• No results found

University of Groningen The long road Hilgendorf, Susan

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen The long road Hilgendorf, Susan"

Copied!
23
0
0

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

Hele tekst

(1)

The long road

Hilgendorf, Susan

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2018

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Hilgendorf, S. (2018). The long road: The autophagic network and TP53/ASXL1 aberrations in hematopoietic malignancies. Rijksuniversiteit Groningen.

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

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.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)
(3)
(4)
(5)

1

1. NORMAL AND MALIGNANT HEMATOPOIESIS

1.1 Normal hematopoiesis

Differentiation within the hematopoietic system cells is based on a hierarchal tree with immense regenerative potential. At the top of the hierarchy reside the most undifferentiated hematopoietic stem cells (HSC) with the greatest self-renewal potential (Figure 1). After

cell division they give rise to more differentiated progenitors, which constitute the entire blood lineage in a multi-step process. Constant cell differentiation and proliferation occurs to reconstitute the hematopoietic system but only a small amount of long-term HSCs is responsible for the total blood development. However, recent studies have also shown that during steady state hematopoiesis the multipotent and lineage-restricted progenitors are highly responsible to drive hematopoiesis. This suggests that dependent on the insult, e.g. transplantation vs steady state hematopoiesis, HSCs vs progenitors mainly drive hematopoiesis1–5. LT-HSC ST-HSC MPP LMPP CLP CMP GMP MEP T cells B cells NK cells dendritic cells granulocytes macrophages erythrocytes platelets

Figure 1: Schematic representation of the hematopoietic system Figure 1: Schematic representation of the hematopoietic system

(6)

The path into different lineages is under control of transcription factors and epigenetic changes within the hematopoietic stem and progenitor population (HSPC) in conjunction with the surrounding micro-environment6,7. Stem cells appear to have an open chromatin structure

to allow for multi-lineage differentiation and the activity of certain chromatin-related genes determines accessibility of lineage specific genes8. Mutations of genes within the primitive

hematopoietic cells can predispose individuals to develop myeloid malignancies.

1.2 Clonal hematopoiesis of indeterminate potential

Several studies have shown that with increased age, blood-specific mutations increase9–11. The

mutation frequency is low in the younger generation but is rising from 1.5% in those aged 50-59 to 19.8% in 90-98 year olds12. Many of the discovered genes play important roles in

leukemogenesis11. The term “clonal hematopoiesis of indeterminate potential” (CHIP) has

been proposed to describe the feature of clonal hematopoiesis with leukemogenic-related somatic mutations in the blood and bone marrow without displaying any dysplasia in the bone marrow (Figure 2)13,14. Many people can present with background mutations, which

are unrelated to hematopoietic expansion13. However, recurrently mutated genes may lead

to clonal hematopoiesis by awarding stem cells and their progenies with survival advantages and allow for clonal expansion but do not induce hematopoietic disease development13,14.

Mutated genes that are thought to contribute to pathogenesis of cancer, such as DNMT3A, TET2, and ASXL1, are part of CHIP and predispose individuals to develop hematological malignancies in particular when they are present in combinations. Nevertheless, persons with CHIP clones can remain healthy for years with clones being stable at low levels9,11. This

suggests that cell intrinsic changes are necessary but not sufficient for disease development and progression. Cell extrinsic factors, such as environmental stresses, inflammation, radio- and chemotherapy, might be important to drive preferential clonal expansion15,16. Evidence

is emerging that chemotherapy may lead to the expansion of an, until then, unrelated hematopoietic population with somatic mutations17,18. Consequently, to transform to overt

malignancy, cells need to acquire additional mutations in the background of cooperating genes. Additionally, the leukemogenic genes in CHIP have been found to lead to an increased risk incidence of ischemic stroke and coronary heart disease in patients11,19. In mice, mutated

TET2 has been demonstrated to induce clonal hematopoiesis and leads to an accelerated development of atherosclerosis with a striking increase in atherosclerotic plaque size due to an altered function of infiltrating monocytes and macrophages.

(7)

1

Figure 2: Schematic representation of the development of hematological malignancies

CHIP MDS AML

Clonal size

Clonal haematopoiesis

Cytopenias & dysplasia

Excess blasts

Normal blood cell Mutation 1 Mutation 2 Mutation 3 Mutation 4

Figure 2: Schematic representation of the development of hematological malignancies

1.3 Myelodysplastic syndrome

Myelodysplastic syndrome (MDS) comprises a set of symptoms, with the most prominent ones being morphologic dysplasia, peripheral cytopenia and subsequently ineffective hematopoiesis20. MDS is characterized by ineffective hematopoiesis of one or several

lineages in the bone marrow21. Patients can present with reduced platelet production due to

enhanced apoptosis of immature megakaryocytes22,23. Evidence suggests that an abnormal

microenvironment can contribute to disease patterns within patients. Inflammation, oxidative stress, and abnormal stromal function can be instrumental to the pathogenesis of MDS24–27.

Similarly, low-risk MDS erythroid-blasts reveal an overexpression of pro-apoptotic genes and are more vulnerable to programmed cell death when removed from the bone marrow environment28,29. With disease progression, a differentiation block of all myeloid lineages

can be observed and ultimately, a blast count of up to 19% can be detected within the bone marrow (Table 1).

(8)

Table 1: Classification of MDS

Name

Blast

BM PB

MDS with single-lineage dysplasia (MDS-SLD) <5% <1%

MDS with multi-lineage dysplasia (MDS-MLD) <5% <1%

MDS with ringed sideroblasts (MDS-RS) <5% <1%

MDS with excess blasts (MDS-EB) 5-19% 2-19%

MDS, unclassifiable (MDS-U) <5% =<1%

BM: Bone marrow; PB: Peripheral blood

A recent model suggested that the cells of origin are MDS-stem cells which arose from CHIP (Figure 2)30,31. With increased severity of MDS, patients appear to acquire more mutations

and are at greater risk to develop secondary acute myeloid leukemia (sAML)32,33. Patients with

sAML are more chemoresistant, have reduced event-free survival, and have an overall poorer outcome34. The genetic aberrations underlying MDS have been heavily investigated and 75

to 90% of patients have at least one known, recurrently mutated gene32,33. The spliceosome

machinery, epigenetic/DNA methylating machinery, and transcription factors are the most commonly affected34–38. Moreover, approximately half of all MDS patients present with

chromosomal rearrangements, such as del(5q),-7/del(7q), trisomy 839. These copy-number

alterations can drive MDS evolution as individual disease subset or by affecting several genes. For example, del(5q) can present with a distinct pathological and clinical disease subset in MDS patients as 5q- syndrome. It has been found that these patients respond very well to

lenalidomide40. EZH2, located at 7q, may present with deletion, missense and frameshift

mutations, leading to inactivating and loss-of-function mutations41,42. In mice, EZH2 loss in

hematopoietic stem cells resulted in myelodysplasia and MDS-like phenotypes43. Changes

in copy number alterations are more likely to occur in intermediate and high-risk MDS and predict for unfavorable outcome44,45. These genetic lesions could help to determine more

precise treatment options to manage MDS, as no known drug therapy is ultimately curative46–48.

Frequently, low-risk MDS patients are only seen in the clinic until symptoms worsen and disease progression occurs49. High-risk MDS patients may receive chemotherapeutic agents

and ultimately hematopoietic stem cell transplantation (HSCT). HSCT is considered the only curative option but is still associated with enhanced mortality rates50,51.

1.4 AML

AML is a disease that remains difficult to treat. Up to 70% of patients above the age of 65 that receive chemotherapy die within one year of diagnosis due to AML52. It manifests itself

as an accumulation of malignant myeloid cells that replace normal bone marrow cells and ultimately present in the peripheral blood53. AML can arise due to the genetic mutations

(9)

1

that may lead to a block of differentiation and disturbed proliferation of clonal populations of the myeloid stem cells (Figure 2). The World Health Organization classified adult AML

with recurrent genetic aberrations into several separate groups, including the t(8;21) group, the inv(16) group, and the MLL fusion gene group54. By sequencing more than 1500 AML

patients, Papaemmanuil et al. established a new genomic classification with non-overlapping subtypes55. The second biggest subgroup established contained RNA splicing, chromatin,

and transcription factor genes. This chromatin-spliceosome group was composed of such complex patterns and co-mutations that no individual genetic aberration was defining this group55. This interplay of genetic lesions reveals the complexity of this AML subgroup.

Genetic aberrations are found in more than 97% of AML patients, affecting mostly the same pathways as found in high- risk MDS patients56,57. Drawing a line between high-risk

MDS and AML patients has been challenging as a 20% blast count in the bone marrow and peripheral blood is supposedly indicative for AML but at 19% for MDS. If applicable, AML and high-risk MDS patients will receive intensive chemotherapy. Distinguishing these two diseases and accurately classifying them is essential for patients to determine therapy but also to assess prognosis of outcome. A deeper understanding into genes leading to MDS and AML development may eventually help improve patient outcome by more targeted and individualized therapy.

2. THE ROLE OF ASXL1 IN NORMAL AND MALIGNANT

HEMATOPOIESIS

2.1 ASXL1

Originally discovered in Drosophila, a human homologue named ASXL1 (addition of sex-comb like 1) was later found at 20q11 by Fisher et al. Under normal conditions, ASXL1 is thought to recruit or stabilize the polycomb repressive complex 2 (PRC2) using its C-terminus-containing PHD finger to interact with chromatin and is therefore being classified as an epigenetic modifier (Figure 3, left). PRC2 controls gene expression patterns

in cells by controlling the chromatin structure. ASXL1 can either stabilize or recruit the PRC2 complex to the chromatin. Using its lysine methyltransferase capabilities, the PRC2 core subunit EZH2 can catalyse H3K27me3, compacting the chromatin further. ASXL1 is thus involved in the silencing of specific target genes, such as the HOXA family. Upon loss of the C-terminus, the interaction between ASXL1 and chromatin is disrupted and H3K27 trimethylation levels are reduced over time58. Therefore, genes such as the HOXA family may

become derepressed and are transcriptionally accessible. Most members of the HOXA cluster are found to be leukemogenic due to their control over the self-renewal and differentiation

(10)

potential of HSPCs59. Inoue et al. and Abdel-Wahab et al. demonstrated an upregulation of

HOXA9 after ASXL1 abrogation, suggesting that derepression of target genes via loss of H3K27me3 may play an important role in myeloid transformation58,60.

The ASX homology (ASXH) domain, which adjoins the N-terminal region of ASXL1, appears to be necessary for protein-protein interaction61. The most prominent interaction partner is

BAP1 with which ASXL1 forms the polycomb repressive deubiquitinase (PR-DUB) complex (Figure 3, right)62. BAP1 is a ubiquitin C-terminal hydrolase (UCH) with deubiquitinating

activity that possesses tumor suppressor functions63,64. Within the PR-DUB complex, BAP1

removes monoubiquitin from H2AK119Ub and prevents hyperubiquitylation of H2A and therefore balances gene expression62. Scheuermann et al. demonstrated in Drosophila that

deubiquitination of H2A requires ASXL1-BAP1 interaction, suggesting that loss of ASXL1 may lead to increased ubiquitination and therefore silencing of target genes62.

RbAp46/48 EZH1/2 SUZ12 Jarid2 EED ASXL1 BAP1 ASXL1

PRC2

PR-DUB

H3K27me3 H2AK119ub

Figure 3: Schematic representation of ASXL1 within the PRC2 & PR-DUB complexes Figure 3: Schematic representation of ASXL1 within the PRC2 & PR-DUB complexes

2.2 ASXL1 in human and mouse hematopoiesis

Initially, ASXL1 was thought not to be required for stem cell and multipotent progenitor function65. However, later studies conducted by Wang et al. and Abdel-Wahab et al. revealed

(11)

1

importance for hematopoiesis was established as a complete and tissue specific knockout in mice lead to myelodysplastic-like phenotype and developmental defects in vivo66,67. Moreover,

loss of ASXL1 was found to compromise the erythroid differentiation of human cord blood CD34+ cells and reduced the number of erythroid progenitors in mice68,69. In patients, ASXL1

abrogation is always heterozygous and most mutations occur as frameshift or missense mutations within exon 12 (Figure 4). This can lead to loss of the C-terminus, including the

PHD finger, and result in truncation of the protein. A deletion of ASXL1 gene expression and therefore loss of protein in mice has enabled myeloid leukemogenesis only in combination with other gene alterations, such as TET2 and NRAS58,67. Whether the truncated form of the

ASXL1 protein has any additional functions that may lead to cancer development is currently under investigation. Inoue et al. demonstrated that ASXL1 truncation mutants are detectable at protein levels, suggesting that the altered proteins may be stable for at least some time to facilitate other functions70. A mouse model overexpressing a mutant ASXL1 protein revealed

an MDS-like disease after a 12 month latency and some mice progressed towards leukemia60.

Underlying epigenetic changes clearly involved reduction of H3K27me3 of the HOXA family members. A study conducted by Balasubramani suggested that ASXL1 mutations could have gain-of-function properties. Overexpression of several mutants together with BAP1 in a mouse cell line enhanced PR-DUB activity and reduced H2AK119Ub and H3K27me3 levels greatly. ASXL1 mutant activity seemed to be completely dependent on BAP1 binding or interaction, suggesting that H2AK119Ub depletion is the main cause of H3K27me3 loss71.

N PHD EXON 12 EXON 11 Frameshift Nonsense G646fs*12 Missense E1102D R693*

ASXH ASXM1 ASXM2

Mutations

Figure 4: Schematic representation of ASXL1 mutations

Figure 4: Schematic representation of ASXL1 mutations in hematopoietic and lymphoid tissue)

(12)

2.3 ASXL1 in MDS and AML development and progression

According to a study conducted by Jaiswal et al., ASXL1 is the third most commonly mutated gene in healthy individuals predisposing these individuals to an increased risk of hematological cancers. However, the absolute risk of developing malignant hematopoiesis remains small11. Interestingly, ASXL1 appears to be mutated mainly in the oldest of the healthy

individuals10. Once an MDS or AML is diagnosed, ASXL1 singularly predicts for adverse

outcome37,72–75. The frequency of ASXL1 distortion varies between myeloid malignancies. In

MDS, approximately 16% of patients have ASXL1 alterations, while this is 6% for de novo AML and 30% for sAML patients75.

It is still unclear whether ASXL1 is a cancer-initiating gene or a mutation contributing to disease development and progression. Initially, studies suggested that ASXL1 alterations occur primarily in high-risk MDS and AML patients33,74. However, new studies conducted

on low-risk MDS found ASXL1 mutations to be present and also predicting for worse overall survival35,76,77. Moreover, ASXL1 mutations appeared to be retained throughout disease

progression and relapse. Nevertheless, mouse models failed to reveal a clear relationship between initiation of malignant myeloid development and ASXL1 functional abrogation, raising the question whether mutations in ASXL1 are initiators of disease37,78,79. ASXL1 could

be a second- or third hit to help shape the epigenetic landscape and thus allow for genes to induce transformative effects. Furthermore, ASXL1 mutations could be acquired throughout disease development. Evidence for these theories comes from the frequencies of co-mutation. Up to 85% of MDS patients with ASXL1 mutation present with at least one additional gene alteration. Patients appear to retain the concurrent gene alterations, suggesting that ASXL1 does not convey initiating events on its own77. Often, ASXL1 is co-mutated with genes

typically occurring in a distinct set of pathways, including transcription factors (RUNX1, TP53), spliceosomes (U2AF1, SRSF2), epigenetic modifiers (TET2, EZH2), and signaling (NRAS)35,74,76,77,80,81. The variety of possible co-mutations and the high percentage of sAML

suggest that ASXL1 alone cannot initiate and drive MDS/AML development and progression.

2.4 ASXL2 and ASXL3

All three ASXL genes are expressed in different tissues and have been found mutated in a variety of cancers82,83. ASXL2 mutations in AML are mainly found in t(8;21)-AML patients

and are mutual exclusive with ASXL1 mutations84. ASXL3 is expressed at rather low levels

in bone marrow cells compared to the other two genes and reports about its mutation in adult AML are rare. The three genes share up to 70% homology between their ASXN, ASXH, ASXM1, ASXM2 and PHD domains83. Sahtoe et al. discovered that the DEUBiquitinase

Adaptor (DEUBAD) domain of ASXL2/3, containing all of ASXH and more, comprises >60% sequence identity to ASXL185. They demonstrated that all three ASXL proteins could

(13)

1

stimulate BAP1 activity to similar extends in E. coli bacteria. In a study conducted by Daou et al., it was found that BAP1 forms two mutually exclusive PR-DUBs with ASXL1 and -286.

Moreover, they found that loss of BAP1 destabilizes ASXL2 but not ASXL1. In murine hearts, knockout of ASXL2 was seen to disrupt PRC2-chromatin interaction due to PRC2s’ failure of trimethylating H3K27me2. Additionally, loss of ASXL2 H2Aub levels increased noticeably82.

Consequently, the three ASXL proteins appear to have similar functional mechanism but might exert them on different target regions due to tissue dependency. How and to what extent ASXL2 and 3 confer their activity in myeloid malignancies remains to be seen.

3. TP53 MUTATIONS IN HEMATOPOIETIC MALIGNANCIES

3.1 Normal and mutant functions of TP53

The normal functions of the tumor suppressor p53 have been extensively studied for more than 30 years in cell cultures and animal models. It has been widely established that this gene plays a pivotal role in many different pathways, such as differentiation, cell cycle, and apoptosis. Located on chromosome 17p13.1, the encoded p53 protein has a half-life of 5-20 minutes, which is tightly regulated via a negative feedback loop involving the N-terminus87,88.

P53 induces MDM2 transcription, which in turn binds to the N-terminus of the p53 protein and marks it for degradation89. Additionally, the N-terminal is necessary for transcriptional

transactivation and repression, and the induction of apoptosis90. The C-terminal region

contains an oligomerization and a DNA damage recognition domain, which is necessary for conformational changes and transactivation of p5388. The central region of the gene contains

the DNA binding domain that can distinguish target sequences for binding88. A wide amount

of genes are under the control of p53, such as autophagy genes (ATG7, FOXO3), DNA repair genes (DDB2, MLH1), and senescence genes (CDKN1A, PML)91. Stressors such as DNA

damaging agents may transiently stabilize and therefore lead to accumulation of the p53 protein, resulting in increased transcriptional activation of downstream targets92. Mutant p53

protein may accumulate in mutant cells via several pathways. Accumulation may occur due to the inability to bind MDM2, due to failed induction of transcription of MDM2, or due to differences in folding and degradation, thereby keeping mutant p53 constitutively stabilized and expressed at augmented protein levels87,93.

Half of all cancers have a deregulation of p53, with many tumors revealing an accumulation of mutant protein94,95. Many p53 mutants are unable to activate the same targets as wild-type

p53 and therefore lose the p53 tumor suppressor function. Additionally, these mutations may confer a dominant-negative effect over the remaining wild-type p53, abolishing wild-type functions over time96. Increased presence of p53 mutant protein can lead to the

(14)

resistance97. Furthermore, mutant proteins that lost their apoptotic functions can interrupt

p53-independent apoptotic pathways to enhance cell survival and diminish apoptosis98.

Taken together, this may lead to tumor initiation and progression.

3.2 TP53 mutations in AML and MDS

Knowledge of the role of TP53 mutations in leukemia and its involvement in leukemia onset and progression is scarce. Overall, only five to ten per cent of hematopoietic malignancies have TP53 dysfunctions, while up to 50-70% are found in solid tumors (for example in ovarian cancer and head and neck cancer)99,100. Rücker et al. conducted a study into complex

karyotype AML (CK-AML), a subgroup of AML101. They demonstrated that TP53 alterations

were the single most important prognostic factor for CK-AML with 70% of the cases having a TP53 deletion and/or mutation. Additionally, Wong et al. recently established the importance of TP53 in therapy related AML/MDS18. By sequencing the genomes of patients

and establishing mouse models with TP53-mutated HSPCs, they uncovered the preferential expansion these clones after chemotherapy. This was recently confirmed in patients who had obtained an autologous stem cell transplantation and were followed over time to determine the evolution of preleukemic clones16. Present data suggests that a TP53 clone may undergo

clonal evolution due to the selective pressure of cytotoxic therapy16,18,102. Generally, AML and

MDS patients presented with TP53 alterations predict for chemotherapy resistance101,103,104.

The median survival among these patients can be as low as four month in comparison to 11 month with TP53 unaltered function101. As conventional chemotherapy does not increase

patient survival, new treatment strategies are currently being exploited. The most prominent success so far has been achieved by the use of hypomethylating agents, such as Decitabine and Azacytidine. Both drugs may lead to favorable clinical responses and to (although incomplete) mutation clearance105,106. However, different types of TP53 mutation may require different

treatment strategies.

TP53 mutations in advanced MDS and sAML have been discovered to be mainly hemizygous or homozygous due to a copy number-neutral loss of heterozygosity (CN-LOH) or deletion of 17p107. Patients with loss of heterozygosity (LOH) of 17p and other forms of TP53

mutations are generally presented with complex karyotypes101,105,108. P53 can present with

many co-mutations, such as DNA methylators (DNMT3A, TET2), spliceosome machinery (U2AF1, SRSF2), transcription factors (ETV6, CEBPA), fusion and gene rearrangements (MLL and EVI1 translocations) and more 57,105,109–111.

In myeloid malignancies, most TP53 mutations are missense substitutions and are located within the DNA binding domain. These mutations can exert an oncogenic gain-of-function (GOF), presumably altering the binding profile of mutant p53 versus the wild-type but still allowing for full-length expression of the protein112. In a panel of breast cancer cell lines, Zhu

(15)

1

et al. discovered that GOF p53 appears to be conveyed via post-translational modification of histones113. Specifically the MLL family of DNA methyltransferases appeared to be under

tight control of GOF p53. It is therefore not surprising that the DNA hypomethylating agent Decitabine revealed a favorable clinical response among AML and MDS patients with TP53 mutations105. A study into several, non-hematopoietic cell lines revealed the

interaction of GOF p53 and the proteasome machinery114. In concert with NRF2, p53

missense substitutions led to the resistance against the proteasome inhibitor carfilzomib. The resistance was overcome by combining carfilzomib with APR-246, a small molecule that restores TP53 wild-type conformation115. Post-translational changes of mutants may bestow

additional treatment options. Activity of wild-type and mutant p53 can be influenced by acetylation and phosphorylation, with the latter shielding the proteins from degradation via MDM2116,117. In triple negative breast cancers, Wang et al. demonstrated that certain histone

deacetylase inhibitors (HDACIs) could decrease phosphorylation, protein, and mRNA levels of mutant p53 in vitro118. Usage of the HDACI SAHA destabilized mutant p53 but not

wild-type p53 levels, killing tumour cell lines in vitro119. A phase 1 study in AML and MDS

patient with SAHA showed first promising results with some patients achieving complete response120. Another encouraging approach is the selective degradation of mutant p53 via

the mevalonate pathway. Parrales et al. demonstrated that statins could reduce mevalonate-5-phosphate, thereby leading to misfolding of mutant proteins by disrupting the binding of the heat-shock protein DNAJA1 to the mutant p53 protein121. The mutant proteins are then

marked for degradation. Using statins based on mutant p53 status might provide additional treatment options.

Unraveling the differences in downstream targets between wild-type and mutant p53 may also play a pivotal role in overcoming negative effects of mutant p53. Some mutants, but not wild-type p53, have been found to enhance NF-ĸB activation thus promoting cancer progression via a chronic inflammatory pathway122. Moreover, mutant p53 proteins may bind

and inactivate the two p53 family member p63 and p73123,124. Mouse models demonstrated

that, when wild-type p53 function is abrogated, p63 and p73 could partially compensate125.

However, their binding by mutant p53 disrupts their tumor suppressive functions. If this p53-p63-p73 axis is targetable remains to be elucidated. More studies into p53 mutants and their role in hematopoietic malignancies are currently conducted and may pave the way for more efficient (and novel) therapeutic agents.

(16)

4. AUTOPHAGY

Autophagy is a process used by cells to degrade and recycle cellular substrates. Three different types of autophagy exist, with macroautophagy being the most intensely studied so far. Eukaryotic cells have a consistent basal level of macroautophagy that can be enhanced under stress conditions. Macroautophagy takes place in several steps. At first, the ULK1/2 complex induces the nucleation of a phagophore126–128. Then, with the help of complexes

and proteins such as LC3-II and ATG9, the phagophore with a double membrane becomes elongated and ultimately matures enough to close around its cargo, which is then known as autophagosome128–130. Thereafter, the outer membrane of the autophagosome fuses with an

endosome and/or lysosome to form an autolysosome. The content within the autolysosome is then degraded and transported through the permease back into the cytoplasm for re-use128,131,132.

While it is widely acknowledged that autophagy is a general housekeeping process necessary to remove misfolded proteins and damaged organelles, evidence has been emerging that autophagy is an important regulator of HSPC maintenance. In mice, Warr et al. discovered that autophagy is essential for maintaining young and old HSCs via FOXO3A induced autophagic gene program133. Gomez-Puerto et al. found that CD34+/CD38- immature HSPCs

appear to have a higher autophagic flux than the more progenitor-like CD34+/CD38+ cells,

as well as differentiated erythroid and myeloid cells134. Upon knockdown of the autophagy

genes ATG5 and ATG7, which are required for elongation and closure of the phagophore double membrane, reduced HSPC frequencies could be observed in vitro as well as in vivo134– 136. Additionally, Mortensen et al. discovered that ATG7-deficient Lin-Sca-1+ c-Kit+ (LSK)

cells were not able to reconstitute the hematopoietic system in lethally irradiated mice137.

Moreover, production of myeloid and lymphoid progenitors was impaired when ATG7 was absent. These findings collectively suggest that autophagy is an important player in regulating HSPC/HSC maintenance. Increasing evidence also points towards autophagy being involved in cancer development and therapy resistance in cancer treatment. Folkerts et al. discovered that leukemic cell lines and primary human CD34+ AML cells have a large variability in

basal autophagic flux (Chapter 3, this thesis)138. Especially poor-risk AMLs, which are

frequently associated with mutations in TP53, have higher autophagic levels than favorable- or intermediate risk AMLs. Knockdown of the autophagy genes ATG5 or ATG7 initiated apoptosis and coincided with increased expression of p53138.

(17)

1

5. SCOPE OF THE THESIS

The aim of the first part of this thesis was to investigate the role of ASXL1 in HSPCs and whether malignant transformation can occur upon combined knockdown of ASXL1 with TP53. Mouse studies into ASXL1 functions suggested that a complete loss of ASXL1 leads to an MDS-like phenotype accompanied with pronounced anemia, a clinical manifestation often observed in MDS and AML patients66,67. However, studies into the function of

ASXL1 and its abrogation in human cells are missing. In Chapter 2 of this thesis, we aimed

to elucidate the role of ASXL1 during human stem and progenitor development in vitro and investigated its effect particularly in erythroid development. It has also been shown that ASXL1 is co-mutated with different transcription factors in MDS and AML patients. Following the detrimental effects of ASXL1 loss, we investigated in Chapter 3 if additional

loss of the transcription factor TP53 could rescue ASXL1 mutated cells and lead to malignant transformation in vitro and in vivo.

Autophagy can be used as a survival mechanism in times of energy crisis or other forms of stress. In treatment of cancer, autophagy has been found to play a pivotal role in therapy resistance. The aim of the second part of this thesis was to highlight the present understanding of the autophagy pathway in cancer, its role in transformation, disease progression and therapy resistance (Chapter 5) and how targeting autophagy might provide a new treatment

strategy in AMLs (Chapter 4). The aim of Chapter 4 was to determine if autophagy

inhibition could be used as a treatment strategy for patients with AML and which underlying genes and pathways are involved. In Chapter 5, the goal was to review the multi-faceted

role of autophagy in regulating cancer initiation and cancer (stem) cell maintenance, how autophagic flux in cancer affects therapeutic outcome, and the role of autophagy in the tumor microenvironment. Moreover, the aim was to give perspectives on selective targeting of cancer (stem) cells for therapy.

Finally, in Chapter 6, a summary of the results and a discussion of future perspectives are

(18)

REFERENCES

1. Sun J, Ramos A, Chapman B, et al. Clonal dynamics of native haematopoiesis. Nature 2014;514(7522):322–327. 2. Harrison DE, Lerner C, Hoppe

PC, Carlson GA, Alling D. Large numbers of primitive stem cells are active simultaneously in aggregated embryo chimeric mice. Blood 1987;69(3):773–777. 3. Busch K, Rodewald H-R.

Unperturbed vs.

post-transplantation hematopoiesis: both in vivo but different. Curr Opin Hematol 2016;23(4):295–303. 4. Busch K, Klapproth K, Barile M,

et al. Fundamental properties of unperturbed haematopoiesis from stem cells in vivo. Nature 2015;518(7540):542–546. 5. Sawai CM, Babovic S, Upadhaya S,

et al. Hematopoietic Stem Cells Are the Major Source of Multilineage Hematopoiesis in Adult Animals. Immunity 2016;45(3):597–609. 6. Bryder D, Rossi DJ, Weissman

IL. Hematopoietic stem cells: the paradigmatic tissue-specific stem cell. Am J Pathol 2006;169(2):338– 346.

7. Doulatov S, Notta F, Laurenti E, Dick JE. Hematopoiesis: A Human Perspective. Cell Stem Cell 2012;10(2):120–136.

8. Akashi K, He X, Chen J, et al. Transcriptional accessibility for genes of multiple tissues and hematopoietic lineages is hierarchically controlled during early hematopoiesis. Blood 2003;101(2):383–389.

9. Genovese G, Kähler AK, Handsaker RE, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med 2014;371(26):2477–2487. 10. Xie M, Lu C, Wang J, et al.

Age-related mutations associated with clonal hematopoietic expansion

and malignancies. Nat Med 2014;20(12):1472–1478. 11. Jaiswal S, Fontanillas P, Flannick

J, et al. Age-Related Clonal Hematopoiesis Associated with Adverse Outcomes. N Engl J Med 2014;371(26):2488–2498. 12. McKerrell T, Park N, Moreno T, et

al. Leukemia-associated somatic mutations drive distinct patterns of age-related clonal hemopoiesis. Cell Rep 2015;10(8):1239–1245. 13. Steensma DP, Bejar R, Jaiswal

S, et al. Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood 2015;126(1):9– 16.

14. Heuser M, Thol F, Ganser A. Clonal Hematopoiesis of Indeterminate Potential. Dtsch Arzteblatt Int 2016;113(18):317–322. 15. Link DC, Walter MJ. ’CHIP’ping

away at clonal hematopoiesis. Leukemia 2016;30(8):1633–1635. 16. Berger G, Kroeze LI,

Koorenhof-Scheele TN, et al. Early detection and evolution of pre-leukemic clones in therapy-related myeloid neoplasms following autologous SCT. Blood [Epub ahead of print]. 17. Wong TN, Miller CA, Klco JM, et al. Rapid expansion of preexisting nonleukemic hematopoietic clones frequently follows induction therapy for de novo AML. Blood 2016;127(7):893–897. 18. Wong TN, Ramsingh G, Young AL,

et al. Role of TP53 mutations in the origin and evolution of therapy-related acute myeloid leukaemia. Nature 2015;518(7540):552–555. 19. Jaiswal S, Natarajan P, Silver

AJ, et al. Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease. N Engl J Med 2017;377(2):111–121.

20. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 2016;127(20):2391–2405. 21. Corey SJ, Minden MD, Barber DL,

Kantarjian H, Wang JCY, Schimmer AD. Myelodysplastic syndromes: the complexity of stem-cell diseases. Nat Rev Cancer 2007;7(2):118– 129.

22. Braun T, Carvalho G, Grosjean J, et al. Differentiating megakaryocytes in myelodysplastic syndromes succumb to mitochondrial derangement without caspase activation. Apoptosis Int J Program Cell Death 2007;12(6):1101–1108. 23. Houwerzijl EJ, Blom NR, van der

Want JJL, et al. Increased peripheral platelet destruction and caspase-3-independent programmed cell death of bone marrow megakaryocytes in myelodysplastic patients. Blood 2005;105(9):3472–3479. 24. Marcondes AM, Ramakrishnan A,

Deeg HJ. Myeloid Malignancies and the Marrow Microenvironment: Some Recent Studies in Patients with MDS. Curr Cancer Ther Rev 2009;5(4):310–314.

25. Rankin EB, Narla A, Park JK, Lin S, Sakamoto KM. Biology of the bone marrow microenvironment and myelodysplastic syndromes. Mol Genet Metab 2015;116(1–2):24– 28.

26. Yang L, Qian Y, Eksioglu E, Epling-Burnette PK, Wei S. The inflammatory microenvironment in MDS. Cell Mol Life Sci CMLS 2015;72(10):1959–1966. 27. Tauro S, Hepburn MD, Peddie

CM, Bowen DT, Pippard MJ. Functional disturbance of marrow stromal microenvironment in the myelodysplastic syndromes. Leukemia 2002;16(5):785–790.

(19)

1

28. Tehranchi R, Invernizzi R, Grandien A, et al. Aberrant mitochondrial iron distribution and maturation arrest characterize early erythroid precursors in low-risk myelodysplastic syndromes. Blood 2005;106(1):247–253. 29. Folkerts H, Hazenberg CLE,

Houwerzijl EJ, et al. Erythroid progenitors from patients with low-risk myelodysplastic syndromes are dependent on the surrounding micro environment for their survival. Exp Hematol 2015;43(3):215–222.e2. 30. Woll PS, Kjällquist U, Chowdhury

O, et al. Myelodysplastic syndromes are propagated by rare and distinct human cancer stem cells in vivo. Cancer Cell 2014;25(6):794–808. 31. Sperling AS, Gibson CJ, Ebert BL. The genetics of myelodysplastic syndrome: from clonal haematopoiesis to secondary leukaemia. Nat Rev Cancer 2017;17(1):5–19. 32. Papaemmanuil E, Gerstung

M, Malcovati L, et al. Clinical and biological implications of driver mutations in

myelodysplastic syndromes. Blood 2013;122(22):3616–3627; quiz 3699.

33. Haferlach T, Nagata Y, Grossmann V, et al. Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia

2014;28(2):241–247. 34. Lindsley RC, Mar BG, Mazzola

E, et al. Acute myeloid leukemia ontogeny is defined by distinct somatic mutations. Blood 2015;125(9):1367–1376. 35. Bejar R, Stevenson KE, Caughey

BA, et al. Validation of a Prognostic Model and the Impact of Mutations in Patients With Lower-Risk Myelodysplastic Syndromes. J Clin Oncol 2012;30(27):3376–3382. 36. Bejar R, Stevenson KE, Caughey

B, et al. Somatic mutations predict poor outcome in

patients with myelodysplastic syndrome after hematopoietic stem-cell transplantation. J Clin Oncol Off J Am Soc Clin Oncol 2014;32(25):2691–2698. 37. Della Porta MG, Gallì A,

Bacigalupo A, et al. Clinical Effects of Driver Somatic Mutations on the Outcomes of Patients With Myelodysplastic Syndromes Treated With Allogeneic Hematopoietic Stem-Cell Transplantation. J Clin Oncol [Epub ahead of print]. 38. Uy GL, Duncavage EJ, Chang

GS, et al. Dynamic changes in the clonal structure of MDS and AML in response to epigenetic therapy. Leukemia [Epub ahead of print]. 39. Haase D, Germing U, Schanz J, et

al. New insights into the prognostic impact of the karyotype in MDS and correlation with subtypes: evidence from a core dataset of 2124 patients. Blood 2007;110(13):4385–4395. 40. Talati C, Sallman D, List A.

Lenalidomide: Myelodysplastic syndromes with del(5q) and beyond. Semin Hematol 2017;54(3):159–166. 41. Nikoloski G, Langemeijer SMC,

Kuiper RP, et al. Somatic mutations of the histone methyltransferase gene EZH2 in myelodysplastic syndromes. Nat Genet 2010;42(8):665–667. 42. Ernst T, Chase AJ, Score J, et al.

Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders. Nat Genet 2010;42(8):722–726. 43. Mochizuki-Kashio M, Aoyama

K, Sashida G, et al. Ezh2 loss in hematopoietic stem cells predisposes mice to develop heterogeneous malignancies in an Ezh1-dependent manner. Blood 2015;126(10):1172–1183. 44. Bernasconi P, Klersy C, Boni M,

et al. Does cytogenetic evolution have any prognostic relevance in myelodysplastic syndromes? A study on 153 patients from a

single institution. Ann Hematol 2010;89(6):545–551. 45. Wang H, Wang X-Q, Xu X-P,

Lin G-W. Cytogenetic evolution correlates with poor prognosis in myelodysplastic syndrome. Cancer Genet Cytogenet 2010;196(2):159–166. 46. Chang C-K, Zhao Y-S, Xu F, et al.

TP53 mutations predict decitabine-induced complete responses in patients with myelodysplastic syndromes. Br J Haematol [Epub ahead of print].

47. Cherian MA, Tibes R, Gao F, et al. A study of high-dose lenalidomide induction and low-dose lenalidomide maintenance therapy for patients with hypomethylating agent refractory myelodysplastic syndrome. Leuk Lymphoma 2016;57(11):2535–2540. 48. Lindsley RC, Saber W, Mar BG,

et al. Prognostic Mutations in Myelodysplastic Syndrome after Stem-Cell Transplantation. N Engl J Med 2017;376(6):536–547. 49. Gangat N, Patnaik MM, Tefferi

A. Myelodysplastic syndromes: Contemporary review and how we treat. Am J Hematol 2016;91(1):76–89. 50. Duarte FB, Santos TE de JD,

Barbosa MC, et al. Relevance of prognostic factors in the decision-making of stem cell transplantation in Myelodysplastic Syndromes. Rev Assoc Medica Bras 1992 2016;62 Suppl 125–28.

51. Konuma T, Miyazaki Y, Uchida N, et al. Outcomes of Allogeneic Hematopoietic Stem Cell Transplantation in Adult Patients with Myelodysplastic Syndrome Harboring Trisomy 8. Biol Blood Marrow Transplant J Am Soc Blood Marrow Transplant 2017;23(1):75– 80.

52. Meyers J, Yu Y, Kaye JA, Davis KL. Medicare fee-for-service enrollees with primary acute myeloid leukemia: an analysis of treatment

(20)

patterns, survival, and healthcare resource utilization and costs. Appl Health Econ Health Policy 2013;11(3):275–286. 53. De Kouchkovsky I, Abdul-Hay

M. “Acute myeloid leukemia: a comprehensive review and 2016 update.” Blood Cancer J 2016;6(7):e441.

54. Vardiman JW, Thiele J, Arber DA, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood 2009;114(5):937–951. 55. Papaemmanuil E, Gerstung

M, Bullinger L, et al. Genomic Classification and Prognosis in Acute Myeloid Leukemia. N Engl J Med 2016;374(23):2209–2221. 56. Patel JP, Gönen M, Figueroa ME,

et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med 2012;366(12):1079–1089. 57. Network TCGAR. Genomic and

Epigenomic Landscapes of Adult De Novo Acute Myeloid Leukemia. N Engl J Med 2013;368(22):2059– 2074.

58. Abdel-Wahab O, Adli M, LaFave LM, et al. ASXL1 Mutations Promote Myeloid Transformation through Loss of PRC2-Mediated Gene Repression. Cancer Cell 2012;22(2):180–193. 59. Bach C, Buhl S, Mueller D,

García-Cuéllar M-P, Maethner E, Slany RK. Leukemogenic transformation by HOXA cluster genes. Blood 2010;115(14):2910–2918. 60. Inoue D, Kitaura J, Togami K, et

al. Myelodysplastic syndromes are induced by histone methylation-altering ASXL1 mutations. J Clin Invest 2013;123(11):4627–4640. 61. Katoh M. Functional and cancer

genomics of ASXL family members. Br J Cancer 2013;109(2):299–306.

62. Scheuermann JC, de Ayala Alonso AG, Oktaba K, et al. Histone H2A deubiquitinase activity of the Polycomb repressive complex PR-DUB. Nature 2010;465(7295):243– 247.

63. Nijman SMB, Luna-Vargas MPA, Velds A, et al. A genomic and functional inventory of deubiquitinating enzymes. Cell 2005;123(5):773–786. 64. Ventii KH, Devi NS, Friedrich KL,

et al. BAP1 is a tumor suppressor that requires deubiquitinating activity and nuclear localization. Cancer Res 2008;68(17):6953– 6962.

65. Fisher CL, Pineault N, Brookes C, et al. Loss-of-function Additional sex combs like 1 mutations disrupt hematopoiesis but do not cause severe myelodysplasia or leukemia. Blood 2010;115(1):38–46. 66. Wang J, Li Z, He Y, et al. Loss of

Asxl1 leads to myelodysplastic syndrome-like disease in mice. Blood 2014;123(4):541–553. 67. Abdel-Wahab O, Gao J, Adli M,

et al. Deletion of Asxl1 results in myelodysplasia and severe developmental defects in vivo. J Exp Med 2013;210(12):2641–2659. 68. Hilgendorf S, Folkerts H, Schuringa

JJ, Vellenga E. Loss of ASXL1 triggers an apoptotic response in human hematopoietic stem and progenitor cells. Exp Hematol [Epub ahead of print]. 69. Shi H, Yamamoto S, Sheng M,

et al. ASXL1 plays an important role in erythropoiesis. Sci Rep 2016;628789.

70. Inoue D, Matsumoto M, Nagase R, et al. Truncation mutants of ASXL1 observed in myeloid malignancies are expressed at detectable protein levels. Exp Hematol 2016;44(3):172–176.e1. 71. Balasubramani A, Larjo A, Bassein

JA, et al. Cancer-associated ASXL1 mutations may act as

gain-of-function mutations of the ASXL1– BAP1 complex. Nat Commun 2015;67307.

72. Thol F, Friesen I, Damm F, et al. Prognostic Significance of ASXL1 Mutations in Patients With Myelodysplastic Syndromes. J Clin Oncol 2011;29(18):2499–2506. 73. Metzeler KH, Becker H, Maharry

K, et al. ASXL1 mutations identify a high-risk subgroup of older patients with primary cytogenetically normal AML within the ELN Favorable genetic category. Blood 2011;118(26):6920–6929. 74. Schnittger S, Eder C, Jeromin S,

et al. ASXL1 exon 12 mutations are frequent in AML with intermediate risk karyotype and are independently associated with an adverse outcome. Leukemia 2013;27(1):82–91. 75. Gelsi-Boyer V, Brecqueville M,

Devillier R, Murati A, Mozziconacci M-J, Birnbaum D. Mutations in ASXL1 are associated with poor prognosis across the spectrum of malignant myeloid diseases. J Hematol Oncol 2012;5(1):12. 76. Bejar R, Stevenson K, Abdel-Wahab

O, et al. Clinical effect of point mutations in myelodysplastic syndromes. N Engl J Med 2011;364(26):2496–2506. 77. Chen T-C, Hou H-A, Chou

W-C, et al. Dynamics of ASXL1 mutation and other associated genetic alterations during disease progression in patients with primary myelodysplastic syndrome. Blood Cancer J 2014;4e177.

78. Bejar R, Lord A, Stevenson K, et al. TET2 mutations predict response to hypomethylating agents in myelodysplastic syndrome patients. Blood 2014;124(17):2705–2712. 79. Cazzola M, Della Porta MG,

Malcovati L. The genetic basis of myelodysplasia and its clinical relevance. Blood 2013;122(25):4021–4034.

(21)

1

80. Walter MJ, Shen D, Shao J, et al. Clonal diversity of recurrently mutated genes in myelodysplastic syndromes. Leukemia 2013;27(6):1275–1282. 81. Devillier R, Gelsi-Boyer V,

Brecqueville M, et al. Acute myeloid leukemia with myelodysplasia-related changes are characterized by a specific molecular pattern with high frequency of ASXL1 mutations. Am J Hematol 2012;87(7):659– 662.

82. Lai H-L, Wang QT. Additional sex combs-like 2 is required for polycomb repressive complex 2 binding at select targets. PloS One 2013;8(9):e73983.

83. Micol J-B, Abdel-Wahab O. The Role of Additional Sex Combs-Like Proteins in Cancer. Cold Spring Harb Perspect Med;6(10):. 84. Micol J-B, Duployez N, Boissel N,

et al. Frequent ASXL2 mutations in acute myeloid leukemia patients with t (8; 21)/RUNX1-RUNX1T1 chromosomal translocations. Blood 2014;124(9):1445–1449. 85. Sahtoe DD, van Dijk WJ, Ekkebus

R, Ovaa H, Sixma TK. BAP1/ ASXL1 recruitment and activation for H2A deubiquitination. Nat Commun 2016;710292. 86. Daou S, Hammond-Martel

I, Mashtalir N, et al. The BAP1/ASXL2 Histone H2A Deubiquitinase Complex Regulates Cell Proliferation and Is Disrupted in Cancer. J Biol Chem 2015;290(48):28643–28663. 87. Midgley CA, Lane DP. p53 protein

stability in tumour cells is not determined by mutation but is dependent on Mdm2 binding. Oncogene 1997;15(10):1179– 1189.

88. May P, May E. Twenty years of p53 research: structural and functional aspects of the p53 protein. Oncogene 1999;18(53):7621– 7636.

89. Levine AJ. p53, the cellular gatekeeper for growth and division. Cell 1997;88(3):323–331. 90. Venot C, Maratrat M, Dureuil C,

Conseiller E, Bracco L, Debussche L. The requirement for the p53 proline-rich functional domain for mediation of apoptosis is correlated with specific PIG3 gene transactivation and with transcriptional repression. EMBO J 1998;17(16):4668–4679. 91. Bieging KT, Mello SS, Attardi LD.

Unravelling mechanisms of p53-mediated tumour suppression. Nat Rev Cancer 2014;14(5):359–370. 92. Kuerbitz SJ, Plunkett BS, Walsh

WV, Kastan MB. Wild-type p53 is a cell cycle checkpoint determinant following irradiation. Proc Natl Acad Sci U S A 1992;89(16):7491– 7495.

93. Brosh R, Rotter V. When mutants gain new powers: news from the mutant p53 field. Nat Rev Cancer 2009;9(10):701–713. 94. Vogelstein B, Lane D, Levine AJ.

Surfing the p53 network. Nature 2000;408(6810):307–310. 95. Terzian T, Suh Y-A, Iwakuma T,

et al. The inherent instability of mutant p53 is alleviated by Mdm2 or p16INK4a loss. Genes Dev 2008;22(10):1337–1344. 96. Milner J, Medcalf EA.

Cotranslation of activated mutant p53 with wild type drives the wild-type p53 protein into the mutant conformation. Cell 1991;65(5):765–774. 97. Chin KV, Ueda K, Pastan I,

Gottesman MM. Modulation of activity of the promoter of the human MDR1 gene by Ras and p53. Science 1992;255(5043):459–462. 98. Li R, Sutphin PD, Schwartz D, et

al. Mutant p53 protein expression interferes with p53-independent apoptotic pathways. Oncogene 1998;16(25):3269–3277.

99. Schuijer M, Berns EMJJ. TP53 and ovarian cancer. Hum Mutat 2003;21(3):285–291. 100. van Ginkel JH, de Leng WWJ, de

Bree R, van Es RJJ, Willems SM. Targeted sequencing reveals TP53 as a potential diagnostic biomarker in the post-treatment surveillance of head and neck cancer. Oncotarget 2016;7(38):61575–61586. 101. Rücker FG, Schlenk RF, Bullinger

L, et al. TP53 alterations in acute myeloid leukemia with complex karyotype correlate with specific copy number alterations, monosomal karyotype, and dismal outcome. Blood 2012;119(9):2114–2121. 102. Greaves M, Maley CC. Clonal

evolution in cancer. Nature 2012;481(7381):306–313. 103. Yin B, Kogan SC, Dickins RA, Lowe

SW, Largaespada DA. Trp53 loss during in vitro selection contributes to acquired Ara-C resistance in acute myeloid leukemia. Exp Hematol 2006;34(5):631–641. 104. Zuber J, Radtke I, Pardee

TS, et al. Mouse models of human AML accurately predict chemotherapy response. Genes Dev 2009;23(7):877–889.

105. Welch JS, Petti AA, Miller CA, et al. TP53 and Decitabine in Acute Myeloid Leukemia and Myelodysplastic Syndromes. N Engl J Med 2016;375(21):2023–2036. 106. van der Helm LH, Berger G,

Diepstra A, Huls G, Vellenga E. Overexpression of TP53 is associated with poor survival, but not with reduced response to hypomethylating agents in older patients with acute myeloid leukaemia. Br J Haematol [Epub ahead of print].

107. Jasek M, Gondek LP, Bejanyan N, et al. TP53 mutations in myeloid malignancies are either homozygous or hemizygous due to copy number-neutral loss of heterozygosity

(22)

or deletion of 17p. Leukemia 2010;24(1):216–219. 108. Middeke JM, Herold S,

Rücker-Braun E, et al. TP53 mutation in patients with high-risk acute myeloid leukaemia treated with allogeneic haematopoietic stem cell transplantation. Br J Haematol 2016;172(6):914–922. 109. Sallman DA, Komrokji R,

Vaupel C, et al. Impact of TP53 mutation variant allele frequency on phenotype and outcomes in myelodysplastic syndromes. Leukemia 2016;30(3):666–673. 110. Goel S, Hall J, Pradhan K, et al.

High prevalence and allele burden-independent prognostic importance of p53 mutations in an inner-city MDS/AML cohort. Leukemia 2016;30(8):1793–1795. 111. Zatkova A, Merk S, Wendehack M,

et al. AML/MDS with 11q/MLL amplification show characteristic gene expression signature and interplay of DNA copy number changes. Genes Chromosomes Cancer 2009;48(6):510–520. 112. Olive KP, Tuveson DA, Ruhe ZC,

et al. Mutant p53 gain of function in two mouse models of Li-Fraumeni syndrome. Cell 2004;119(6):847– 860.

113. Zhu J, Sammons MA, Donahue G, et al. Gain-of-function p53 mutants co-opt chromatin pathways to drive cancer growth. Nature 2015;525(7568):206–211. 114. Walerych D, Lisek K, Sommaggio

R, et al. Proteasome machinery is instrumental in a common gain-of-function program of the p53 missense mutants in cancer. Nat Cell Biol 2016;18(8):897–909. 115. Lambert JMR, Gorzov P, Veprintsev

DB, et al. PRIMA-1 reactivates mutant p53 by covalent binding to the core domain. Cancer Cell 2009;15(5):376–388. 116. Li D, Marchenko ND, Schulz R,

et al. Functional inactivation of

endogenous MDM2 and CHIP by HSP90 causes aberrant stabilization of mutant p53 in human cancer cells. Mol Cancer Res MCR 2011;9(5):577–588. 117. Alsheich-Bartok O, Haupt S,

Alkalay-Snir I, Saito S, Appella E, Haupt Y. PML enhances the regulation of p53 by CK1 in response to DNA damage. Oncogene 2008;27(26):3653– 3661.

118. Wang Z-T, Chen Z-J, Jiang G-M, et al. Histone deacetylase inhibitors suppress mutant p53 transcription via HDAC8/YY1 signals in triple negative breast cancer cells. Cell Signal 2016;28(5):506–515. 119. Li D, Marchenko ND, Moll

UM. SAHA shows preferential cytotoxicity in mutant p53 cancer cells by destabilizing mutant p53 through inhibition of the HDAC6-Hsp90 chaperone axis. Cell Death Differ 2011;18(12):1904–1913. 120. Garcia-Manero G, Yang H,

Bueso-Ramos C, et al. Phase 1 study of the histone deacetylase inhibitor vorinostat (suberoylanilide hydroxamic acid [SAHA]) in patients with advanced leukemias and myelodysplastic syndromes. Blood 2008;111(3):1060–1066. 121. Parrales A, Ranjan A, Iyer SV, et al. DNAJA1 controls the fate of misfolded mutant p53 through the mevalonate pathway. Nat Cell Biol 2016;18(11):1233–1243. 122. Weisz L, Damalas A, Liontos M, et

al. Mutant p53 Enhances Nuclear Factor κB Activation by Tumor Necrosis Factor α in Cancer Cells. Cancer Res 2007;67(6):2396–2401. 123. Di Como CJ, Gaiddon C, Prives

C. p73 function is inhibited by tumor-derived p53 mutants in mammalian cells. Mol Cell Biol 1999;19(2):1438–1449. 124. Gaiddon C, Lokshin M, Ahn J,

Zhang T, Prives C. A subset of tumor-derived mutant forms of p53 down-regulate p63 and p73

through a direct interaction with the p53 core domain. Mol Cell Biol 2001;21(5):1874–1887. 125. Flores ER, Sengupta S, Miller JB,

et al. Tumor predisposition in mice mutant for p63 and p73: evidence for broader tumor suppressor functions for the p53 family. Cancer Cell 2005;7(4):363–373. 126. Ganley IG, Lam DH, Wang J,

Ding X, Chen S, Jiang X. ULK1. ATG13.FIP200 complex mediates mTOR signaling and is essential for autophagy. J Biol Chem 2009;284(18):12297–12305. 127. Hara T, Takamura A, Kishi C, et al.

FIP200, a ULK-interacting protein, is required for autophagosome formation in mammalian cells. J Cell Biol 2008;181(3):497–510. 128. Parzych KR, Klionsky DJ.

An overview of autophagy: morphology, mechanism, and regulation. Antioxid Redox Signal 2014;20(3):460–473. 129. He C, Klionsky DJ. Regulation

mechanisms and signaling pathways of autophagy. Annu Rev Genet 2009;4367–93.

130. Mijaljica D, Prescott M, Devenish RJ. The intriguing life of autophagosomes. Int J Mol Sci 2012;13(3):3618–3635. 131. Yang Z, Klionsky DJ. An overview

of the molecular mechanism of autophagy. Curr Top Microbiol Immunol 2009;3351–32. 132. Yorimitsu T, Klionsky DJ.

Autophagy: molecular machinery for self-eating. Cell Death Differ 2005;12 Suppl 21542–1552. 133. Warr MR, Binnewies M, Flach

J, et al. FOXO3A directs a protective autophagy program in haematopoietic stem cells. Nature 2013;494(7437):323–327. 134. Gomez-Puerto MC, Folkerts H,

Wierenga ATJ, et al. Autophagy Proteins ATG5 and ATG7 Are Essential for the Maintenance of

(23)

1

Human CD34(+) Hematopoietic Stem-Progenitor Cells. Stem Cells Dayt Ohio 2016;34(6):1651–1663. 135. Mizushima N. Autophagy:

process and function. Genes Dev 2007;21(22):2861–2873. 136. Kroemer G, Mariño G, Levine

B. Autophagy and the integrated stress response. Mol Cell 2010;40(2):280–293. 137. Mortensen M, Soilleux EJ,

Djordjevic G, et al. The autophagy protein Atg7 is essential for hematopoietic stem cell maintenance. J Exp Med 2011;208(3):455–467. 138. Folkerts H, Hilgendorf S, Wierenga

ATJ, et al. Inhibition of autophagy as a treatment strategy for p53 wild-type acute myeloid leukemia. Cell Death Dis 2017;8(7):e2927.

Referenties

GERELATEERDE DOCUMENTEN

Whether a stem cell divides symmetrically or asymmetrically depends on intrinsic and extrinsic factors. Research has confirmed that the interplay with the bone marrow

On day 24, cells with a double knockdown had similar cumulative cell count to control cells and a significantly higher cell count than ASXL1 single cells (p&lt;0.05)... TP53

(A) Left panel: table showing relative expression of ATG5, ATG7 or p53 as determined by q-PCR in shSCR, shATG5, shATG7 and shP53 transduced cell lines or right panel: p53 protein

Another pathway involved in autophagy-mediated resistance to therapy is the MAPK pathway, with chemotherapeutic treatment of hepatocellular carcinoma cell lines leading to increased

Poor-risk AMLs are often associated with TP53 mutations but knockdown or overexpression of wild-type or mutant TP53 did not affect the flux.. When using hydoxychloroquine (HCQ),

Autophagy in cancer associated fibroblasts promotes tumor cell survival: Role of hypoxia, HIF1 induction and NFκB activation in the tumor stromal microenvironment..

Expression of ASXL1 mutations rather than knockdown of ASXL1 may lead to transformation in xenografts – Inoue (J Clin Invest 2013), Inoue (JEM 2018), this thesis8. AMLs with

To arrive at the T cell score, vaccination-induced T cell immune responses were scored in four different categories, i.e., (1) T cell proliferative response to the DCP-001, (2)