Hematological malignancies are an aggregate of diverse populations of cells that arise following a complex process of clonal evolution and selection. Recent approaches have facilitated the study of clonal populations and their evolution over time across multiple phenotypic cell populations. In this review, we present current concepts on the role of clonal evolution in leukemic initiation, disease progression, and relapse. We highlight recent advances and unanswered questions about the contribution of the hematopoietic stem cell population to these processes.

Leukemias, like many tumors, contain diverse populations of cellular clones. This tumor heterogeneity is further shaped by clonal evolution, a process by which accumulated aberrations, both genetic and nongenetic, generate diversity that is subject to selective pressures. The clonal makeup changes during disease pathogenesis, whether it be initial malignant transformation, disease progression, or relapse. Historically, clonal evolution has been thought of as a primarily linear process in which a series of subsequent mutations give rise to novel subclones that drive disease progression.1  Recently, many lines of evidence point to a highly dynamic process of clonal evolution in leukemia that originates long before diagnosis, including at the stem cell level, and a clonal landscape that is continuously reshaped in response to changing selective pressures. Having a greater understanding of clonal evolution and its origins is of particular importance in myelodysplastic syndromes (MDSs) and acute myeloid leukemia (AML), where poor clinical outcomes are common, in large part because of high rates of progression and relapse even after complete remission.2-4  This review provides an overview of emerging concepts of clonal evolution, specifically in the context of myeloid malignancies and highlights recent work proposing premalignant stem cells as a clinically relevant source of clonal heterogeneity.

Over time, competition between hematopoietic stem cells (HSCs) drives clonal diversification of the hematopoietic system. In some cases, a series of transforming events give rise to preleukemic stem cells (pre-LSCs), which precedes the transformation of leukemia stem cells (LSCs) from which the tumor bulk is derived.5,6  Early evidence for this concept was found in murine models of preleukemic-to-overt leukemia transition,7-9  followed by the discovery of transcriptional and other epigenetic changes in phenotypically “normal” stem cells from patients8,10-13  and, later, genomic identification of pre-LSCs in patients with AML6,14,15  and MDS.16 

Myeloid malignancies are often preceded by a premalignant state in which accumulated cellular alterations can exist years prior to the development of symptoms. In some cases, the earliest events arise as germline mutations, as seen in familial leukemias.17  Often, somatic mutations resulting in clonal hematopoiesis (CH) can precede transformation.18-23  Although most cases of CH do not progress to malignancy, recent studies directly linking CH mutations to AML suggest that AML can be descended from long-lived premalignant CH clones and provides additional evidence supporting the stem cell origin of leukemias.23-25 

The effects of clonal selection during initiation can be exemplified by therapy-related MDS and AML. Patients who have undergone previous chemotherapy for unrelated malignancies demonstrate a higher rate of CH, especially in genes related to DNA damage response, such as TP53 and PPM1D.26,27  These mutations suggest that genotoxic stress caused by therapy can lead to the expansion of DNA damage-resistant clones. This is further supported by the enrichment of these mutations in therapy-related MDS and AML compared with de novo MDS/AML, without an increase in the absolute number of mutations.28,29 

Collectively, these findings, combined with the longevity and long-term self-renewal capacity of stem cells,30,31  suggest that the HSC population naturally diversifies over time. Continued accumulation of aberrations leads to the transformation of pre-LSCs and the appearance of a clinically detectable tumor bulk, or blast cells. Although the majority of blast cells usually belong to a limited number of dominant clones, the processes of evolution, competition, and selection during premalignancy and transformation result in a complex and heterogeneous clonal architecture, often including numerous minor subclones. This concept has been reflected in analysis of the clonal makeup of MDS and AML bulk cell populations by single-cell or deep next-generation sequencing, revealing diverse clones with patterns of linear and branching evolution.32-40 

Although mutations provide an easy way to track clones and their evolution, nongenetic factors also play an important role in clonal selection and disease initiation.41-44  For example, transcriptional and other RNA regulatory changes appear to be key events in leukemic transformation, with genetic mutations being only a part of that dysregulation.18,45-51  Additionally, cell-extrinsic mediators can affect how HSCs respond to cell-intrinsic aberrations.52-55  These effectors can include extrinsic selective pressures, such as chemotherapy, as well as a chronic inflammatory milieu and decreased supportive function of an aging bone marrow niche.56,57  A deteriorating hematopoietic microenvironment may promote the preferential expansion of certain mutated clones that would have had no advantage in an unperturbed system. To that end, it is unclear whether CH is necessarily a pathological step on the path to leukemogenesis or simply a result of natural clonal diversification in response to a changing environment. This idea is also supported by the persistence of some common CH mutations after complete remission without an increased risk for relapse.58 

Clonal evolution continues as part of the natural history of the disease, but the evolutionary patterns and role of the stem cell compartment become more difficult to track at disease stages when a large dominant bulk population is present. Cytotoxic and targeted treatments can impose major selective pressures, overpowering cell-intrinsic advantages to direct clonal evolution. Paired MDS and AML sequencing studies have generally shown persistence of the dominant mutations and karyotype in progressive disease, often with the emergence of a newly detected subclone.32-36  For example, treatment with lenalidomide showed successful suppression of the del(5q) clone in MDS; however, in most cases treatment was associated with selection and expansion of a preexisting minor subclone.32,33  This suggests that relapse following treatment may often be a result of clonal selection of an existing clone and not induced mutagenesis. Although, in many cases, the dominant clone after treatment contained most or all mutations found in the dominant clone at diagnosis, in some cases a distinct mutational profile with few common mutations was detected, indicating early divergence from the dominant lineage at diagnosis.34,36  Conceptually similar phenomena have also been observed in myeloproliferative neoplasms.59-62 

In AML, early studies of paired diagnosis-relapse samples demonstrated that most cases of AML with chromosomal abnormalities remained cytogenetically stable, sometimes acquiring additional chromosomal abnormalities.63  However, there were some cases in which relapse clones were cytogenetically distinct. Genomic analysis of paired samples further established that AML relapse can be due to evolution, often linear, of the dominant disease clone. In other cases, AML relapse originated from a related, but distinct, clone.15,37  In either of these cases, the relapse clones have sometimes been detectable in primary disease as a minor clone.38,64,65 

These studies and others suggest that preexisting minor clones may be critical to disease progression, whether they are directly derived from the dominant clone or not. Even in cases in which the minor clones are not detected, they may be present. Classical methodologies of investigating the tumor bulk make it difficult to distinguish whether a newly detected clone at progression is an expansion of an existing leukemic clone below detection limits in primary disease or genuinely due to mutational acquisition since prior sampling. Distinguishing between these important, but not necessarily mutually exclusive, possibilities will have obvious implications for strategies of targeted diagnostics and precision therapeutics to achieve lasting disease control or even prevention of relapse and progression.

The stem cell compartment plays an important role in the pathogenesis of myeloid malignancies, not only as the source of the cancer, but also because LSCs and pre-LSCs persist throughout the course of the disease. LSC burden has been shown to be an excellent predictor of clinical outcomes in AML,66  and LSCs are thought to cause relapse when not fully eradicated.67,68  HSCs are also subject to various cell-extrinsic selective pressures, such as niche effects, inflammation, and immunological control, which can codrive clonal evolution.42,54,69-72  However, their role during disease progression, including in the context of therapeutic interventions, has not been fully appreciated until recently.

The origin of a clone can be traced back by using specific somatically acquired genetic variants as lineage-tracing markers. Cell populations that share the same variants can be assumed to originate from the same founding cell, irrespective of whether the specific variant is assumed to confer altered functionality.73  To determine the origin of secondary disease, Shlush et al,74  Chen et al,16  and Quek et al75  applied this approach to directly investigate the role of stem cell populations in AML relapse, MDS to secondary AML (sAML) progression, and targeted therapy of AML, respectively.

Shlush et al determined the mutational profile of bulk AML samples at diagnosis and relapse in 11 patients, as well as sequencing nonleukemic T cells to distinguish between leukemic and preleukemic mutations. Most patients had a large number of unique relapse variants not detected in the diagnosis sample, consistent with a clonal switch at relapse. To characterize the genetic diversity of the stem cell population, they performed xenotransplantation of the diagnosis sample and assayed the resultant xenografts for the presence of relapse variants. In addition, sorted progenitor populations from the primary sample were assayed when available. Variant allele frequencies were then used to infer the clonal evolution from diagnosis to relapse. This analysis demonstrated that most relapse variants were already present at the time of diagnosis to varying degrees, suggesting that chemotherapy-induced mutations were not the cause of relapse; instead, the stem cells destined to give rise to relapse clones were preexisting and resistant to therapy. Interestingly, relapse variants were never detected in primary T-cell populations or in lymphoid xenograft populations, suggesting LSCs as the most primitive probable source of these clones, and not pre-LSCs, in relapsed AML. Shlush et al identified 2 major patterns of LSC source populations responsible for relapse: relapse origin‐primitive and relapse origin‐committed. In the relapse origin‐primitive group, mutations found only in the relapse blasts, but not in the diagnosis blasts, were detectable in early progenitors, as well as in the diagnosis xenografts. In these patients, the LSCs responsible for relapse are exceedingly rare at diagnosis and seemingly do not contribute to the blasts at that time. However, they are apparently resistant to standard treatment and instead are positively selected and reinitiate the leukemia during relapse. In the relapse origin‐committed group, the relapse clones are more closely related to the major diagnosis clone present and originate from more committed progenitors; however, they still possess a strong stemness signature.

In their study of MDS progression, Chen et al assayed longitudinal paired samples from 7 patients with high-risk MDS who had progressed to sAML to interrogate the clonal architecture of the stem and blast populations. Phenotypically defined and functionally validated pre–MDS-stem cells (pre–MDS-SCs), MDS-SCs, pre-LSCs, LSCs, and blast populations were sorted from bone marrow samples at both time points. The mutational profiles of these samples were determined by targeted deep sequencing, followed by single-cell validation. The stem populations showed greater mutational diversity and an increased number of mutational clusters (ie, clones) than did the blast population. This further demonstrates that the stem population harbors increased heterogeneity that is not appreciated in mature populations.

By tracking the variant allele frequencies in each population, Chen et al were able to infer the clonal evolution that occurs during MDS to sAML progression. Most patients (6/7) showed a demonstrable clonal shift in the blast populations upon progression. However, all patients showed persistence of ≥1 dominant clone after transition, indicating that the bulk of sAML cells likely originated from the same founding population, consistent with previous results. In 4 patients, mutations in a dominant clone that seemed to newly emerge in the AML blasts (ie, was not detectable in the MDS blasts) were detectable in the MDS-SC or pre–MDS-SC population at the MDS time point. In 1 of these cases, the clone was highly represented (49%-52% clonal prevalence) in the MDS stem populations, pointing to possibly complex differential competition and selection mechanisms of stem cell subclones at the MDS and sAML disease stages, respectively, that may not be predicted based on clone size alone. Similar results, of seemingly new sAML mutations being detected in the stem cell compartment at the MDS stage, were reported by Simonsen et al76  in 2 of 3 paired MDS-sAML samples assayed.

In their study, Quek et al detailed the clonal basis of response and acquired resistance to enasidenib. In patients with IDH2-mutant AML, treatment with enasidenib produces a clinical response by promoting differentiation of arrested leukemic cells. By sorting multiple stem and progenitor populations of longitudinal samples, Quek et al tracked clonal changes across different cell populations and in response to treatment. In some cases, the clonal compositions differed across cell populations, especially when comparing more committed cells to early stem and progenitor cells. In most responding patients, the terminally differentiated mature cells arose from the dominant AML clone, retaining the same mutational profile. In other cases, treatment induced differentiation and selection of ancestral or seemingly unmutated clones. Relapse and reimposition of the differentiation block were found to be a result of clonal evolution or selection. In 1 patient with 2 divergent dominant clones (both IDH2 mutant) present at diagnosis, treatment induced a response in only 1 of the clones. At relapse, the nonresponding clone shrunk in most populations and was only found with high prevalence in lymphoid-primed multipotent progenitors. However, accumulation of additional mutations in the persistent nonresponding clone led to selection of that clone and relapse. These findings highlight the complexities of clonal evolution that can take place during AML disease course and targeted therapy and show the importance of studying stem and progenitor populations directly in the clinical context.

These findings point to a model in which some clones diverge from the dominant leukemic lineage very early in disease pathogenesis and including at the level of premalignant stem cell subclones. These coexisting stem cell clones continue to evolve in parallel with the clinically apparent dominant clone and have the capacity to be selected for at a later point (Figure 1).

Figure 1.

Model of HSC clonal evolution leading to MDS and AML pathogenesis and progression. Schematic depiction of a hypothetical evolutionary history in the stem compartment and its representation in bulk samples. Clonal evolution begins with the acquisition of mutations or other nongenetic aberrations that result in clonal expansion and further mutation of the founder clone. A descendent HSC may gain a new aberration, causing it to branch into a new subclone. Branching subclones continue to evolve in parallel and may have disparate responses to selective pressures. Some subclones expand rapidly upon acquisition of a new mutation, whereas others may remain stable for longer periods. Extrinsic pressures, such as targeted therapies, may affect only some clones, whereas nontargeted therapies may have a more widespread, but often also less deep, effect. Clonal outgrowth is represented with time on the x-axis and relative abundance of HSC clones on the y-axis. The colors represent accumulated aberrations. The lower panels show the relative frequency of each alteration found in the bulk blood population at selected time points. Bulk sampling may not reflect the complete diversity and complexity found in HSCs, because variants below the detection limit (dotted line) will not be accounted for but may be shown to be disease relevant at later time points.

Figure 1.

Model of HSC clonal evolution leading to MDS and AML pathogenesis and progression. Schematic depiction of a hypothetical evolutionary history in the stem compartment and its representation in bulk samples. Clonal evolution begins with the acquisition of mutations or other nongenetic aberrations that result in clonal expansion and further mutation of the founder clone. A descendent HSC may gain a new aberration, causing it to branch into a new subclone. Branching subclones continue to evolve in parallel and may have disparate responses to selective pressures. Some subclones expand rapidly upon acquisition of a new mutation, whereas others may remain stable for longer periods. Extrinsic pressures, such as targeted therapies, may affect only some clones, whereas nontargeted therapies may have a more widespread, but often also less deep, effect. Clonal outgrowth is represented with time on the x-axis and relative abundance of HSC clones on the y-axis. The colors represent accumulated aberrations. The lower panels show the relative frequency of each alteration found in the bulk blood population at selected time points. Bulk sampling may not reflect the complete diversity and complexity found in HSCs, because variants below the detection limit (dotted line) will not be accounted for but may be shown to be disease relevant at later time points.

Close modal

With the current ease of DNA sequencing and the implementation of newer methods with increasingly higher cellular resolution, it has become clear that clonal heterogeneity is present in all hematological malignancies,77-80  including at the stem cell level.16,61,62,74,75  The clonal architecture found in the tumor bulk is a result of the continuous accumulation and selection of genetic and nongenetic variants, first in HSCs and later in the cancer cells themselves. These recent findings highlight the stem cell compartment as an additional space where clinically relevant clonal evolution is occurring during the disease process.

The evolutionary pattern observed in a given case after treatment likely depends on the treatment’s efficacy. If the dominant clone persists after treatment, the secondary clone will probably show a linear evolutionary pattern with greater genetic similarity to the primary clone. New targeted therapies, such as IDH and FLT3 inhibitors, or other emerging therapies resulting in broader and deeper responses, such as venetoclax plus azacitidine, will likely be more successful in ablating the initial dominant clone and may result in relapse clones that are more divergent. Ultimately, treatments that target the tumor evolutionary process will likely need to be considered to achieve more effective and lasting disease control. Such approaches could include treatments directly targeting phenotypic pre-LSCs70  or specific vulnerabilities of clones carrying CH mutations81  or treatments affecting mutational acquisition and subclonal diversification.82-84  The targeting of the inflammatory environment that drives selection is also emerging as an interesting new avenue.54,85-90  Furthermore, the direct targeting of transcriptional regulators, previously deemed “undruggable,” is becoming increasingly feasible through chemical and biophysical advances and the design and optimization of novel therapeutic substance classes,82,91-94  some of which have recently advanced to the clinic. Lastly, the ability to detect relevant minor subclones and understand what drives the expansion of a seemingly inactive pre-LSC clone may lead to additional anticipation-based treatment strategies.

However, there is much left unknown, especially at the stem cell level. The degree of heterogeneity is almost certainly still underestimated as a result of technical limitations. There are likely hundreds, and possibly more, stem cell subclones that exist and contribute to the fitness of the HSC pool, and it may be difficult to determine which are clinically relevant. Deep-sequencing or single-cell analysis of multiple longitudinal stem cell and blast samples collected during disease course and treatment could help to determine the variants that contribute to cellular fitness and subsequent expansion and in relation to the specific treatment context. Because of the observed long-lived presence of minor stem cell subclones that do not obtain dominance, despite well-known genetic driver mutations, the mechanisms are unlikely to be solely genetic and may include cell-extrinsic mechanisms. Therefore, comprehensive functional studies, with human (pre)cancer stem cells, but also newly emerging polyclonal (and nonlinear) models of leukemia pathogenesis (eg, in the mouse, zebrafish, or others), are warranted to better understand the factors governing fitness of the heterogeneous stem cell pool and, ultimately, how it can be influenced pharmacologically, including in a preemptive manner.

The authors thank Emily Schwenger and Amit Verma for helpful comments and critical discussion of data and ideas discussed in this article. They apologize to all scientists whose work could not be cited because of space limitations in this spotlight format.

J.S. is supported by National Institutes of Health (NIH)/National Institute of General Medical Sciences grant T32GM007288. U.S. is supported by NIH/National Cancer Institute grant R01CA217092, the Albert Einstein Cancer Center core support grant P30CA013330, and is the Diane and Arthur B. Belfer Faculty Scholar in Cancer Research of the Albert Einstein College of Medicine.

Contribution: J.S., J.M.G., and U.S. wrote the manuscript.

Conflict-of-interest disclosure: U.S. has received research funding from GlaxoSmithKline, Bayer Healthcare, Aileron Therapeutics, and Novartis; has received compensation for consultancy services and for serving on scientific advisory boards from Novartis, GlaxoSmithKline, Bayer Healthcare, Celgene, Aileron Therapeutics, Stelexis Therapeutics, and Pieris Pharmaceuticals; and has equity ownership in and serves on the board of directors of Stelexis Therapeutics. The remaining authors declare no competing financial interests.

Correspondence: Ulrich Steidl, Albert Einstein College of Medicine, 1300 Morris Park Ave, Chanin Bldg, Rm 601-605, The Bronx, NY 10461; e-mail: ulrich.steidl@einsteinmed.org.

1.
Kreso
A
,
Dick
JE
.
Evolution of the cancer stem cell model
.
Cell Stem Cell
.
2014
;
14
(
3
):
275
-
291
.
2.
Pastore
F
,
Dufour
A
,
Benthaus
T
, et al
.
Combined molecular and clinical prognostic index for relapse and survival in cytogenetically normal acute myeloid leukemia
.
J Clin Oncol
.
2014
;
32
(
15
):
1586
-
1594
.
3.
Della Porta
MG
,
Tuechler
H
,
Malcovati
L
, et al
.
Validation of WHO classification-based Prognostic Scoring System (WPSS) for myelodysplastic syndromes and comparison with the revised International Prognostic Scoring System (IPSS-R). A study of the International Working Group for Prognosis in Myelodysplasia (IWG-PM)
.
Leukemia
.
2015
;
29
(
7
):
1502
-
1513
.
4.
Döhner
H
,
Estey
E
,
Grimwade
D
, et al
.
Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel
.
Blood
.
2017
;
129
(
4
):
424
-
447
.
5.
Bonnet
D
,
Dick
JE
.
Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell
.
Nat Med
.
1997
;
3
(
7
):
730
-
737
.
6.
Jan
M
,
Snyder
TM
,
Corces-Zimmerman
MR
, et al
.
Clonal evolution of preleukemic hematopoietic stem cells precedes human acute myeloid leukemia
.
Sci Transl Med
.
2012
;
4
(
149
):
149ra118
.
7.
Kuo
YH
,
Landrette
SF
,
Heilman
SA
, et al
.
Cbf β-SMMHC induces distinct abnormal myeloid progenitors able to develop acute myeloid leukemia
.
Cancer Cell
.
2006
;
9
(
1
):
57
-
68
.
8.
Steidl
U
,
Rosenbauer
F
,
Verhaak
RGW
, et al
.
Essential role of Jun family transcription factors in PU.1 knockdown-induced leukemic stem cells
.
Nat Genet
.
2006
;
38
(
11
):
1269
-
1277
.
9.
Bereshchenko
O
,
Mancini
E
,
Moore
S
, et al
.
Hematopoietic stem cell expansion precedes the generation of committed myeloid leukemia-initiating cells in C/EBPalpha mutant AML
.
Cancer Cell
.
2009
;
16
(
5
):
390
-
400
.
10.
Steidl
U
,
Steidl
C
,
Ebralidze
A
, et al
.
A distal single nucleotide polymorphism alters long-range regulation of the PU.1 gene in acute myeloid leukemia
.
J Clin Invest
.
2007
;
117
(
9
):
2611
-
2620
.
11.
Majeti
R
,
Becker
MW
,
Tian
Q
, et al
.
Dysregulated gene expression networks in human acute myelogenous leukemia stem cells
.
Proc Natl Acad Sci USA
.
2009
;
106
(
9
):
3396
-
3401
.
12.
Mitchell
K
,
Barreyro
L
,
Todorova
TI
, et al
.
IL1RAP potentiates multiple oncogenic signaling pathways in AML
.
J Exp Med
.
2018
;
215
(
6
):
1709
-
1727
.
13.
Will
B
,
Zhou
L
,
Vogler
TO
, et al
.
Stem and progenitor cells in myelodysplastic syndromes show aberrant stage-specific expansion and harbor genetic and epigenetic alterations
.
Blood
.
2012
;
120
(
10
):
2076
-
2086
.
14.
Shlush
LI
,
Zandi
S
,
Mitchell
A
, et al;
HALT Pan-Leukemia Gene Panel Consortium
.
Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia [published correction appears in Nature. 2014;508(7496):420]
.
Nature
.
2014
;
506
(
7488
):
328
-
333
.
15.
Corces-Zimmerman
MR
,
Hong
WJ
,
Weissman
IL
,
Medeiros
BC
,
Majeti
R
.
Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission
.
Proc Natl Acad Sci USA
.
2014
;
111
(
7
):
2548
-
2553
.
16.
Chen
J
,
Kao
YR
,
Sun
D
, et al
.
Myelodysplastic syndrome progression to acute myeloid leukemia at the stem cell level [published correction appears in Nat Med. 2019;25(3):529]
.
Nat Med
.
2019
;
25
(
1
):
103
-
110
.
17.
Godley
LA
,
Shimamura
A
.
Genetic predisposition to hematologic malignancies: management and surveillance
.
Blood
.
2017
;
130
(
4
):
424
-
432
.
18.
Gu
X
,
Ebrahem
Q
,
Mahfouz
RZ
, et al
.
Leukemogenic nucleophosmin mutation disrupts the transcription factor hub that regulates granulomonocytic fates
.
J Clin Invest
.
2018
;
128
(
10
):
4260
-
4279
.
19.
Busque
L
,
Patel
JP
,
Figueroa
ME
, et al
.
Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis
.
Nat Genet
.
2012
;
44
(
11
):
1179
-
1181
.
20.
Genovese
G
,
Kähler
AK
,
Handsaker
RE
, et al
.
Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence
.
N Engl J Med
.
2014
;
371
(
26
):
2477
-
2487
.
21.
Jaiswal
S
,
Fontanillas
P
,
Flannick
J
, et al
.
Age-related clonal hematopoiesis associated with adverse outcomes
.
N Engl J Med
.
2014
;
371
(
26
):
2488
-
2498
.
22.
Bowman
RL
,
Busque
L
,
Levine
RL
.
Clonal hematopoiesis and evolution to hematopoietic malignancies
.
Cell Stem Cell
.
2018
;
22
(
2
):
157
-
170
.
23.
Desai
P
,
Mencia-Trinchant
N
,
Savenkov
O
, et al
.
Somatic mutations precede acute myeloid leukemia years before diagnosis
.
Nat Med
.
2018
;
24
(
7
):
1015
-
1023
.
24.
Abelson
S
,
Collord
G
,
Ng
SWK
, et al
.
Prediction of acute myeloid leukaemia risk in healthy individuals
.
Nature
.
2018
;
559
(
7714
):
400
-
404
.
25.
Luis
TC
,
Wilkinson
AC
,
Beerman
I
,
Jaiswal
S
,
Shlush
LI
.
Biological implications of clonal hematopoiesis
.
Exp Hematol
.
2019
;
77
:
1
-
5
.
26.
Coombs
CC
,
Zehir
A
,
Devlin
SM
, et al
.
Therapy-related clonal hematopoiesis in patients with non-hematologic cancers is common and associated with adverse clinical outcomes
.
Cell Stem Cell
.
2017
;
21
(
3
):
374
-
382.e4
.
27.
Gibson
CJ
,
Lindsley
RC
,
Tchekmedyian
V
, et al
.
Clonal hematopoiesis associated with adverse outcomes after autologous stem-cell transplantation for lymphoma
.
J Clin Oncol
.
2017
;
35
(
14
):
1598
-
1605
.
28.
Wong
TN
,
Ramsingh
G
,
Young
AL
, et al
.
Role of TP53 mutations in the origin and evolution of therapy-related acute myeloid leukaemia
.
Nature
.
2015
;
518
(
7540
):
552
-
555
.
29.
Lindsley
RC
,
Saber
W
,
Mar
BG
, et al
.
Prognostic mutations in myelodysplastic syndrome after stem-cell transplantation
.
N Engl J Med
.
2017
;
376
(
6
):
536
-
547
.
30.
Lee-Six
H
,
Øbro
NF
,
Shepherd
MS
, et al
.
Population dynamics of normal human blood inferred from somatic mutations
.
Nature
.
2018
;
561
(
7724
):
473
-
478
.
31.
Trumpp
A
,
Essers
M
,
Wilson
A
.
Awakening dormant haematopoietic stem cells
.
Nat Rev Immunol
.
2010
;
10
(
3
):
201
-
209
.
32.
da Silva-Coelho
P
,
Kroeze
LI
,
Yoshida
K
, et al
.
Clonal evolution in myelodysplastic syndromes
.
Nat Commun
.
2017
;
8
(
1
):
15099
.
33.
Mossner
M
,
Jann
JC
,
Wittig
J
, et al
.
Mutational hierarchies in myelodysplastic syndromes dynamically adapt and evolve upon therapy response and failure
.
Blood
.
2016
;
128
(
9
):
1246
-
1259
.
34.
Makishima
H
,
Yoshizato
T
,
Yoshida
K
, et al
.
Dynamics of clonal evolution in myelodysplastic syndromes
.
Nat Genet
.
2017
;
49
(
2
):
204
-
212
.
35.
Stosch
JM
,
Heumüller
A
,
Niemöller
C
, et al
.
Gene mutations and clonal architecture in myelodysplastic syndromes and changes upon progression to acute myeloid leukaemia and under treatment
.
Br J Haematol
.
2018
;
182
(
6
):
830
-
842
.
36.
Walter
MJ
,
Shen
D
,
Ding
L
, et al
.
Clonal architecture of secondary acute myeloid leukemia
.
N Engl J Med
.
2012
;
366
(
12
):
1090
-
1098
.
37.
Parkin
B
,
Ouillette
P
,
Li
Y
, et al
.
Clonal evolution and devolution after chemotherapy in adult acute myelogenous leukemia
.
Blood
.
2013
;
121
(
2
):
369
-
377
.
38.
Ding
L
,
Ley
TJ
,
Larson
DE
, et al
.
Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing
.
Nature
.
2012
;
481
(
7382
):
506
-
510
.
39.
Jacoby
MA
,
Duncavage
EJ
,
Chang
GS
, et al
.
Subclones dominate at MDS progression following allogeneic hematopoietic cell transplant
.
JCI Insight
.
2018
;
3
(
5
):
e98962
.
40.
Morita
K
,
Wang
F
,
Jahn
K
, et al
.
Clonal evolution of acute myeloid leukemia revealed by high-throughput single-cell genomics
.
Nat Commun
.
2020
;
11
(
1
):
5327
.
doi:10.1101/2020.02.07.925743
41.
Li
S
,
Garrett-Bakelman
FE
,
Chung
SS
, et al
.
Distinct evolution and dynamics of epigenetic and genetic heterogeneity in acute myeloid leukemia
.
Nat Med
.
2016
;
22
(
7
):
792
-
799
.
42.
Paczulla
AM
,
Rothfelder
K
,
Raffel
S
, et al
.
Absence of NKG2D ligands defines leukaemia stem cells and mediates their immune evasion [published correction appears in Nature. 2019;572(7770):E19]
.
Nature
.
2019
;
572
(
7768
):
254
-
259
.
43.
Bell
CC
,
Fennell
KA
,
Chan
YC
, et al
.
Targeting enhancer switching overcomes non-genetic drug resistance in acute myeloid leukaemia
.
Nat Commun
.
2019
;
10
(
1
):
2723
.
44.
Guo
L
,
Li
J
,
Zeng
H
, et al
.
A combination strategy targeting enhancer plasticity exerts synergistic lethality against BETi-resistant leukemia cells
.
Nat Commun
.
2020
;
11
(
1
):
740
.
45.
Tenen
DG
.
Disruption of differentiation in human cancer: AML shows the way
.
Nat Rev Cancer
.
2003
;
3
(
2
):
89
-
101
.
46.
Will
B
,
Vogler
TO
,
Narayanagari
S
, et al
.
Minimal PU.1 reduction induces a preleukemic state and promotes development of acute myeloid leukemia
.
Nat Med
.
2015
;
21
(
10
):
1172
-
1181
.
47.
Sive
JI
,
Basilico
S
,
Hannah
R
,
Kinston
SJ
,
Calero-Nieto
FJ
,
Göttgens
B
.
Genome-scale definition of the transcriptional programme associated with compromised PU.1 activity in acute myeloid leukaemia
.
Leukemia
.
2016
;
30
(
1
):
14
-
23
.
48.
Jiang
Q
,
Crews
LA
,
Holm
F
,
Jamieson
CHM
.
RNA editing-dependent epitranscriptome diversity in cancer stem cells
.
Nat Rev Cancer
.
2017
;
17
(
6
):
381
-
392
.
49.
Loberg
MA
,
Bell
RK
,
Goodwin
LO
, et al
.
Sequentially inducible mouse models reveal that Npm1 mutation causes malignant transformation of Dnmt3a-mutant clonal hematopoiesis
.
Leukemia
.
2019
;
33
(
7
):
1635
-
1649
.
50.
Thoms
JAI
,
Beck
D
,
Pimanda
JE
.
Transcriptional networks in acute myeloid leukemia
.
Genes Chromosomes Cancer
.
2019
;
58
(
12
):
859
-
874
.
51.
Prieto
C
,
Kharas
MG
.
RNA regulators in leukemia and lymphoma
.
Cold Spring Harb Perspect Med
.
2020
;
10
(
5
):
a034967
.
52.
DeGregori
J
.
Connecting cancer to its causes requires incorporation of effects on tissue microenvironments
.
Cancer Res
.
2017
;
77
(
22
):
6065
-
6068
.
53.
Barreyro
L
,
Chlon
TM
,
Starczynowski
DT
.
Chronic immune response dysregulation in MDS pathogenesis
.
Blood
.
2018
;
132
(
15
):
1553
-
1560
.
54.
Pietras
EM
.
Inflammation: a key regulator of hematopoietic stem cell fate in health and disease
.
Blood
.
2017
;
130
(
15
):
1693
-
1698
.
55.
Verovskaya
EV
,
Dellorusso
PV
,
Passegué
E
.
Losing sense of self and surroundings: hematopoietic stem cell aging and leukemic transformation
.
Trends Mol Med
.
2019
;
25
(
6
):
494
-
515
.
56.
King
KY
,
Huang
Y
,
Nakada
D
,
Goodell
MA
.
Environmental influences on clonal hematopoiesis
.
Exp Hematol
.
2020
;
83
:
66
-
73
.
57.
Cook
EK
,
Luo
M
,
Rauh
MJ
.
Clonal hematopoiesis and inflammation: partners in leukemogenesis and comorbidity
.
Exp Hematol
.
2020
;
83
:
85
-
94
.
58.
Jongen-Lavrencic
M
,
Grob
T
,
Hanekamp
D
, et al
.
Molecular minimal residual disease in acute myeloid leukemia
.
N Engl J Med
.
2018
;
378
(
13
):
1189
-
1199
.
59.
Beer
PA
,
Jones
AV
,
Bench
AJ
, et al
.
Clonal diversity in the myeloproliferative neoplasms: independent origins of genetically distinct clones
.
Br J Haematol
.
2009
;
144
(
6
):
904
-
908
.
60.
Beer
PA
,
Delhommeau
F
,
LeCouédic
JP
, et al
.
Two routes to leukemic transformation after a JAK2 mutation-positive myeloproliferative neoplasm
.
Blood
.
2010
;
115
(
14
):
2891
-
2900
.
61.
Van Egeren
D
,
Escabi
J
,
Nguyen
M
, et al
. Reconstructing the lineage histories and differentiation trajectories of individual cancer cells in JAK2-mutant myeloproliferative neoplasms. bioRxiv.
2020
;.
62.
Williams
N
,
Lee
J
,
Moore
L
, et al
Phylogenetic reconstruction of adult blood cancer reveals early origins and lifelong evolution.
bioRxiv
.
2020
. .
63.
Estey
E
,
Keating
MJ
,
Pierce
S
,
Stass
S
.
Change in karyotype between diagnosis and first relapse in acute myelogenous leukemia
.
Leukemia
.
1995
;
9
(
6
):
972
-
976
.
64.
Krönke
J
,
Bullinger
L
,
Teleanu
V
, et al
.
Clonal evolution in relapsed NPM1-mutated acute myeloid leukemia
.
Blood
.
2013
;
122
(
1
):
100
-
108
.
65.
Garg
M
,
Nagata
Y
,
Kanojia
D
, et al
.
Profiling of somatic mutations in acute myeloid leukemia with FLT3-ITD at diagnosis and relapse
.
Blood
.
2015
;
126
(
22
):
2491
-
2501
.
66.
Ng
SWK
,
Mitchell
A
,
Kennedy
JA
, et al
.
A 17-gene stemness score for rapid determination of risk in acute leukaemia
.
Nature
.
2016
;
540
(
7633
):
433
-
437
.
67.
Pollyea
DA
,
Jordan
CT
.
Therapeutic targeting of acute myeloid leukemia stem cells
.
Blood
.
2017
;
129
(
12
):
1627
-
1635
.
68.
Tehranchi
R
,
Woll
PS
,
Anderson
K
, et al
.
Persistent malignant stem cells in del(5q) myelodysplasia in remission
.
N Engl J Med
.
2010
;
363
(
11
):
1025
-
1037
.
69.
Hoggatt
J
,
Kfoury
Y
,
Scadden
DT
.
Hematopoietic stem cell niche in health and disease
.
Annu Rev Pathol
.
2016
;
11
(
1
):
555
-
581
.
70.
Mitchell
K
,
Steidl
U
.
Targeting immunophenotypic markers on leukemic stem cells: How lessons from current approaches and advances in the leukemia stem cell (LSC) model can inform better strategies for treating acute myeloid leukemia (AML)
.
Cold Spring Harb Perspect Med
.
2020
;
10
(
1
):
a036251
.
71.
Méndez-Ferrer
S
,
Bonnet
D
,
Steensma
DP
, et al
.
Bone marrow niches in haematological malignancies
.
Nat Rev Cancer
.
2020
;
20
(
5
):
285
-
298
.
72.
Yamashita
M
,
Dellorusso
PV
,
Olson
OC
,
Passegué
E
.
Dysregulated haematopoietic stem cell behaviour in myeloid leukaemogenesis
.
Nat Rev Cancer
.
2020
;
20
(
7
):
365
-
382
.
73.
Miller
CA
,
White
BS
,
Dees
ND
, et al
.
SciClone: inferring clonal architecture and tracking the spatial and temporal patterns of tumor evolution
.
PLoS Comput Biol
.
2014
;
10
(
8
):
e1003665
.
doi:10.1371/journal.pcbi.1003665
74.
Shlush
LI
,
Mitchell
A
,
Heisler
L
, et al
.
Tracing the origins of relapse in acute myeloid leukaemia to stem cells
.
Nature
.
2017
;
547
(
7661
):
104
-
108
.
75.
Quek
L
,
David
MD
,
Kennedy
A
, et al
.
Clonal heterogeneity of acute myeloid leukemia treated with the IDH2 inhibitor enasidenib
.
Nat Med
.
2018
;
24
(
8
):
1167
-
1177
.
76.
Simonsen
AT
,
Bill
M
,
Rosenberg
CA
, et al
.
Unraveling clonal heterogeneity at the stem cell level in myelodysplastic syndrome: In pursuit of cell subsets driving disease progression
.
Leuk Res
.
2020
;
92
:
106350
.
77.
Ferrando
AA
,
López-Otín
C
.
Clonal evolution in leukemia
.
Nat Med
.
2017
;
23
(
10
):
1135
-
1145
.
78.
Loh
JW
,
Khiabanian
H
.
Leukemia’s clonal evolution in development, progression, and relapse
.
Curr Stem Cell Rep
.
2019
;
5
(
2
):
73
-
81
.
79.
Miles
LA
,
Bowman
RL
,
Merlinsky
TR
, et al
.
Single-cell mutation analysis of clonal evolution in myeloid malignancies
.
Nature
.
2020
;
587
(
7834
):
477
-
482
.
80.
Potter
N
,
Miraki-Moud
F
,
Ermini
L
, et al
.
Single cell analysis of clonal architecture in acute myeloid leukaemia
.
Leukemia
.
2019
;
33
(
5
):
1113
-
1123
.
81.
Cimmino
L
,
Dolgalev
I
,
Wang
Y
, et al
.
Restoration of TET2 function blocks aberrant self-renewal and leukemia progression
.
Cell
.
2017
;
170
(
6
):
1079
-
1095.e20
.
82.
Carvajal
LA
,
Neriah
DB
,
Senecal
A
, et al
.
Dual inhibition of MDMX and MDM2 as a therapeutic strategy in leukemia
.
Sci Transl Med
.
2018
;
10
(
436
):
eaa3003
.
83.
Konopleva
M
,
Letai
A
.
BCL-2 inhibition in AML: an unexpected bonus?
Blood
.
2018
;
132
(
10
):
1007
-
1012
.
84.
Mullard
A
.
p53 programmes plough on
.
Nat Rev Drug Discov
.
2020
;
19
(
8
):
497
-
500
.
85.
Reynaud
D
,
Pietras
E
,
Barry-Holson
K
, et al
.
IL-6 controls leukemic multipotent progenitor cell fate and contributes to chronic myelogenous leukemia development
.
Cancer Cell
.
2011
;
20
(
5
):
661
-
673
.
86.
Schinke
C
,
Giricz
O
,
Li
W
, et al
.
IL8-CXCR2 pathway inhibition as a therapeutic strategy against MDS and AML stem cells [published correction appears in Blood. 2015;126(3):425]
.
Blood
.
2015
;
125
(
20
):
3144
-
3152
.
87.
Pietras
EM
,
Mirantes-Barbeito
C
,
Fong
S
, et al
.
Chronic interleukin-1 exposure drives haematopoietic stem cells towards precocious myeloid differentiation at the expense of self-renewal
.
Nat Cell Biol
.
2016
;
18
(
6
):
607
-
618
.
88.
Çelik
H
,
Lindblad
KE
,
Popescu
B
, et al
.
Highly multiplexed proteomic assessment of human bone marrow in acute myeloid leukemia
.
Blood Adv
.
2020
;
4
(
2
):
367
-
379
.
89.
Sjövall
D
,
Staffas
A
.
The origin of leukemia: genetic alterations and inflammatory factors in the development of premalignant clonal hematopoiesis
.
Semin Hematol
.
2020
;
57
(
1
):
7
-
12
.
90.
Zhang
TY
,
Dutta
R
,
Benard
B
,
Zhao
F
,
Yin
R
,
Majeti
R
.
IL-6 blockade reverses bone marrow failure induced by human acute myeloid leukemia
.
Sci Transl Med
.
2020
;
12
(
538
):
eaax5104
.
91.
Illendula
A
,
Pulikkan
JA
,
Zong
H
, et al
.
A small-molecule inhibitor of the aberrant transcription factor CBFβBSMMHC delays leukemia in mice
.
Science
.
2015
;
347
(
6223
):
779
-
784
.
doi:10.1126/science.aaa0314
92.
Stephens
DC
,
Kim
HM
,
Kumar
A
,
Farahat
AA
,
Boykin
DW
,
Poon
GM
.
Pharmacologic efficacy of PU.1 inhibition by heterocyclic dications: a mechanistic analysis
.
Nucleic Acids Res
.
2016
;
44
(
9
):
4005
-
4013
.
93.
Tan
YS
,
Lane
DP
,
Verma
CS
.
Stapled peptide design: principles and roles of computation
.
Drug Discov Today
.
2016
;
21
(
10
):
1642
-
1653
.
94.
Antony-Debré
I
,
Paul
A
,
Leite
J
, et al
.
Pharmacological inhibition of the transcription factor PU.1 in leukemia
.
J Clin Invest
.
2017
;
127
(
12
):
4297
-
4313
.
Sign in via your Institution