Вы находитесь на странице: 1из 9

CE: Tripti; CCO/300606; Total nos of Pages: 9;

CCO 300606

REVIEW

CURRENT
OPINION Current management of patients with chronic
myelomonocytic leukemia
Elvira Mora a and Guillermo F. Sanz a,b

Purpose of review
The present review focuses on the current management of patients with chronic myelomonocytic leukemia
(CMML) and the most recent developments in the field.
Recent findings
CMML is a heterogeneous malignant myeloid disorder sharing features of myelodysplastic syndromes
(MDS) and myeloproliferative neoplasms and characterized by peripheral blood monocytosis and
increased risk of progression to acute leukemia. Its natural course is highly variable and use of CMML-
specific prognostic scoring systems is strongly recommended for tailoring treatment. Multiple recent studies
have showed that somatic mutations, which are almost always present have a relevant and independent
impact on survival but lack a clear role in predicting the response to currently available drugs.
Summary
The incorporation of somatic mutations to prognostic scoring systems has improved the prediction of
patients’ outcomes. Current treatment for CMML remains unsatisfactory. Allogeneic hematopoietic cell
transplantation is the only curative option but is applicable to a minority of patients. Usually higher-risk
patients displaying MDS-like characteristics are treated with hypomethylating agents (HMAs), whereas
those with myeloproliferative features generally receive hydroxyurea or HMAs but none of these drugs
substantially modify the natural history of CMML. Newer therapies are clearly needed.
Keywords
chronic myelomonocytic leukemia, hematopoietic cell transplantation, hypomethylating agents, mutations,
prognosis, treatment

INTRODUCTION exclusion of reactive monocytosis. Differential diag-


Chronic myelomonocytic leukemia (CMML) is a rare nosis should also include other malignant myelo-
clonal hematological malignancy with characteris- proliferative disorders as outlined in the 2008 WHO
tics of both myelodysplastic and myeloproliferative criteria [1] and the 2016 revision [2] for the diagnosis
disorders and a highly variable clinical course. For- of CMML, as shown in Table 1. The latter revision
merly classified among myelodysplastic syndromes incorporated the CMML-0 category (<5% bone mar-
(MDS), CMML is currently included in the MDS/ row blasts), based on a study that showed that
myeloproliferative (MPN) neoplasms by the World CMML-0 had a significantly better prognosis than
Health Organization (WHO) classification [1]. CMML-1 [3]. Interestingly, this finding has not been
In the present article, we do not intend to pro- confirmed in a large series recently reported [4].
vide an exhaustive description of the disease, but Careful cytomorphological evaluation of blast
rather to focus on recent relevant issues related to percentage, including myeloblasts, monoblasts, and
the management of patients with CMML and offer promonocytes, in the peripheral blood and bone
currently available evidence to assist physicians in
decision-making. a
Hospital Universitario y Politécnico La Fe, Valencia and bCIBERONC,
Instituto Salud Carlos III, Madrid, Spain
Correspondence to Guillermo F. Sanz, MD, PhD, Hematology Depart-
Diagnosis ment, Hospital Universitario y Politécnico La Fe, Torre F, Planta 7, Avenida
The diagnosis of CMML requires a 3-month persis- Fernando Abril Martorell, 106, 46026 Valencia, Spain.
tent monocytosis, defined as an absolute monocyte Tel: +34 96 124 4925; e-mail: sanz_gui@gva.es
count more than 1  109/l that should represent Curr Opin Oncol 2018, 30:000–000
more than 10% of the WBC differential [2], after DOI:10.1097/CCO.0000000000000486

1040-8746 Copyright ß 2018 Wolters Kluwer Health, Inc. All rights reserved. www.co-oncology.com

Copyright © 2018 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; CCO/300606; Total nos of Pages: 9;
CCO 300606

Hematologic malignancies

marrow aspirate smears as well as the detection of


KEY POINTS myelodysplastic features in at least one hematopoi-
 Flow cytometry immunophenotyping of peripheral etic cell lineage are essential for diagnosis. A bone
blood monocytes and the presence of cytogenetic marrow biopsy is strongly recommended by many
abnormalities or somatic gene mutations are powerful experts and considered as mandatory in case of ‘dry
tools for establishing the diagnosis of CMML. tap’ bone marrow aspiration to assess bone marrow
fibrosis. The presence of myelofibrosis in CMML has
 Several specific somatic mutations are clearly
associated with long-term outcomes and may be recently been associated with inferior overall sur-
&

valuable for establishing a more accurate prognosis in vival (OS) [5 ,6], presence of myeloproliferative
the individual patient. In contrast, the relevance of (MP)-like features, and more frequent JAK2 muta-
somatic mutations for predicting response to HMAs tions [6].
is unclear. Flow cytometry immunophenotyping (FCI) in
 Allogeneic hematopoietic cell transplantation is the only CMML has been recognized as a valuable tool for
potentially curative treatment approach for patients with CMML diagnosis and for monitoring response to
CMML but relevant questions regarding the procedure therapy [7,8]. The presence of classical CD14þ/
remain unanswered. CD16- monocytes in peripheral blood more than
94% of total monocytes constitutes a simple, rapid
 Hydroxyurea and HMAs are the most commonly used
treatment alternatives for CMML patients who are not and robust diagnostic marker to distinguish CMML
candidates to allo-HCT but they do not modify the from other causes of monocytosis, with a sensitivity
natural history of the disorder. and specificity of 90.6 and 95.1%, respectively [9].
This approach has been recently validated in two
 The development of new treatment strategies &
smaller studies [10,11 ], which stress the importance
is essential.
of performing the analysis within the first 24 h after
sampling to avoid false negative results [10] and the

Table 1. WHO diagnostic criteria for chronic myelomonocytic leukemia and categories

WHO 2008 [1] WHO 2016 [2]

Criteria Persistent (3 months) peripheral blood Persistent peripheral blood monocytosis 1  109/l, with
monocytosis 1  109/l monocytes accounting for 10% of the WBC count
Lack of Philadelphia chromosome or BCR-ABL Not meeting WHO criteria for BCR-ABL1þ CML, PMF,
fusion gene polycythemia vera, or ET
No evidence of PDGFRA or PDGFRB No evidence of PDGFRA, PDGFRB, or FGFR1 rearrangements
rearrangements in cases with eosinophilia or PCM1-JAK2 fusions in cases with eosinophilia
<20% blasts in the peripheral blood and bone <20% blasts in the peripheral blood and bone marrowa
marrowa
At least one of the following: (a) Myelodysplastic At least one of the following: (a) Myelodysplastic features in
features in at least one cell line. (b) Presence of at least one cell line. (b) Presence of an acquired clonal
an acquired clonal cytogenetic or mutational cytogenetic or mutational abnormality in hematopoietic
abnormality in hematopoietic cells. (c) All other cells. (c) All other causes of monocytosis have been
causes of monocytosis have been excluded. excluded.
WHO categories
CMML-0 – <2% blasts in peripheral blood and <5% blasts in bone
marrow
CMML-1 <5% blasts in peripheral blood and/or <10% 2–4% blasts in peripheral blood and/or 5–9% blasts in
blasts in bone marrow bone marrow
CMML-2 5–19% blasts in peripheral blood, 10–19% in 5–19% blasts in peripheral blood, 10–19% in bone marrow,
bone marrow, and/or when any Auer rods are and/or when any Auer rods are present
present
FAB categories
MD-CMML WBC <13  109/l WBC <13  109/l
9
MP-CMML WBC 13  10 /l WBC 13  109/l

Changes in the 2016 revision are in bold.


WHO, World Health Organization; FAB, French-American-British; WBC, white blood cell; CML, chronic myeloid leukemia; PMF, primary myelofibrosis; ET:
essential thrombocythemia.
a
Count of blasts includes myeloblasts, monoblasts, and promonocytes (mature monocytes displaying atypical cytologic features are common and are excluded
from the blast count).

2 www.co-oncology.com Volume 30  Number 00  Month 2018

Copyright © 2018 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; CCO/300606; Total nos of Pages: 9;
CCO 300606

Current management of patients with CMML Mora and Sanz

capacity to distinguish CMML from other MPN with chromosomal abnormalities, resulting in shorter
& &
associated monocytosis [11 ]. It has therefore been OS and leukemia-free survival (LFS) [15 ].
suggested that the current cautious recommenda- A large number of gene mutations have been
tion of a 3-month delay for CMML diagnosis could found in CMML, illustrating its molecular heteroge-
be avoided by using this FCI criteria [9,10]. neity and improving our knowledge of the biology of
the disease. As shown in Table 2, which depicts the
relative frequencies of these mutations, the most
Cytogenetic and mutational profiles frequently mutated genes are TET2, SRSF2, and
Cytogenetic abnormalities and somatic mutations ASXL1, followed by mutations in RUNX1 and RAS
&&
are found respectively in 25–30% [12–14] and up to pathway [16,17 ,18]. The range of cellular pathways
& &&
95% [15 ,16,17 ,18–25] of CMML patients, provid- affected by those gene mutations includes epigenetic,
ing definitive evidence of clonality and supporting splicing, transcription, tumor suppression, and cell
the correct diagnosis in doubtful cases. signaling regulation. Although none of them is dis-
The most frequent cytogenetic abnormalities ease-specific, the high frequency of some mutations
are trisomy 8, loss of Y chromosome, abnormalities certainly confirms that CMML is an MDS-indepen-
of chromosome 7, and complex karyotypes. Cyto- dent entity. Recent findings suggest that ancestral
genetic findings have a strong influence in OS and a mutations that drive the pathogenesis of the disease
validated CMML-specific cytogenetic risk classifica- are frequently TET2, SRSF2, and ASXL1 while others
tion recognizes three risk categories: low risk (nor- are predominantly secondary mutations, that can
& &
mal karyotype or loss of Y chromosome as a single trigger disease progression [19 ,20 ].
anomaly), high risk (presence of trisomy 8 or abnor- Signaling-associated mutations (JAK2 and RAS
malities of chromosome 7, or complex karyotype), pathway mutations) are more common in MP-
& &
and intermediate risk (all other abnormalities) [13]. CMML [11 ,16,20 ]. The distribution of gene muta-
On the other hand, therapy-related CMML (t- tions or frequency of mutations in TP53 in t-CMML
&
CMML) has been associated with a higher frequency and de-novo CMML are similar [15 ]. Although
of both abnormal karyotypes and higher-risk TET2 mutations are broadly prevalent, only one

Table 2. Relative frequencies of somatic mutations in patients with chronic myelomonocytic leukemia

Class of genetic Itzykson et al. Elena et al. Patnaik et al.


&& &
mutation Gene 2013 [16] (n ¼ 173–312) 2016 [17 ] (n ¼ 214) 2018 [15 ] (n ¼ 265)

Detection of 1 somatic mutation 95% 93% NA


Epigenetic regulators TET2 58 44 58
DNMT3A 2 NA 6
IDH1 0.4 NA 2
IDH2 6 6 6
Chromatin regulation ASXL1a 40 37 51
EZH2 5 7 3
Splicing SRSF2 46 39 48
SF3B1 6 6 6
U2AF1 5 4 7
ZRSR2 8 4 3
Cell signaling NRAS 11 12 15
KRAS 8 9 5
CBL 10 8 13
PTPN11 NA NA 3
JAK2 V617F 8 7 8
FLT3 3 3
Transcription factors RUNX1 15 8 8
Tumor suppressor genes TP53 1 7 3
Other SETBP1 NA 9 14

NA, not available.


a
Nonsense and frameshift mutations.

1040-8746 Copyright ß 2018 Wolters Kluwer Health, Inc. All rights reserved. www.co-oncology.com 3

Copyright © 2018 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; CCO/300606; Total nos of Pages: 9;
CCO 300606

Hematologic malignancies

marrow blasts 5%;

usseldorf scoring system; MPM, Mayo prognostic model; CPSS, CMML-specific prognostic scoring system; GFMM, Groupe Français des Myélodysplasies model; MMM,
WBC 13  109/l;
study suggests a favorable OS in the absence of

(CPSS cytogenetics
CPSS-Mol [17 ]

þ ASXL1, NRAS,
Gene risk groups

mutations.; bone
SETBP1, RUNX1
&&

RBC transfusion
ASLX1 mutation [21]. Frameshift and nonsense

dependence

Mayo molecular model; CPSS-Mol, Molecular CMML-specific prognostic scoring system; Hb, hemoglobin; Plt, platelets; IMC, immature myeloid cells; ALC, absolute lymphocyte count, LDH: lactate dehydrogenase;
ASXL1 mutations have been correlated to adverse

214

NR
64
37
18
OS in several studies [16,18]. In addition, the co-
occurrence of EZH2 and ASXL1 has been associated
with shorter OS in comparison to ASXL1mt patients
alone [22]. Similarly, DNMT3A mutations may

dl; Plt <100  109/


AMC >10  109/l;

blood; Hb <10 g/
&
adversely impact on OS and LFS [23 ]. The number

l; ASXL1 mutations
IMC in peripheral
of driver mutations also determines survival out-

MMM [18]
comes, as recently demonstrated in a study where

466

59
97

31
16
a shorter OS was observed in CMML patients with
&
three or more concomitant mutations [24 ]. Not
unexpectedly, TP53 mutation, although uncom-
mon in CMML (4%), has been also associated with

<10 or 11 g/dl (by sex);


WBC >15  109/l; Hb
&
poorer OS [24 ]. Conversely, a recent meta-analysis

Plt <100  109/l;


ASXL1 mutations
Age >65 years;
revealed no significant effect on prognosis of SRSF2

GFMM [16]
mutations in CMML patients [25].

312

NR
38

14
Assessment of prognosis
The use of one of the prognostic scoring indexes that
are offered in Table 3 is strongly recommended for risk

CMML WHO type;


assessment of the individual patient with CMML.

CMML FAB type;

RBC transfusion
CMML-specific
cytogenetics;

dependence
Most of them have been externally validated and have CPSS [29]
shown a comparable performance when tested in a
578

31
72

13
5
large international dataset [26]. Among those CMML-

CMML, chronic myelomonocytic leukemia; WBC, white blood cell; AMC, absolute monocyte count; NR, not reached.
specific prognostic tools, some of them include only
peripheral blood and bone marrow characteristics,
which are readily available for practically all CMML
AMC >10  109/l; IMC

patients [12,27,28]. The more recently developed also


in peripheral blood;

Plt <100  109/l


Hb <10 g/dl;

include cytogenetics [29,30], gene mutations [16,18],


MPM [28]

&&
or both cytogenetics and gene mutations [17 ]. The
226

18
32

10
Table 3. Prognostic scoring systems for chronic myelomonocytic leukemia

mutational status of ASXL1 has recently been incor-


porated into two models developed by the Groupe
Francophone des Myelodysplasies [16] and Mayo
Clinic [18]. The so-called CMML-specific prognostic
scoring system (CPSS)-Molecular, integrates the
>5%; LDH >200 U/l;
bone marrow blasts

genetic score (that includes the CPPS cytogenetic risk


Hb <9 g/dl; Plt
<100  109/l
DUSS [27]

score and the mutational status of not only ASXL1 but


288

also NRAS, SETBP1, and RUNX1) along with RBC


26
93

11

transfusion requirements, WBC and bone marrow


&&
blast counts [17 ]. Potential reasons for using the
classical CPSS [29] is that is relatively simple and the
MDAPS, MD Anderson prognostic score; DUSS, D€

only one that has also been validated in the transplan-


Hb < 12 g/dl; IMC
in peripheral blood;
ALC >2.5  109/l;

&&
tation setting [31,32 ]. Furthermore, this prognostic
bone marrow
MDAPS [12]

blasts >10%

score is able to segregate 4 risk groups with signifi-


Risk groups, median OS in months
213

cantly different OS. Patients with low- or intermedi-


15
24

8
5

ate-1 risk by CPSS are considered as lower-risk patients,


whereas those with intermediate-2 or high-risk are
deemed as higher-risk and, thus, as potential candi-
dates for a curative treatment approach.
Prognostic score

Intermediate-1
Intermediate-2
No. of patients

included

Current treatment approach


Variables

High
Low

There is a lack of evidence-based recommendations


for the selection of treatment in patients with

4 www.co-oncology.com Volume 30  Number 00  Month 2018

Copyright © 2018 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; CCO/300606; Total nos of Pages: 9;
CCO 300606

Current management of patients with CMML Mora and Sanz

CMML, since most data on efficacy and safety are source. An international expert panel position
from small uncontrolled studies or from clinical trials report recommends to perform allo-HCT in CMML
designed for MDS. Further, the criteria used for assess- patients with intermediate-2 or high-risk score by
&&
ing response in these trials did not consider end CPSS [43 ] after assessing the allo-HCT associated
points related to the control of the myeloproliferative mortality risk by the HCT-comorbidity index [44].
component of CMML. An international consortium In lower-risk patients, transplantation could be
proposed in 2015 specific response criteria for MDS/ delayed until disease progression. All symptomatic
MPN [33] that has been recently retrospectively vali- younger patients should be offered the opportunity
dated for CMML [34]. Finally, the variable clinical to discuss the indication of allo-HCT shortly after
course of the disease, advanced age, and disappoint- diagnosis. Regarding disease status at transplanta-
ing long-term efficacy of currently available treat- tion, a large registry-based study [40] reported a
ment alternatives, complicate treatment choice. better outcome in patients who were transplanted
in complete remission compared to those with
active disease, whereas another study [39] did not
Allogeneic hematopoietic cell transplantation find this relationship. The potential advantage of
Allogeneic hematopoietic cell transplantation (allo- using a particular treatment modality before trans-
HCT) is the only potentially curative therapy for plant is unclear. A recent single-institution retro-
patients with CMML, but only a minority of patients spective study on 83 patients showed a lower
are candidates for transplantation. In addition, most incidence of relapse and better disease-free survival
available data regarding the role of allo-HCT in in patients treated with HMAs before transplanta-
CMML come from a limited number of retrospective tion than in those receiving chemotherapy or sup-
&&
studies [32 ,35–42] that are shown in Table 4. Over- portive care [42]. In contrast, in a large CIBMTR
&&
all, these studies show that allo-HCT is a valid study [32 ] on 209 patients, outcomes of treated
treatment option for selected patients, resulting in or untreated patients prior to transplantation were
long-term remission in up to 40% of transplanted similar. Reasons for these discrepant results have
patients. However, they do not allow drawing defin- been the subject of a recent commentary [45]. The
itive conclusions on the timing of transplantation, presence of RAS-pathway mutations has been signif-
patients who may benefit most from this procedure, icantly associated with worse OS after transplanta-
need and type of pretreatment, most appropriate tion in patients with MDS/MPN [46] but this finding
conditioning regimen, and best hematopoietic cell requires confirmation.

Table 4. Allogeneic hematopoietic cell transplantation in the treatment of chronic myelomonocytic leukemia
Condition- Type of Outcome
ing (%) donor (%) (%)
Complete
Study No. of Median remission
References Institution period patients age (Range) at HCT (%) MAC RIC Related Unrelated OS DFS

Elliot et al. 2006 [35] Mayo Clinic 1992–2004 17 50 (26–60) 6 94 6 82 18 18 (3y) 18 (3y)
Ocheni et al. Hamburg 2003–2007 12a 56 (37–66) 17 54 46 15 85 75 (3y) 50 (3y)
2009 [36] University
Krishnamurthy et al. Kings College 1998–2007 18 54 (38–66) <50% 17 83 39 61 31 (3y) 31 (3y)
2010 [37]
Eissa et al. FHCRC 1986–2008 85 51.7 (1–69) NA 68 32 45 55 40 (10y) 38 (10y)
2011 [38]
Park et al. SFGM-TC 1992–2009 73 53 (29–67) 21 41 59 58 42 32 (3y) 29 (3y)
2013 [39]
Symeonidis et al. EBMT 1988–2009 513 53 (18–75) 26 52 48 56 44 33 (4y) 27 (4y)
2016 [40]
Sharma et al. Mayo Clinic 1990–2014 36 53 (18–66) NA 61 39 NA NA NA NA
2015 [41]
Kongtim et al. MDACC 1991–2013 83 57 (18–78) 29 77 23 36 64 32–36 (3y) NA
2016 [42]
&&
Liu et al. 2017 [32 ] CIBMTR 2001–2012 209 57 (23–74) NA 51 49 35 64 38 (3y) 27 (3y)

NA, not available; MAC, myeloablative conditioning; RIC, reduced intensity conditioning; OS, overall survival; DFS, disease-free survival; FHCRC, Fred
Hutchinson Cancer Research Center; SFGM-TC, Societé Francaise de Greffe de Moelle et Therapie Cellulaire; EBMT, European Blood and Marrow Transplant;
MDACC, MD Anderson Cancer Center; CIBMTR, Center for International Blood and Marrow Transplant Research.
a
One patient received two transplants.

1040-8746 Copyright ß 2018 Wolters Kluwer Health, Inc. All rights reserved. www.co-oncology.com 5

Copyright © 2018 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; CCO/300606; Total nos of Pages: 9;
CCO 300606

Hematologic malignancies

Hypomethylating agents and other retrospective study compared azacitidine with


treatments hydroxyurea as first-line treatment and showed a
The currently available treatment for those CMML trend for better OS with azacitidine [51]. The results
patients who are not candidates for allo-HCT is of the ongoing prospective randomized DACOTA
symptom-directed and does not substantially mod- trial (NCT02214407), comparing decitabine to
ify the natural history of the disease. Schematically, hydroxiurea in MP-CMML patients, are eagerly
treatment options are limited to hypomethylating awaited.
agents (HMAs), such as azacitidine and decitabine; A major goal of research is the identification of
cytoreductive therapy (hydroxyurea and other biomarkers to predict treatment response after
cytotoxic drugs), as well as supportive care, includ- HMAs. However, up to date the relationship
ing erythropoiesis-stimulating agents (ESAs) and between somatic mutations and response to HMAs
& &
transfusions. in CMML is unclear [20 ,56 ,57]. A large study of 174
Myelodysplastic (MD)-CMML patients are usu- CMML patients has showed that ASXL1 mutations
ally managed as those with MDS. In the only avail- predicted lower ORR to HMAs, whereas TET2mut/
able study specifically analyzing the efficacy of ESAs ASXL1wt genotype predicted a higher complete
in CMML, the erythroid response rate was 64% remission rate and OS [21]. Nonetheless, those
(median duration, 8 months) and RBC transfusion results were not confirmed by another study that,
independence was 31%. The CPSS and EPO levels remarkably, found that patients with at least three
were found adequate tools to select CMML patients somatic mutations had a shorter median response
&
with symptomatic anemia who may benefit from duration [24 ]. Interestingly, it seems that response
treatment with ESAs (low and intermediate-1 risk by to azacitidine has no effect on somatic mutation
the CPSS and low endogenous serum EPO levels) burden and early clonal evolution with expansion of
[47]. subclones has been detected even under treatment
&
The use of HMAs in CMML patients is supported and irrespective of clinical response [19 ]. One study
by several noncomparative clinical trials [48– has found that a specific DNA methylation pattern
&
58,59 ] (Table 5). In these studies, the proportion could predict response to decitabine in CMML [57],
of FAB and WHO subtypes were heterogeneous and but this result requires confirmation. New method-
HMAs were frequently used after prior treatment ologies to determine the intracellular dynamics of
had failed (35–70%). The overall response rate azacitidine therapy, such as multiparameter mass
(ORR) ranged from 38 to 69% with decitabine and spectrometry, are under development, aiming to
from 39% to 54% with azacitidine, but complete understand the mechanisms of azacitidine refracto-
remission rates were significantly more modest. riness [58].
Recently, a large retrospective study compared Intensive chemotherapy is used for fitted
CMML patients treated with azacitidine or decita- patients with extreme myeloproliferative features
bine as first line therapy, with an ORR of 75%, or those progressing to AML, especially as a bridge
including a complete remission of 41% and a to allo-HCT. However, their survival is dismal,
&
median OS of 20 months [55 ]. Patients treated with regardless of response to therapy, and with minimal
&
decitabine showed significantly higher complete benefit for patients undergoing an allo-HCT [59 ].
remission rates (54.8 vs. 23.0%, P < 0.001), although
no statistically significant differences between both
drugs were observed in OS. Yet, another recent study Novel therapies in chronic myelomonocytic
that compared complete remission between azaci- leukemia
tidine and decitabine found no differences [21]. Given the lack of therapeutic options, multiple clini-
Therefore, currently there are not sufficient data cal trials in CMML are ongoing. Some of them are
to support the choice of one HMA over the other. assessing drugs such as rigosertib or guadecitabine as
Cytoreductive therapy with hydroxyurea is con- well as combinations of HMAs with the NEDD8-
sidered the mainstay of therapy for symptomatic activating enzyme inhibitor pevonedistat. Luspater-
MP-CMML patients. Although the specific role of cept and sotatercept, two modified activin receptor
HMAs in MP-CMML is not yet established, it is IIB fusion proteins, are under investigation for ane-
generally assumed that these agents are less active mic patients [60]. Other studies are evaluating the
&
in MP-CMML compared with MD-CMML [56 ]. combination of HMAs with histone deacetylase
However, the ORR reported in three studies with inhibitors (vorinostat or panobinostat [61]) or
azacitidine [48,50,51] for patients with MP-CMML immune checkpoint inhibitors. While RAS inhibitors
(32, 48, and 68%) were not significantly different like tipifarnib or MEK inhibitors such as trametinib
from those observed in the MD-CMML setting could be useful due to the high prevalence of RAS-
(55, 40, and 35%, respectively) (Table 5). A recent pathway mutations [62], other specific molecular

6 www.co-oncology.com Volume 30  Number 00  Month 2018

Copyright © 2018 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
Table 5. Hypomethylating agents in the treatment of chronic myelomonocytic leukemia

Response (myelodys-
plastic/
WHO CMML sub- myeloprolif
CE: Tripti; CCO/300606; Total nos of Pages: 9;

type (%) FAB CMML subtype (%) erative) (%)


Drug No. of Median Prior Median Overall Complete Median
References (schedule) patients age (range) therapy (%) cycles (range) CMML-1 CMML-2 Myelodysplastic Myeloproliferative response remission OS (months)

Costa et al. Azacitidine 38 70.5 (36–83) 56 5 (1–>50) 73 27 30 70 39 (55/32) 11 12


2011 [48] (75D7/100D5)
Fianchi et al. Azacitidine (50D7/ 31 69 (53–84) 35 6 (2–31) 44 56 65 35 51 42a 37
2013 [49] 75D7/F100D5/
F100D7)
Adès et al. Azacitidine 76 70 (33–85) 46 6 (1–40) 55 45 54 46 43 (40/48) 17 29
2013 [50] (75D5/75D7)
Pleyer et al. Azacitidine (75D7/ 48 71 (38–87) 50 5.5 (1–65) 40 60 42 58 54 (35/68) 13 10
CCO 300606

2014 [51] 75D5/ F100D7)


Aribi A et al. Decitabine 19 66 (44–82) 47 9 (1–18) 79 21 68 32 69 58a 19

1040-8746 Copyright ß 2018 Wolters Kluwer Health, Inc. All rights reserved.
2007 [52] (20D5/ 10D10)
Wijermans Decitabine (45D3) 31 71 (53–81) NA 4 (0–6) 58 42 24 76 44 10 15
et al. 2008 [53]
Braun et al. Decitabine (20D5) 39 71 (54–88) 41 10 (1–24) 44 56 18 82 38 10 18.5
2011 [54]
Alfonso et al. Azacitidine 151 69 (50–88) 0 8 (1–71) 39 35 43 57 75 41 24
&
2017 [55 ] Decitabine
Santini et al. Decitabine (20D5) 42 72 (42–84) NA 6 (1–34) 62 38 33 67 48 17 17
&
2018 [56 ]

NA, not available; OS, overall survival; 50D7, azacitidine 50 mg/m2 daily for 7 days; 75D5, azacitidine 75 mg/m2 daily for 5 days; 75D7, azacitidine 75 mg/m2 daily for 7 days; 100D5, azacitidine 100 mg/m2
daily for 5 days; F100D5/F100D7, ‘flat’ azacitidine 100 mg daily for 5 or 7 days, 20D5, decitabine 20 mg/m2 daily intravenous or subcutaneous for 5 days; 10D10, decitabine 10 mg/m2 daily intravenous for 10
days; 45D3, decitabine 15 mg/m2/8 h intravenous for 3 days.
a
Complete remission and marrow complete remission were considered together.

www.co-oncology.com
7
Current management of patients with CMML Mora and Sanz

Copyright © 2018 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; CCO/300606; Total nos of Pages: 9;
CCO 300606

Hematologic malignancies

4. Xicoy B, Triguero A, Such E, et al. The division of chronic myelomonocytic


targets, such as IDH1/2 inhibitors, might be a thera- leukemia (CMML)-1 into CMML-0 and CMML-1 according to 2016 World
peutic option in occasional patients. Finally, other Health Organization (WHO) classification has no impact in outcome in a large
series of patients from the Spanish group of MDS. Leuk Res 2018; 70:34–36.
clinical trials are currently assessing the potential role 5. Khan M, Muzzafar T, Kantarjian H, et al. Association of bone marrow fibrosis
of agents targeting the JAK/STAT pathway or granu- & with inferior survival outcomes in chronic myelomonocytic leukemia. Ann
Hematol 2018; 97:1183–1191.
locyte macrophage colony stimulating factor (GM- CMML with bone marrow fibrosis is associated with inferior survival and a trend for
CSF), such as lenzilumab [63]. Regarding the former, more frequent myeloproliferative features.
6. Gur HD, Loghavi S, Garcia-Manero G, et al. Chronic myelomonocytic leukemia
the preliminary results of a phase I clinical trial with with fibrosis is a distinct disease subset with myeloproliferative features and
the JAK inhibitor ruxolitinib are encouraging, with frequent JAK2 p.V617F mutations. Am J Surg Pathol 2018; 42:799–806.
7. Porwit A, van de Loosdrecht AA, Bettelheim P, et al. Revisiting guidelines for
an ORR up to 35% and showing some benefit for the integration of flow cytometry results in the WHO classification of myelodys-
management of constitutional symptoms or spleno- plastic syndromes-proposal from the International/European LeukemiaNet
Working Group for Flow Cytometry in MDS. Leukemia 2014; 28:1793–1798.
megaly [64]. Therefore, the combination of ruxoliti- 8. Shen Q, Ouyang J, Tang G, et al. Flow cytometry immunophenotypic findings
nib with azacitidine is currently being explored. in chronic myelomonocytic leukemia and its utility in monitoring treatment
response. Eur J Haematol 2015; 95:168–176.
9. Selimoglu-Buet D, Wagner-Ballon O, Saada V, et al. Characteristic repartition
of monocyte subsets as a diagnostic signature of chronic myelomonocytic
CONCLUSION leukemia. Blood 2015; 125:3618–3626.
10. Picot T, Aanei CM, Flandrin Gresta P, et al. Evaluation by flow cytometry of
Treatment of CMML remains largely unsatisfactory mature monocyte subpopulations for the diagnosis and follow-up of chronic
myelomonocytic leukemia. Front Oncol 2018; 8:109.
for most patients. For this reason, the design of 11. Patnaik MM, Timm MM, Vallapureddy R, et al. Flow cytometry based monocyte
CMML-specific clinical trials aiming to find more & subset analysis accurately distinguishes chronic myelomonocytic leukemia
from myeloproliferative neoplasms with associated monocytosis. Blood Cancer
effective agents able to modify the natural history of J 2017; 7:e584.
the disorder are clearly needed. For this purpose, a The assessment of mature monocyte subsets by flow cytometry is useful as part of
the diagnostic workup for CMML patients.
large international cooperative effort will be 12. Onida F, Kantarjian HM, Smith TL, et al. Prognostic factors and scoring
required. A better understanding of the molecular systems in chronic myelomonocytic leukemia: a retrospective analysis of 213
patients. Blood 2002; 99:840–849.
mechanisms involved in the pathogenesis of CMML 13. Such E, Cervera J, Costa D, et al. Cytogenetic risk stratification in chronic
and in driving disease progression will undoubtedly myelomonocytic leukemia. Haematologica 2011; 96:375–383.
14. Tang G, Zhang L, Fu B, et al. Cytogenetic risk stratification of 417 patients
contribute to identifying new therapeutic targets with chronic myelomonocytic leukemia from a single institution. Am J Hematol
and to develop more accurate prognostic models 2014; 89:813–818.
15. Patnaik MM, Vallapureddy R, Yalniz FF, et al. Therapy related-chronic mye-
for better stratifying CMML patients regarding & lomonocytic leukemia (CMML): Molecular, cytogenetic, and clinical distinc-
treatment. tions from de novo CMML. Am J Hematol 2018; 93:65–73.
Therapy-related CMML is associated with a higher frequency of both abnormal
karyotypes and higher-risk chromosomal abnormalities, resulting in shorter OS and
Acknowledgements LFS. Conversely, there were no differences in the distribution of somatic mutations
between both cohorts.
None. 16. Itzykson R, Kosmider O, Renneville A, et al. Prognostic score including gene
mutations in chronic myelomonocytic leukemia. J Clin Oncol 2013;
31:2428–2436.
Financial support and sponsorship 17. Elena C, Gallı̀ A, Such E, et al. Integrating clinical features and genetic lesions
&& in the risk assessment of patients with chronic myelomonocytic leukemia.
This work was partially financed with FEDER funds Blood 2016; 128:1408–1417.
The CPSS-Mol is a new CMML-specific powerful scoring system to assess
(CIBERONC (CB16/12/00284)). prognosis and plan risk-adapted therapy and the only one that integrates cyto-
genetic data as well as mutational data of ASXL1, NRAS, SETBP1, and RUNX1.
18. Patnaik MM, Itzykson R, Lasho TL, et al. ASXL1 and SETBP1 mutations and
Conflicts of interest their prognostic contribution in chronic myelomonocytic leukemia: a two-
center study of 466 patients. Leukemia 2014; 28:2206–2212.
G.F.S. has participated as consultant for Abbvie, Amgen, 19. Than H, Qiao Y, Huang X, et al. Ongoing clonal evolution in chronic
Boehringer Ingelheim, Celgene, Helsinn Healthcare, & myelomonocytic leukemia on hypomethylating agents: a computational per-
spective. Leukemia 2018. [Epub ahead of print]
Janssen, Novartis, and Roche. E.M. has no conflicts of Early clonal evolution with expansion of subclones has been detected in CMML
interest to disclose. patients, even under treatment and irrespective of clinical response to azacitidine
20. Patel BJ, Przychodzen B, Thota S, et al. Genomic determinants of chronic
& myelomonocytic leukemia. Leukemia 2017; 49:2815–2823.
Ancestral mutations that drive the pathogenesis of the disease are frequently
REFERENCES AND RECOMMENDED TET2, SRSF2, and ASXL1 while others, are usually secondary mutations that can
trigger disease progression.
READING 21. Duchmann M, Yalniz FF, Sanna A, et al. Prognostic role of gene mutations in
Papers of particular interest, published within the annual period of review, have chronic myelomonocytic leukemia patients treated with hypomethylating
been highlighted as: agents. EBioMedicine 2018; 31:174–181.
& of special interest 22. Patnaik MM, Vallapureddy R, Lasho TL, et al. EZH2 mutations in chronic
&& of outstanding interest
myelomonocytic leukemia cluster with ASXL1 mutations and their co-occur-
rence is prognostically detrimental. Blood Cancer J 2018; 8:12.
1. Orazi A, Bennett JM, Germing U, et al. Chronic myelomonocytic leukaemia. In: 23. Patnaik MM, Barraco D, Lasho TL, et al. DNMT3A mutations are associated
Swerdlow S, Campo E, Harris NL, editors. International Agency for Research & with inferior overall and leukemia-free survival in chronic myelomonocytic
on Cancer. WHO Classification of Tumours of Haematopoietic and Lymphoid leukemia. Am J Hematol 2017; 92:56–61.
Tissue. Geneva, Switzerland: World Health Organization; 2008. pp. 76–79. The presence of DNMT3A mutations had an adverse impact on OS and LFS.
2. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the World Health 24. Montalban-Bravo G, Takahashi K, Patel K, et al. Impact of the number of
Organization classification of myeloid neoplasms and acute leukemia. Blood & mutations in survival and response outcomes to hypomethylating agents in
2016; 127:2391–2405. patients with myelodysplastic syndromes or myelodysplastic/myeloprolifera-
3. Schuler E, Schroeder M, Neukirchen J, et al. Refined medullary blast and white tive neoplasms. Oncotarget 2018; 9:9714–9727.
blood cell count based classification of chronic myelomonocytic leukemias. A higher number of somatic mutations is associated with poorer outcomes and
Leuk Res 2014; 38:1413–1419. shorter median response duration to HMAs in CMML patients.

8 www.co-oncology.com Volume 30  Number 00  Month 2018

Copyright © 2018 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; CCO/300606; Total nos of Pages: 9;
CCO 300606

Current management of patients with CMML Mora and Sanz

25. Jafari PA, Ayatollahi H, Sadeghi R, et al. Prognostic significance of SRSF2 44. Sorror ML, Storb RF, Sandmaier BM, et al. Comorbidity-age index: a clinical
mutations in myelodysplastic syndromes and chronic myelomonocytic leuke- measure of biologic age before allogeneic hematopoietic cell transplantation.
mia: a meta-analysis. Hematology 2018; 1–7. [Epub ahead of print] J Clin Oncol 2014; 32:3249–3256.
26. Padron E, Garcia-Manero G, Patnaik MM, et al. An international data set for 45. Sanz GF. A lot to learn about allogeneic hematopoietic cell transplantation for
CMML validates prognostic scoring systems and demonstrates a need for chronic myelomonocytic leukemia. Biol Blood Marrow Transplant 2017;
novel prognostication strategies. Blood Cancer J 2015; 5:e333. 23:713–714.
27. Germing U, K€ undgen A, Gattermann N. Risk assessment in chronic myelo- 46. Yoshizato T, Nannya Y, Atsuta Y, et al. Genetic abnormalities in myelodys-
monocytic leukemia (CMML). Leuk Lymphoma 2004; 45:1311–1318. plasia and secondary acute myeloid leukemia: impact on outcome of stem cell
28. Patnaik MM, Padron E, LaBorde RR, et al. Mayo prognostic model for WHO- transplantation. Blood 2017; 129:2347–2358.
defined chronic myelomonocytic leukemia: ASXL1 and spliceosome compo- 47. Xicoy B, Germing U, Jimenez M-J, et al. Response to erythropoietic-stimulat-
nent mutations and outcomes. Leukemia 2013; 27:1504–1510. ing agents in patients with chronic myelomonocytic leukemia. Eur J Haematol
29. Such E, Germing U, Malcovati L, et al. Development and validation of a 2016; 97:33–38.
prognostic scoring system for patients with chronic myelomonocytic leuke- 48. Costa R, Abdulhaq H, Haq B, et al. Activity of azacitidine in chronic myelo-
mia. Blood 2013; 121:3005–3015. monocytic leukemia. Cancer 2010; 117:2690–2696.
30. Wassie EA, Itzykson R, Lasho TL, et al. Molecular and prognostic correlates of 49. Fianchi L, Criscuolo M, Breccia M, et al. High rate of remissions in chronic
cytogenetic abnormalities in chronic myelomonocytic leukemia: a Mayo Clinic- myelomonocytic leukemia treated with 5-azacytidine: results of an Italian
French Consortium Study. Am J Hematol 2014; 89:1111–1115. retrospective study. Leuk Lymphoma 2012; 54:658–661.
31. Kenji M, Shin F, Noriko D, et al. Cytogenetic risk stratification may predict 50. Adès L, Sekeres MA, Wolfromm A, et al. Predictive factors of response and
allogeneic hematopoietic stem cell transplantation outcomes for chronic survival among chronic myelomonocytic leukemia patients treated with aza-
myelomonocytic leukemia. Leuk Lymphoma 2018; 59:1332–1337. citidine. Leuk Res 2013; 37:609–613.
32. Liu HD, Ahn KW, Hu Z-H, et al. Allogeneic hematopoietic cell transplantation 51. Pleyer L, Germing U, Sperr WR, et al. Azacitidine in CMML: matched-pair
&& for adult chronic myelomonocytic leukemia. Biol Blood Marrow Transplant analyses of daily-life patients reveal modest effects on clinical course and
2017; 23:767–775. survival. Leuk Res 2014; 38:475–483.
The CPSS estimated at the time of transplantation strongly and independently 52. Aribi A, Borthakur G, Ravandi F, et al. Activity of decitabine, a hypomethylating
influences OS after allogeneic-HCT. This large CIBMTR study was unable to show agent, in chronic myelomonocytic leukemia. Cancer 2007; 109:713–717.
a clear benefit of pretreatment (intensive chemotherapy or HMAs) on outcomes. 53. Wijermans PW, R€ uter B, Baer MR, et al. Efficacy of decitabine in the treatment
33. Savona MR, Malcovati L, Komrokji R, et al. An international consortium of patients with chronic myelomonocytic leukemia (CMML). Leuk Res 2008;
proposal of uniform response criteria for myelodysplastic/myeloproliferative 32:587–591.
neoplasms (MDS/MPN) in adults. Blood 2015; 125:1857–1865. 54. Braun T, Itzykson R, Renneville A, et al. Molecular predictors of response to
34. Duchmann M, Braun T, Micol J-B, et al. Validation of response assessment decitabine in advanced chronic myelomonocytic leukemia: a phase 2 trial.
according to international consortium for MDS/MPN criteria in chronic Blood 2011; 118:3824–3831.
myelomonocytic leukemia treated with hypomethylating agents. Blood Can- 55. Alfonso A, Montalban-Bravo G, Takahashi K, et al. Natural history of chronic
cer J 2017; 7:e562. & myelomonocytic leukemia treated with hypomethylating agents. Am J Hematol
35. Elliott MA, Tefferi A, Hogan WJ, et al. Allogeneic stem cell transplantation and 2017; 92:599–606.
donor lymphocyte infusions for chronic myelomonocytic leukemia. Bone The largest series to date on the use of HMAs in CMML patients. The median
Marrow Transplant 2006; 37:1003–1008. overall response rate was 75%. Responding patients had a better outcome than
36. Ocheni S, Kröger N, Zabelina T, et al. Outcome of allo-SCT for chronic nonresponders.
myelomonocytic leukemia. Bone Marrow Transplant 2008; 43:659–661. 56. Santini V, Allione B, Zini G, et al. A phase II, multicentre trial of decitabine in
37. Krishnamurthy P, Lim ZY, Nagi W, et al. Allogeneic haematopoietic SCT for & higher-risk chronic myelomonocytic leukemia. Leukemia 2017; 32:413–418.
chronic myelomonocytic leukaemia: a single-centre experience. Bone Marrow Decitabine treatment may be also useful for CMML patients with myeloproliferative
Transplant 2010; 45:1502–1507. features.
38. Eissa H, Gooley TA, Sorror ML, et al. Allogeneic hematopoietic cell trans- 57. Meldi K, Qin T, Buchi F, et al. Specific molecular signatures predict decitabine
plantation for chronic myelomonocytic leukemia: relapse-free survival is response in chronic myelomonocytic leukemia. J Clin Invest 2015; 125:
determined by karyotype and comorbidities. Biol Blood Marrow Transplant 1857–1872.
2011; 17:908–915. 58. Unnikrishnan A, Vo ANQ, Pickford R, et al. AZA-MS: a novel multiparameter
39. Park S, Labopin M, Yakoub-Agha I, et al. Allogeneic stem cell transplantation mass spectrometry method to determine the intracellular dynamics of azaci-
for chronic myelomonocytic leukemia: a report from the Societe Francaise de tidine therapy in vivo. Leukemia 2018; 32:900–910.
Greffe de Moelle et de Therapie Cellulaire. Eur J Haematol 2013; 59. Patnaik MM, Pierola AA, Vallapureddy R, et al. Blast phase chronic myelo-
90:355–364. & monocytic leukemia: Mayo-MDACC collaborative study of 171 cases. Leu-
40. Symeonidis A, van Biezen A, de Wreede L, et al. Achievement of complete kemia 2018. [Epub ahead of print]
remission predicts outcome of allogeneic haematopoietic stem cell trans- Survival after progression to acute leukemia is dismal, regardless of response to
plantation in patients with chronic myelomonocytic leukaemia. A study of the therapy. The benefit of allogeneic-HCT in this setting is very limited.
Chronic Malignancies Working Party of the European Group for Blood and 60. Platzbecker U, Germing U, Götze KS, et al. Luspatercept for the treatment of
Marrow Transplantation. Br J Haematol 2015; 171:239–246. anaemia in patients with lower-risk myelodysplastic syndromes (PACE-MDS):
41. Sharma P, Shinde S, Patnaik M. Outcomes of allogeneic stem cell a multicentre, open-label phase 2 dose-finding study with long-term extension
transplant in patients with chronic myelomonocytic leukemia (CMML) – a study. Lancet Oncol 2017; 18:1338–1347.
single institution experience. Biol Blood Marrow Transplant 2015; 21: 61. Kobayashi Y, Munakata W, Ogura M, et al. Phase I study of panobinostat and
S193–S194. 5-azacitidine in Japanese patients with myelodysplastic syndrome or chronic
42. Kongtim P, Popat U, Jiménez A, et al. Treatment with hypomethylating agents myelomonocytic leukemia. Int J Hematol 2018; 107:83–91.
before allogeneic stem cell transplant improves progression-free survival for 62. Fenaux P, Raza A, Mufti GJ, et al. A multicenter phase 2 study of the
patients with chronic myelomonocytic leukemia. Biol Blood Marrow Trans- farnesyltransferase inhibitor tipifarnib in intermediate- to high-risk myelodys-
plant 2016; 22:47–53. plastic syndrome. Blood 2007; 109:4158–4163.
43. de Witte T, Bowen D, Robin M, et al. Allogeneic hematopoietic stem cell 63. Padron E, Painter JS, Kunigal S, et al. GM-CSF-dependent pSTAT5 sensitivity
&& transplantation for MDS and CMML: recommendations from an international is a feature with therapeutic potential in chronic myelomonocytic leukemia.
expert panel. Blood 2017; 129:1753–1762. Blood 2013; 121:5068–5077.
Recommendations from an international expert panel on the use of allogeneic 64. Padron E, Dezern A, Andrade-Campos M, et al. A multiinstitution phase I
stem cell transplantation in CMML patients. Early transplant is recommended for trial of ruxolitinib in patients with chronic myelomonocytic leukemia (CMML).
patients with intermediate-2 or high-risk score by the CPSS. Clin Cancer Res 2016; 22:3746–3754.

1040-8746 Copyright ß 2018 Wolters Kluwer Health, Inc. All rights reserved. www.co-oncology.com 9

Copyright © 2018 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Вам также может понравиться