was associated with a median 4-day reduc- the same can be said of morphologic evaluation). Nonetheless, despite
tion in time to ANC >500 from start of chemotherapy and with fewer these issues MRD detection has unambiguously been shown to have
platelet transfusions than in 176 historical patients given the 1, 3, and a high positive predictive value for subsequent morphologic relapse,
5 schedule.89 Cytarabine doses of 1 g/m2 twice daily for 3-4 days for which typically occurs within 12 months of detection of MRD. Ter-
three courses after CR is achieved are very probably as effective as wijn et al.94 showed higher rates of relapse at 1 year in patients who
higher doses and/or more courses irrespective of cytogenetic group.90 remained MFC- positive after 2 cycles of therapy regardless of whether
Furthermore, after randomizing 221 adults age 18-59 in the interme- cytogenetics were “favorable” (40% vs 5%), “intermediate” (60% vs 20%),
diate or adverse ELN 2010 groups, but seemingly without a donor for or “adverse” (100% vs 50%). The unfavorable effect of MRD by MFC
HCT, between cytarabine in the usual 3 g/m2 schedule and cytarabine remains after allogeneic HCT; thus Araki et al.95 have reported risk of
2 2
1 g/m daily X5 plus clofarabine 30 mg/m daily X5 (CLARA) both relapse following HCT in people who have <5% morphologic blasts but
preceded by G-CSF, Thomas et al. reported a lower incidence of are MFC positive pre-HCT is closer to that seen in people receiving HCT
relapse (34% vs 46% at 2 years P = .025) accompanied by longer RFS with >5% blasts (“active AML”) than to people who are MFC negative pre
(but not survival) with CLARA regardless of risk group, raising the HCT (Figure 6). Regarding PCR, Ivey at al. demonstrated the 15% of
question whether CLARA should constitute standard post-remission patients with intermediate-risk AML and pretreatment NPM mutations
91
therapy in such patients. who were in presumed CR but remained NPM positive in blood after
However, basing post-remission therapy exclusively on risk of 2 cycles of induction therapy had an 86% cumulative incidence of
relapse determined by pretreatment information is problematic, since, relapse vs 34% for NPM negative patients96 (Figure 7). Multivariate anal-
as discussed above, in these instances we likely overestimate our abil- ysis indicated persistent NPM positivity in blood was the sole predictor
37
ity to forecast prognosis. It is intuitive knowledge gained after treat- of relapse and had similar effects in people with or without FLT3 muta-
ment begins might increase this ability. Means to this end might for tions (Figure 7). Likewise, less than 3 log reduction in PCR transcripts for
example include assessment of disease status 2-3 weeks post treat- RUNX1-RUNX1T1 or CBFB-MYH11 after high-dose cytarabine “consol-
ment. However, most attention has focused on the presence of mea- idation” was associated with a 3-year cumulative incidence of relapse of
surable residual disease (MRD) even though conventional criteria for 54% vs 22% for patients with the presumably favorable CBF AML and
CR are met. Although criteria for CR are often considered to include reduction in transcripts was the only factor identified with relapse in mul-
only neutrophil and platelet count >1000/μL and >100 000/μL, tivariate analysis, including pretreatment KIT mutations.97
respectively, Danish investigators have noted the association between Attention has more recently turned to assessment of post-
continued receipt of red cell transfusions at time of CR and increased treatment persistence of a variety of mutations assessed using NGS.
risk of relapse.92 Jongen-Lavrenic et al.98 found one or more mutations amenable to
MRD is assessed using MFC or molecular means. The 2018 ELN NGS testing in 430 of 482 adults age <66 years (average 2.9 muta-
MRD guidelines provide technical details.93 Using at least 8-colors to tions per patient). Testing in these 430 was done in remission (CR or
detect abnormal antigen patterns MFC can detect one abnormal cell CRi) within 21 days to 4 months after receipt of a second course of
among 10 000 normal ones (sensitivity 10[−4]) and using “different therapy, with mutations found to persist in 51% of patients. These

/
ESTEY 1289

40. Estey E, Gale R, Sekeres M. New drug approvals in acute myeloid leu- 59. Serve H, Krug U, Wagner R, et al. Sorafenib in combination with
kemia: uses and abuses. Leukemia. 2018 Jun 6. https://doi.org/10. intensive chemotherapy in elderly patients with acute myeloid leuke-
1038/s41375-018-0168-z In press. mia: results from a randomized, placebo-controlled trial. J Clin Oncol.
41. Boddu PC, Kantarjian HM, Ravandi F, et al. Characteristics and out- 2013;31:3110-3118.
comes of older patients with secondary acute myeloid leukemia 60. Levis M. The current therapeutic landscape of FLT3 inhibitors. Blood
according to treatment approach. Cancer. 2017;123:3050-3060. Adv. 2017;1:1944.
42. Quintas-Cardama A, Ravandi F, Liu-Dumlao T, et al. Epigenetic ther- 61. Galanis A, Ma H, Rajkhowa T, et al. Crenolanib is a potent inhibitor
apy is associated with similar survival compared with intensive che- of FLT3 with activity against resistance-conferring point mutants.
motherapy in older patients with newly diagnosed acute myeloid Blood. 2014;123:94-100.
leukemia. Blood. 2012;120:4840-4845. 62. Gallipoli P, Giotopoulos G, Tzelepis K, et al. Glutaminolysis is a meta-
43. Sorror M, Storer B, Elsawy M, et al. Intensive versus non-intensive bolic dependency in FLT3 ITD acute myeloid leukemia unmasked by
induction therapy for patients (pts) with newly diagnosed acute mye- FLT3 tyrosine kinase inhibition. Blood. 2018;131:1639-1653.
loid leukemia (AML) using two different novel prognostic models. 63. Walter RB, Appelbaum FR, Estey EH, et al. Acute myeloid leukemia
Blood. 2016;128:216. stem cells and CD33-targeted immunotherapy. Blood. 2012;119:
44. Desoutter J, Gay J, Berthon C, et al. Molecular prognostic factors in 6198-6208.
acute myeloid leukemia receiving first-line therapy with azacitidine. 64. Lo-Coco F, Cimino G, Breccia M, et al. Gemtuzumab ozogamicin
Leukemia. 2016;30:1416-1418. (Mylotarg) as a single agent for molecularly relapsed acute promyelo-
45. Prata PH, Bally C, Prebet T, et al. NPM1 mutation is not associated

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


cytic leukemia. Blood. 2004;104:1995-1999.
with prolonged complete remission in acute myeloid leukemia 65. Larson RA, Sievers EL, Stadtmauer EA, et al. Final report of the effi-
patients treated with hypomethylating agents. Haematologica. 2018 cacy and safety of gemtuzumab ozogamicin (Mylotarg) in patients
May 10. https://doi.org/10.3324/haematol.2018.189886 In press. with CD33-positive acute myeloid leukemia in first recurrence. Can-
46. Bello C, Yu D, Komrokji RS, et al. Outcomes after induction chemo- cer. 2005;104:1442-1452.
therapy in patients with acute myeloid leukemia arising from myelo- 66. Hills RK, Castaigne S, Appelbaum FR, et al. Addition of gemtuzumab
dysplastic syndrome. Cancer. 2011;117:1463-1469. ozogamicin to induction chemotherapy in adult patients with acute
47. Krug U, Berdel WE, Gale RP, et al. Increasing intensity of therapies myeloid leukaemia: a meta-analysis of individual patient data from
assigned at diagnosis does not improve survival of adults with acute
randomised controlled trials. Lancet Oncol. 2014;15:986-996.
myeloid leukemia. Leukemia . 2016;30:1230-1236. 67. Amadori S, Suciu S, Selleslag D, et al. Gemtuzumab Ozogamicin ver-
48. Garcia-Manero G, Othus M, Pagel J, et al. A randomized phase III sus best supportive Care in Older Patients with Newly Diagnosed
study of standard Cytarabine plus Daunorubicin (7+3) therapy versus
Acute Myeloid Leukemia Unsuitable for intensive chemotherapy:
Idarubicin with high dose Cytarabine (IA) with or without Vorinostat
results of the randomized phase III EORTC-GIMEMA AML-19 trial. J
(IA+V) in younger patients with previously untreated acute myeloid
Clin Oncol. 2016;34:972-979.
leukemia. Blood. 2016;128:901.
68. Olombel G, Guerin E, Guy J, et al. The level of blast CD33 expression
49. Burnett AK, Russell NH, Hills RK, et al. Optimization of chemother-
positively impacts the effect of gemtuzumab ozogamicin in patients
apy for younger patients with acute myeloid leukemia: results of the
with acute myeloid leukemia. Blood. 2016;127:2157-2160.
medical research council AML15 trial. J Clin Oncol. 2013;31:
69. Khan N, Hills RK, Virgo P, et al. Expression of CD33 is a predictive
3360-3368.
factor for effect of gemtuzumab ozogamicin at different doses in
50. Burnett AK, Russell NH, Hills RK, et al. A randomized comparison of
adult acute myeloid leukaemia. Leukemia. 2017;31:1059-1068.
daunorubicin 90 mg/m2 vs 60 mg/m2 in AML induction: results from
70. Laszlo GS, Harrington KH, Gudgeon CJ, et al. Expression and func-
the UK NCRI AML17 trial in 1206 patients. Blood. 2015;125:
tional characterization of CD33 transcript variants in human acute
3878-3885.
myeloid leukemia. Oncotarget . 2016;7:43281-43294.
51. Burnett AK, Russell NH, Hills RK. Higher daunorubicin exposure ben-
71. Lamba JK, Chauhan L, Shin M, et al. CD33 splicing polymorphism
efits FLT3 mutated acute myeloid leukemia. Blood. 2016;128:
determines Gemtuzumab Ozogamicin response in De novo acute
449-452.
myeloid leukemia: report from randomized phase III Children's oncol-
52. Löwenberg B, Pabst T, Maertens J, et al. Therapeutic value of clofar-
abine in younger and middle-aged (18-65 years) adults with newly ogy group trial AAML0531. J Clin Oncol . 2017;35:2674-2682.
diagnosed AML. Blood. 2017;129:1636-1645. 72. Gale RE, Popa T, Wright M, et al. No evidence that CD33 splicing
53. Halpern AB, Othus M, Huebner EM, et al. Phase 1/2 trial of GCLAM SNP impacts the response to GO in younger adults with AML treated
with dose-escalated mitoxantrone for newly diagnosed AML or other on UK MRC/NCRI trials. Blood. 2018;131:468-471.
high-grade myeloid neoplasms. Leukemia. 2018 Apr 17. https://doi. 73. Walter RB. Investigational CD33-targeted therapeutics for acute
org/10.1038/s41375-018-0135-8. myeloid leukemia. Expert Opin Investig Drugs. 2018;27:339-348.
54. Wei AH, Tiong IS. Midostaurin, enasidenib, CPX-351, gemtuzumab 74. Nikanjam M, Capparelli EV, Lancet JE, et al. Persistent cytarabine
ozogamicin, and venetoclax bring new hope to AML. Blood. 2017; and daunorubicin exposure after administration of novel liposomal
130:2469-2474. formulation CPX-351: population pharmacokinetic assessment. Can-
55. Lazenby M, Gilkes AF, Marrin C, et al. The prognostic relevance of cer Chemother Pharmacol. 2018;81:171-178.
flt3 and npm1 mutations on older patients treated intensively or 75. Lancet J, Uy G, Cortes J, et al. CPX-351(cytarabine and daunorubicin)
non-intensively: a study of 1312 patients in the UK NCRI AML16 liposome for injection vs. conventional cytarabine plus daunorubicin
trial. Leukemia. 2014;28:1953-1959. in older patients with newly diagnosed secondary acute myeloid leu-
56. Stone RM, Mandrekar SJ, Sanford BL, et al. Midostaurin plus chemo- kemia. J Clin Oncol . 2018;36(26):2684-2692.
therapy for acute myeloid leukemia with a FLT3 mutation. N Engl J 76. Lancet JE, Hoering A, Uy GL, et al. Survival following allogeneic
Med. 2017;377:454-464. hematopoietic cell transplantation in older high-risk acute myeloid
57. Larson RA, Mandrekar SJ, Sanford BL, et al. An analysis of mainte- leukemia patients initially treated with CPX-351 liposome injection
nance therapy and post-Midostaurin outcomes in the international versus standard Cytarabine and Daunorubicin: subgroup analysis of a
prospective randomized, placebo-controlled, double-blind trial large phase III trial. Blood. 2016;128:906-906.
(CALGB 10603/RATIFY [alliance]) for newly diagnosed acute mye- 77. Uy GL, Lancet JE, Cortes JE, et al. Multivariate efficacy analysis of a
loid leukemia (AML) patients with FLT3 mutations. Blood. 2017; randomized, phase 3 study of CPX-351 versus 7+3 in older adults
130:145. with treatment-related acute myeloid leukemia (AML) or AML with
58. Röllig C, Serve H, Huttmann A, et al. Addition of sorafenib versus pla- myelodysplasia-related changes. Blood. 2017;130:2647-2647.
cebo to standard therapy in patients aged 60 years or younger with 78. Nardi V, Winkfield KM, Ok CY, et al. Acute myeloid leukemia and
newly diagnosed acute myeloid leukaemia (SORAML): a multicentre, myelodysplastic syndromes after radiation therapy are similar to de
phase 2, randomised controlled trial. Lancet Oncol. 2015;16: novo disease and differ from other therapy-related myeloid neo-
1691-1699. plasms. J Clin Oncol. 2012;30:2340-2347.

/
ESTEY 1281

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].

FIGURE 7 Outcomes in people according to whether they had persistence of NPM1 mutations (MRD-positive) in blood after initial 2 cycles of
chemotherapy (see reference 96)

mutations could be divided into those associated with ARCH7 (ie, in 28% of patients and usually in VAF consistent with MRD. Persis-
mutations in DNMT3a, TET2, and ASXL1) and others. The ARCH- tence of non-ARCH, but not ARCH, mutations were associated with
related mutations were often present in varying allele frequencies higher cumulative incidence of relapse and shorter survival and
(VAF) suggesting presence in >5% of marrow cells consistent with relapse-free survival. These associations were also found in a “valida-
presence of non-leukemic clones. Non-ARCH mutations were found tion” population and were independent of ELN 2017, age, number of

/
1290 ESTEY

79. Walter RB, Othus M, Orlowski KF, et al. Unsatisfactory efficacy in allogeneic transplantation. Biol Blood Marrow Transplant. 2017;23:
randomized study of reduced-dose CPX-351 for medically less fit 1064-1071.
adults with newly diagnosed acute myeloid leukemia or other 100. Morita K, Kantarjian HM, Wang F, et al. Clearance of somatic muta-
high-grade myeloid neoplasm. Haematologica. 2018;103:e106-e109. tions at remission and the risk of relapse in acute myeloid leukemia. J
80. DiNardo CD, Pollyea DA, Jonas BA, et al. Updated safety and effi- Clin Oncol. 2018 Jun 20;36(18):1788-1797.
cacy of Venetoclax with Decitabine or Azacitidine in treatment- 101. Boddu P, Jorgensen J, Kantarjian H, et al. Achievement of a negative
naive, elderly patients with acute myeloid leukemia. Blood. 2017;130: minimal residual disease state after hypomethylating agent therapy
2628-2628. in older patients with AML reduces the risk of relapse. Leukemia.
81. Wei A, Strickland SA, Roboz GJ, et al. Phase 1/2 study of Venetoclax 2018;32:241-244.
with low-dose Cytarabine in treatment-naive, elderly patients with 102. Othus M, Wood BL, Stirewalt DL, et al. Effect of measurable ('mini-
acute myeloid leukemia unfit for intensive chemotherapy: 1-year mal') residual disease (MRD) information on prediction of relapse and
outcomes. Blood. 2017;130:890-890. survival in adult acute myeloid leukemia. Leukemia. 2016;30:
82. Welch JS, Petti AA, Miller CA, et al. TP53 and Decitabine in acute 2080-2083.
myeloid leukemia and myelodysplastic syndromes. N Engl J Med. 103. Salk JJ, Schmitt MW, Loeb LA. Enhancing the accuracy of
2016;375:2023-2036. next-generation sequencing for detecting rare and subclonal muta-
83. Paschka P, Schlenk RF, Weber D, et al. Adding dasatinib to intensive tions. Nat Rev Genet . 2018;19:269-285.
treatment in core-binding factor acute myeloid leukemia-results of 104. Terwijn M, Zeijlemaker W, Kelder A, et al. Leukemic stem cell fre-
quency: a strong biomarker for clinical outcome in acute myeloid leu-

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


the AMLSG 11-08 trial. Leukemia. 2018 Apr 17. https://doi.org/10.
1038/s41375-018-0129-6 In press. kemia. PloS one. 2014;9:e107587.
84. Marcucci G, Geyer S, Zhao W, et al. Adding KIT inhibitor Dasatinib 105. Freeman SD, Virgo P, Couzens S, et al. Prognostic relevance of treat-
(DAS) to chemotherapy overcomes the negative impact of KIT ment response measured by flow cytometric residual disease detec-
mutation/over-expression in Core binding factor (CBF) acute myeloid tion in older patients with acute myeloid leukemia. J Clin Oncol.
leukemia (AML): results from CALGB 10801 (alliance). Blood. 2014; 2013;31:4123-4131.
106. stgård LSG, Lund JL, Norgaard JM, et al. Impact of allogeneic stem
124:8.
85. Cancer Genome Atlas Research Network, Ley TJ, Miller C, cell transplantation in first complete remission in acute myeloid leu-
kemia: a national population-based cohort study. Biol Blood Marrow
et al. Genomic and epigenomic landscapes of adult de novo acute
Transplant. 2018;24:314-323.
myeloid leukemia. N Engl J Med. 2013;368(22):2059-2074.
107. Wheatley K, Gray R. Commentary: Mendelian randomization—an
86. Bruker B, Byrd J, Levine R. Beat AML. http://www.lls.org/beat-aml/
update on its use to evaluate allogeneic stem cell transplantation in
beat-aml-master-trial-more-information.
leukaemia. Int J Epidemiol. 2004;33:15-17.
87. Estey E, Levine RL, Löwenberg B. Current challenges in clinical devel-
108. Freeman SD, Hills RK, Virgo P, et al. Measurable residual disease at
opment of “targeted therapies”: the case of acute myeloid leukemia.
induction redefines partial response in acute myeloid leukemia and
Blood. 2015;125:2461-2466.
stratifies outcomes in patients at standard risk without NPM1 muta-
88. Montalban-Bravo G, Huang X, Naqvi K, et al. A clinical trial for
tions. J Clin Oncol . 2018;36:1486-1497.
patients with acute myeloid leukemia or myelodysplastic syndromes
109. Burnett AK, Goldstone A, Hills RK, et al. Curability of patients with
not eligible for standard clinical trials. Leukemia . 2017;31:318-324.
acute myeloid leukemia who did not undergo transplantation in first
89. Jaramillo S, Benner A, Krauter J, et al. Condensed versus standard
remission. J Clin Oncol. 2013;31:1293-1301.
schedule of high-dose cytarabine consolidation therapy with pegfil-
110. Zhu HH, Zhang XH, Qin YZ, et al. MRD-directed risk stratification
grastim growth factor support in acute myeloid leukemia. Blood Can-
treatment may improve outcomes of t(8,21) AML in the first com-
cer J. 2017;7:e564.
plete remission: results from the AML05 multicenter trial. Blood.
90. Löwenberg B. Sense and nonsense of high-dose cytarabine for acute
2013;121:4056-4062.
myeloid leukemia. Blood . 2013;121:26-28. 111. Elsawy M, Sorror ML. Up-to-date tools for risk assessment before
91. Thomas X, de Botton S, Chevret S, et al. Randomized phase II study
allogeneic hematopoietic cell transplantation. Bone Marrow Trans-
of Clofarabine-based consolidation for younger adults with acute
plant. 2016;51:1283-1300.
myeloid leukemia in first remission. J Clin Oncol. 2017;35: 112. Sorror ML, Storb RF, Sandmaier BM, et al. Comorbidity-age index: a
1223-1230. clinical measure of biologic age before allogeneic hematopoietic cell
92. vlisen AK, Oest A, Bendtsen MD, et al. Stringent or nonstringent transplantation. J Clin Oncol. 2014;32:3249-3256.
complete remission and prognosis in acute myeloid leukemia: a Dan- 113. Appelbaum FR. Impact of allogeneic hematopoietic cell transplanta-
ish population-based study. Blood Adv. 2018;2:559-564. tion on the outcome of older patients with acute myeloid leukemia.
93. Schuurhuis GJ, Heuser M, Freeman S, et al. Minimal/measurable Best Pract Res Clin Haematol. 2017;30:320-326.
residual disease in AML: a consensus document from the European 114. Muffly L, Pasquini MC, Martens M, et al. Increasing use of allogeneic
LeukemiaNet MRD working party. Blood. 2018;131:1275-1291. hematopoietic cell transplantation in patients aged 70 years and
94. Terwijn M, van Putten WL, Kelder A, et al. High prognostic impact of older in the United States. Blood . 2017;130:1156-1164.
flow cytometric minimal residual disease detection in acute myeloid 115. Gooley TA, Chien JW, Pergam SA, et al. Reduced mortality after allo-
leukemia: data from the HOVON/SAKK AML 42A study. J Clin Oncol. geneic hematopoietic-cell transplantation. N Engl J Med. 2010;363:
2013;31:3889-3897. 2091-2101.
95. Araki D, Wood BL, Othus M, et al. Allogeneic hematopoietic cell 116. Scott BL, Pasquini MC, Logan BR, et al. Myeloablative versus
transplantation for acute myeloid leukemia: time to move toward a reduced-intensity hematopoietic cell transplantation for acute mye-
minimal residual disease-based definition of complete remission? J loid leukemia and myelodysplastic syndromes. J Clin Oncol. 2017;35:
Clin Oncol. 2016;34:329-336. 1154-1161.
96. Ivey A, Hills RK, Simpson MA, et al. Assessment of minimal residual 117. Fasslrinner F, Schetelig J, Burchert A, et al. Long-term efficacy of
disease in standard-risk AML. N Engl J Med. 2016;374:422-433. reduced-intensity versus myeloablative conditioning before alloge-
97. Jourdan E, Boissel N, Chevret S, et al. Prospective evaluation of gene neic haemopoietic cell transplantation in patients with acute myeloid
mutations and minimal residual disease in patients with core binding leukaemia in first complete remission: retrospective follow-up of an
factor acute myeloid leukemia. Blood. 2013;121:2213-2223. open-label, randomised phase 3 trial. Lancet Haematol. 2018;5:
98. Jongen-Lavrencic M, Grob T, Hanekamp D, et al. Molecular minimal e161-e169.
residual disease in acute myeloid leukemia. N Engl J Med. 2018;378: 118. Kroger N, Iacobelli S, Franke GN, et al. Dose-reduced versus stan-
1189-1199. dard conditioning followed by allogeneic stem-cell transplantation
99. Getta BM, Devlin SM, Levine RL, et al. Multicolor flow cytometry for patients with myelodysplastic syndrome: a prospective random-
and multigene next-generation sequencing are complementary and ized phase III study of the EBMT (RICMAC trial). J Clin Oncol. 2017;
highly predictive for relapse in acute myeloid leukemia after 35:2157-2164.

/
1282 ESTEY

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


FIGURE 8 Relation between relapse and MRD as detected by MFC and NGS (see reference 98)

courses needed to attain CR/CRi, and when in the 21 day to 4 month complementary fashion. These approaches might improve the predic-
window testing was done. It remains unclear whether persistence of tive value of a positive MRD test (PPV) for subsequent morphologic
some non-ARCH related mutations is more prognostic than persis- relapse which averages about 80% and the predictive value of a nega-
tence of others; VAF thresholds for positivity may vary by gene. tive MRD test for remaining in CR (NPV) which is approximately 60%-
Examining the 340 of the 430 adults in whom MFC as well as 80%, that is, 20%-40% of patients deemed MRD negative will relapse,
NGS testing was performed, Jongen-Lavrenic et al. found concor- with PPV higher and NPV lower in older patients 105 and perhaps after
98
dance between MFC and NGS in 69%. In the discordant cases, NGS less intense therapy (see figure 1 in reference 101). But even today, it
was positive in about 60% and MFC in about 40%. Combining NGS is clear MRD detection using PCR and MFC has a role in modifying
and MFC appeared to improve the sensitivity/predictive value of a post-remission therapy, previously based solely on pretreatment vari-
positive MRD test (Figure 8). 98 Similar results have been reported for ables, for example, ELN 2017. Indeed, the recognition by ELN 2017 of
prediction of post HCT relapse by Getta et al, 99 while Morita CR with MRD as distinct entity18 implies the desirability of treating it
et al. reported post-treatment clearance of mutations might help strat- in different fashion than CR without MRD. Allogeneic HCT provides
ify MFC negative patients according to risk of relapse.100 an example.
The great majority of the results noted above have been obtained
after “intensive” therapy  HCT. However, Boddu et al. have sug-
5.2 | Allogeneic HCT
gested persistence of MFC-detected MRD after 3 months in CR or
CRi is associated with relapse after therapy with azacitidine or decita- Without accounting for MRD, HCT in CR1 is often recommended for
bine in a population whose median age was 75 years (Supporting fit patients, usually age <70, with ELN 2017 intermediate- and
101 adverse-, but not favorable-risk disease. For example, stgård
Information Figure 4). Nonetheless, while it is clear detection of
MRD provides invaluable information, at least after intensive therapy et al. used the Danish Acute Leukemia Registry assuring virtual com-
 HCT, addition of MFC status at time of CR to pretreatment vari- plete data collection on virtually all 1031 adults with intermediate—or
ables had only modest quantitative effect on assessing chance of adverse-risk cytogenetics age <70 who entered CR between 2000
relapse, 102 suggesting the likely need to consider changes in MRD and 2014, of whom 19% received HCT in CR1. 106 As expected, HCT
over time rather than MRD at only a single instance, to develop new patients tended to be younger with fewer comorbidities but more
103
means for MRD detection for example, “duplex sequencing” or often had adverse cytogenetics. A total of 70%-75% of patients
analysis of leukemia “stem cells” 104 and to use these methods in received at least 1 “consolidation” chemotherapy course, with 40% of

/
ESTEY 1291

119. Deeg HJ, Stevens EA, Salit RB, et al. Transplant conditioning with 138. DiNardo CD, Rausch CR, Benton C, et al. Clinical experience with
Treosulfan/Fludarabine with or without Total body irradiation: a ran- the BCL2-inhibitor venetoclax in combination therapy for relapsed
domized phase II trial in patients with myelodysplastic syndrome and and refractory acute myeloid leukemia and related myeloid malignan-
acute myeloid leukemia. Biol Blood Marrow Transplant. 2018;24: cies. Am J Hematol. 2018;93:401-407.
956-963. 139. Feldman EJ, Lancet JE, Kolitz JE, et al. First-in-man study of
120. Jurcic JG, Rosenblat TL. Targeted alpha-particle immunotherapy for CPX-351: a liposomal carrier containing cytarabine and daunorubicin
acute myeloid leukemia. Am Soc Clin Oncol Educ Book. ASCO 50th in a fixed 5:1 molar ratio for the treatment of relapsed and refractory
Annual Meeting. 2014;34:e126-e131. acute myeloid leukemia. J Clin Oncol. 2011;29:979-985.
121. Appelbaum FR. Alternative donor transplantation for adults with 140. Stein EM, DiNardo CD, Pollyea DA, et al. Enasidenib in mutant IDH2
acute leukemia. Best Pract Res Clin Haematol . 2014;27:272-277. relapsed or refractory acute myeloid leukemia. Blood . 2017;130:
122. Tsai SB, Rhodes J, Liu H, et al. Reduced-intensity allogeneic trans- 722-731.
plant for acute myeloid leukemia and myelodysplastic syndrome 141. Fathi AT, DiNardo CD, Kline I, et al. Differentiation syndrome associ-
using combined CD34-selected Haploidentical graft and a single ated with Enasidenib, a selective inhibitor of mutant Isocitrate dehy-
umbilical cord unit compared with matched unrelated donor stem drogenase 2: analysis of a phase 1/2 study. JAMA Oncol . 2018;4(8):
cells in older adults. Biol Blood Marrow Transplant. 2018;24: 1106-1110.
997-1004. 142. Paschka P, Schlenk R, Weber D, et al. Outcome of patients with
123. Lee SJ, Logan B, Westervelt P, et al. Comparison of patient-reported refractory or relapsed AML with IDH1 and IDH2 mutations after
outcomes in 5-year survivors who received bone marrow vs periph- conventional salvage therapy: a study of the German-Austrian AML

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


eral blood unrelated donor transplantation: long-term follow-up of a study group; 21 st congress of the European Hematology Association.
randomized clinical trial. JAMA Oncol . 2016;2:1583-1589. Copenhagen. 2016; Abstract S809. Leukemia. 2018 Apr 17. https://
124. Milano F, Appelbaum FR, Delaney C. Cord-blood transplantation in doi.org/10.1038/s41375-018-0129-6.
patients with minimal residual disease. N Engl J Med. 2016;375: 143. Stein EM, Garcia-Manero G, Rizzieri DA, et al. The DOT1L inhibitor
2204-2205. pinometostat reduces H3K79 methylation and has modest clinical activ-
125. Estey E, Gale RP. Acute myeloid leukemia therapy and the chosen ity in adult acute leukemia. Blood. 2018 Jun 14;131(24):2661-2669.
people. Leukemia. 2017;31:269-271. 144. DiNardo C, Stein E, de Botton S, et al. Durable remissions with ivosi-
126. Pagel JM, Othus M, Garcia-Manero G, et al. Feasibility of allogeneic denib in IDH1-mutated relapsed or refractory AML. New Engl J Med.
hematopoietic cell transplantation among high-risk AML patients in 2018;378:2386-2398.
first complete remission: results of the transplant objective from the 145. Stein EM, DiNardo CD, Mims AS, et al. Ivosidenib or Enasidenib com-
SWOG (S1203) randomized phase III study of induction therapy bined with standard induction chemotherapy is well tolerated and
using standard 7+3 therapy or Idarubicin with high-dose Cytarabine active in patients with newly diagnosed AML with an IDH1 or IDH2
(IA) versus IA plus Vorinostat. Blood. 2016;128:1166-1166. mutation: initial results from a phase 1 trial. Blood. 2017;130:726-726.
127. Mawad R, Gooley TA, Sandhu V, et al. Frequency of allogeneic 146. Quek L, David M, Kennedy A, et al. Clonal heterogeneity in differen-
hematopoietic cell transplantation among patients with high- or tiation response and resistance to the IDH2 inhibitor Enasidenib in
intermediate-risk acute myeloid leukemia in first complete remission. acute myeloid leukemia. Blood. 2017;130:724-724.
J Clin Oncol. 2013;31:3883-3888. 147. Lichtenegger FS, Krupka C, Kohnke T, et al. Immunotherapy for
128. Cornelissen JJ, Blaise D. Hematopoietic stem cell transplantation for acute myeloid leukemia. Semin Hematol. 2015;52:207-214.
patients with AML in first complete remission. Blood. 2016;127: 148. Daver N, Boddu P, Garcia-Manero G, et al. Hypomethylating agents in
62-70. combination with immune checkpoint inhibitors in acute myeloid leuke-
129. Schlenk RF, Taskesen E, van Norden Y, et al. The value of allogeneic mia and myelodysplastic syndromes. Leukemia. 2018;32:1094-1105.
and autologous hematopoietic stem cell transplantation in prognosti- 149. Middeke JM, Herbst R, Parmentier S, et al. Long-term follow-up and
cally favorable acute myeloid leukemia with double mutant CEBPA. impact of comorbidity before allogeneic hematopoietic stem cell
Blood. 2013;122:1576-1582. transplantation in patients with relapsed or refractory acute myeloid
130. Othus M, Mukherjee S, Sekeres MA, et al. Prediction of CR following leukemia-lessons learned from the prospective BRIDGE trial. Biol
a second course of ‘7+3’ in patients with newly diagnosed acute Blood Marrow Transplant. 2017;23:1491-1497.
myeloid leukemia not in CR after a first course. Leukemia. 2016;30: 150. Chen X, Xie H, Wood BL, et al. Relation of clinical response and mini-
1779-1780. mal residual disease and their prognostic impact on outcome in acute
131. Ravandi F, Cortes J, Faderl S, et al. Characteristics and outcome of myeloid leukemia. J Clin Oncol. 2015;33:1258-1264.
patients with acute myeloid leukemia refractory to 1 cycle of 151. Craddock C, Labopin M, Robin M, et al. Clinical activity of azacitidine
high-dose cytarabine-based induction chemotherapy. Blood. 2010; in patients who relapse after allogeneic stem cell transplantation for
116:5818-5823. quiz 6153. acute myeloid leukemia. Haematologica. 2016;101:879-883.
132. Walter RB, Othus M, Löwenberg B, et al. Empiric definition of eligi- 152. Mielcarek M, Storer BE, Flowers ME, et al. Outcomes among patients
bility criteria for clinical trials in relapsed/refractory acute myeloid with recurrent high-risk hematologic malignancies after allogeneic
leukemia: analysis of 1,892 patients from HOVON/SAKK and hematopoietic cell transplantation. Biol Blood Marrow Transplant.
SWOG. Haematologica. 2015;100:e409-e411. 2007;13:1160-1168.
133. Thol F, Schlenk RF, Heuser M, et al. How I treat refractory and early 153. Zhou Y, Wood BL, Walter RB, et al. Is there a need for morphologic
relapsed acute myeloid leukemia. Blood . 2015;126:319-327. exam to detect relapse in AML if multi-parameter flow cytometry is
134. Breems DA, Van Putten WL, Huijgens PC, et al. Prognostic index for employed? Leukemia. 2017;31:2536-2537.
adult patients with acute myeloid leukemia in first relapse. J Clin
Oncol. 2005;23:1969-1978.
135. Wong TN, Miller CA, Klco JM, et al. Rapid expansion of preexisting SUPPORTING I NFORMATION
nonleukemic hematopoietic clones frequently follows induction ther- Additional supporting information may be found online in the Sup-
apy for de novo AML. Blood . 2016;127:893-897.
136. Roboz GJ, Rosenblat T, Arellano M, et al. International randomized
porting Information section at the end of the article.
phase III study of elacytarabine versus investigator choice in patients
with relapsed/refractory acute myeloid leukemia. J Clin Oncol. 2014;
32:1919-1926. How to cite this article: Estey EH. Acute myeloid leukemia:
137. Kantarjian HM, DiNardo CD, Nogueras-Gonzalez GM, et al. Results 2019 update on risk-stratification and management. Am
of second salvage therapy in 673 adults with acute myelogenous leu- J Hematol. 2018;93:1267–1291. https://doi.org/10.1002/ajh.
kemia treated at a single institution since 2000. Cancer . 2018;124:
2534-2540. 25214

/
ESTEY 1283

FIGURE 9 Effect of HCT in CR1 on survival. HR <1.0 indicates HCT better than continued chemotherapy. Adjusted HR accounts for age (up to
70 years), PS, comorbidities, de novo vs secondary AML and cytogenetics (see reference 106)

chemotherapy-only and 24% of HCT patients receiving two such in CR1, at relapse, or in CR2.109 However, it has been pointed out that

Printed by [Wiley Online Library - 123.253.099.020 - /doi/epdf/10.1002/ajh.25214] at [13/02/2021].


courses, corresponding to a median time from CR of to HCT of about the CR2 rate in such patients was unusually high (54%),109 suggesting
4 months. Two-thirds of transplanted patients received reduced the influence of selection bias. Nonetheless, since this report included
intensity conditioning (RIC), the remainder myeloablative conditioning patients with or without MRD, it suggests there, at least, may be no
(MAC); approximately half the donors were matched siblings (MSD), need for allogeneic HCT in intermediate risk patients without MRD
the remainder matched unrelated (MUD). Using Mantel-Byar method- after the first two courses of therapy. Certainly, the expectation of
ology and landmark analyses to account for the “guarantee time” HCT post-HCT NRM would need to be very low to justify HCT in such
patients, but not chemotherapy only patients, must live before patients. In contrast, in people in the ELN intermediate group with
107
HCT, cumulative incidence of relapse was lower, and survival and MRD, ELN adverse group without, and particularly ELN adverse risk
relapse-free survival longer, in HCT recipients with either intermedi- group with MRD, higher risks of post-HCT NRM might be acceptable
ate or adverse cytogenetics or age <60 or 60-69 (Figure 9). Median given HCT's greater potential benefit. For example, Thomas
follow up was 2.2 years in all patients, with relapses occurring in 49% et al. demonstrated the beneficial effect of addition of clofarabine to
and deaths in 68% of chemotherapy patients vs 24% and 42% for post-remission therapy was substantially abrogated by receipt of
HCT patients. Median RFS was 293 days (interquartile range HCT 91 (Supporting Information Figure 5).
142-910) for chemotherapy only vs 1067 (IQ range 397-2696) for Investigating the potential utility of using MRD to decide
HCT with median survival times of 476 days (IQ 167-1589) and 1173 between transplant and nontransplant approaches, Zhu et al.110
(IQ 474-2859) assigned patients with t(8;21) who were PCR positive after two “con-
Might accounting for MRD affect the recommendation for HCT solidation” courses (high risk) to receive HCT while PCR negative
in ELN adverse or intermediate but not “favorable” patients? Specifi- patients (low risk) were to receive autologous HCT or further chemo-
cally, it seems plausible the presence of MRD might be used to therapy. High risk patients who received HCT had better outcomes
“upstage,” and the absence of MRD to “downstage,” the pretreatment than high risk patients who did not, while outcomes were similar in
ELN risk score, although it appears the influence of MRD status may low risk patients regardless of whether they received HCT
be less in the ELN adverse risk group. 108 More attention would be (Supporting Information Figure 6). Only MRD (yes/no) and MRD
given the possibility of allogeneic HCT in upstaged patients and less in adapted therapy (yes/no) affected outcome. However, confounding
down-staged patients (Table 5). For example, patients in the ELN factors exist, and the study was not randomized. It remains to be seen
2017 “favorable” group pretreatment but who have MRD after whether MRD-directed therapy will sufficiently reduce MRD, thus
2 cycles of therapy, as assessed by MFC or PCR, have risk of morpho- making it possible to examine whether patients in whom such reduc-
logic relapse within 1-2 years of approximately 50%, not dramatically tion, occurs, for example pre-HCT, will have better outcomes than
different than the 60% seen in patients initially in the ELN adverse patients who receive HCT without prior attempts to reduce MRD.
group but who have no MRD after 2 cycles. The risk of nonrelapse Although there are alternatives, the hematopoietic cell transplan-
mortality (NRM) patients/physicians might be willing to accept after tation comorbidity index (HCT-CI) effectively estimates NRM post-
HCT depends not only on the risk of relapse without HCT but on the HCT.111 Although as with induction therapy, age alone even up to
degree to which HCT might reduce this risk. It is generally accepted 75-80 appears less important than comorbidities in influencing post
HCT reduces relapse risk, as in stgärd et al.106 and numerous other HCT NRM, 112 the proportion of adults with AML receiving HCT
publications. It is also well-known pre HCT MRD is highly associated (approximately 30% at any time in their course), while unaffected by
with post HCT relapse, much as is the case without HCT. What seems age up to 70, falls considerably once age 70 is reached. 113 Nonethe-
less clear is whether the reduction in relapse risk following HCT varies less Muffly et al.114 have noted the increasing use of HCT for AML
by MRD status. Preliminary data from the NCRI group in the UK sug- patients in their 70s (Supporting Information Figure 7A) concomitant
gest HCT in CR1 has a beneficial effect in intermediate risk patients in with better survival and disease-free survival in such patients
the presence but not the absence of MRD.108 These data are in partial (Supporting Information Figure 7B). The same is true for HCT in gen-
accord with the NCRI's report that survival was the same in people eral. 115 Attempts to improve the HCT-CI's performance include incor-
with intermediate risk AML regardless of whether they received HCT poration of frailty indices and markers of inflammation.111

You might also like