• No results found

Modeling of MLL-AF9-rearranged pediatric leukemia

N/A
N/A
Protected

Academic year: 2021

Share "Modeling of MLL-AF9-rearranged pediatric leukemia "

Copied!
233
0
0

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

Hele tekst

(1)

University of Groningen

Modeling of MLL-AF9-rearranged pediatric leukemia Carretta, Marco

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):

Carretta, M. (2018). Modeling of MLL-AF9-rearranged pediatric leukemia: Identification of mechanisms and potential targets. University of 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)

!!

!

!

!

!

!!

!

!

!

Modeling of MLL-AF9-rearranged pediatric leukemia

Identification of mechanisms and potential targets

Marco Carretta

(3)

The studies described in this thesis were financially supported by the grant: Marie Curie Euro Cancer Stem Cell Training Network FP7-PEOPLE-2010-ITN-264361.

The publication of this thesis was financially supported by University of Groningen.

Cover design: Oscar ‘Odd’ Diodoro, odd-house.com Printed by: Ridderprint BV, www.ridderprint.nl

ISBN 978-94-034-0440-0 (printed) ISBN 978-94-034-0439-4 (e-book)

Copyright © 2018 by Marco Carretta. All rights reserved. No parts of this book may be reproduced or transmitted in any form or by any means without prior permission of the author.

(4)

!

Modeling of MLL-AF9-rearranged pediatric leukemia

Identification of mechanisms and potential targets

PhD thesis

to obtain the degree of PhD at the University of Groningen

on the authority of the Rector Magnificus Prof. E. Sterken

and in accordance with

the decision by the College of Deans.

This thesis will be defended in public on Monday 5 February 2018 at 14.30 hours

by

Marco Carretta born on 7 May 1985

in Mantova, Italië

(5)

Supervisors Prof. J.J. Schuringa Prof. E. Vellenga

Assessment Committee Prof. S. de Jong

Prof. G. de Haan Prof. J. Jansen

!

(6)

!

Alla mia famiglia

(7)

Paranymphs Vincenzo Terlizzi Bauke de Boer

(8)

!

TABLE OF CONTENTS

CHAPTER 1 9

General introduction and scope of the thesis

CHAPTER 2 45

Modeling BCR-ABL and MLL-AF9 leukemia in a human bone marrow-like scaffold-based xenograft model

Leukemia. 2016 Oct;30(10):2064-2073

CHAPTER 3 89

Genetically engineered mesenchymal stromal cells produce IL-3 and TPO to further improve human scaffold-based xenograft models

Exp Hematol. 2017 Jul;51:36-46.

CHAPTER 4 123

BRD3/4 inhibition and FLT3-ligand deprivation target pathways that are essential for the survival of human MLL-AF9+ leukemic cells

PLoS One. 2017 Dec 14;12(12):e0189102.

CHAPTER 5 159

The cell of maintenance in MLL-rearranged B-ALL is multipotent and retains myeloid potential while MLL-rearranged AML is maintained by myeloid restricted LSCs

Manuscript in preparation

CHAPTER 6 183

Summary, general discussion and future perspectives

CHAPTER 7 211

Nederlandse samenvatting Acknowledgements

Curriculum Vitae List of publications

(9)
(10)

!

!

!

!

!

!!

!

!

!

CHAPTER

1

General introduction and scope of the thesis

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!!

!

!

!

!

!

!

(11)

Introduction

Over the last decades, the continuous advancement of in vitro and in vivo models has greatly improved our understanding of normal and malignant hematopoiesis. The development of progressively more immunodeficient mice and xenotransplantation models has led to the identification of normal and leukemic hematopoietic stem cells, the detailed characterization of the hierarchical organization of human hematopoiesis, and also provided pre-clinical models for drug testing. Insights from these models also directly contributed to transplantation procedures and treatment options in the clinical practice. In this chapter, a general introduction on normal and malignant hematopoiesis is provided, with a particular focus on MLL-rearranged leukemias. Furthermore, the in vitro and the in vivo models that are being used to study leukemia are reviewed from an historical perspective. Lastly, the general scope and aim of this thesis are outlined.

!

!

!

!

!!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

(12)

CHAPTER 1 11 Hematopoietic stem cells and their niche

Hematopoiesis is defined as the formation and development of blood cells. This essential process, which in adults occurs mainly in the bone marrow, ensures the daily generation of over 500 billion cells in response to the turnover of the short-lived red and white blood cells. The production of this impressive amount of new blood cells can even further increase in case of bleeding, infection or pathological loss, making the hematopoietic system one of the most actively regenerating tissues of the whole body 1,2. Despite the heterogeneity in both morphology and function of all the mature elements present in the blood, all these different cells originate from a rare population of primitive hematopoietic stem cells (HSCs). Adult HSCs possess multi- lineage potential, which is the ability to develop into any of the three types of blood cells: red cells, white cells and platelets. Furthermore, HSCs possess self-renewal capacity, which maintains the stem cells pool and ensures the continuous production of blood cells throughout the lifetime of an organism. Decades of studies on the hematopoietic system in mice and humans showed that stem cells are rare and quiescent and they display heterogeneity with respect to their self-renewal potential.

With the use of specific molecular surface markers, in mice HSCs were subdivided in long-term reconstituting HSCs (LT-HSCs) and short-term reconstituting HSCs (ST- HSCs) 1,3–5. Long-term HSCs are considered to be the least abundant and most quiescent population, only diving few times during life and able to reconstitute lethally irradiated mice for the lifetime of the animal. Analysis of the kinetics of BrdU incorporation revealed that over 99% of LT-HSCs incorporated BrdU after long periods of administration and it was postulated that LT-HSCs enter the cell cycle and divide on average every 57 days 6,7. In a more recent study by Wilson and colleagues

8, mathematical modelling of the results obtained from pulse-chase experiments

(13)

combined with the use of six different molecular markers phenotypically identifying LT-HSCs (Lin-, Sca+, cKit+, CD150+, CD48-, and CD34-) uncovered the existence of a subpopulation of dormant HSCs, which represented about one seventh of the studied population. These HSCs divide every 145 days, which is equivalent to five divisions per average C57/BL6 mouse lifetime 9.

The direct descendants of the LT-HSCs are the so-called short-term HSCs (ST- HSCs) and multipotent progenitors (MPPs). While these two cell populations still possess multilineage potential, they appear to have less self-renewal potential and are able to reconstitute lethally irradiated mice for only short periods of time.

However, all the evidence supporting these concepts has been obtained by using in vivo transplantation assays and therefore they might not reflect the real mechanisms

governing life-long blood cell production in unperturbed animals. Recent papers suggested that in a native setting without transplantation, where murine cells were uniquely and genetically labelled in situ, also committed progenitors can contribute to life-long hematopoiesis rather than classically defined hematopoietic stem cells 10,11. However, this concept of progenitor cells instead of LT-HSCs that sustain life-long hematopoiesis was again recently challenged by a study showing that, by genetic labelling of a subset of self-renewing LT-HSCs in the adult bone marrow, it is the most primitive HSCs that sustain endogenous hematopoiesis 12. No doubt, further future studies will be aimed at clarifying these different observations.

Upon progression down the hematopoietic tree, cells become more committed towards either the lymphoid (Common Lymphoid Progenitors; CLPs) or myeloid lineage (Common Myeloid Progenitors; CMPs). In the last stage of differentiation, CLPs generate B- and T-lymphocytes as well as Natural Killer (NK) and dendritic cells. On the other side, CMPs give rise to megakaryocyte erythrocyte progenitors

(14)

CHAPTER 1 13 (MEPs) that will terminally differentiate in erythrocytes and platelets, and give rise to granulocyte-macrophage progenitors (GMPs) that will differentiate into the mature granulocytes and macrophages. More recent studies have shown that the hierarchical organization of the hematopoietic tree might be different, whereby HSCs differentiate into a previously unidentified population of lymphoid-primed multipotent progenitors (LMPPs), which possess lymphoid, granulocyte and macrophage differentiation potential. Furthermore, an early MEP population would be able to give rise to megakaryocytes and erythrocytes 13–17.

Figure 1 Hierarchical organization of the hematopoietic tree Long Term Hematopoietic Stem Cells (LT-HSCs); Short Term Hematopoietic Stem Cells (ST- HSC); Multipotent Progenitor (MPP); Lymphoid-primed Multipotent Progenitor (LMPP); Common Lymphoid Progenitor (CLP); Common Myeloid Progenitor (CMP);

Granulocyte-Macrophage Progenitor (GMP); Megakaryocyte-Erythrocyte Progenitor (MEP).

(15)

In adults, HSCs are residing in a specific anatomical location in the bone marrow denominated the hematopoietic stem cell niche 18. These niches are constituted of various cell types, extracellular matrix and soluble chemical factors. The niche provides a specific physiological microenvironment that plays a key role in the regulation of the balance between differentiation and self-renewal of the HSCs. Early studies proposed that HSCs are mainly located in the endosteal region, but more recent studies have shown that quiescent HSCs are also perivascular and distributed throughout the bone marrow 19,20. In the perivascular space, several cell types have been associated with HSC regulation, including megakaryocyte 21, endothelial cells

22, arteriolar pericytes 23, sympathetic nerves 24 and non-myelinating Schwann cells

25. The perivascular space in the BM also includes perivascular mesenchymal stromal cells (MSCs) defined as CXCL12-abundant reticular (CAR) cells 26. CAR cells express high levels of niche factors such as CXCL12, also known as stroma derived factor 1 (SDF-1). By binding to the CXCR4 receptor, SFD-1 plays an important role in HSCs homing and retention in the BM 27. Furthermore, it has also been shown that CAR cells in the BM are producing stem cell factor (SCF), a crucial cytokine involved in the maintenance and proliferation of HSCs 26,28. In human BM, CD146-expressing cells might represent the counterpart of murine CAR cells 29. The role of osteolineage cells in HSC maintenance is still debated, but current evidence suggests that immature osteolineage cells rather than mature osteoblasts may contribute to the regulation of HSCs 30. Intriguingly, in a study published by Ding et al.31 it has been shown that deletion of CXCL12 from perivascular stromal cells depleted HSCs and certain restricted progenitors and mobilized these cells into circulation. Deletion of CXCL12 from osteoblasts instead depleted certain early lymphoid progenitors, but not HSCs or myelo-erythroid progenitors and did not

(16)

CHAPTER 1 15 mobilize these cells into circulation. The authors concluded that different stem/progenitor cells occupy distinct cellular niches in the BM: HSCs are residing in a perivascular niche and early lymphoid progenitors in an endosteal niche.

Collectively, these findings are contributing to a better understanding of the hematopoietic BM niche but several aspects of HSC-niche interaction still remain to be elucidated 32.

Leukemic stem cells and their niche

In normal hematopoiesis, the differentiation of healthy HSCs into mature blood elements is a highly regulated and quantitatively controlled process. Disruption of these regulatory mechanisms by sequential acquisition of genetic alterations and/or epigenetic changes in stem or progenitor cells will first interfere with cell production and then lead to clonal expansion. Ultimately, such expanding clones may result in the development of leukemia.

Different types of leukemia can be distinguished according to their lineage, aggressiveness and dynamics. In acute leukemia, the disease evolves rapidly and aberrant blood cells are blasts that remain immature and cannot carry out their normal functions. Clinical signs of acute leukemia are derived from the infiltration of leukemic clones in the BM, which mainly causes cytopenia, impaired hemostasis and impaired immune functions. In chronic leukemia instead, the progression of the disease is slower and while some blast cells are present, most cells are mature and can carry out some of their normal functions. Both acute and chronic leukemia can develop along the myeloid or lymphoid lineages, giving rise to acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL), chronic myeloid leukemia (CML) or chronic lymphocytic leukemia (CLL) 33.

(17)

For these hematological malignancies there is experimental evidence supporting the concept of the cancer stem cell (CSC), which is defined as a subset of neoplastic stem cells that propagate malignant clones indefinitely and give rise to cancer 34. The existence of leukemic stem cells (LSC) was first shown in AML by John Dick, which represented the first direct identification of a cancer driven by cancer stem cells 35. In this study, non-obese diabetic, severely combined immunodeficiency disease (NOD/SCID) mice were used as a model to study the engraftment and proliferation of primary human patient-derived leukemic cells. By transplanting sorted CD34+CD38 cells, which often represents only 0.1 to 1% of the total AML cell population, the leukemic cell morphology found in patients was recapitulated, whereas the CD34+CD38+ and CD34 fractions contained no cells with these properties. This study conclusively established that, similarly to the healthy counterpart, also leukemia is hierarchically organized and sustained by a rare population of LSCs. However, consecutive studies have highlighted that LSCs may also reside in other compartments than the CD34+CD38- compartment. For example, LSCs were also found in the CD34+CD38+ cells as well as CD34- fraction in the majority of NPM1-mutated AML 36–38. In recent years, LSCs were found in even more mature fractions; for example, oncogenes such MLL fusion proteins can re-install aberrant self-renewing properties in committed MPPs and transform them into LSCs

39,40.

While for different AML subtypes the characterization of the LSCs has progressed significantly, the phenotype of LSCs in ALL is less clear. One study suggests that for pre–B cell lineage ALL the LSCs display an immature phenotype lacking the expression of CD19 41, whereas others showed that ALL-LSCs are more mature and do express CD19 42–44. Currently, little is known about similarities and differences

(18)

CHAPTER 1 17 between LSCs that maintain myeloid versus lymphoid malignancies, an aspect that is addressed in the context of MLL-rearranged leukemia in Chapter 5 of this thesis.

The experimental evidence so far suggests that leukemia, in analogy to normal hematopoiesis, is hierarchically organized and is produced by relatively few LSCs that are impaired in their differentiation potential. Regardless of the ‘cell of origin’

(HSCs or more committed progenitors), tumorigenic leukemic cells are able to give rise to phenotypically diverse progeny including a few LSCs with indefinite proliferation potential, as well as cells within the leukemia that may have limited or no proliferative potential 45. A better understanding of leukemia biology is fundamental to improve therapeutic strategies. Current approaches to cure leukemia tend in fact to target the bulk of the tumor cells, but are not effectively targeting the pool of LSCs.

Most LSCs are quiescent the majority of the time, and are therefore resistant to standard chemotherapeutic options that target cells undergoing replication.

While we have learned a lot about the role of the niche in normal hematopoiesis, the exact role of the niche in leukemogenesis is still less clear and only in recent years experimental work has started to investigate specific roles of the microenvironment in the pathophysiological context. In spite of the technical limitations which still hinder our understanding of the BM niche physiology, it is becoming increasingly clear that BM stromal changes and the formation of a self-reinforcing malignant niche is more than a mere bystander effect of disease development and can directly contribute to myeloid malignancies 46. Stromal abnormalities and structural changes in the BM cavities due to impaired angiogenesis and bone-loss are documented in both AML and Myelodysplastic Syndrome (MDS) 47. A growing body of literature has investigated how leukemic cells remodel their niche into an aberrant environment that provides support to malignant cells at the expense of normal hematopoietic cells.

(19)

BM-derived MSCs from AML patients have been shown to have a decreased production of CXCL12, which could potentially affect also the maintenance of healthy HSCs 48. More recently, intra vital imaging and xenograft transplantation were used to show how human leukemic cells drive the formation of specialized malignant niches that directly impair healthy HSC function due to overproduction of Stem Cell Factor (SCF) 49,50. Another concept that has emerged is the possible existence of different niches between CML and AML. For example, experimental evidence suggests that while parathyroid hormone impaired CML growth, it augmented MLL- AF9-induced AML proliferation 51. Furthermore, Schepers et al. 52 showed that CML cells stimulate MSCs to proliferate and adopt an abnormal differentiation program resulting in the overproduction of functionally altered osteoblastic cells, which accumulate in the BM cavity as inflammatory myelofibrotic cells. For AML instead, it has been reported that leukemic cells create neuropathic changes in the BM niche, which affect the activity of perivascular MSCs and alter the function of the HSC niche

53,54. It is thus clear that the disease can be the initiator of niche changes but, conversely, genetic alterations of the niche may also represent driving mutations during malignant transformation. In support of this concept, it has been shown that targeted deletion of the miRNA-processing endonuclease Dicer1 from osteoprogenitor cells disrupts the integrity of the hematopoietic system, recapitulating key features of human myelodysplastic syndrome. This included the propensity to develop, although in low frequency, AML55,56. Lastly, but importantly, the leukemic niche in the BM is shown to support LSC in the maintenance of their quiescent state, thus contributing to protect the cells from chemotherapeutic agents 57–59.

Taken these pieces of evidence together, it immediately becomes clear that manipulating the leukemic niche might represent an advantageous therapeutic

(20)

CHAPTER 1 19 strategy, particularly in malignancies for which targeting the hematopoietic cells has proven inefficient.

Characteristics of MLL-rearranged leukemias

Acute leukemia is a highly heterogeneous disease and comprises a wide variety of cytogenetic aberrations and molecular mutations. To date, hundreds of genetic markers have been identified, serving as basis for the categorization of cases, prediction of prognosis and therapeutic strategies.

Due to their unique biological and clinical features, MLL-related leukemias are of particular interest. The MLL (Mixed Lineage Leukemia) gene on chromosome 11q23 encodes for a Histone-Lysine N-methyltransferase, an epigenetic regulator which plays an essential role in early development and hematopoiesis. Chromosomal translocations bearing the MLL gene define a unique group of leukemias, which can give rise to AML, ALL or biphenotypic (mixed lineage) leukaemia (BAL). MLL-related leukemias account for approximately 70% of infant leukemia 60, 10% of adult AML and are frequently found in therapy-related acute leukemia cases that arise following treatment with topoisomerase II inhibitors 61. Patients with MLL-rearranged AML have a similar poor prognosis as others AML, but children with MLL-rearranged ALL have a particularly poor outcome compared with children with other forms of ALL. At a molecular level, two main aberrations of the MLL gene are found. One of these creates a short repeat within the N-terminal MLL coding sequence which results in an internal partial tandem duplication (PTD). Thus, an extra amino-terminus is added in- frame to full-length MLL, resulting in MLL-PTD driven myeloid dysplastic syndrome (MDS) or AML. In the other type of disruption of the MLL gene, the genomic sequences encoding the N-terminal portion of MLL are fused to sequences encoding

(21)

the C-terminus of another translocation partner. These types of translocation are the most common and to date, nearly 100 different chromosome bands have been described in rearrangements involving chromosome 11q23 and more than 70 fusion genes have been characterized at the molecular level 62.

Figure 2 Distribution of major MLL fusion partner genes in de novo childhood and adult leukemias Mixed lineage leukaemia (MLL) rearrangements are found in approximately 5% of acute lymphoblastic leukaemias (ALL), approximately 5–10% of acute myeloid leukaemias (AML) and virtually all cases of mixed lineage (or

biphenotypic) leukaemias (MLL). Adapted from 63

Among the numerous fusion translocation partners, four out of the five most frequent MLL rearrangements belong to a family of serine/proline rich nuclear proteins (AF4, AF9, AF10, ENL). Different MLL fusion can give rise to different leukemia subtypes

(22)

CHAPTER 1 21 and the same translocation can give rise to different leukemia in pediatric or adult patients. For example, the MLL-AF9 fusion gene is of particular interest since it is found in pediatric cases in similar proportions between AML (33%) and ALL cases (18%) however, for adults this translocation is mainly found in AML (32%) compared to ALL (2%) (Fig.2). MLL fusions are transcriptional regulators that take control of targets normally controlled by MLL. In the wild type form of the MLL gene, the SET domain confers H3K4 methyltransferase activity, allowing transcription initiation by polymerase II. When the MLL gene is fused with one of its partners, the SET domain is in most cases lost together with its catalytic activity. However, MLL fusion proteins gain the ability to methylate H3K79, which results in an aberrant gene expression of homeobox genes such HOXA9 and MEIS1 64. The H3K79 methyltransferase DOT1L and the MLL-interacting protein Menin have emerged as important mediators of MLL fusion-mediated leukemic transformation. In work by Okada et al. 65, a unique function of an MLL fusion (MLL–AF10) was identified for the first time. The recruitment of the H3K79 methyltransferase DOT1L by AF10 is in fact shown to be indispensable for transformation and responsible for the aberrant H3K79 methylation at the HOXA9 locus. Other MLL fusion partner genes, for example the ones involved in transcriptional elongation such AF4 and AF9, were also reported to interact directly with DOT1L 66,67. Furthermore, fusion partner genes such ELL, ENL and others have been reported to interact with DOT1L via common binding proteins 68,69. Although inhibition of DOT1L is currently under clinical investigation, downstream signalling is still far from clear and a more detailed characterization of molecular mechanisms underlying MLL-fusion leukemia is still greatly needed. In Chapter 4 of this thesis, by taking advantage of state of the art molecular techniques, we aimed to investigate

(23)

the downstream targets in human MLL-AF9 leukemias and identify targetable signaling networks.

As briefly mentioned before, MLL fusions offer a valuable tool to study LSCs biology.

By transducing the MLL-ENL fusion oncogene in BM-isolated GMP cells, Cozzio et al. demonstrated that murine myeloid leukemia can also originate from committed

myeloid progenitors that have no intrinsic self-renewal capabilities, at least in the mouse. Despite the fact that 10 times more GMP-transduced cells were needed compared to HSC-transduced cells to generate AML, this experimental evidence shows that committed myeloid progenitor cells are also capable of being transformed into LSCs. The same concept was demonstrated also upon transduction of GMPs with MLL–AF9 fusion 40,70. Gene expression analysis revealed that these LSCs preserve the overall GMP expression profile, but the MLL fusion is capable of re- conferring a self-renewal programme normally present only in the HSCs compartment. However, another study focused on the malignant transformation initiated by the fusion gene MLL-AF9 and compared when expressed at physiologic levels in a knock-in model or at supraphysiologic levels in a retroviral model. The data from the physiologic model showed highest levels of MLL and MLL-AF9 in the most transformable HSCs and lower levels in the more resistant committed myeloid progenitor GMPs. The authors conclude that while it is possible that the transforming human MLL-AF9 translocation may take place at a maturation stage later than the HSC, murine studies suggest that this is much less likely to be functionally meaningful than a “hit” within the HSC population 71. Future studies will be necessary to further define this issue, but similar conclusion were also found by Horton et al. 72. Transcriptome analysis identified enrichment of HSC but not progenitor gene signatures in MLL-AF9-expressing cells and highlighted the differences in MLL-AF9-

(24)

CHAPTER 1 23 mediated immortalization in neonatal cells versus adult cells, reinforcing the notion that intrinsic properties of the cell of origin, in addition to extrinsic cues, might dictate lineage of the immortalized cell.

Modeling of human leukemia in vivo

The in vitro and in vivo models available for studying normal and malignant hematopoiesis have constantly improved over the last four decades, directly contributing not only to a better understanding of human biology but also significantly impacting on drug design and the development of therapeutic strategies.

Progressively improved xenograft mouse models supporting the engraftment and development of human hematopoietic cells have been generated and they now represent essential tools in the field, making the disease accessible to experimentation73–77. In this section, a historical review of the most relevant mouse xenograft models and in vitro assays to study leukemia are provided.

In order to generate humanized mice in which successful xenotransplantation can be achieved three major requirements need to be met. First, the recipient mouse needs to be immunodeficient in both its adaptive and innate immune compartments in order to tolerate the human graft and prevent xeno-rejection. Second, an appropriate spatial location (or niche) needs to be provided in the bone marrow, in which transplanted human cells can home and develop. Lastly, the host needs to provide the appropriate cross-reactive human growth factors, cytokines and major histocompatibility complex molecules that can act on human cells 76.

The development of mouse strains that support human hematopoiesis and AML in vivo started in the late 1960’s and a schematic historical overview of the development of the different immunodeficient mouse models is shown in Figure 3.

(25)

Figure 3 History of the development of immunodeficient mice for establishing leukemia mice models (Adapted from Goyama, S., Wunderlich, M., & Mulloy, J.

C. (2015). Xenograft models for normal and malignant stem cells. Blood, 125(17), 2630-2640.)

The nude mice (nu) were first generated in 1962 and they represent the first immunodeficient mouse strain ever described and utilized for xenotransplantation 78. These mice are athymic and lack functional T and – only partially - B cells 79. Initial attempts to engraft human AML consisted in the subcutaneous injection of BM derived leukemic cells. However, even when the nude mice were immunosuppressed with radiation, transplantation of leukemic tissue proved to be inconsistent and limited to the formation of localized myelosarcomas with little evidence of BM engraftment

80Moving to the late 1980s, in an attempt to further improve immunodeficient models, beige mice (bg) and X-linked immunodeficiency (xid) models were generated 81–83. In the bg/nu/xid model, mice are athymic and have a reduced number of natural killers (NK) and so-called lymphokine activated killer cells. Engraftment of human myeloid

(26)

CHAPTER 1 25 cells indicated better seeding, proliferation and differentiation of human stem cells, but attempts to engraft peripheral blood derived human cells proved difficult84,85. In 1983 Bosma et al. described the SCID (severe combined immunodeficiency) mice, which are deficient in mature B and T cells. In this model, germ line DNA segments encoding immunoglobulin and T lymphocyte antigen receptor molecule fail to undergo rearrangement resulting in lack of functional antigen receptors 86. The first successful xenotransplantation of AML into SCID was reported in 199187, followed by several other studies demonstrating the superiority of this model over the be/nu/xid

88,89. In spite of the clear benefits of the SCID model, the lack of cross-reactive human cytokines and innate host resistance still posed major practical limitations in modeling AML in vivo 87,90–96. Researchers have tried to overcome these issues by injecting endogenous human cytokines and growth factors such as IL3, IL6, GM-CSF and EPO into the SCID recipients 97–99. However, although this approach was simple and resulted in enhanced engraftment, it is expensive, time-consuming and not suited for long-term experiments. Furthermore, antigen-presenting and NK cells were not affected in the SCID strains, and moreover, SCID mice occasionally express mature B- or T-cells indicating a certain degree of ‘leakiness’ of the system

100,101. Further improvements of this immunodeficient strain led to the generation of the NOD/SCID mouse strain.

The NOD/SCID mice were generated by backcrossing the SCID gene onto the non- obese diabetic (NOD) background and displayed impaired innate and adaptive immunological functions. Besides lacking mature B- and T-cells, NOD/SCID mice possess impaired NK and antigen-presenting cell functions. Moreover, these mice are insulitis- and diabetes-free throughout their lifespan 102. These mice showed appreciably higher engraftment rate also when a limited amount of cells were injected

(27)

103,104. Importantly, the NOD/SCID model allowed the formal demonstration of the concept of leukemic stem cells (LSCs) 104. In fact it has been shown that only a rare subset of the most immature cells termed SCID-Leukemia Initiating Cells (SL-ICs) is capable of initiating and sustaining leukemic growth in vivo. SL-ICs are able to recapitulate the morphological, genotypic and biological characteristics of the donor and, as demonstrated in serial transplantation experiments, possess high self- renewal capacity 104,105. Despite the clear advantages of the NOD/SCID model, major limitations are represented by the frequent occurrence of thymic lymphoma and spontaneous NK cells activity, which frequently hindered long-term enduring engraftment 102.

In more recent years, in order to overcome the limitations of the previous models, NSG (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ) 106 and NOG (NOD/Shi-scid/IL-2Rγnull)107 mice have been developed. In these mice the complete deficiency of NK cells is achieved by disrupting the signalling downstream of IL-15, which is essential for the NK-cell development and maturation. The common gamma chain of cytokine receptor (IL2Ry) is shared by multiple members of the IL2 family of cytokines, including IL15. Thus, deficiency in the IL2Ry gene results in the complete absence of NK cells and limits the spontaneous development of murine thymoma, a phenomenon often observed in NOD/SCID mice 57,108–110. Several reports have highlighted the superiority in engraftability of AML cells compared to previous models

110–112 and to date the NSG model is still considered the ‘golden standard’ for xenograft models. The generation of immunodeficient mouse strains like the NSG allowed researchers to functionally define hematopoietic stem cells and their malignant counterpart and also to serve as preclinical models for drug testing.

(28)

CHAPTER 1 27 Despite the ability of engrafting different subtypes of primary acute lymphoblastic leukemia samples, NSG mice still possess major limitations for myeloblastic leukemia, since the engraftment of at least 30-40% of AML is not achieved 113 and overall this model is clearly lymphoid biased 72,114. This may be explained by the high influence of the murine microenvironment and the absence of species-specific human factors over AML. In order to overcame these limitations, in the last decade researchers have developed several immunodeficient mice where the transgenic expression of cytokine-encoding genes is under the control of a strong constitutive promoter in order to provide support for human hematopoietic (or leukemic) cell development 115–119. For example, NSG transgenic mice expressing human factors such SCF, GM-CSF, IL-3, and TPO have been developed, showing an increase in the engraftability rate of primary tumors. However, one limitation of this approach consists of the non-physiological levels of human cytokines expressed by these transgenic mice that eventually lead to myeloid biasness or exhaustion of self- renewal capacity 115. Thus, novel approaches are greatly needed in order to improve the current xenotransplantation procedures and in particular the ones that consider the importance of providing multiple human factors at physiological concentrations.

During the time I performed the studies presented in this thesis, an innovative mouse model called MISTRG has been generated in which human versions of four genes encoding cytokines important for innate immune cell development (M-CSF, IL-3, GM- CSF and TPO) are knocked into their respective mouse loci 120. These mice support the development of human NK cell, myelo-monocytic cells and show a strong human innate immune response in response to viral and bacterial infections. The possible implications of the use of this mouse model for leukemia research will be discussed in the final chapter of this thesis.

(29)

Modeling of human leukemia in vitro

Although the in vivo models (in particular the NOD-SCID/NSG models) have been very instrumental, in most cases these systems have limitations such as their cost- effectiveness and duration. Thus, in order to further understand the differences in molecular mechanisms that are involved in leukemic transformation of hematopoietic stem cells, accessible assays are also required, in which gene-function analyses of the leukemic stem cell can be performed and in which phenotypes such as long-term expansion, self-renewal, and apoptosis can be monitored. It is clear that self-renewal of normal stem cells heavily depends on extrinsic cues they obtain from the bone marrow microenvironment. Culture systems aiming to recapitulate the bone marrow microenvironment are thus essential to study niche-mediated regulation of normal and malignant hematopoietic stem cells at a molecular level and much effort has been put in the development of such in vitro systems.

Over three decades ago, Dexter et. al developed a 2D co-culture system where murine HSC where cultured on a basal cell layer comprised of mixed BM stromal cells obtained by flushing out the marrow from mouse femurs and sub-culturing the adherent cell fraction121. Viability and stemness of murine HSCs cultured on stroma were improved and cultures could be maintained over longer periods of time (7–12 weeks) compared to pure HSC cultures, especially when the culture medium was supplemented with cytokines. The Dexter model paved the way for many HSC and AML ex vivo culture models that have been developed subsequently.

AML cocultures with bone marrow stromal and supplemented with human cytokines could also be established, in which a long-term leukemic expansion for 7 to >24 weeks could be achieved122–132. In these assays, Leukemic Cobblestone Area Forming Cells (L-CAFCs) readily form and self-renewal capacity can be addressed

(30)

CHAPTER 1 29 by serial replating of cultures onto new stroma, whereby new L-CAFCs are generated. This in vitro assay also allows for the evaluation of leukemic stem cell markers that identify populations with long-term self-renewal potential 122,128,131. Moreover, it is also possible to introduce or downmodulate genes in the AML LTC- ICs by lentiviral (RNAi) approaches 122,125–127,130,131,133.

Besides using patient samples to gain further insight into LSCs, model systems have been generated as well in which oncogenes such as MLL-AF9114,134,135, BCR-ABL

125,136, FLT3-ITDs137, KRAS138, STAT5 139 and NUP98-HOXA9 137 are introduced into healthy human HSCs. For many of these studies fetal cord blood (CB) CD34+ cells have been used, but distinct phenotypes can be obtained when oncogenes are introduced into fetal versus adult human CD34+ cells135. Myeloid and lymphoid transformation of transduced human CD34+ can be achieved upon changes of composition of the culturing media 72.

(31)

Scope of the thesis

As discussed in this introductory chapter, mouse xenograft models have significantly contributed to a better understanding of normal and malignant hematopoiesis but major limitations still remain in the currently available and widely used xenograft models. In Chapter 2 of this thesis, we describe a novel in vivo approach aimed to reconstruct a humanized BM microenvironment in immunodeficient mice. We hypothesized that the presence of a human niche would provide human niche- specific factors that might still be missing in the murine recipient and would also provide interaction of the hematopoietic cells with various niche components. To test this hypothesis, we focus our studies on two leukemic CB models that possess distinct biology: BCR-ABL and MLL-AF9. For BCR-ABL, a serially transplantable CD19+ B-ALL could be induced in NSG mice only when additional hits were provided, such as co-expression of BMI1. In this study, we wished to evaluate if in the humanized BM xenotransplant model would allow BCR-ABL-only expressing cells to engraft and induce leukemia. Also for MLL-AF9, a serially transplantable CD19+ B- ALL could be induced in NSG mice while the AML transformation was less frequently observed. Since the susceptibility of MLL-AF9 to microenvironmental cues had already been shown 140, we wanted to evaluate if the presence of a human BM niche would favor myeloid engraftment. Furthermore, we also set out to test the engraftment efficiency of primary material derived from patients with the same mutations. Lastly, we investigated whether this model could also be used to evaluate the efficacy of inhibitors in vivo.

The ectopic implantation of a humanized BM niche in immunodeficient mice allowed the faithful recapitulation of some of the key features of BCR-ABL and MLL-AF9 leukemia. Furthermore, in parallel studies from our lab, we showed that the BM

(32)

CHAPTER 1 31 humanized model allowed the engraftment of leukemic samples that failed to engraft in NSG mice without humanized niches and resulted in a better preservation of leukemic stem cell self-renewal properties 141. Although the presence of an ectopic human BM niche presented clear advantages compared to normal NSG mice, some key issues still remain such for example the engraftment of myeloid transformed cells in secondary recipients. Transcriptome studies of primary human BM-derived MSCs revealed that a variety of cytokines and growth factors are produced, but some critically important cytokines such IL3 and TPO are not. In Chapter 3, we investigated whether we could genetically engineer MSCs to produce such factors and evaluated these modified MSCs in vitro and in vivo in humanized scaffold xenograft models.

In Chapter 4, we aimed to identify targetable signaling networks by taking advantage of our in vitro and in vivo models for human MLL-AF9 leukemia. Progress in DNA- sequencing technologies has reinforced the notion that cancer is initiated and maintained by alterations in the genome and it has also become more evident that epigenetic regulators are among the most frequent aberrancies in hematopoietic malignancies 142. Here, we show that MLL-AF9 cells critically depend on FLT3-ligand induced pathways as well as on BRD3/4 for their survival. We evaluated the in vitro and in vivo efficacy of the BRD3/4 inhibitor I-BET151 in various human MLL-AF9 (primary) models and patient samples and analyzed the transcriptome changes following treatment

Switches between lymphoid and myeloid lineages can occur during treatment or during relapse of pediatric leukemia patients bearing MLL-translocations. The mechanisms behind these phenomena have remained elusive, but published studies suggest that both intrinsic and environmental cues may inspire changes in cell fate

(33)

decisions of leukemic stem cell clones. Although these complete lineage switches occur infrequently, it is relevant to evaluate whether the leukemic blasts originate from the same leukemic clone or whether new clones emerge as a consequence of the treatment. In chapter 5, by making use of a lentiviral MLL-AF9 CB model as well as primary MLL-rearranged pediatric patient samples, we aimed to model lineage switching in vitro and in vivo in xenograft mice. Furthermore, we used ligation mediated PCR (LM-PCR) and RNA-sequencing to investigate clonality of leukemia and gene expression programs.

We conclude in Chapter 6 with a summary of the results and a discussion on the future perspectives of modeling of human leukemia in vitro and in vivo.

(34)

CHAPTER 1 33 Reference list

1. Bryder, D., Rossi, D. J. & Weissman, I. L. Hematopoietic Stem Cells. Am. J.

Pathol. 169, 338–346 (2006).

2. Gordon, M. Y., Lewis, J. L. & Marley, S. B. Of mice and men ... and elephants.

Blood 100, 4679–4679 (2002).

3. Adolfsson, J. et al. Upregulation of Flt3 expression within the bone marrow Lin(-)Sca1(+)c-kit(+) stem cell compartment is accompanied by loss of self- renewal capacity. Immunity 15, 659–69 (2001).

4. Christensen, J. L. & Weissman, I. L. Flk-2 is a marker in hematopoietic stem cell differentiation: A simple method to isolate long-term stem cells. Proc. Natl.

Acad. Sci. 98, 14541–14546 (2001).

5. Osawa, M., Hanada, K., Hamada, H. & Nakauchi, H. Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell. Science 273, 242–5 (1996).

6. Cheshier, S. H., Morrison, S. J., Liao, X. & Weissman, I. L. In vivo proliferation and cell cycle kinetics of long-term self-renewing hematopoietic stem cells.

Proc. Natl. Acad. Sci. U. S. A. 96, 3120–5 (1999).

7. Passegué, E., Wagers, A. J., Giuriato, S., Anderson, W. C. & Weissman, I. L.

Global analysis of proliferation and cell cycle gene expression in the regulation of hematopoietic stem and progenitor cell fates. J. Exp. Med. 202, 1599–611 (2005).

8. Wilson, A. et al. Hematopoietic Stem Cells Reversibly Switch from Dormancy to Self-Renewal during Homeostasis and Repair. Cell 135, 1118–1129 (2008).

9. Sottocornola, R. & Lo Celso, C. Dormancy in the stem cell niche. Stem Cell Res. Ther. 3, 10 (2012).

10. Sun, J. et al. Clonal dynamics of native haematopoiesis. Nature 514, 322–7 (2014).

11. Busch, K. et al. Fundamental properties of unperturbed haematopoiesis from stem cells in vivo. Nature 518, 542–546 (2015).

12. Sawai, C. M. et al. Hematopoietic Stem Cells Are the Major Source of

Multilineage Hematopoiesis in Adult Animals. Immunity 45, 597–609 (2016).

13. Adolfsson, J. et al. Identification of Flt3+ Lympho-Myeloid Stem Cells Lacking Erythro-Megakaryocytic Potential. Cell 121, 295–306 (2005).

(35)

14. Arinobu, Y. et al. Reciprocal Activation of GATA-1 and PU.1 Marks Initial Specification of Hematopoietic Stem Cells into Myeloerythroid and

Myelolymphoid Lineages. Cell Stem Cell 1, 416–427 (2007).

15. Lai, A. Y. & Kondo, M. Asymmetrical lymphoid and myeloid lineage

commitment in multipotent hematopoietic progenitors. J. Exp. Med. 203, 1867–

73 (2006).

16. Abdel-Wahab, O. et al. ASXL1 Mutations Promote Myeloid Transformation through Loss of PRC2-Mediated Gene Repression. Cancer Cell 22, 180–193 (2012).

17. Yoshida, T., Yao-Ming Ng, S., Zuniga-Pflucker, J. C. & Georgopoulos, K. Early hematopoietic lineage restrictions directed by Ikaros. Nat. Immunol. 7, 382–391 (2006).

18. Scadden, D. T. The stem cell niche in health and leukemic disease. Best Pract.

Res. Clin. Haematol. 20, 19–27 (2007).

19. Acar, M. et al. Deep imaging of bone marrow shows non-dividing stem cells are mainly perisinusoidal. Nature 526, 126–30 (2015).

20. Chen, J. Y. et al. Hoxb5 marks long-term haematopoietic stem cells and reveals a homogenous perivascular niche. Nature 530, 223–7 (2016).

21. Bruns, I. et al. Megakaryocytes regulate hematopoietic stem cell quiescence through CXCL4 secretion. Nat. Med. 20, 1315–20 (2014).

22. Hooper, A. T. et al. Engraftment and reconstitution of hematopoiesis is

dependent on VEGFR2-mediated regeneration of sinusoidal endothelial cells.

Cell Stem Cell 4, 263–74 (2009).

23. Kunisaki, Y. et al. Arteriolar niches maintain haematopoietic stem cell quiescence. Nature 502, 637–43 (2013).

24. Katayama, Y. et al. Signals from the Sympathetic Nervous System Regulate Hematopoietic Stem Cell Egress from Bone Marrow. Cell 124, 407–421 (2006).

25. Yamazaki, S. et al. Nonmyelinating Schwann cells maintain hematopoietic stem cell hibernation in the bone marrow niche. Cell 147, 1146–58 (2011).

26. Omatsu, Y. et al. The essential functions of adipo-osteogenic progenitors as the hematopoietic stem and progenitor cell niche. Immunity 33, 387–99 (2010).

27. Sugiyama, T., Kohara, H., Noda, M. & Nagasawa, T. Maintenance of the Hematopoietic Stem Cell Pool by CXCL12-CXCR4 Chemokine Signaling in

(36)

CHAPTER 1 35 Bone Marrow Stromal Cell Niches. Immunity 25, 977–988 (2006).

28. Ding, L., Saunders, T. L., Enikolopov, G. & Morrison, S. J. Endothelial and perivascular cells maintain haematopoietic stem cells. Nature 481, 457–462 (2012).

29. Sacchetti, B. et al. Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell 131, 324–36 (2007).

30. Calvi, L. M. & Link, D. C. The hematopoietic stem cell niche in homeostasis and disease. Blood 126, 2443–2451 (2015).

31. Ding, L. & Morrison, S. J. Haematopoietic stem cells and early lymphoid progenitors occupy distinct bone marrow niches. Nature 495, 231–235 (2013).

32. Boulais, P. E. & Frenette, P. S. Making sense of hematopoietic stem cell niches. Blood 125, 2621–9 (2015).

33. Grimwade, D., Ivey, A. & Huntly, B. J. P. Molecular landscape of acute myeloid leukemia in younger adults and its clinical relevance. Blood 127, 29–41 (2016).

34. Valent, P. et al. Cancer stem cell definitions and terminology: the devil is in the details. Nat. Rev. Cancer 12, 767–75 (2012).

35. Lapidot, T. et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 367, 645–648 (1994).

36. Taussig, D. C. et al. Anti-CD38 antibody-mediated clearance of human repopulating cells masks the heterogeneity of leukemia-initiating cells. Blood 112, 568–75 (2008).

37. Taussig, D. C. et al. Leukemia-initiating cells from some acute myeloid

leukemia patients with mutated nucleophosmin reside in the CD34(-) fraction.

Blood 115, 1976–84 (2010).

38. Eppert, K. et al. Stem cell gene expression programs influence clinical outcome in human leukemia. Nat. Med. 17, 1086–93 (2011).

39. Cozzio, A. et al. Similar MLL-associated leukemias arising from self-renewing stem cells and short-lived myeloid progenitors. Genes Dev. 17, 3029–35 (2003).

40. Krivtsov, A. V. et al. Transformation from committed progenitor to leukaemia stem cell initiated by MLL–AF9. Nature 442, 818–822 (2006).

41. Cox, C. V et al. Characterization of acute lymphoblastic leukemia progenitor cells. Blood 104, 2919–25 (2004).

(37)

42. Hotfilder, M. et al. Immature CD34+CD19- progenitor/stem cells in TEL/AML1- positive acute lymphoblastic leukemia are genetically and functionally normal.

Blood 100, 640–6 (2002).

43. le Viseur, C. et al. In Childhood Acute Lymphoblastic Leukemia, Blasts at Different Stages of Immunophenotypic Maturation Have Stem Cell Properties.

Cancer Cell 14, 47–58 (2008).

44. Bomken, S., Fišer, K., Heidenreich, O. & Vormoor, J. Understanding the cancer stem cell. Br. J. Cancer 103, 439–445 (2010).

45. Passegué, E., Jamieson, C. H. M., Ailles, L. E. & Weissman, I. L. Normal and leukemic hematopoiesis: are leukemias a stem cell disorder or a reacquisition of stem cell characteristics? Proc. Natl. Acad. Sci. U. S. A. 100 Suppl 1, 11842–9 (2003).

46. Schepers, K., Campbell, T. B. & Passegué, E. Normal and leukemic stem cell niches: insights and therapeutic opportunities. Cell Stem Cell 16, 254–67 (2015).

47. Dührsen, U. & Hossfeld, D. K. Stromal abnormalities in neoplastic bone marrow diseases. Ann. Hematol. 73, 53–70 (1996).

48. Ge, J., Hou, R., Liu, Q., Zhu, R. & Liu, K. Stromal-derived factor-1 deficiency in the bone marrow of acute myeloid leukemia. Int. J. Hematol. 93, 750–9 (2011).

49. Sipkins, D. A. et al. In vivo imaging of specialized bone marrow endothelial microdomains for tumour engraftment. Nature 435, 969–973 (2005).

50. Colmone, A. et al. Leukemic Cells Create Bone Marrow Niches That Disrupt the Behavior of Normal Hematopoietic Progenitor Cells. Science (80-. ). 322, 1861–1865 (2008).

51. Krause, D. S. et al. Differential regulation of myeloid leukemias by the bone marrow microenvironment. Nat. Med. 19, 1513–1517 (2013).

52. Schepers, K. et al. Myeloproliferative neoplasia remodels the endosteal bone marrow niche into a self-reinforcing leukemic niche. Cell Stem Cell 13, 285–99 (2013).

53. Arranz, L. et al. Neuropathy of haematopoietic stem cell niche is essential for myeloproliferative neoplasms. Nature 512, 78–81 (2014).

54. Hanoun, M. et al. Acute Myelogenous Leukemia-Induced Sympathetic Neuropathy Promotes Malignancy in an Altered Hematopoietic Stem Cell

(38)

CHAPTER 1 37 Niche. Cell Stem Cell 15, 365–375 (2014).

55. Raaijmakers, M. H. G. P. et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature 464, 852–7 (2010).

56. Raaijmakers, M. H. G. P. Myelodysplastic syndromes: revisiting the role of the bone marrow microenvironment in disease pathogenesis. Int. J. Hematol. 95, 17–25 (2012).

57. Ishikawa, F. et al. Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region. Nat. Biotechnol. 25, 1315–21 (2007).

58. Kaplan, R. N., Rafii, S. & Lyden, D. Preparing the "soil": the premetastatic niche. Cancer Res. 66, 11089–93 (2006).

59. Nestor, C. E. et al. Rapid reprogramming of epigenetic and transcriptional profiles in mammalian culture systems. Genome Biol. 16, 11 (2015).

60. Biondi, A., Cimino, G., Pieters, R. & Pui, C.-H. Biological and therapeutic aspects of infant leukemia. Blood 96, 24–33 (2000).

61. Felix, C. A. Secondary leukemias induced by topoisomerase-targeted drugs.

Biochim. Biophys. Acta 1400, 233–55 (1998).

62. Meyer, C. et al. New insights to the MLL recombinome of acute leukemias.

Leukemia 23, 1490–9 (2009).

63. Krivtsov, A. V & Armstrong, S. A. MLL translocations, histone modifications and leukaemia stem-cell development. Nat. Rev. Cancer 7, 823–33 (2007).

64. Krivtsov, A. V. & Armstrong, S. A. MLL translocations, histone modifications and leukaemia stem-cell development. Nat. Rev. Cancer 7, 823–833 (2007).

65. Okada, Y. et al. hDOT1L links histone methylation to leukemogenesis. Cell 121, 167–78 (2005).

66. Zhang, W., Xia, X., Reisenauer, M. R., Hemenway, C. S. & Kone, B. C. Dot1a- AF9 complex mediates histone H3 Lys-79 hypermethylation and repression of ENaCalpha in an aldosterone-sensitive manner. J. Biol. Chem. 281, 18059–68 (2006).

67. Bitoun, E., Oliver, P. L. & Davies, K. E. The mixed-lineage leukemia fusion partner AF4 stimulates RNA polymerase II transcriptional elongation and mediates coordinated chromatin remodeling. Hum. Mol. Genet. 16, 92–106 (2007).

(39)

68. So, C. W. et al. The interaction between EEN and Abi-1, two MLL fusion partners, and synaptojanin and dynamin: implications for leukaemogenesis.

Leukemia 14, 594–601 (2000).

69. García-Cuéllar, M. P. et al. ENL, the MLL fusion partner in t(11;19), binds to the c-Abl interactor protein 1 (ABI1) that is fused to MLL in t(10;11)+.

Oncogene 19, 1744–51 (2000).

70. Somervaille, T. C. P. & Cleary, M. L. Identification and characterization of leukemia stem cells in murine MLL-AF9 acute myeloid leukemia. Cancer Cell 10, 257–268 (2006).

71. Chen, W. et al. Malignant transformation initiated by Mll-AF9: gene dosage and critical target cells. Cancer Cell 13, 432–40 (2008).

72. Horton, S. J. et al. MLL-AF9-mediated immortalization of human hematopoietic cells along different lineages changes during ontogeny. Leukemia 27, 1116–26 (2013).

73. Laurenti, E. & Dick, J. E. Molecular and functional characterization of early human hematopoiesis. Ann. N. Y. Acad. Sci. 1266, 68–71 (2012).

74. Doulatov, S., Notta, F., Laurenti, E. & Dick, J. E. Hematopoiesis: a human perspective. Cell Stem Cell 10, 120–36 (2012).

75. Miller, P. H., Knapp, D. J. H. F. & Eaves, C. J. Heterogeneity in hematopoietic stem cell populations. Curr. Opin. Hematol. 20, 257–264 (2013).

76. Rongvaux, A. et al. Human hemato-lymphoid system mice: current use and future potential for medicine. Annu. Rev. Immunol. 31, 635–674 (2013).

77. Akkina, R. New generation humanized mice for virus research: comparative aspects and future prospects. Virology 435, 14–28 (2013).

78. Kim, J. B., O’Hare, M. J. & Stein, R. Models of breast cancer: is merging human and animal models the future? Breast Cancer Res. 6, 22–30 (2004).

79. Flanagan, S. P. ‘Nude’, a new hairless gene with pleiotropic effects in the mouse. Genet. Res. 8, 295–309 (1966).

80. Nilsson, K., Giovanella, B. C., Stehlin, J. S. & Klein, G. Tumorigenicity of

human hematopoietic cell lines in athymic nude mice. Int. J. cancer 19, 337–44 (1977).

81. Kamel-Reid, S. & Dick, J. E. Engraftment of immune-deficient mice with human hematopoietic stem cells. Science 242, 1706–9 (1988).

(40)

CHAPTER 1 39 82. Dick, J. E., Kamel-Reid, S., Murdoch, B. & Doedens, M. Gene transfer into

normal human hematopoietic cells using in vitro and in vivo assays. Blood 78, 624–34 (1991).

83. Mulé, J. J., Jicha, D. L. & Rosenberg, S. A. The use of congenitally

immunodeficient mice to study human tumor metastases and immunotherapy.

J. Immunother. (1991). 12, 196–8 (1992).

84. Mosier, d. E., gulizia, r. J., torbett, b. E., baird, s. M. & wilson, d. B. Break for scids. Nature 353, 509–509 (1991).

85. Pollock, P. L., Germolec, D. R., Comment, C. E., Rosenthal, G. J. & Luster, M.

I. Development of human lymphocyte-engrafted SCID mice as a model for immunotoxicity assessment. Fundam. Appl. Toxicol. 22, 130–8 (1994).

86. Bosma, G. C., Custer, R. P. & Bosma, M. J. A severe combined

immunodeficiency mutation in the mouse. Nature 301, 527–530 (1983).

87. De Lord, C. et al. Growth of primary human acute leukemia in severe combined immunodeficient mice. Exp. Hematol. 19, 991–3 (1991).

88. Kawata, A. et al. Establishment of new SCID and nude mouse models of human B leukemia/lymphoma and effective therapy of the tumors with

immunotoxin and monoclonal antibody: marked difference between the SCID and nude mouse models in the antitumor efficacy of monoclonal antibody.

Cancer Res. 54, 2688–94 (1994).

89. Paine-Murrieta, G. D. et al. Human tumor models in the severe combined immune deficient ( scid ) mouse. Cancer Chemother. Pharmacol. 40, 209–214 (1997).

90. Cesano, A. et al. The severe combined immunodeficient (SCID) mouse as a model for human myeloid leukemias. Oncogene 7, 827–36 (1992).

91. Ratajczak, M. Z. et al. In vivo treatment of human leukemia in a scid mouse model with c-myb antisense oligodeoxynucleotides. Proc. Natl. Acad. Sci. U. S.

A. 89, 11823–7 (1992).

92. Namikawa, R., Ueda, R. & Kyoizumi, S. Growth of human myeloid leukemias in the human marrow environment of SCID-hu mice. Blood 82, 2526–36 (1993).

93. Cesano, A. et al. Reversal of acute myelogenous leukemia in humanized SCID mice using a novel adoptive transfer approach. J. Clin. Invest. 94, 1076–84 (1994).

(41)

94. Chelstrom, L. M. et al. Childhood acute myeloid leukemia in mice with severe combined immunodeficiency. Blood 84, 20–6 (1994).

95. Pirruccello, S. J., Jackson, J. D. & Sharp, J. G. The leukemic myeloid cell line OMA-AML-1: an in vitro model of hematopoietic cell differentiation. Leuk.

Lymphoma 13, 169–78 (1994).

96. Yan, Y. et al. Growth pattern and clinical correlation of subcutaneously inoculated human primary acute leukemias in severe combined

immunodeficiency mice. Blood 88, 3137–46 (1996).

97. Goan, S. R. et al. The severe combined immunodeficient-human peripheral blood stem cell (SCID-huPBSC) mouse: a xenotransplant model for huPBSC- initiated hematopoiesis. Blood 86, 89–100 (1995).

98. Cashman, J. D. et al. Kinetic evidence of the regeneration of multilineage hematopoiesis from primitive cells in normal human bone marrow transplanted into immunodeficient mice. Blood 89, 4307–16 (1997).

99. Lapidot, T. et al. Cytokine stimulation of multilineage hematopoiesis from immature human cells engrafted in SCID mice. Science 255, 1137–41 (1992).

100. Bosma, G. C. et al. Evidence of functional lymphocytes in some (leaky) scid mice. J. Exp. Med. 167, 1016–33 (1988).

101. Carroll, A. M., Hardy, R. R. & Bosma, M. J. Occurrence of mature B (IgM+, B220+) and T (CD3+) lymphocytes in scid mice. J. Immunol. 143, 1087–93 (1989).

102. Shultz, L. D. et al. Multiple defects in innate and adaptive immunologic function in NOD/LtSz-scid mice. J. Immunol. 154, 180–91 (1995).

103. Dick, J. E. Human stem cell assays in immune-deficient mice.$: Current Opinion in Hematology. Curr. Opin. Hematol. (1996).

104. Bonnet, D. & Dick, J. E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat. Med. 3, 730–

7 (1997).

105. Dick, J. Normal and leukemic human stem cells assayed in SCID mice. Semin.

Immunol. 8, 197–206 (1996).

106. Shultz, L. D. et al. Human lymphoid and myeloid cell development in NOD/LtSz-scid IL2R gamma null mice engrafted with mobilized human hemopoietic stem cells. J. Immunol. 174, 6477–89 (2005).

(42)

CHAPTER 1 41 107. Ito, M. et al. NOD/SCID/gamma(c)(null) mouse: an excellent recipient mouse

model for engraftment of human cells. Blood 100, 3175–82 (2002).

108. Kato, C. et al. Spontaneous thymic lymphomas in the non-obese diabetic/Shi- scid, IL-2R gamma (null) mouse. Lab. Anim. 43, 402–4 (2009).

109. Katano, I., Ito, R., Eto, T., Aiso, S. & Ito, M. Immunodeficient NOD-scid IL- 2Rγ(null) mice do not display T and B cell leakiness. Exp. Anim. 60, 181–6 (2011).

110. Woiterski, J. et al. Engraftment of low numbers of pediatric acute lymphoid and myeloid leukemias into NOD/SCID/IL2Rcγnull mice reflects individual

leukemogenecity and highly correlates with clinical outcome. Int. J. cancer 133, 1547–56 (2013).

111. Agliano, A. et al. Human acute leukemia cells injected in NOD/LtSz-scid/IL- 2Rgamma null mice generate a faster and more efficient disease compared to other NOD/scid-related strains. Int. J. cancer 123, 2222–7 (2008).

112. Brehm, M. A. et al. Parameters for establishing humanized mouse models to study human immunity: analysis of human hematopoietic stem cell engraftment in three immunodeficient strains of mice bearing the IL2rgamma(null) mutation.

Clin. Immunol. 135, 84–98 (2010).

113. Pearce, D. J. et al. AML engraftment in the NOD/SCID assay reflects the outcome of AML: implications for our understanding of the heterogeneity of AML. Blood 107, 1166–1173 (2005).

114. Wei, J. et al. Microenvironment Determines Lineage Fate in a Human Model of MLL-AF9 Leukemia. Cancer Cell 13, 483–495 (2008).

115. Nicolini, F. E., Cashman, J. D., Hogge, D. E., Humphries, R. K. & Eaves, C. J.

NOD/SCID mice engineered to express human IL-3, GM-CSF and Steel factor constitutively mobilize engrafted human progenitors and compromise human stem cell regeneration. Leukemia 18, 341–347 (2004).

116. Takagi, S. et al. Membrane-bound human SCF/KL promotes in vivo human hematopoietic engraftment and myeloid differentiation. Blood 119, 2768–2777 (2012).

117. Ito, R. et al. Establishment of a Human Allergy Model Using Human IL-3/GM- CSF-Transgenic NOG Mice. J. Immunol. 191, 2890–2899 (2013).

118. Brehm, M. A. et al. Engraftment of human HSCs in nonirradiated newborn

Referenties

GERELATEERDE DOCUMENTEN

Upon transplantation of MLL-AF9-transduced CB CD34 + cells, acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) developed in engineered scaffolds, in which

Next, a Tet-regulated miR-E PCGF1 human xenograft MLL-AF9 leukemia mouse model was established, which revealed that early knockdown of PCGF1 at the onset of

Modeling of MLL-AF9-rearranged pediatric leukemia: Identification of mechanisms and potential targets!. University

Parameters for establishing humanized mouse models to study human immunity: analysis of human hematopoietic stem cell engraftment in three immunodeficient strains of mice bearing

A) Schematic representation of the different models injected with CB MLL-AF9 cells: IV injected NSG mice, ISC injected huBM-sc and ISC injected cytoBM-sc. Kaplan– Meier

Moreover, and once again as observed for the I- BET151 treated cells, none of the HOXA cluster of genes was affected by the ligand deprivation as confirmed also by qPCR for

In contrast, target genes specific for MLL-rearranged leukemias were rather comparable in all 4 populations, further confirming the presence of MLL-AF4- drivene gene

Efficient engraftment in the huBM-sc xenograft model was also observed when primary CD34 + CML patient cells were injected and, in contrast to the murine niche, leukemic