You are on page 1of 23

The Biology and Pathogenesis

of Chronic Myeloid Leukemia 2


Bradley Chereda and Junia V. Melo

2.1 Clinical Overview Mortality (ACIM) books: Chronic myeloid leu-


kemia 2014; Mendizabal et al. 2013; Quintas-
2.1.1 Diagnosis Cardama and Cortes 2006). The average age at
presentation segregates with geographical loca-
Chronic myeloid leukemia (CML) is usually tion. For example, in Africa and Latin America,
diagnosed in chronic phase (CP) (Quintas- CML patients are diagnosed at least 15 years
Cardama and Cortes 2006). The main symptoms younger compared to Australia (median age 55
and signs at presentation are fatigue, anaemia, years), Europe and the USA (Australian Cancer
splenomegaly, abdominal discomfort and epi- Incidence and Mortality (ACIM) books: Chronic
sodes of infections (Quintas-Cardama and Cortes myeloid leukemia 2014; Mendizabal et al. 2013;
2006). However, a significant proportion of Quintas-Cardama and Cortes 2006). Differences
patients are asymptomatic, with diagnosis occur- in life expectancy do not entirely explain the age
ring after unrelated medical examination of onset; thus, future investigation could identify
(Quintas-Cardama and Cortes 2006). Males have additional determinants of CML. Currently, the
an increased incidence of CML at a ratio of 1.3– only proven risk factor is exposure to high-dose
1.5 to 1.0 (Australian Cancer Incidence and ionising radiation (Corso et al. 1995). The BCR-
ABL1 gene is observed in all cases of CML, and
PCR-based detection of this gene, together with
Parts of this chapter have formerly been published within karyotyping to identify the Ph chromosome, is
the journal Annals of Hematology in Volume 94, Issue 2, used to confirm the diagnosis if a complete blood
supplement, April 2015, “Chronic Myeloid Leukemia”, count reports the number of granulocytes to be
doi:10.1007/s00277-015-2325-z. abnormally high.
B. Chereda (*)
Department of Genetics and Molecular Pathology,
Centre for Cancer Biology, SA Pathology, 2.1.2 CML Evolution and Prognosis
Frome Road, Adelaide 5000, Australia
e-mail: bradley.chereda@outlook.com
Without therapeutic intervention, the disease will
J.V. Melo progress from CP (generally after 3–5 years) to
Department of Haematology, Centre for Cancer
Biology, University of Adelaide, Adelaide 5000, blast crisis (BC), often via an accelerated phase
Australia (AP). Disease progression is defined by the blast
Department of Haematology, Imperial College cell count in the peripheral blood as 10–20 % in
London, London, UK AP and >20 % in BC. The BC phenotype can be

© Springer International Publishing Switzerland 2016 17


R. Hehlmann (ed.), Chronic Myeloid Leukemia, Hematologic Malignancies,
DOI 10.1007/978-3-319-33198-0_2
18 B. Chereda and J.V. Melo

myeloid or lymphoid or, in rare cases, both, greater, a 4-log reduction in BCR-ABL (%IS))
although myeloid BC is predominantly observed achieved in some patients have allowed clinicians
(on a 2:1 ratio) compared with lymphoid BC to consider therapy cessation – a new goal for the
(Kantarjian et al. 1987). Aggressive haematolog- treatment of CML (Mahon et al. 2010).
ical symptoms occur in BC, including infection, Despite successful advances in CML treat-
thrombosis or anaemia – a consequence of bone ment, imatinib resistance is observed in approxi-
marrow failure due to the lack of cell differentia- mately 25 % of patients (Milojkovic and
tion and massive infiltration with immature blasts Apperley 2009). The most common known
(Ilaria 2005). As a result, the patient’s health rap- resistance mechanism is mutations in the BCR-
idly deteriorates in advanced disease, which ABL1 protein, which are observed in 25–30 % of
nearly invariably leads to mortality within 1 year early CP and 70–80 % of BC patients (Soverini
of progression (Hehlmann 2012). et al. 2011). Current strategies to circumvent
Leukaemic cells in advanced disease lose the suboptimal response include the use of more
ability to undergo terminal differentiation, result- potent BCR-ABL1 TKIs, such as nilotinib,
ing in an expansion of primitive cells rather than dasatinib and bosutinib (Weisberg et al. 2007).
mature granulocytes. The exact mechanism for In addition, a newly developed TKI, ponatinib,
disease progression is unknown. However, muta- has showed promising activity against a BCR-
tions in genes other than BCR-ABL1 are com- ABL1 mutant (T315I) which is totally resistant
monly detected following BC transformation to other TKIs (Soverini et al. 2015). Another
(Melo and Barnes 2007b), which suggests that a therapeutic shortcoming is the lack of efficient
second hit is important for the transformation options in BC-CML. The outcome for patients in
into acute leukemia. advanced disease is still almost unchanged as
compared to the natural course of disease. Stem
cell transplantation provides the best option for
2.1.3 Treatment patients on AP/BC. However, not all patients are
eligible to undergo transplantation, and long-
Introduction of interferon-α therapy and stem term remission rates still remain poor (Hehlmann
cell transplantation marked the first era when sur- 2012).
vival and quality of life noticeably improved for
a large proportion of patients (Kantarjian et al.
2012; Quintas-Cardama and Cortes 2006). 2.2 The Molecular Biology
During the peak of interferon treatment, the of CML
median survival doubled to 6 years (Kantarjian
et al. 2012). Previously, cytotoxic agents (e.g. 2.2.1 The t(9;22) Translocation
arsenic, radiotherapy, busulfan and hydroxyurea) and the BCR-ABL1 Gene
were primarily used to treat the symptoms of
CML, but did not alter the course of the disease. The Ph chromosome is formed by a reciprocal
The recent development of tyrosine kinase inhib- t(9;22)(q34;q11) translocation between the long
itors (TKIs) has greatly improved patient out- arms of chromosomes 9 and 22, causing the jux-
come by inhibiting the constitutive kinase activity taposition of the BCR (breakpoint cluster region)
of BCR-ABL1. The TKI imatinib is currently the and ABL1 (Abelson) genes. The BCR-ABL1
first-line therapy for CML. Its selective inhibition fusion gene consists of the 5′ end of the BCR
of BCR-ABL1’s kinase activity significantly gene and the 3′ end of the ABL1 gene (Fig. 2.1a).
reduces the frequency of progression to BC and The location of the BCR and ABL1 genomic
eliminates the symptoms of CP (Druker et al. breakpoints is highly variable, but the recombi-
2006). TKI treatment has led to overall survival nation usually involves fusion of intron 13 or 14
rates of >80 % after 8 years (Deininger et al. of BCR with a 140 kilobase (kb) region of ABL1
2009). Moreover, deep responses (MR4 or surrounding exons 1b and 2 (Fig. 2.1a) (Score
2 The Biology and Pathogenesis of Chronic Myeloid Leukemia 19

a BCR gene There has been much debate regarding the


(22q11) exon 13 14 15 consequence of a patient expressing either the
e13a2 or e14a2 transcripts (Melo 1996). Before
the TKI era, most reports on large series refuted
ABL1 gene the importance of the BCR breakpoint (Dowding
(9q34) 140 kb et al. 1991; Fioretos et al. 1993; Rozman et al.
1995; Shepherd et al. 1995). However, a recent
exon 1b 1a 2
revival of this debate has found consistent evi-
dence that patients with either the e14a2 tran-
BCR-ABL1 transcripts script or both the e14 and e13a2 transcripts
e13a2
exhibit a higher platelet count approximately 1.5
times higher than that in the e13a2 group
e14a2 (Balatzenko et al. 2011; Hanfstein et al. 2014;
Jain et al. 2015). Furthermore, transcript type has
b BCR-ABL1 protein structure
also been shown to impact on the response to TKI
therapy. Several laboratories have found that
C-C DNA Actin
domain patients with the e14a2 transcript exhibit a faster
PxxP binding binding
Y177 Y rate of achieving a deeper response (both MMR,
i.e., a 3-log reduction, and MR4, the therapy ces-
SH3
SH2
SH1

sation target). In contrast to the relationship to


NES

NLS platelet count, the TKI response in patients with


Ser/Thr Rho/GEF
both transcripts tracks with the e13a2 group (Jain
Fig. 2.1 The gene and protein structure of BCR-ABL1. et al. 2015; Bonifacio et al. 2015; Hanfstein et al.
(a) The BCR-ABL1 fusion gene consists of the 5′ end of 2014).
the BCR gene and the 3′end of ABL1. The location of the Interestingly, the BCR-ABL1 transcript can
translocation usually involves fusion of intron 13 or 14 of
BCR with a 140 kilobase (kb) region of ABL1 surround- also be detected, by specially sensitive PCR
ing exons 1b and 2. Examples of the two BCR-ABL1 methods, in healthy individuals without CML
major mRNA isoforms are shown to highlight the BCR symptoms (Bose et al. 1998; Biernaux et al.
breakpoint variants. Depending on the breakpoint on the 1995). It is hypothesised that in these cases the
ABL1 gene, exons 1a and 1b may be included in the pri-
mary transcript but are always excluded from the mRNA translocation occurs in a haemoatpoietic cell or
because they lack a splice acceptor sequence. (b) The environment that is unable to support leukemia
BCR-ABL1 protein contains the dimerisation or coiled transformation. The BCR-ABL1 signal from this
coil (C-C), the Ser/Thr kinase and the Rho/GEF domains phenomenon is very low and thus does not pose a
of BCR, as well as the SH domain; proline-rich (PxxP),
nuclear localisation signal (NLS); DNA-binding, nuclear concern for diagnostic laboratories (Bose et al.
export signal (NES) and actin-binding domains from 1998).
ABL. The tyrosine residues in the Ser/Thr and SH1 kinase
domains have been highlighted with a Y. The diagrams in
a and b are not to scale
2.2.2 Protein Structure

et al. 2010; Melo 1996). Regardless of the break- The 210 kDa BCR-ABL1 protein observed in
point location on the ABL1 gene, mRNA splicing CML contains more than ten protein domains
gives rise to major BCR-ABL1 transcripts with (Fig. 2.1b). The SH1 tyrosine kinase region is the
e13a2 (BCR exon 13 and ABL1 exon 2) or e14a2 most studied BCR-ABL1 domain due to its
junctions. These transcripts were originally inherent role in CML pathogenesis and the func-
referred to as b2a2 and b3a2, respectively. Both tional domain targeted by TKIs. However, other
transcripts result in the expression of a 210 kDa features such as tyrosine 177 in the BCR Ser/Thr
BCR-ABL1 protein, with a 75-amino-acid kinase domain (Chu et al. 2007; Hantschel 2012;
difference. Pendergast et al. 1993; Zhang et al. 2001) and the
20 B. Chereda and J.V. Melo

coil-coil protein dimerisation domain also influ- 1990; Kelliher et al. 1990). Additional work
ence the function of BCR-ABL1 (Zhao et al. established BCR-ABL1’s ability to transform
2002). cells, cause growth factor-independent cell
Although BCR-ABL1 contains the majority growth and block apoptosis (Bedi et al. 1994;
of the ABL1 gene, it lacks an amino acid from Daley and Baltimore 1988; Hariharan et al.
the ABL1 N-terminal region that is myris- 1988). The first studies to specifically target
toylated. For the endogenous ABL1 protein, BCR-ABL1 by antisense oligonucleotides
myristoylation of this residue and subsequent cis (Ratajczak et al. 1992; Skorski et al. 1991;
binding within ABL1’s myristoylation binding Szczylik et al. 1991) and disruption of BCR-
pocket causes autoinhibition of the SH1 kinase ABL1 kinase activity (Engelman and Rosenberg
activity (Hantschel 2012). Thus, it is thought that 1990b) showed that BCR-ABL1 was essential
the loss of this moiety is, in part, responsible for for maintenance of leukemia. These initial
the pathogenic constitutive kinase activity of observations underpinned the function of BCR-
BCR-ABL1. Since BCR-ABL1 retains the ABL1 and affirmed this gene as the driver of
myristoylation binding pocket, compounds tar- CP-CML. Since expression of the BCR-ABL1
geting this motif have been trialled to inhibit its coding sequence in an HSC is sufficient to gen-
kinase activity. These compounds exhibit prom- erate CML-like disease, it is generally accepted
ising allosteric inhibition of BCR-ABL1 activity that BCR-ABL1 is the sole lesion required for
and may enhance the capabilities of therapeutic CP-CML, and it is unlikely that an additional
targeting of BCR-ABL1 (Zhang et al. 2010; event is required (however, this has not been
Wylie et al. 2014). formally ruled out). As will be discussed herein,
BCR-ABL1 has remarkable properties that can
control almost every cellular event to function
2.2.3 The Consequence in its favour for promoting CP-CML.
of BCR-ABL1

The BCR-ABL1 protein gives rise to aberrant 2.3 Important Pathways


activation of cell signalling pathways and a shift Affected by BCR-ABL1
to a cellular environment that supports leukemia. Activity
For example, CML cells exhibit changes in
growth factor dependence, apoptosis, prolifera- After understanding the phenotypic changes
tion and cell adhesion. These changes result in caused by the formation of BCR-ABL1, the focus
excessive proliferation of granulocytes that in of research shifted to identifying BCR-ABL1’s
turn cause the clinical features observed in targets. Initial studies found that JAK/STAT,
CP-CML (Daley et al. 1990; Deininger et al. PI3K/AKT and Ras/MEK signalling proteins are
2000). The importance of BCR-ABL1 signalling at the forefront of pathogenic signalling via
(particularly via the tyrosine kinase domain) is BCR-ABL1 (Goldman and Melo 2003). A ‘next
demonstrated by the success of TKI therapy. wave’ of knowledge for CML biology was accu-
Furthermore, the ability to silence BCR-ABL1 mulated from research that focused on specific
activity in the laboratory has also given scientists effectors known to have oncogenic roles (e.g.
the capacity to further dissect the biology of p53, MYC, β-catenin, C/EBPα). With the advent
CML. of next-generation (next-gen) sequencing,
Early CML models focused on BCR-ABL1’s another ‘wave’ of research is expected to charac-
primary mechanism of disease. Reconstitution terise genes that are mutated and/or their expres-
of mouse bone marrow with BCR-ABL1 sion altered in CML patients. The next sections
expressing haemopoietic stem cells (HSCs) summarise well-studied CML pathways and
caused affected mice to display a CML-like introduce examples of exciting and novel CML
phenotype (Daley et al. 1990; Heisterkamp et al. research.
2 The Biology and Pathogenesis of Chronic Myeloid Leukemia 21

2.3.1 JAK/STAT and inhibition of cell death (Zhao et al. 2006).


AKT is a downstream effector of PI3K and plays
Signalling from the JAK/STAT pathway is com- a major role in its signalling (Zhao et al. 2006).
monly exploited to promote pathogenesis in leu- BCR-ABL1 can stimulate PI3K signalling
kemia (Lin et al. 2000). STAT proteins are through the adaptor proteins Grb2/Gab2 (Sattler
transcription factors commonly activated by the et al. 1996) and CBL (Sattler et al. 2002) and
JAK cell receptor (Hennighausen and Robinson PTEN inhibition (Morotti et al. 2014). One of the
2008). CML models demonstrated that BCR- first reports revealing PI3K’s role in CML was
ABL1 kinase activity directly enhances JAK2/ the observation that PI3K was required for BCR-
STAT activation to promote cell growth/survival ABL1-mediated transformation of haemopoietic
(Chai et al. 1997; Warsch et al. 2013). Two piv- cells (Skorski et al. 1997). Subsequent work
otal studies showed that STAT5 plays a crucial found that PI3K/AKT is also important for CML
role in development and maintenance of CML. A maintenance, and inhibition of PI3K signalling
mouse model was established which showed that can circumvent BCR-ABL1 oncogenesis and kill
expression of BCR-ABL1 in STAT5 knockout primary CML cells (Klejman et al. 2002a).
bone marrow cells failed to induce CML in recip- Another consequence of PI3K activation is stim-
ient mice after both primary and secondary trans- ulation of the mTOR pathway (Mayerhofer et al.
plantation (Walz et al. 2012). In another model, 2002), which is responsible for controlling pro-
BCR-ABL1-transformed bone marrow cells tein synthesis, cell growth/size and autophagy.
were allowed to cause initial disease in mice and Autophagy involvement in CML is a new area
then STAT5 was deleted, which caused a remark- of interest. Autophagy can occur following cell
able loss of BCR-ABL1-expressing cells and stress (i.e. loss of BCR-ABL1 signalling) to send
restored healthy haematological parameters the cell into hibernation rather than apoptosis and
(Hoelbl et al. 2010). The importance of STAT5 can be reversed when the environment becomes
not only centres on its requirement for CML dis- favourable again. Recent studies have observed
ease but also as a therapeutic target because that whilst BCR-ABL1 inhibits autophagy, TKI
STAT5 is dispensable for (adult) haemopoiesis. treatment restores this pathway and may allow
Interestingly, whilst JAK2 is upstream of for protection of leukemia cells and resistance to
STAT5, it was found that JAK2 was not essential therapy (Sheng et al. 2011). Co-inhibition of
for myeloid (but was required for lymphoid) leu- autophagy and BCR-ABL1 considerably
kemia in BCR-ABL1 mouse models (Hantschel enhances eradication of primitive CML cells
et al. 2012). It was proposed that BCR-ABL1 compared to TKI alone (Salomoni and Calabretta
may directly activate STAT5 (Hantschel et al. 2009). Therefore, this approach appears to be a
2012) and bypass endogenous regulation by promising method to counter the unwanted TKI-
JAK2 to promote leukaemogenesis (Hansen et al. mediated inhibition of autophagy. Current work
2013; Schafranek et al. 2014). Nevertheless, JAK is focused on discovering the genes responsible
inhibitors exhibit efficacy against primary CML for autophagy control in CML.
cells, including TKI-resistant cells (Samanta
et al. 2009), and recent work suggests that JAK
signalling is important for stem cell biology (dis- 2.3.3 Ras/MEK Pathway
cussed later).
Activation of Ras GTPases/MEK kinases stimu-
lates cell growth via membrane receptor-binding
2.3.2 PI3K/AKT and Autophagy cascade to activate transcription of a number of
growth factor genes and is a key pathway deregu-
PI3K proteins communicate extracellular signals lated in cancer (Steelman et al. 2011). BCR-ABL1
to modulate transcription factor activation and activates Ras via Grb2/Gab2 phosphorylation to
programming that favour cell growth/survival promote cell growth (Chu et al. 2007; Puil et al.
22 B. Chereda and J.V. Melo

1994). Disruption of Ras signalling impairs devel- 2.3.5 Crkl


opment of BCR-ABL1-induced CML-like dis-
ease in mice (Baum and Ren 2008; Sattler et al. The adaptor protein Crkl is constitutively acti-
2002). In addition, small-molecule inhibitors vated by BCR-ABL1 (ten Hoeve et al. 1994).
against MEK can target primitive CML cells Protein networks involving BCR-ABL1 and Crkl
(Packer et al. 2011; Pellicano et al. 2011). include Cbl, STAT, PI3K, paxillin and Ras (Birge
However, there is limited knowledge of how the et al. 2009). Indeed, loss of the interaction
Ras-effector repertoire contributes to disease and between Ckrl and BCR-ABL1 impaired BCR-
which effectors in particular are important. One ABL1-induced transformation in mice (Seo et al.
exception is NF-κB, which is a transcription fac- 2010). The potent phosphorylation of Crkl by
tor activated by BCR-ABL1/Ras (Reuther et al. BCR-ABL1 allows the measurement of the per-
1998) and required for BCR-ABL1-induced CML centage of phospho-Crkl as a surrogate to BCR-
(Hsieh and Van Etten 2014). ABL1 phosphorylation levels (which are more
difficult to measure) in order to experimentally
examine patient response to TKI therapy and to
2.3.4 Src Kinases predict outcome (White et al. 2005).

The Src family kinases (SFKs) are another group


of widely studied downstream targets of BCR- 2.3.6 Long Noncoding (lnc)
ABL1. Their role is to coordinate cell growth, RNA-BGL3
differentiation and motility in response to extra-
cellular signals (Kim et al. 2009). Initial CML The discovery of microRNAs resulted in exten-
cell line models showed that BCR-ABL1 expres- sive expression profiling and functional analysis
sion significantly activated the Hck and Lyn in the context of CML. These studies found that
SFKs (Danhauser-Riedl et al. 1996). Subsequent microRNA regulation of gene networks is likely
studies demonstrated that Hck, Lyn and Fyn were to control aspects of CML pathogenesis and
required for BCR-ABL1 cell line transformation, treatment response. Similarly, lncRNA research
as well as functionally phosphorylating several has led to the first description of their involve-
BCR-ABL1 tyrosines (Lionberger et al. 2000; ment in CML biology.
Wilson et al. 2002). One mechanism by which The general mechanism by which lncRNAs
SFKs contribute to disease is in assisting BCR- function is not yet fully understood, but lncRNA-
ABL1 in its activation of STAT5 and AKT BGL3 was reported to play an important role in
(Klejman et al. 2002b; Warmuth et al. 2003). In BCR-ABL1 transformation. In K562 and pri-
addition, knockdown of Lyn exhibited impressive mary CML cells, BCR-ABL1 inhibits the expres-
killing of BC cells and its upregulation in sion of this lncRNA in a kinase-dependent
BC-CML suggested a potential role for promot- manner via the MYC transcription factor (Guo
ing disease progression (Ban et al. 2008; Ptasznik et al. 2014b). Forced expression of lncRNA-
et al. 2004). However, the importance of SFKs in BGL3 in K562 cells induced apoptosis and
CML remains unclear because CML mouse mod- reduced the ability of these cells to engraft in
els show that SFKs are not required for initiation mice. It was subsequently found that this lncRNA
of CML and instead support the generation of acted as a decoy for several microRNAs that tar-
acute lymphoid leukemia (Engelman and get the tumour suppressor gene PTEN, leading to
Rosenberg 1990a; Hu et al. 2004). Second- its stabilisation and associated inhibition of leu-
generation TKIs, dasatinib and bosutinib, are kaemogenesis. In addition, the same group also
dual Src/Abl1 inhibitors, so defining the role of reported that another lncRNA, lncRNA-H19,
SFKs in CML could have an impact on both biol- may also have an involvement in CML biology.
ogy and treatment (Rusconi et al. 2014; Lindauer This lncRNA is positively regulated by BCR-
and Hochhaus 2014). ABL1/MYC, and its knockdown perturbs the
2 The Biology and Pathogenesis of Chronic Myeloid Leukemia 23

pathogenicity of BCR-ABL1 in CML cell lines 2.4 CML Stem Cells


(Guo et al. 2014a). Further work is required to
understand the full mechanisms and impact of It is well established that quiescent leukaemic
these lncRNAs in CML. stem cells (LSCs) within the CD34+ population
are resistant to TKIs (Bhatia et al. 2003; Copland
et al. 2006; Jorgensen et al. 2007). This phenom-
2.3.7 Apoptosis Deregulation enon is believed to be responsible for relapse in
approximately half of all patients eligible for
In addition to promoting cell proliferation, BCR- therapy cessation (Mahon et al. 2010). As a
ABL1 can disrupt cell death. An example of this result, CML stem cells have been thrust into the
involves a BCR-ABL1, Bad, BCL2 and BCL-XL limelight. Prior to this, research was focused on
circuit (Fig. 2.2). Expression of BCR-ABL1 can characterising the differences between normal
inhibit apoptosis by increasing expression of the HSCs and LSCs. One goal was to understand
anti-apoptotic proteins BCL2 and BCL-XL exactly how BCR-ABL1 altered normal haemo-
(Salomoni et al. 2000). Both STAT5 and PI3K poiesis to drive CP-CML, leading to the identifi-
signalling are important mediators of BCR- cation of several haemopoietic markers and
ABL1’s anti-apoptotic function. STAT5 activa- oncogenes that differed between the two popula-
tion by BCR-ABL1 causes increased BCL-XL tions (reviewed in Kabarowski and Witte 2000).
expression (de Groot et al. 2000; Horita et al. The improvement of strategies to isolate primi-
2000). Furthermore, phosphorylation of the pro- tive cells increased accessibility to this very rare
apoptotic protein Bad by PI3K/AKT facilitates (less than 2 % of PB-MNCs) cell population
the interaction between the chaperone protein (Holyoake et al. 1999; Silvestri et al. 1992).
14-3-3 and Bad, which restricts Bad to the cyto- These early studies also acknowledged the
plasm (Neshat et al. 2000). This prevents Bad importance of LSCs in the quest for a cure in
opposing BCL2 and BCL-XL inhibition of apop- CML (Eaves et al. 1993), which became a more
tosis in the mitochondrion. viable possibility after the availability of potent
TKIs. Interestingly, there is a significant overlap
between genes involved in LSC biology and BC
development (e.g., p53, MYC, β-catenin), thus
T5 PI3
STA BCR-ABL1
K linking the two key unsolved areas of CML
K research.
PI3
P
BCL2 Bad Bad
2.4.1 LSCs Are Refractory to TKIs
BCL-XL
A seminal paper from the Holyoake laboratory
showed that kinase inhibition reduced LSC pro-
BCL-XL
liferation, but did not kill quiescent LSCs
Bad
BCL2 (Graham et al. 2002). Further work from the
same group demonstrated that LSCs were also
ndrion
Mitocho insensitive to more potent second-generation
TKIs, even though the BCR-ABL1 kinase activ-
Anti-apoptotic cell environment ity was silenced (Copland et al. 2006; Jorgensen
et al. 2007). These studies warned of the possibil-
Fig. 2.2 An example of apoptotic circuitry controlled by ity of early relapse, but long-term TKI usage has
BCR-ABL1. BCR-ABL1 promotes both the expressions
quelled these concerns. Subsequent studies have
of anti-apoptotic genes BCL2 and BCL-XL and inhibits
the function of pro-apoptotic protein Bad via phosphory- strengthened the notion that LSCs do not rely on
lation (grey circle) and cytoplasmic sequestration BCR-ABL1 kinase activity for survival
24 B. Chereda and J.V. Melo

(Chakraborty et al. 2012; Corbin et al. 2011). to its receptor Frizzled, β-catenin is protected
They showed that potent TKIs failed to eliminate from ubiquitin-mediated degradation and is free
CML-LSCs, that the bone marrow environment to translocate to the nucleus and activate its target
may offer sanctuary against TKIs and that with- genes (Moon et al. 2004). β-Catenin tyrosine
drawal of TKIs leads to reconstitution of leukae- phosphorylation by BCR-ABL1 also leads to its
mic expansion (Chakraborty et al. 2012; Corbin stabilisation and increased levels and activity in
et al. 2011). It was recently reported that therapy- CML (Coluccia et al. 2007). Although dispens-
refractory LSCs exhibit a bias for low BCR- able for maintenance of LSCs and HSCs (Cobas
ABL1 expression (Chomel et al. 2012; Kumari et al. 2004; Heidel et al. 2012; Koch et al. 2008),
et al. 2012). Several lines of evidence discussed dual targeting of β-catenin and BCR-ABL1 can
herein show that LSCs benefit from tempered synergise to delay disease onset and deplete
signalling from BCR-ABL1. Therefore, it is pos- CML-LSCs in CML mouse models (Heidel et al.
sible that LSCs may not require BCR-ABL1 for 2012). The β-catenin pathway has also been
survival, and/or rely on non-kinase activity of implicated in BC-CML. Enhanced β-catenin sig-
BCR-ABL1, and/or prefer moderate kinase activ- nalling in BC-CML is thought to confer stem
ity. Several pathways have been shown to play cell-like properties to progenitor cells leading to
key roles in stem cell biology (Fig. 2.3), and tar- their expansion – a feature of advanced disease
geting them could lead to a promising strategy to (Jamieson et al. 2004). The control of β-catenin
eliminate the LSC in CML. in CML is complex and is involved in many of
the pathways discussed in the next sections
(Fig. 2.4).
2.4.2 β-Catenin

β-Catenin signalling is important for develop- 2.4.3 Smo


ment and self-renewal of both HSCs and LSCs
(Zhao et al. 2007). β-Catenin is a component of The Smoothened (Smo)/hedgehog pathway gov-
the Wnt signalling pathway. When Wnt is bound erns developmental and homeostasis decisions
conserved from Drosophila to humans. Smo is a
membrane receptor for the hedgehog ligand.
BCR-ABL1
Analogous to Wnt/β-catenin, activation of Smo, in
PTEN turn, activates Gli transcription factors, which drive
RAS
BCL2 JAK2 PI3K TGF-β STAT5
expression of their downstream transcriptional tar-
gets (Briscoe and Therond 2013). Whilst there is
hnRNP-K β-cat
FOXO MSI2 debate surrounding the role of this pathway in nor-
MYC mal HSC development, there is a consensus that it
BCL6
SIRT1 is important for CML-LSCs (Mar et al. 2011). Smo
Fbw7
signalling is enhanced in CML compared to nor-
LSC turnover P53 LSC maintenance mal HSCs, and both the loss and inhibition of Smo
impair the development and maintenance of BCR-
Fig. 2.3 LSC circuitry of genes discussed in this chapter. ABL1-induced CML in mice (Zhao et al. 2009).
STAT5, JAK2 and PI3K all feature to control LSC effector Aberrant hedgehog signalling has been recently
genes. However, quiescent stem cells have intrinsic coun-
reported to drive BC-CML. Studies on primary
ters to prevent potent BCR-ABL1 signalling depleting the
LSC population, such as MSI2/TGF-β, PTEN, FOXO CML cells found that Smo/Gli2 promoted LSC
transcription factors and Fbw7. In the context of BC-CML, dormancy via cell cycle arrest, and an enhanced
the reliance on countering BCR-ABL1 is not as important hedgehog pathway signature is observed in BC
due to the incapacity of leukaemic progenitor cells to dif-
patients. Inhibition of Gli2 was able to restore LSC
ferentiate. This may explain how enhanced pathway acti-
vation (JAK2/β-catenin) is compatible with expansion of cycling and sensitise LSCs to TKI eradication
the stem/progenitor compartment in BC (Sadarangani et al. 2015). The availability of Smo
2 The Biology and Pathogenesis of Chronic Myeloid Leukemia 25

BCR-ABL1
Alo
x5 LSC survival (Neviani et al. 2013). At the centre
is PP2A, a tyrosine phosphatase whose activity is
impaired in CML. ‘Active’ PP2A has the ability
JAK2
PI3K to silence key pathways that are activated by
IRF8
BCR-ABL1, including BCR-ABL1 itself
SET (Neviani et al. 2005). In CML-LSCs, BCR-
ABL1/JAK2 signalling overcomes PP2A activity
GAS2
PP2A b-catenin by enhancing the activity of SET, a PP2A inhibi-
tor. Blocking the PP2A inhibitory role of SET
restores PP2A function and impairs the self-
GSK-3β β-cat Transcription
renewal and survival of CML-LSCs, but not nor-
U U mal HSCs (Neviani et al. 2013). A major
mechanism by which PP2A activation affects
b-catenin U
LSC maintenance is thought to be the loss of
Transcriptional
program for a stem-
β-catenin signalling via GSK-3β-mediated ubiq-
Β-catenin degradation cell-like environment uitination. This is coupled with PP2A silencing
of BCR-ABL1 to allow for LSC turnover and
Fig. 2.4 Complex control of β-catenin in CML. BCR- reduced leukaemic potential.
ABL1 stabilises β-catenin signalling via PI3K and JAK2
and inhibition of IRF8. Canonical stability of β-catenin is
controlled by protein ubiquitination (grey circles). Thus in
CML, this pathway is activated to promote a stem cell-like
2.4.5 FoxO
environment. However, inhibition of, for example, PP2A
activation can reverse pathogenic β-catenin signalling and The FoxO transcription factors, in particular
synergise with BCR-ABL1 inhibition to enhance treat- FoxO3a, have also been linked to LSC biology.
ment efficacy
BCR-ABL1 promotes nuclear export and deacti-
vation of these transcription factors via PI3K/AKT
inhibitors and their synergy with TKIs has allowed (Atfi et al. 2005). In mature cells, AKT signalling
for clinical trials to determine the efficacy of Smo/ is strong and assists propagation of BCR-ABL1’s
TKI therapy (Mar et al. 2011). The largest of these proliferative advantage. However, in LSCs, AKT
trials investigated dasatinib in combination with signalling is inhibited by PTEN (Hurtz et al. 2011)
the Smo inhibitor (BMS-833923) for treatment of and TGF-β (Naka et al. 2010). This reverses BCR-
patients in advanced disease and who had subopti- ABL1 inactivation of FoxO3a and allows for
mal response to another TKI. This trial found no BCL6 transcription, which favours quiescence and
combinatorial effect for Smo inhibition on patient self-renewal (Hurtz et al. 2011). Targeting this
outcome (Shah et al. 2014). Reporting the results of mechanism with BCL6 or TGF-β inhibitors
other studies probing dual inhibitory effects on effi- together with TKIs perturbed CML development
cacy (including as a first-line treatment) should fur- and induced cell death/turnover of primitive CML
ther clarify the potential of Smo inhibition in CML. cells (Hurtz et al. 2011; Naka et al. 2010). These
studies also provide evidence that potent BCR-
ABL1 signalling is detrimental to LSCs.
2.4.4 PP2A-JAK2-SET

BCR-ABL1 was reported to circumvent the 2.4.6 Alox5 and Alox15


requirement of JAK2 for activation of STAT5
(Hantschel et al. 2012), but a recent study has Alox5 was identified as an important gene for
implicated a role for JAK2 within the LSC com- LSCs by an innovative screen that looked for
partment. A network involving PP2A/JAK2/ BCR-ABL1-induced genes in LSCs which did
SET/GSK-3β was shown to play a critical role in not rely on BCR-ABL1 kinase activity (Chen
26 B. Chereda and J.V. Melo

et al. 2009). Other genes validated from the faster accumulation of mutations (compared to
experiment were CD121b, Asprv1, Hdc, MS4A2, LSCs) required for transformation.
MaoB, Mctp2 and Tph1 (which are either stress
or immune-related genes). Functional experi-
ments found that loss or inhibition of Alox5 2.5 Biology of Blast Crisis
impaired BCR-ABL1-induced myeloid, but not
lymphoid, leukemia in mice and did not affect It is currently unknown exactly how the transition
normal haemopoiesis. Alox5 supports both PI3K to BC occurs. This stage of the disease is charac-
and β-catenin signalling in stem cells by inhibi- terised by the expansion of haemopoietic progen-
tion of the Msr1 gene, and this, in turn, drives itors that can no longer differentiate and can
LSC self-renewal (Chen et al. 2011). The same invade the peripheral blood. These progenitor
group also investigated the function of Alox15 in cells gain self-renewal capacity, differentiation
CML-LSCs. Loss of this gene prevented BCR- arrest and survival properties that lead to their
ABL1-induced CML in mice, and administration uncontrolled proliferation (Jamieson et al. 2004).
of an Alox15 inhibitor prolonged survival of pri- Thus, BC progenitors exhibit more stem cell-like
mary and secondary BCR-ABL1 LSC recipient characteristics compared to CP progenitors. This
mice. A key downstream target of Alox15, is partially attributed to increased β-catenin activ-
P-selectin was also identified. Increased ity, which is also thought to provide BC progeni-
P-selectin expression was observed upon Alox15 tors with the capacity to initiate leukemia in mice
deletion, and loss of both Alox15 and P-selectin (Jamieson et al. 2004). Genomic and genetic
rescued BCR-ABL1’s ability to induce CML in instability is another feature of advanced disease
mice (Chen et al. 2014). (Perrotti et al. 2010; Skorski 2012). Extra-
chromosomal abnormalities are observed in
approximately 80 % of BC patients (e.g. Ph
2.4.7 Bone Marrow duplication, trisomy 8 or 19, loss of 17p)
Microenvironment (Johansson et al. 2002). Pathogenic alterations of
tumour suppressor and oncogenes have also been
HSCs reside in the bone marrow, which provides detected in advanced CML (Melo and Barnes
an environment that controls haemopoiesis by 2007a). Thus, it is hypothesised that these addi-
coordinating HSC renewal and differentiation tional hits are responsible in part for the transi-
into functional blood cells. The bone marrow tion into BC (Melo and Barnes 2007a; Skorski
supportive environment comprises the osteoblast 2012). The changes in cell biology in BC may
and vascular niches (Ellis and Nilsson 2012; Ema explain why TKIs have diminished efficacy in
and Suda 2012). The former promotes self- BC, reflecting reduced reliance on BCR-ABL1
renewal and quiescence, whilst the vascular niche activity in the presence of other mutations, and/or
is permissive of differentiation into progenitor stem cell-like progenitors becoming refractive to
and then functional cells. Furthermore, signalling TKIs similar to CP-LSCs.
molecules and membrane receptors are also vital
for legitimate haemopoiesis. In CML, it is
thought that the osteoblast niche nurtures LSCs 2.5.1 BCR-ABL1 and BC-CML
and may explain why LSCs do not require BCR-
ABL1 kinase activity to survive TKI exposure Inhibition of BCR-ABL1 kinase activity effec-
(Hazlehurst et al. 2007; Zhang et al. 2013). This tively delays the onset of BC, but does not elimi-
may also contribute to BC. Since progenitor cells nate the primitive population that establishes
attain stem cell-like properties (discussed later), a advanced disease. One interpretation is that
progenitor contingent may retreat towards the BCR-ABL1 signalling is required for transition
osteoblast niche for protection against TKIs to BC, especially since progression to BC is rare
whilst retaining cycling properties that allow for in TKI-responsive patients. A number of studies
2 The Biology and Pathogenesis of Chronic Myeloid Leukemia 27

have found increased expression of BCR- 2006). Further experiments revealed that BCR-
ABL1 in BC compared to CP. This was observed ABL1 negatively regulates the expression of C/
when comparing matched CP and BC samples EBPα via upregulation of hnRNP-E2, an RNA-
(from the same patient) at both the mRNA binding protein which inhibits C/EBPα expres-
(Barnes et al. 2005; Gaiger et al. 1995; Jiang sion (Chang et al. 2007). Interestingly, analysis
et al. 2007; Marega et al. 2010) and protein levels of CML patient cells found that loss of C/EBPα
(Andrews and Collins 1987; Barnes et al. 2005; and expression of hnRNP-E2 were restricted to
Neviani et al. 2005). Additionally, it has been BC (Chang et al. 2007). In addition, hnRNP-E2
shown that cells expressing higher amounts of upregulation and C/EBPα downregulation were
BCR-ABL1 have an increase in genomic insta- directly proportional to increasing levels of BCR-
bility as well as perturbed differentiation, which ABL1 (Chang et al. 2007). To add extra complex-
are intrinsic properties of BC-CML (Chang et al. ity to this pathway, it was recently shown that the
2007; Skorski 2012). These findings imply more microRNA miR-328 acts in a non-canonical way
than a passenger role for BCR-ABL1 in BC to block hnRNP-E2 regulation of C/EBPα and
transformation, but this has yet to be determined. promotes myeloid differentiation (Eiring et al.
The next sections outline direct involvement of 2010). The expression of miR-328 negatively
BCR-ABL1 in two key features of BC: genetic correlates with BCR-ABL1 expression levels and
instability and differentiation arrest. is thus downregulated in BC (Eiring et al. 2010).
These experiments provide evidence of a sophis-
2.5.1.1 DNA Damage/Repair ticated circuit by which enhanced BCR-ABL1
BCR-ABL1 has been shown to facilitate genomic expression can facilitate a switch to BC by dis-
instability via disrupting DNA repair pathways, rupting myeloid differentiation.
generating reactive oxygen species and inhibiting
DNA damage-induced apoptosis, which may 2.5.1.3 IRF-8
lead to retention of genomic mutations (Amos Remarkably, genomic deletion of interferon reg-
et al. 1995; Bedi et al. 1995; Dierov et al. 2009; ulatory factor 8 (IRF-8) (also known as ICSBP)
Koptyra et al. 2008; Slupianek et al. 2013). These in mice was sufficient to generate a CML-like
events are in part tied to the level of BCR-ABL1 myeloproliferative disease (Holtschke et al.
expression (Deutsch et al. 2001). CML CD34+ 1996). The mice developed splenomegaly and
cells express high levels of BCR-ABL1 as com- WBC counts consistent with those of CML
pared to mature cells (Jiang et al. 2007), and they patients, and one-third of them succumbed to a
are highly susceptible to genomic instability as BC-like pathology. Conversely, over-expression
compared to their healthy counterparts of IRF-8 produces the opposite effect – induc-
(Chakraborty et al. 2012). Although not formally tion of apoptosis in myeloid cell lines (Gabriele
shown, it is reasonable to suggest that BCR- et al. 1999). Expression of this gene is com-
ABL1 provides progenitor cells with the genomic monly reduced in CML patients CML patients
plasticity required for malignant transformation (Schmidt et al. 1998) and a new study found that
(Skorski 2007, 2008, 2012). this occurs via the BCR-ABL1/STAT5 signal-
ling axis (Waight et al. 2014). A mouse model
2.5.1.2 C/EBPα and hnRNP-E2 co-expressing BCR-ABL1 and IRF-8 demon-
Required for myeloid differentiation (Zhang strated IRF-8’s tumour suppressor role in vivo.
et al. 2004), C/EBPα expression is reduced in cell Mice transplanted with BCR-ABL1/IRF-8 cells
lines expressing BCR-ABL1 (Guerzoni et al. survived much longer than those with BCR-
2006). These lines responded poorly to growth ABL1 alone, but the former showed increased
factor-induced differentiation (Chang et al. incidence of lymphoid leukemia (Burchert et al.
2007), but ectopic expression of C/EBPα and 2004; Hao and Ren 2000). IRF-8’s reversal of
BCR-ABL1 kinase inhibition were able to BCR-ABL1’s anti-apoptotic effects partially
reverse this differentiation block (Guerzoni et al. explained IRF-8’s suppressor role (Gabriele
28 B. Chereda and J.V. Melo

et al. 1999; Tamura et al. 2003). As interferon els show that BCR-ABL1 activation of BCL2 can
activates IRF-8 expression (Schmidt et al. 1998), inhibit MYC apoptotic activity whilst retaining
IRF-8 may underpin the mechanism behind its proliferative advantage (Sanchez-Garcia and
interferon treatment efficacy in CML. Grutz 1995). This is one of many examples by
Recent work has identified a putative role for which BCR-ABL1 creates ‘a perfect storm’ to
IRF-8 in CML progenitor cells and disease pro- promote leukaemogenesis.
gression via β-catenin disruption. In the absence BCR-ABL1 can control MYC expression via
of BCR-ABL1, IRF-8 destabilises the β-catenin PI3K, JAK2 and the transcription factor E2F1
protein via the GAS2 protease to promote normal (Birchenall-Roberts et al. 1997; Skorski et al.
haemopoiesis (Huang et al. 2010; Scheller et al. 1997; Stewart et al. 1995; Xie et al. 2002) and
2013). As discussed previously, β -catenin is dis- protein stability via MEK and hnRNP-K (Notari
pensable for CML maintenance and aberrant et al. 2006). A recent CML mouse model demon-
β -catenin activity cannot cause CML by itself. strated that MYC expression is required for CML
Actually, it is possible that enhanced β -catenin maintenance and progression. They further
signalling is toxic to HSCs (Scheller et al. 2013). showed that high levels of MYC are harmful for
However, transgenic mice that have both high LSCs, and ubiquitination (degradation) of MYC
β -catenin activity and deletion of the IRF-8 gene by ubiquitin ligase Fbw7 keeps MYC levels in
rapidly develop acute leukemia (Scheller et al. check in LSCs (Reavie et al. 2013). This provides
2013). A similar situation is achieved by BCR- a rationale for the constrained BCR-ABL1 kinase
ABL1 in CML. It can downregulate IRF-8, whilst activity observed in quiescent LSCs (Neviani
enhancing β -catenin signalling. Based on the et al. 2013) and selection of low BCR-ABL1
Scheller et al. study, this will create a cellular expression in TKI-refractive LSCs (Chomel et al.
environment that is permissive of disease pro- 2012; Kumari et al. 2012) (suggesting that
gression. BCR-ABL1’s effect on IRF-8/β -catenin enhanced BCR-ABL1 signalling is toxic for qui-
is more subtle than the genomic deletions of the escent cells). These findings, coupled with
aforementioned IRF-8/β -catenin mouse model MYC’s established role in myeloid differentia-
and highlights the requirement of additional tion (Delgado and Leon 2010), present deregula-
event(s) for acute leukemia transformation. tion of MYC as a strong candidate for BC
transformation in CML.

2.5.2 Examples of Important 2.5.2.2 p53


Pathways Involved in BC-CML The normal function of p53 is to respond to cell
stress events, where it becomes activated and
2.5.2.1 MYC drives transcription of genes that decide cell fate
The MYC proto-oncogene was one of the first (apoptosis, DNA repair, cell cycle arrest or senes-
genes implicated in CML disease progression. cence) (Pant et al. 2012). Early CML genetic stud-
MYC is a transcription factor which governs the ies observed inactivating mutations of p53 in
expression of genes enabling cell growth and approximately 20 % of patients who progressed to
proliferation and, thus, commonly activated in BC-CML (Guinn and Mills 1997; Stuppia et al.
cancer (Dang 2012). It was originally observed 1997). Regulation of p53 by BCR-ABL1 is com-
that patients with BC exhibited higher levels of plex and unclear, with p53 activation (Sionov
MYC as compared to CP patients (Preisler et al. et al. 1999; Stoklosa et al. 2004) and inactivation
1988). This was followed by reports that ABL1 (Trotta et al. 2003; Wendel et al. 2006) being
expression enhances MYC expression and that reported. However, loss or inhibition of p53 pro-
MYC is required for BCR-ABL1-induced trans- motes BC-like disease in mice (Honda et al. 2000;
formation (Cleveland et al. 1989; Sawyers et al. Velasco-Hernandez et al. 2013; Wendel et al.
1992). Although excess MYC can induce apopto- 2006), and stabilisation of p53 in BC cells induces
sis (Bissonnette et al. 1992), early cell line mod- apoptosis (Velasco-Hernandez et al. 2013;
2 The Biology and Pathogenesis of Chronic Myeloid Leukemia 29

Peterson et al. 2011). It has also been shown that whereas short-term inhibition shut down STAT5,
MYC overexpression is only toxic to LSCs if p53 AKT and MEK signalling prior to affecting
is present (Reavie et al. 2013). With the knowl- BCR-ABL1 activity (Walker et al. 2013). This
edge that MYC activity is enhanced in BC, this suggests that both BCR-ABL1-dependent and
may explain the high frequency (20 %) of p53 BCR-ABL1-independent cell deaths result
mutations observed in BC (Stuppia et al. 1997). through XPO1 inhibition. Remarkably, an XPO1
inhibitor reversed CML symptoms (WBC count/
2.5.2.3 Musashi-2 (Msi2) splenomegaly) in a patient who was resistant to
Msi2 is an RNA-binding protein (Nakamura et al. TKI therapy and had progressed to AP-CML –
1994), which has been recently linked to HSC highlighting an exciting strategy to treat advanced
development (de Andres-Aguayo et al. 2011). disease (Walker et al. 2013).
Two groups have also reported the involvement of
Msi2 in promoting BC-like disease (Ito et al. 2.5.2.5 SIRT1
2010; Kharas et al. 2010). These studies found the Expression of SIRT1 is enhanced in CML and is,
RNA-binding protein’s expression is markedly in part, regulated by BCR-ABL1/STAT5 (Yuan
elevated in BC compared to CP. In addition, CML et al. 2012). This protein deacetylase has been
mouse models demonstrated that enhanced Msi2 linked to BC-CML due to its disruption of LSC
expression promoted aggressive leukemia via turnover and DNA repair. SIRT1 suppression of
impaired myeloid differentiation and progenitor p53/FoxO-controlled LSC maintenance is
expansion (Ito et al. 2010; Kharas et al. 2010). It believed to prolong the survival of CML-LSCs
was initially thought that high Msi2 expression (Li et al. 2012; Yuan et al. 2012). In contrast,
inhibited the Msi2’s downstream target NUMB, knockout or inhibition of SIRT1 impairs CML
leading to disruption of cell differentiation (Ito development and disease progression in mice by
et al. 2010; Kharas et al. 2010). However, a new reducing proliferative and self-renewal capacity
study identified direct mRNA targets of Msi2 and of LSCs (Li et al. 2012; Yuan et al. 2012). SIRT1
discovered an interactome consisting of genes regulation of the DNA repair protein Ku70 in
associated with self-renewal. Surprisingly, this CML cell lines causes enhancement of less faith-
study found no connection between Msi2 and ful non-homologous end joining to enhance DNA
NUMB and, instead, proposed that Msi2 cooper- mutations (Wang et al. 2013). The knowledge
ates with TGF-β to propagate self-renewal signals that SIRT1 provides a route for LSC survival and
important for promoting advanced disease (Park genomic instability – the key drivers of
et al. 2014). BC-CML – provides strong evidence that SIRT1
has a major role in BC development.
2.5.2.4 XPO1
The nuclear export protein, XPO1, is another 2.5.2.6 ADAR1
novel candidate for regulation of BC. Its expres- ADAR1 is an RNA editor whose enzymatic
sion is enhanced in BC patients, and pharmaco- activity converts adenosine to inosine in RNA,
logical blockade of its function was shown as resulting in these nucleotides being interpreted as
sufficient to kill both CP and BC primary CD34+ guanine in the ribosome, thus altering RNA
cells (Walker et al. 2013). Inhibition of XPO1 in behaviour and protein amino acid composition.
BCR-ABL1-positive cell lines demonstrated that Analysis of ADAR1 expression in CML patients
impaired nuclear transport could explain XPO1 showed a marked increase in expression from CP
inhibition lethality. For example, both SET and to BC and was correlated with BCR-ABL1 levels
p53 were abnormally enriched in the nucleus (Jiang et al. 2013).
leading to their inactivation (Walker et al. 2013). The BC samples also had enhanced A to I edit-
Additional experiments revealed that long-term ing and altered expression of RNA-edited genes,
XPO1 inhibition caused BCR-ABL1 degradation providing evidence that the increased expression
(via loss of SET control of PP2A activity), of ADAR1 in BC had a functional effect on its
30 B. Chereda and J.V. Melo

downstream targets (Jiang et al. 2013). Two 2.6 Concluding Remarks


mouse models have been developed which suc-
cessfully demonstrate the important role that The biology of CML is centred on BCR-ABL1’s
ADAR1 plays in CML stem cells. Following dis- constitutive kinase activity, which is sufficient to
ruption of ADAR1 expression in CML mouse cause the clinical features of CP. The ability to
models, CML development, maintenance and BC readily model CML in both cell lines and mice
onset were all impaired due to the loss of primi- has allowed for a large accumulation of knowl-
tive leukaemic cells (Steinman et al. 2013). In edge regarding the molecular network of
contrast, ADAR1 overexpression caused myeloid CML. These studies have shown that BCR-ABL1
progenitor expansion (Jiang et al. 2013). is implicated in altering almost every process
Moreover, specific deletion of ADAR1’s RNA- within the cell to drive CML pathogenesis. This
editing moiety demonstrated that RNA editing is extends to dampening its own excessive signal-
vital for CML progenitor self-renewal (Steinman ling in LSCs, which would be otherwise unfa-
et al. 2013). It is known that the RNA-editing vourable. Current literature has shown that
activity of ADAR1 is required for HSC survival STAT5 stands out as a vital component of BCR-
(XuFeng et al. 2009), so it is speculated that the ABL1’s induction of CML as demonstrated by
enhanced activity of ADAR1 in BC locks the two conditional knockout models (Hoelbl et al.
LSCs in a primitive state. 2010; Walz et al. 2012). The recent investigation
of primitive CML cell biology has benefitted
2.5.2.7 Screening for Novel BC Driver from the utilisation of new and powerful tech-
Genes niques to identify a number of important genes
In order to identify putative BC driver genes, within this compartment. The best studied are
Giotopoulos et al. utilised an impressive mouse p53, MYC and β-catenin, which have prominent
model. The experiment centred on a transposable roles in both stem cell biology and BC
cassette array in the presence or absence of BCR- transformation.
ABL1. Transposition of the cassette(s) can either The link between LSCs and BC and treatment
activate or deactivate the gene(s) in proximity to the response has put the LSC and progenitor popula-
genomic insertion site. Gene activation is achieved tions at the forefront of CML biology. Of particu-
by a transposition event within the 5′ region of the lar interest is the finding that LSCs do not rely on
gene due to enhancer/promoter sequences in the BCR-ABL1 kinase activity for survival. It is
cassette. Conversely, intragenic transposition can unknown if another protein domain of BCR-
disrupt genes causing loss of function. ABL1 confers LSC survival properties. Another
Mice with a BCR-ABL1-only genetic back- possibility is that BCR-ABL1 can programme
ground succumbed to a CP-CML phenotype, LSCs in such a way that its kinase activity is no
whilst 85 % of the BCR-ABL1/transposon mice longer required. Primitive CML cells also con-
exhibited BC-CML, 5 % CP and 10 % ‘AP’-like tain the answer to the mechanisms of disease pro-
disease. Microarray gene expression analysis of gression. It is unknown whether the HSC or
the mice showed clustering within disease type progenitor compartment gives rise to the clone(s)
and inter-type separation. These data identified responsible for BC-CML. Pinpointing the cell(s)
several genes that correctly segregated with dis- responsible is important because each of these
ease type from a known role in BC development. compartments has discrete biological properties
Next-generation sequencing was performed to and thus requires alternate therapeutic strategies.
identify transposon integration sites mirroring the It remains to be seen if BC and LSCs are the
clustering patterns of the gene expression dataset. last bastions for understanding CML biology and
There were 78 gene transposition events within the how big of an impact epigenetics will contribute
BC sample cohort, some of which have been dis- to CML pathogenesis. Technical advances such
cussed herein, e.g. STAT5, XPO4 (herein: XPO1), as next-generation sequencing and powerful
PTEN, MYC-target genes and JAK1 (JAK2). experimental modelling tools will no doubt pro-
2 The Biology and Pathogenesis of Chronic Myeloid Leukemia 31

vide a flood of information regarding CML biol- mechanisms behind CML pathogenesis and the
ogy. Rapid and accurate sequencing of whole potential for application to other diseases.
genomes, exomes and epigenomes is becoming
increasingly accessible to most laboratories. It is Conflict of Interest The authors declare that they have
expected that next-generation long-read sequenc- no conflict of interest for the writing of this manuscript.
ing will answer questions related to RNA splice
isoforms and complex genomic regions. These
advances should generate evidence of recurrent References
mutations and epigenetic marks that favour or
Agirre X, Jimenez-Velasco A, San Jose-Eneriz E, Garate
hinder CML pathogenesis or response to treat-
L, Bandres E, Cordeu L, Aparicio O, Saez B, Navarro
ment. One example is a polymorphism in the Bim G, Vilas-Zornoza A, Perez-Roger I, Garcia-Foncillas
gene, which perturbs apoptosis induced by ima- J, Torres A, Heiniger A, Calasanz MJ, Fortes P,
tinib to impair TKI efficacy (Ng et al. 2012). This Roman-Gomez J, Prosper F (2008) Down-regulation
of hsa-miR-10a in chronic myeloid leukemia CD34+
mutation was uncovered by next-generation
cells increases USF2-mediated cell growth. Mol
sequencing and examined in mouse models using Cancer Res 6(12):1830–1840
new gene editing techniques. Amos TA, Lewis JL, Grand FH, Gooding RP, Goldman
In the proteomics field, improved methods to JM, Gordon MY (1995) Apoptosis in chronic myeloid
leukaemia: normal responses by progenitor cells to
study proteins (SILAC (Kapoor et al. 2012)) and
growth factor deprivation, X-irradiation and glucocor-
more powerful mass spectrometers have the ticoids. Br J Haematol 91(2):387–393
potential to uncover post-translational modifica- Andrews DF 3rd, Collins SJ (1987) Heterogeneity in
tions and protein interactomes. The study of pro- expression of the bcr-abl fusion transcript in CML
blast crisis. Leukemia 1(10):718–724
teome networks is relatively untapped in CML
Atfi A, Abecassis L, Bourgeade MF (2005) Bcr-Abl acti-
(although elegant examples do exist (Halbach vates the AKT/Fox O3 signalling pathway to restrict
et al. 2013; Winter et al. 2012)), making this an transforming growth factor-beta-mediated cytostatic
attractive area of interest to improve the knowl- signals. EMBO Rep 6(10):985–991
Australian Institute of Health and Welfare (2014).
edge of CML biology. The same can be said of
Australian Cancer Incidence and Mortality books:
noncoding RNA (ncRNA) involvement in Chronic Myeloid Leukaemia. Canberra: AIHW. http://
CML. It is known that ncRNA deregulation www.aihw.gov.au/acim-books
occurs in CML, for example, in CP vs. BC, and in Balatzenko G, Vundinti BR, Margarita G (2011)
Correlation between the type of bcr-abl transcripts and
primitive cells vs. granulocytes (Agirre et al.
blood cell counts in chronic myeloid leukemia – a pos-
2008; Machova Polakova et al. 2011). However, sible influence of mdr1 gene expression. Hematol Rep
most functional work is limited to a single 3(1):e3
microRNA and target. Further work is required to Ban K, Gao Y, Amin HM, Howard A, Miller C, Lin Q,
Leng X, Munsell M, Bar-Eli M, Arlinghaus RB,
understand the global ncRNA circuitry in key
Chandra J (2008) BCR-ABL1 mediates up-regulation
areas within this disease. It is also anticipated of Fyn in chronic myelogenous leukemia. Blood
that the recent study on lncRNA-BGL3 will spark 111(5):2904–2908
interest into researching the impact of lncRNAs Barnes DJ, Palaiologou D, Panousopoulou E, Schultheis
B, Yong AS, Wong A, Pattacini L, Goldman JM, Melo
in CML. These areas of interest are bolstered
JV (2005) Bcr-Abl expression levels determine the
with the emerging accessibility to high-powered rate of development of resistance to imatinib mesylate
fluorescence microscopy, which can monitor the in chronic myeloid leukemia. Cancer Res
spatiotemporal behaviour of proteins and RNA. 65(19):8912–8919
Baum KJ, Ren R (2008) Effect of Ras inhibition in hema-
Finally, availability of pathway inhibitors and
topoiesis and BCR/ABL leukemogenesis. J Hematol
genome editing (TALEN and crispR) systems Oncol 1:5
(Gaj et al. 2013) is a powerful option to function- Bedi A, Barber JP, Bedi GC, El-Deiry WS, Sidransky D,
ally validate pathways identified by next- Vala MS, Akhtar AJ, Hilton J, Jones RJ (1995) BCR-
ABL-mediated inhibition of apoptosis with delay of
generation sequencing/proteomic studies in both
G2/M transition after DNA damage: a mechanism of
cell lines and mouse models. These technologies resistance to multiple anticancer agents. Blood
will make for an exciting time to uncover novel 86(3):1148–1158
32 B. Chereda and J.V. Melo

Bedi A, Zehnbauer BA, Barber JP, Sharkis SJ, Jones RJ the BCR/ABL oncoprotein are required for the
(1994) Inhibition of apoptosis by BCR-ABL in MAPK-hnRNP-E2 dependent suppression of C/
chronic myeloid leukemia. Blood 83(8):2038–2044 EBPalpha-driven myeloid differentiation. Blood
Bhatia R, Holtz M, Niu N, Gray R, Snyder DS, Sawyers 110(3):994–1003
CL, Arber DA, Slovak ML, Forman SJ (2003) Chen Y, Hu Y, Zhang H, Peng C, Li S (2009) Loss of the
Persistence of malignant hematopoietic progenitors in Alox5 gene impairs leukemia stem cells and prevents
chronic myelogenous leukemia patients in complete chronic myeloid leukemia. Nat Genet 41(7):
cytogenetic remission following imatinib mesylate 783–792
treatment. Blood 101(12):4701–4707 Chen Y, Peng C, Abraham SA, Shan Y, Guo Z, Desouza N,
Biernaux C, Loos M, Sels A, Huez G, Stryckmans P Cheloni G, Li D, Holyoake TL, Li S (2014)
(1995) Detection of major bcr-abl gene expression at a Arachidonate 15-lipoxygenase is required for chronic
very low level in blood cells of some healthy individu- myeloid leukemia stem cell survival. J Clin Invest
als. Blood 86(8):3118–3122 124(9):3847–3862
Birchenall-Roberts MC, Yoo YD, Bertolette DC 3rd, Lee Chen Y, Sullivan C, Peng C, Shan Y, Hu Y, Li D, Li S
KH, Turley JM, Bang OS, Ruscetti FW, Kim SJ (1997) (2011) A tumor suppressor function of the Msr1 gene
The p120-v-Abl protein interacts with E2F-1 and reg- in leukemia stem cells of chronic myeloid leukemia.
ulates E2F-1 transcriptional activity. J Biol Chem Blood 118(2):390–400
272(14):8905–8911 Chomel JC, Sorel N, Guilhot J, Guilhot F, Turhan AG
Birge RB, Kalodimos C, Inagaki F, Tanaka S (2009) Crk (2012) BCR-ABL expression in leukemic progenitors
and CrkL adaptor proteins: networks for physiological and primitive stem cells of patients with chronic
and pathological signaling. Cell Commun Signal 7:13 myeloid leukemia. Blood 119(12):2964–2965; author
Bissonnette RP, Echeverri F, Mahboubi A, Green DR reply 2965–2966
(1992) Apoptotic cell death induced by c-myc is inhib- Chu S, Li L, Singh H, Bhatia R (2007) BCR-tyrosine 177
ited by bcl-2. Nature 359(6395):552–554 plays an essential role in Ras and Akt activation and in
Bonifacio M, Scaffidi L, Binotto G, De Marchi F, Maino human hematopoietic progenitor transformation in
E, Calistri E, Bonalumi A, Frison L, Marin L, Medeot chronic myelogenous leukemia. Cancer Res
M, De Matteis G, Fanin R, Semenzato G, Ambrosetti 67(14):7045–7053
A, Tiribelli M (2015) Predictive factors of stable deep Cleveland JL, Dean M, Rosenberg N, Wang JY, Rapp UR
molecular response in chronic myeloid leukemia (1989) Tyrosine kinase oncogenes abrogate interleu-
patients treated with imatinib standard dose: a study kin-3 dependence of murine myeloid cells through
from the Gruppo Triveneto LMC. Blood 126(23):597 signaling pathways involving c-myc: conditional regu-
Bose S, Deininger M, Gora-Tybor J, Goldman JM, Melo lation of c-myc transcription by temperature-sensitive
JV (1998) The presence of typical and atypical BCR- v-abl. Mol Cell Biol 9(12):5685–5695
ABL fusion genes in leukocytes of normal individuals: Cobas M, Wilson A, Ernst B, Mancini SJ, MacDonald
biologic significance and implications for the assess- HR, Kemler R, Radtke F (2004) Beta-catenin is dis-
ment of minimal residual disease. Blood pensable for hematopoiesis and lymphopoiesis. J Exp
92(9):3362–3367 Med 199(2):221–229
Briscoe J, Therond PP (2013) The mechanisms of Coluccia AM, Vacca A, Dunach M, Mologni L, Redaelli
Hedgehog signalling and its roles in development and S, Bustos VH, Benati D, Pinna LA, Gambacorti-
disease. Nat Rev Mol Cell Biol 14(7):416–429 Passerini C (2007) Bcr-Abl stabilizes beta-catenin in
Burchert A, Cai D, Hofbauer LC, Samuelsson MK, Slater chronic myeloid leukemia through its tyrosine phos-
EP, Duyster J, Ritter M, Hochhaus A, Muller R, Eilers phorylation. EMBO J 26(5):1456–1466
M, Schmidt M, Neubauer A (2004) Interferon consen- Copland M, Hamilton A, Elrick LJ, Baird JW, Allan EK,
sus sequence binding protein (ICSBP; IRF-8) antago- Jordanides N, Barow M, Mountford JC, Holyoake TL
nizes BCR/ABL and down-regulates bcl-2. Blood (2006) Dasatinib (BMS-354825) targets an earlier
103(9):3480–3489 progenitor population than imatinib in primary CML
Chai SK, Nichols GL, Rothman P (1997) Constitutive but does not eliminate the quiescent fraction. Blood
activation of JAKs and STATs in BCR-Abl-expressing 107(11):4532
cell lines and peripheral blood cells derived from leu- Corbin AS, Agarwal A, Loriaux M, Cortes J, Deininger
kemic patients. J Immunol 159(10):4720–4728 MW, Druker BJ (2011) Human chronic myeloid leu-
Chakraborty S, Stark JM, Sun CL, Modi H, Chen W, kemia stem cells are insensitive to imatinib despite
O’Connor TR, Forman SJ, Bhatia S, Bhatia R (2012) inhibition of BCR-ABL activity. J Clin Invest
Chronic myelogenous leukemia stem and progenitor 121(1):396–409
cells demonstrate chromosomal instability related to Corso A, Lazzarino M, Morra E, Merante S, Astori C,
repeated breakage-fusion-bridge cycles mediated by Bernasconi P, Boni M, Bernasconi C (1995) Chronic
increased nonhomologous end joining. Blood myelogenous leukemia and exposure to ionizing radi-
119(26):6187–6197 ation--a retrospective study of 443 patients. Ann
Chang JS, Santhanam R, Trotta R, Neviani P, Eiring Hematol 70(2):79–82
AM, Briercheck E, Ronchetti M, Roy DC, Calabretta Daley GQ, Baltimore D (1988) Transformation of an
B, Caligiuri MA, Perrotti D (2007) High levels of interleukin 3-dependent hematopoietic cell line by the
2 The Biology and Pathogenesis of Chronic Myeloid Leukemia 33

chronic myelogenous leukemia-specific P210bcr/abl Eaves C, Udomsakdi C, Cashman J, Barnett M, Eaves A


protein. Proc Natl Acad Sci U S A 85(23):9312–9316 (1993) The biology of normal and neoplastic stem
Daley GQ, Van Etten RA, Baltimore D (1990) Induction cells in CML. Leuk Lymphoma 11(Suppl 1):245–253
of chronic myelogenous leukemia in mice by the Eiring AM, Harb JG, Neviani P, Garton C, Oaks JJ, Spizzo
P210bcr/abl gene of the Philadelphia chromosome. R, Liu S, Schwind S, Santhanam R, Hickey CJ, Becker
Science 247(4944):824–830 H, Chandler JC, Andino R, Cortes J, Hokland P,
Dang CV (2012) MYC on the path to cancer. Cell Huettner CS, Bhatia R, Roy DC, Liebhaber SA,
149(1):22–35 Caligiuri MA, Marcucci G, Garzon R, Croce CM,
Danhauser-Riedl S, Warmuth M, Druker BJ, Emmerich B, Calin GA, Perrotti D (2010) miR-328 functions as an
Hallek M (1996) Activation of Src kinases p53/56lyn RNA decoy to modulate hnRNP E2 regulation of
and p59hck by p210bcr/abl in myeloid cells. Cancer mRNA translation in leukemic blasts. Cell
Res 56(15):3589–3596 140(5):652–665
de Andres-Aguayo L, Varas F, Kallin EM, Infante JF, Ellis SL, Nilsson SK (2012) The location and cellular
Wurst W, Floss T, Graf T (2011) Musashi 2 is a regula- composition of the hemopoietic stem cell niche.
tor of the HSC compartment identified by a retroviral Cytotherapy 14(2):135–143
insertion screen and knockout mice. Blood Ema H, Suda T (2012) Two anatomically distinct niches
118(3):554–564 regulate stem cell activity. Blood 120(11):2174–2181
de Groot RP, Raaijmakers JA, Lammers JW, Koenderman Engelman A, Rosenberg N (1990a) bcr/abl and src but not
L (2000) STAT5-dependent CyclinD1 and Bcl-xL myc and ras replace v-abl in lymphoid transformation.
expression in Bcr-Abl-transformed cells. Mol Cell Mol Cell Biol 10(8):4365–4369
Biol Res Commun 3(5):299–305 Engelman A, Rosenberg N (1990b) Temperature-sensitive
Deininger M, O’Brien SG, Guilhot F, Goldman JM, mutants of Abelson murine leukemia virus deficient in
Hochhaus A, Hughes TP, Radich JP, Hatfield AK, protein tyrosine kinase activity. J Virol
Mone M, Filian J, Reynolds J, Gathmann I, Larson 64(9):4242–4251
RA, Druker BJ (2009) International randomized study Fioretos T, Nilsson PG, Aman P, Heim S, Kristoffersson
of interferon vs STI571 (IRIS) 8-year follow up: sus- U, Malm C, Simonsson B, Turesson I, Mitelman F
tained survival and low risk for progression or events (1993) Clinical impact of breakpoint position within
in patients with newly diagnosed Chronic Myeloid M-bcr in chronic myeloid leukemia. Leukemia
Leukemia in Chronic Phase (CML-CP) treated with 7(8):1225–1231
imatinib. ASH annual meeting abstracts 114(22):1126 Gabriele L, Phung J, Fukumoto J, Segal D, Wang IM,
Deininger MW, Goldman JM, Melo JV (2000) The Giannakakou P, Giese NA, Ozato K, Morse HC 3rd
molecular biology of chronic myeloid leukemia. (1999) Regulation of apoptosis in myeloid cells by
Blood 96(10):3343–3356 interferon consensus sequence-binding protein. J Exp
Delgado MD, Leon J (2010) Myc roles in hematopoiesis Med 190(3):411–421
and leukemia. Genes Cancer 1(6):605–616 Gaiger A, Henn T, Horth E, Geissler K, Mitterbauer G,
Deutsch E, Dugray A, AbdulKarim B, Marangoni E, Maier-Dobersberger T, Greinix H, Mannhalter C,
Maggiorella L, Vaganay S, M’Kacher R, Rasy SD, Haas OA, Lechner K, Lion T (1995) Increase of bcr-
Eschwege F, Vainchenker W, Turhan AG, Bourhis abl chimeric mRNA expression in tumor cells of
J (2001) BCR-ABL down-regulates the DNA repair patients with chronic myeloid leukemia precedes dis-
protein DNA-PKcs. Blood 97(7):2084–2090 ease progression. Blood 86(6):2371–2378
Dierov J, Sanchez PV, Burke BA, Padilla-Nash H, Putt Gaj T, Gersbach CA, Barbas CF 3rd (2013) ZFN, TALEN,
ME, Ried T, Carroll M (2009) BCR/ABL induces and CRISPR/Cas-based methods for genome engi-
chromosomal instability after genotoxic stress and neering. Trends Biotechnol 31(7):397–405
alters the cell death threshold. Leukemia Goldman JM, Melo JV (2003) Chronic myeloid leuke-
23(2):279–286 mia--advances in biology and new approaches to treat-
Dowding C, Guo AP, Maisin D, Gordon MY, Goldman ment. N Engl J Med 349(15):1451–1464
JM (1991) The effects of interferon-alpha on the pro- Graham SM, Jorgensen HG, Allan E, Pearson C, Alcorn
liferation of CML progenitor cells in vitro are not MJ, Richmond L, Holyoake TL (2002) Primitive, qui-
related to the precise position of the M-BCR break- escent, Philadelphia-positive stem cells from patients
point. Br J Haematol 77(2):165–171 with chronic myeloid leukemia are insensitive to
Druker BJ, Guilhot F, O’Brien SG, Gathmann I, Kantarjian STI571 in vitro. Blood 99(1):319–325
H, Gattermann N, Deininger MW, Silver RT, Goldman Guerzoni C, Bardini M, Mariani SA, Ferrari-Amorotti G,
JM, Stone RM, Cervantes F, Hochhaus A, Powell BL, Neviani P, Panno ML, Zhang Y, Martinez R, Perrotti
Gabrilove JL, Rousselot P, Reiffers J, Cornelissen JJ, D, Calabretta B (2006) Inducible activation of
Hughes T, Agis H, Fischer T, Verhoef G, Shepherd J, CEBPB, a gene negatively regulated by BCR/ABL,
Saglio G, Gratwohl A, Nielsen JL, Radich JP, inhibits proliferation and promotes differentiation of
Simonsson B, Taylor K, Baccarani M, So C, Letvak L, BCR/ABL-expressing cells. Blood 107(10):
Larson RA (2006) Five-year follow-up of patients 4080–4089
receiving imatinib for chronic myeloid leukemia. N Guinn BA, Mills KI (1997) p53 mutations, methylation
Engl J Med 355(23):2408–2417 and genomic instability in the progression of chronic
34 B. Chereda and J.V. Melo

myeloid leukaemia. Leuk Lymphoma 26(3–4): Heidel FH, Bullinger L, Feng Z, Wang Z, Neff TA, Stein
211–226 L, Kalaitzidis D, Lane SW, Armstrong SA (2012)
Guo G, Kang Q, Chen Q, Chen Z, Wang J, Tan L, Chen Genetic and pharmacologic inhibition of beta-catenin
JL (2014a) High expression of long non-coding targets imatinib-resistant leukemia stem cells in
RNA H19 is required for efficient tumorigenesis CML. Cell Stem Cell 10(4):412–424
induced by Bcr-Abl oncogene. FEBS Lett 588(9): Heisterkamp N, Jenster G, ten Hoeve J, Zovich D,
1780–1786 Pattengale PK, Groffen J (1990) Acute leukaemia in
Guo G, Kang Q, Zhu X, Chen Q, Wang X, Chen Y, bcr/abl transgenic mice. Nature 344(6263):251–253
Ouyang J, Zhang L, Tan H, Chen R, Huang S, Chen JL Hennighausen L, Robinson GW (2008) Interpretation of
(2014b) A long noncoding RNA critically regulates cytokine signaling through the transcription factors
Bcr-Abl-mediated cellular transformation by acting as STAT5A and STAT5B. Genes Dev 22(6):711–721
a competitive endogenous RNA. Oncogene 34(14): Hoelbl A, Schuster C, Kovacic B, Zhu B, Wickre M,
1768–1779 Hoelzl MA, Fajmann S, Grebien F, Warsch W, Stengl
Halbach S, Rigbolt KT, Wohrle FU, Diedrich B, G, Hennighausen L, Poli V, Beug H, Moriggl R, Sexl
Gretzmeier C, Brummer T, Dengjel J (2013) V (2010) Stat5 is indispensable for the maintenance of
Alterations of Gab2 signalling complexes in imatinib bcr/abl-positive leukaemia. EMBO Mol Med
and dasatinib treated chronic myeloid leukaemia cells. 2(3):98–110
Cell Commun Signal 11(1):30 Holtschke T, Lohler J, Kanno Y, Fehr T, Giese N,
Hanfstein B, Lauseker M, Hehlmann R, Saussele S, Erben Rosenbauer F, Lou J, Knobeloch KP, Gabriele L,
P, Dietz C, Fabarius A, Proetel U, Schnittger S, Waring JF, Bachmann MF, Zinkernagel RM, Morse
Haferlach C, Krause SW, Schubert J, Einsele H, Hanel HC 3rd, Ozato K, Horak I (1996) Immunodeficiency
M, Dengler J, Falge C, Kanz L, Neubauer A, Kneba and chronic myelogenous leukemia-like syndrome in
M, Stegelmann F, Pfreundschuh M, Waller CF, mice with a targeted mutation of the ICSBP gene. Cell
Spiekermann K, Baerlocher GM, Pfirrmann M, 87(2):307–317
Hasford J, Hofmann WK, Hochhaus A, Muller MC Holyoake T, Jiang X, Eaves C, Eaves A (1999) Isolation
(2014) Distinct characteristics of e13a2 versus e14a2 of a highly quiescent subpopulation of primitive leu-
BCR-ABL1 driven chronic myeloid leukemia under kemic cells in chronic myeloid leukemia. Blood
first-line therapy with imatinib. Haematologica 94(6):2056–2064
99(9):1441–1447 Honda H, Ushijima T, Wakazono K, Oda H, Tanaka Y,
Hansen N, Agerstam H, Wahlestedt M, Landberg N, Aizawa S, Ishikawa T, Yazaki Y, Hirai H (2000)
Askmyr M, Ehinger M, Rissler M, Lilljebjorn H, Acquired loss of p53 induces blastic transformation in
Johnels P, Ishiko J, Melo JV, Alexander WS, Bryder p210(bcr/abl)-expressing hematopoietic cells: a trans-
D, Jaras M, Fioretos T (2013) SOCS2 is dispensable genic study for blast crisis of human CML. Blood
for BCR/ABL1-induced chronic myeloid leukemia- 95(4):1144–1150
like disease and for normal hematopoietic stem cell Horita M, Andreu EJ, Benito A, Arbona C, Sanz C, Benet
function. Leukemia 27(1):130–135 I, Prosper F, Fernandez-Luna JL (2000) Blockade of
Hantschel O (2012) Structure, regulation, signaling, and the Bcr-Abl kinase activity induces apoptosis of
targeting of abl kinases in cancer. Genes Cancer chronic myelogenous leukemia cells by suppressing
3(5–6):436–446 signal transducer and activator of transcription
Hantschel O, Warsch W, Eckelhart E, Kaupe I, Grebien F, 5-dependent expression of Bcl-xL. J Exp Med
Wagner KU, Superti-Furga G, Sexl V (2012) BCR- 191(6):977–984
ABL uncouples canonical JAK2-STAT5 signaling in Hsieh MY, Van Etten RA (2014) IKK-dependent activa-
chronic myeloid leukemia. Nat Chem Biol 8(3): tion of NF-kappaB contributes to myeloid and lym-
285–293 phoid leukemogenesis by BCR-ABL1. Blood
Hao SX, Ren R (2000) Expression of interferon consen- 123(15):2401–2411
sus sequence binding protein (ICSBP) is downregu- Hu Y, Liu Y, Pelletier S, Buchdunger E, Warmuth M,
lated in Bcr-Abl-induced murine chronic myelogenous Fabbro D, Hallek M, Van Etten RA, Li S (2004)
leukemia-like disease, and forced coexpression of Requirement of Src kinases Lyn, Hck and Fgr for
ICSBP inhibits Bcr-Abl-induced myeloproliferative BCR-ABL1-induced B-lymphoblastic leukemia but
disorder. Mol Cell Biol 20(4):1149–1161 not chronic myeloid leukemia. Nat Genet
Hariharan IK, Adams JM, Cory S (1988) bcr-abl onco- 36(5):453–461
gene renders myeloid cell line factor independent: Huang W, Zhou W, Saberwal G, Konieczna I, Horvath E,
potential autocrine mechanism in chronic myeloid leu- Katsoulidis E, Platanias LC, Eklund EA (2010)
kemia. Oncogene Res 3(4):387–399 Interferon consensus sequence binding protein
Hazlehurst LA, Argilagos RF, Dalton WS (2007) Beta1 (ICSBP) decreases beta-catenin activity in myeloid
integrin mediated adhesion increases Bim protein deg- cells by repressing GAS2 transcription. Mol Cell Biol
radation and contributes to drug resistance in leukae- 30(19):4575–4594
mia cells. Br J Haematol 136(2):269–275 Hurtz C, Hatzi K, Cerchietti L, Braig M, Park E, Kim YM,
Hehlmann R (2012) How I treat CML blast crisis. Blood Herzog S, Ramezani-Rad P, Jumaa H, Muller MC,
120(4):737–747 Hofmann WK, Hochhaus A, Ye BH, Agarwal A,
2 The Biology and Pathogenesis of Chronic Myeloid Leukemia 35

Druker BJ, Shah NP, Melnick AM, Muschen M (2011) Kapoor I, Pal P, Lochab S, Kanaujiya JK, Trivedi AK
BCL6-mediated repression of p53 is critical for leuke- (2012) Proteomics approaches for myeloid leukemia
mia stem cell survival in chronic myeloid leukemia. drug discovery. Expert Opin Drug Discov
J Exp Med 208(11):2163–2174 7(12):1165–1175
Ilaria RL Jr (2005) Pathobiology of lymphoid and myeloid Kelliher MA, McLaughlin J, Witte ON, Rosenberg N
blast crisis and management issues. Hematology Am (1990) Induction of a chronic myelogenous leukemia-
Soc Hematol Educ Program 1(1):188–194 like syndrome in mice with v-abl and BCR/ABL. Proc
Ito T, Kwon HY, Zimdahl B, Congdon KL, Blum J, Lento Natl Acad Sci U S A 87(17):6649–6653
WE, Zhao C, Lagoo A, Gerrard G, Foroni L, Goldman Kharas MG, Lengner CJ, Al-Shahrour F, Bullinger L, Ball
J, Goh H, Kim SH, Kim DW, Chuah C, Oehler VG, B, Zaidi S, Morgan K, Tam W, Paktinat M, Okabe R,
Radich JP, Jordan CT, Reya T (2010) Regulation of Gozo M, Einhorn W, Lane SW, Scholl C, Frohling S,
myeloid leukaemia by the cell-fate determinant Fleming M, Ebert BL, Gilliland DG, Jaenisch R,
Musashi. Nature 466(7307):765–768 Daley GQ (2010) Musashi-2 regulates normal hema-
Jain P, Kantarjian HM, Luthra R, Nogueras Gonzalez G, topoiesis and promotes aggressive myeloid leukemia.
Jabbour E, Sasaki K, Guillermo Romo C, Patel KP, Nat Med 16(8):903–908
Kadia T, Borthakur G, Estrov Z, Ravandi F, O’Brien S, Kim LC, Song L, Haura EB (2009) Src kinases as thera-
Cortes JE (2015) Impact of the type of BCR-ABL peutic targets for cancer. Nat Rev Clin Oncol
fusion transcripts on response and survival in patients 6(10):587–595
with chronic phase CML treated with four frontline Klejman A, Rushen L, Morrione A, Slupianek A, Skorski
TKI modalities. Blood 126(23):598 T (2002a) Phosphatidylinositol-3 kinase inhibitors
Jamieson CH, Ailles LE, Dylla SJ, Muijtjens M, Jones C, enhance the anti-leukemia effect of STI571. Oncogene
Zehnder JL, Gotlib J, Li K, Manz MG, Keating A, 21(38):5868–5876
Sawyers CL, Weissman IL (2004) Granulocyte- Klejman A, Schreiner SJ, Nieborowska-Skorska M,
macrophage progenitors as candidate leukemic stem Slupianek A, Wilson M, Smithgall TE, Skorski T
cells in blast-crisis CML. N Engl J Med 351(7): (2002b) The Src family kinase Hck couples BCR/
657–667 ABL to STAT5 activation in myeloid leukemia cells.
Jiang Q, Crews LA, Barrett CL, Chun HJ, Court AC, EMBO J 21(21):5766–5774
Isquith JM, Zipeto MA, Goff DJ, Minden M, Koch U, Wilson A, Cobas M, Kemler R, Macdonald HR,
Sadarangani A, Rusert JM, Dao KH, Morris SR, Radtke F (2008) Simultaneous loss of beta- and
Goldstein LS, Marra MA, Frazer KA, Jamieson CH gamma-catenin does not perturb hematopoiesis or
(2013) ADAR1 promotes malignant progenitor repro- lymphopoiesis. Blood 111(1):160–164
gramming in chronic myeloid leukemia. Proc Natl Koptyra M, Cramer K, Slupianek A, Richardson C,
Acad Sci U S A 110(3):1041–1046 Skorski T (2008) BCR/ABL promotes accumulation
Jiang X, Zhao Y, Smith C, Gasparetto M, Turhan A, Eaves of chromosomal aberrations induced by oxidative and
A, Eaves C (2007) Chronic myeloid leukemia stem genotoxic stress. Leukemia 22(10):1969–1972
cells possess multiple unique features of resistance to Kumari A, Brendel C, Hochhaus A, Neubauer A, Burchert
BCR-ABL targeted therapies. Leukemia A (2012) Low BCR-ABL expression levels in hemato-
21(5):926–935 poietic precursor cells enable persistence of chronic
Johansson B, Fioretos T, Mitelman F (2002) Cytogenetic myeloid leukemia under imatinib. Blood
and molecular genetic evolution of chronic myeloid 119(2):530–539
leukemia. Acta Haematol 107(2):76–94 Li L, Wang L, Wang Z, Ho Y, McDonald T, Holyoake TL,
Jorgensen HG, Allan EK, Jordanides NE, Mountford JC, Chen W, Bhatia R (2012) Activation of p53 by SIRT1
Holyoake TL (2007) Nilotinib exerts equipotent antip- inhibition enhances elimination of CML leukemia
roliferative effects to imatinib and does not induce stem cells in combination with imatinib. Cancer Cell
apoptosis in CD34+ CML cells. Blood 21(2):266–281
109(9):4016–4019 Lin TS, Mahajan S, Frank DA (2000) STAT signaling in
Kabarowski JH, Witte ON (2000) Consequences of BCR- the pathogenesis and treatment of leukemias.
ABL expression within the hematopoietic stem cell in Oncogene 19(21):2496–2504
chronic myeloid leukemia. Stem Cells 18(6):399–408 Lindauer M, Hochhaus A (2014) Dasatinib. Recent
Kantarjian H, O’Brien S, Jabbour E, Garcia-Manero G, Results Cancer Res 201:27–65
Quintas-Cardama A, Shan J, Rios MB, Ravandi F, Lionberger JM, Wilson MB, Smithgall TE (2000)
Faderl S, Kadia T, Borthakur G, Huang X, Champlin Transformation of myeloid leukemia cells to cytokine
R, Talpaz M, Cortes J (2012) Improved survival in independence by Bcr-Abl is suppressed by kinase-
chronic myeloid leukemia since the introduction of defective Hck. J Biol Chem 275(24):18581–18585
imatinib therapy: a single-institution historical experi- Machova Polakova K, Lopotova T, Klamova H, Burda P,
ence. Blood 119(9):1981–1987 Trneny M, Stopka T, Moravcova J (2011) Expression
Kantarjian HM, Keating MJ, Talpaz M, Walters RS, Smith patterns of microRNAs associated with CML phases
TL, Cork A, McCredie KB, Freireich EJ (1987) and their disease related targets. Mol Cancer 10:41
Chronic myelogenous leukemia in blast crisis. Mahon FX, Rea D, Guilhot J, Guilhot F, Huguet F,
Analysis of 242 patients. Am J Med 83(3):445–454 Nicolini F, Legros L, Charbonnier A, Guerci A, Varet
36 B. Chereda and J.V. Melo

B, Etienne G, Reiffers J, Rousselot P (2010) is mediated by Bad-dependent and -independent path-


Discontinuation of imatinib in patients with chronic ways: roles for phosphatidylinositol 3-kinase and Raf.
myeloid leukaemia who have maintained complete Mol Cell Biol 20(4):1179–1186
molecular remission for at least 2 years: the prospec- Neviani P, Harb JG, Oaks JJ, Santhanam R, Walker CJ,
tive, multicentre Stop Imatinib (STIM) trial. Lancet Ellis JJ, Ferenchak G, Dorrance AM, Paisie CA,
Oncol 11(11):1029–1035 Eiring AM, Ma Y, Mao HC, Zhang B, Wunderlich M,
Mar BG, Amakye D, Aifantis I, Buonamici S (2011) The con- May PC, Sun C, Saddoughi SA, Bielawski J, Blum W,
troversial role of the Hedgehog pathway in normal and Klisovic RB, Solt JA, Byrd JC, Volinia S, Cortes J,
malignant hematopoiesis. Leukemia 25(11):1665–1673 Huettner CS, Koschmieder S, Holyoake TL, Devine S,
Marega M, Piazza RG, Pirola A, Redaelli S, Mogavero A, Caligiuri MA, Croce CM, Garzon R, Ogretmen B,
Iacobucci I, Meneghetti I, Parma M, Pogliani EM, Arlinghaus RB, Chen CS, Bittman R, Hokland P, Roy
Gambacorti-Passerini C (2010) BCR and BCR-ABL DC, Milojkovic D, Apperley J, Goldman JM, Reid A,
regulation during myeloid differentiation in healthy Mulloy JC, Bhatia R, Marcucci G, Perrotti D (2013)
donors and in chronic phase/blast crisis CML patients. PP2A-activating drugs selectively eradicate TKI-
Leukemia 24(8):1445–1449 resistant chronic myeloid leukemic stem cells. J Clin
Mayerhofer M, Valent P, Sperr WR, Griffin JD, Sillaber C Invest 123(10):4144–4157
(2002) BCR/ABL induces expression of vascular Neviani P, Santhanam R, Trotta R, Notari M, Blaser BW,
endothelial growth factor and its transcriptional acti- Liu S, Mao H, Chang JS, Galietta A, Uttam A, Roy
vator, hypoxia inducible factor-1alpha, through a path- DC, Valtieri M, Bruner-Klisovic R, Caligiuri MA,
way involving phosphoinositide 3-kinase and the Bloomfield CD, Marcucci G, Perrotti D (2005) The
mammalian target of rapamycin. Blood tumor suppressor PP2A is functionally inactivated in
100(10):3767–3775 blast crisis CML through the inhibitory activity of the
Melo JV (1996) The diversity of BCR-ABL fusion pro- BCR/ABL-regulated SET protein. Cancer Cell
teins and their relationship to leukemia phenotype. 8(5):355–368
Blood 88(7):2375–2384 Ng KP, Hillmer AM, Chuah CT, Juan WC, Ko TK, Teo
Melo JV, Barnes DJ (2007a) Chronic myeloid leukaemia AS, Ariyaratne PN, Takahashi N, Sawada K, Fei Y,
as a model of disease evolution in human cancer. Nat Soh S, Lee WH, Huang JW, Allen JC Jr, Woo XY,
Rev Cancer 7(6):441–453 Nagarajan N, Kumar V, Thalamuthu A, Poh WT, Ang
Melo JV, Barnes DJ (2007b) Chronic myeloid leukemia: AL, Mya HT, How GF, Yang LY, Koh LP, Chowbay
biology of advanced phase. In: Myeloproliferative dis- B, Chang CT, Nadarajan VS, Chng WJ, Than H, Lim
orders. Springer Berlin Heidelberg, New York, LC, Goh YT, Zhang S, Poh D, Tan P, Seet JE, Ang
pp 37–59 MK, Chau NM, Ng QS, Tan DS, Soda M, Isobe K,
Mendizabal AM, Garcia-Gonzalez P, Levine PH (2013) Nothen MM, Wong TY, Shahab A, Ruan X, Cacheux-
Regional variations in age at diagnosis and overall sur- Rataboul V, Sung WK, Tan EH, Yatabe Y, Mano H,
vival among patients with chronic myeloid leukemia Soo RA, Chin TM, Lim WT, Ruan Y, Ong ST (2012)
from low and middle income countries. Cancer A common BIM deletion polymorphism mediates
Epidemiol 37(3):247–254 intrinsic resistance and inferior responses to tyrosine
Milojkovic D, Apperley J (2009) Mechanisms of resis- kinase inhibitors in cancer. Nat Med 18(4):521–528
tance to imatinib and second-generation tyrosine Notari M, Neviani P, Santhanam R, Blaser BW, Chang JS,
inhibitors in chronic myeloid leukemia. Clin Cancer Galietta A, Willis AE, Roy DC, Caligiuri MA,
Res 15(24):7519–7527 Marcucci G, Perrotti D (2006) A MAPK/HNRPK
Moon RT, Kohn AD, Ferrari GVD, Kaykas A (2004) pathway controls BCR/ABL oncogenic potential by
WNT and [beta]-catenin signalling: diseases and ther- regulating MYC mRNA translation. Blood
apies. Nat Rev Genet 5(9):691–701 107(6):2507–2516
Morotti A, Panuzzo C, Crivellaro S, Pergolizzi B, Packer LM, Rana S, Hayward R, O’Hare T, Eide CA,
Familiari U, Berger AH, Saglio G, Pandolfi PP (2014) Rebocho A, Heidorn S, Zabriskie MS, Niculescu-
BCR-ABL disrupts PTEN nuclear-cytoplasmic shut- Duvaz I, Druker BJ, Springer C, Marais R (2011)
tling through phosphorylation-dependent activation of Nilotinib and MEK inhibitors induce synthetic lethal-
HAUSP. Leukemia 28(6):1326–1333 ity through paradoxical activation of RAF in drug-
Naka K, Hoshii T, Muraguchi T, Tadokoro Y, Ooshio T, resistant chronic myeloid leukemia. Cancer Cell
Kondo Y, Nakao S, Motoyama N, Hirao A (2010) 20(6):715–727
TGF-beta-FOXO signalling maintains leukaemia- Pant V, Quintas-Cardama A, Lozano G (2012) The p53
initiating cells in chronic myeloid leukaemia. Nature pathway in hematopoiesis: lessons from mouse mod-
463(7281):676–680 els, implications for humans. Blood
Nakamura M, Okano H, Blendy JA, Montell C (1994) 120(26):5118–5127
Musashi, a neural RNA-binding protein required for Park SM, Deering RP, Lu Y, Tivnan P, Lianoglou S,
Drosophila adult external sensory organ development. Al-Shahrour F, Ebert BL, Hacohen N, Leslie C, Daley
Neuron 13(1):67–81 GQ, Lengner CJ, Kharas MG (2014) Musashi-2 con-
Neshat MS, Raitano AB, Wang HG, Reed JC, Sawyers CL trols cell fate, lineage bias, and TGF-beta signaling in
(2000) The survival function of the Bcr-Abl oncogene HSCs. J Exp Med 211(1):71–87
2 The Biology and Pathogenesis of Chronic Myeloid Leukemia 37

Pellicano F, Simara P, Sinclair A, Helgason GV, Copland Wall R, McPherson JD, Moore RA, Pu M, Bao L,
M, Grant S, Holyoake TL (2011) The MEK inhibitor Jackson-Fisher A, Munchhof M, VanArsdale T, Reya
PD184352 enhances BMS-214662-induced apoptosis T, Morris SR, Minden MD, Messer K, Mikkola HK,
in CD34+ CML stem/progenitor cells. Leukemia Marra MA, Hudson TJ, Jamieson CH (2015) GLI2
25(7):1159–1167 inhibition abrogates human leukemia stem cell dor-
Pendergast AM, Quilliam LA, Cripe LD, Bassing CH, Dai mancy. J Transl Med 13:98
Z, Li N, Batzer A, Rabun KM, Der CJ, Schlessinger Salomoni P, Calabretta B (2009) Targeted therapies and
J et al (1993) BCR-ABL-induced oncogenesis is autophagy: new insights from chronic myeloid leuke-
mediated by direct interaction with the SH2 domain of mia. Autophagy 5(7):1050–1051
the GRB-2 adaptor protein. Cell 75(1):175–185 Salomoni P, Condorelli F, Sweeney SM, Calabretta B
Perrotti D, Jamieson C, Goldman J, Skorski T (2010) (2000) Versatility of BCR/ABL-expressing leukemic
Chronic myeloid leukemia: mechanisms of blastic cells in circumventing proapoptotic BAD effects.
transformation. J Clin Invest 120(7):2254–2264 Blood 96(2):676–684
Peterson LF, Mitrikeska E, Giannola D, Lui Y, Sun H, Samanta AK, Chakraborty SN, Wang Y, Kantarjian H, Sun
Bixby D, Malek SN, Donato NJ, Wang S, Talpaz M X, Hood J, Perrotti D, Arlinghaus RB (2009) Jak2
(2011) p53 stabilization induces apoptosis in chronic inhibition deactivates Lyn kinase through the SET-
myeloid leukemia blast crisis cells. Leukemia PP2A-SHP1 pathway, causing apoptosis in drug-
25(5):761–769 resistant cells from chronic myelogenous leukemia
Preisler HD, Sato H, Yang PM, Wilson M, Kaufman C, patients. Oncogene 28(14):1669–1681
Watt R (1988) Assessment of c-myc expression in Sanchez-Garcia I, Grutz G (1995) Tumorigenic activity of
individual leukemic cells. Leuk Res 12(6):507–516 the BCR-ABL oncogenes is mediated by BCL2. Proc
Ptasznik A, Nakata Y, Kalota A, Emerson SG, Gewirtz Natl Acad Sci U S A 92(12):5287–5291
AM (2004) Short interfering RNA (siRNA) targeting Sattler M, Mohi MG, Pride YB, Quinnan LR, Malouf NA,
the Lyn kinase induces apoptosis in primary, and drug- Podar K, Gesbert F, Iwasaki H, Li S, Van Etten RA,
resistant, BCR-ABL1(+) leukemia cells. Nat Med Gu H, Griffin JD, Neel BG (2002) Critical role for
10(11):1187–1189 Gab2 in transformation by BCR/ABL. Cancer Cell
Puil L, Liu J, Gish G, Mbamalu G, Bowtell D, Pelicci PG, 1(5):479–492
Arlinghaus R, Pawson T (1994) Bcr-Abl oncoproteins Sattler M, Salgia R, Okuda K, Uemura N, Durstin MA,
bind directly to activators of the Ras signalling path- Pisick E, Xu G, Li JL, Prasad KV, Griffin JD (1996)
way. EMBO J 13(4):764–773 The proto-oncogene product p120CBL and the
Quintas-Cardama A, Cortes JE (2006) Chronic myeloid adaptor proteins CRKL and c-CRK link c-ABL,
leukemia: diagnosis and treatment. Mayo Clin Proc p190BCR/ABL and p210BCR/ABL to the phospha-
81(7):973–988 tidylinositol-3′ kinase pathway. Oncogene 12(4):
Ratajczak MZ, Kant JA, Luger SM, Hijiya N, Zhang J, 839–846
Zon G, Gewirtz AM (1992) In vivo treatment of Sawyers CL, Callahan W, Witte ON (1992) Dominant
human leukemia in a scid mouse model with c-myb negative MYC blocks transformation by ABL onco-
antisense oligodeoxynucleotides. Proc Natl Acad Sci genes. Cell 70(6):901–910
U S A 89(24):11823–11827 Schafranek L, Nievergall E, Powell JA, Hiwase DK,
Reavie L, Buckley SM, Loizou E, Takeishi S, Aranda- Leclercq T, Hughes TP, White DL (2014) Sustained
Orgilles B, Ndiaye-Lobry D, Abdel-Wahab O, Ibrahim inhibition of STAT5, but not JAK2, is essential for
S, Nakayama KI, Aifantis I (2013) Regulation of TKI-induced cell death in chronic myeloid leukemia.
c-Myc ubiquitination controls chronic myelogenous Leukemia 29(1):76–85
leukemia initiation and progression. Cancer Cell Scheller M, Schonheit J, Zimmermann K, Leser U,
23(3):362–375 Rosenbauer F, Leutz A (2013) Cross talk between
Reuther JY, Reuther GW, Cortez D, Pendergast AM, Wnt/beta-catenin and Irf8 in leukemia progression and
Baldwin AS Jr (1998) A requirement for NF-kappaB drug resistance. J Exp Med 210(11):2239–2256
activation in Bcr-Abl-mediated transformation. Genes Schmidt M, Nagel S, Proba J, Thiede C, Ritter M, Waring
Dev 12(7):968–981 JF, Rosenbauer F, Huhn D, Wittig B, Horak I,
Rozman C, Urbano-Ispizua A, Cervantes F, Rozman M, Neubauer A (1998) Lack of interferon consensus
Colomer D, Feliz P, Pujades A, Vives Corrons JL sequence binding protein (ICSBP) transcripts in
(1995) Analysis of the clinical relevance of the break- human myeloid leukemias. Blood 91(1):22–29
point location within M-BCR and the type of chimeric Score J, Calasanz MJ, Ottman O, Pane F, Yeh RF,
mRNA in chronic myelogenous leukemia. Leukemia Sobrinho-Simoes MA, Kreil S, Ward D, Hidalgo-
9(6):1104–1107 Curtis C, Melo JV, Wiemels J, Nadel B, Cross NC,
Rusconi F, Piazza R, Vagge E, Gambacorti-Passerini C Grand FH (2010) Analysis of genomic breakpoints in
(2014) Bosutinib : a review of preclinical and clinical p190 and p210 BCR-ABL indicate distinct mecha-
studies in chronic myelogenous leukemia. Expert nisms of formation. Leukemia 24(10):1742–1750
Opin Pharmacother 15(5):701–710 Seo J-H, Wood LJ, Agarwal A, O’Hare T, Elsea CR, Griswold
Sadarangani A, Pineda G, Lennon KM, Chun HJ, Shih A, IJ, Deininger MWN, Imamoto A, Druker BJ (2010) A
Schairer AE, Court AC, Goff DJ, Prashad SL, Geron I, specific need for CRKL in p210BCR-ABL-induced
38 B. Chereda and J.V. Melo

transformation of mouse hematopoietic progenitors. ABL kinase domain mutation analysis in chronic myeloid
Cancer Res 70(18):7325–7335 leukemia patients treated with tyrosine kinase inhibitors:
Shah NP, Cortes JE, Martinelli G, Smith BD, Clarke E, recommendations from an expert panel on behalf of
Copland M, Strauss L, Talpaz M (2014) Dasatinib plus European LeukemiaNet. Blood 118(5):1208–1215
smoothened (SMO) inhibitor BMS-833923 in Chronic Steelman LS, Franklin RA, Abrams SL, Chappell W,
Myeloid Leukemia (CML) with resistance or subopti- Kempf CR, Basecke J, Stivala F, Donia M, Fagone P,
mal response to a prior Tyrosine Kinase Inhibitor Nicoletti F, Libra M, Ruvolo P, Ruvolo V, Evangelisti
(TKI): phase I study CA180323. Blood 124(21):4539 C, Martelli AM, McCubrey JA (2011) Roles of the
Sheng Z, Ma L, Sun JE, Zhu LJ, Green MR (2011) BCR- Ras/Raf/MEK/ERK pathway in leukemia therapy.
ABL suppresses autophagy through ATF5-mediated Leukemia 25(7):1080–1094
regulation of mTOR transcription. Blood Steinman RA, Yang Q, Gasparetto M, Robinson LJ, Liu
118(10):2840–2848 X, Lenzner DE, Hou J, Smith C, Wang Q (2013)
Shepherd P, Suffolk R, Halsey J, Allan N (1995) Analysis Deletion of the RNA-editing enzyme ADAR1 causes
of molecular breakpoint and m-RNA transcripts in a regression of established chronic myelogenous leuke-
prospective randomized trial of interferon in chronic mia in mice. Int J Cancer 132(8):1741–1750
myeloid leukaemia: no correlation with clinical fea- Stewart MJ, Litz-Jackson S, Burgess GS, Williamson EA,
tures, cytogenetic response, duration of chronic phase, Leibowitz DS, Boswell HS (1995) Role for E2F1 in
or survival. Br J Haematol 89(3):546–554 p210 BCR-ABL downstream regulation of c-myc
Silvestri F, Banavali S, Yin M, Gopal V, Savignano C, transcription initiation. Studies in murine myeloid
Baccarani M, Preisler HD (1992) CD34-positive cell cells. Leukemia 9(9):1499–1507
selection by immunomagnetic beads and chymopap- Stoklosa T, Slupianek A, Datta M, Nieborowska-Skorska
ain. Haematologica 77(4):307–310 M, Nowicki MO, Koptyra M, Skorski T (2004) BCR/
Sionov RV, Moallem E, Berger M, Kazaz A, Gerlitz O, ABL recruits p53 tumor suppressor protein to induce
Ben-Neriah Y, Oren M, Haupt Y (1999) c-Abl neutral- drug resistance. Cell Cycle 3(11):1463–1472
izes the inhibitory effect of Mdm2 on p53. J Biol Stuppia L, Calabrese G, Peila R, Guanciali-Franchi P,
Chem 274(13):8371–8374 Morizio E, Spadano A, Palka G (1997) p53 loss and
Skorski T (2007) Genomic instability: the cause and effect point mutations are associated with suppression of
of BCR/ABL tyrosine kinase. Curr Hematol Malig apoptosis and progression of CML into myeloid blas-
Rep 2(2):69–74 tic crisis. Cancer Genet Cytogenet 98(1):28–35
Skorski T (2008) BCR/ABL, DNA damage and DNA Szczylik C, Skorski T, Nicolaides NC, Manzella L,
repair: implications for new treatment concepts. Leuk Malaguarnera L, Venturelli D, Gewirtz AM, Calabretta
Lymphoma 49(4):610–614 B (1991) Selective inhibition of leukemia cell
Skorski T (2012) Genetic mechanisms of chronic myeloid proliferation by BCR-ABL antisense oligodeoxynu-
leukemia blastic transformation. Curr Hematol Malig cleotides. Science 253(5019):562–565
Rep 7(2):87–93 Tamura T, Kong HJ, Tunyaplin C, Tsujimura H, Calame
Skorski T, Bellacosa A, Nieborowska-Skorska M, K, Ozato K (2003) ICSBP/IRF-8 inhibits mitogenic
Majewski M, Martinez R, Choi JK, Trotta R, activity of p210 Bcr/Abl in differentiating myeloid
Wlodarski P, Perrotti D, Chan TO, Wasik MA, Tsichlis progenitor cells. Blood 102(13):4547–4554
PN, Calabretta B (1997) Transformation of hemato- ten Hoeve J, Arlinghaus RB, Guo JQ, Heisterkamp N,
poietic cells by BCR/ABL requires activation of a Groffen J (1994) Tyrosine phosphorylation of CRKL
PI-3k/Akt-dependent pathway. EMBO in Philadelphia + leukemia. Blood 84(6):1731–1736
J 16(20):6151–6161 Trotta R, Vignudelli T, Candini O, Intine RV, Pecorari L,
Skorski T, Szczylik C, Malaguarnera L, Calabretta B Guerzoni C, Santilli G, Byrom MW, Goldoni S, Ford
(1991) Gene-targeted specific inhibition of chronic LP, Caligiuri MA, Maraia RJ, Perrotti D, Calabretta B
myeloid leukemia cell growth by BCR-ABL antisense (2003) BCR/ABL activates mdm2 mRNA translation
oligodeoxynucleotides. Folia Histochem Cytobiol via the La antigen. Cancer Cell 3(2):145–160
29(3):85–89 Velasco-Hernandez T, Vicente-Duenas C, Sanchez-Garcia
Slupianek A, Falinski R, Znojek P, Stoklosa T, Flis S, I, Martin-Zanca D (2013) p53 restoration kills primi-
Doneddu V, Pytel D, Synowiec E, Blasiak J, Bellacosa tive leukemia cells in vivo and increases survival of
A, Skorski T (2013) BCR-ABL1 kinase inhibits uracil leukemic mice. Cell Cycle 12(1):122–132
DNA glycosylase UNG2 to enhance oxidative DNA Waight JD, Banik D, Griffiths EA, Nemeth MJ, Abrams
damage and stimulate genomic instability. Leukemia SI (2014) Regulation of the interferon regulatory fac-
27(3):629–634 tor-8 (IRF-8) tumor suppressor gene by the Signal
Soverini S, de Benedittis C, Mancini M, Martinelli G Transducer and Activator of Transcription 5 (STAT5)
(2015) Mutations in the BCR-ABL1 kinase domain transcription factor in chronic myeloid leukemia.
and elsewhere in chronic myeloid leukemia. Clin J Biol Chem 289(22):15642–15652
Lymphoma Myeloma Leuk 15(Suppl):S120–S128 Walker CJ, Oaks JJ, Santhanam R, Neviani P, Harb JG,
Soverini S, Hochhaus A, Nicolini FE, Gruber F, Lange T, Ferenchak G, Ellis JJ, Landesman Y, Eisfeld AK,
Saglio G, Pane F, Muller MC, Ernst T, Rosti G, Porkka Gabrail NY, Smith CL, Caligiuri MA, Hokland P, Roy
K, Baccarani M, Cross NC, Martinelli G (2011) BCR- DC, Reid A, Milojkovic D, Goldman JM, Apperley J,
2 The Biology and Pathogenesis of Chronic Myeloid Leukemia 39

Garzon R, Marcucci G, Shacham S, Kauffman MG, allosteric inhibitor of BCR-ABL, prevents emer-
Perrotti D (2013) Preclinical and clinical efficacy of gence of resistant disease when administered in
XPO1/CRM1 inhibition by the karyopherin inhibitor combination with nilotinib in an in vivo murine
KPT-330 in Ph + leukemias. Blood model of chronic myeloid leukemia. Blood
122(17):3034–3044 124(21):398
Walz C, Ahmed W, Lazarides K, Betancur M, Patel N, Xie S, Lin H, Sun T, Arlinghaus RB (2002) Jak2 is
Hennighausen L, Zaleskas VM, Van Etten RA (2012) involved in c-Myc induction by Bcr-Abl. Oncogene
Essential role for Stat5a/b in myeloproliferative neo- 21(47):7137–7146
plasms induced by BCR-ABL1 and JAK2(V617F) in XuFeng R, Boyer MJ, Shen H, Li Y, Yu H, Gao Y, Yang Q,
mice. Blood 119(15):3550–3560 Wang Q, Cheng T (2009) ADAR1 is required for
Wang Z, Yuan H, Roth M, Stark JM, Bhatia R, Chen WY hematopoietic progenitor cell survival via RNA edit-
(2013) SIRT1 deacetylase promotes acquisition of ing. Proc Natl Acad Sci U S A 106(42):17763–17768
genetic mutations for drug resistance in CML cells. Yuan H, Wang Z, Li L, Zhang H, Modi H, Horne D,
Oncogene 32(5):589–598 Bhatia R, Chen W (2012) Activation of stress response
Warmuth M, Simon N, Mitina O, Mathes R, Fabbro D, gene SIRT1 by BCR-ABL promotes leukemogenesis.
Manley PW, Buchdunger E, Forster K, Moarefi I, Blood 119(8):1904–1914
Hallek M (2003) Dual-specific Src and Abl kinase Zhang B, Li M, McDonald T, Holyoake TL, Moon RT,
inhibitors, PP1 and CGP76030, inhibit growth and Campana D, Shultz L, Bhatia R (2013) Microenvironmental
survival of cells expressing imatinib mesylate-resistant protection of CML stem and progenitor cells from tyro-
Bcr-Abl kinases. Blood 101(2):664–672 sine kinase inhibitors through N-cadherin and Wnt-beta-
Warsch W, Walz C, Sexl V (2013) JAK of all trades: catenin signaling. Blood 121(10):1824–1838
JAK2-STAT5 as novel therapeutic targets in BCR- Zhang J, Adrian FJ, Jahnke W, Cowan-Jacob SW, Li AG,
ABL1+ chronic myeloid leukemia. Blood Iacob RE, Sim T, Powers J, Dierks C, Sun F, Guo G-R,
122(13):2167–2175 Ding Q, Okram B, Choi Y, Wojciechowski A, Deng X,
Weisberg E, Manley PW, Cowan-Jacob SW, Hochhaus A, Liu G, Fendrich G, Strauss A, Vajpai N, Grzesiek S,
Griffin JD (2007) Second generation inhibitors of Tuntland T, Liu Y, Bursulaya B, Azam M, Manley PW,
BCR-ABL for the treatment of imatinib-resistant Engen JR, Daley GQ, Warmuth M, Gray NS (2010)
chronic myeloid leukaemia. Nat Rev Cancer Targeting Bcr-Abl by combining allosteric with ATP-
7(5):345–356 binding-site inhibitors. Nature 463(7280):501–506
Wendel HG, de Stanchina E, Cepero E, Ray S, Emig M, Zhang P, Iwasaki-Arai J, Iwasaki H, Fenyus ML, Dayaram
Fridman JS, Veach DR, Bornmann WG, Clarkson B, T, Owens BM, Shigematsu H, Levantini E, Huettner CS,
McCombie WR, Kogan SC, Hochhaus A, Lowe SW Lekstrom-Himes JA, Akashi K, Tenen DG (2004)
(2006) Loss of p53 impedes the antileukemic response Enhancement of hematopoietic stem cell repopulating
to BCR-ABL inhibition. Proc Natl Acad Sci U S A capacity and self-renewal in the absence of the transcrip-
103(19):7444–7449 tion factor C/EBP alpha. Immunity 21(6):853–863
White D, Saunders V, Lyons AB, Branford S, Grigg A, To Zhang X, Subrahmanyam R, Wong R, Gross AW, Ren R
LB, Hughes T (2005) In vitro sensitivity to imatinib- (2001) The NH(2)-terminal coiled-coil domain and
induced inhibition of ABL kinase activity is predictive tyrosine 177 play important roles in induction of a
of molecular response in patients with de novo myeloproliferative disease in mice by Bcr-Abl. Mol
CML. Blood 106(7):2520–2526 Cell Biol 21(3):840–853
Wilson MB, Schreiner SJ, Choi HJ, Kamens J, Smithgall Zhao C, Blum J, Chen A, Kwon HY, Jung SH, Cook JM,
TE (2002) Selective pyrrolo-pyrimidine inhibitors Lagoo A, Reya T (2007) Loss of beta-catenin impairs
reveal a necessary role for Src family kinases in Bcr- the renewal of normal and CML stem cells in vivo.
Abl signal transduction and oncogenesis. Oncogene Cancer Cell 12(6):528–541
21(53):8075–8088 Zhao C, Chen A, Jamieson CH, Fereshteh M, Abrahamsson
Winter GE, Rix U, Carlson SM, Gleixner KV, Grebien F, A, Blum J, Kwon HY, Kim J, Chute JP, Rizzieri D,
Gridling M, Muller AC, Breitwieser FP, Bilban M, Munchhof M, VanArsdale T, Beachy PA, Reya T
Colinge J, Valent P, Bennett KL, White FM, Superti- (2009) Hedgehog signalling is essential for mainte-
Furga G (2012) Systems-pharmacology dissection of a nance of cancer stem cells in myeloid leukaemia.
drug synergy in imatinib-resistant CML. Nat Chem Nature 458(7239):776–779
Biol 8(11):905–912 Zhao JJ, Cheng H, Jia S, Wang L, Gjoerup OV, Mikami A,
Wylie A, Schoepfer J, Berellini G, Cai H, Caravatti G, Roberts TM (2006) The p110alpha isoform of PI3K is
Cotesta S, Dodd S, Donovan J, Erb B, Furet P, essential for proper growth factor signaling and onco-
Gangal G, Grotzfeld R, Hassan Q, Hood T, Iyer V, genic transformation. Proc Natl Acad Sci U S A
Jacob S, Jahnke W, Lombardo F, Loo A, Manley 103(44):16296–16300
PW, Marzinzik A, Palmer M, Pelle X, Salem B, Zhao X, Ghaffari S, Lodish H, Malashkevich VN, Kim PS
Sharma S, Thohan S, Zhu S, Keen N, Petruzzelli L, (2002) Structure of the Bcr-Abl oncoprotein oligomer-
Vanasse KG, Sellers WR (2014) ABL001, a potent ization domain. Nat Struct Biol 9(2):117–120

You might also like