Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

REVIEW

CURRENT
OPINION Progress in the problem of relapsed or refractory
acute myeloid leukemia
Alice S. Mims a and William Blum b

Purpose of review
The majority of patients with acute myeloid leukemia (AML) die from disease recurrence and historically,
treatment options in both the relapsed and refractory settings of this disease have been limited. However,
new insights into the molecular characterization and biology of relapsed and refractory AML have led to
novel therapeutics and improvement in outcomes in these settings. The current understanding of
mechanisms of disease resistance and status of treatment options both currently available and under
exploration in relapsed and refractory AML are summarized in this review.
Recent findings
The rapid approval of multiple therapeutic agents since 2017 has led to improvement in selected
Downloaded from http://journals.lww.com/co-hematology by BhDMf5ePHKbH4TTImqenVCscvrd+bRBwk4m9f/LleuiLJYCu/ukBASh+3il94Nc3 on 02/11/2019

populations such as isocitrate dehydrogenase and fms-like tyrosine kinase 3-mutated relapsed and
refractory AML with agents such as enasidenib, ivosidenib, and gilteritinib. Despite these advancements,
the only current curative approach remains allogeneic transplantation and only for those minority of
patients that are candidates. However, encouraging results are being seen with a multitude of novel small
molecular inhibitors and immunotherapeutic approaches currently in clinical trials both as single agents and
combination strategies in both upfront and relapsed/refractory AML.
Summary
Continued advancements in the knowledge of various mechanisms of relapse and resistance in AML are
ongoing, leading to the realization that diverse treatment strategies are needed to both prevent and
manage relapsed and refractory disease.
Keywords
acute myeloid leukemia, clonal architecture, immunotherapy agents, relapsed/refractory, small molecule
inhibitors

INTRODUCTION AML is defined as the inability to achieve complete


Acute myeloid leukemia (AML) is a devastating remission after two cycles of intensive induction
diagnosis that is curative only in the minority of therapy (reported 25–30% incidence in younger
patients. Current standard treatment results in 5- adults) [4,5] and 30–50% of older adults [6,7]. Early
year overall survival (OS) for 35–40% of younger relapse is defined as disease recurrence within
patients (<60 years) and 5–15% of older patients 6 months after achievement of initial complete
(60 years) [1,2]. Though chemotherapy has con- remission. Patients with primary refractory disease
siderable toxicities, the risk of treatment-related or early relapse are predicted to have low response
death is actually quite low; most patients die of rates when given salvage chemotherapy and short
leukemia. Allogeneic transplantation (alloHCT) OS compared with those patients with later relapse.
remains the most effective means to reduce the risk This is demonstrated in Table 1 by findings of Walter
of relapse. Vasu et al. recently published results from
2551 AML adult patients treated with standard a
Division of Hematology, Department of Medicine, The Ohio State
intensive chemotherapy regimens without alloHCT
& University Comprehensive Cancer Center, Columbus, Ohio and bDepart-
in first remission [3 ]. Results showed 10-year dis- Department of Hematology and Medical Oncology, Emory University
ease-free survival estimated at only 15% in younger School of Medicine, Winship Cancer Institute, Atlanta, Georgia, USA
patients and less than 2% of the older population, Correspondence to Alice S. Mims, MD, The Ohio State University 320 W
demonstrating the enormity of the problem of AML 10th Avenue, Columbus, OH 43210, USA. Tel: +1 614 685 6031;
relapse with standard chemotherapy alone. e-mail: alice.mims@osumc.edu
Several terms are commonly used to describe Curr Opin Hematol 2019, 26:88–95
and define relapse in AML. Primary refractory DOI:10.1097/MOH.0000000000000490

www.co-hematology.com Volume 26  Number 2  March 2019

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Relapsed or refractory acute myeloid leukemia Mims and Blum

Table 1. Predicted median survival after acute myeloid


KEY POINTS
leukemia treatment failure
 Ivosidenib for IDH1m and enasidenib for IDH2m AML Outcome Median survival (95% CI)
have complete remission rates of approximately 20% All patients n ¼ 1026
and in responders, median OS of at least 18 months in
the relapsed/refractory setting. Failed induction 4 (3–5) months

 Gilteritinib shows single-agent ORR of 51% in FLT3-ITD CR 0–3 months 3 (2–5) months
and TKD mutated AML and allowed some patients to CR 3–6 months 3 (2–4) months
bridge to alloHCT. CR 6–9 months 5 (4–7) months

 Understanding the clonal architecture of AML will aid CR 9–12 months 12 (7–18) months
in determining resistance mechanisms and innovative CR 1–2 years 15 (9–28) months
therapeutic strategies to both prevent and treat relapse
and refractory disease. (Abbreviated version from Walter et al.)
CR, complete remission.

and ongoing research in relapsed and refractory


et al. [8] after analyzing 1026 patients with relapsed/ AML.
refractory AML. Also faring very poorly are patients
who relapse after alloHCT, with estimated 3-year OS
of 12% or less if relapse occurs within 2 years of LESSONS LEARNED FROM STUDIES IN
alloHCT [9]. Regardless of the timeframe or setting, CLONAL ARCHITECTURE
there is no consensus standard of care treatment The ideal approach to relapsed/refractory AML is to
regimen for patients with relapsed/refractory dis- prevent occurrence in the first place. Knowledge
ease. Historically, treatment options have been lim- gained from studies of clonal architecture of AML
ited to intensive chemotherapy regimens or more may help to develop superior therapeutic strategies
palliative approaches with hypomethylating agents at diagnosis and beyond to increase the likelihood of
(HMA), low-dose cytarabine (LDAC), or supportive cure. Ding et al. performed seminal work with
care/comfort measures [1]. However, since 2017, whole-genome sequencing of primary AML tumor
there have been four agents approved by the Federal samples for eight patients at both diagnosis and
Drug Administration (FDA) for patients with relapse [10]. The authors discovered two major
relapsed/refractory AML, and many other agents mutational patterns: the founding clone gained
currently in clinical trials are showing promise. mutations that evolved at time of relapse, and a
Herein, we discuss the progress in clinical care subclone of the founding clone gained additional

FIGURE 1. Schematic of clonal architecture at diagnosis and two subsequent relapses in an AML patient. A FISH plot
representing clonal evolution of a patient throughout the disease course. Diagnosis, first relapse pre-allogeneic transplant, and
second relapse post-allogeneic transplant are depicted. The dominant subclone at both post-chemotherapy relapse and post-
transplantation relapse was derived from a small subclone that was detected at presentation (in red), which evolved with new
mutations of unknown significance after each therapy. AML, acute myeloid leukemia. Reprinted with permission from NEJM.

1065-6251 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-hematology.com 89

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Myeloid disease

mutations and expanded at relapse (similar to that therapy are more likely to respond to subsequent
observed in Fig. 1). In either scenario, the initial therapy. As reported by Keating et al. [13], if AML
choice of chemotherapy failed to eradicate the first complete remission duration was greater than
founding clone; in both scenarios, it is possible that 1 year the likelihood of second complete remission
chemotherapy itself contributed to the expansion or was 60%, versus 19% for those patients with first
emergence of clones with the new mutations [10]. complete remission less than 1 year. These data from
Whether new mutations emerge, or were present at the 1980s unfortunately still largely applies today,
diagnosis but below detectable thresholds, remain an with some modest improvements in supportive
important question. Similarly, there is heterogeneity care. However, promising new results with molecu-
among patients, of course, on this question. In a larly targeted therapies (which will be discussed in
mouse model of AML, Shlush et al. further analyzed the next section) have substantially improved out-
both diagnosis and relapse AML patient samples comes for patients with targetable mutations.
through whole-genome sequencing and transcrip- Without targeted therapy, for patients who have
&&
tional profiling [11 ]. They similarly showed that first complete remission lasting over 1 year, rein-
the origin cell at relapse can vary among patients duction with the same induction regimen used for
with some arising from a more primitive leukemic initial remission can be considered [14]. Typically,
stem cell or a more committed leukemia cell clone high dose cytarabine-based salvage regimens have
that has retained the stem cell transcriptional pro- been recommended for first-line salvage treatment
gram. Their findings highlight that successful thera- for primary refractory or early relapse disease. There
pies to prevent relapse should not only target the are a variety of combination chemotherapy regi-
dominant leukemic clone but also must effectively mens [i.e., mitoxantrone, etoposide, and cytarabine,
target putative leukemic stem cells. At relapse, based fludarabine, cytarabine, and granulocyte colony-
on the origin cell, patients may need different thera- stimulating factor), CLAG-M (cladribine, cytara-
peutic strategies. Christopher et al. investigated bine, granulocyte colony-stimulating factor, and
another potential mechanism of relapse, this time mitoxantrone)] that have been used in this setting
in the post-alloHCT setting. The authors again exam- with estimated response rates of approximately 40–
ined paired diagnostic and relapse AML patient sam- 50% [15–18]. Clearly, there is selection bias in these
&&
ples [12 ]. By performing both exome and RNA individual reports, as older or infirm patients who
sequencing, they determined that in patients who would be unlikely to respond to such therapies are
relapsed after alloHCT, downregulation of major (appropriately) unlikely to receive them.
histocompatibility complex class II genes was There is evidence to support the use of alloHCT in
observed in AML cells along with dysregulation of carefully selected patients with primary refractory or
other pathways that impact immune function. Resid- relapsed AML, with reported success rates of 20% or
ual leukemic cells evolved to downregulate major less [19]. Most centers agree on a role for myeloabla-
histocompatibility complex class II genes (involved tive alloHCT for fit younger patients with primary
in antigen presentation), thus reducing graft-versus- refractory AML. In contrast, though there is no con-
leukemia activity. Together, all of these different sensus, the low likelihood of cure for patients with
articles highlight the complexity of AML and the relapsed AML who are transplanted not in remission
many different resistance and escape mechanisms suggests that such patients (especially those with
that leukemic cells employ to survive. As we continue proliferative disease and circulating blasts) may be
to learn more about the biology of the disease both at better served with alternative treatment approaches.
diagnosis and at relapse, hopefully individualized The goal would be to achieve second complete remis-
therapeutic strategies can be implemented to elimi- sion first rather than proceeding directly to alloHCT
nate risk of recurrence. with active leukemia. Useful predictive models for
alloHCT in patients not in remission help transplant
decision-making by providing risk/benefit estimates
CURRENT TREATMENT STRATEGIES based on clinical and disease-specific factors. One
such method is the Duval score which showed the
Intensive chemotherapy importance of five pre-transplant factors in regards to
For relapsed AML patients who are considered can- survival after alloHCT. Adverse factors are first com-
didates for intensive therapy, treatment selection plete remission duration of less than 6 months, pres-
decisions have historically been based on duration ence of circulating blasts, donor other than a human
of prior remission. A simple rule of thumb is the leukocyte antigen (HLA)-identical sibling, Karnofsky
shorter the remission duration, the lower the likeli- or Lansky score less than 90, and poor-risk cytoge-
hood of achieving subsequent remission. Said in netics. A predictive score of 0 has OS of 42% at 3 years
another way, patients who respond the best to prior versus 10% for a score at least 3 [20].

90 www.co-hematology.com Volume 26  Number 2  March 2019

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Relapsed or refractory acute myeloid leukemia Mims and Blum

New approaches designed to improve outcomes relapsed/refractory disease. Both agents may cause
with intensive chemotherapy include combination a differentiation syndrome with capillary leak and
with small molecule inhibitors, immunotherapies, leukocytosis, at an incidence of approximately 10%.
or other targeted therapeutics and will be discussed This requires careful monitoring during treatment.
in subsequent sections. Importantly, both drugs were approved following
completion of first-in-human, non-randomized clin-
Nonintensive chemotherapy ical trials in relapsed/refractory AML.
The phase 1/2 study of enasidenib included 109
Nonintensive chemotherapy including HMAs such
relapsed/refractory IDH2 mutated AML patients at the
as azacitidine and decitabine, or LDAC has also been
recommended phase 2 dosing schedule of 100 mg
used in relapsed/refractory AML. HMA therapy
daily. The overall response rate (ORR) was 38.5%
given in the salvage setting led to poor complete
which included responses of complete remission,
remission rates between 15–20% with median OS of
complete remission with incomplete count recovery
approximately 6–9 months [1,21,22]. LDAC after
(CRi), partial remission, and morphological leukemia
intensive chemotherapy had complete remission
free state. The complete remission rate was 20.2% with
rates of less than 10% and median post-relapse
an additional 6.4% of patients achieving CRi. The time
survival of 5–6 months [23]. Clearly, none of these
to complete remission was 3.7 months with median
treatment strategies is ideal, and all are typically
duration of response (DoR) of 8.8 months. The median
considered to be of palliative intent. Two drugs were
OS in patients with relapsed/refractory AML was 9.3
recently FDA approved in the upfront setting in
months with estimated 1 year OS of 39%. For those
combination with lower intensity treatment for
patients who achieved a complete remission, the
older patients not candidates for intensive therapy: &&
median OS was 19.7 months [28 ].
venetoclax or glasdegib, respectively. Whether the
For ivosidenib, the phase 1 dose escalation and
addition of either drug to similar regimens in the
expansion clinical study included 125 patients with
relapsed setting will prove similarly efficacious
relapsed/refractory IDH1-mutated AML treated at
remains to be determined.
the clinical efficacy dose of 500 mg daily. Of these
125 patients, the ORR was 41.6% defined as in the
Small molecule inhibitors enasidenib study with a 21.6% complete remission
Small molecule inhibitors including drugs targeting rate and 8.8% complete remission with partial
specific molecular aberrations are new to the arsenal hematologic recovery (CRh). CRh is defined as bone
of AML therapy. These agents are appealing because marrow myeloblasts of less than 5% combined with
of decreased toxicity compared to intensive both absolute neutrophil count more than 500/ml
chemotherapy, the ability to bridge some patients and platelet count more than 50 000/ml. The median
to transplant, increased duration of remissions, and time to complete remission and CRh was 2.7
hematological improvements leading to improve- months with median DoR of 8.2 months. All
ment of quality of life. Both isocitrate dehydrogenase patients who achieved complete remission or CRh
(IDH1 or IDH2) and FMS-like tyrosine kinase 3 (FLT3) had responses occurring within 6 months of starting
inhibitors have been recently approved in AML. ivosidenib. The median OS was 8.8 months and not
reached at data cut-off at 18 months for patients
Isocitrate dehydrogenase inhibitors with complete remission or CRh [29 ].
&&

IDH1 and 2 are critical enzymes involved in the citric With these exciting single-agent findings, IDH
acid cycle. The two major isoforms that are targets in inhibitors are now being explored in upfront and
AML therapeutics include IDH1 which is located in relapsed/refractory settings in combination with
the cytoplasm and IDH2 located in the mitochon- HMA or intensive treatment as well as in the
dria. Both IDH1 and IDH2 catalyze the conversion of post-transplant setting as maintenance therapy
isocitrate to a-ketoglutarate under normal circum- (NCT03683433, NCT02577406, NCT03515512,
stances. Somatic mutations in IDH1 and IDH2 result NCT03728335). New IDH1 inhibitors such as FT-
in converting isocitrate to the oncometabolite 2- 2102 and IDH305 are currently in clinical trials
hydroxyglutarate which leads to impairment of nor- (NCT02719574 and NCT02381886).
mal hematopoietic differentiation through induc-
tion of epigenetic dysregulation [24–27]. The FMS-like tyrosine kinase 3 inhibitors
incidence of IDH mutations in AML is 5–10% for FLT3 is mutated in approximately 30% of AML
IDH1 and 10–15% for IDH2. Inhibitors targeting the patients with the majority being FLT3 internal tandem
abnormal enzyme from mutated IDH2 (enasidenib) duplications (ITD) and less frequently FLT3 tyrosine
and IDH1 (ivosidenib) are oral agents and have kinase domain (TKD). Midostaurin was the first FLT3
recently been FDA approved in the setting of inhibitor FDA approved for FLT3-mutated AML but

1065-6251 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-hematology.com 91

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Myeloid disease

only in the setting of upfront treatment in combina- and HMA, complete remission and CRi rates were
tion with intensive chemotherapy [30]. Midostaurin 70% at the 400 mg dose with azacitidine and 74% at
has not shown significant single agent activity and not the 400 mg dose with decitabine, with time to
felt to be beneficial in the relapsed/refractory setting, response of 1–2 months. This agent has been stud-
at least not as monotherapy [31]. Sorafenib, a multi- ied as monotherapy in patients with relapsed/refrac-
targeted kinase inhibitor, has shown activity in the tory AML, but activity is low. The recent phase 2
relapsed/refractory FLT3 mutated AML setting and has monotherapy study allowed patients with either
been used off-label both as a single agent and in recently relapsed AML (n ¼ 30) or untreated AML
combination with HMA therapy. This is based on unfit for intensive chemotherapy (n ¼ 2) with dose
reported monotherapy complete remission rates of escalation up to 800 mg daily. The complete remis-
6–48% [32,33] and complete remission rates in com- sion rate was 6% and CRi rate was 13% with a
bination with HMA of 16–80% in small series of median OS of 4.7 months. For responders, the
patients [34,35]. median leukemia-free survival was 2.3 months
&
Gilteritinib was recently FDA-approved as mono- [40 ]. Venetoclax is also being explored currently
therapy for relapsed/refractory AML with activity in in the relapsed/refractory setting with multiple
both FLT3-ITD and TKD-mutated disease. In the phase novel–novel combination studies including gilter-
1/2 study, 169 patients with FLT3-mutated AML were itinib and ivosidenib (NCT03484520,
treated at dose of 80 mg/day or above, a dose where NCT03441555, NCT03625505, NCT03471260).
90% of FLT3 phosphorylation inhibition was observed TP53-mutated (TP53m) patients fare extremely
by day 8 of treatment. Of these patients, the ORR of poor with conventional intensive chemotherapy.
52% included rates of complete remission at 11%, CRi However, there have been recent data that show
at 30%, and partial remission at 11%. DoR was potential benefit from HMA therapy, in particular
20 weeks and OS at 31 weeks with 19% of patients the 10-day decitabine approach as shown by Welch
&&
able to bridge to alloHCT [36 ]. These findings led to et al. [41]. There is a planned study comparing the 10-
the approval of this agent in November 2018. Two day decitabine approach versus intensive reinduc-
other second-generation FLT3 inhibitors are also tion chemotherapy for patients with TP53m AML
showing promise in the relapsed/refractory setting, who fail first induction. A potential target in AML
quizartinib and crenolanib. Quizartinib has shown a that includes the TP53m subtype is neural cell devel-
composite complete remission rate of 44–54% in opmentally downregulated 8 (NEDD8)-activating
FLT3-ITD relapsed/refractory AML (does not target enzyme which processes NEDD8 for binding to its
FLT3-TKD). However, responses averaged approxi- target substrates such as Cullin-RING E3 ubiquitin
mately 10 weeks, with resistance notably including ligases. Pevonedistat binds to NEDD8 forming an
&
acquisition of FLT3-TKD mutations [37 ]. Crenolanib adduct tightly bound to NEDD8-activating enzyme
targets FLT3 and platelet-derived growth factor recep- which is then unable to process NEDD8, leading to
tor and has shown single agent activity with CRi rates antiproliferative effects in AML. Treatment appears to
of approximately 39 and 11% partial remission in 18 simultaneously induce DNA damage and DNA dam-
relapsed/refractory FLT3-mutated AML patients with- age responses with compromised DNA repair sensi-
out prior exposure to other FLT3 inhibitors [38]. All tizing transformed cells to DNA-damaging agents
three second-generation FLT3 inhibitors are also being [42]. This agent has shown single-agent responses
explored in combination with induction chemother- in AML with an ORR of approximately 10% (two
apy and HMA therapy in the upfront and relapsed/ complete remission and three partial remissions)
refractory settings (NCT02236013, NCT02668653, [43]; combination with azacitidine in relapsed/refrac-
NCT03661307, NCT01892371, NCT02283177, tory AML showed an ORR of 50% with 20 complete
NCT03250338, NCT02400281). There is also a ran- remissions, two CRi, and seven partial remissions. In
domized placebo-controlled trial underway assessing patients with TP53m, the composite complete remis-
the role of gilteritinib as maintenance post-transplant sion/partial remission rate was 80% with this combi-
for patients with FLT3-ITD mutations to attempt to nation [44]. This agent is being studied in
mitigate the risk of relapse (NCT02997202). combination with 10-day decitabine in newly diag-
nosed TP53m AML patients [NCT03013998].

Additional small molecule inhibitors


Venetoclax is a highly selective, orally bioavailable IMMUNOTHERAPEUTIC STRATEGIES
b-cell lymphoma 2 (Bcl-2) BCL2 inhibitor that was
recently FDA approved for newly diagnosed AML in Targeting CD33
combination with HMA and LDAC therapy for older Gemtuzumab is a humanized CD33 monoclonal
&&
(75 years) or unfit patients [39 ]. For venetoclax antibody linked to cytotoxic calicheamicin, which

92 www.co-hematology.com Volume 26  Number 2  March 2019

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Relapsed or refractory acute myeloid leukemia Mims and Blum

underwent voluntary withdrawal from the market likely, however, that the relatively low mutational
in 2010 because of safety concerns. However, this burden observed in AML (relative to melanoma and
agent was reapproved in 2017 after a revamped non-small cell lung cancer) will limit the use of these
dosing schedule in combination with induction agents in AML, at least in combination with cyto-
chemotherapy showed improvements in clinical toxic therapies. Addition of such agents to immu-
outcome [45]. In relapsed/refractory AML, a phase nologic targeting approach such as BiTEs will be an
2 study enrolled 57 patients with CD33þ AML in first interesting approach for study. There are many
relapse, with initial remission durations between 3 additional other immunotherapeutic agents includ-
and 18 months. Fractionated doses of gemtuzumab ing off the shelf normal donor CAR T cells (targeting
monotherapy were given on days 1, 4, and 7 as this CD123) and natural killer (NK) cell CAR therapy (NK
repeated exposure was thought to enhance internal- CARs with activating NK Group 2D receptors for
ization of the drug and lead to increased intracellular ligands on leukemic cells) [53].
accumulation. With single agent dosing, 15 patients
achieved complete remission with median relapse- CONCLUSION
free survival of 11.6 months and four patients
The treatment arsenal for AML grows with the addi-
achieved CRi with median relapse-free survival of
tion of nonchemotherapeutic strategies such as
8.6 months. No episodes of veno-occlusive disease
were seen with seven patients who underwent small molecular inhibitors and immunotherapeutic
alloHCT after treatment [46]. Other CD33-targeted targeting approaches. Although small molecule
inhibitors do not appear to be curative as mono-
agents are in study including BI 836858, a fully
therapies, these agents may be able to bridge
human CD33 monoclonal antibody; AMV564,
patients to transplant or improve quantity along
CD33xC3 bispecific antibody (BiTE); IMGN779, a
with quality of life in nontransplant candidates.
CD33 antibody-drug conjugate (ADC); AMG330,
Critical to successful treatment of AML will be con-
CD3xCD33 BiTE [NCT03207191, NCT03144245,
tinued evolution of our understanding of the clonal
NCT02674763, NCT02520427]. SGN-CD33A, a
CD33 ADC, was being tested in AML but unfortu- architecture and biology of the disease at diagnosis
nately hepatotoxicity including several cases of veno- through to relapse and hopefully to cure.
occlusive disease was observed. There are no current
Acknowledgements
plans for future clinical trials in AML for this agent.
None.

Other potential immunotherapy targets Financial support and sponsorship


CD123 is the a-subunit of the interleukin-3 receptor A.S.M. reports consulting with Agios, Astellas, and Abb-
and is more highly expressed in AML than normal vie.
hematopoietic stem cells. There are multiple agents W.B. reports research funding with Xencor, Immu-
currently in phase 1/2 clinical trials targeting CD123 noGen, Boehringer Ingelheim (BI). Consulting with BI,
in relapsed/refractory AML including IMGN632, a Astellas, Pfizer, Tolero.
CD123 ADC coupled via peptide linker to unique
DNA alkylating payload; Flotetuzumab, a Conflicts of interest
CD123xCD3 BiTE; and Xmab-14045, a second There are no conflicts of interest.
CD3xCD123 BiTE. Preliminary data for these agents
show promising complete remission/CRi rates rang-
ing from 19 to 25% with final response results REFERENCES AND RECOMMENDED
pending [47–49]. There is also a clinical trial under- READING
Papers of particular interest, published within the annual period of review, have
way testing chimeric antigen receptor (CAR) T cells been highlighted as:
targeting CD123 (NCT03114670). & of special interest
&& of outstanding interest
The success of programmed death-ligand (PD-
L1) and programmed death protein 1 (PD-1) target- 1. Döhner H, Estey E, Grimwade D, et al. Diagnosis and management of AML in
adults: 2017 ELN recommendations from an international expert panel. Blood
ing in other malignances has raised interest in AML. 2017; 129:424–447.
Single agent therapy results were disappointing but 2. Döhner H, Weisdorf DJ, Bloomfield CD. Acute myeloid leukemia. N Engl J
Med 2015; 373:1136–1152.
now these antibodies are being explored in combi- 3. Vasu S, Kohlschmidt J, Mrózek K, et al. Ten-year outcome of patients with
nation with HMA therapy and high-dose cytarabine & acute myeloid leukemia not treatment with allogeneic transplantation in first
complete remission. Blood Adv 2018; 2:1645–1650.
in the relapsed/refractory population. In combina- This large retrospective review illustrates poor disease free survival for the majority of
tion with HMA therapy, early complete remission AML patients who did not undergo allogeneic transplant in first complete remission.
4. Othus M, Appelbaum FR, Petersdorf SH, et al. The fate of patients with newly
rates are being reported at 10–43% and at 35% with diagnosed acute myeloid leukemia who fail primary induction therapy. Biol
high-dose cytarabine combination [50–52]. It is Blood Marrow Transplant 2015; 21:559–564.

1065-6251 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-hematology.com 93

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Myeloid disease

5. Fernadez HF, Sun Z, Yao X, et al. Anthracycline dose intensification of acute 30. Stone RM, Madrekar SJ, Sanford BL, et al. Midostaurin plus chemotherapy for
myeloid leukemia. N Engl J Med 2009; 361:1249–1259. acute myeloid leukemia with a FLT3 mutation. N Engl J Med 2017;
6. Löwenberg B, Ossenkoppele GH, van Putten W, et al. High-dose daunor- 377:454–464.
ubicin in older patients with acute myeloid leukemia. N Engl J Med 2009; 31. Fischer T, Stone RM, Deangelo DJ, et al. Phase IIB trial of oral midostaurin
361:1235–1248. (PCK412), the FMS-like tyrosine kinase 3 receptor (FLT3) and multi-targeted
7. Estey E. Why are there so few randomized trials for patients with primary kinase inhibitor, in patients with acute myeloid leukemia and high-risk mye-
refractory acute myeloid leukemia? Best Pract Res Clin Haematol 2016; lodysplastic syndrome with either wild-type or mutated FLT3. J Clin Oncol
29:324–328. 2010; 28:4339–4345.
8. Walter RB, Othus M, Löwenberg B, et al. Empiric definition of eligibility criteria 32. Borthakur G, Kantarjian H, Ravandi F, et al. Phase I study of sorafenib in
for clinical trials in relapsed/refractory acute myeloid leukemia: analysis of patients with refractory or relapsed acute leukemias. Haematologica 2011;
1,892 patients from HOVON/SAKK and SWOG. Haematologica 2015; 96:62–68.
100:e490–e411. 33. Metzelder SK, Schroeder T, Finck A, et al. High activity of sorafenib in FLT3-
9. Bejanyan N, Weisdorf DJ, Logan BR, et al. Survival of patients with acute ITD-positive acute myeloid leukemia synergizes with allo-immune effects to
myeloid leukemia relapsing after allogeneic hematopoietic cell transplanta- induce sustained responses. Leukemia 2012; 26:2353–2359.
tion: a center for international blood and marrow transplant research study. 34. Ravandi F, Alattar ML, Grunwalk MR, et al. Phase 2 study of azacytidine plus
Biol Blood Marrow Transplant 2015; 21:454–459. sorafenib in patients with acute myeloid leukemia and FLT-3 internal tandem
10. Ding L, Ley TJ, Larson DE, et al. Clonal evolution in relapsed acute myeloid duplication mutation. Blood 2013; 121:1655–1662.
leukaemia revealed by whole-genome sequencing. Nature 2012; 35. Muppidi MR, Portwood S, Griffiths EA, et al. Decitabine and sorafenib therapy
481:506–510. in FLT-3 ITD-mutant acute myeloid leukemia. Clin Lymphoma Myeloma Leuk
11. Shlush LI, Mitchell A, Heisler L, et al. Tracing the origins of relapse in acute 2015; 15:S73–S79.
&& myeloid leukemia to stem cells. Nature 2017; 547:104–108. 36. Perl AE, Altman JK, Cortes J, et al. Selective inhibition of FLT3 by
Whole-genome sequencing and profiling demonstrates differences between the && gilteritinib in relapsed or refractory acute myeloid leukaemia: a multicenter,
origin cell of relapse in patients with relapsed AML. first-in-human, open-label, phase 1-2 study. Lancet Oncol 2017;
12. Christopher MJ, Petti AA, Rettig MP, et al. Immune escape of relapsed AML 18:1061–1075.
&& cells after allogeneic transplantation. N Engl J Med 2018. [Epub ahead of The Phase 1/2 study that led to the FDA-approval of gilteritinib in relapsed and
print]. PMID 30380364. refractory FLT3 mutated AML.
This study demonstrates a mechanism of AML relapse after allogeneic stem cell 37. Cortes JE, Tallman MS, Schiller GJ, et al. Phase 2b study of 2 dosing regimens
transplant through downregulation of major histocompatibility complex class II & of quizartinib monotherapy in FLT3-ITD-mutated, relapsed or refractory AML.
genes. Blood 2018; 132:598–607.
13. Keating MJ, Kantarjian H, Smith TL, et al. Response to salvage therapy and The Phase 2b study showing single-agent response with quizartinib in relapsed
survival after relapse in acute myeloid leukemia. J Clin Oncol 1989; and refractory FLT3-ITD mutated AML.
7:1071–1080. 38. Cortes JE, Kantarjian HM, Kadia TM, et al. Crenolanib besylate, a type I pan-
14. Breems DA, Van Putten WL, Huijgens PC, et al. Prognostic index for adult FLT3 inhibitor, to demonstrate clinical activity in multiply relapsed FLT3-ITD
patients with acute myeloid leukemia in first relapse. J Clin Oncol 2005; and D835 AML. Presented at the American Society of Clinical Oncology
23:1969–1978. Meeting 2016, Chicago, IL. Abstract 7008
15. Herzig RH, Lazarus HM, Wolff SN, et al. High-dose cytosine arabinoside 39. DiNardo CD, Pratz K, Pullarkat V, et al. Venetoclax combined with decitabine
therapy with and without anthracycline antibiotics for remission reinduction of && or azacitidine in treatment-naı̈ve, elderly patients with acute myeloid leukemia.
acute nonlymphoblastic leukemia. J Clin Oncol 1985; 3:992–997. Blood 2018. [Epub ahead of print]. PMID 30361262.
16. Amadori S, Arcese W, Isacchi G, et al. Mitoxantrone, etoposide, and inter- Phase 1b dose escalation and expansion study of venetoclax in combination with
mediate-dose cytarabine: an effective and tolerable regimen for the treatment hypomethylating agents in newly diagnosed older AML patients that led to the
of refractory acute myeloid leukemia. J Clin Oncol 1991; 9:1210–1214. FDA-approval of this combination therapy in this setting.
17. Montillo M, Mirto S, Petti MC, et al. Fludarabine, cytarabine, and G-CSF 40. Konopleva M, Pollyea DA, Potluri J, et al. Efficacy and biological correlates of
(FLAG) for the treatment of poor risk acute myeloid leukemia. Am J Hematol & response in a phase II study of venetoclax monotherapy in patients with acute
1998; 58:105–109. myeloid leukemia. Cancer Discov 2016; 60:1106–1117.
18. Wierzbowska A, Robak T, Pluta A, et al. Cladribine combined with high doses This Phase II study demonstrated overall low clinical responses to venetoclax
of arabinoside cytosine, mitoxantrone, and G-CSF (CLAG-M) is a highly monotherapy in the setting of relapsed and refractory AML.
effective salvage regimen in patients with refractory and relapsed acute 41. Welch JS, Petti AA, Miller CA, et al. TP53 and decitabine in acute myeloid
myeloid leukemia of the poor risk: a final report of the Polish Adult Leukemia leukemia and myelodysplastic syndromes. N Engl J Med 2016;
Group. Eur J Haematol 2008; 80:115–126. 375:2023–2036.
19. Gyurkocza B, Lazarus HM, Giralt S. Allogeneic hematopoietic cell transplan- 42. Bhalla KN, Fiskus W. NEDD8 and HDACs: promising cotargets in AML.
tation in patients with AML not achieving remission: potentially curative Blood 2016; 127:2168–2170.
therapy. Bone Marrow Transplant 2017; 52:1083–1090. 43. Swords RT, Erba HP, DeAngelo DJ, et al. Pevonedistat (MLN4924), a first-in-
20. Duval M, Klein JP, He W, et al. Hematopoietic stem-cell transplantation for class NEDD8-activating enzyme inhibitor, in patients with acute myeloid
acute leukemia in relapse or primary induction failure. J Clin Oncol 2010; leukemia and myelodysplastic syndromes: a phase 1 study. Br J Haematol
28:3730–3738. 2015; 169:534–543.
21. Ivanoff S, Gruson B, Chantepie SP, et al. 5-Azacytidine treatment for relapsed 44. Swords RT, Coutre S, Maris MB, et al. Pevonedistat, a first-in-class NEDD8-
or refractory acute myeloid leukemia after intensive chemotherapy. Am J activating enzyme inhibitor, combined with azacitidine in patients with AML.
Hematol 2013; 88:601–605. Blood 2018; 131:1415–1424.
22. Ritchie EK, Feldman EJ, Christos PJ, et al. Decitabine in patients with newly 45. Lambert J, Pautas C, Terré C, et al. Gemtuzumab ozogamicin for de novo
diagnosed and relapsed acute myeloid leukemia. Leuk Lymphoma 2013; acute myeloid leukemia: final efficacy and safety updates from the open-label,
54:2003–2007. phase 3 ALFA-0701 trial. Haematologica 2018. [Epub ahead of print]. PMID
23. Sarkozy C, Gardin C, Gachard N, et al. Outcome of older patients with acute 30076173.
myeloid leukemia in first relapse. Am J Hematol 2013; 88:758–764. 46. Taksin AL, Legrand O, Raffoux E, et al. High efficacy and safety profile of
24. Dang L, White DW, Gross S, et al. Cancer-associated IDH1 mutation fractionated doses of mylotarg as induction therapy in patients with relapsed
produce 2-hydroxygluturate. Nature 2009; 462:739–744. acute myeloblastic leukemia: a prospective study of the alfa group. Leukemia
25. Ward PS, Patel J, Wise DR, et al. The common feature of leukemia-associated 2007; 21:66–71.
IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha- 47. Daver NG, Erba HP, Papadantonakis N, et al. A Phase I, First-in-Human Study
ketoglutarate to 2-hydroxygluturate. Cancer Cell 2010; 17:225–234. evaluating the safety and preliminary antileukemia activity of IMGN632, a
26. Figueroa ME, Abdel-Wahab O, Lu C, et al. Leukemic IDH1 and IDH2 novel CD123-targeting antibody-drug conjugate, in patients with relapsed/
mutations results in a hypermethylation phenotype, disrupt TET2 function, refractory acute myeloid leukemia and other CD213-opositive hematologic
and impair hematopoietic differentiation. Cancer Cell 2010; 18:553–567. malignancies. Presented at the American Society of Hematology Meeting
27. Lu C, Ward PS, Kapoor GS, et al. IDH mutation impairs histone demethylation 2018, San Diego, CA. Abstract 27
and results in a block to cell differentiation. Nature 2012; 483:474–478. 48. Uy GL, Rettig MP, Vey N, et al. Phase 1 Cohort expansion of flotetuzumab, a
28. Stein EM, DiNardo CD, Pollyea DA, et al. Enasidenib in mutant IDH2 relapsed CD123xCD3 Bispecific Dart protein in patients with relapsed/refractory
&& or refractory acute myeloid leukemia. Blood 2017; 130:722–731. acute myeloid leukemia (AML). Presented at the American Society of Hema-
The Phase 1/2 study that led to the FDA-approval of enasidenib in relapsed and tology Meeting 2018, San Diego, CA. Abstract 764
refractory IDH2 mutated AML. 49. Ravandi F, Bashey A, Foran JM, et al. Complete responses in relapsed/
29. DiNardo CD, Stein EM, de Botton S, et al. Durable remissions with ivosidenib in refractory acute myeloid leukemia (AML) patients on a weekly dosing sche-
&& IDH1-mutated relapsed or refractory AML. N Engl J Med 2018; 378:2386–2398. dule of XmAB14045, a CD123 x CD3 T cell-engaging bispecific antibody:
The Phase 1 dose escalation and expansion study that led to the FDA-approval of Initial results of a phase 1 study. Presented at the American Society of
ivosidenib in relapsed and refractory IDH1 mutated AML. Hematology Meeting 2018, San Diego, CA. Abstract 736

94 www.co-hematology.com Volume 26  Number 2  March 2019

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Relapsed or refractory acute myeloid leukemia Mims and Blum

50. Assi R, Kantarjian HM, Daver NG, et al. Results of a phase 2, open-label study 52. Zeidner JF, Vincent BG, Ivanova A, et al. Genomics reveal potential
of idarubicin (I), cytarabine (A) and nivolumab (Nivo) in patients with newly biomarkers of response to pembrolizumab after high dose cytarabine in
diagnosed Acute Myeloid Leukemia (AML) and high-risk myelodysplastic an ongoing phase II trial in relapsed/refractory AML. Presented at the
syndrome (MDS). Presented at the American Society of Hematology Meeting American Society of Hematology Meeting 2018, San Diego, CA. Abstract
2018, San Diego, CA. Abstract 905 4054
51. Lindblad KE, Thompson J, Gui G, et al. Pembrolizumab and decitabine for 53. Sallman DA, Brayer J, Sagatys EM, et al. NKG2D-Based chimeric antigen
refractory or relapsed acute myeloid leukemia. Presented at the American receptor therapy induced remission in a relapsed/refractory acute myeloid
Society of Hematology Meeting 2018, San Diego, CA. Abstract 1437 leukemia patient. Haematologica 2018; 103:e424–e426.

1065-6251 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-hematology.com 95

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.

You might also like