• No results found

Targeting cancer stem cells: Modulating apoptosis and stemness - Chapter 2: Cancer stem cells; important players in tumor therapy resistance

N/A
N/A
Protected

Academic year: 2021

Share "Targeting cancer stem cells: Modulating apoptosis and stemness - Chapter 2: Cancer stem cells; important players in tumor therapy resistance"

Copied!
23
0
0

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

Hele tekst

(1)

Targeting cancer stem cells: Modulating apoptosis and stemness

Çolak, S.

Publication date

2016

Document Version

Final published version

Link to publication

Citation for published version (APA):

Çolak, S. (2016). Targeting cancer stem cells: Modulating apoptosis and stemness.

General rights

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), other than for strictly personal, individual use, unless the work is under an open

content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please

let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material

inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter

to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You

will be contacted as soon as possible.

(2)

S. Colak & J.P. Medema Published in FEBS Journal, 2014

Cancer stem cells; important players

in tumor therapy resistance

(3)

Abstract

Resistance to tumor therapy is an unsolved problem in cancer treatment. A plethora

of studies have attempted to explain this phenomenon and many mechanisms of

resistance have been suggested over the last decades. The concept of cancer stem cells

(CSCs), which describes tumors as hierarchically organized, has added a new level of

complexity to therapy failure. CSCs are the root of cancers and resist chemo- and

irra-diation therapy explaining cancer recurrence even many years after the therapy ended.

This review discusses briefly CSCs in cancers, gives an overview of the role of CSCs

in therapy resistance, and discusses the potential means of targeting these therapy

resistant tumor cells.

(4)

2

Introduction

Well over 40 years ago president Nixon signed the national cancer act and officially

started the war on cancer.

1

Despite this act and resulting significant improvements

in therapy, the war has not ended and cancer is still one of the leading causes of death

worldwide. To illustrate, in 2012 14.1 million adults in the world were diagnosed with

cancer and in the same year an estimated 8.2 million people died from the disease

2

The

main problem we face in cancer treatment is the presence or development of resistance

to therapy for which a multitude of distinct reasons have been defined. For instance,

acquisition of mutations in key signalling molecules, enhanced anti-apoptotic protein

expression, presence of quiescent and/or resistant tumor cells or high expression of drug

efflux pumps are all potential means that impair therapy efficacy.

3, 4

In the last decade

a lot of attention has gone to the role of a specific subset of cancer cells called cancer

stem cells (CSCs). In analogy with their normal counterparts, the stem cells, these cells

display a high level of therapy resistance and can effectively repopulate the tumor.

CSCs are the tumorigenic core of tumors

CSCs are defined based on their tumor forming capacity in xenograft studies.

5

These

cells normally represent a minority of the tumor cells and can be identified by a long

list of markers, although most of these are not restricted solely to the CSC

popula-tion.

6

CSCs can be selected in vitro using spheroid growth in suspension and defined

media compositions. Upon injection in mice CSCs, but not their more differentiated

counterparts, can very efficiently form tumors that resemble the original tumor from

which they were derived, including all differentiated cells. Moreover, re-isolation of

the CSCs from xenografts allows for serial transplantation to secondary and tertiary

mice, which is the gold-standard assay to prove that tumor cells are indeed CSCs.

5

CSCs were first defined in acute myeloid leukemia (AML) in 1994.

7

CD34

+

/CD38

-

expression has long been used to mark progenitor and pluripotent stem cells in the bone

marrow. Intriguingly, a similar subpopulation was detected in AML and

xenotrans-plantation of specifically this CD34

+

/CD38

-

leukemia cells resulted in leukemia in mice

that reproduced many features of human AML.

7

A decade later CSCs were detected in

solid tumors. Breast, glioblastoma, prostate and colorectal tumors are only some of the

tumor types where CSCs were identified.

6, 8-12

The variety of tumors in which CSCs

were identified suggests that it is a common feature in most cancers, although some

observations indicate that it may not occur in all tumor types or alternatively at all

stages of disease.

13-16

(5)

As mentioned above, CSCs are highly tumorigenic and therefore are also referred to

as tumor-initiating cells. The name “CSC” does not refer to the fact that CSCs can be

derived from normal stem cells, but rather points to the idea that CSCs display

prop-erties normally attributed to stem cells. Firstly, stem cells have the capacity to

self-renew, i.e. to form a new stem cell upon division, and secondly stem cells can

differ-entiate into the more specialized cell types that make up a tissue.

17

Self-renewal and

differentiation of stem cells is regulated by morphogenic pathways and interestingly

these signaling pathways are also highly active in many CSCs, suggesting that equal

regulatory principles exist in CSCs. One of the morphogenic pathways that is active in

stem cells is the Wnt signaling.

18

This pathway determines self-renewal and cell fate

of hematopoietic stem cell (HSCs).

19

High activity of this pathway is also observed in

stem cells of other tissues like breast and colon.

18, 20, 21

Next to Wnt signaling, Notch

signaling is shown to be essential for stem cell maintenance. Notch signaling is highly

active in HSC when compared to more differentiated cells and inhibition of Notch

signaling promotes differentiation of HSC.

22

Likewise, Hedgehog (HH) signaling

regu-lates proliferation and self-renewal of stem cells and activation of HH signaling is able

to expand HSC and brain stem cells in vitro and in vivo.

23, 24

In contrast to Wnt, Notch,

and HH signaling BMP signaling is inhibiting stem cell expansion. Activation of BMP

signalling results in suppression of Wnt signaling and this controls stem cells numbers.

25-27

Combined these morphogenic pathways regulate stem cell fate and

differentia-tion cues. Intriguingly, this regulatory network appears to extend to CSCs. In a high

throughput screening in breast CSCs salinomycin was identified as a compound that

eliminates CSCs.

28

This antibiotic was shown to inhibit Wnt signaling and as a result

is capable to differentiate breast CSCs.

28

High Wnt pathway activity is also shown

to be important for cell fate of CSCs from many tumors like CLL, breast, CRC,

squa-mous cell carcinoma, and lung cancer.

28-32

In these tumors inhibition of Wnt pathway

activity, with e.g. salinomycin is detrimental for CSCs.

28-32

Beside Wnt signaling,

Notch signaling can also regulate CSC self-renewal. Inhibition of Notch signals can

be achieved by neutralizing antibodies against DLL4, or treatment with a g-secretase

inhibitor (DBZ). GBM, CRC and breast cancer stem cells need high Notch activity

and inhibition of Notch results in loss of CSCs.

33-36

Furthermore, Hedgehog signalling

is highly active in CSCs, which is shown to be required for self-renewal of CSCs in

various cancers like breast, lung, and CRC.

37-39

Morphogenic pathways and inhibitors

are depicted in figure 1.

In addition to the analogous usage of morphogenic pathways, stem cells and CSCs

also appear to share high activity of DNA repair pathways. For example, HSC can

(6)

2

Figure 1: Targeting morphogenic pathways in CSCs

Morphogenic pathways Notch (blue), Wnt (green) and Hedgehog (red) signaling pathways are highly active and important for self-renewal of stem cell and CSCs. Blue: Notch signaling is activated via direct cell-cell contact. A cell expressing Notch ligand (e.g. DLL4) contacts with another cell that expresses Notch receptor. When bound by a Notch ligand, the intracellular domain of the Notch receptor (IC-Notch) is cleaved by γ-secretase (γsec) and is targeted to the nucleus to activate transcription of downstream target gene that enhance self-renewal of CSCs. This pathway can be inhibited with a DLL4 antibody that neutralizes Notch ligand DLL4. Also γ-secretase inhibitors like DBZ are efficient in blocking Notch signaling. Green: Wnt ligands binds to the Frizzled-LRP receptor and inhibit a cytoplasmic destruction complex (APC-GSK3β-Axin) of β-catenin, which then enters the nucleus to activate transcription of Wnt target genes that are known to be important for CSCs maintenance. Wnt signaling can be inhibited with salinomycin. Red: The Hedgehog pathway is activated by Hedgehog ligands binding to the Patched receptor, which releases its inhibition of the Smoothened (Smo) transmembrane receptor. Smoothened can then in turn activates Gli transcription factors, the final effectors of the Hedgehog pathway. The natural occuring compound cyclopamine is used to inhibit Smo receptor and thereby block Hedgehog signaling.

(7)

repair UV induced single-strand breaks faster when compared with more

differenti-ated cells

40

and this was reported in CSCs as well.

41

It is important to mention that

more and more evidence indicates that CSCs are not a fixed cell population, but rather

represents a state of tumor cells that appears inducible. Giving the right signals from

the micro-environment or introduction of new mutations can result in

de-differentia-tion of more differentiated tumor cells into CSCs.

31, 42-44

Not only signaling pathways, but also cell surface molecules are similarly expressed on

stem cells and CSCs. The pentaspan membrane glycoprotein CD133, also known as

Prominin-1, is expressed in normal stem cells, e.g. hematopoietic, neural, and intestinal

stem cells,

45-47

but CD133 was also used to identify CSCs from different tumor types.

8, 9, 41, 48

Moreover, in the last decade the G-protein coupled receptor Lgr5 received a lot

of attention because high Lgr5 expression was reported to mark stem cells in various

organs. This target gene of the Wnt signaling pathway is exclusively expressed by stem

cells of various organs

49-52

and we and others have shown that Lgr5 also marks CSCs

in various tumors.

53, 54

Most of the currently used markers have no known role in CSC

biology. In contrast, Aldehyde dehydrogenase isoform 1 (ALDH1) oxidizes aldehydes

to carboxylic acids and thus for instance catalyzes the conversion of retinol (vitamin

A) to retinoid acid. Inhibition of ALDH1 reduces retinoic acid levels and thereby

promotes HSC and breast CSCs self-renewal.

55, 56

ALDH1 is highly expressed in many

stem cells and CSCs, and ALDH1 expression and its activity were used to isolate stem

cells and CSCs.

57-59

Taken together, it appears that stem cells and CSCs are wired in

the same way and share expression of several cell surface markers. Unfortunately, this

similarity extends to a more detrimental property, namely therapy resistance.

The extreme survivors; CSCs and their therapy resistance

The concept that CSCs selectively resist therapy stems from a multitude of

observa-tions in cell culture, animal models and cancer patients. In cell culture direct analysis

of apoptosis revealed that differentiated colon cancer cells are induced to die upon

chemotherapy treatment, while colon CSCs from the same cultures survive the toxic

insults.

60

This differential sensitivity was not due to proliferation differences between

CSCs and more differentiated cells as it was also observed when using treatments that

are not dependent on cycling cells.

60

Moreover, these surviving CSCs can

re-estab-lish the culture, confirming the idea that they are responsible for therapy failure.

60

Chemotherapy resistant CD133

+

CSCs were described in liver and lung cancer as

well,

61, 62

Similar observations were derived in pancreatic cancer where CSCs were

(8)

2

this tumor type in vitro cell death was more pronounced in the differentiated CD133

-cells as compared to the CD133

+

cells.

48

Finally, GBM CSCs and breast CSCs isolated

from patient specimens, displayed selective resistance to various chemotherapies.

63, 64

Next to the in vitro evidence, escape from therapy was also evident from xenograft

studies. Chemotherapy treatment of xenotransplanted CRCs resulted in an increase

in CD133

+

in the tumor.

65

This indicates that CD133

+

CSCs are more resistant to

oxaliplatin in vivo when compared to differentiated CD133

-

cells. In vivo resistance

of CRC CSCs is not restricted to oxaliplatin as mice bearing human CRC tumors

treated with irinotecan show an increase in cells that express ESA

+

CD44

+

CD166

+

,

distinct markers for CSCs.

66

Moreover, in vivo gemcitabine treatment of

xenotrans-planted pancreatic cancer induced the CD133

+

fraction, pointing to a gemcitabine

resistant CSC population.

48

Finally, also in xenotransplants of AML enhanced

CD34

+

/CD38

-

CSCs were observed in vivo upon cytosine arabinoside treatment.

67

Intriguingly, therapy resistance appears to be a general feature of these cells

and is not restricted to chemotherapy, but observed with radiotherapy as well.

68

Irradiated glioblastoma, either implanted subcutaneously or intracranially

showed an increase of CD133

+

cells when compared to non-irradiated tumors.

This distinction was suggested to be clinically relevant as the authors extended

these findings to ex vivo irradiation of surgically removed glioblastomas.

41

Also for irradiation examples encompass other tumor types. MMTV-Wnt1 mice bearing

breast tumors showed an increase in the CSC (

Thy1

+

CD24

+

Lin

-

)

fraction after

irradi-ation and in the same study CSCs from primary human head and neck cancers proved

radioresistant.

69

Combined these data indicate that cell line or primary tumor-derived

cells with CSC markers display decreased sensitivity to chemo and radiotherapy. It

is important to realize though that one potential caveat with this conclusion is the

fact that CSC markers are heavily debated, suggesting that the increases observed in

marker expression may not represent enhanced stemness(for a review see Medema

2013).

6

Moreover, xenotransplantation models may not adequately represent the

normal situation in patients

6

and select for distinct traits and/or markers.

Neverthe-less, a first direct hint that this CSC resistance concept could explain minimal residual

disease and therapy failure in patients came from a study of the group of Luis Parada

who used a genetically modified mouse model to study endogenously growing tumors

in which CSCs could be traced using a Nestin reporter construct. Nestin

+

tumor cells,

which represent a quiescent CSC population, could fully repopulate the tumor after

temozolomide chemotherapy, while selective deletion of these cells prevented tumor

outgrowth.

70

These data indicate that CSCs resist therapy and might be a

(9)

poten-tial cause of tumor relapse. In line with this observation, an increasing list of

obser-vations in patients support the crucial role of CSCs in tumor relapse after therapy.

In patients with GBM, CRC, or breast cancer, increased CSC fractions using marker

expression were measured after chemotherapy treatment.

71-74

A more direct evidence

for increases in true functional CSCs came from a study in breast cancer. In contrast to

other reports, here authors studied patient samples and performed functional assays.

Increase in mammosphere formation capacity was seen after chemotherapy

treat-ment,

72, 73

proving that stemness rather showed a relative increase than decrease upon

therapeutic intervention. The growing body of evidence that points to a role for CSCs

in resistance warrants a more detailed survey to increase our understanding of the

mechanisms that determine resistance in order to target these survivors of therapy.

Mechanisms behind therapy resistance

Normal stem cells contain multiple mechanisms to control cell death, which aids to

protect these crucial cells from cytotoxic insults. Elevated apoptosis resistance,

drug-efflux pumps, enhanced DNA repair efficiency, detoxification enzyme expression and

quiescence are all identified as pro-survival mechanisms. Intriguingly, all these

mecha-nisms appear to be hijacked by CSCs. For instance, mitochondrial apoptosis is

associ-ated with loss of mitochondrial membrane integrity, which is maintained by a strict

balance of anti-apoptotic BCL2 proteins (e.g. BCL2, BCLXL, and MCL1), pro-apoptotic

BCL2 family members (BAX and BAK) and BH3 proteins (e.g. BIM, BAD, and NOXA).

A cytotoxic insult-induced imbalance in the ratio of these molecules results in

permea-bilization of the mitochondrial outer membrane and subsequent activation of a caspase

cascade.

75

In stem cells, but also in CSCs, an elevated anti-apoptotic protein expression

increases the threshold for apoptosis induction and thereby directly protects the cells

against apoptosis. For instance, in breast and AML CSCs, BCL2 and BCLXL are highly

expressed.

76, 77

Similarly, in primary GBM cultures CD133

+

CSCs had elevated

expres-sion of BCL2 and BCLXL compared to their more differentiated CD133

-

progeny.

78

In

agreement with a role for apoptosis regulation in CSCs, direct proteomic analysis of

CRC CSCs and differentiated cells revealed “apoptosis” as one of the main molecular

pathways affected, involving differential expression of key anti-apoptotic proteins,

including BIRC6.

79

Combined this suggests that CSCs have an elevated anti-apoptotic

threshold. Recent data confirm this idea using so-called BH3 profiling, an assay to

directly measure the apoptosis priming state of cells.

80

This revealed that CRC CSCs

were less-primed as compared to differentiated cells, which at least in part explains

their resistance to conventional chemotherapy.

60

In agreement, sublethal doses of BH3

(10)

2

mimetics can change this threshold and strongly sensitize CSCs to chemotherapy.

Besides an elevated apoptotic threshold, CSCs display high expression of drug efflux

pumps, like ATP-binding cassette (ABC) transporter family proteins.

71, 81-83

These

proteins are important for efflux of chemotherapy across the plasma membrane.

4

Various

ABC transporter proteins are highly expressed in HSC and in AML CSCs (CD34

+

/

CD38

-

) compared to the non stem (CD34

+

/CD38

+

) cells [81]. Also in GBM and

mela-noma high expression of drug efflux pumps in CSCs are reported.

82

In the latter,

expression of ABC transporter ABCB5 in fact serves as a marker for CSCs.

83

Surpris-ingly, in CRC a different scenario is reported. Here not CSCs, but rather the

differenti-ated cells express high levels of the drug efflux pump ABCB1. The authors suggested

that differentiated cells protect CSCs from chemotherapy treatment by forming a

protective rim around the CSCs.

84

The above points to the fact that CSCs employ means to avoid the impact of therapy,

which we can potentially circumvent using combination therapy. However a

poten-tially more challenging problem is the recent observation that CSCs may exist that

display quiescent properties. Selectivity of chemotherapy for cancer cells relies on

the fact that chemotherapy mainly kills cells that are highly proliferative. As rapid

uncontrolled proliferation is a standard feature of many tumor cells, chemotherapy is

thought to target tumor cells selectively over non-proliferating normal cells, consistent

with the observed toxicity in organs with rapidly dividing cells, such as bone marrow,

digestive tract, and hair follicles. In contrast, slow proliferating or quiescent normal

cells are largely protected from chemotherapy treatment. Importantly, this

resist-ance also extends to quiescent tumor cells.

In ovarian cancer CD24

+

CSCs are less

proliferative and more resistant to chemotherapy when compared to CD24

-

cells.

85

Recent data point to the existence of CSCs that are quiescent. These can be

identi-fied using the dye PKH26, which dilutes out when cells proliferate and therefore only

low or non-proliferative cells will retain the label. In primary melanoma cultures label

retaining cells were detected with a very low doubling time of around 4 weeks in vitro.

Although these cells are slow dividing they have increased sphere forming capacity in

vitro

suggesting that these label retaining cells are enriched in CSCs.

86

Such quiescent

cells are also identified in pancreatic adenocarcinoma and shown to be enriched for

CSC markers like CD133, CD24

+

/CD44

+

and ALDH. In agreement with this notion,

these label retaining cells are more tumorigenic, indicating that cancer is not a disease

of homogeneously rapidly proliferating cells, but also contains quiescent cells that

can escape classical chemotherapy and subsequently induce regrowth of the tumor.

87

(11)

contain enhanced potential and/or time to repair the damage that is inflicted to them.

As many chemotherapeutic agents as well as radiotherapy work by inducing DNA

damage, cells that effectively repair DNA damage can potentially survive

chemo-therapy. Various reports have shown that CSCs, for instance from GBM, possess high

DNA repair activity, which makes them resistant to radiation and chemotherapy.

41

Similarly, in breast CSCs there is increased expression of DNA repair genes, indicating

that high DNA repair pathway activity may aid in making CSCs resistant to tumor

therapy. In conclusion, there are many ways for CSCs to resist tumor therapy. Figure

2 illustrates the reasons for therapy resistance in CSCs.

Killing CSCs, magic bullets or combination cocktails?

Although considered bad news, the efficient DNA repair of CSCs may also point to a

dependency for these mechanisms and as such offer a means to target these cells. For

example, CSCs in GBM have elevated activity of Chk1 and ATM and survive irradiation,

but inhibition of the cell cycle checkpoint kinases Chk1 and Chk2 is sufficient to sensitize

CSCs towards irradiation.

41

Recently, it has been reported that a combined Chk1 and

PDK1 inhibition is required to kill CSCs in GBM.

88

Similarly, non-small cell lung cancer

CSCs can be sensitized to chemotherapy by combining treatment with Chk1 and Chk2

inhibitors SB218078 or AZD7762.

89

Mechanistically, inhibition of Chk1 results in active

Cdc2-cyclin B complex that is followed by mitotic catastrophe.

90

Although effective, these

compounds are also relatively toxic and combination of the Chk inhibitor AZD7762 with

gemcitabine showed cardiac toxicity.

91

To overcome this toxicity an inhibitor of a

down-stream target of Chk1, Wee1, was developed. In the presence of DNA damage Wee1 arrests

cells in G2 phase and allows cells to repair DNA before entering into mitosis. Interestingly,

Wee1 is reported to be overexpressed in GBM CSCs. In the same report, the authors show

that inhibition of Wee1 with PD0166285 sensitizes GBM CSCs towards irradiation.

92

Besides targeting the core of the repair machinery, a lot of effort is put into targeting the

execution machinery in cancer cells. Previously, we have used an inducible caspase-9 to

target colon CSCs. Upon activation of caspase-9, colon CSCs were killed efficiently in

vitro

and in vivo suggesting that activation of caspases are sufficient to efficiently kill

CSCs.

93

As described, anti-apoptotic proteins are highly expressed in various cancers and

especially in CSCs. Targeting these anti-apoptotic proteins using small molecules that

have been developed therefore forms an attractive mechanism. For instance, ABT-737,

a small molecule inhibitor that targets BCL2, BCLXL, and BCLW tips the apoptotic

balance to a more pro-apoptotic state and reverts the resistance of colon CSCs.

60, 94

(12)

2

Figure 2: Mechanisms of therapy resistance in CSCs

a) Four mechanisms that are used by CSCs to resist chemotherapy. Efficient DNA repair (orange), quies-cence (red), increase ABC transporter expression (green), and decreased mitochondrial priming (blue). b) Potential means of targeting therapy resistant CSCs.

(13)

the same specificity, seems to exert high toxicity for platelets, which depend on BCLXL

for survival.

95

Although more selective inhibitors, such as ABT-199 targeting only BCL2,

have been developed to overcome this problem, our recent data indicates that also colon

CSCs are dependent on BCLXL for survival. In agreement inhibition of BCLXL with

ABT-737 or a BCLXL specific inhibitor (WEHI-539) is sufficient to kill colon CSCs.

Simi-larly, lung CSCs are shown to be dependent on BCLXL and also in these cells

inhibi-tion eliminates lung CSCs in vitro and in vivo.

96

Although this still raises the problem of

toxicity, it is possible to use sublethal amounts of BCLXL inhibition, which is sufficient

to strongly sensitize CSCs towards chemotherapy.

60

It is currently not completely clear

why colon CSCs acquire this dependency on BCLXL. One possible explanation is the

observation by Todaro and colleagues showing an autocrine loop of IL4/IL4R in colon

CSCs, which appears to maintain BCLXL levels and protect CSCs from chemotherapy.

65

One thing that allows for the identification of CSCs is the presence of several cell

surface markers. Various groups and companies therefore developed immunotoxins

that directly target such CSC markers. For instance, antibodies against for instance the

stem cell marker CD133 conjugated to paclitaxel or cytolethal distending toxin (Cdt)

target CD133 expressing cells and show in vitro and in vivo elimination of tumors.

97, 98

Similarly, targeting of CD133

+

cells can be achieved by generation of CD133 specific

Measlus viruses. These oncolytic viruses infect CD133 expressing cells and destroy

them by lysis.

99

Moreover, selective killing of CD133

+

GBM cells was shown with

CD133 antibodies coupled to singe walled carbon nanotubes (SWNTs). These

anti-CD133-SWNTs induce thermal destruction of cancer cells when it is combined with

nearIR laser light.

100

However, CD133 expression is not specific for CSCs but also

expressed on normal stem cells, which should be protected from such therapies at

all times. To minimize toxicity and deliver drug to cancer selectivity photochemical

internalization (PCI) was developed. This technique makes it possible to release

drug in the tumor area specifically.

101

Next to CD133, there is an increasing effort

to target other cell surface molecules including the stem cell marker Lgr5.

Neverthe-less, as is true for CD133, toxicity with such an approach can be expected.

Surpris-ingly, antibodies without toxins targeting other cell surface molecules are shown to

be efficient in killing CSCs as well. Antibodies against CD47 give promising effects

in various cancers. CD47 is a receptor that is involved in inhibition of so called

“eat-me” signals and is highly expressed on CSCs compared to more differentiated

cells. Blocking of this CD47 receptor with an antibody enables the phagocytosis of

AML CSCs and thereby blocks tumor growth.

102

In addition, CD47 inhibition also

(14)

2

blocks tumor growth in solid cancers, like breast cancer, CRC, ovarian cancer and

GBM, which is also suggested to depend on facilitating phagocytosis of CSCs.

103

Next to phagocytosis induction, several antibodies have shown to delete essential

signals from CSCs. For instance, direct targeting of breast CSCs can be achieved by

using an antibody against CXCR1. The IL8 receptor CXCR1 is expressed almost

exclu-sively on CSCs and Repertaxin, an inhibitor of CXCR1/2, or anti-CXCR1 treatment

induces cell death in CXCR1

+

breast CSCs, which appears to be mediated by AKT

signaling inhibition.

64

Intriguingly, PI3K/AKT signaling addiction in colon CSCs was

also reported in colon CSCs, where a CD44v6-positive subset was identified that is

exclusively metastatic. These cells express high levels of PI3K, which, if inhibited, alter

the viability of the cells and impede the capacity to migrate,

104

suggesting that PI3K

signaling is crucial for CSCs.

As described, CSCs require signaling through morphogenic pathways for their

main-tenance, suggesting that these may be attractive targets for therapy as well. In

agree-ment, a screen for CSC sensitizing compounds identified salinomycin, which inhibits

Wnt signaling and eliminates breast and CLL CSCs.

28, 32

As CSCs in CML, AML and

skin tumors are dependent on the Wnt pathway, inhibition can be clinically relevant.

29, 105-107

Furthermore, inhibition of Notch signalling pathway using a neutralizing

anti-body against DLL4 results in less tumor engraftment in secondary tumors suggesting

in vivo

differentiation of CSCs. Importantly, DLL4 antibody was also able to

sensi-tize the tumor to irinotecan in vivo.

33

Inhibition of Notch signaling was also

suffi-cient to deplete GBM CSCs and sensitize ovarian CSCs to chemotherapy.

35, 108

Lastly,

HH signaling can be inhibited by using cyclopamine and this Smoothened

antago-nist sensitizes AML CSCs to Ara-c treatment.

109

Similarly, in GBM and in

pancre-atic cancer decreases in CSCs are observed after treatment with Smoothened

inhibi-tors cyclopamine or CUR199691.

110, 111

These data point to a crucial role for HH

signalling in cancer stemness and this is confirmed by knockdown of Smoothened,

which results in loss of CML CSCs.

112

Antibodies can also be used to target CSC

niche. Blood vessels maintain GBM CSCs in a stem like state. Targeting

microenvi-ronment with Bevacizumab, an antibody against VEGF is able to differentiate GBM

CSCs.

113

In 2006 Jin et al showed that using an antibody against CD44 decreased

homing of AML cells and thereby promoting the AML CSCs to differentiate to a

more mature cancer cell progeny. This antibody inhibited AML growth in mice.

114

(15)

reported to happen with bone morphogenetic protein 4 (BMP4) as well. In CRC BMP4

expression is exclusively expressed by differentiated cancer cells and shown to induce

differentiation of CSCs and sensitization to oxaliplatin in vivo.

115

In addition, BMP4

also forces GBM CSCs to differentiate and thereby inhibits their tumorigenicity.

116

Not only inhibition of morphogenic pathways, but activation of signaling pathways

can change CSCs cell fate as well. Activation of the unfolded protein response (UPR)

induces differentiation of stem cells in the mouse intestine.

117

In line with this,

salu-brinal induces UPR in colon CSCs and forces them to differentiate. In addition to

inducing differentiation, UPR sensitizes cells to chemotherapy in vitro and in vivo

(M.C.B. Wielenga and S. Colak unpublished observations). Although targeting CSCs

by forcing them to differentiate or by the induction of apoptosis seem to be a

attrac-tive therapeutic options, the suggested flexibility of the system is a clear caveat. Even

when CSCs are eliminated within a tumor, differentiated cells can de-differentiated

and take the place of the CSCs that were deleted.

31, 43

Targeting the cues that induce

de-differentiation or simply attacking both CSCs and more-differentiated cells needs

to be achieved to eradicate a tumor.

6

Altogether, direct CSCs targeting or CSC

differ-entiation therapy are promising means to improve tumor therapy. Further studies are

needed to investigate the most promising combination treatments that does not give

severe toxicities.

Summary

Identification of CSCs in many tumors allowed for a better understanding as to why

even many years after therapy tumors can relapse. There is increasing evidence that

targeting these CSCs is important to improve therapies. Here we reviewed mechanisms

that make CSCs resistant to therapy A better understanding of these mechanisms

and the way CSCs retain their tumorigenic stem cell capacities is crucial. The exiting

new insight will undoubtedly provide new therapeutic tools in the years to come.

Acknowledgements

We thank the members of the laboratory for useful discussion. JPM is

sponsored by grants from the Netherlands Organization for Scientific

Research (NWO; Gravitation-Cancer Genomics Center The Netherlands

Zwaartekracht), from the Dutch Cancer Society (UVA2009-4416 and

UVA2012-5735), from MLDS (FP13-07) and Alpe dHuzes/KWF (CONNECTION).

(16)

2

References

1. Weinhouse, S. (1972) National Cancer Act of 1971--an editorial, Cancer research. 32, i-ii.

2. Ferlay J, S. I., Ervik M, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, & Forman D, B., F (2013) GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 11 [Internet]. Lyon, France: International Agency for Research on Cancer. Available from http://globocan. iarc.fr. Accessed June 2014

3. Holohan, C., Van Schaeybroeck, S., Longley, D. B. & Johnston, P. G. (2013) Cancer drug resistance: an evolving paradigm, Nature reviews Cancer. 13, 714-26.

4. Gottesman, M. M., Fojo, T. & Bates, S. E. (2002) Multidrug resistance in cancer: role of ATP-dependent transporters, Nature reviews Cancer. 2, 48-58.

5. Clarke, M. F., Dick, J. E., Dirks, P. B., Eaves, C. J., Jamieson, C. H., Jones, D. L., Visvader, J., Weissman, I. L. & Wahl, G. M. (2006) Cancer stem cells--perspectives on current status and future directions: AACR Workshop on cancer stem cells, Cancer research. 66, 9339-44.

6. Medema, J. P. (2013) Cancer stem cells: the challenges ahead, Nature cell biology. 15, 338-44.

7. Lapidot, T., Sirard, C., Vormoor, J., Murdoch, B., Hoang, T., Caceres-Cortes, J., Minden, M., Paterson, B., Caligiuri, M. A. & Dick, J. E. (1994) A cell initiating human acute myeloid leukaemia after transplanta-tion into SCID mice, Nature. 367, 645-8.

8. Ricci-Vitiani, L., Lombardi, D. G., Pilozzi, E., Biffoni, M., Todaro, M., Peschle, C. & De Maria, R. (2007) Identification and expansion of human colon-cancer-initiating cells, Nature. 445, 111-5.

9. O’Brien, C. A., Pollett, A., Gallinger, S. & Dick, J. E. (2007) A human colon cancer cell capable of initi-ating tumour growth in immunodeficient mice, Nature. 445, 106-10.

10. Singh, S. K., Hawkins, C., Clarke, I. D., Squire, J. A., Bayani, J., Hide, T., Henkelman, R. M., Cusimano, M. D. & Dirks, P. B. (2004) Identification of human brain tumour initiating cells, Nature. 432, 396-401. 11. Al-Hajj, M., Wicha, M. S., Benito-Hernandez, A., Morrison, S. J. & Clarke, M. F. (2003) Prospective iden-tification of tumorigenic breast cancer cells, Proceedings of the National Academy of Sciences of the United States of America. 100, 3983-8.

12. Collins, A. T., Berry, P. A., Hyde, C., Stower, M. J. & Maitland, N. J. (2005) Prospective identification of tumorigenic prostate cancer stem cells, Cancer research. 65, 10946-51.

13. Kelly, P. N., Dakic, A., Adams, J. M., Nutt, S. L. & Strasser, A. (2007) Tumor growth need not be driven by rare cancer stem cells, Science. 317, 337.

14. Quintana, E., Shackleton, M., Sabel, M. S., Fullen, D. R., Johnson, T. M. & Morrison, S. J. (2008) Efficient tumour formation by single human melanoma cells, Nature. 456, 593-8.

15. Meacham, C. E. & Morrison, S. J. (2013) Tumour heterogeneity and cancer cell plasticity, Nature. 501, 328-37.

16. Magee, J. A., Piskounova, E. & Morrison, S. J. (2012) Cancer stem cells: impact, heterogeneity, and uncer-tainty, Cancer cell. 21, 283-96.

17. Blanpain, C., Horsley, V. & Fuchs, E. (2007) Epithelial stem cells: turning over new leaves, Cell. 128, 445-58.

18. Medema, J. P. & Vermeulen, L. (2011) Microenvironmental regulation of stem cells in intestinal homeo-stasis and cancer, Nature. 474, 318-26.

19. Reya, T., Duncan, A. W., Ailles, L., Domen, J., Scherer, D. C., Willert, K., Hintz, L., Nusse, R. & Weissman, I. L. (2003) A role for Wnt signalling in self-renewal of haematopoietic stem cells, Nature. 423, 409-14.

(17)

20. Zeng, Y. A. & Nusse, R. (2010) Wnt proteins are self-renewal factors for mammary stem cells and promote their long-term expansion in culture, Cell stem cell. 6, 568-77.

21. Kuhnert, F., Davis, C. R., Wang, H. T., Chu, P., Lee, M., Yuan, J., Nusse, R. & Kuo, C. J. (2004) Essential requirement for Wnt signaling in proliferation of adult small intestine and colon revealed by adenoviral expression of Dickkopf-1, Proceedings of the National Academy of Sciences of the United States of America. 101, 266-71.

22. Duncan, A. W., Rattis, F. M., DiMascio, L. N., Congdon, K. L., Pazianos, G., Zhao, C., Yoon, K., Cook, J. M., Willert, K., Gaiano, N. & Reya, T. (2005) Integration of Notch and Wnt signaling in hematopoietic stem cell maintenance, Nature immunology. 6, 314-22.

23. Bhardwaj, G., Murdoch, B., Wu, D., Baker, D. P., Williams, K. P., Chadwick, K., Ling, L. E., Karanu, F. N. & Bhatia, M. (2001) Sonic hedgehog induces the proliferation of primitive human hematopoietic cells via BMP regulation, Nature immunology. 2, 172-80.

24. Lai, K., Kaspar, B. K., Gage, F. H. & Schaffer, D. V. (2003) Sonic hedgehog regulates adult neural progen-itor proliferation in vitro and in vivo, Nature neuroscience. 6, 21-7.

25. Haramis, A. P., Begthel, H., van den Born, M., van Es, J., Jonkheer, S., Offerhaus, G. J. & Clevers, H. (2004) De novo crypt formation and juvenile polyposis on BMP inhibition in mouse intestine, Science. 303, 1684-6.

26. He, X. C., Zhang, J., Tong, W. G., Tawfik, O., Ross, J., Scoville, D. H., Tian, Q., Zeng, X., He, X., Wiede-mann, L. M., Mishina, Y. & Li, L. (2004) BMP signaling inhibits intestinal stem cell self-renewal through suppression of Wnt-beta-catenin signaling, Nature genetics. 36, 1117-21.

27. Jamora, C., DasGupta, R., Kocieniewski, P. & Fuchs, E. (2003) Links between signal transduction, tran-scription and adhesion in epithelial bud development, Nature. 422, 317-22.

28. Gupta, P. B., Onder, T. T., Jiang, G., Tao, K., Kuperwasser, C., Weinberg, R. A. & Lander, E. S. (2009) Iden-tification of selective inhibitors of cancer stem cells by high-throughput screening, Cell. 138, 645-59. 29. Malanchi, I., Peinado, H., Kassen, D., Hussenet, T., Metzger, D., Chambon, P., Huber, M., Hohl, D.,

Cano, A., Birchmeier, W. & Huelsken, J. (2008) Cutaneous cancer stem cell maintenance is dependent on beta-catenin signalling, Nature. 452, 650-3.

30. Teng, Y., Wang, X., Wang, Y. & Ma, D. (2010) Wnt/beta-catenin signaling regulates cancer stem cells in lung cancer A549 cells, Biochemical and biophysical research communications. 392, 373-9.

31. Vermeulen, L., De Sousa, E. M. F., van der Heijden, M., Cameron, K., de Jong, J. H., Borovski, T., Tuynman, J. B., Todaro, M., Merz, C., Rodermond, H., Sprick, M. R., Kemper, K., Richel, D. J., Stassi, G. & Medema, J. P. (2010) Wnt activity defines colon cancer stem cells and is regulated by the microenvi-ronment, Nature cell biology. 12, 468-76.

32. Lu, D., Choi, M. Y., Yu, J., Castro, J. E., Kipps, T. J. & Carson, D. A. (2011) Salinomycin inhibits Wnt sign-aling and selectively induces apoptosis in chronic lymphocytic leukemia cells, Proceedings of the National Academy of Sciences of the United States of America. 108, 13253-7.

33. Hoey, T., Yen, W. C., Axelrod, F., Basi, J., Donigian, L., Dylla, S., Fitch-Bruhns, M., Lazetic, S., Park, I. K., Sato, A., Satyal, S., Wang, X., Clarke, M. F., Lewicki, J. & Gurney, A. (2009) DLL4 blockade inhibits tumor growth and reduces tumor-initiating cell frequency, Cell stem cell. 5, 168-77.

34. Sikandar, S. S., Pate, K. T., Anderson, S., Dizon, D., Edwards, R. A., Waterman, M. L. & Lipkin, S. M. (2010) NOTCH signaling is required for formation and self-renewal of tumor-initiating cells and for repression of secretory cell differentiation in colon cancer, Cancer research. 70, 1469-78.

35. Fan, X., Khaki, L., Zhu, T. S., Soules, M. E., Talsma, C. E., Gul, N., Koh, C., Zhang, J., Li, Y. M., Maci-aczyk, J., Nikkhah, G., Dimeco, F., Piccirillo, S., Vescovi, A. L. & Eberhart, C. G. (2010) NOTCH pathway blockade depletes CD133-positive glioblastoma cells and inhibits growth of tumor neurospheres and xenografts, Stem cells. 28, 5-16.

(18)

2

36. Prasetyanti, P. R., Zimberlin, C. D., Bots, M., Vermeulen, L., Melo Fde, S. & Medema, J. P. (2013) Regu-lation of stem cell self-renewal and differentiation by Wnt and Notch are conserved throughout the adenoma-carcinoma sequence in the colon, Molecular cancer. 12, 126.

37. Liu, S., Dontu, G., Mantle, I. D., Patel, S., Ahn, N. S., Jackson, K. W., Suri, P. & Wicha, M. S. (2006) Hedgehog signaling and Bmi-1 regulate self-renewal of normal and malignant human mammary stem cells, Cancer research. 66, 6063-71.

38. Varnat, F., Duquet, A., Malerba, M., Zbinden, M., Mas, C., Gervaz, P. & Ruiz i Altaba, A. (2009) Human colon cancer epithelial cells harbour active HEDGEHOG-GLI signalling that is essential for tumour growth, recurrence, metastasis and stem cell survival and expansion, EMBO molecular medicine. 1, 338-51. 39. Justilien, V., Walsh, M. P., Ali, S. A., Thompson, E. A., Murray, N. R. & Fields, A. P. (2014) The PRKCI

and SOX2 oncogenes are coamplified and cooperate to activate Hedgehog signaling in lung squamous cell carcinoma, Cancer cell. 25, 139-51.

40. Myllyperkio, M. H. & Vilpo, J. A. (1999) Increased DNA single-strand break joining activity in UV-irra-diated CD34+ versus CD34- bone marrow cells, Mutation research. 425, 169-76.

41. Bao, S., Wu, Q., McLendon, R. E., Hao, Y., Shi, Q., Hjelmeland, A. B., Dewhirst, M. W., Bigner, D. D. & Rich, J. N. (2006) Glioma stem cells promote radioresistance by preferential activation of the DNA damage response, Nature. 444, 756-60.

42. Friedmann-Morvinski, D., Bushong, E. A., Ke, E., Soda, Y., Marumoto, T., Singer, O., Ellisman, M. H. & Verma, I. M. (2012) Dedifferentiation of neurons and astrocytes by oncogenes can induce gliomas in mice, Science. 338, 1080-4.

43. Gupta, P. B., Fillmore, C. M., Jiang, G., Shapira, S. D., Tao, K., Kuperwasser, C. & Lander, E. S. (2011) Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer cells, Cell. 146, 633-44.

44. Chaffer, C. L., Brueckmann, I., Scheel, C., Kaestli, A. J., Wiggins, P. A., Rodrigues, L. O., Brooks, M., Rein-hardt, F., Su, Y., Polyak, K., Arendt, L. M., Kuperwasser, C., Bierie, B. & Weinberg, R. A. (2011) Normal and neoplastic nonstem cells can spontaneously convert to a stem-like state, Proceedings of the National Academy of Sciences of the United States of America. 108, 7950-5.

45. Yin, A. H., Miraglia, S., Zanjani, E. D., Almeida-Porada, G., Ogawa, M., Leary, A. G., Olweus, J., Kearney, J. & Buck, D. W. (1997) AC133, a novel marker for human hematopoietic stem and progenitor cells, Blood. 90, 5002-12.

46. Zhu, L., Gibson, P., Currle, D. S., Tong, Y., Richardson, R. J., Bayazitov, I. T., Poppleton, H., Zakharenko, S., Ellison, D. W. & Gilbertson, R. J. (2009) Prominin 1 marks intestinal stem cells that are susceptible to neoplastic transformation, Nature. 457, 603-7.

47. Snippert, H. J., van Es, J. H., van den Born, M., Begthel, H., Stange, D. E., Barker, N. & Clevers, H. (2009) Prominin-1/CD133 marks stem cells and early progenitors in mouse small intestine, Gastroenterology. 136, 2187-2194 e1.

48. Hermann, P. C., Huber, S. L., Herrler, T., Aicher, A., Ellwart, J. W., Guba, M., Bruns, C. J. & Heeschen, C. (2007) Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer, Cell stem cell. 1, 313-23.

49. Barker, N., van Es, J. H., Kuipers, J., Kujala, P., van den Born, M., Cozijnsen, M., Haegebarth, A., Korving, J., Begthel, H., Peters, P. J. & Clevers, H. (2007) Identification of stem cells in small intestine and colon by marker gene Lgr5, Nature. 449, 1003-7.

50. Barker, N., Tan, S. & Clevers, H. (2013) Lgr proteins in epithelial stem cell biology, Development. 140, 2484-94.

51. Barker, N., Huch, M., Kujala, P., van de Wetering, M., Snippert, H. J., van Es, J. H., Sato, T., Stange, D. E., Begthel, H., van den Born, M., Danenberg, E., van den Brink, S., Korving, J., Abo, A., Peters, P. J., Wright, N., Poulsom, R. & Clevers, H. (2010) Lgr5(+ve) stem cells drive self-renewal in the stomach and build

(19)

52. Jaks, V., Barker, N., Kasper, M., van Es, J. H., Snippert, H. J., Clevers, H. & Toftgard, R. (2008) Lgr5 marks cycling, yet long-lived, hair follicle stem cells, Nature genetics. 40, 1291-9.

53. Kemper, K., Prasetyanti, P. R., De Lau, W., Rodermond, H., Clevers, H. & Medema, J. P. (2012) Mono-clonal antibodies against Lgr5 identify human colorectal cancer stem cells, Stem cells. 30, 2378-86. 54. Hirsch, D., Barker, N., McNeil, N., Hu, Y., Camps, J., McKinnon, K., Clevers, H., Ried, T. & Gaiser, T.

(2014) LGR5 positivity defines stem-like cells in colorectal cancer, Carcinogenesis. 35, 849-58.

55. Chute, J. P., Muramoto, G. G., Whitesides, J., Colvin, M., Safi, R., Chao, N. J. & McDonnell, D. P. (2006) Inhibition of aldehyde dehydrogenase and retinoid signaling induces the expansion of human hematopoietic stem cells, Proceedings of the National Academy of Sciences of the United States of America. 103, 11707-12.

56. Ginestier, C., Wicinski, J., Cervera, N., Monville, F., Finetti, P., Bertucci, F., Wicha, M. S., Birnbaum, D. & Charafe-Jauffret, E. (2009) Retinoid signaling regulates breast cancer stem cell differentiation, Cell cycle. 8, 3297-302.

57. Ma, I. & Allan, A. L. (2011) The role of human aldehyde dehydrogenase in normal and cancer stem cells, Stem cell reviews. 7, 292-306.

58. Ginestier, C., Hur, M. H., Charafe-Jauffret, E., Monville, F., Dutcher, J., Brown, M., Jacquemier, J., Viens, P., Kleer, C. G., Liu, S., Schott, A., Hayes, D., Birnbaum, D., Wicha, M. S. & Dontu, G. (2007) ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome, Cell stem cell. 1, 555-67.

59. Huang, E. H., Hynes, M. J., Zhang, T., Ginestier, C., Dontu, G., Appelman, H., Fields, J. Z., Wicha, M. S. & Boman, B. M. (2009) Aldehyde dehydrogenase 1 is a marker for normal and malignant human colonic stem cells (SC) and tracks SC overpopulation during colon tumorigenesis, Cancer research. 69, 3382-9. 60. Colak, S., Zimberlin, C. D., Fessler, E., Hogdal, L., Prasetyanti, P. R., Grandela, C. M., Letai, A. & Medema,

J. P. (2014) Decreased mitochondrial priming determines chemoresistance of colon cancer stem cells, Cell death and differentiation.

61. Ma, S., Lee, T. K., Zheng, B. J., Chan, K. W. & Guan, X. Y. (2008) CD133+ HCC cancer stem cells confer chemoresistance by preferential expression of the Akt/PKB survival pathway, Oncogene. 27, 1749-58. 62. Bertolini, G., Roz, L., Perego, P., Tortoreto, M., Fontanella, E., Gatti, L., Pratesi, G., Fabbri, A., Andriani,

F., Tinelli, S., Roz, E., Caserini, R., Lo Vullo, S., Camerini, T., Mariani, L., Delia, D., Calabro, E., Pastorino, U. & Sozzi, G. (2009) Highly tumorigenic lung cancer CD133+ cells display stem-like features and are spared by cisplatin treatment, Proceedings of the National Academy of Sciences of the United States of America. 106, 16281-6.

63. Eramo, A., Ricci-Vitiani, L., Zeuner, A., Pallini, R., Lotti, F., Sette, G., Pilozzi, E., Larocca, L. M., Peschle, C. & De Maria, R. (2006) Chemotherapy resistance of glioblastoma stem cells, Cell death and differentia-tion. 13, 1238-41.

64. Ginestier, C., Liu, S., Diebel, M. E., Korkaya, H., Luo, M., Brown, M., Wicinski, J., Cabaud, O., Charafe-Jauffret, E., Birnbaum, D., Guan, J. L., Dontu, G. & Wicha, M. S. (2010) CXCR1 blockade selectively targets human breast cancer stem cells in vitro and in xenografts, The Journal of clinical investigation. 120, 485-97.

65. Todaro, M., Alea, M. P., Di Stefano, A. B., Cammareri, P., Vermeulen, L., Iovino, F., Tripodo, C., Russo, A., Gulotta, G., Medema, J. P. & Stassi, G. (2007) Colon cancer stem cells dictate tumor growth and resist cell death by production of interleukin-4, Cell stem cell. 1, 389-402.

66. Dylla, S. J., Beviglia, L., Park, I. K., Chartier, C., Raval, J., Ngan, L., Pickell, K., Aguilar, J., Lazetic, S., Smith-Berdan, S., Clarke, M. F., Hoey, T., Lewicki, J. & Gurney, A. L. (2008) Colorectal cancer stem cells are enriched in xenogeneic tumors following chemotherapy, PloS one. 3, e2428.

(20)

2

67. Ishikawa, F., Yoshida, S., Saito, Y., Hijikata, A., Kitamura, H., Tanaka, S., Nakamura, R., Tanaka, T., Tomiyama, H., Saito, N., Fukata, M., Miyamoto, T., Lyons, B., Ohshima, K., Uchida, N., Taniguchi, S., Ohara, O., Akashi, K., Harada, M. & Shultz, L. D. (2007) Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region, Nature biotechnology. 25, 1315-21. 68. Baumann, M., Krause, M. & Hill, R. (2008) Exploring the role of cancer stem cells in radioresistance,

Nature reviews Cancer. 8, 545-54.

69. Diehn, M., Cho, R. W., Lobo, N. A., Kalisky, T., Dorie, M. J., Kulp, A. N., Qian, D., Lam, J. S., Ailles, L. E., Wong, M., Joshua, B., Kaplan, M. J., Wapnir, I., Dirbas, F. M., Somlo, G., Garberoglio, C., Paz, B., Shen, J., Lau, S. K., Quake, S. R., Brown, J. M., Weissman, I. L. & Clarke, M. F. (2009) Association of reactive oxygen species levels and radioresistance in cancer stem cells, Nature. 458, 780-3.

70. Chen, J., Li, Y., Yu, T. S., McKay, R. M., Burns, D. K., Kernie, S. G. & Parada, L. F. (2012) A restricted cell population propagates glioblastoma growth after chemotherapy, Nature. 488, 522-6.

71. Wilson, B. J., Schatton, T., Zhan, Q., Gasser, M., Ma, J., Saab, K. R., Schanche, R., Waaga-Gasser, A. M., Gold, J. S., Huang, Q., Murphy, G. F., Frank, M. H. & Frank, N. Y. (2011) ABCB5 identifies a therapy-refractory tumor cell population in colorectal cancer patients, Cancer research. 71, 5307-16.

72. Li, X., Lewis, M. T., Huang, J., Gutierrez, C., Osborne, C. K., Wu, M. F., Hilsenbeck, S. G., Pavlick, A., Zhang, X., Chamness, G. C., Wong, H., Rosen, J. & Chang, J. C. (2008) Intrinsic resistance of tumori-genic breast cancer cells to chemotherapy, Journal of the National Cancer Institute. 100, 672-9.

73. Yu, F., Yao, H., Zhu, P., Zhang, X., Pan, Q., Gong, C., Huang, Y., Hu, X., Su, F., Lieberman, J. & Song, E. (2007) let-7 regulates self renewal and tumorigenicity of breast cancer cells, Cell. 131, 1109-23. 74. Tamura, K., Aoyagi, M., Wakimoto, H., Ando, N., Nariai, T., Yamamoto, M. & Ohno, K. (2010)

Accumu-lation of CD133-positive glioma cells after high-dose irradiation by Gamma Knife surgery plus external beam radiation, Journal of neurosurgery. 113, 310-8.

75. Tait, S. W. & Green, D. R. (2010) Mitochondria and cell death: outer membrane permeabilization and beyond, Nature reviews Molecular cell biology. 11, 621-32.

76. Madjd, Z., Mehrjerdi, A. Z., Sharifi, A. M., Molanaei, S., Shahzadi, S. Z. & Asadi-Lari, M. (2009) CD44+ cancer cells express higher levels of the anti-apoptotic protein Bcl-2 in breast tumours, Cancer immunity. 9, 4. 77. Konopleva, M., Zhao, S., Hu, W., Jiang, S., Snell, V., Weidner, D., Jackson, C. E., Zhang, X., Champlin,

R., Estey, E., Reed, J. C. & Andreeff, M. (2002) The anti-apoptotic genes Bcl-X(L) and Bcl-2 are over-expressed and contribute to chemoresistance of non-proliferating leukaemic CD34+ cells, British journal of haematology. 118, 521-34.

78. Liu, G., Yuan, X., Zeng, Z., Tunici, P., Ng, H., Abdulkadir, I. R., Lu, L., Irvin, D., Black, K. L. & Yu, J. S. (2006) Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma, Molecular cancer. 5, 67.

79. Van Houdt, W. J., Emmink, B. L., Pham, T. V., Piersma, S. R., Verheem, A., Vries, R. G., Fratantoni, S. A., Pronk, A., Clevers, H., Borel Rinkes, I. H., Jimenez, C. R. & Kranenburg, O. (2011) Comparative proteomics of colon cancer stem cells and differentiated tumor cells identifies BIRC6 as a potential therapeutic target, Molecular & cellular proteomics : MCP. 10, M111 011353.

80. Del Gaizo Moore, V. & Letai, A. (2013) BH3 profiling--measuring integrated function of the mitochon-drial apoptotic pathway to predict cell fate decisions, Cancer letters. 332, 202-5.

81. de Grouw, E. P., Raaijmakers, M. H., Boezeman, J. B., van der Reijden, B. A., van de Locht, L. T., de Witte, T. J., Jansen, J. H. & Raymakers, R. A. (2006) Preferential expression of a high number of ATP binding cassette transporters in both normal and leukemic CD34+CD38- cells, Leukemia. 20, 750-4.

82. Shervington, A. & Lu, C. (2008) Expression of multidrug resistance genes in normal and cancer stem cells, Cancer investigation. 26, 535-42.

(21)

83. Schatton, T., Murphy, G. F., Frank, N. Y., Yamaura, K., Waaga-Gasser, A. M., Gasser, M., Zhan, Q., Jordan, S., Duncan, L. M., Weishaupt, C., Fuhlbrigge, R. C., Kupper, T. S., Sayegh, M. H. & Frank, M. H. (2008) Identification of cells initiating human melanomas, Nature. 451, 345-9.

84. Emmink, B. L., Van Houdt, W. J., Vries, R. G., Hoogwater, F. J., Govaert, K. M., Verheem, A., Nijkamp, M. W., Steller, E. J., Jimenez, C. R., Clevers, H., Borel Rinkes, I. H. & Kranenburg, O. (2011) Differenti-ated human colorectal cancer cells protect tumor-initiating cells from irinotecan, Gastroenterology. 141, 269-78.

85. Gao, M. Q., Choi, Y. P., Kang, S., Youn, J. H. & Cho, N. H. (2010) CD24+ cells from hierarchically organ-ized ovarian cancer are enriched in cancer stem cells, Oncogene. 29, 2672-80.

86. Roesch, A., Fukunaga-Kalabis, M., Schmidt, E. C., Zabierowski, S. E., Brafford, P. A., Vultur, A., Basu, D., Gimotty, P., Vogt, T. & Herlyn, M. (2010) A temporarily distinct subpopulation of slow-cycling mela-noma cells is required for continuous tumor growth, Cell. 141, 583-94.

87. Dembinski, J. L. & Krauss, S. (2009) Characterization and functional analysis of a slow cycling stem cell-like subpopulation in pancreas adenocarcinoma, Clinical & experimental metastasis. 26, 611-23.

88. Signore, M., Pelacchi, F., di Martino, S., Runci, D., Biffoni, M., Giannetti, S., Morgante, L., De Majo, M., Petricoin, E. F., Stancato, L., Larocca, L. M., De Maria, R., Pallini, R. & Ricci-Vitiani, L. (2014) Combined PDK1 and CHK1 inhibition is required to kill glioblastoma stem-like cells in vitro and in vivo, Cell death & disease. 5, e1223.

89. Bartucci, M., Svensson, S., Romania, P., Dattilo, R., Patrizii, M., Signore, M., Navarra, S., Lotti, F., Biffoni, M., Pilozzi, E., Duranti, E., Martinelli, S., Rinaldo, C., Zeuner, A., Maugeri-Sacca, M., Eramo, A. & De Maria, R. (2012) Therapeutic targeting of Chk1 in NSCLC stem cells during chemotherapy, Cell death and differentiation. 19, 768-78.

90. Niida, H., Tsuge, S., Katsuno, Y., Konishi, A., Takeda, N. & Nakanishi, M. (2005) Depletion of Chk1 leads to premature activation of Cdc2-cyclin B and mitotic catastrophe, The Journal of biological chemistry. 280, 39246-52.

91. Sausville, E., Lorusso, P., Carducci, M., Carter, J., Quinn, M. F., Malburg, L., Azad, N., Cosgrove, D., Knight, R., Barker, P., Zabludoff, S., Agbo, F., Oakes, P. & Senderowicz, A. (2014) Phase I dose-escalation study of AZD7762, a checkpoint kinase inhibitor, in combination with gemcitabine in US patients with advanced solid tumors, Cancer chemotherapy and pharmacology. 73, 539-49.

92. Mir, S. E., De Witt Hamer, P. C., Krawczyk, P. M., Balaj, L., Claes, A., Niers, J. M., Van Tilborg, A. A., Zwinderman, A. H., Geerts, D., Kaspers, G. J., Peter Vandertop, W., Cloos, J., Tannous, B. A., Wesseling, P., Aten, J. A., Noske, D. P., Van Noorden, C. J. & Wurdinger, T. (2010) In silico analysis of kinase expres-sion identifies WEE1 as a gatekeeper against mitotic catastrophe in glioblastoma, Cancer cell. 18, 244-57. 93. Kemper, K., Rodermond, H., Colak, S., Grandela, C. & Medema, J. P. (2012) Targeting colorectal cancer

stem cells with inducible caspase-9, Apoptosis : an international journal on programmed cell death. 17, 528-37. 94. Oltersdorf, T., Elmore, S. W., Shoemaker, A. R., Armstrong, R. C., Augeri, D. J., Belli, B. A., Bruncko,

M., Deckwerth, T. L., Dinges, J., Hajduk, P. J., Joseph, M. K., Kitada, S., Korsmeyer, S. J., Kunzer, A. R., Letai, A., Li, C., Mitten, M. J., Nettesheim, D. G., Ng, S., Nimmer, P. M., O’Connor, J. M., Oleksijew, A., Petros, A. M., Reed, J. C., Shen, W., Tahir, S. K., Thompson, C. B., Tomaselli, K. J., Wang, B., Wendt, M. D., Zhang, H., Fesik, S. W. & Rosenberg, S. H. (2005) An inhibitor of Bcl-2 family proteins induces regression of solid tumours, Nature. 435, 677-81.

95. Wilson, W. H., O’Connor, O. A., Czuczman, M. S., LaCasce, A. S., Gerecitano, J. F., Leonard, J. P., Tulpule, A., Dunleavy, K., Xiong, H., Chiu, Y. L., Cui, Y., Busman, T., Elmore, S. W., Rosenberg, S. H., Krivoshik, A. P., Enschede, S. H. & Humerickhouse, R. A. (2010) Navitoclax, a targeted high-affinity inhibitor of BCL-2, in lymphoid malignancies: a phase 1 dose-escalation study of safety, pharmacokinetics, pharma-codynamics, and antitumour activity, The lancet oncology. 11, 1149-59.

(22)

2

96. Zeuner, A., Francescangeli, F., Contavalli, P., Zapparelli, G., Apuzzo, T., Eramo, A., Baiocchi, M., De Angelis, M. L., Biffoni, M., Sette, G., Todaro, M., Stassi, G. & De Maria, R. (2014) Elimination of quies-cent/slow-proliferating cancer stem cells by Bcl-X inhibition in non-small cell lung cancer, Cell death and differentiation.

97. Swaminathan, S. K., Roger, E., Toti, U., Niu, L., Ohlfest, J. R. & Panyam, J. (2013) CD133-targeted paclitaxel delivery inhibits local tumor recurrence in a mouse model of breast cancer, Journal of controlled release : official journal of the Controlled Release Society. 171, 280-7.

98. Damek-Poprawa, M., Volgina, A., Korostoff, J., Sollecito, T. P., Brose, M. S., O’Malley, B. W., Jr., Akintoye, S. O. & DiRienzo, J. M. (2011) Targeted inhibition of CD133+ cells in oral cancer cell lines, Journal of dental research. 90, 638-45.

99. Bach, P., Abel, T., Hoffmann, C., Gal, Z., Braun, G., Voelker, I., Ball, C. R., Johnston, I. C., Lauer, U. M., Herold-Mende, C., Muhlebach, M. D., Glimm, H. & Buchholz, C. J. (2013) Specific elimination of CD133+ tumor cells with targeted oncolytic measles virus, Cancer research. 73, 865-74.

100. Wang, C. H., Chiou, S. H., Chou, C. P., Chen, Y. C., Huang, Y. J. & Peng, C. A. (2011) Photothermolysis of glioblastoma stem-like cells targeted by carbon nanotubes conjugated with CD133 monoclonal anti-body, Nanomedicine : nanotechnology, biology, and medicine. 7, 69-79.

101. Bostad, M., Berg, K., Hogset, A., Skarpen, E., Stenmark, H. & Selbo, P. K. (2013) Photochemical inter-nalization (PCI) of immunotoxins targeting CD133 is specific and highly potent at femtomolar levels in cells with cancer stem cell properties, Journal of controlled release : official journal of the Controlled Release Society. 168, 317-26.

102. Majeti, R., Chao, M. P., Alizadeh, A. A., Pang, W. W., Jaiswal, S., Gibbs, K. D., Jr., van Rooijen, N. & Weissman, I. L. (2009) CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells, Cell. 138, 286-99.

103. Willingham, S. B., Volkmer, J. P., Gentles, A. J., Sahoo, D., Dalerba, P., Mitra, S. S., Wang, J., Contreras-Trujillo, H., Martin, R., Cohen, J. D., Lovelace, P., Scheeren, F. A., Chao, M. P., Weiskopf, K., Tang, C., Volkmer, A. K., Naik, T. J., Storm, T. A., Mosley, A. R., Edris, B., Schmid, S. M., Sun, C. K., Chua, M. S., Murillo, O., Rajendran, P., Cha, A. C., Chin, R. K., Kim, D., Adorno, M., Raveh, T., Tseng, D., Jaiswal, S., Enger, P. O., Steinberg, G. K., Li, G., So, S. K., Majeti, R., Harsh, G. R., van de Rijn, M., Teng, N. N., Sunwoo, J. B., Alizadeh, A. A., Clarke, M. F. & Weissman, I. L. (2012) The CD47-signal regula-tory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors, Proceedings of the National Academy of Sciences of the United States of America. 109, 6662-7.

104.Todaro, M., Gaggianesi, M., Catalano, V., Benfante, A., Iovino, F., Biffoni, M., Apuzzo, T., Sperduti, I., Volpe, S., Cocorullo, G., Gulotta, G., Dieli, F., De Maria, R. & Stassi, G. (2014) CD44v6 is a marker of constitutive and reprogrammed cancer stem cells driving colon cancer metastasis, Cell stem cell. 14, 342-56.

105.Zhao, C., Blum, J., Chen, A., Kwon, H. Y., Jung, S. H., Cook, J. M., Lagoo, A. & Reya, T. (2007) Loss of beta-catenin impairs the renewal of normal and CML stem cells in vivo, Cancer cell. 12, 528-41. 106. Wang, Y., Krivtsov, A. V., Sinha, A. U., North, T. E., Goessling, W., Feng, Z., Zon, L. I. & Armstrong, S. A.

(2010) The Wnt/beta-catenin pathway is required for the development of leukemia stem cells in AML, Science. 327, 1650-3.

107.Heidel, F. H., Bullinger, L., Feng, Z., Wang, Z., Neff, T. A., Stein, L., Kalaitzidis, D., Lane, S. W. & Armstrong, S. A. (2012) Genetic and pharmacologic inhibition of beta-catenin targets imatinib-resistant leukemia stem cells in CML, Cell stem cell. 10, 412-24.

108. McAuliffe, S. M., Morgan, S. L., Wyant, G. A., Tran, L. T., Muto, K. W., Chen, Y. S., Chin, K. T., Partridge, J. C., Poole, B. B., Cheng, K. H., Daggett, J., Jr., Cullen, K., Kantoff, E., Hasselbatt, K., Berkowitz, J., Muto, M. G., Berkowitz, R. S., Aster, J. C., Matulonis, U. A. & Dinulescu, D. M. (2012) Targeting Notch, a key pathway for ovarian cancer stem cells, sensitizes tumors to platinum therapy, Proceedings of the National Academy of Sciences of the United States of America. 109, E2939-48.

(23)

109. Kobune, M., Takimoto, R., Murase, K., Iyama, S., Sato, T., Kikuchi, S., Kawano, Y., Miyanishi, K., Sato, Y., Niitsu, Y. & Kato, J. (2009) Drug resistance is dramatically restored by hedgehog inhibitors in CD34+ leukemic cells, Cancer science. 100, 948-55.

110.Mueller, M. T., Hermann, P. C., Witthauer, J., Rubio-Viqueira, B., Leicht, S. F., Huber, S., Ellwart, J. W., Mustafa, M., Bartenstein, P., D’Haese, J. G., Schoenberg, M. H., Berger, F., Jauch, K. W., Hidalgo, M. & Heeschen, C. (2009) Combined targeted treatment to eliminate tumorigenic cancer stem cells in human pancreatic cancer, Gastroenterology. 137, 1102-13.

111.Bar, E. E., Chaudhry, A., Lin, A., Fan, X., Schreck, K., Matsui, W., Piccirillo, S., Vescovi, A. L., DiMeco, F., Olivi, A. & Eberhart, C. G. (2007) Cyclopamine-mediated hedgehog pathway inhibition depletes stem-like cancer cells in glioblastoma, Stem cells. 25, 2524-33.

112.Zhao, C., Chen, A., Jamieson, C. H., Fereshteh, M., Abrahamsson, A., Blum, J., Kwon, H. Y., Kim, J., Chute, J. P., Rizzieri, D., Munchhof, M., VanArsdale, T., Beachy, P. A. & Reya, T. (2009) Hedgehog signal-ling is essential for maintenance of cancer stem cells in myeloid leukaemia, Nature. 458, 776-9. 113.Calabrese, C., Poppleton, H., Kocak, M., Hogg, T. L., Fuller, C., Hamner, B., Oh, E. Y., Gaber, M. W.,

Finklestein, D., Allen, M., Frank, A., Bayazitov, I. T., Zakharenko, S. S., Gajjar, A., Davidoff, A. & Gilbertson, R. J. (2007) A perivascular niche for brain tumor stem cells, Cancer cell. 11, 69-82.

114.Jin, L., Hope, K. J., Zhai, Q., Smadja-Joffe, F. & Dick, J. E. (2006) Targeting of CD44 eradicates human acute myeloid leukemic stem cells, Nature medicine. 12, 1167-74.

115.Lombardo, Y., Scopelliti, A., Cammareri, P., Todaro, M., Iovino, F., Ricci-Vitiani, L., Gulotta, G., Dieli, F., de Maria, R. & Stassi, G. (2011) Bone morphogenetic protein 4 induces differentiation of colorectal cancer stem cells and increases their response to chemotherapy in mice, Gastroenterology. 140, 297-309. 116.Piccirillo, S. G., Reynolds, B. A., Zanetti, N., Lamorte, G., Binda, E., Broggi, G., Brem, H., Olivi, A.,

Dimeco, F. & Vescovi, A. L. (2006) Bone morphogenetic proteins inhibit the tumorigenic potential of human brain tumour-initiating cells, Nature. 444, 761-5.

117.Heijmans, J., van Lidth de Jeude, J. F., Koo, B. K., Rosekrans, S. L., Wielenga, M. C., van de Wetering, M., Ferrante, M., Lee, A. S., Onderwater, J. J., Paton, J. C., Paton, A. W., Mommaas, A. M., Kodach, L. L., Hardwick, J. C., Hommes, D. W., Clevers, H., Muncan, V. & van den Brink, G. R. (2013) ER stress causes rapid loss of intestinal epithelial stemness through activation of the unfolded protein response, Cell reports. 3, 1128-39.

Referenties

GERELATEERDE DOCUMENTEN

Grigg and coworkers showed that intramolecular carbocyclization of aliènes leads to the formation of carbo- and heterocycles and an excellent review has appeared about this

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), other than for strictly

Uit een t-toets van Herkenning van de kernwaarden bleek er geen significant verschil te zijn tussen het bekende merk FrieslandCampina en het onbekende merk Parmalat wat betreft

Maar daarvan biedt TIN-20 eigenlijk geen voorbeelden, want in de voorbeelden die ik hier- voor genoemd heb, is de techniek voornamelijk gebruikt om de maatschappelijke context scherp

Een nieuwe nexus, gebaseerd op een significante (digitale) aanwezigheid, kan bijdragen aan een beter systeem, maar dan dient deze mogelijkheid eerst verder te worden uitgewerkt

[r]

Besides the expression of cell cycle inhibitors and DNA damage proteins, senescent cells are charac- terised by increased lysosomal activity, exemplified by induction of the

[r]