• No results found

VU Research Portal

N/A
N/A
Protected

Academic year: 2021

Share "VU Research Portal"

Copied!
20
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

VU Research Portal

Target identification to selectively eradicate acute myeloid leukemia stem cells

Verhagen, H.J.M.P.

2018

document version

Publisher's PDF, also known as Version of record

Link to publication in VU Research Portal

citation for published version (APA)

Verhagen, H. J. M. P. (2018). Target identification to selectively eradicate acute myeloid leukemia stem cells.

General rights

Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights.

• Users may download and print one copy of any publication from the public portal for the purpose of private study or research. • You may not further distribute the material or use it for any profit-making activity or commercial gain

• You may freely distribute the URL identifying the publication in the public portal ?

Take down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

E-mail address:

(2)

C H A P T E R

1

(3)
(4)

1

G E N E R A L I N T RO D U C T I O N

Leukemia

Leukemia is a malignancy characterized by the accumulation of aberrant leukocytes in the bone

marrow and peripheral blood (Figure 1). This increase in aberrant white blood cells hampers

the function and development of normal hematopoiesis, causing symptoms related to anaemia,

thrombocytopenia and dysfunctional granulocytes, which is insurmountable fatal when patients

are left untreated. Mid 1800s, John Bennet and Rudolf Virchow established the fundament for

the recognition of leukemia and proposed that the aberrant presence of white blood cells in

the patient’s blood was not due to inflammation but rather a result of transformation of the complete

blood system

1

. As leukemia was gradually accepted as a distinct disease and many novel cases

were diagnosed, by studying morphology of the leukemic cells, a high degree of heterogeneity

among patients was observed. Although leukemia was generally considered to be a chronic disease,

the time between diagnosis and mortality for some patients was only a few weeks, therefore these

cases were referred to as acute leukemia. In the next decade, it became evident that the bone

marrow was the source for both the normal hematopoietic system and leukemia. In 1900, leukemia

was further categorised in myeloid and lymphoid neoplasms by detection of either myeloblasts

or lymphoblasts. This formed the basis of what we still recognize today as four primary types of

leukemia; chronic lymphoid leukemia (CLL), acute lymphoid leukemia (ALL), chronic myeloid

leukemia (CML) and acute myeloid leukemia (AML)

2

.

Diagnosis and classification of AML

AML is the most common form of the myeloid malignancies, with an average incidence of 3.7

cases per 100.000 persons each year in Europe

3

. Diagnosis of AML occurs by the detection of >20%

Figure 1. Illustration of a normal healthy and AML bone marrow aspirate. Healthy normal bone marrow (left panel) contains a wide variety of distinct differentiated myeloid cell types that all share a common ancestor, the myeloblast. The bone marrow of myeloid leukemia patients (right panel) shows accumulation of myeloblasts and the lack of differentiated cells which can result in symptoms like easy bleedings, pale skin, fatigue, weight

Normal hematopoiesis

Acute Myeloid Leukemia

(5)

1

myeloblasts in bone marrow or blood, or the presence of myeloid cells with genetic abnormalities

that include t(8;21), inv(16)/t(16;16), t(15;17)

4

. AML can be divided into various subgroups (according

to the WHO2016 classification) by integration of cell morphology, immunophenotyping, gene

expression, gene mutations and cytogenetics. Based on these features, AML cases can be classified

as either good, intermediate or adverse risk

5

. Elderly patients have in general a dismal prognosis

due to a poor condition and co-morbidities, as well as the presence of additional poor prognostic

factors like complex cytogenetics

6,7

. Chromosomal abnormalities are detected in ~55% of the AML

patients and have a strong impact on prognosis. These abnormalities include t(8;21), inv(16) and

t(15;17) which are good prognostic, and t(9;11), inv(3)(q21q26), t(3;3)(q21q26) del(7) or del(5) which

are poor prognostic. Patients lacking cytogenetic abnormalities are diagnosed as normal karyotype

(NK) and the presence of >3 abnormalities is referred to as a complex karyotype (CK). In addition to

chromosomal aberrations, somatic mutations are frequently found in AML. These include mutations

in genes like NPM1, FLT3, DNMT3A, CEBPA, MLL, RUNX1, TET2, WT1, KIT, RAS and p53. Some, such as

NMP1 and bi allelic CEBPA mutations are associated with a good prognosis, while others such as p53

mutations are associated with a poor prognosis. Moreover, high expression of genes such as ERG,

EVI-1, BAALC and MN1 are also linked to poor AML treatment outcome

8

.

Aberrant expression of genes, caused by translocations, somatic mutations, or epigenetic

changes can result in a variety of functional consequences driving the development of AML.

Mutations found in AML affect signalling pathways (59%), DNA methylation (44%), chromatin

modification (30%), and myeloid transcription factors (22%), and in lesser extend tumor suppressors,

the spliceosome complex and cohesins

9

. This aberrant gene expression can lead to aberrant

immunophenotypes, including expression of lymphoid plasma membrane markers on myeloid cells

that normally lack these markers. The aberrant expression of several of these markers, including

CD25,CD2,CD7 and CD56, correlate to a poor prognosis

10–12

. Other aberrant immunophenotypes are

the lack of a myeloid marker on myeloid cells, for instance the absence of the myeloid marker CD13

on CD33 expressing cells. Also aberrant expression of CD34 and MDR1 on myeloid cells is frequently

found for AML and both correlate to a poor prognosis

13,14

.

A well-defined prognostic marker for AML patients that predicts a poor response to

chemotherapy and a poor overall survival, is high expression of the Ecotropic Viral Integration

site-1 (EVI-1) oncogene. Despite treatment with chemotherapy and a hematopoietic stem cell

transplantation if eligible, >95% of AML patients with high EVI-1 expression do not survive two

years after diagnosis

15

. EVI-1 was initially discovered as a common retroviral integration site for

murine haematological malignancies

16

and enhanced expression of EVI-1 occurs in ∼10% of AML

patients

17

. High EVI-1 expression is sometimes driven by 3q26 abnormalities, as a result of aberrantly

positioned transcriptional enhancers

18

, however, in most cases EVI-1 is found to be overexpressed

due to an unknown cause

19

. In healthy tissue, EVI-1 is predominantly expressed in embryonic and

hematopoietic stem cells (HSCs) by acting as a multi-functional transcriptional regulator and is

essential for the proliferation and maintenance of hematopoietic stem cells (HSCs)

20,21

. Although

(6)

1

Chemotherapy response and resistance in AML

Although AML is a very heterogeneous disease, most patients are treated with a standard

combination chemotherapy that consists of cytarabine, and an anthracycline such as daunorubicin

and etoposide

5

. This treatment results in a complete remission (CR), defined as <5% myeloblasts, and

recovery of normal hematopoiesis, in approximately 55-85% of adult AML patients

22–25

. If eligible,

adult AML patients with an intermediate or adverse prognostic score are offered an allogenic

hematopoietic stem cell transplantation after achieving CR

4

. Despite these high initial CR rates, 50%

of AML patients experience a relapse within 5 years post diagnosis

26

. Recurrent disease is difficult to

treat and is the main reason for the in general poor overall survival rates (30%) of AML patients

27,26

.

Once the patient has developed a relapse its 3-year overall survival is 12% and 0% for intermediate

and adverse risk groups, respectively

28

. Refractory AML patients, patients that still present >5%

leukemia in the bone marrow after standard chemotherapy, can be offered alternative (salvage)

therapy. Commonly used salvage therapies include fludarabine, idarubcin and mitoxantrone, which

can potentially result in CR

5,29

.

The large number of clinical trials (>200), that are currently testing the efficacy of tyrosine

kinase -, epigenetic - and many other inhibitors as well as immunotherapeutic modalities (Clinical

trial register.eu), suggest increased numbers of potential alternative therapeutic options for AML

patients in the next decades. Some of these novel therapeutic strategies are predicted to act

on subgroups of AML patients, for instance, inhibition of Disrupter of Telomeric Silencing1-like

(DOT1L). This histone 3 lysine 79 (H3K79) methyltransferase is highly active in DNMT3a mutated as

well as MLL rearranged AML and inhibition of DOT1L demonstrated to induce apoptosis and tumor

reducing activity in vivo

30,31

. Other promising options are targeted therapy for specific subgroups of

AML patients with high FLT3 and SYK activity. Recently a dual inhibitor for both kinases in addition to

chemotherapy has demonstrated to improve survival of AML patients

32,33

. Other novel therapeutic

options are predicted to have a broader applicability, for instance anti-CD33 antibodies that

potentially eradicate all myeloid cells

34

.

Despite all the promising novel strategies, relapse is currently still the main cause for

the poor outcome of AML patients. Residual leukemic cells in the bone marrow of the patient after

chemotherapy treatment, referred to as minimal residual disease (MRD), initiate this relapse. MRD

can be detected using flow cytometric immunophenotyping and mutation analysis using molecular

techniques like PCR and next generation sequencing. High MRD levels strongly correlate with

increased relapse rates and measuring MRD is included in clinical studies as a novel risk parameter

to predict treatment outcome

35,36

.

Cytarabine, daunorubicin and etoposide are currently the most commonly used drugs to treat

AML, and mainly act on the DNA replication machinery by inhibition of DNA polymerase activity,

by cooperation into DNA during S-phase or by fixation of topoisomerase II to the DNA

37,38

. As

a result, DNA replication is terminated and double strand breaks are generated that cannot be

sufficiently repaired, hence leading to the induction of apoptosis. Recently it has been shown that

anthracyclines induce histone eviction and changes the transcriptional landscape

39

, indicating that

(7)

1

of escaping chemotherapy induced cell death are illustrated in Figure 2 and include (I) enhanced

DNA repair, (II) enhanced drug efflux, (III) enhanced survival signalling, (IV) target modification, (V)

apoptotic block and (VI) quiescence

40–42

.

Recently it has been shown that inactivation of the SWI/

SNF chromatin remodeling complex, that is involved in loading the Topoisomerase II to the DNA,

can also mediate resistance to chemotherapy

43,44

.

Figure 2. Mechanisms of resistance to chemotherapy in AML. (I) Genes that enhance DNA repair activity can prevent that cells undergo apoptosis44–47. High INPP4B expression is a biomarker for chemotherapy resistance

and is associated with a poor overall survival of AML patients. IBPP4B regulates ATM dependent DNA repair by activating BRCA1and RAD5145,46. Another gene that provides enhanced DNA repair is CHEK1. High CHEK1

expression is associated with a poor response to chemotherapy and when activated by ATR it still induces DNA replication upon cytarabine treatment47. (II) Chemotherapeutic agents that enter the cell can be efficiently

exported by drug transporters like multi-drug resistance 1 (MDR1), which prevent that cytostatic drugs reach the nucleus and induce DNA damage48. High MDR1 expression, as well as functional efflux capacity,

is observed in ~30% of the AML cases and correlates to a poor prognosis49. MDR1 is also frequently found

(8)

1

Leukemic and normal hematopoietic stem cells

AML is a clonal disease which is like normal haematopoiesis hierarchically organised

69

and originates

from mutations that occur in a multipotent progenitor or a HSC

70,71

(Figure 3). The development

of leukemia has been suggested to originate from environmental stress induced by chemicals and

radiation, but also genotoxic stress driven by the bone marrow microenvironment, both inducing

DNA aberrancies

72,73

. To date, the best accepted model describing the onset of AML is the “two

hit model” hypothesis

74,75

. This hypothesis states that the cell of origin needs at least two impaired

genes in order to transform into leukemia. The so called class I mutations can confer cells with

a proliferation advantage whereas the class II mutations direct a block in differentiation.

Within AML, tumor heterogeneity is observed since rare subpopulations are detected with

enhanced self-renewal capacity

69

. These cells are often referred to as leukemic stem cells (LSCs) and

are functionally defined by their long term tumor-initiating capacity in immunodeficient mice using

serial transplantations, or by their colony forming capacity using long term colony forming unit

(CFU) assays in vitro

69,76–78

. Although the cell of origin can be a LSC, these two are not necessarily

directly correlated to each other

70

. LSCs co-reside with HSCs in the bone marrow of AML patients

and are initially identified as having the CD34

+

CD38

-

immunophenotype, similar to HSCs

78

. Later,

LSCs showed to be more heterogeneous and to also reside in the CD34

-

population

79

.

CD34

+

CD38

-

LSCs are mainly quiescent

66

like HSCs

80

, however, recently a novel non-quiescent

LSC population was reported indicating the heterogeneity of the LSC pool

81

. The frequency of

CD34

+

CD38

-

LSCs at diagnosis and after chemotherapy treatment is predictive for overall survival

and relapse

82,83

. Importantly CD34

+

CD38

-

LSCs show decreased sensitivity to chemotherapy and

reside in the endosteal regions of the bone marrow

66,84

. The hypothesis that LSCs preferentially

therapy-induced drug resistance50. (III) Activated signalling routes can trigger survival responses that

overcome therapy induced cell death51–55. The best known example for AML is the FLT-3 receptor which is

frequently found highly expressed due to internal tandem duplications in the DNA (FLT3-ITDs). FLT3-ITDs are associated with a poorer survival of AML patients and result in increased resistance to cytarabine54,56. Activated

Insulin Growth Factor 1 receptor (IGF1R) can also promote AML cell survival during chemotherapy treatment by facilitating resistance to chemotherapy52. (IV) Targets of chemotherapy can either be downregulated in

expression levels or mutate during treatment, leading to therapy unresponsiveness40. Classically, etoposide and

anthracyclines like daunorubcin target topo isomerase II activity, leading to double strand breaks in the DNA and consequently apoptosis. However, in vitro studies revealed that expression of topoisomerase II is reduced in leukemic cells that are resistant to etoposide57. Furthermore, topoisomerase II expression levels in AML cells

have been shown to determine sensitivity to chemotherapy58. Moreover, a point mutation in topoisomerase II

demonstrated to decrease sensitivity to cytotoxic agents more than 50 fold In AML cell lines59. (V) The activation

or overexpression of anti-apoptotic proteins can overcome chemotherapy-induced cell death by blocking apoptosis60,61. BCL2 is a key protein in the regulation of apoptosis and its elevated expression is correlated to

a poor response to chemotherapy62. Also loss of the apoptotic regulators Puma and Noxa decreased sensitivity

to cytarabine and anthracyclines61. (VI) Quiescent (non-cycling) cells remain in the G0 cell cycle state and are

therefore less vulnerable for chemotherapy and have more time to repair potential DNA defects63–67. The loss of

(9)

1

survive chemotherapy is confirmed by a study that shows that LSC frequency is increased at relapse

compared to diagnosis

85

. However, conflicting data shows no increase in the number of LSCs after

chemotherapy treatment of AML in a xenograft mouse model

86

.

Various molecular mechanisms can contribute to the presence of stem cell-like features

(“stemness”), such as self-renewing capacity and chemotherapy resistance in AML. “Stemness”

might be imposed on AML cells by (I) genetic changes, (II) epigenetic changes and by the (III)

tumor microenvironment

87

. Genetic changes, caused by irreversible somatic mutations, can result

in distinct cell populations with different sensitivities to chemotherapy

88,89

. Epigenetically regulated

events and the tumor microenvironment might cause dynamically regulated AML heterogeneity in

the sense of chemotherapy sensitivity. Soluble factors secreted by mesenchymal stromal cells, like

CXCL12 (SDF-1) and JAG1, but also fatty acids derived from gonadal adipose tissue have been linked

to the survival of LSCs and the facilitation of chemotherapy resistance

90,91

. Besides the induction of

“stemness”, LSCs can also arise as a consequence of cell intrinsic changes

92,93

. Recently it has been

shown that epigenetic reprogramming of H3K27m3 modifications is important for LSC survival

94

.

The central role for LSCs in AML leukemogenesis, therapeutic responses and the initiation

of recurrent disease, drives the hypothesis that their elimination is key to improve AML therapy

(10)

1

outcome. The development of therapies that eradicate LSCs is challenging since these should spare

healthy HSCs that are needed to restore normal hematopoiesis after therapy. To develop such

specific anti-LSC therapies, the identification of genes that are differently expressed between HSCs,

LSCs and the AML bulk is crucial. Several studies have already explored transcriptional differences

between LSCs and non-LSCs from AML patients and differences between LSCs and normal HSCs

derived from healthy donors (Table 1). Eppert and colleagues generated gene expression profiles

(GEPs) comparing functionally defined LSCs to non-LSCs fractions and showed that high expression

of a LSC signature in AML correlates to a dismal outcome

95

. Furthermore, in this study cord

blood derived CD34

+

CD38

-

HSCs were compared with CD38

+

CD38

-

LSCs from AML bone marrow

and revealed that LSCs and HSCs share a core transcriptional program. Most of the studies that

are listed in Table 1, compared the expression profile of CD34

+

CD38

-

LSCs with that of CD34

+

CD38

-HSCs derived from healthy donors, however, since gene expression in -HSCs is influenced by both

AML cells

96

and the leukemic microenvironment

90

, comparing LSCs GEPs with that of HSCs is most

relevant when both stem cell fractions are derived from the same patient sample.

In the past decade, our lab identified biomarkers, that can distinguish LSCs from HSCs both

residing in the AML bone marrow. CD45RA

97

, CLL-1

98

and other lineage markers such as CD11b,

CD7 and CD56

99

can be present on LSCs, however, are always absent on HSCs. Also other labs have

identified such markers, including CD123

100

and Tim3

101

. Importantly, our lab has identified that HSCs

can be functionally discriminated from CD34

+

CD38

-

LSCs using aldehyde dehydrogenase activity

(ALDH)

102

. ALDH was initially identified as a cancer stem cell (CSC) marker in solid tumors and high

ALDH activity generally correlates to a poor prognosis

103

. In most FLT3-ITD and NPM1 mutated

AML cases, ALDH

bright

CD34

+

CD38

-

populations (HSCs) lack leukemia associated mutations and

immunophenotypic aberrancies, while ALDH

low

CD34

+

CD38

-

cells (LSCs) have these mutations and

immunophenotypic aberrancies

102

. Hoang and colleagues showed that 80 out of 104 AML cases

had low numbers (<1.9%) of CD34

+

ALDH

+

cells and were all annotated as HSCs since there was no

aberrancy present in mutation analysis, this in contrast to the matched CD34

+

ALDH

-

fractions

104

.

The other 24 cases had relative high numbers of CD34

+

ALDH

+

cells and were mostly LSCs indicating

that only CD34

+

ALDH

+

cells from AML with rare CD34

+

ALDH

+

fractions are suitable for HSC

discrimination. CD34

+

CD38

-

ALDH

bright

populations derived from ‘ALDH rare AML’ gave rise to

multi-lineage engraftment in contrast to CD34

+

CD38

-

ALDH

low

populations which gave myeloid leukemic

engraftment in a xenograft mouse model.

LSCs in AML patients BM are heterogonous in multiple ways. Accumulation of mutations in LSCs

and hence Darwinian selection during disease progression and after chemotherapy treatment, can

lead to a complete different clone that is present at relapse compared to diagnosis

88

. The same

holds true for aberrant immunophenotype which is in some AML patients different at MRD or

relapse than at diagnosis

105

. Importantly, cancer cells can show different sensitivities to therapy that

is regulated by non-genetically induced events, thereby adding another complexity to intratumoral

heterogeneity

106

. For example, small small populations of EGF receptor tolerant cells, with a stem

(11)

1

Insulin-like Growth Factor-1 (IGF-1) signaling

The IGF-1 receptor (IGF1R) is frequently found overexpressed in cancer cells and can regulate a wide

variety of cellular processes including survival, differentiation, proliferation, quiescence and therapy

resistance

114

. Although the role of the IGF-1 system has been extensively studied in solid tumors, less

is known about this pathway in hematological malignancies. In multiple myeloma (MM), IGF-1 has

been shown to stimulate the growth and survival of leukemic cells

115

and several anti-IGF1R therapies

were shown to inhibit MM proliferation

116

. In AML, expression of IGF-1 and the IGF1R was detected in

a panel of human AML blasts and cell lines and IGF-1 could promote the growth of AML blasts, AML

cell lines as well as AML progenitors

 

in vitro

117–119

.

 

For AML, inhibition of the IGF-1 system has been

performed by neutralizing anti-IGF1R antibodies and IGF1R tyrosine kinase inhibitors (NVP-AEW541

and NVP-ADW742) which both inhibited cell proliferation and induced apoptosis

51,120

. The effect of

IGF-1/IGF1R inhibition on the survival of AML LSCs is currently unknown. Anti-IGF-1/IGF1R strategies

seem to mimic the molecular effects caused by calorie restriction, which is known to decrease IGF-1

signaling. Combining calorie restriction (starvation) with chemotherapy treatment demonstrated

to specifically reduce the survival of cancer cells, while enhancing that of normal cells

121,122

. IGF1R

signaling can be regulated by expression of the components of IGF1R pathway, ligand bioavailability

and by receptor trafficking

123

. Moreover, IGF1R activity can be regulated by insulin-like growth

factor binding proteins (IGFBPs)

124

. To date, ten IGFBPs have been identified from which IGFBP1-6

are considered to have a function in inhibition of IGF1R signaling by binding IGF-1 and thus affecting

ligand bioavailability. IGFBP 7-10 do not bind with high affinity to the IGFs and might have additional

functions next to inhibition of IGF1R signaling

125

. An important role of IGFBP7 in IGF1R signaling is

Table 1. Gene expression profiles of HSCs, leukemic progenitors and LSCs. NBM = normal bone marrow, CB = Cord Blood, PB = peripheral blood.

Source Sorted fractions Reference

(12)

1

shown in multiple studies

126,127

. The loss of secreted IGFBP7 by tumor associated endothelial cells

contributes to the transformation of naïve tumor cells to chemotherapy resistant stem cells

127

.

Importantly, treatment of breast cancer and melanoma cells with purified recombinant human

IGFBP7 (rhIGFBP7) led to the induction of senescence and apoptosis and reduced tumor growth

in vivo

126,128

.

Retinoic acid based therapy for AML patients

AML cases that harbor a t(15;17) translocation, leading to expression of the fusion gene between

promyelocytic leukemia (PML) and the retinoic acid receptor alpha (RARA), are a separate entity of

AML cases, referred to as acute promyelocytic leukemia (APL). APL patients are treated with all- trans

retinoic acid (ATRA) differentiation therapy in combination with arsenic trioxide (ATO), resulting in

overall survival rates around 95%

129

. ATRA is a vitamin A derivative that belongs to the best known

examples of a successful targeted treatment and turned APL from a poor prognostic malignancy

into a curable disease

129,130

. In APL patients, expression of PML-RARA results in a differentiation

block by repression of genes that regulate differentiation

131

. RARs function as molecular switches

by recruiting N-Cor, SMRT and a histone deacetylase (HDAC), resulting in closed chromatin

and transcriptional silencing. Application of ATRA leads to conformational changes of RARs and

release of the transcriptional repressors and recruitment of transcriptional activators such as

the histone acetyl transferase p300. The increase of histone acetylation results in decompressed

chromatin leading to active gene transcription facilitating myeloid differentiation. Various clinical

trials have explored the applicability of ATRA for the non-APL AML subgroups. however, in general

ATRA treatment is not superior over chemotherapy treatment

132,133

. Previously, it has been shown

that inhibition of the H3K4 demethylase LSD1 (KDM1A), using tranylcypromine (TCP), can unlock

the ATRA differentiation pathway for non-APL AML indicating the importance of the epigenetic

landscape of AML cells in ATRA sensitivity

134

. The combination TCP and ATRA results in expression

of the myeloid differentiation marker CD11b in HL60 cells and reduced primary AML engraftment in

vivo, indicating a novel therapeutic opportunity to extend ATRA based therapy for AML. Recently,

several studies have explored the susceptibility to ATRA in smaller subgroups of AML patients and

reported a partial response to ATRA in vitro as well as in vivo for IDH1 mutant AML

135

, NPM1 mutant

AML

136,137

and FLT3-ITD

+

AML

138

. Altogether these data indicate that ATRA sensitivity is determined

on a transcriptional or epigenetic level and treatment of AML patients might work for well-defined

subgroups of AML patients.

O U T L I N E O F T H I S T H E S I S

(13)

1

enhancing insulin-like growth factor binding protein 7 (IGFBP7), by overexpression or addition of

recombinant human IGFBP7 (rhIGFBP7), induces apoptosis and enhances chemotherapy sensitivity,

similar as the inhibition of IGF1R activity by a tyrosine kinase inhibitor. The tumor reducing effect of

IGFBP7 is at least partly explained by an IGF1R independent mechanism. Furthermore we report that

low IGFBP7 expression levels are more indicative for a poor overall survival compared to high IGF-1

or high IGF1R expression levels in AML patients.

Since LSCs play a central role in the treatment outcome of AML, the eradication of these cells

at diagnosis, while sparing HSCs, is of critical importance for the development of novel therapeutic

strategies. In chapter 3 we have studied GEPs of LSCs relative to HSCs that are both derived from

the bone marrow of an AML patient. Moreover, we compared GEPs of CD34

+

CD38

-

leukemic cells

with those of leukemic CD34

+

CD38

+

cells and identified IGFBP7 as differently expressed. IGFBP7

expression levels correlate with chemotherapy sensitivity and self-renewal capacity. Importantly,

we show that rhIGFBP7 induces differentiation and apoptosis in primary AML cells in vitro and

eradicates LSCs while sparing HSCs. Treatment of AML with rhIGFBP7 in patient derived xenograft

models reduces engraftment and LSC activity, this in contrast to HSCs derived from healthy BM

which remain unaffected by rhIGFBP7 treatment.

In chapter 4 we have studied anthracycline resistance in a subpopulation of myeloid leukemic

cells residing within a sensitive leukemia cell line. This small subpopulation of leukemic cells

possesses stem cell-like features and a reversible drug tolerant state. Using GEP we identified

KDM6B as a potential regulator of this drug tolerant state. Inhibition of KDM6B activity using GSK-J4

resulted in the elimination of the drug-tolerant cells, indicating that KDM6B inhibition might be

a potential successful therapeutic strategy to eliminate stem cell-like chemotherapy resistant

AML cells.

In chapter 5 we explored the effect of ATRA on AML patients that have high expression of

EVI-1. Here we show that part of the EVI-1 positive AML cases respond to ATRA by induction of

differentiation and in some cases induction of apoptosis in vitro. Importantly, we show that EVI-1

positive AML can be successfully treated with ATRA in vivo, suggesting that treatment with ATRA

might be a promising strategy for EVI-1 positive AML patients.

In chapter 6 we have reviewed the current knowledge on susceptibility of non-APL AML for

ATRA. Non-APL AML cases are less sensitive for ATRA therapy as compared to APL, however,

epigenetic reprogramming as well as the identification of biomarkers that predict ATRA response

can potentially change this dogma.

(14)

1

R E F E R E N C E S

1. Piller GJ. Leukaemia - A brief historical review from ancient times to 1950. Br. J. Haematol. 112, 282–292 (2001).

2. Arber DA, Orazi A, Hasserjian R, Borowitz MJ, Beau MM Le,et al. The 2016 revision to the World Health Organization classi fi cation of myeloid neoplasms and acute leukemia. Blood 127, 2391–2406 (2016).

3. Visser O, Trama A, Maynadié M, Stiller C, Marcos-Gragera R,et al. Incidence, survival and prevalence of myeloid malignancies in Europe. Eur. J. Cancer 48, 3257–3266 (2012).

4. Döhner H, Estey EHE, Amadori S, Appelbaum FRFR, Büchner T,et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood 115, 453–474 (2010).

5. Döhner H, Estey E & Grimwade D. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. 129, 424–448 (2017).

6. Juliusson G, Antunovic P, Derolf Å, Lehmann S, Möllgård L,et al. Age and acute myeloid leukemia: real world data on decision to treat and outcomes from the Swedish Acute Leukemia Registry Age and acute myeloid leukemia: real world data on decision to treat and outcomes from the Swedish Acute Leukemia Registry. Blood 113, 4179–4187 (2014). 7. Østgård LSG, Nørgaard JM, Sengeløv H,

Severinsen M, Friis LS,et al. Comorbidity and performance status in acute myeloid leukemia patients: a nation-wide population-based cohort study. Leukemia 29, 548–55 (2015). 8. Papaemmanuil E, Gerstung M, Bullinger L,

Gaidzik VI, Paschka P,et al. Genomic Classification and Prognosis in Acute Myeloid Leukemia. N. Engl. J. Med. 374, 2209–2221 (2016).

9. Khwaja A, Bjorkholm M, Gale RE, Levine RL, Jordan CT,et al. Acute myeloid leukaemia. Nat. Rev. Dis. Prim. 2, 16010 (2016).

10. Fujiwara S, Muroi K, Yamamoto C, Hatano K, Okazuka K,et al. CD25 as an adverse prognostic factor in elderly patients with acute myeloid leukemia. Hematology 0, 1–7 (2017).

11. Perea G, Domingo A, Villamor N, Palacios C, Juncà J,et al. Adverse prognostic impact of CD36 and

CD2 expression in adult de novo acute myeloid leukemia patients. Leuk. Res. 29, 1109–1116 (2005). 12. Suzuki R, Ohtake S, Takeuchi J, Nagai M, Kodera Y,et al. The clinical characteristics of CD7+ CD56+ acute myeloid leukemias other than M0. Int. J. Hematol. 91, 303–309 (2010).

13. Zeijlemaker W, Kelder A, Wouters R, Valk PJM, Witte BI,et al. Absence of leukaemic CD34+ cells in acute myeloid leukaemia is of high prognostic value: A longstanding controversy deciphered. Br. J. Haematol. 171, 227–238 (2015).

14. Van Der Pol M, Feller N, Ossenkoppele GJ, Weijers G, Westra A,et al. Minimal residual disease in acute myeloid leukemia is predicted by P-Glycoprotein activity but not by multidrug resistance protein activity at diagnosis. Leukemia 17, 1674–1677 (2003).

15. Doorn-khosrovani SBVW Van, Erpelinck C, Putten WLJ Van, Valk PJM, Luytgaarde SVDP De,et al. High EVI1 expression predicts poor survival in acute myeloid leukemia : a study of 319 de novo AML patients. 101, 837–845 (2003). 16. Mucenski ML, Taylor BA, Ihle JN, Hartley JW,

Morse HC,et al. Identification of a common ecotropic viral integration site, Evi-1, in the DNA of AKXD murine myeloid tumors. Mol. Cell. Biol. 8, 301–8 (1988).

17. Lugthart S, Van Drunen E, Van Norden Y, Van Hoven A, Erpelinck CAJ,et al. High EVI1 levels predict adverse outcome in acute myeloid leukemia: Prevalence of EVI1 overexpression and chromosome 3q26 abnormalities underestimated. Blood 111, 4329–4337 (2008). 18. Gröschel S, Sanders MA, Hoogenboezem R, De

Wit E, Bouwman BAM,et al. A single oncogenic enhancer rearrangement causes concomitant EVI1 and GATA2 deregulation in Leukemia. Cell 157, 369–381 (2014).

19. Gröschel S, Lugthart S, Schlenk RF, Valk PJM, Eiwen K,et al. High EVI1 Expression Predicts Outcome in Younger Adult Patients with Acute Myeloid Leukemia and Is Associated with Distinct Cytogenetic Abnormalities Cytogenetics and Molecular Analyses Routine cytogenetics analyses included chromosome banding. Reverse t. J. Clin. Oncol. 28, 1–18 (2010).

(15)

1

Hematopoietic Stem Cells and Transformed Leukemic Cells. Cell Stem Cell 3, 207–220 (2008). 21. Hinai AA & Valk PJM. Review: Aberrant EVI1

expression in acute myeloid leukaemia. Br. J. Haematol. 172, 870–878 (2016).

22. Cheson BD, Cassileth PA, Head DR, Schiffer CA, Bennett JM,et al. Report of the national cancer institute-sponsored workshop on definitions of diagnosis and response in acute myeloid leukemia. J. Clin. Oncol. 8, 813–819 (1990). 23. Cheson BD, Bennett JM, Kopecky KJ, Büchner T,

Willman CL,et al. Revised Recommendations of the International Working Group for diagnosis, standardization of response criteria, treatment outcomes, and reporting standards for therapeutic trials in acute myeloid leukemia. J. Clin. Oncol. 21, 4642–4649 (2003).

24. Löwenberg B, Pabst T, Vellenga E, van Putten W, Schouten HC,et al. Cytarabine dose for acute myeloid leukemia. N. Engl. J. Med. 364, 1027–1036 (2011). 25. Fernandez HF, Sun Z, Yao X, Litzow MR, Luger

SM,et al. Anthracycline Dose Intensification in Acute Myeloid Leukemia. 1249–1259 (2009). 26. Verma D, Kantarjian H, Faderl S, O’Brien S, Pierce

S,et al. Late relapses in acute myeloid leukemia: analysis of characteristics and outcome. Leuk. Lymphoma 51, 778–82 (2010).

27. Estey E & Gale RP. Acute myeloid leukemia therapy and the chosen people. Leukemia 1–3 (2016). 28. Weltermann a, Fonatsch C, Haas O a, Greinix

HT, Kahls P,et al. Impact of cytogenetics on the prognosis of adults with de novo AML in first relapse. Leuk. Off. J. Leuk. Soc. Am. Leuk. Res. Fund, U.K 18, 293–302 (2004).

29. Thol F, Schlenk RF, Heuser M & Ganser A. How I treat refractory and early relapsed acute myeloid leukemia. Blood 126, 319–327 (2015). 30. Rau RE, Rodriguez B, Luo M, Jeong M, Rosen A,et

al. DOT1L as a therapeutic target for the treatment of DNMT3A-mutant acute myeloid leukemia. Blood 128, blood-2015-11-684225 (2016).

31. Daigle SR, Olhava EJ, Therkelsen CA, Basavapathruni A, Jin L,et al. Potent inhibition of DOT1L as treatment of MLL-fusion leukemia. Blood 122, 1017–1025 (2013).

32. Weisberg EL, Puissant A, Stone R, Sattler M, Buhrlage SJ,et al. Characterization of midostaurin as a dual inhibitor of FLT3 and SYK and potentiation of FLT3 inhibition against

FLT3-ITD-driven leukemia harboring activated SYK kinase. 8, 52026–52044 (2017).

33. Stone RM, Mandrekar SJ, Sanford BL, Laumann K, Geyer S,et al. Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a FLT3 Mutation. N. Engl. J. Med. 377, 454–464 (2017). 34. Laing AA, Harrison CJ, Gibson BES & Keeshan K.

Unlocking the potential of anti-CD33 therapy in adult and childhood acute myeloid leukemia. Exp. Hematol. (2017).

35. Terwijn M, Kelder A, Huijgens PC, Dr??ger AM, Oussoren YJM,et al. High prognostic impact of flow cytometric minimal residual disease detection in acute myeloid leukemia: Data from the HOVON/SAKK AML 42A study. J. Clin. Oncol. 31, 3889–3897 (2013).

36. Ossenkoppele G & Schuurhuis GJ. MRD in AML: does it already guide therapy decision-making? ASH Educ. Progr. B. 2016, 356–365 (2016). 37. Galmarini CM, Mackey JR & Dumontet C. Nucleoside

analogues: mechanisms of drug resistance and reversal strategies. Leukemia 15, 875–890 (2001). 38. Nitiss J. Targeting DNA topoisomerase II in cancer

chemotherapy. Nat. Rev. Cancer 9, 338–350 (2009). 39. Pang B, Qiao X, Janssen L, Velds A, Groothuis

T,et al. Drug-induced histone eviction from open chromatin contributes to the chemotherapeutic effects of doxorubicin. Nat. Commun. 4, 1908 (2013).

40. Wijdeven RH, Pang B, Assaraf YG & Neefjes J. Old drugs , novel ways out : Drug resistance toward cytotoxic chemotherapeutics. Drug Resist. Updat. 28, 65–81 (2016).

41. Borst P. Cancer drug pan-resistance: pumps, cancer stem cells, quiescence, epithelial to mesenchymal transition, blocked cell death pathways, persisters or what? Open Biol 2, 120066 (2012).

42. Colak S & Medema JP. Cancer stem cells - Important players in tumor therapy resistance. FEBS J. 281, 4779–4791 (2014).

43. Hohmann A & Vakoc C. NIH Public Access. Trends Genet. 30, 356–363 (2014).

44. Wijdeven RH, Pang B, Van Der Zanden SY, Qiao X, Blomen V,et al. Genome-wide identification and characterization of novel factors conferring resistance to topoisomerase II poisons in cancer. Cancer Res. 75, 4176–4187 (2015). 45. Wang P, Ma D, Wang J, Fang Q, Gao R,et al.

(16)

1

resistance to chemotherapy in acute myeloid leukemia. Tumour Biol. 12513–12523 (2016). 46. Rijal S, Fleming S, Cummings N, Rynkiewicz

NK, Ooms LM,et al. Inositol polyphosphate 4-phosphatase II (INPP4B) is associated with chemoresistance and poor outcome in AML. Blood 125, 2815–2824 (2015).

47. David L, FernandezVidal A, Bertoli S, Grgurevic S, Lepage B,et al. CHK1 as a therapeutic target to bypass chemoresistance in AML. Sci. Signal. 9, 1–13 (2016). 48. Shaffer BC, Gillet JP, Patel C, Baer MR, Bates

SE,et al. Drug resistance: Still a daunting challenge to the successful treatment of AML. Drug Resist. Updat. 15, 62–69 (2012).

49. Leith CP, Kopecky KJ, Chen I, Eijdems L, Slovak ML,et al. Frequency and Clinical Significance of the Expression of the Multidrug Resistance Proteins MDR1/P-Glycoprotein, MRP1, and LRP in Acute Myeloid Leukemia. A Southwest Oncology Group Study. 94, 1086–1099 (1999). 50. Pisco AO, Brock A, Zhou J, Moor A, Mojtahedi M,et al.

Non-Darwinian dynamics in therapy-induced cancer drug resistance. Nat. Commun. 4, 2467 (2013). 51. Tazzari PL, Tabellini G, Bortul R, Papa V,

Evangelisti C,et al. The insulin-like growth factor-I receptor kinase inhibitor NVP-AEW541 induces apoptosis in acute myeloid leukemia cells exhibiting autocrine insulin-like growth factor-I secretion. Leukemia 21, 886–96 (2007). 52. Doepfner KT, Spertini O & Arcaro a. Autocrine

insulin-like growth factor-I signaling promotes growth and survival of human acute myeloid leukemia cells via the phosphoinositide 3-kinase/Akt pathway. Leuk. Off. J. Leuk. Soc. Am. Leuk. Res. Fund, U.K 21, 1921–1930 (2007). 53. Hong CC, Lay JD, Huang JS, Cheng AL, Tang

JL,et al. Receptor tyrosine kinase AXL is induced by chemotherapy drugs and overexpression of AXL confers drug resistance in acute myeloid leukemia. Cancer Lett. 268, 314–324 (2008). 54. Jin G, Matsushita H, Asai S, Tsukamoto H, Ono

R,et al. FLT3-ITD induces ara-C resistance in myeloid leukemic cells through the repression of the ENT1 expression. Biochem. Biophys. Res. Commun. 390, 1001–1006 (2009).

55. Morita K, Masamoto Y, Kataoka K, Koya J, Kagoya Y,et al. BAALC potentiates oncogenic ERK pathway through interactions with MEKK1 and KLF4. Leukemia 29, 2248–56 (2015).

56. Gorin N-C, Labopin M, Meloni G, Pigneux A, Esteve J,et al. Impact of FLT3 ITD/NPM1 mutation status in adult patients with acute myelocytic leukemia autografted in first remission. Haematologica 98, 12–14 (2013). 57. Ritke M k., Allan WP, Fattman C, Gunduz NN

& Yalowich JC. Reduced Phosphorylation of Topoisomerase Resistant Human Leukemia K562 Cells. Mol. Pharmacol. 46, 58–66 (1994). 58. Burgess DJ, Doles J, Zender L, Xue W, Ma B,et al.

Topoisomerase levels determine chemotherapy response in vitro and in vivo. Proc. Natl. Acad. Sci. U. S. A. 105, 9053–9058 (2008).

59. Hinds M, Deisseroth K, Mayes J, Altschuler E, Jansen R,et al. Identification of a point mutation in the topoisomerase II gene from a human leukemia cell line containing an amsacrine-resistant form of topoisomerase II. Medicine (Baltimore). 4729–4731 (1991).

60. Lagadinou ED, Sach A, Callahan K, Rossi RM, Neering SJ,et al. BCL-2 inhibition targets oxidative phosphorylation and selectively eradicates quiescent human leukemia stem cells. Cell Stem Cell 12, 329–341 (2013).

61. Bilardi RA, Anstee NS, Glaser SP, Robati M, Vandenberg CJ,et al. Impact of loss of BH3-only proteins on the development and treatment of MLL-fusion gene-driven AML in mice. Cell Death Dis. 7, e2351 (2016).

62. Campos L, Rouault JP, Sabido O, Oriol P, Roubi N,et al. High expression of bcl-2 protein in acute myeloid leukemia cells is associated with poor response to chemotherapy. Blood 81, 3091–3096 (1993). 63. Raffel S & Trumpp A. MiR-126 Drives Quiescence

and Self-Renewal in Leukemic Stem Cells. Cancer Cell 29, 133–135 (2016).

64. Ehninger A, Boch T, Medyouf H, Müdder K, Orend G,et al. Loss of SPARC protects hematopoietic stem cells from chemotherapy toxicity by accelarting their return to quiescene. Blood 123, 4054–4063 (2014).

65. Cogle CR, Bosse RC, Brewer T, Migdady Y, Shirzad R,et al. Acute myeloid leukemia in the vascular niche. Cancer Lett. 380, 552–560 (2016). 66. Ishikawa F, Yoshida S, Saito Y, Hijikata A,

(17)

1

67. Essers MAG & Trumpp A. Targeting leukemic stem cells by breaking their dormancy. Mol. Oncol. 4, 443–450 (2010).

68. Lechman ER, Gentner B, Ng SWK, Schoof EM, van Galen P,et al. MiR-126 Regulates Distinct Self-Renewal Outcomes in Normal and Malignant Hematopoietic Stem Cells. Cancer Cell 29, 214–228 (2016).

69. Bonnet D & Dick J. human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopeietic cell. Nat. Med. 3, 730–737 (1997).

70. Visvader JE. Cells of origin in cancer. (2011). 71. Shlush LI, Zandi S, Mitchell A, Chen WC, Brandwein

JM,et al. Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature (2014).

72. Zambetti NA, Ping Z, Chen S, Kenswil KJG, Mylona MA,et al. Mesenchymal Inflammation Drives Genotoxic Stress in Hematopoietic Stem Cells and Predicts Disease Evolution in Human Pre-leukemia. Cell Stem Cell 19, 613–627 (2016). 73. Matsuo M, Iwanaga M, Kondo H, Soda M,

Jo T,et al. Clinical features and prognosis of patients with myelodysplastic syndromes who were exposed to atomic bomb radiation in Nagasaki. Cancer Sci. 107, 1484–1491 (2016). 74. Kelly LM & Gilliland DG. Genetics of myeloid

leukemias. Annu. Rev. Genomics Hum. Genet. 3, 179–198 (2002).

75. Shih AH, Abdel-Wahab O, Patel JP & Levine RL. The role of mutations in epigenetic regulators in myeloid malignancies. Nat Rev Cancer 12, 599–612 (2012).

76. Udomsakdi C, Lansdorp PM, Hogge DE, Reid DS, Eaves a C,et al. Characterization of primitive hematopoietic cells in normal human peripheral blood. Blood 80, 2513–2521 (1992). 77. Sutherland H, Blair A & Zapf RW. Characterization

of a hierarchy in human acute myeloid leukemia progenitor cells . Blood 87, 4754–4761 (1996). 78. Lapidot T, Sirard C, Vormoor J, Murdoch B,

Hoang T,et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 367, 645–648 (1994). 79. Taussig DC, Vargaftig J, Miraki-moud F,

Griessinger E, Sharrock K,et al. Leukemia initiating cells from some acute myeloid leukemia patients with mutated nucleophosmin

reside in the CD34 - fraction Leukemia initiating cells from some acute myeloid leukemia patients with mutated nucleophosmin reside in the CD34 fraction. 115, 1976–1985 (2010). 80. Laurenti E, Frelin C, Xie S, Ferrari R, Dunant

CF,et al. CDK6 levels regulate quiescence exit in human hematopoietic stem cells. Cell Stem Cell 16, 302–313 (2015).

81. Iwasaki M, Liedtke M, Gentles AJ & Cleary ML. CD93 Marks a Non-Quiescent Human Leukemia Stem Cell Population and Is Required for Development of MLL-Rearranged Acute Myeloid Leukemia. Cell Stem Cell 17, 412–421 (2015). 82. Rhenen A Van, Feller N, Kelder A, Westra AH,

Rombouts E,et al. High Stem Cell Frequency in Acute Myeloid Leukemia at Diagnosis Predicts High Minimal Residual Disease and Poor Survival. 11, 6520–6527 (2005).

83. Terwijn M, Zeijlemaker W, Kelder A, Rutten AP, Snel AN,et al. Leukemic stem cell frequency: A strong biomarker for clinical outcome in acute myeloid leukemia. PLoS One 9, 7–9 (2014). 84. Costello R t, Mallet F, Gaugler B, Sainty D,

Arnoulet C,et al. Human Acute Myeloid LeukemiaCD34+CD38- Progenitor Cells Have Decreased Sensitivity to Chemotherapy and Fas-induced Apoptosis, Reduced Immunogenicity , and Impaired Dendritic Cell Re. 4403–4411 (2000). 85. Ho T-C, LaMere M, Stevens BM, Ashton JM,

Myers JR,et al. Evolution of acute myelogenous leukemia stem cell properties following treatment and progression. Blood 128, blood-2016-02-695312 (2016).

86. Farge T, saland E, de Toni F, Aroua N, Hosseini M,et al. Chemotherapy Resistant Human Acute Myeloid Leukemia Cells are Not Enriched for Leukemic Stem Cells but Require Oxidative Metabolism. Cancer Discov. CD-16-0441 (2017). 87. Kreso A & Dick JE. Evolution of the cancer stem

cell model. Cell Stem Cell 14, 275–291 (2014). 88. Ding L, Ley TJ, Larson DE, Miller CA, Koboldt

DC,et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature 481, 506–510 (2012). 89. Bachas C, Schuurhuis GJ, Assaraf YG,

(18)

1

diagnosis in the development of relapse. Leukemia 26, 1313–1320 (2012).

90. Kim JA, Shim JS, Lee GY, Yim HW, Kim TM,et al. Microenvironmental remodeling as a parameter and prognostic factor of heterogeneous leukemogenesis in acute myelogenous leukemia. Cancer Res. 75, 2222–2231 (2015). 91. Ye H, Adane B, Khan N, Sullivan T, Minhajuddin

M,et al. Leukemic Stem Cells Evade Chemotherapy by Metabolic Adaptation to an Adipose Tissue Niche. Cell Stem Cell 1–15 (2015).

92. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T,et al. Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors. Cell 131, 861–872 (2007).

93. Chaffer CL, Brueckmann I, Scheel C, Kaestli AJ, Wiggins PA,et al. Normal and neoplastic nonstem cells can spontaneously convert to a stem-like state. Proc. Natl. Acad. Sci. U. S. A. 108, 7950–7955 (2011). 94. Scott MT, Korfi K, Saffrey P, Hopcroft LEM, Kinstrie

R,et al. Epigenetic reprogramming sensitizes CML stem cells to combined EZH2 and tyrosine kinase inhibition. Cancer Discov. 6, 1248–1257 (2016). 95. Eppert K, Takenaka K, Lechman ER, Waldron

L, Nilsson B,et al. Stem cell gene expression programs influence clinical outcome in human leukemia. Nat. Med. 17, 1086–1093 (2011). 96. Hornick NI, Doron B, Abdelhamed S, Huan J,

Harrington CA,et al. AML suppresses hematopoiesis by releasing exosomes that contain microRNAs targeting c-MYB. Sci. Signal. 9, 1–10 (2016).

97. Kersten B, Valkering M, Wouters R, van Amerongen R, Hanekamp D,et al. CD45RA, a specific marker for leukaemia stem cell sub-populations in acute myeloid leukaemia. Br. J. Haematol. 173, 219–235 (2016).

98. Van Rhenen A, Dongen GAMS Van, Kelder A, Rombouts EJ, Feller N,et al. discrimination between normal and leukemic stem cells The novel AML stem cell – associated antigen CLL-1 aids in discrimination between normal and leukemic stem cells. Blood 110, 2659–2666 (2007). 99. Zeijlemaker W, Kelder A, Oussoren-Brockhoff

YJM, Scholten WJ, Snel AN,et al. A simple one-tube assay for immunophenotypical quantification of leukemic stem cells in acute myeloid leukemia. Leukemia 1–8 (2015).

100. Jordan CT, Upchurch D, Szilvassy SJ, Guzman ML, Howard DS,et al. The interleukin-3 receptor alpha chain is a unique marker for human acute myelogenous leukemia stem cells. Leukemia 14, 1777–1784 (2000).

101. Jan M, Chao MP, Cha AC, Alizadeh AA, Gentles AJ,et al. Prospective separation of normal and leukemic stem cells based on differential expression of TIM3, a human acute myeloid leukemia stem cell marker. Proc. Natl. Acad. Sci. U. S. A. 108, 5009–14 (2011).

102. Schuurhuis GJ, Meel MH, Wouters F, Min LA, Terwijn M,et al. Normal hematopoietic stem cells within the AML bone marrow have a distinct and higher ALDH activity level than co-existing leukemic stem cells. PLoS One 8, (2013). 103. Tomita H, Tanaka K, Tanaka T & Hara A.

Aldehyde dehydrogenase 1A1 in stem cells and cancer. Oncotarget 7, 11018–32 (2016). 104. Hoang VT, Buss EC, Wang W, Hoffmann I, Raffel S,et

al. The rarity of ALDH+ cells is the key to separation of normal versus leukemia stem cells by ALDH activity in AML patients. Int. J. Cancer 137, 525–536 (2015). 105. Zeijlemaker W, Gratama JW & Schuurhuis GJ.

Tumor heterogeneity makes AML a ‘moving target’ for detection of residual disease. Cytom. Part B - Clin. Cytom. 86, 3–14 (2014).

106. Sharma S V., Lee DY, Li B, Quinlan MP, Takahashi F,et al. A Chromatin-Mediated Reversible Drug-Tolerant State in Cancer Cell Subpopulations. Cell 141, 69–80 (2010).

107. Gal H, Amariglio N, Trakhtenbrot L, Jacob-Hirsh J, Margalit O,et al. Gene expression profiles of AML derived stem cells; similarity to hematopoietic stem cells. Leuk. Off. J. Leuk. Soc. Am. Leuk. Res. Fund, U.K 20, 2147–2154 (2006).

108. Gentles AJ, Plevritis SK & Page P. Association of a Leukemic Stem Cell Gene in Acute Myeloid Leukemia. JAMA 304, (2012).

109. Saito Y, Kitamura H, Hijikata A, Tomizawa-Murasawa M, Tanaka S,et al. Identification of therapeutic targets for quiescent, chemotherapy-resistant human leukemia stem cells. Sci. Transl. Med. 2, 17ra9 (2010).

(19)

1

111. de Jonge HJM, Woolthuis CM, Vos a Z, Mulder a, van den Berg E,et al. Gene expression profiling in the leukemic stem cell-enriched CD34+ fraction identifies target genes that predict prognosis in normal karyotype AML. Leukemia 25, 1825–1833 (2011).

112. Ng SWK, Mitchell A, Kennedy JA, Chen WC, McLeod J,et al. A 17-gene stemness score for rapid determination of risk in acute leukaemia. Nature 540, 433–437 (2016).

113. Laurenti E, Doulatov S, Zandi S, Plumb I, Chen J,et al. Pub Med Central CANADA. Nat. Immunol. 14, 756–763 (2013).

114. Pollak M. Insulin and insulin-like growth factor signalling in neoplasia. Nat Rev Cancer 8, 915–928 (2008).

115. Ge N-L & Rudikoff S. Insulin-like growth factor I is a dual effector of multiple myeloma cell growth. Blood 96, 2856–2861 (2000).

116. Mitsiades CS, Mitsiades NS, McMullan CJ, Poulaki V, Shringarpure R,et al. Inhibition of the insulin-like growth factor receptor-1 tyrosine kinase activity as a therapeutic strategy for multiple myeloma, other hematologic malignancies, and solid tumors. Cancer Cell 5, 221–230 (2004). 117. Estrov BZ, Meir R, Barak Y, Zaizov R & Zadik Z.

Human Growth Hormone and Insulin-like Growth Factor-i Enhance the Proliferation of Human Leukemic Blasts. J. Clin. Oncol. 9, 394–399 (1991). 118. Frostad S, Bjerknes R, Hervig T, Nesthus

I, Olweus J BØ. Malignancy: Insulin-like Growth Factor-1 (IGF-1) is a Costimulator of the Expansion of Lineage Committed Cells Derived from Peripheral Blood Mobilized CD34+ Cells in Multiple Myeloma Patients. Hematology 4, 217–229 (1999).

119. Zadik Z, Estrov Z, Karov Y, Hahn T & Barak Y. The effect of growth hormone and IGF-I on clonogenic growth of hematopoietic cells in leukemic patients during active disease and during remission--a preliminary report. J Pediatr Endocrinol 6, 79–83 (1993).

120. Chapuis N, Tamburini J, Cornillet-Lefebvre P, Gillot L, Bardet V,et al. Autocrine IGF-1/IGF-1R signaling is responsible for constitutive PI3K/ Akt activation in acute myeloid leukemia: Therapeutic value of neutralizing anti-IGF-1R antibody. Haematologica 95, 415–423 (2010).

121. Raffaghello L, Lee C, Safdie FM, Wei M, Madia F,et al. Starvation-dependent differential stress resistance protects normal but not cancer cells against high-dose chemotherapy. Proc. Natl. Acad. Sci. 105, 8215–8220 (2008).

122. Lee C, Safdie FM, Raffaghello L, Wei M, Madia F,et al. Reduced levels of IGF-I mediate differential protection of normal and cancer cells in response to fasting and improve chemotherapeutic index. Cancer Res. 70, 1564–1572 (2010).

123. Pollak M. The insulin and insulin-like growth factor receptor family in neoplasia: an update. Nat. Rev. Cancer 12, 159–69 (2012).

124. Baxter RC. IGF binding proteins in cancer: mechanistic and clinical insights. Nat Rev Cancer 14, 329–341 (2014).

125. Baxter RC, Binoux MA, Clemmons DR, Conover CA, Drop SLS,et al. Recommendations for Nomenclature of the Insulin-Like. J. Clin. Endocrinol. Metab. 83, 12986 (1998).

126. Benatar T, Yang W, Amemiya Y, Evdokimova V, Kahn H,et al. IGFBP7 reduces breast tumor growth by induction of senescence and apoptosis pathways. Breast Cancer Res. Treat. 133, 563–573 (2012). 127. Cao Z, Scandura JM, Inghirami GG, Shido K,

Ding B-S,et al. Molecular Checkpoint Decisions Made by Subverted Vascular Niche Transform Indolent Tumor Cells into Chemoresistant Cancer Stem Cells. Cancer Cell 1–17 (2016). 128. Wajapeyee N, Serra RW, Zhu X, Mahalingam M &

Green MR. Oncogenic BRAF Induces Senescence and Apoptosis through Pathways Mediated by the Secreted Protein IGFBP7. Cell 132, 363–374 (2008). 129. Lo-Coco F, Avvisati G, Vignetti M, Thiede C,

Orlando SM,et al. Retinoic acid and arsenic trioxide for acute promyelocytic leukemia. N Engl J Med 369, 111–121 (2013).

130. Fenaux BP, Cbcile M, Deley L, Castaigne S, Archimbaud E,et al. Effect of All Transretinoic Acid in Newly Diagnosed Acute Promyelocytic. 3241–3250 (2014).

131. Schenk T, Stengel S & Zelent A. Unlocking the potential of retinoic acid in anticancer therapy. Br. J. Cancer 111, 2039–2045 (2014). 132. Schlenk RF, Fröhling S, Hartmann F, Fischer JT,

(20)

1

133. Schlenk RF, L??bbert M, Benner A, Lamparter A, Krauter J,et al. All-trans retinoic acid as adjunct to intensive treatment in younger adult patients with acute myeloid leukemia: results of the randomized AMLSG 07-04 study. Ann. Hematol. 95, 1931–1942 (2016).

134. Schenk T, Chen WC, Göllner S, Howell L, Jin L,et al. Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-trans-retinoic acid differentiation pathway in acute myeloid leukemia. Nat. Med. 18, 605–11 (2012).

135. Boutzen H, Saland E, Larrue C, de Toni F, Gales L,et al. Isocitrate dehydrogenase 1 mutations prime the all-trans retinoic acid myeloid differentiation pathway in acute myeloid leukemia. J. Exp. Med. 213, 483–497 (2016).

136. Martelli MP, Gionfriddo I, Mezzasoma F, Milano F, Pierangeli S,et al. Arsenic trioxide and all-trans retinoic acid target NPM1 mutant oncoprotein levels and induce apoptosis in NPM1-mutated AML cells. Blood 125, 3455–3465 (2015). 137. Hajj H El, Dassouki Z, Berthier C, Raffoux

E, Ades L,et al. Retinoic acid and arsenic trioxide trigger degradation of mutated NPM1, resulting in apoptosis of AML cells. B lood 125, 3447–3454 (2015).

Referenties

GERELATEERDE DOCUMENTEN

We found that simultaneous PU.1/CITED2 deregulation in human CD34 + cord blood (CB) cells, as well as CITED2 upregulation in preleukemic murine PU.1-knockdown (PU.1 KD/KD )

by the increasing knowledge about characteristic ROS-low LSC properties, since features such as low metabolic activity, increased BCL2 expression and high levels of

(i) Expression of indicated proteins is shown for the ROS-low and ROS-high cell fraction sorted from CD34 + AML cells.. (ii) Expression of indicated proteins is shown from the

We demonstrated that ROS- low LSCs have a reduced mitochondrial content, low levels of ATP and a different gene expression signature that suggests an increased stemness

Latere gebeurtenissen in de AML- evolutie veroorzaken differentiatie- en proliferatie-afwijkingen en resulteren in de vorming van leukemische stamcellen, welke voor een

(A) mRNA expression fold change of indicated genes in the ROS-low vs the ROS-high cell fraction of normal (both CB and PBSCs) CD34 + stem- and progenitor cells (n=4).. Genes

Normal hematopoietic- and leukemic stem cells are very similar regarding their morphology, metabolic activity and gene expression, however, they have an altered

Human acute myeloid leukemia reprogramming to pluripotency is a rare event and selects for patient hematopoietic cells devoid of leukemic mutations..