You are on page 1of 13

Current Oncology Reports (2021) 23:120

https://doi.org/10.1007/s11912-021-01108-9

LEUKEMIA (A AGUAYO, SECTION EDITOR)

3+7 Combined Chemotherapy for Acute Myeloid Leukemia: Is It Time


to Say Goodbye?
Kenny Tang 1 & Andre C Schuh 1 & Karen WL Yee 1

Accepted: 16 February 2021


# The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature 2021

Abstract
Purpose of Review With the recent approval of multiple new drugs for the treatment of acute myeloid leukemia (AML), the
relevance of conventional treatment approaches, such as daunorubicin and cytarabine (“3+7”) induction chemotherapy, has been
challenged. We review the AML risk stratification, the efficacy of the newly approved drugs, and the role of “3+7”.
Recent Findings Treatment of AML is becoming more niched with specific subtypes more appropriately treated with
gemtuzumab, midostaurin, and CPX-351. Although lower intensity therapies can yield high response rates, they are less efficient
at preventing relapses. The only curative potential for poor-risk AML is still an allogeneic stem cell transplant.
Summary The number of AML subtypes where 3+7 alone is an appropriate therapeutic option is shrinking. However, it remains
the backbone for combination therapy with newer agents in patients suitable for intensive chemotherapy.

Keywords Acute myeloid leukemia . cytarabine . CPX-351 . Daunorubicin . Midostaurin . gemtuzumab ozogamicin .
Venetoclax

Introduction general treatment approach has remained largely unchanged


over the last 50 years. Although there is no standard induction
Acute myeloid leukemia (AML) is an aggressive, genetically regimen for patients with untreated AML who are suitable for
heterogeneous group of malignant disorders characterized by intensive therapy, the most widely used regimen has been
infiltration of the bone marrow, blood, and other tissues by daunorubicin (DNR) intravenously for 3 days and cytarabine
clonally related myeloblasts. AML is cured in 35–40% of at a dose of 100–200 mg/m2 intravenously by continuous
patients ≤ 60 years of age and in 5–15% of patients >60 years. infusion for 7 days (i.e., 3+7) [8, 9•]. This regimen produces
Although the cytogenetic heterogeneity of AML has been a complete remission (CR) in 60–80% of younger adults with
recognized for more than 30 years, the molecular heterogene- 5-year overall survival (OS) of ~ 40% [10–12]. Among older
ity of this disease has only become increasingly apparent over adults (≥ 60 years), 40–60% will achieve a CR, but most will
the past 15 years [1, 2, 3••, 4, 5, 6••, 7]. Nonetheless, the relapse with few surviving beyond 2 years [13, 14].
Most patients with AML who achieve a CR will eventually
relapse due to the existence of subpopulations of AML cells
resistant to standard chemotherapy. A smaller subset of patients
This article is part of the Topical collection on Leukemia
will not achieve a CR. Attempts to overcome or prevent resis-
tance have led to the development of drugs targeting specific
* Karen WL Yee
karen.yee@uhn.ca mechanisms of resistance to be administered with traditional
intensive (e.g., 3+7 backbone) and lower intensity chemother-
Kenny Tang apy regimens or as single or combination targeted therapies.
kenny.tang@uhn.ca
While the recent approval of multiple new drugs for the treat-
Andre C Schuh ment of AML is changing conventional treatment approaches,
andre.schuh@uhn.ca
and their successes have raised the notion that “‘3+7’ is dead”,
1 we would argue that in spite of these advances, “3+7” will be
Division of Medical Oncology and Hematology, University Health
Network – Princess Margaret Cancer Centre, 610 University Avenue, with us for the foreseeable future (Table 1) [15••, 16••, 17, 18••,
700 U 6th Floor, Toronto, Ontario M5G 2M9, Canada 19••, 20, 21, 22••, 23, 24••, 25••].
120 Page 2 of 13 Curr Oncol Rep (2021) 23:120

In this article, we will review the AML risk stratification, may be due, in part, to lack of a standardized assay. However,
the efficacy of the newly approved drugs, and finally discuss the prognostic impact of FLT3-ITD mutations in a NPM1-
why these new drugs are not yet able to replace “3+7”. mutated patient is also modified by co-occurrence of other
mutations. For example, the effect of a FLT3-ITD mutation
on OS may be more pronounced in patients with concomitant
Risk Stratification of AML NPM1 and DNMT3A mutations as opposed to an isolated
NPM1 mutation, regardless of the FLT3-ITD allelic ratio
In recent years, the European LeukemiaNet (ELN) risk cate- [30]. Therefore, the significance of the FLT3-ITD allelic ratio
gory, based on cytogenetics and mutational status of specific and its role in guiding treatment decisions (e.g., allogeneic
genes, has become the most widely used risk classification stem cell transplant (alloSCT) in a patient with a low FLT3-
system to guide AML treatment-making decisions (Table 2) ITD allelic ratio) and/or determining prognosis remains
[3••]. However, there remain a number of questions unclear. Similarly, RUNX1 [6••, 31–34] and ASXL1 [6••,
concerning the ELN genetic categories. FLT3-ITD mutations 35, 36, 37–41] mutations, in the absence of favorable
are associated with poor OS and a higher risk of relapse [7, 26, genetic risk features, have been shown to be independent
27]. While ELN defines the risk of patients based on both prognostic factors predicting an inferior response rate and
NPM1 mutational status and FLT3-ITD allelic burden, i.e., OS in some, but not all, studies.
low FLT3-ITD: FLT3WT allelic ratio (< 0.5) vs high allelic Other prognostic factors that contribute to prediction of
ratio (> 0.5), the prognostic significance of FLT3-ITD allelic outcome are not necessarily captured in the ELN risk catego-
burden has not been consistently demonstrated [28, 29]. This ries, including a history of antecedent hematological disorders

Table 1 Recent drug approvals for first-line therapy for AML

Drug Indication Regulatory status Reference

Intensive therapy – first line


CPX-351 (liposomal Treatment of adults with newly diagnosed therapy-related AML (t-AML) FDA (2017); EMA Lancet 2018 [15••]
daunorubicin + or AML with myelodysplasia-related changes (AML-MRC)a (2018)
cytarabine)
Gemtuzumab Treatment of newly diagnosed CD33-positive AML in adults in FDA (2017); EMA Castaigne 2012;
ozogamicin (GO) combination with daunorubicin and cytarabine for adults with newly (2018); HC (2019) Lambert 2019
diagnosed AML, except acute promyelocytic leukemia (APL)b [16••, 17]
Midostaurin For use with standard cytarabine and daunorubicin induction and FDA (2017); EMA Stone 2017 [18••]
cytarabine consolidation for the treatment of adults with newly (2017); HC (2017)
diagnosed FLT3-mutated AMLa,c,d
Less intensive therapy – first line
Ivosidenib For newly-diagnosed AML with a susceptible IDH1 mutation, as detected FDA (2019) Roboz 2020 [19••]
by an FDA-approved test, in patients who are at least 75 years old or who
have comorbidities that preclude the use of intensive induction
chemotherapy
Venetoclax (VEN) + Venetoclax in combination with AZA or DEC or LDAC for the treatment FDA (accelerated 2018; DiNardo 2018,
azacitidine (AZA) of newly diagnosed AML in adults who are > 75 years, or who have regular 2020); HC 2019, 2020 [20,
comorbidities that preclude use of intensive induction chemotherapy (2020) 21, 22••]
Venetoclax (VEN) + Venetoclax in combination with AZA or DEC or LDAC for the treatment FDA (accelerated 2018; DiNardo 2018, 2019
decitabine (DEC) of newly diagnosed AML in adults who are > 75 years, or who have regular 2020) [20, 21]
comorbidities that preclude use of intensive induction chemotherapy
Venetoclax (VEN) + Venetoclax in combination with AZA or DEC or LDAC for the treatment FDA (accelerated 2018; Wei 2019, 2020 [23,
low-dose cytarabine of newly diagnosed AML in adults who are > 75 years, or who have regular 2020); HC 24••]
(LDAC) comorbidities that preclude use of intensive induction chemotherapy (2020)
Low-dose cytarabine For use in combination with LDAC, for newly diagnosed AML in patients FDA (2018); EMA Cortes 2019 [25••]
(LDAC) + glasdegib who are > 75 years or who have comorbidities that preclude intensive (2020); HC (2020)
induction chemotherapy

EMA European Medicine Agencies; FDA US Food and Drug Administration; HC Health Canada
a
No age restriction; b EMA approval for age > 15 years; c drug is FDA approved for use with a companion diagnostic, the LeukoStrat CDx FLT3
mutation assay, which is used to detect the FLT3 mutation in patients with AML; d for patients in complete response, the EMA has approved midostaurin
as single agent maintenance therapy
Curr Oncol Rep (2021) 23:120 Page 3 of 13 120

Table 2 2017 ELN risk


classification of AML (Dohner Risk category Genetic abnormality
et al. 2017) [3••]
Favorable t(8;21)(q22;q22.1); RUNX1-RUNX1T1
inv(16)(p13.1q22) or t(16;16)(p13.1;q22); CBFB-MYH11
Mutated NPM1 without FLT3-ITD
Mutated NPM1 with FLT3-ITDlow†
Biallelic mutated CEBPA
Intermediate Mutated NPM1 and FLT3-ITDhigh†
Wild-type NPM1 without FLT3-ITD (without adverse-risk genetic lesions)
Wild-type NPM1 with FLT3-ITDlow† (without adverse-risk genetic lesions)
t(9;11)(p21.3;q23.3); MLLT3-KMT2A‡
Cytogenetic abnormalities not classified as favorable or adverse
Adverse t(6;9)(p23;q34.1); DEK-NUP214
t(v;11q23.3); KMT2A rearranged
t(9;22)(q34.1;q11.2); BCR-ABL1
inv(3)(q21.3q26.2) or t(3;3)(q21.3;q26.2); GATA2, MECOM (EVI1)
−5 or del(5q); −7; −17/abn(17p)
Complex karyotype,§ monosomal karyotype||
Wild-type NPM1 and FLT3-ITDhigh†
Mutated RUNX1¶
Mutated ASXL1¶
Mutated TP53#

Low low allelic ratio (< 0.5); high high allelic ratio (> 0.5)

The presence of t(9;11)(p21.3;q23.3) takes precedence over rare, concurrent adverse-risk gene mutations
§
Three or more unrelated chromosome abnormalities in the absence of 1 of the WHO-designated recurring
translocations or inversions, that is, t(8;21), inv [16] or t(16;16), t(9;11), t(v;11)(v;q23.3), t(6;9), inv(3) or
t(3;3); t(9;22)
||
Defined by the presence of 1 single monosomy (excluding loss of X or Y) in association with at least 1 additional
monosomy or structural chromosome abnormality (excluding core-binding factor AML)

These markers should not be used as an adverse prognostic marker if they co-occur with favorable-risk AML
subtypes.
#
TP53 mutations are significantly associated with AML with complex and monosomal karyotype.

(e.g., myelodysplastic syndrome [MDS], myelodysplastic Newly Approved Drugs


syndrome/myeloproliferative neoplasm, myeloproliferative
neoplasm [MPN]) or prior exposure to cytotoxic chemother- Intensive Therapy
apy or radiation for treatment of other disorders or malignan-
cies [5, 42–48]. Some of these genetic abnormalities and/or Gemtuzumab ozogamicin (GO) plus 3+7
clinical variables have been captured in a proposed genomic
classification of AML [6••] in the updated 2016 World Gemtuzumab ozogamicin (GO) is a humanized antibody con-
Health Organization (WHO) classification of myeloid jugated with the toxin calicheamicin that targets CD33. The
neoplasm and acute leukemia [5]. However, this classifi- Acute Leukemia French Association (ALFA)-0701 study in
cation system is not without controversy. For example, 280 older patients with de novo treatment-naïve (TN) AML
both therapy-related myeloid neoplasms and AML with demonstrated a survival benefit when GO 3 mg/m2 was ad-
myelodysplasia-related changes (AML-MRC) portend a ministered in 3 fractionated doses on days 1, 4, and 7 in con-
poor prognosis. However, recent data indicate not only junction with 3+7 induction chemotherapy compared to stan-
that morphologic criteria for AML-MRC may not carry dard 3+7 induction alone (OS2y 53.2% vs 41.9%; P = 0.0368)
the same weight as cytogenetic and mutational criteria, (Table 3) [16••, 17]. This was attributable to fewer relapses in
but that there is significant heterogeneity in outcomes of patients with favorable- or intermediate-risk cytogenetics.
these patients and that specific subgroups may benefit Thirty-three percent of patients had an NPM1 mutation and
preferentially from different therapies [49]. 6% CEBPA mutations. FLT3-ITD allelic frequency and
RUNX1, ASXL1, and TP53 mutational analysis were not
120 Page 4 of 13 Curr Oncol Rep (2021) 23:120

Table 3 Outcomes with intensive regimens

CPX-351 GO + 3+7 Midostaurin + 3+7


(N = 153) (N = 139) (N = 360)
Phase 3 Phase 3 Phase 3
Lancet 2018 [15] Castaigne 2012; Stone 2017 [18]
Lambert 2019 [16••, 17]

Characteristics
Median age, y (range) 67.8a 62.8 (59.3–66.8)b 48.6 (18–60.9) d
Age > 75y Not provided but no patients > 75 y 0b 0d
De novo AML 41 (26.8%) 139 (100%) Not provided
Therapy-related AML (tAML) 30 (19.6%) 0c 0c
Secondary AML (sAML) 82 (53.6%) 0c Not provided
Prior HMA 21 (13.7%) 0c 0
Cytogenetic risk group
Good 7 (4.9%) 3 (2%) 16/269 (5.9%)
Intermediate 64 (44.8%) 91 (65%) 231/269 (85.9%)
Poor 72 (50.3%) 28 (20%)d 22/269 (8.2%)
Gene mutations
FLT3-ITD/TKD 22/138 (15.9%) FLT3-ITD 22/137 (16%) FLT3-ITD 279 (77.5%) /
FLT3-TKD 81 (22.5%)
IDH1/2 Not provided Not provided Not provided
TP53 Not provided Not provided Not provided
Outcomes
CR + CRi
All 73 (47.7%; CR 37.3%) CR + CRp 113 (81%; CR 73%) CR 212/360 (59%); expanded
CR 244/360 (68%)
tAML 14/30 (46.7%; CR 36.7%) Not applicable Not applicable
sAML 36/82 (43.9%: CR 32.9%) Not applicable Not provided
Poor risk cytogenetics 31/72 (43.1%; CR 34.7%) 14/28 (50%; CR not provided) Not provided
Prior HMA 18/50 (36%; CR 26%) Not applicable Not applicable
FLT3-ITD/TKD 15/22 (68.2%; CR 54.5%) FLT3-ITD 21/22 CR 212/360 (59%); expanded
(95.5%; CR not provided) CR 244/360 (68%)
IDH1/2 Not provided Not done Not provided
TP53 Not provided Not done Not provided
Median duration of response, mos (95% CI) 6.93 Not specified Not specified
Median follow-up, mos 20.7 47.6 59
Median OS, mos (95% CI) 9.56 (6.60–11.86) 27.5 (21.4–45.6) 74.7 (31.5-not reached)
30-day mortality 5.9% 3.8% Not specified

CI confidence interval; CR complete remission; CRp CR with incomplete platelet recovery; CRi complete remission with incomplete count recovery; GO
gemtuzumab ozogamicin; HMA hypomethylating agent; mos months
a
Eligible age for enrolment onto study: 60–75 y; b eligible age for enrolment onto study: 50–70 y; c this group of patients were not eligible for the study;
d
cytogenetics not available in 17 patients (12%); d eligible age for enrolment onto study: 18- <60 y

performed. There was no difference in composite CR (CR + used with 3+7 was hemorrhage and infection. Persistent
CR without platelet recovery [CRp]) or CR rates (81% vs thrombocytopenia was more common in the GO arm (20.4%
75%; P = 0.25 and 73% vs 72%; P = not significant [NS]) vs 2%). Veno-occlusive disease/sinusoidal obstruction syn-
between the 2 groups. On long-term follow-up, although OS drome (VOD/SOS), including after alloSCT, was observed
favored GO, this was no longer statistically significant (medi- in 4.6% patients in the GO arm and 1.5% patients in the
an OS 27.5 months vs 21.8 months; P = 0.16) [17]. Subgroup control arm (P = 0.165) [17]. Thirty-nine patients received
analysis demonstrated a survival benefit for FLT3-ITD-mutat- an alloSCT in first CR or CRp (17 in the GO arm; 22 in the
ed AML patients. The most common side effect of GO when control arm), whereas 46 patients received an alloSCT without
Curr Oncol Rep (2021) 23:120 Page 5 of 13 120

achieving a CR or CRp (2 GO arm; 9 control arm) or after 30 days of ending protocol therapy were considered (68% vs
relapse (13 GO arm; 22 control arm). 59%; P = 0.04). Midostaurin improved OS (74.7 months vs
An individual patient data meta-analysis of 3325 patients 25.6 months; OS4y 51.4% vs 44.3%; P = 0.009) in both FLT3-
from 5 randomized trials [16••, 50–53] demonstrated an im- TKD- and FLT3-ITD-mutated (high [> 0.7] and low [0.05–
proved 5-year OS with GO, irrespective of patient age, due to 0.7] allele burden) AML, regardless of whether patients re-
decreased risk of relapse [54]. The benefit was observed in ceived alloSCT. Analyses of subgroups according to FLT3
patients with favorable (survival difference 20.7%; odds ratio subtype and FLT3-ITD allelic burden showed that OS did
[OR] 0.47; 95% CI: 0.31–0.73; P = 0.0006) or intermediate- not differ significantly according to treatment regimen within
risk karyotype (survival difference 5.7%; OR 0.84; 95% CI: each subgroup. As only a small proportion of patients received
0.75–0.95; P = 0.005), but not in adverse-risk patients. maintenance, the benefit of midostaurin maintenance is un-
Overall, 29% and 20% of patients had NPM1 and FLT3-ITD clear. Twenty-eight percent of patients in the midostaurin
mutations. Information on other gene mutations was not pro- arm received an alloSCT in first CR compared with 22.7%
vided. There was no survival benefit observed with GO in in the PBO arm (P = 0.10).
patients with secondary AML (sAML), FLT3-ITD, or NPM1 The phase 2 German-Austrian AMLSG 16-10 trial ad-
mutations. Doses of 3 mg/m2 were associated with equal ef- ministered midostaurin with 3+7 followed by alloSCT and
ficacy and less toxicity than higher doses. single agent midostaurin maintenance in 286 patients (age
As a proportion of patients with ELN intermediate risk 18–70 years) with FLT3-ITD-mutated AML (88% de
AML may require an alloSCT, the concern of developing novo, 8% sAML and 5% t-AML) [60]. Patients with core
VOD/SOS may influence choice of induction therapy. In an binding factor (CBF) AML were excluded. The composite
attempt to mitigate this risk, investigators in the ALFA-0701 CR (CR + CR with incomplete count recovery [CRi]) rate
study recommended an interval of 2 months between the last was 76.4% (CR 49.3%). The 2-year event-free survival
dose of GO and alloSCT [16••]. A retrospective safety analy- and OS were 39% and 34%, respectively, for all patients
sis of clinical trial data identified 99 (11.4%) cases of VOD/ and 53% and 46%, respectively, for the older patients.
SOS among 870 GO-treated patients (221 of whom received Seventy-three percent of patients underwent alloSCT.
alloSCT) [55]. Rates of VOD/SOS were 3% when GO was This demonstrated the safety and efficacy of midostaurin,
administered at doses < 6 mg/m2 as monotherapy or with non- at least in patients up to age 70.
hepatotoxic agents and 28% when administered with 6-
thioguanine (a hepatotoxic agent), 15% when administered CPX-351 (VYXEOS™ (Cytarabine: Daunorubicin) Liposome
as monotherapy at doses of 9 mg/m2, and between 15–40% for Injection)
if alloSCT was performed within 3 months of GO. Death from
VOD/SOS occurred in 60% of the cases. The European CPX-351 is a liposomal formulation of cytarabine and DNR
Society for Blood and Marrow Transplantation (EBMT) and fixed at a 5:1 molar ratio within 100-nm diameter liposomes
the Center for International Blood and Marrow Transplant [61, 62]. A randomized phase 3 trial compared CPX-351 with
Research (CIBMTR) documented VOD/SOS incidences of 3+7 in 309 older patients with TN high-risk AML (i.e., t-
4% and 8% among patients who previously received GO prior AML, sAML (preceding MDS or chronic myelomonocytic
to alloSCT and concluded that the incidence was not higher leukemia (CMML)), or AML-MRC) [15••]. The overall re-
than a matched control group [56, 57]. sponse rate (ORR: CR + CRi) was higher with CPX-351
compared with 3+7 (47.7% vs 33.3%; P = 0.016) with CR
Midostaurin Plus 3+7 rates of 37.3% and 25.6% in favor of CPX-351 (P = 0.04).
With a median follow-up of 20.7 months, median OS was
Midostaurin is a small molecule multikinase inhibitor with increased in patients treated with CPX-351 (9.56 months vs
activity against FLT3. FLT3 mutations occur in approximately 5.95 months; P = 0.003; OS2y 31.1% vs 12.3%). Subgroup
30% of cases of AML (~ 20–25% FLT3-ITD; ~ 7% FLT3- analyses demonstrated improved OS compared with 3+7 in
TKD) [7, 58]. The RATIFY trial evaluated midostaurin/ patients with wild-type FLT3, t-AML, sAML, and favorable/
placebo (PBO) administered with 3+7 followed by 4 cycles intermediate karyotype. There was no survival benefit ob-
of consolidation therapy with midostaurin/PBO and high-dose served in patients who had prior hypomethylating agent
cytarabine, then a 1-year midostaurin/PBO maintenance phase (HMA) exposure or unfavorable cytogenetics. Mutational
in 717 younger patients with TN FLT3-mutated AML analysis of CEBPA and NPM1 genes was performed, but no
(Table 3) [18••, 59]. Patients with therapy-related AML (t- information has been provided. CPX-351 may not be effective
AML) and patients with sAML who had received therapy in patients with TP53 mutations [63–65]. The toxicity profile
for preceding MDS were ineligible. CR rates were comparable (including incidence of reduced ejection fraction) of CPX-351
between the 2 arms (58.9% vs 53.5%; P = 0.15), but were was similar to that seen with 3+7 except for more prolonged
increased in the midostaurin arm when all CRs reported within neutropenia and thrombocytopenia in the CPX-351 arm. The
120 Page 6 of 13 Curr Oncol Rep (2021) 23:120

30-day mortality was 5.9% with CPX-351 and 10.6% with 3+ control (84% and 42% vs 67% and 19%, respectively); how-
7 (P = 0.149). Quality of life (QoL) analysis was not per- ever, incidence of pneumonia was comparable between the 2
formed, but a post-hoc Quality-Adjusted Time without arms (17% vs 22%, respectively). Tumor lysis syndrome
Symptoms of Disease and Toxicity (Q-TWiST) analysis dem- (TLS; all laboratory) was reported in 1% of patients in the
onstrated improved clinical benefit with CPX-351 over 3+7 VEN + AZA arm and none in the control arm. Only 2 patients
[66]. Thirty-four percent of patients in the CPX-351 arm in the VEN + AZA arm and 1 patient in the control arm
underwent alloSCT compared with 25% in the 3+7 arm received an alloSCT after discontinuing assigned study treat-
(P = 0.098) with less non-relapse mortality [67]. Although ment. Mortality at 30 days was the same between the 2 groups
this study excluded patients with MPN in blast phase (7% vs 6%). There was no observed difference between the 2
(MPN-BP), CPX-351 has yielded responses in this group groups with respect to QoL.
of patients [65]. A phase 3 randomized trial comparing Although no patients with CBF AML or MPN-BP were
CPX-351 with 3+7 in adult patients with ELN defined included in this trial, responses have been observed in patients
intermediate or adverse genetic risk AML is ongoing with MPN-BP and CBF AML [70, 71]. Furthermore, high
(NCT03897127) [68]. response rates and long remission durations appear to be as-
sociated with NPM1 or IDH2 mutations, with resistance asso-
Less Intensive Therapy ciated with emergence of FLT3 or RAS mutations, biallelic
TP53 loss [72], and/or monocytic subclones via loss of
Venetoclax and Azacitidine BCL-2 expression and increased dependency on MCL-1 for
oxidative phosphorylation and survival [73]. Outcomes for
Venetoclax (VEN) is a selective BH3-mimetic that targets patients with AML who did not respond, or lost response, to
B cell lymphoma 2 (BCL-2) resulting in programmed cell front-line VEN and HMAs have a median OS of 2.4 months
death. The phase 3 VIALE-A trial comparing VEN and [74].
azacitidine (AZA) with PBO and AZA in 433 patients with
TN AML who were not suitable for intensive chemotherapy Venetoclax and Low-Dose Cytarabine
(as defined by age and/or comorbidities) confirmed an im-
proved median OS in favor of VEN + AZA (14.7 months vs The phase 3 VIALE-C study evaluated VEN and low dose
9.6 months; P < 0.001) (Table 4) [22••]. Patients with MPN- cytarabine (LDAC) compared with PBO and LDAC in 211
BP or prior HMA exposure were excluded. Patients in the patients with TN AML who were not suitable for intensive
VEN + AZA arm received a median of 7 treatment cycles chemotherapy (as defined by age and/or comorbidities)
compared to 4.5 in the control arm. Composite CR (CR + (Table 4) [24••]. Patients with MPN-BP or who had prior
CRi) and CR rates were higher with VEN + AZA compared cytarabine exposure were excluded. Median treatment dura-
with PBO + AZA (66.4% [CR 36.7%] vs 28.3 % [CR 17.9%]; tion was 3.9 months, and median number of cycles adminis-
P < 0.001). Median time to first response (either CR or CRi) tered was 4 in the VEN + LDAC arm compared with 1.7
was 1.3 months vs 2.8 months, respectively. Composite CR months and 2, respectively, in the control arm. Median OS
before initiation of cycle 2 was achieved in 43.4% of patients at the planned analysis was not statistically different between
receiving VEN + AZA vs 7.6% in those receiving PBO + the 2 arms (7.2 months vs 4.1 months; P = 0.11). There were
AZA. Subgroup analysis demonstrated higher composite CR several imbalances in baseline characteristics (i.e., sAML, pri-
rates with VEN + AZA compared with PBO + AZA in the or hematologic disorder-related secondary AML, and poor-
following: (a) IDH1/2 mutated (75.4% vs 10.7%; P < 0.001); risk cytogenetics) as well as increased administrative censor-
(b) FLT3-ITD/TKD mutated (72.4% vs 36.4%; P = 0.02); (c) ing in the VEN + LDAC arm compared to control arm (i.e.,
TP53 mutated (55.3% vs 0%; P < 0.001); (d) therapy-related enrolment was ongoing 3.4 months before the pre-planned OS
and/or secondary AML (t/s-AML; 66.7% vs 22.9%; P not analysis; censoring of patients still alive at time of analysis and
specified); and (e) poor-risk cytogenetics (52.9% vs 23.2%; who had not reached the median OS by study cut-off date
P not specified). In patients with composite CR, measurable happened more frequently in the VEN + LDAC arm [12%
residual disease (MRD) negativity occurred in 24.4% of pa- vs 6%]). Hence, a longer-term unplanned analysis with an
tients receiving VEN + AZA compared with 7.6% in the con- additional 6 months of patient follow-up was performed and
trol arm (P not specified). OS was improved in favor of VEN demonstrated an improved median OS in favor of VEN +
+ AZA compared with the control arm in IDH1/2-mutated LDAC (8.4 months vs 4.1 months; P = 0.04). Composite
AML (OS1y 66.8% vs 35.7%; P < 0.001), t/s-AML (median CR (CR + CRi) rates were higher in patients receiving VEN
16.4 months vs 10.6 months; P not specified), and AML with + LDAC than PBO + LDAC (48% [CR 27%] vs 13% [CR
poor-risk cytogenetics (median 7.6 months vs 6 months; P not 7%]; P < 0.001). Composite CR before initiation of cycle 2
specified). Infections of any grade, including febrile neutrope- was achieved in 34% of patients receiving VEN + LDAC
nia, were higher in the VEN + AZA group compared with compared with 3% in those receiving PBO + LDAC (P <
Curr Oncol Rep (2021) 23:120 Page 7 of 13 120

Table 4 Outcomes with less intensive agents/regimens

VEN + AZA VEN + LDAC LDAC + glasdegib Ivosidenib Enasidenib


(N = 286) (N = 143) (N = 88)b (N = 34) (N = 39)
Phase 3 Phase 3 Phase 2 Phase 1 Phase 1/2
DiNardo 2020 [22] Wei 2020 [24] Cortes 2019 [25] Roboz 2020 [19••] Pollyea 2019 [69]

Characteristics
Median age, y (range) 76 (59–91) 76 (36–93) 77 (63–92)b 76.5 (64–87) 77 (58–87)
Age > 75y 174 (61%) 82 (57%) 53 (60.2%)b 19 (56%) 24 (62%)
De novo AML 214 (75%) 85 (59%) Not provided 8 (24%) Not provided
Therapy-related AML 26 (9%) 6/58 (10%) Not provided 3 (9%) 2 (5%)
(tAML)
Secondary AML (sAML) 46 (16%) 52/58 (90%) Not provided 23 (68%) 23 (59%)
Prior HMA 0 28 (20%) 13 (14.8%)b 16 (47%) Not provided
Cytogenetic risk group
Good 0 1 (1%) Good/Intermediate 0 0
52
Intermediate 182 (64%) 90 (63%) (59.1%)b 24 (71%) 19 (49%)
Poor 104 (36%) 47 (33%) 36 (40.9%)b 9 (26%) 10 (26%)
Gene mutations
FLT3-ITD/TKD 29/206 (14%) 20/112 (18%) 8/61 (13%)b 1/33 (3%) 2/34 (6%)
IDH1/2 61/245 (25%) 21/112 (19%) 22/61 (36%)b IDH2 2/33 (6%) IDH1 1/34 (3%)
TP53 38/163 (23%) 22/112 (20%) Not provided 3/33 (9%) 3/34 (9%)
Outcomes
CR + CRi
All 190/286 (66.4%; 69/143 (48%; CR 27%) 19/78 (24%; CR CR + CRh 14/33 CR + CRi 8/39
CR 36.7%) 17.9%) (42.4%; CR 30.3%) (20.5%; CR 18%)
tAML s/tAML 48/72 s/tAML 21/58 (36%; CR Not provided Not provided Not provided
(66.7%) 16%)
sAML Not provided Not provided CR 3/23 (13%)
Poor risk cytogenetics 55/104 (52.9%) 13/47 (28%; CR 17%) CR 5/36 (13.9%)b Not provided Not provided
Prior HMA Not applicable 7/28 (25%; CR 7%) Not provided CR + CRh 4/15 (26.7%; Not provided
CR 20%)
FLT3-ITD/TKD 21/29 (72.4%) 9/20 (45%; CR25%) ORR 2/8 (25%)b 0 0
IDH1/2 46/61 (75.4%)a 12/21 (57%; CR 38%) ORR 7/22 IDH2 0 IDH1 0
(31.8%)b
TP53 21/38 (55.3%) 4/22 (18%; CR 5%) Not provided CRh 1/3 (33%) 0
Time to response, mos 1.3 (0.6–9.9) for Before initiation cycle 2: Not provided 2.8 (1.9–4.6) for CR 5.6 (3.4–12.9) for CR
(range) CR + CRi 34% for CR +CRi
Median duration of 17.5 (13.6-NR) for Not provided 9.9 (0.03–28.8) for NR (proportion in CR at NR (3.7-NR) for CR
response, mos (95% CI) CR + CRi CRb 1y 77.8%)
Median follow-up, mos 20.5 Planned: 12 21.7b 23.5 8.4
Unplanned: 18
Median OS, mos (95% CI) 14.7 (11.9–18.7) Planned: 7.2 (5.6–10.1) 8.3 (6.6–9.5) 12.6 (4.5–25.7) 11.3 (5.7–15.1)
Unplanned: 8.4 (5.9–10.1)
30-day mortality 21/286 (7%) 18/143 (13%) 5/84 (6%)b Not provided 3/39 (8%)

CI confidence interval; CR complete remission; CRh CR with partial hematologic improvement; CRi complete remission with incomplete count
recovery; HMA hypomethylating agent; MDS myelodysplastic syndrome; mos months; NR not reached; ORR overall response rate (CR + CRi +
morphologic leukemic-free state [MLFS]); OS overall survival; y year
a
CR + CRi for IDH1 mutated 13/23 (56.5%) and for IDH2 mutated 34/40 (85%); b includes 10 patients with MDS

0.001). Subgroup analysis demonstrated higher composite CR AML (36% vs 4%; P not specified); and (d) poor-risk cytoge-
rates with VEN + LDAC compared with placebo + LDAC in netics (28% vs 10%; P not specified). There was no difference
the following: (a) IDH1/2 mutated (57% vs 33%; P not spec- in median OS in the different subgroups between the 2 treat-
ified); (b) TP53 mutated (18% vs 0%; P not specified); (c) t/s- ment arms. In patients with co-existing FLT3 and NPM1
120 Page 8 of 13 Curr Oncol Rep (2021) 23:120

mutations, median OS was 10.2 months in the PBO arm and Ivosidenib
not yet reached in the VEN + LDAC arm (P not specified).
The outcomes for patients with FLT3-ITD/TKD-mutated Ivosidenib is an oral, small molecule inhibitor of mutant
AML were comparable between the 2 arms. In patients with isocitrate dehydrogenase 1 (IDH1). Mutations in the IDH1
composite CR, MRD negativity as determined by flow cytom- gene occur in ~ 6–10% of AML [7]. Ivosidenib monotherapy
etry occurred in 6% of patients receiving VEN + LDAC com- induced durable remissions in 34 patients with newly diag-
pared with 1% in the control arm. Grade > 3 febrile neutrope- nosed mutated IDH1 AML not eligible for intensive chemo-
nia and serious pneumonias were comparable between the 2 therapy who were treated as part of a phase 1 study (Table 4)
arms (32% and 13% vs 29% and 10%, respectively). TLS was [19••, 75]. Sixty-eight percent had secondary AML [12% had
observed in 8 patients (6%; 4 clinical and 4 laboratory) in the preceding MPN]. Differentiation syndrome occurred in 18%
VEN + LDAC arm and none in the control arm. No patient of patients, but did not require treatment discontinuation.
received alloSCT after study therapy. The 30-day mortality Grade > 3 febrile neutropenia and QTc prolongation
rate was the same between the 2 groups (13% vs 11%). occurred in 2 (6%) patients each. The composite CR (CR +
There was no observed difference between the 2 groups with CR with incomplete hematologic recovery [CRh]) was 42.4%
respect to QoL. (CR 30.3%). None of the patients achieving a CR/CRh had a
FLT3 mutation. IDH1 mutation clearance occurred in 9/14
(64%) of patients achieving CR+CRh (CR 50%; CRh
Glasdegib and Low-Dose Cytarabine 100%). Median duration of response had not been reached
with 77.8% of patients remaining in CR at 1 year. The median
Glasdegib is a selective oral inhibitor of the hedgehog signal- time to CR was 3.7 months. After a follow-up of 23.5 months,
ing pathway by binding to smoothened (SMO). A randomized median OS was 12.6 months. Three (9%) patients received an
phase 2 study evaluated LDAC with or without glasdegib in alloSCT.
132 patients with newly diagnosed AML or higher risk MDS
unsuitable for intensive chemotherapy (as defined by age and/ Enasidenib
or comorbidities) (Table 4) [25••]. Patients could not have
preceding chronic myeloid leukemia and needed to be > 55 Enasidenib is an oral inhibitor of mutant IDH2 proteins. IDH2
years. The proportion of patients with t/s-AML was not re- mutations occur in 6–19% of AML [7]. Thirty-nine patients
ported. Median treatment duration was low in both arms (2.7 with newly diagnosed IDH2-mutated AML were treated with
months vs 1.5 months, respectively). Median OS was 8.8 enasidenib monotherapy on a phase 1/2 trial (Table 4) [69,
months with LDAC + glasdegib and 4.9 months with LDAC 76]. Twenty-three (59%) patients had secondary AML arising
alone (P = 0.0004) with 1-year OS of 39.5% and 9.5%, re- from prior MDS, CMML, or MPN. Differentiation syndrome
spectively. The composite CR (CR + CRi) rates in patients occurred in 13% of patients and QTc prolongation in 10%.
with AML were higher in the LDAC + glasdegib arm com- The composite CR (CR + CRp) was 20.5% (CR 18%). None
pared to single agent LDAC (24% [CR 17.9%] vs 10.5% [CR of the patients with FLT3 or TP53 mutations achieved a re-
2.6%]; P < 0.05 for CR). Febrile neutropenia occurred in sponse. Median duration of response had not been reached.
35.7% of patients treated with LDAC + glasdegib and Median OS was 11.3 months. Three (8%) patients received an
24.4% with LDAC. Dysguesia and muscle spasms occurred alloSCT. Although there is no FDA approval for enasidenib in
in 25% and 22.6%, respectively, in patients receiving LDAC the front-line setting at this time, it is approved in the relapsed/
+ glasdegib compared with 2.4% and 4.9%, respectively, in refractory AML setting.
the control arm. On subgroup analysis, ORR (CR + CRi +
morphological leukemia free state (MLFS) for patients with
AML and CR + marrow CR for those with MDS) favored Evolving Treatment Strategies Based on Risk
LDAC + glasdegib in the following: (a) IDH1/2 mutated Group
(31.8% vs 0%; P = NS); (b) FLT3-ITD/TKD mutated (25%
vs 0%; P not specified); (c) RUNX1 mutated (36% vs 0%; P = The recent drug approvals, on the most part, irrespective of
NS); and (d) poor-risk cytogenetics (13.9% vs 5.3%; P not age, have created challenges in deciding which is the most
specified). There was no difference in response rates for appropriate therapy if several overlapping treatment options
TP53-mutated AML. Outcomes for t/s-AML were not report- are available (Tables 1 and 5). These newer therapies highlight
ed. Only 1 patient in the LDAC + glasdegib arm and none in the need for timely identification of cytogenetic abnormalities
the control arm received an alloSCT after discontinuing and genetic mutations. Despite the availability of these new
assigned study treatment. The 30-day mortality rate for pa- drugs, there continues to be a lack of highly effective therapy
tients with AML was 6.7% in the LDAC + glasdegib arm in patients with adverse/poor-risk AML, including RUNX1-
and 13.9% in the LDAC only arm. and ASXL1-mutated AML (Tables 3, 4, and 5).
Curr Oncol Rep (2021) 23:120 Page 9 of 13 120

Table 5 First-line treatment strategies for patients with AML who are suitable for intensive therapy

Risk group Genetic abnormality or 2016 WHO classification First-line therapya

Favorable Core binding factor (CBF) 3+7 + GO


Intermediate FLT3-TKD mutation (exclusive of adverse/poor risk cytogenetics) 3+7 + midostaurin
Intermediate risk cytogenetics (exclusive of therapy-related or 3+7 + GO; intensive regimens (e.g., FLAG-Ida, MEC)b
secondary AML or AML-MRC)
Adverse/poor TP53 mutation VEN + AZA (DEC)c; intensive regimens (e.g., FLAG-Ida, MEC)b
Adverse/poor risk cytogeneticsd VEN + AZA (DEC)c; intensive regimens (e.g., FLAG-Ida, MEC)b
MPN-BC VEN + AZA (DEC)c; intensive regimens (e.g., FLAG-Ida, MEC)b
FLT3-ITD mutation (exclusive of adverse/poor risk cytogenetics) 3+7 + midostaurin
Therapy-relatede or secondary AML or AML-MRC CPX-351b; intensive regimens (e.g., FLAG-Ida, MEC)b

AML-MRC AML with myelodysplasia-related changes; AZA azacitidine; DEC decitabine; FLAG-Ida fludarabine, cytarabine, G-CSF, and idarubicin;
GO gemtuzumab ozogamicin; MEC mitoxantrone, etoposide and cytarabine; MPN-BC myeloproliferative neoplasm in blast crisis; VEN venetoclax
a
Patients should be enrolled onto clinical trials if possible; b transplant eligible patients; c may be off-label use; d for AML-MRC with poor risk
cytogenetics, administration of CPX-351 would also be appropriate; e for CBF administration of 3+7 + GO would also be appropriate

AlloSCT is routinely recommended in first CR for trans- regimens/agents, faster and higher CR rates and/or MRD neg-
plant eligible patients with poor-risk AML, as it is potentially ativity appear to be achieved with VEN + AZA (Table 4),
curative. Intensive chemotherapy regimens other than 3+7 including in HMA-naïve patients with poor-risk cytogenetics
have been utilized in an attempt to increase the CR rates and and t/s-AML. Although patients with MPN-BP were excluded
eligibility for alloSCT. Subgroup analyses in randomized tri- from most of the studies [15••, 17, 18••, 22••, 24••] due to their
als with 3+7 have demonstrated CR rates of 51–65% in youn- extremely poor response to intensive chemotherapy [46, 84],
ger patients (age < 60 years) and 34–56% in older patients VEN-based therapies may be effective in this group of pa-
(age > 60 years) with poor-risk cytogenetics; however, OS is tients [71]. The high response rates of VEN + HMA compared
poor in both age groups [9, 11, 12, 14]. Published studies with contemporary intensive chemotherapy and other lower
comparing different regimens have not been powered to dem- intensity regimens with low rates of early deaths/induction
onstrate significant differences in outcomes in poor-risk sub- deaths and/or infections have led to more off-label use of these
groups [9•]; however, in exploratory ad hoc analyses, regimens in higher-risk patients for whom alloSCT is deemed
flavopiridol (alvocidib), cytarabine, and mitoxantrone appropriate (Tables 3, 4, and 5) [85, 86, 87, 88]. However,
(FLAM) yielded higher CR rates in patients with > 1 poor- limited data is available for outcomes (such as relapse risk and
risk feature compared with 3+7 (61% vs 34%; P = 0.04) [77]. OS) when VEN + HMAs are administered as a bridge to
Also, the UK MRC AML15 trial compared 2 induction alloSCT [87, 88]. A phase 2 trial is examining the safety and
courses of fludarabine, cytarabine, G-CSF, and idarubicin tolerability of VEN + AZA in younger patients (< 60 years)
(FLAG-Ida) (n = 635) with daunorubicin and cytarabine with with ELN defined poor-risk AML compared to historical con-
etoposide (ADE) (n = 633) in the front-line setting in patients trols who received induction chemotherapy (NCT03573024)
with TN AML [78]. FLAG-Ida was associated with a signif- [89]. Ultimately, a randomized prospective trial will be re-
icantly higher CR rate after 1 cycle of induction therapy (77% quired to determine the most effective strategy in this high-
vs 67%; P < 0.001) with improved RFS (45% vs 34%; P = risk group of patients who are eligible for intensive chemo-
0.01), overall and in subgroups, but there was no OS benefit. therapy and alloSCT.
Based on the higher CR rate after 1 induction, several centers
have adopted FLAG-Ida as front-line therapy for patients with
poor-risk AML in order to enable more of these patients to Why It Is Not Time to Say Goodbye to “3+7”
proceed to alloSCT [79].
More effective and less toxic treatments could deliver more The number of AML subtypes where “unadulterated” 3+7
patients to transplant who otherwise might not respond to (i.e., 3+7 alone) is an appropriate therapeutic option is shrink-
first-line therapy, relapse before reaching transplant, or devel- ing. A meta-analysis demonstrated a small incremental surviv-
op treatment-related complications that impair their ability to al benefit with the addition of GO to 3+7 for intermediate-risk
receive an alloSCT (Tables 3, 4, and 5). Therapy with single cytogenetics (excluding t/s-AML or AML-MRC) with no im-
agent LDAC, AZA, and decitabine (DEC) in patients with de pact on CR rates; hence, administration of 3+7 without GO
novo or sAML has yielded low CR rates (range, 8–20%) may be warranted in patients with risk factors for VOD/SOS.
[80–83]. Among the newly approved lower intensity AML subtypes where there is no longer a role for treatment
120 Page 10 of 13 Curr Oncol Rep (2021) 23:120

with 3+7 only include t/s-AML, FLT3-mutated AML, and References


CBF AML. However, unlike for acute promyelocytic leuke-
mia, where there is a high CR and cure rate with all-trans Papers of particular interest, published recently, have been
retinoic acid and arsenic trioxide [90], there is no curative highlighted as:
non-chemotherapeutic treatment option for patients with other • Of importance
subtypes of AML. The lower intensive therapies, including •• Of major importance
VEN + HMA, may be very effective in remission induction,
but they are not very effective at preventing relapse. 1. Slovak ML, Kopecky KJ, Cassileth PA, Harrington DH, Theil KS,
Therefore, 3+7 will remain the intensive chemotherapy back- Mohamed A, et al. Karyotypic analysis predicts outcome of
preremission and postremission therapy in adult acute myeloid leu-
bone onto which newer therapies have been/will be added to
kemia: a Southwest Oncology Group/Eastern Cooperative
manage specific subsets of AML (e.g., quizartinib Oncology Group Study. Blood. 2000;96(13):4075–83.
(NCT02668653), gilteritinib (NCT02236013), and 2. Grimwade D, Hills RK, Moorman AV, Walker H, Chatters S,
venetoclax (NCT03709758)). Goldstone AH, et al. Refinement of cytogenetic classification in
acute myeloid leukemia: determination of prognostic significance
of rare recurring chromosomal abnormalities among 5876 younger
adult patients treated in the United Kingdom Medical Research
Conclusion Council trials. Blood. 2010;116(3):354–65.
3.•• Dohner H, Estey E, Grimwade D, Amadori S, Appelbaum FR,
Buchner T, et al. Diagnosis and management of AML in adults:
The treatment of AML has been propelled into the foreground
2017 ELN recommendations from an international expert panel.
in the last 5 years with the approval of multiple new drugs. Blood. 2017;129(4):424–47. The ELN risk category for AML
These are exciting times for treating clinicians as they have an is used by most clinicians and in clinical trials.
unprecedented number of therapeutic options at their disposal, 4. National Comprehensive Cancer Network. Acute myeloid leuke-
but at the same time, AML treatment is becoming increasingly mia (Version 1.2021). https://wwwnccnorg/professionals/
physician_gls/pdf/amlpdf. Accessed Nov 28, 2020.
complex. Naturally, the relevance and value of intensive che- 5. Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau
motherapeutic regimens have been challenged as our under- MM, et al. The 2016 revision to the World Health Organization
standing of the underlying pathobiology of AML pathogene- classification of myeloid neoplasms and acute leukemia. Blood.
sis expands and the spotlight turns towards targeted therapies. 2016;127(20):2391–405.
6.•• Papaemmanuil E, Gerstung M, Bullinger L, Gaidzik VI, Paschka P,
As highlighted in this review, 3+7 continues to play a pivotal
Roberts ND, et al. Genomic classification and prognosis in acute
role in AML treatment, especially in patients < 60 years, and myeloid leukemia. N Engl J Med. 2016;374(23):2209–21. This
will remain a cornerstone of AML therapy for individuals who manuscript proposes a genetic based classifcation of AML
are suitable for intensive chemotherapy, at least until there are (which correlates with prognosis).
randomized, head-to-head trials that demonstrate a clear supe- 7. Bullinger L, Dohner K, Dohner H. Genomics of acute myeloid
leukemia diagnosis and pathways. J Clin Oncol. 2017;35(9):934–
riority of novel therapies over the 3+7 backbone. Currently, 46.
clinical trials are attempting to augment the established effica- 8. Yates JW, Wallace HJ Jr, Ellison RR, Holland JF. Cytosine arabi-
cy of 3+7/intensive chemotherapy regimens though combina- noside (NSC-63878) and daunorubicin (NSC-83142) therapy in
tion with novel therapies. It will be fascinating to see how well acute nonlymphocytic leukemia. Cancer Chemother Rep.
1973;57(4):485–8.
combinations of targeted, lower intensity therapies perform
9.• Murphy T, Yee KWL. Cytarabine and daunorubicin for the treat-
against traditional intensive chemotherapy regimens in the ment of acute myeloid leukemia. Expert Opin Pharmacother.
front-line setting. Until then, it would be premature to say 2017;18(16):1765–80. This manuscript provides a more in
goodbye to 3+7 chemotherapy altogether. depth review of the 3+7 regimen.
10. Burnett AK, Russell NH, Hills RK, Kell J, Cavenagh J, Kjeldsen L,
et al. A randomized comparison of daunorubicin 90 mg/m2 vs 60
mg/m2 in AML induction: results from the UK NCRI AML17 trial
Human and Animal Rights and Informed Consent This article does not in 1206 patients. Blood. 2015;125(25):3878–85.
contain any studies with human or animal subjects performed by any of
11. Fernandez HF, Sun Z, Yao X, Litzow MR, Luger SM, Paietta EM,
the authors.
et al. Anthracycline dose intensification in acute myeloid leukemia.
N Engl J Med. 2009;361(13):1249–59.
Declarations 12. Lee JH, Joo YD, Kim H, Bae SH, Kim MK, Zang DY, et al. A
randomized trial comparing standard versus high-dose daunorubi-
Conflict of Interest KWLY consulted for and/or received honorarium cin induction in patients with acute myeloid leukemia. Blood.
from Novartis, F. Hoffmann La Roche, Takeda, Pfizer, TaiHo, Bristol- 2011;118(14):3832–41.
Myers Squib/Celgene, Paladin, Astex, and Otsuka. Received research 13. Buchner T, Hiddemann W, Wormann B, Loffler H, Gassmann W,
funding from Astex, Novartis, Forma Therapeutics, Jazz, Onconova, F. Haferlach T, et al. Daunorubicin 60 instead of 30 mg/sqm improves
Hoffmann La Roche, Genentech, and Tolero. ACS has consulted for and/ response and survival in elderly patients with AML. Blood.
or received honoraria from and/or received research support from 1997;90(supplement 1):583a.
AbbVie, Agios, Amgen, Astellas, Celgene/BMS, GlycoMimetics, Jazz, 14. Lowenberg B, Ossenkoppele GJ, van Putten W, Schouten HC,
Novartis, Paladin, Pfizer, and Teva. Graux C, Ferrant A, et al. High-dose daunorubicin in older patients
Curr Oncol Rep (2021) 23:120 Page 11 of 13 120

with acute myeloid leukemia. N Engl J Med. 2009;361(13):1235– duplication in patients with acute myeloid leukemia (AML) adds
48. important prognostic information to cytogenetic risk group and re-
15.•• Lancet JE, Uy GL, Cortes JE, Newell LF, Lin TL, Ritchie EK, et al. sponse to the first cycle of chemotherapy: analysis of 854 patients
CPX-351 (cytarabine and daunorubicin) liposome for injection ver- from the United Kingdom Medical Research Council AML 10 and
sus conventional cytarabine plus daunorubicin in older patients 12 trials. Blood. 2001;98(6):1752–9.
with newly diagnosed secondary acute myeloid leukemia. J Clin 27. Gale RE, Green C, Allen C, Mead AJ, Burnett AK, Hills RK, et al.
Oncol. 2018;36(26):2684–92. This phase 3 study led to the ap- The impact of FLT3 internal tandem duplication mutant level, num-
proval of CPX-351 for t-AML, sAML and AML-MRC. ber, size, and interaction with NPM1 mutations in a large cohort of
16.•• Castaigne S, Pautas C, Terre C, Raffoux E, Bordessoule D, Bastie young adult patients with acute myeloid leukemia. Blood.
JN, et al. Effect of gemtuzumab ozogamicin on survival of adult 2008;111(5):2776–84.
patients with de-novo acute myeloid leukaemia (ALFA-0701): a 28. Linch DC, Hills RK, Burnett AK, Khwaja A, Gale RE. Impact of
randomised, open-label, phase 3 study. Lancet. 2012;379(9825): FLT3(ITD) mutant allele level on relapse risk in intermediate-risk
1508–16. This study led to the approval of gemtuzumab for acute myeloid leukemia. Blood. 2014;124(2):273–6.
CD33-positive favorable and intermediate cytogenetic risk 29. Thiede C, Steudel C, Mohr B, Schaich M, Schakel U, Platzbecker
AML. U, et al. Analysis of FLT3-activating mutations in 979 patients with
17. Lambert J, Pautas C, Terre C, Raffoux E, Turlure P, Caillot D, et al. acute myelogenous leukemia: association with FAB subtypes and
Gemtuzumab ozogamicin for de novo acute myeloid leukemia: identification of subgroups with poor prognosis. Blood.
final efficacy and safety updates from the open-label, phase III 2002;99(12):4326–35.
ALFA-0701 trial. Haematologica. 2019;104(1):113–9. 30. Bezerra MF, Lima AS, Pique-Borras MR, Silveira DR, Coelho-
18.•• Stone RM, Mandrekar SJ, Sanford BL, Laumann K, Geyer S, Silva JL, Pereira-Martins DA, et al. Co-occurrence of DNMT3A,
Bloomfield CD, et al. Midostaurin plus chemotherapy for acute NPM1, FLT3 mutations identifies a subset of acute myeloid leuke-
myeloid leukemia with a FLT3 mutation. N Engl J Med. mia with adverse prognosis. Blood. 2020;135(11):870–5.
2017;377(5):454–64. This study led to the approval of 31. Metzeler KH, Herold T, Rothenberg-Thurley M, Amler S,
midostaurin for FLT3 mutated AML. Sauerland MC, Gorlich D, et al. Spectrum and prognostic relevance
19.•• Roboz GJ, DiNardo CD, Stein EM, de Botton S, Mims AS, Prince of driver gene mutations in acute myeloid leukemia. Blood.
GT, et al. Ivosidenib induces deep durable remissions in patients 2016;128(5):686–98.
with newly diagnosed IDH1-mutant acute myeloid leukemia. 32. Gaidzik VI, Teleanu V, Papaemmanuil E, Weber D, Paschka P,
Blood. 2020;135(7):463–71 This subgroup analysis led to the Hahn J, et al. RUNX1 mutations in acute myeloid leukemia are
FDA approval of ivosidenib in treatment-naïve IDH1 mutated associated with distinct clinico-pathologic and genetic features.
AML patients, but it is not approved in Canada or Europe. Leukemia. 2016;30(11):2160–8.
20. DiNardo CD, Pratz KW, Letai A, Jonas BA, Wei AH, Thirman M, 33. Gaidzik VI, Bullinger L, Schlenk RF, Zimmermann AS, Rock J,
et al. Safety and preliminary efficacy of venetoclax with decitabine Paschka P, et al. RUNX1 mutations in acute myeloid leukemia:
or azacitidine in elderly patients with previously untreated acute results from a comprehensive genetic and clinical analysis from
myeloid leukaemia: a non-randomised, open-label, phase 1b study. the AML study group. J Clin Oncol. 2011;29(10):1364–72.
Lancet Oncol. 2018;19(2):216–28. 34. Tang JL, Hou HA, Chen CY, Liu CY, Chou WC, Tseng MH, et al.
21. DiNardo CD, Pratz K, Pullarkat V, Jonas BA, Arellano M, Becker AML1/RUNX1 mutations in 470 adult patients with de novo acute
PS, et al. Venetoclax combined with decitabine or azacitidine in myeloid leukemia: prognostic implication and interaction with oth-
treatment-naive, elderly patients with acute myeloid leukemia. er gene alterations. Blood. 2009;114(26):5352–61.
Blood. 2019;133(1):7–17. 35. Patel JP, Gonen M, Figueroa ME, Fernandez H, Sun Z, Racevskis J,
22.•• DiNardo CD, Jonas BA, Pullarkat V, Thirman MJ, Garcia JS, Wei et al. Prognostic relevance of integrated genetic profiling in acute
AH, et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N Engl J Med. 2012;366(12):1079–89.
myeloid leukemia. N Engl J Med. 2020;383(7):617–29. 36. Sasaki K, Kanagal-Shamanna R, Montalban-Bravo G, Assi R,
Landmark phase 3 study comparing venetoclax and azacitidine Jabbour E, Ravandi F, et al. Impact of the variant allele frequency
to placebo and azacitidine in treatment-naïve AML patients. of ASXL1, DNMT3A, JAK2, TET2, TP53, and NPM1 on the
23. Wei AH, Strickland SA Jr, Hou JZ, Fiedler W, Lin TL, Walter RB, outcomes of patients with newly diagnosed acute myeloid leuke-
et al. Venetoclax Combined with low-dose cytarabine for previous- mia. Cancer. 2020;126(4):765–74.
ly untreated patients with acute myeloid leukemia: results from a 37. Tsai CH, Hou HA, Tang JL, Liu CY, Lin CC, Chou WC, et al.
phase Ib/II study. J Clin Oncol. 2019;37(15):1277–84. Genetic alterations and their clinical implications in older patients
24.•• Wei AH, Montesinos P, Ivanov V, DiNardo CD, Novak J, Laribi K, with acute myeloid leukemia. Leukemia. 2016;30(7):1485–92.
et al. Venetoclax plus LDAC for newly diagnosed AML ineligible 38. Metzeler KH, Becker H, Maharry K, Radmacher MD, Kohlschmidt
for intensive chemotherapy: a phase 3 randomized placebo- J, Mrozek K, et al. ASXL1 mutations identify a high-risk subgroup
controlled trial. Blood. 2020;135(24):2137–45. Landmark phase of older patients with primary cytogenetically normal AML within
3 study comparing venetoclax and low dose cytarabine to pla- the ELN Favorable genetic category. Blood. 2011;118(26):6920–9.
cebo and low dose cytarabine in treatment-naïve AML patients. 39. Schnittger S, Eder C, Jeromin S, Alpermann T, Fasan A,
It included patients who had previo usly received Grossmann V, et al. ASXL1 exon 12 mutations are frequent in
hypomethylating agents. AML with intermediate risk karyotype and are independently as-
25.•• Cortes JE, Heidel FH, Hellmann A, Fiedler W, Smith BD, Robak T, sociated with an adverse outcome. Leukemia. 2013;27(1):82–91.
et al. Randomized comparison of low dose cytarabine with or with- 40. Paschka P, Schlenk RF, Gaidzik VI, Herzig JK, Aulitzky T,
out glasdegib in patients with newly diagnosed acute myeloid leu- Bullinger L, et al. ASXL1 mutations in younger adult patients with
kemia or high-risk myelodysplastic syndrome. Leukemia. acute myeloid leukemia: a study by the German-Austrian Acute
2019;33(2):379–89. Phase 2 study comparing glasdegib and Myeloid Leukemia Study Group. Haematologica. 2015;100(3):
low dose cytarabine to low dose cytarabine in treatment-naïve 324–30.
AML patients. Led to FDA, EMA and Health Canada approval 41. Pratcorona M, Abbas S, Sanders MA, Koenders JE, Kavelaars FG,
of glasdegib. Erpelinck-Verschueren CA, et al. Acquired mutations in ASXL1 in
26. Kottaridis PD, Gale RE, Frew ME, Harrison G, Langabeer SE, acute myeloid leukemia: prevalence and prognostic value.
Belton AA, et al. The presence of a FLT3 internal tandem Haematologica. 2012;97(3):388–92.
120 Page 12 of 13 Curr Oncol Rep (2021) 23:120

42. Kayser S, Dohner K, Krauter J, Kohne CH, Horst HA, Held G, et al. 57. Ho VT, Martin AS, Perez WS, Steinert P, Zhang MJ, Chirnomas D,
The impact of therapy-related acute myeloid leukemia (AML) on et al. Prior gemtuzumab ozogamicin exposure in adults with acute
outcome in 2853 adult patients with newly diagnosed AML. Blood. myeloid leukemia does not increase hepatic veno-occlusive disease
2011;117(7):2137–45. risk after allogeneic hematopoietic cell transplantation: a center for
43. Morton LM, Dores GM, Schonfeld SJ, Linet MS, Sigel BS, Lam international blood and marrow transplant research analysis. Biol
CJK, et al. Association of Chemotherapy for solid tumors with Blood Marrow Transplant. 2020;26(5):884–92.
development of therapy-related myelodysplastic syndrome or acute 58. Perl AE. Availability of FLT3 inhibitors: how do we use them?
myeloid leukemia in the modern era. JAMA Oncol. 2019;5(3):318– Blood. 2019;134(9):741–5.
25. 59. Stone RM, Mandrekar S, Sanford BL, Geyer S, Bloomfield CD,
44. Granfeldt Ostgard LS, Medeiros BC, Sengelov H, Norgaard M, Dohner K, et al. The multi-kinase inhibitor midostaurin (M) pro-
Andersen MK, Dufva IH, et al. Epidemiology and clinical signifi- longs survival compared with placebo (P) in combination with dau-
cance of secondary and therapy-related acute myeloid leukemia: a norubicin (D)/cytarabine (C) induction (ind), high-dose C consoli-
national population-based cohort study. J Clin Oncol. 2015;33(31): dation (consol), and as maintenance (maint) therapy in newly diag-
3641–9. nosed acute myeloid leukemia (AML) patients (pts) age 18-60 with
45. Bello C, Yu D, Komrokji RS, Zhu W, Wetzstein GA, List AF, et al. FLT3 mutations (muts): an international prospective randomized
Outcomes after induction chemotherapy in patients with acute my- (rand) P-controlled double-blind trial (CALGB 10603/RATIFY
eloid leukemia arising from myelodysplastic syndrome. Cancer. [Alliance]). Blood. 2015:126.
2011;117(7):1463–9. 60. Schlenk RF, Weber D, Fiedler W, Salih HR, Wulf G, Salwender H,
46. Kennedy JA, Atenafu EG, Messner HA, Craddock KJ, Brandwein et al. Midostaurin added to chemotherapy and continued single-
JM, Lipton JH, et al. Treatment outcomes following leukemic trans- agent maintenance therapy in acute myeloid leukemia with FLT3-
formation in Philadelphia-negative myeloproliferative neoplasms. ITD. Blood. 2019;133(8):840–51.
Blood. 2013;121(14):2725–33. 61. Mayer LD, Harasym TO, Tardi PG, Harasym NL, Shew CR,
47. Yogarajah M, Tefferi A. Leukemic transformation in myeloprolif- Johnstone SA, et al. Ratiometric dosing of anticancer drug combi-
erative neoplasms: a literature review on risk, characteristics, and nations: controlling drug ratios after systemic administration regu-
outcome. Mayo Clin Proc. 2017;92(7):1118–28. lates therapeutic activity in tumor-bearing mice. Mol Cancer Ther.
48. Tefferi A, Mudireddy M, Mannelli F, Begna KH, Patnaik MM, 2006;5(7):1854–63.
Hanson CA, et al. Blast phase myeloproliferative neoplasm: 62. Feldman EJ, Lancet JE, Kolitz JE, Ritchie EK, Roboz GJ, List AF,
Mayo-AGIMM study of 410 patients from two separate cohorts. et al. First-in-man study of CPX-351: a liposomal carrier containing
Leukemia. 2018;32(5):1200–10. cytarabine and daunorubicin in a fixed 5:1 molar ratio for the treat-
49. Montalban-Bravo G, Kanagal-Shamanna R, Class CA, Sasaki K, ment of relapsed and refractory acute myeloid leukemia. J Clin
Ravandi F, Cortes JE, et al. Outcomes of acute myeloid leukemia Oncol. 2011;29(8):979–85.
with myelodysplasia related changes depend on diagnostic criteria 63. Goldberg AD, Talati C, Desai P, Famulare C, Devlin SM, Farnoud
and therapy. Am J Hematol. 2020;95(6):612–22. N, et al. TP53 mutations predict poorer responses to CPX-351 in
50. Petersdorf SH, Kopecky KJ, Slovak M, Willman C, Nevill T, acute myeloid leukemia. Blood. 2018;132:1433.
Brandwein J, et al. A phase 3 study of gemtuzumab ozogamicin 64. Madarang E, Lykon J, Nguyen N, Watts JM, Bradley TJ, Chandhok
during induction and postconsolidation therapy in younger patients NS. Real world outcomes of liposomal daunorubicin and cytarabine
with acute myeloid leukemia. Blood. 2013;121(24):4854–60. versus 7+3 in patients with secondary acute myeloid leukemia.
51. Burnett AK, Hills RK, Milligan D, Kjeldsen L, Kell J, Russell NH, Blood. 2020;136:5–6.
et al. Identification of patients with acute myeloblastic leukemia 65. Kim GY, Koprivnikar JL, Testi R, McCabe T, Perry G, Marcotulli
who benefit from the addition of gemtuzumab ozogamicin: results D, et al. Treatment with CPX-351 induces deep responses and TP53
of the MRC AML15 trial. J Clin Oncol. 2011;29(4):369–77. mutation clearance in patients with t-AML and AML MRC, includ-
52. Burnett AK, Russell NH, Hills RK, Kell J, Freeman S, Kjeldsen L, ing younger patients and those with pre-existing MPNs: a real-
et al. Addition of gemtuzumab ozogamicin to induction chemother- world experience. Blood. 2020;136:49–50.
apy improves survival in older patients with acute myeloid leuke- 66. Cortes JE, Lin T, Uy GL, Ryan RJ, Faderl S, Lancet JE. Quality-
mia. J Clin Oncol. 2012;30(32):3924–31. adjusted time without symptoms of disease and toxicity (Q-TWiST)
53. Delaunay J, Recher C, Pigneux A, Witz F, Vey N, Blanchet O, et al. analysis of CPX-351 versus 7+3 in older adults with newly diag-
Addition of gemtuzumab ozogamycin to chemotherapy improves nosed high-risk/secondary acute myeloid leukemia (AML). Blood.
event-free survival but not overall survival of AML patients with 2020;136:55–6.
intermediate cytogenetics not eligible for allogeneic transplantation. 67. Uy GL, Newell LF, Lin T, Goldberg SL, Wieduwilt MJ, Ryan RJ,
Results of the GOELAMS AML 2006 IR study. Blood. et al. Long-term outcomes of allogeneic hematopoietic cell trans-
2011;118(21):79. plantation in patients enrolled in CPX-351-301, a randomized phase
54. Hills RK, Castaigne S, Appelbaum FR, Delaunay J, Petersdorf S, 3 study of CPX-351 versus 7+3 in older adults with newly diag-
Othus M, et al. Addition of gemtuzumab ozogamicin to induction nosed, high-risk and/or secondary AML. Blood. 2020;136:44–5.
chemotherapy in adult patients with acute myeloid leukaemia: a 68. Gaidzik VI, Mayr-Benedikter V, Weber D, Schrade A, Krauter J,
meta-analysis of individual patient data from randomised controlled Walz JS, et al. Higher dose of CPX-351 is associated with
trials. Lancet Oncol. 2014;15(9):986–96. prolonged hematologic recovery: results from an interim safety
55. Magwood-Golston JS, Kessler S, Bennett CL. Evaluation of analysis of the randomized, phase III AMLSG 30-18 trial. Blood.
gemtuzumab ozogamycin associated sinusoidal obstructive syn- 2020;136:46–7.
drome: Findings from an academic pharmacovigilance program 69. Pollyea DA, Tallman MS, de Botton S, Kantarjian HM, Collins R,
review and a pharmaceutical sponsored registry. Leuk Res. Stein AS, et al. Enasidenib, an inhibitor of mutant IDH2 proteins,
2016;44:61–4. induces durable remissions in older patients with newly diagnosed
56. Battipaglia G, Labopin M, Candoni A, Fanin R, El Cheikh J, Blaise acute myeloid leukemia. Leukemia. 2019;33(11):2575–84.
D, et al. Risk of sinusoidal obstruction syndrome in allogeneic stem 70. Arslan S, Zhang J, Dhakal P, Moran J, Naidoo N, Lombardi J, et al.
cell transplantation after prior gemtuzumab ozogamicin treatment: a Outcomes of therapy with venetoclax combined with a
retrospective study from the acute leukemia working party of the hypomethylating agent in favorable-risk acute myeloid leukemia.
EBMT. Bone Marrow Transplant. 2017;52(4):592–9. Am J Hematol. 2020.
Curr Oncol Rep (2021) 23:120 Page 13 of 13 120

71. Wolach O, Levi I, Canaani J, Tadmor T, Tavor S, Hellmann I, et al. conventional care regimens in older patients with newly diagnosed
First results from a nationwide prospective non-interventional study AML with >30% blasts. Blood. 2015;126(3):291–9.
of venetoclax-based 1st line therapies in patients with acute myeloid 82. Kantarjian HM, Thomas XG, Dmoszynska A, Wierzbowska A,
leukemia (AML) - Revive study. Blood. 2020;136:27–8. Mazur G, Mayer J, et al. Multicenter, randomized, open-label,
72. DiNardo CD, Tiong IS, Quaglieri A, MacRaild S, Loghavi S, phase III trial of decitabine versus patient choice, with physician
Brown FC, et al. Molecular patterns of response and treatment advice, of either supportive care or low-dose cytarabine for the
failure after frontline venetoclax combinations in older patients treatment of older patients with newly diagnosed acute myeloid
with AML. Blood. 2020;135(11):791–803. leukemia. J Clin Oncol. 2012;30(21):2670–7.
73. Pei S, Pollyea DA, Gustafson A, Stevens BM, Minhajuddin M, Fu 83. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Santini V, Gattermann
R, et al. Monocytic subclones confer resistance to venetoclax-based N, Germing U, et al. Azacitidine prolongs overall survival com-
therapy in patients with acute myeloid leukemia. Cancer Discov. pared with conventional care regimens in elderly patients with
2020;10(4):536–51. low bone marrow blast count acute myeloid leukemia. J Clin
74. Maiti A, Rausch CR, Cortes JE, Pemmaraju N, Daver NG, Ravandi Oncol. 2010;28(4):562–9.
F, et al. Outcomes of relapsed or refractory acute myeloid leukemia 84. Mesa RA, Li CY, Ketterling RP, Schroeder GS, Knudson RA,
after frontline hypomethylating agent with venetoclax regimens. Tefferi A. Leukemic transformation in myelofibrosis with myeloid
Blood. 2019;134:738. metaplasia: a single-institution experience with 91 cases. Blood.
75. DiNardo CD, Stein EM, de Botton S, Roboz GJ, Altman JK, Mims 2005;105(3):973–7.
AS, et al. Durable remissions with ivosidenib in IDH1-mutated 85. Tadros M, Koehne G, Zahra T, Unzaga J, Shwin M, Chow N.
relapsed or refractory AML. N Engl J Med. 2018;378(25):2386–98. Evaluating acute myeloid leukemia induction regimens in elderly
76.•• Stein EM, DiNardo CD, Pollyea DA, Fathi AT, Roboz GJ, Altman patients with unfavorable risk cytogenetics that are candidates for
JK, et al. Enasidenib in mutant IDH2 relapsed or refractory acute intensive remission therapy. Blood. 2020;136:17.
myeloid leukemia. Blood. 2017;130(6):722–31. This Phase 1/2
86. Salhotra A, Ngo D, Zhang J, Sandhu KS, Al Malki MM, Aribi A,
study led to the FDA approval of enasidenib for treatment of
et al. Clinical outcomes of patients with secondary acute myeloid
IDH2-mutated, relapsed/refractory AML.
leukemia (sAML) treated with hypomethylating agent plus
77. Zeidner JF, Foster MC, Blackford AL, Litzow MR, Morris LE,
venetoclax (HMA-Ven) or liposomal daunorubicin cytarabine
Strickland SA, et al. Randomized multicenter phase II study of
(CPX-351). Blood. 2020;136:37–8.
flavopiridol (alvocidib), cytarabine, and mitoxantrone (FLAM) ver-
sus cytarabine/daunorubicin (7+3) in newly diagnosed acute mye- 87. Kennedy VE, Hui G, Gaut D, Mittal V, Oliai C, Muffly LS, et al.
loid leukemia. Haematologica. 2015;100(9):1172–9. Hypomethylating agents in combination with venetoclax as a
78. Burnett AK, Russell NH, Hills RK, Hunter AE, Kjeldsen L, Yin J, bridge to allogeneic transplant in acute myeloid leukemia. Blood.
et al. Optimization of chemotherapy for younger patients with acute 2020;136:32–3.
myeloid leukemia: results of the medical research council AML15 88. Pollyea DA, Winters A, Jordan CT, Smith C, Gutman JA.
trial. J Clin Oncol. 2013;31(27):3360–8. Allogeneic transplant improves AML outcomes compared to main-
79. Bhella SD, Atenafu EG, Schuh AC, Minden MD, Schimmer AD, tenance venetoclax and azacitidine following response to initial
Gupta V, et al. Front-line FLAG-IDA and Nove-HiDAC chemo- venetoclax and azacitidine therapy. Blood. 2020;136:24.
therapy improves overall survival (OS) and complete remission 89. Gutman JA, Winters A, Amaya ML, McMahon CM, Schowinsky J,
rates (CR) for patients with secondary or therapy-related AML Abbott D, et al. Venetoclax and azacitidine for newly diagnosed
compared to 3&7. Blood. 2015;126(23):4893. non-elderly adult patients with acute myeloid leukemia and adverse
80. Burnett AK, Milligan D, Prentice AG, Goldstone AH, McMullin risk features. Blood. 2020;136:9.
MF, Hills RK, et al. A comparison of low-dose cytarabine and 90. Lo-Coco F, Avvisati G, Vignetti M, Thiede C, Orlando SM,
hydroxyurea with or without all-trans retinoic acid for acute mye- Iacobelli S, et al. Retinoic acid and arsenic trioxide for acute
loid leukemia and high-risk myelodysplastic syndrome in patients promyelocytic leukemia. N Engl J Med. 2013;369(2):111–21.
not considered fit for intensive treatment. Cancer. 2007;109(6):
1114–24. Publisher’s Note Springer Nature remains neutral with regard to jurisdic-
81. Dombret H, Seymour JF, Butrym A, Wierzbowska A, Selleslag D, tional claims in published maps and institutional affiliations.
Jang JH, et al. International phase 3 study of azacitidine vs

You might also like