PHD Kanishka Thesis
PHD Kanishka Thesis
Kanishka Tiwary
Sareshkunda, India
2023
Dean of the medical faculty:
Prof. Dr. Thomas Wirth
External reviewer:
Dr. Shiv K. Singh
Prof. Dr. Dr. Daniel Stange
1. Walter, K., Tiwary, K., Trajkovic-Arsic, M., Hidalgo-Sastre, A., Dierichs, L.,
T. Liffers, S., Gu, J., Gout, J., Schulte, L.-A., Münch, J., Seufferlein, T., Sainz,
B.-Jr., Hermann, P.-C. MEK inhibition targets cancer stem cells and impedes
migration of pancreatic cancer cells in vitro and in vivo. Stem Cells Int. 2019
Jun 2, 8475389. doi: 10.1155/2019/8475389.
1. Godbole, M., Togar, T., Patel K., Dharavath B., Yadav, N., Janjuha, S., Gardi,
N., Tiwary, K., Terwadkar, P., Desai, R.-P., Dhamne, H., Karve, K.,
Salunkhe, S., Kawle, D., Chandrani, P., Dutt, S., Gupta, S., Badwe, R.-A.,
Dutt, A. Up-regulation of the kinase gene SGK1 by progesterone activates
the AP-1–NDRG1 axis in both PR-positive and -negative breast cancer cells.
J. Biol. Chem. 2018 Dec 14, 293 (50): 19263-19276. doi:
10.1074/jbc.RA118.002894.
2. Valle, S., Alcalá, S., Martin-Hijano, L., Cabezas-Sáinz, P., Navarro, D.,
Ramos Muñoz, E., Yuste, L., Tiwary, K., Walter, K., Ruiz-Cañas, L., Alonso-
Nocelo, M., Rubiolo, J.-A., González-Arnay, E., Heeschen, C., Garcia-
Bermejo, L., Hermann, P.-C., Sánchez , L., Sancho, P., Fernández-Moreno
M.- Á., Sainz, B.-Jr. Exploiting oxidative phosphorylation to promote the stem
and immunoevasive properties of pancreatic cancer stem cells. Nat.
Commun. 2020 Oct 16, 11, 5265. https://doi.org/10.1038/s41467-020-
18954-z.
3. Walter, K., Rodriguez-Aznar, E., Ferreira, M.S.V., Frappart, P.-O., Dittrich,
T., Tiwary, K., Meessen, S., Lerma, L., Daiss, N., Schulte, L.-A., Najafova,
Z., Arnold, F., Usachov, V., Azoitei, N., Erkan, M., Lechel, A., Brümmendorf,
T.H., Seufferlein, T., Kleger, A., Tabarés, E., Günes, C., Johnsen, S.A., Beier,
F., Sainz, B.-Jr., Hermann, P.-C. Telomerase and Pluripotency Factors
Jointly Regulate Stemness in Pancreatic Cancer Stem Cells. Cancers. 2021
Jun 23, 13, 3145. https://doi.org/10.3390/cancers13133145.
4. Tsesmelis, M., Tiwary, K., Steiger, K., Sperb, N., Gerstenlauer, M., Manfras,
U., Maier, H.J., Hermann, P.C., Chan, L.K., Wirth, T. Deletion of NEMO
Inhibits EMT and Reduces Metastasis in KPC Mice. Cancers. 2021 Sep
10, 13, 4541. https://doi.org/10.3390/cancers13184541.
TABLE OF CONTENTS
1 TABLE OF CONTENTS
2 LIST OF ABBREVIATIONS III
3 INTRODUCTION 1
3.1 Pancreatic ductal adenocarcinoma (PDAC) 1
3.1.1 KRAS: the driver mutation and an elusive target 1
3.1.2 Cellular plasticity and metastasis in PDAC 3
3.1.3 Current modalities in PDAC treatment 4
3.2 Cancer Stem Cells (CSCs) 5
3.2.1 CD133: A bona fide CSC marker in PDAC 5
3.2.2 Impact of CSCs on therapy resistance 7
3.2.3 Migrating CSCs: invasive front of metastasis 8
3.3 PDAC and its tumor microenvironment (TME) 10
3.3.1 TME and active players in metastasis 10
3.3.2 TME – CSC interplay 12
3.3.3 The tumor microenvironment mediates resistance to therapy 13
3.4 CXCR4 – CXCL12 signaling 15
3.4.1 CXCR4 and CXCL12 relevance in the metastasis paradigm 16
3.4.2 Importance of CXCR4 – CXCL12 signaling during PDAC development 17
3.4.3 Current therapies and recent advances in targeting CXCR4 18
3.5 Hypothesis and aims 21
4 MATERIAL AND METHODS 23
4.1 Chemicals and reagents 23
4.2 Consumables 24
4.3 Devices 26
4.4 Mice and Cell lines 27
4.5 Co-culture technique 29
4.5.1 Co-culture with PSCs 29
4.5.2 Condition media for PSCs maintenance 30
4.5.3 Primed and Non-primed PSCs – CM preparation 30
4.6 Organoid culture 30
4.7 Treatments 31
4.8 MTT assay 31
4.9 Clonogenic assay 31
4.10 Sphere formation assay 31
4.11 Scratch wound healing assay 32
4.12 Migration assay 32
4.12.1 Migration towards serum 32
4.12.2 Migration towards CXCL12 32
4.13 Molecular Cloning 33
4.13.1 Bacterial culture and plasmid isolation 33
4.13.2 Lentiviral particle production in Lenti-X cells 33
4.13.3 Lentiviral transduction and selection of target cells 34
4.14 RNA isolation and real-time PCR 34
4.14.1 RNA Isolation and cDNA synthesis 34
4.14.2 Real-time quantitative PCR 34
4.15 RNA Seq 36
4.16 Protein extraction and western blot analysis 36
4.16.1 Protein sample preparation 36
4.16.2 Western Blot 37
4.17 Immunofluorescence 38
4.17.1 E-cadherin 38
4.17.2 Nile Red and Phalloidin 38
4.17.3 Phalloidin 39
4.18 Flow Cytometry 39
I
TABLE OF CONTENTS
II
List of abbreviations
2 List of abbreviations
5-FU 5- Fluorouracil
ADM Acinar to ductal metaplasia
ALDH Aldehyde dehydrogenase
BMI1 B lymphoma Mo-MLV insertion region 1 homolog
CAF Cancer associated fibroblast
CSC Cancer stem cell
CXCL12 C-X-C motif chemokine 12
CXCR4 C-X-C chemokine receptor type 4
DAPI 4´,6-diamidin-2-phenylindol
DCs Dendritic cells
DFS Diesease free survival
DMSO Dimethyl sulfoxide
ECM Extra cellular matrix
EMT Epithelial to mesenchymal transition
EPI-X4 Endogenous Peptide Inhibitor of CXCR4
FBS Fetal bovine serum
FCS Fetal calf serum
FOLFIRINOX Folic acid, 5 Fluorouracil, irinotecan, oxaliplatin
GEMM Genetically engineered mouse model
GEPIA Gene expression profiling interactive analysis
GSH Glutathione
iCAF inflammatory cancer associated fibroblast
IF Immunofluorescence
IL Interleukins
IPMN Intraductal papillary mucinous neoplasm
KRAS Kirsten rat sarcoma virus
MAPK Mitogen-activated protein kinase
MCN Mucinous cystic neoplasm
MDSCs Myeloid-derived suppressor cells
III
List of abbreviations
IV
Introduction
3 Introduction
3.1 Pancreatic ductal adenocarcinoma (PDAC)
The pancreas is a spongy, tube-shaped organ that is about 15 cm long and located
in the retroperitoneum between the stomach and the spine, in the upper left
abdomen2. A normal healthy pancreas is comprised of acinar cells that secrete
digestive enzymes, ductal cells that secrete bicarbonate, centro-acinar cells as the
transitional region between acinar and ductal cells, endocrine islets which produce
hormones, and relatively inactive stellate cells3. Mutations in the pancreas can result
in pancreatic cancer, causing the pancreatic cells to uncontrollably grow and divide,
forming tumors4. Pancreatic cancer is characterized as a fatal disease and is one of
the most aggressive and lethal malignancies worldwide5. It accounts for more than
a fifth of all gastrointestinal cancer-related deaths and is projected to be the 2nd most
lethal cancer by 20306. Most cases (90%) are pancreatic ductal adenocarcinomas
(PDAC), which arise from the pancreatic epithelium7. Due to the lack of early
symptoms at the time of diagnosis, PDAC patients mostly present at an advanced
and/or metastasized stage8,9. Nearly 80% patients undergoing surgery with curative
intent ultimately relapse, with 2 out of 3 succumbing to distant recurrence9.
Moreover, 5-year overall survival is 3% for patients with metastasis, an outcome
that has largely remained unchanged in the past 15 years10,11.
1
Introduction
PanINs are part of a multistep tumor progression model in PDAC: Three stages of
PanINs show increasing cellular atypia and typically harbour step-specific
mutations20,21. Changes in the epithelium also result in increasing desmoplasia (Fig
3.1.1A). One of the earliest genetic alterations in this process is the activation of
Kirsten rat sarcoma viral oncogene homolog (KRAS), accompanied by the
inactivation of tumor suppressor genes cyclin-dependent kinase inhibitor p16
(p16)/cyclin-dependent kinase inhibitor 2A (CDKN2A)22,23. Activating mutations in
KRAS are the driver for additional inactivating mutations in tumor suppressor genes
in a rate-limiting step for tumor progression. Therefore, during later PanIN stages,
oncogenic hits in TP53 and inactivating mutations in SMAD4 pave the way for
invasiveness and metastatic burden24 (Fig 3.1.1). Moreover, epigenetic regulation
can repress key tumor suppressor genes and upregulate oncogenes25. In this
regard, tumor progression and metastasis formation have also been linked to
epigenetic (re-)programing26,27.
Given that mutations in one RAS protein isoform, KRAS, are found in nearly 90% of
pancreatic cancers, RAS signaling appears to play a critical role in both pancreatic
cancer initiation and maintenance28,29. Activated RAS can cause cell transformation,
proliferation, and metastasis by activating effector signaling pathways and
transcription factors30. RAS activation can also promote pro-inflammatory signaling
via activation of NF-κB, STAT3, and glycogen synthase kinase-3/nuclear factor of
activated T cell signaling31-33. Knockdown of KRAS in cancer cell lines diminishes
cell proliferation and activates apoptosis34, making KRAS a prime target for
therapeutic interventions in PDAC. Unfortunately, attempts to develop
adequate KRAS inhibitors have been hindered due to various unique features of
oncogenic KRAS, e.g., its high affinity for GTP and redundant pathways for
membrane localization. In this regard, Muzumdar et al. have shown that most PDAC
cell lines can survive in spite of CRISPR/Cas-mediated genetic ablation of KRAS,
pointing towards a potential for resistance to even the very best KRAS inhibitors35.
Hence, despite its central role in PDAC, KRAS still remains “undruggable”, shifting
the therapeutic focus to new approaches targeting crucial downstream effectors.
2
Introduction
Figure 3.1.1 PanIN stages and tumor progression model for PDAC. (Reprinted with permission
from SPRINGER NATURE. Nature Reviews Cancer. Ref No.24. KRAS, Hedgehog, Wnt and the
twisted developmental biology of pancreatic ductal adenocarcinoma. Morris, J.-P. IV., Wang S.-C.,
Hebrok, M. ©2010).
3
Introduction
to determine the kinetics of EMT and dissemination during the natural evolution of
PDAC in vivo and demonstrated that EMT occurs as early as during premalignant
lesions. They further discovered that the spread of pancreatic epithelial cells in mice
in fact precedes tumor formation46.
Figure 3.1.2 EMT activating signaling pathways. (Reprinted with permission from SPRINGER
NATURE. Nature Reviews Molecular Cell Biology. Ref No.47. New insights into the mechanisms of
epithelial–mesenchymal transition and implications for cancer. Dongre, A., Weinberg, R.-A. ©2018).
4
Introduction
regimen are associated with higher toxicity16,53, and can therefore be used only in
patients with high performance scores.
Although checkpoint inhibition has been approved for treatment of the small subset
of PDAC tumors with high microsatellite instability (1-2% of all cases54,55), the
therapeutic efficacy of immunotherapeutic approaches in PDAC has been
unfortunately limited56,57. Interestingly, approximately 7% of patients have mutations
involving DNA repair genes (e.g., BRCA2 and PALB2), which might allow the use
of PARP inhibitors in this subset of patients58. While new drugs are continuously
developed and improved, some of the major hurdles in PDAC therapy are the highly
heterogeneous tumor system consisting of cells with varying degrees of
differentiation (e.g., poorly differentiated cancer stem cells) and physical
inaccessibility of the tumors due to highly desmoplastic stroma.
5
Introduction
6
Introduction
Figure 3.2.1 Pancreatic cancer stem cell markers for stemness, metastatic potential, EMT,
immune evasion and chemoresistance. (Reprinted from Valle, S., Martin-Hijano, L., Alcalá, S.,
Alonso-Nocelo, M., Sainz, B.-Jr. The ever-evolving concept of the cancer stem cell in pancreatic
cancer. doi: 10.3390/cancers10020033. Ref No.63. Licensed under a Creative Commons Attribution
License (CC BY 4.0) licence https://creativecommons.org/licenses/by/4.0/).
7
Introduction
drugs from these cells61,63,84,85. Recent studies also show that knockdown of NRF2
(a gene responsible for glutathione metabolism and antioxidant metabolism), re-
sensitizes PDAC cells towards 5-FU by downregulating CD133 and ABCG2
expression86.
Mani et al. described the role of EMT induction in immortalized mammary epithelial
cells and subsequently increased stem cell marker expression, mammosphere
formation and tumorigenicity in vivo, showing a direct association between EMT and
the development of CSCs characteristics91. More recent studies have discovered
the underlying mechanisms by which EMT programs control stemness:
Pastushenko et al. identified transition states that occur in the tumor during the
process of EMT. In their model, hybrid EMT populations displayed a 5-fold increase
in tumor propagation as compared to epithelial cancer cells and demonstrated that
the intermediate EMT subpopulations were the most plastic92. Furthermore, the
EMT International Association (TEMTIA) uses the term ‘epithelial–mesenchymal
plasticity’ (EMP) to describe the ability of cells to adopt mixed
8
Introduction
Expanding this concept, Hermann et al. indeed identified not only a CD133+ CSC
population with tumor initiating properties in PDAC62 but their study also identified a
CD133+CXCR4+ subpopulation of CSCs that was highly and exclusively
metastatic. Due to this exclusive metastatic activity, they were defined as “migrating
CSC (miCSC)”. The expression of CXCR4 in miCSCs has significant therapeutic
potential: Hermann et al. used AMD3100 or CXCR4 neutralizing antibodies to inhibit
CXCR4, and thus were able to abrogate metastasis. A clinical correlation between
migrating CSCs and advanced disease stages was established by the significantly
higher presence of (CD133+ CXCR4+) population in patients with lymph node
metastases (pN1+). After this landmark study by Hermann et al., migrating CSCs
have been described in a number of other cancer types99-101. This provides
convincing evidence for the crucial role of CXCR4 signaling in metastasis.
Figure 3.2.3 The metastatic cascade. From Chaffer CL, Weinberg RA. A perspective on cancer
cell metastasis. Science. 2011 Mar 25;331(6024):1559-64. doi: 10.1126/science.1203543. Ref.
No.102. Reprinted with permission from AAAS.
9
Introduction
Fundamentals for determining the capacity of cancer cells to escape from primary
tissues and subsequently seed and colonize distant tissues are (i) a permissive
tumor microenvironment and (ii) a receptive distant organ niche106. PDAC tumor
microenvironment components (Fig. 3.3.1) and factors secreted by them participate
in modulating metastasis. Tumor-associated macrophages (TAMs) are one of the
major immune cell types and cancer-associated fibroblasts (CAFs) are the primary
source of extracellular matrix deposition in the TME107. Studies have suggested a
crucial role of these components in cancer progression, metastasis, and
chemoresistance108. Furthermore, there is substantial evidence for tumor– stroma
crosstalk promoting metastasis109-111.
CAFs typically arise from tissue-resident fibroblasts (e.g., PSCs), but also from bone
marrow-derived cells and adipose tissue-derived mesenchymal stem cells (MSCs).
Interestingly, the heterogeneity in CAFs occurs based on their expression profile
and localization within the tumor microenvironment (e.g., myofibrotic CAFs,
[myCAFs]; inflammatory CAFs [iCAFs] and antigen presenting CAFs
10
Introduction
Figure 3.3.1 Components of PDAC microenvironment. (Reprinted from Truong, L.-H.; Pauklin, S.
Pancreatic Cancer Microenvironment and Cellular Composition: Current Understandings and
11
Introduction
12
Introduction
Clinical studies have demonstrated that the expression of certain TME components
can correlate with a poor patient prognosis and can facilitate desmoplasia and
immunosuppression or promote metastasis in PDAC120. Non-specific targeting of
the ECM alone is not effective in pancreatic cancer as MMP inhibitors such as
marimastat and tanomastat or enzymatic degradation of hyaluronan by human
recombinant PH20 hyaluronidase (PEGPH20) failed to show significant clinical
benefit in patients with advanced stage pancreatic cancer143-146. Another approach
to target the TME involves cancer-associated stromal cells or cancer-associated
fibroblasts (CAFs), which are one of the major producers of ECM proteins. CAFs
are generally spindle-shaped cells and express one or more activated fibroblast
markers (e.g., fibroblast activation protein [FAP] and α-smooth muscle actin [α-
SMA])147. Specific targeting of fibroblasts was first assessed in colorectal cancer
using the FAP targeting inhibitor sibrotuzumab, which failed to meet the end point
in a phase II trial148. Other small molecule inhibitors of FAP have shown limited
efficacy against PDAC149.
In mouse models of PanIN or PDAC, Erkan et al. showed that genetic deletion of α-
SMA expressing fibroblasts resulted in more aggressive disease150 and
demonstrated that high CAF activity with low collagen deposition is associated with
poor prognosis. However, it also appears that the desmoplastic reaction might
restrain tumor cells from expanding outside the pancreas151. Therefore, instead of
eliminating the stromal fibroblasts from the TME, a more sophisticated approach
could be to alter CAF phenotype. Stromal reprogramming, as opposed to ablation,
is gaining acceptance in the field of stroma-targeting approaches for the treatment
of PDAC152-154. Chronopoulos et al. showed that ATRA is an active metabolite of
vitamin A that restores quiescence in PSCs. In 3D organotypic models, the authors
depicted ATRA treatment inhibits local cancer cell invasion153. Some of the
attractive targets for TME-related therapy in PDAC are depicted in (Fig. 3.3.3).
With regard to the immune compartment of the TME, studies show that CXCL12
secreted from PSCs reduces the migration of CD8+ T cells into the peritumoral
stroma, and galectin-1 induces T-cell apoptosis and Th2 cytokine secretion, thereby
13
Introduction
Figure 3.3.3 Potential cancer stem cell and ECM targeting approaches in PDAC therapy.
(Reprinted from Truong, L.-H.; Pauklin, S. Pancreatic Cancer Microenvironment and Cellular
Composition: Current Understandings and Therapeutic Approaches. doi:
127
10.3390/cancers13195028. Ref No. . Licensed under a Creative Commons Attribution License (CC
BY 4.0) licence https://creativecommons.org/licenses/by/4.0/).
14
Introduction
15
Introduction
Figure 3.4 CXCR4-related signaling. (Reprinted from Truong, Xu C, Zhao H, Chen H, Yao Q.
CXCR4 in breast cancer: oncogenic role and therapeutic targeting. doi: 10.2147/DDDT.S84932. Ref
No.177. Licensed under a Creative Commons Attribution 3.0 Unported License (CC BY-NC 3.0)
licence https://creativecommons.org/licenses/by-nc/3.0/).
16
Introduction
CXC12 is present in the basal lamina of endothelial venules and helps CXCR4+
cancer cells to migrate to blood vessels from the primary tumor192. Furthermore,
liver, bone marrow, lungs and lymph nodes exhibit peak expression levels of
CXCL12 and are most common target organs for metastasis193. Kim et al. showed
the liver to be a selective target of CXCR4+ colorectal cancer cells due to the
abundance of CXCL12194. Similarly, Gelmini et al. reported that treatment with a
CXCR4 neutralizing antibody dramatically reduced the number and size of
metastases in vivo in endometrial cancer195. This also indicates the relevance of
chemokine modulation for organ-specific migration of cancer cells, ultimately
leading to metastasis196. Moreover, tumors such as PDAC are often hypoxic, and
HIF-1α induces CXCL12 upregulation, assisting in the recruitment of CXCR4+
CSCs to peripheral vessels as a pool for metastasis197,198. All these findings indicate
that after being activated by its ligand CXCL12, CXCR4 initiates signaling in tumor
cells that promotes metastasis in a variety of cancer types.
In terms of patient outcome, blood vessel density and perineural invasion are
prognostic markers for pancreatic cancer and are strongly associated with CXCR4
expression185,208. CXCR4 levels also correlate with lymph node and distant organ
17
Introduction
Apart from AMD3100, researchers have sought other methods to target CXCR4.
Some of them include utilizing neutralizing antibodies for immunotherapy for
colorectal cancer226, copolymer drug incorporating BKT140, for inhibiting prostate
cancer227, LFC131 peptide inhibitor conjugated PGLA nanoparticles for drug
delivery in lung cancer228 and HPMA block copolymer of hydroxychloroquine for
decreasing lung metastasis in breast cancer229. Compared to AMD3100, the
CXCR4 antagonist BL-8040 displayed a greater effect on the retention–mobilization
balance of bone marrow stem cells by providing a higher affinity and longer receptor
occupancy in mouse studies230,231. In preclinical models of pancreatic cancer,
inhibition of the CXCL12/CXCR4 axis enhanced T cell access to the TME and
increased tumor sensitivity to anti-PD-1 ligand-1 (PD-L1) therapy160,232. In this
18
Introduction
Only recently, Zirafi et al. screened an HF-derived (hemofiltrate) peptide library for
inhibitors of CXCR4-tropic (X4) HIV-1 and identified an endogenous antagonist of
CXCR4 generated by limited proteolysis of serum albumin (Fig. 3.4.3A), the most
abundant protein in human serum234. They identified EPI-X4 as an evolutionarily
conserved endogenous antagonist of CXCR4 that may play important roles in
physiological processes and diseases. Since its discovery and characterization in
2015 several derivatives of EPI-X4 have been generated by chemical modification,
most importantly WSCO2 and JM#21 (Fig. 3.4.3B)235. WSCO2 showed decreased
migratory capacity in vitro236 and while no immediate toxicity was observed, further
analysis for long-term toxicity of EPI-X4 and its derivatives is imperative. Recent
studies with Waldenström’s Macroglobulinemia (WM, B-cell lymphoma) confirmed
EPI-X4, and its optimized derivatives bind to CXCR4 on WM cells, and that they are
able to diminish growth of lymphoma cells in vivo237. Therefore, optimized
derivatives of EPI-X4 may represent a promising approach in targeting
CXCL12/CXCR4 signaling.
19
Introduction
Figure 3.4.3 (A) EPI-X4 is generated from serum albumin under acidic conditions by cathepsin
D or E. (Reprinted with permission from John Wiley and Sons. JOURNAL OF LEUKOCYTE
BIOLOGY. Ref No.238. Proteolytic processing of human serum albumin generates EPI-X4, an
endogenous antagonist of CXCR4. Zirafi, O., Hermann, P.-C, Münch, J. ©2016). (B) Intramolecular
H-bonds in EPI-X4, WSCO2 and JM#21. (Color code: C = pink/blue, H = white, N = blue, and O =
red. H-bonds are shown as dotted lines in orange. (EPI-X4 from the previously published thesis
Harms M: Endogenous CXCR4 Antagonists: Role in HIV-1 Transmission and Therapy of CXCR4-
linked diseases239, PhD Dissertation, Institute of Molecular Virology, University of Ulm, 2021.
WSCO2 and JM#21 from Harms M, Habib MMW, Nemska S, Nicolò A, Gilg A, Preising N, Sokkar
P, Carmignani S, Raasholm M, Weidinger G, Kizilsavas G, Wagner M, Ständker L, Abadi AH, Jumaa
H, Kirchhoff F, Frossard N, Sanchez-Garcia E, Münch J. An optimized derivative of an endogenous
CXCR4 antagonist prevents atopic dermatitis and airway inflammation. doi:
235
10.1016/j.apsb.2020.12.005. Ref No. . Licensed under a Creative Commons Attribution License
(CC BY-NC-ND 4.0) licence https://creativecommons.org/licenses/by-nc-nd/4.0/).
20
Introduction
The overall hypothesis of the present study is to discern molecular pathways that
are accountable for maintaining migrating cancer stem cells and their characteristic
features, and to exploit these targets in order to establish novel therapetic strategies.
Metastasis is still the major cause of mortality in patients with pancreatic ductal
adenocarcinoma (PDAC), yet this process is one of the most enigmatic aspects of
the diesease. KRAS, the key oncogenic driver mutation in PDAC, has been
implicated in promoting metastasis. During the different stages of metastasis,
circulating tumor cells (CTCs) act as a transition stage. We hypothesize that
RAS/MEK/ERK signaling may regulate CTC initiation. To address this issue, we
evaluated
(i) the role of MEK signaling downstream of RAS in aiding growth, self-renewal
and migratory propensity in PDAC cells,
(ii) the capacity of MEK signaling to inflect active EMT program, initiated by
TGFβ and
(iii) the effect of MEK inhibition on CTC numbers.
21
Introduction
22
Material and Methods
23
Material and Methods
4.2 Consumables
Table2: Consumables with type and the companies they were ordered from.
24
Material and Methods
25
Material and Methods
4.3 Devices
Table3: Devices with type and the companies they were ordered from.
Device Type Company
CASY TT Cell Counter (#5651697) OLS OMNI Life Science
Cell Counting chamber 0,100 mm depth, 0,0025 mm² Neubauer
Cell culture incubator Heracell Thermo Fisher Scientific
Centrifuges Centrifuge RC 6+ Thermo Fisher Scientific
Heraeus Multifuge X3R
Microcentrifuge 5417R Eppendorf
Chemiluminescence Fusion SL Vilber lourmat
imager
Flow cytometer LSR II BD
Fluorescence EVOS® FL AMF4300 Invitrogen
microscopes Axio Observer 7 Zeiss
Freezer/Refrigerator -20 °C/+4 °C (Premium Kombi) Liebherr
Freezer -80 °C (Hera Freeze KS 102) Thermo Fisher Scientific
Freezing container Mr. Frosty Thermo Fisher Scientific
Incubator (bacteria) Certomat® R Sartorius
Laminar airflow Herasafe HS9 (S/N 98105547) Heraeus
Magnetic stirrer #36560.00 IKA RCT classic
Microplate reader #185911 BioTek® Instruments
NanoDrop 2000 Thermo Fisher Scientific
PCR cycler DNAEngine® Bio-Rad
26
Material and Methods
All cell lines were maintained at 37°C in a humidified atmosphere with 5% CO2 in
the indicated culture media (Table4) supplemented with 10% FBS (ThermoFischer
Scientific, 10270-106) (20% FBS + 10% PSCs - CM for pancreatic stellate cells),
1% Penicillin-Streptomycin (PAN Biosystems, P06-07100) and 1% Glutamine
(Gibco, 41965-039). For experiments, cells were used in early passages (Pass. 3 -
20) and were recovered from frozen stocks on a regular basis. Single cells were
counted with Neubauer chambers and seeded accordingly.
Primary murine pancreatic cancer cell lines were generated from PDAC tumors
resected from Kraswt/LSL-G12D; Trp53loxP/loxP; Ptf1awt/Cre;LSL-tdRFPKI/KI;Slug-YFP
(KPCRS)240. These mice express an oncogenic Kras mutation241, a conditional loss
of Trp53242, an R26-LSL-tdRFP243, a Cre recombinase under the control of a Ptf1a
promoter244, and a Slug-YFP reporter system245. Slug-YFP mice were generously
provided by Robert A. Weinberg, Whitehead Institute for Biomedical Research,
Cambridge, MA. Primary tumors were minced and digested with collagenase
(STEMCELL Technologies, 07902). After fibroblast removal, adherent pancreatic
cancer cells were expanded and cultured246.
27
Material and Methods
The detailed experimental setup, tumor growth data, and imaging of the primary
tumor pertaining to circulating tumor cell counting in vivo (Fig. 5.1.5C) have already
been published in Ref. No.247.
Primary human pancreatic cell lines were generated from resected excess
pancreatic carcinoma tissue that was subcutaneously implanted into nude mice in
vivo. The PDO-derived cell line MetPO1 and MetPO2 originates from resected
excess liver metastasis grown into organoids in vitro. The organoids for generating
the MetPO1 and MetPO2 cell line were kindly provided by the Department of Internal
Medicine I at Ulm University (AG Kleger). Bo80 cell line was kindly provided by the
Department of Molecular Gastroenterological Oncology at Ruhr University Bochum
(AG Hahn). Gö5, Gö7 and Gö13 cell lines were kindly provided by the Department
of Gastroenterology (AG Heßmann) at Göttingen University. Human pancreatic
stellate cell line was kindly provided by Department of Internal Medicine I at Ulm
University (AG Seufferlein).
28
Material and Methods
PDX/O derived cell lines were cultured in 6-well plates at a density of 1 x 105 cells
per well. PSCs were cultured in inserts with 1μm pore size PET membranes at a
density of 1 x 105 cells per insert. Cells were kept separately at 37°C in a humidified
atmosphere with 5% CO2 in the indicated culture media respectively (section 4.1)
for 24 hours. Subsequently, media for both 6-well plate and inserts were replaced
to RPMI (with 10% FBS, 1% Pen-Strep and 1% Glutamine) and inserts with PSCs
were placed over PDX/O derived cell lines on 6-well plate and incubated additionally
for 72 hours to establish dual cultures. For control PDX/O derived cells were cultured
at the density of 1 x 105 cells per insert.
29
Material and Methods
PSCs were thawed and cultured in a T75 flask in DMEM : F12 (1:1) together with
20% FBS, 1% Pen-Strep and 1% Glutamine for 7 days at 37°C in a humidified
atmosphere with 5% CO2. Media was collected in a 50ml tube and centrifuged at
1000 rpm for 5 minutes to remove debris. Media was next filter-sterilized (0.45μm
filter), labelled and stored as PSCs conditioned media (PSCs - CM) at 4°C. 10% of
PSCs – CM is used every time for PSCs media preparation.
PDX/O derived cell lines and PSCs were cultured separately in 10cm dishes with
their respective media (section 4.1) for 24 hours at 37°C in a humidified atmosphere
with 5% CO2. Subsequently, media from PDX/O derived cell lines was collected in
a 50ml tube and centrifuged at 1000 rpm for 5 minutes to remove debris. Media was
next filter-sterilized (0.45μm filter) and was labelled as PDX/O – CM. PSCs media
was replaced with this PDX/O – CM and was allowed to grow for additional 24 hours
to prime the PSCs. After 24 hours media from primed PSCs was collected in a 50ml
tube and centrifuged at 1000 rpm for 5 minutes to remove debris. Media was next
filter-sterilized (0.45μm filter) and was labelled as primed PSCs – CM. Non-primed
PSCs conditioned media was prepared by removing the PDX/O – CM based priming
step and adding fresh RPMI (with 10% FBS, 1% Pen-Strep and 1% Glutamine)
media to PSCs instead.
5,000 single murine primary adherent cells in 25μl medium249 were mixed with equal
amounts of Matrigel GFR (growth factor reduced, Corning) per well and cultured at
37°C in a humidified atmosphere with 5% CO2. Treatment with MEK inhibitors was
performed on day 1 (for effect on organoid formation) or on day 4 (for effect on
established organoids) for 3 consecutive days. Medium was changed daily.
Metabolically active cells were measured with the Cell Titer-Glo Luminescent Cell
Viability Assay (Promega, G9681) according to the manufacturer’s instructions.
30
Material and Methods
4.7 Treatments
PD0325901 was used at 0.5μM (5493 cells) or 5μM (8926 and 9228 cells), and
trametinib was used at 0.035μM (5493) or 0.175μM (8926 and 9228 cells) unless
stated otherwise. TGFβ, CXL12 and OSM was used at 10nM. Cell lines are pre-
treated for 30 minutes with EPI-X4, WSCO2, JM#21, inactive peptide before
experimental procedures unless otherwise stated. Concentrations used for EPI-X4,
WSCO2, JM#21, inactive peptide, paclitaxel and gemcitabine are mentioned in the
section 5.Results.
1,000 cells were seeded in a 96-well plate and incubated for 24 hours at 37°C. After
treatment (as described in section 5.Results), cells were incubated for 3 hours with
5 mg/ml MTT (Merck, M2128). DMSO (Roth, A994) was added, and the optical
density was measured using an Infinite 200 PRO plate reader (Tecan, Switzerland)
at 560 nm.
10,000 cells per milliliter were seeded in a 24-well plate. After the treatment (as
described in section 5.Results), the media was removed, and the wells were washed
three times with PBS. After washing, 1 ml of diluted Giemsa solution (Merck,
1.09204.0100) (1: 20 in PBS) was added to each well and incubated for 20 min on
a shaker with 150 rpm. Later, Giemsa solution was removed, and the wells were
rinsed with water slowly. Dried plates were then used for imaging. For quantification,
10% acetic acid was added to Giemsa-stained cells and incubated for 20 min on a
shaker with 150 rpm for blue color to develop. 100μl of this solution was then
transferred into a 96-well plate, absorbance was measured using Infinite 200 PRO
plate reader (Tecan, Switzerland) at 590 nm.
31
Material and Methods
supplemented with B-27 (Thermo Fisher, 17504044) and basic fibroblast growth
factor (Novoprotein, CO46). After 7 days of incubation, > 40μm and < 120μm were
quantified using CASY TT (OMNI Life Science, 5651697). For second-generation
spheres, after counting the first-generation spheres, they were disrupted using
trypsin for 15 minutes at 37°C to make single cells. Equal number of cells per
milliliter were seeded back in ultralow attachment plates. After 7 days of
incubation, > 40μm and < 120μm were quantified using CASY TT.
Cells were grown to confluency and then serum-starved for 24 hours before scratch
wounds were made using a sterile 10μl pipette tip. The cells were then cultured with
medium containing vehicle or PD0325901 for 24 hours. Images were captured after
24 hours and quantified using ImageJ (version 1.49, https://imagej.nih.gov/ij/).
Migration assays were performed using inserts with 8μm pore size PET membranes
(Corning, 353097). After 24 hours, invaded cells were fixed with 4% PFA and
stained with DAPI (Merck, 10236276001). Ten random 10x fields were chosen and
photographed (Keyence), and the pictures were quantified using ImageJ.
Cells in serum-free medium were added to the inserts. In the bottom well of the
companion plate, media containing 10% FBS was added.
32
Material and Methods
Lenti-X (293T) cells were seeded at 5 x 106 cells per 10cm dish, cultured for 24
hours were transfected with 8µg plasmids of interest (shBMI1; shCXCR4; shSCR),
5.5µg psPAX2 (lentiviral packaging), 2µg pMD2 (lentiviral envelope) and 8.75% PEI
transfection reagent (50 mg PEI, 20 mM HEPES and 150mM NaCl; for DNA charge
opposition) in serum-free DMEM. After 4 hours at 37 °C, media was replaced with
DMEM supplemented with 10% FBS, 1% Pen-strep and 1% Glutamine. Virus was
harvested after 2 and 4 days after transfection, pooled and centrifuged at 1500 rpm,
33
Material and Methods
room temperature for 2 min to pellet remaining cells. Supernatants were sterile
filtered (0.45µm), mixed with Lenti-X Concentrator (3:1) (Clontech, 631231), and
incubated at 4°C for 30 min prior to 45 min centrifugation at 1500 rpm and 4°C for
virus precipitation. Virus pellets were resuspended in DMEM.
Human PDAC cell lines were seeded at a density of 1 x 106 cells per 2ml in 6 well
plates. They were then infected with 0 - 25µl virus polybrene (10µg/ml enhances
retroviral infection) containing RPMI after 24 hours, as well as 48 hours of culture.
Cell culture media was replaced 24 hours after the second infection and the cells
were allowed to recover for 48 hours. For drug selection, media was substituted with
puromycin (3mg/ml) supplemented RPMI every three days, until non-transduced
cells (0µl virus) in control wells died. Consequently, only successfully transduced
cells retaining the puromycin resistance gene survived the treatment. Upon
confluency, samples of each transduction were collected for qRT-PCR analysis to
assess the minimal virus concentration with the strongest downregulation. Cells with
the optimal virus concentration were expanded for conducting experiments.
Total RNA was prepared using the RNeasy kit with on-column genomic DNA
digestion following the manufacturer’s instructions (Qiagen, 74106). First-strand
cDNA was prepared using the QuantiTect Reverse Transcription Kit (Qiagen,
205314).
Reactions were performed with the PerfeCTa SYBR Green FastMix PCR Reagent
(Qiagen, 204057) using a QuantStudioTM 3 machine (Applied Biosystems) using
primers (Table 6 and 7). Results were analyzed using the 2-ddCt method relative
to Ywhaz expression (for murine primary cell lines) and HPRT or GAPDH (for
human primary cell lines). Reactions were carried out from at least three
independent experiments.
34
Material and Methods
35
Material and Methods
Harvested cells were lysed in ice-cold RIPA buffer (Cell Signaling, 9806S)
supplemented with PhosSTOp™ (Merck, 4906845001) and a protease inhibitor
cocktail (Merck, 11836170001).
36
Material and Methods
For each sample, equal amounts of protein were applied to a 10% SDS-
polyacrylamide gel and immunoblotted onto PVDF membranes (GE Healthcare,
10600021). Membranes were blocked for 2 hours in 5% BSA in 1x TBST, probed
with the primary antibodies (Table 8) overnight at 4°C, washed with 1x TBST, and
incubated with indicated secondary antibody (Table 8) for 2 hours. The
chemiluminescence detection was performed according to the manufacturer’s
instructions (Merck, WBKLS0500). Stripping of blots was performed when required
with RestoreTM Western Blot Stripping Buffer according to the manufacturer’s
instructions (Thermo Scientific, 21059). Blots were re-blocked after stripping
overnight at 4°C in 5% BSA in 1x TBST before probing with the primary antibody.
37
Material and Methods
4.17 Immunofluorescence
4.17.1 E-cadherin
38
Material and Methods
4.17.3 Phalloidin
PDX/O cell lines were cultured at a density of 50,000 on coverslips in a 6-well plate.
After 24 hours or treatment (mentioned in section 5.Results), cells were washed
three times with PBS. Subsequently, they were fixed with 2% PFA for 20 min at
room temperature and then washed again three times with PBS. Next cells were
permeabilized with 0.7% TritonX-100 solution for 15 min at room temperature
followed by three PBS washing. Coverslips with cells were then incubated with
Phalloidin-Atto (1:500 diluted in PBS) inside a dark humid chamber for 60 min. Cells
were washed three times in PBS and mounted on slides using Prolong™ Gold
reagent with DAPI. Fluorescence at 565 nm was visualized and photographed Zeiss
Axio Vert.A1(Zen Blue).
Flow cytometry analyses were performed using LSR II (BD). Dead cells were
excluded using DAPI.
Annexin V staining was performed using a BD Annexin V APC kit according to the
manufacturer’s instructions (BD, 550474). Data were analyzed using FlowJo v10
(Ashland, OR).
For the identification and quantification of circulating tumor cells in the blood of mice,
counting beads (Thermo Fisher Scientific, C36950) were added to the whole blood
aspirated from the right ventricle. After red blood cell lysis, samples were stained
with an EpCAM-APC antibody (Thermo Fisher) or an appropriate isotype control
(BD). The number of cells and beads in the final sample was recorded, and the total
quantity of cells in the original sample was calculated.
For surface protein staining (CD133; CXCR4), 1 x 106 cells were resuspended in
100µl ice-cold FACS buffer supplemented with 6 µl Gamunex for 15 min at 4°C.
39
Material and Methods
Cells were then incubated with specific antibodies or an appropriate isotype control
(Table 9) for 30mins at 4°C. After PBS wash, cells were resuspended in 500µl of
ice-cold FACS buffer with DAPI in FACS tubes for analysis.
Media levels of secreted CXCL12 were determined using CXCL12A Human ELISA
kit (Thermo Fischer Scientific, EHCXCL12A) according to the manufacturer
guidelines. Conditioned media from PSCs monoculture, PDX/O – PDX/O and
PDX/O – PSCs co-cultures was collected after 72 hours and stored at -80°C.
Thawed conditioned media samples were diluted 2-fold with assay diluent B.
Absorbance was measured using Infinite 200 PRO plate reader (Tecan,
Switzerland) at 450 nm.
50,000 SupT1 cells/well were seeded in 96-well V-bottom microtiter plates in FACS
buffer. Buffer was removed by centrifugation and cells precooled at 4°C for 15 min.
In the meantime, compounds were serially diluted in ice cold PBS and 12G5-APC
antibody was diluted in cold FACS buffer at a concentration of 0.49nM. Afterwards,
15µl compound directly followed by 15µl antibody was added to the cells. For
stability experiments, the mixture was carefully mixed after addition to the cells.
Cells were incubated at 4°C for 2 hours before unbound antibody and compounds
were removed by 2 washing steps followed by fixation in 2% PFA buffer. Cells were
analyzed for MFI values of bound antibody by flow cytometry using FACS
CytoFLEX. For calculation, the isotype control (Table 9) was subtracted, and values
40
Material and Methods
JM#21 peptide was 150-fold diluted in media supplemented with PBS, 10% FCS or
to reach final concentrations of 20µM. The t = 0 sample was immediately taken and
stored at -80°C. Media/peptide mixture was then transferred to 37°C and shook at
350 rpm. At given time points samples were taken and stored at -80°C. For
measuring the functional activity of the media/peptide samples, the mixtures were
thawed and serially diluted in ice cold PBS (starting with 100 % sample). 12G5-APC
antibody competition was then performed as described before.
JM#21 conjugation to palmitic acid (JM#143 and JM#194) has been previously
described in the published thesis Harms M: Endogenous CXCR4 Antagonists: Role
in HIV-1 Transmission and Therapy of CXCR4-linked diseases239, PhD Dissertation,
Institute of Molecular Virology, University of Ulm, 2021.
The mesoporous silica nanoparticles with a radially (MSN) and dendritic (DMSN)
254
oriented pore system were synthesized similar to as mentioned in Ref. No. and
255
Ref. No. respectively. In brief, cetyltrimethylammonium bromide (CTAB) was
used as a structure directing agent and a mixture of tetramethyl orthosilicate
(TMOS) and (3-aminopropyl) trimethoxysilane (APTMS) acted as silica precursors.
The final synthesis mixture had a molar ratio of 0.68 TMOS: 0.1 APTMS: 1.00 CTAB:
0.21 NaOH: 1746 MeOH: 4142 H2O. Instead of extraction with acidic ethanol, the
surfactant was removed via calcination at 550°C for 5.5 hours. JM#21 was adsorbed
onto the radial mesoporous silica nanoparticles (MSN) and dendritic mesoporous
255
silica nanoparticles (DMSN) similar to as described previously in Ref. No. .
41
Material and Methods
4.25 Software
4.26 Statistics
Results for continuous variables are presented as mean SEM unless stated
otherwise or as median with quartiles and min/max values for box plots.
Statistical analysis was performed using the GraphPad Prism software. In order
to detect normality Shapiro-wilk test was performed. Normally distributed data
was analyzed using two-way ANOVA (or one-way ANOVA for knockdown
experiments) otherwise Mann-Whitney U test was applied. Fold change data
was analyzed using student’s t test. *p values < 0.05 were considered
statistically significant. For each experiment, the sample size is indicated in the
figure legend.
42
Results
5 Results
5.1 MEK signaling contributes to maintenance of CSCs phenotype
and promotes migration of pancreatic cancer cells
Figure 5.1.1. MEK inhibition reduces the growth of murine PDAC cells: (A) MTT assays to
43
Results
determine the respective IC50 for three mouse tumor-derived primary cell lines (5493, 8926 and
9228) treated with PD0325901 (PD). The respective IC50 is depicted for each cell line. (B & C)
Apoptosis induction under MEK inhibitor treatment (3 days) as measured by Annexin V staining and
analyzed by flow cytometry in (B) 5493 and (C) 8926 cell lines respectively. (D) Western blot analysis
for phosphorylated and total ERK 1/2 was performed on the three cell lines treated with PD0325901
at the indicated concentrations. GAPDH was used as a loading control. n ≥ 3 for all experiments, ∗
p < 0.05 vs. control. ns = not significant. (Figures and figure legend are licensed under a Creative
Commons Attribution License (CC BY 4.0) https://creativecommons.org/licenses/by/4.0/ ©2019,
Walter et al. 2019 Ref. No. 1).
Sphere cultures enrich for cancer stem cells (CSCs)258. We therefore performed
sphere formation assays after 72 hours of pretreatment with PD0325901 to
elucidate the functional effects of MEK inhibition on CSCs. MEK inhibition
significantly reduced the number of spheres formed after treatment (Fig. 5.1.2A).
Additionally, the size of the spheres formed was notably smaller compared to the
vehicle-treated control (Fig. 5.1.2B). We next investigated the expression of genes
associated with pluripotency and stemness. Indeed, we observed a significant
downregulation in Sox9, Sox2, CD44, and (only 5493 cell line) Sca1 in adherent cell
cultures after MEK inhibition (Fig. 5.1.2C). In 3D sphere cultures, we observed
comparable, albeit slightly less pronounced effects of PD0325901 on stemness-
associated genes as in adherent cultures (Fig. 5.1.2D). Furthermore, treatment with
trametinib, a clinical-grade MEK inhibitor, also showed a significant down regulation
of stemness-associated genes (Figure 5.1.2E) in adherent cells. In order to
generalize our approach to a larger set of primary cell lines, we performed RNAseq
on 4 additional KPC-derived primary mouse cell lines treated with trametinib or
vehicle. The subsequent analysis revealed downregulation of Nanog, Sox9, and
Klf4 (Fig. 5.1.2 F), matching the qRT-PCR dataset.
44
Results
Figure 5.1.2. Effects of MEK inhibition on stemness: (A) Sphere formation capacity of cells
pretreated with PD0325901 at the indicated concentrations. (B) Quantification of large spheres
(>120μm) and representative micrographs of sphere cultures after 7 days. Gene expression of the
indicated stemness-associated genes in (C) adherent cultures or (D) spheres after treatment with
vehicle or PD0325901 (PD). (E) Gene expression in adherent cultures and (F) RNA seq analysis of
murine primary cancer cell lines treated with DMSO vehicle (white) or trametinib (grey) for 48h. n ≥
3 for all experiments. ∗ p < 0.05 vs. control. (Figures and figure legend are licensed under a Creative
Commons Attribution License (CC BY 4.0) https://creativecommons.org/licenses/by/4.0/ ©2019,
Walter et al. 2019 Ref. No. 1).
45
Results
46
Results
Figure 5.1.3. Effect of MEK inhibition on Migration: (A) The percentage of wound closure 24h
after scratch wound induction in primary cells treated with vehicle or PD0325901 at the indicated
concentrations. (B) Representative micrographs of wound closure at 0 hours and 24 hours in the cell
line 5493. (C) Quantification and (D) representative micrographs (10x, DAPI nuclear staining) of
Transwell migration assays with vehicle or PD0325901 treated cells in the indicated concentrations.
n ≥ 3 for all experiments. ∗ p < 0.05 vs. control. (Figures and figure legend are licensed under a
Creative Commons Attribution License (CC BY 4.0) https://creativecommons.org/licenses/by/4.0/
©2019, Walter et al. 2019 Ref. No. 1).
The treatment effects were then measured by western blotting of Slug and Vimentin
as key signaling molecules of EMT activation. Both proteins were strongly
upregulated following TGFβ treatment. As hypothesized, Slug protein levels were
reduced by subsequent MEK inhibition (Figure 5.1.4B). Interestingly, MEK inhibition
was unable to overcome the effects of continuous TGFβ stimulation (evident via
both Slug and E-cadherin protein levels). Furthermore, we utilized the endogenous
Slug-YFP reporter system contained in the mouse model in our cells: TGFβ
treatment resulted in robust Slug-YFP expression, as shown by the increase in
RFP+YFP+ cells after 3 days and 6 days (Figure 5.1.4C, D). Interestingly, MEK
However, matching the observation in western blotting, MEK inhibition was not able
to abrogate the effects of continuous TGFβ treatment. In line with these
experiments, immunofluorescence staining revealed that while E-cadherin is
suppressed by TGFβ treatment, PD0325901 treatment induces re-expression of E-
cadherin, indicating the induction of a more epithelial and thus more differentiated
phenotype (Figure 5.1.4E). Altogether, the results indicate that pharmacological
MEK inhibition using PD0325901 inhibits TGFβ-induced EMT and migration in vitro.
47
Results
Figure 5.1.4. Effects of MEK inhibition on TGFβ-induced EMT: (A) Experimental overview for
TGFβ and PD0325901 co–treatment. (B) Western blot analysis of key proteins involved in EMT with
treatment in vitro. Gapdh was used as a loading control. (C) Quantification of RFP+YFP+ cells under
treatment as indicated and (D) representative cytometry blots. (E) Immunofluorescence micrographs
of E-cadherin expression with treatment as indicated. n ≥ 3 for all experiments, n ≥ 2 for Western
blots. ∗ P < 0.05 vs. control, # P < 0.05 vs. TGFβ. (Figures and figure legend are licensed under a
48
Results
5.1.5 MEK Inhibitors Prevent Organoid Formation and Decrease CTCs in vivo
49
Results
Figure 5.1.5. MEK inhibitors diminish organoid formation and eliminate CTCs in vivo: (A)
Representative micrograph of RFP+ organoid cultures (left, 5493 cells). Organoid formation under
treatment with PD0325901 (PD) (middle) and trametinib (Tra) (right) in 3 primary cell lines treated as
indicated. (B) Viability of preformed organoids with PD0325901 treatment in vitro. (C) Absolute
numbers of circulating tumor cells (CTCs) in KPC mice treated with either vehicle control or
refametinib (as described in Ref. No. 247). n≥3 for all experiments. ∗ p < 0.05 vs. control. (Figures and
figure legend are licensed under a Creative Commons Attribution License (CC BY 4.0)
https://creativecommons.org/licenses/by/4.0/ ©2019, Walter et al. 2019 Ref. No. 1).
So far we described that MAPK signaling pathway, acting through MEK, confers
invasive properties to pancreatic cancer cells via transcription factor Slug. However,
MEK inhibition could not overcome the effects of sustained TGFβ-indued EMT
activation. These results further shed light on the fact that single agent therapies,
even if targetted, may lead to resistance. Moreover, different cell types in the tumor
microenvironment (TME) release factors such as cytokines (e.g., TGFβ, SHH) and
chemokines which can interact with CSCs thereby promoting chemo-resistance and
metastasis. One such chemokine secreted in the TME is CXCL12 (or SDF1) which
is a ligand for the chemokine receptor CXCR4. As a next step we tried to delineate
the role of the CXCL12 – CXCR4 axis in the maintenance of migrating CSCs
(miCSCs).
50
Results
(Fig. 5.2.1C), and high PROM1 expression correlates with low overall survival of
patients (Fig. 5.2.1D).
Figure 5.2.1. CD133 and CXCR4 are highly expressed in PDAC: (A) Box plots represent relative
mRNA expression for the gene signatures PROM1, CXCR4 or PROM1 and CXCR4 in normal tissue
compared to PDAC tumor tissue (TCGA database, GEPIA). (B) Protein – protein interactions of
PROM1 (CD133) and CXCR4 with relevant factors involved in metastasis (green) and CSCs (red)
(STRING). (C) Kaplan – Meier curves showing disease-free survival for patients with high compared
to low relative mRNA expression of PROM1, CXCR4, or PROM1 and CXCR4 in PDAC (TCGA
database, GEPIA). (D) Kaplan – Meier curves showing overall survival for patients with high
compared to low relative mRNA expression of PROM1, CXCR4, or PROM1 and CXCR4 in PDAC
(TCGA database, GEPIA). The threshold for splitting high and low expression cohorts was set at the
median for box plots, *p < 0.05.
51
Results
Secreted factors from these activated PSCs play a crucial role in the development
of cellular heterogeneity in PDACs104. In order to evaluate the relative contribution
of factors secreted by stellate cells to CSC and miCSC maintenance, we
performed flow cytometry analysis for CD133 and CXCR4 in Panc354 (PDX cell
line) and MetPO1 (PDO cell line) that were exposed to different media conditioned
by PSCs (conditioned medium, CM) at three time points as depicted in (Fig.
5.2.2C). Interestingly, we observed a shift towards higher levels of CD133+,
CXCR4+ and CD133+CXCR4+ cells (Fig. 5.2.2D, E, F) in the presence of CM
from primed PSCs (pink) when compared to CM from non-primed PSCs (blue) or
no CM (gray) in all the time points tested. Since media from PDX/O cell lines was
used to prime PSCs, these results suggest that although secreted factors from
PSCs alone upregulate CD133 and CXCR4, tumor cell-primed PSCs either
secrete more or specific factors that can increase CSCs (CD133+) and miCSCs
(CD133+ CXCR4+) (Fig. 5.2.2G).
52
Results
Figure 5.2.2. PSCs secreted factors upregulate CD133 and CXCR4 surface expression: (A)
Screening for percent CD133+ cells and (B) CD133+ CXCR4 + found in adherent cell culture of
indicated cell lines. (C) Experimental scheme to evaluate CD133 and CXCR4 surface expression to
identify patient derived xenografts (PDX) or patient derived organoids (PDO) crosstalk with
pancreatic stellate cells (PSCs) at the protein level. (D) FACS analysis performed on Panc354 and
MetPO1 when exposed to no conditioned media (grey), conditioned media from non - primed PSCs
(blue) or conditioned media from primed PSCs (pink) for CD133+ cells, (E) CXCR4+ cells and (F)
CD133+ CXCR4+ cells represented as fold change against no conditioned media. (G)
Representative cytometry blots. Error bars represent the standard deviation. n≥3 for all experiments.
*p < 0.05, ns = not significant.
53
Results
5.2.3 Tumor cell – stellate cell crosstalk upregulates CSCs and miCSCs
population, and potentiates CXCL12 release
In order to delineate the extent of tumor cell – stellate cell crosstalk in maintaining
the CSC and miCSC subpopulations and their features, we used three PDX cell
lines (Panc354, Gö13 and Bo80) and one PDO cell line (MetPO1) for the
subsequent experiments. PDX/O cell lines were co-cultured with pancreatic stellate
cells (or PDX/O cell lines as control) for 72 hours. Then cells were harvested to
perform further analysis as depicted in (Fig. 5.2.3A). In line with our previous data,
flow cytometry analysis for CD133 showed a significantly increased CSC (CD133+)
population in Panc354 and MetPO1 cell lines (co-cultured with PSCs (pink)
compared to respective control (grey)) (Fig. 5.2.3B, D). Furthermore, flow cytometry
analysis for CD133 and CXCR4 also revealed a significant increase in miCSCs
(CD133+ CXCR4+) subpopulation in Panc354, MetPO1 and Gö13 cells (co-cultured
with PSCs compared to respective control) (Fig. 5.2.3C, D).
Gene expression analysis from PDX/O derived cell lines co-cultured with PSCs
(compared to control) (Fig. 5.2.3E) revealed significant upregulation of stemness-
associated genes (NANOG, POU5F1, ALDH1a1, BMI1). We also found DCK1, a
gene catalyzing the rate-limiting step in gemcitabine metabolism261 to be
downregulated, whereas genes involved in the metabolism of reactive oxygen
species (ROS), such as SOD1 and (only for Panc354 - PSCs) GPX1 were
upregulated. Moreover, ABCC5, which is involved in the transport of nucleotide
analogues, and which has been speculated to perform excessive efflux of nucleotide
analogue-based drugs such as 5-FU or gemcitabine262, was found to be
downregulated in all cell lines tested. Additionally, the epithelial marker CDH1 was
found to be significantly downregulated, while EMT markers (VIM, SNAI1, SNAI2)
were upregulated. This further confirms stark changes in the transcriptional profile
of PDX/O cell lines during co-culture with PSCs (compared to control). (Fig. 5.2.3E).
54
Results
derived cell lines co-cultured with PSCs showed tremendously increased migratory
capacity as well as sphere forming propensity.
To confirm activation of PSCs when co-cultured with PDX/O cell lines, we performed
immunofluorescence (IF) for lipid droplets (Fig. 5.2.3K) using Nile Red staining (red
dots) (Fig. 5.2.3L). IF revealed a loss of lipid droplets (marked by yellow
arrowheads) in PSCs co-cultured with PDX/O when compared to PSC monocultures
demonstrating an activation of PSCs. Altogether this confirms that tumor cell –
stellate cell crosstalk promotes self-renewal capacity and invasiveness of tumor
cells by promoting CSCs and miCSCs. Furthermore, the crosstalk promotes
CXCL12 release from stellate cells.
55
Results
Figure 5.2.3. Tumor cell – stellate cell crosstalk upregulates CSC and miCSC population and
potentiates CXCL12 release: (A) Experimental scheme to evaluate CD133 and CXCR4 surface
expression, qRT-PCR, migration assay, sphere assay, ELISA and IF for co-culture PDX/O – PSCs
and PDX/O – PDX/O. (B) Flow cytometry analysis performed for percent CD133+ cells and (C)
CD133+ CXCR4+ cells for depicted cell lines. (D) Representative cytometry plots. (E) Gene
expression analysis for indicated genes using qRT-PCR for total RNA extracted from PDX/O cell
lines (indicated) co-cultured with PSCs or PDX/O cell lines represented as log2 fold change. (F)
Transwell migration assays for PDX/O cell lines (indicated) cultivated in dual culture with PSCs or
PDX/O cell lines and (G) representative micrographs (10x, DAPI nuclear staining). (H) Sphere
formation assays for PDX/O cell lines (indicated) cultivated in dual culture with PSCs or PDX/O cell
lines and (I) representative micrographs of sphere cultures after 7 days. (J) ELISA performed for
CXCL12 concentration (pg/ml) in media from cultures as indicated. (K) Quantifications and (L)
representative micrographs with yellow arrowheads marking lipid droplets stained with Nile Red (red
dots), nucleus with DAPI (blue) and actin filaments with Phalloidin (red). Error bars represent the
standard deviation. n≥3 for all experiments. *p < 0.05, ns = not significant.
56
Results
Our previous result suggests that CXCL12 is important in the activation of CSCs
and miCSCs. In order to confirm that CXCL12 indeed acts through CXCR4 to
maintain or promote CSCs and miCSCs and their functional characteristics, we
employed sh-RNA mediated knockdown of CXCR4 in one PDX cell line (Panc354)
and one PDO cell line (MetPO1). We confirmed the successful knockdown of
CXCR4 on gene (Fig. 5.2.4A), protein (Fig. 5.2.4B) and cell surface (Fig. 5.2.4C)
expression level for both cell lines.
Next, we selected the cell carrying the most efficient knockdown (sh1_CXCR4 and
sh2_CXCR4) and cultured Panc354 and MetPO1 (sh_SCR, sh1_CXCR4 and
sh2_CXCR4) in the presence or absence of CXCL12 for 24 hours or co-cultured the
cells with PSCs for 72 hours. Afterwards the harvested cells were subjected to
further experiments as depicted (Fig. 5.2.4D). Flow cytometry analysis for CD133
showed an increase in CSCs (CD133+) population for CXCL12-treated MetPO1
sh_SCR (but not in Panc354 sh_SCR) compared to sh_SCR control. A further
increase in CSCs was observed in MetPO1 sh_SCR (but not in Panc354 sh_SCR)
co-cultured with PSCs. Interestingly, after CXCR4 knockdown (sh1_CXCR4 and
sh2_CXCR4) in both cell lines, neither CXCL12 treatment nor PSCs co-culture were
able to upregulate CSCs (CD133+) population to sh_SCR levels (Fig. 5.2.4E,F,I).
supporting the critical role of the CXCR4-CXCL12 axis in CSC propagation. In line
with our previous data (Fig. 5.2.3C), an increase in miCSCs (CD133+ CXCR4+)
subpopulation was observed in both CXCL12-treated and PSCs co-cultured
Panc354 sh_SCR and MetPO1 sh_SCR cell lines. As expected, knockdown of
CXCR4 in both Panc354 and MetPO1 (sh1_CXCR4 and sh2_CXCR4) resulted in a
significant decrease in miCSCs (CD133+ CXCR4+), which neither CXCL12
treatment nor PSCs co-culture was able to “rescue”, i.e., upregulate to sh_SCR
levels (Fig. 5.2.4G,H,I).
57
Results
58
Results
knockdown in both cell lines downregulated this increase in expression with (or
without) CXCL12 treatment or with PSC co-culture. Thus, we conclude that CXCL12
can recapitulate most of the downstream effects observed with PSC co-culture,
which were reversed or abrogated by CXCR4 knockdown. Altogether, these results
confirm that the regulatory axis between CXCL12 and CXCR4 maintains CSCs and
miCSCs.
59
Results
Figure 5.2.4. CXCL12 maintains CSC and miCSC population through CXCR4: (A) CXCR4 gene
expression analysis for the depicted cell lines. (B) Western blot analysis of CXCR4. GAPDH was
used as a loading control. (C) Flow cytometry analysis of CXCR4 surface expression in depicted cell
lines. (D) Experimental setup to evaluate CD133 and CXCR4 surface expression and qRT-PCR for
Panc354 and MetPO1 (sh_SCR, sh1_CXCR4 and sh2_CXCR4). (E) Flow cytometry analysis of
CD133+ CSCs in Panc354 (sh_SCR, sh1_CXCR4 and sh2_CXCR4) treated with (or without)
CXCL12 or co-cultured with PSCs. (F) Flow cytometry analysis of CD133+ CSCs in MetPO1
(sh_SCR, sh1_CXCR4 and sh2_CXCR4) treated with (or without) CXCL12 or co-cultured with PSCs.
(G) Flow cytometry analysis of CD133+ CXCR4+ miCSCs in Panc354 (sh_SCR, sh1_CXCR4 and
sh2_CXCR4) treated with (or without) CXCL12 or co-cultured with PSCs. (H) Flow cytometry analysis
of CD133+ CXCR4+ miCSCs in MetPO1 (sh_SCR, sh1_CXCR4 and sh2_CXCR4) treated with (or
without) CXCL12 or co-cultured with PSCs and (I) representative cytometry plots for MetPO1 cell
line. (J) Targeted gene expression analysis for indicated genes in Panc354 (sh_SCR, sh1_CXCR4
and sh2_CXCR4) treated with (or without) CXCL12 or co-culture with PSCs. (K) Targeted gene
expression analysis for indicated genes in MetPO1 (sh_SCR, sh1_CXCR4 and sh2_CXCR4) treated
with (or without) CXCL12 or co-culture with PSCs. Error bars represent the standard deviation. n≥3
for all experiments. *p < 0.05, ns = not significant.
We further wanted to validate our findings regarding the role of CXCL12 in the
maintenance of key functional characteristics of miCSCs, and to delineate
downstream pathways by which CXCL12 – CXCR4 signaling is able to impart these
abilities. In order to do so, we performed sphere formation assays with Panc354 and
MetPO1 (sh_SCR, sh1_CXCR4 and sh2_CXCR4) after 24 hours of CXCL12
treatment (Fig. 5.2.5A). We observed that CXCR4 knockdown indeed significantly
hampered sphere forming capacity of both Panc354 and MetPO1 (sh1_CXCR4 and
sh2_CXCR4) cells compared to sh_SCR (Fig. 5.2.5A). To confirm the effects on
60
Results
We next looked into phosphorylation of AKT and IκB-α protein in Panc354 and
MetPO1 cells (sh_SCR, sh1_CXCR4 and sh2_CXCR4) treated with and without
CXCL12 (Fig. 5.2.5E). A decrease in phosphorylation of AKT was found in both
Panc354 and MetPO1 cells (sh1_CXCR4 and sh2_CXCR4) upon CXCL12
treatment. Similarly, phosphorylation of IκB-α was decreased in MetPO1
(sh1_CXCR4 and sh2_CXCR4) upon CXCL12 treatment (Fig. 5.2.5E). In order to
find the converging point through which CXCL12 – CXCR4 signaling maintains both
self-renewal and metastatic potential, we generated a protein – protein interaction
(STRING) network of CXCL12 – CXCR4 (green) with other known relevant factors
involved in metastasis (red), stemness (purple), sonic hedgehog signaling (blue),
AKT signaling (grey) and NFκB signaling (yellow) (Fig. 5.2.5F). These interactions
suggest that BMI1 (red arrow) could be one of the entities through which CXCL12 –
CXCR4 is acting, providing a mechanistic explanation of the relationship between
EMT and stemness in cancer cells. Taken together, these results demonstrate that
61
Results
Figure 5.2.5. The CXCL12 - CXCR4 axis sustains self-renewal capacity and metastatic
propensity of miCSCs: (A) Sphere formation assay for depicted cell lines after CXCL12 treatment
and representative micrographs of sphere cultures after 7 days. (B) Western blot analysis of NANOG
and BMI1 for depicted cell lines and treatment conditions. GAPDH was used as a loading control.
62
Results
(C) Migration assay towards CXCL12 for depicted cell lines and representative micrographs (10x,
DAPI nuclear staining). (D) Western blot analysis of CADHERIN-1 and VIMENTIN for depicted cell
lines and treatment conditions. GAPDH was used as a loading control. (E) Western blot analysis of
p-AKT, AKT, p- IκB-α and IκB-α for depicted cell lines and treatment conditions. GAPDH was used
as a loading control. (F) Protein – protein interactions of CXCL12 and CXCR4 with relevant factors
involved in metastasis (red), stemness (purple), sonic hedgehog signaling (blue), AKT signaling
(grey) and NFκB pathway (yellow) (STRING). Error bars represent the standard deviation. n≥3 for all
experiments. *p < 0.05, ns = not significant.
BMI1 is part of the PRC1 complex maintaining self-renewal and stemness together
with EZH2, which is a component of PRC2265. In order to delineate the underlying
role of BMI1 in EMT and stemness downstream of CXCL12 – CXCR4 signaling, we
employed sh-RNA mediated knockdown of BMI1 in one PDX cell line (Panc354)
and one PDO cell line (MetPO1). We confirmed knockdown of CXCR4 on gene (Fig.
5.2.6A) and protein (Fig. 5.2.6B) expression level for both cell lines.
In order to understand the effect of BMI1 on EMT and migration capacity, gene
expression analysis on key EMT factors was performed (Fig. 5.2.6F). On the one
hand, BMI1 knockdown in MetPO1 (sh1_BMI1 and sh2_BMI1) showed a relevant
increase in expression of the epithelial marker CDH1, on the other hand it showed
63
Results
64
Results
Figure 5.2.6. BMI1 downstream CXCL12 – CXCR4 regulates EMT and stemness: (A) BMI1 gene
65
Results
expression analysis for the depicted cell lines. (B) Western blot analysis of BMI1. GAPDH was used
as a loading control. (C) Gene expression analysis for indicated cell lines with genes involved in
stemness using qRT-PCR. (D) Western blot analysis of NANOG, c-MYC and SOX9 for indicated cell
lines. GAPDH was used as a loading control. (E) Sphere formation assays for depicted cell lines. (F)
Gene expression analysis for indicated cell lines with genes involved in EMT using qRT-PCR. (G)
Western blot analysis of CADHERIN-1, CADHERIN-2 and SLUG for indicated cell lines. GAPDH was
used as a loading control. (H) Migration assays towards serum containing media for depicted cell
lines. (I) Quantifications and representative micrographs for indicated cell lines with white
arrowheads marking mesenchymal structures of actin filaments stained with Phalloidin (pink) and
nucleus stained with DAPI (blue). (J) Western blot analysis of p-ERK for indicated cell lines. Total
ERK was used as a control. Error bars represent the standard deviation. n≥3 for all experiments. *p
< 0.05, ns = not significant.
5.2.7 CXCL12 – CXCR4 maintains CSCs and miCSCs population via BMI1
BMI1 knockdown exhibited its relevance in some of the most important pathways
involved in EMT and stemness. We further wanted to confirm that signaling via the
CXCL12 - CXCR4 axis critically involves BMI1 to maintain both CSCs and miCSCs.
Therefore, we treated Panc354 and MetPO1 (sh_SCR, sh1_BMI1 and sh2_BMI1)
cells with (or without) CXCL12 for 24 hours. The harvested cells were used to
perform flow cytometry for CD133 and CXCR4 (Fig. 5.2.6A).
In line with our hypothesis, we found a significant decrease in the CSC (CD133+)
population in BMI1 knockdown Panc354 and MetPO1 cells (sh1_BMI1 and
sh2_BMI1) compared to the respective sh_SCR after treatment with CXCL12 (Fig.
5.2.6B, D). Similarly, a significant decrease in the miCSC (CD133+ CXCR4+)
population was also observed in MetPO1 cells with BMI1 knockdown (sh1_BMI1
and sh2_BMI1) compared to the respective sh_SCR cells after CXCL12 treatment
(Fig. 5.2.6C, D). As expected, Panc354 sh_SCR showed a strong increase in
miCSCs (CD133+ CXCR4+) when treated with CXCL12 (compared to control-
treated sh_SCR). Surprisingly, we observed an increase in miCSC (CD133+
CXCR4+) after BMI1 knockdown in Panc354 (sh1_BMI1 and sh2_BMI1). But
CXCL12 treatment in Panc354 (sh1_BMI1 and sh2_BMI1) resulted in a strong
decrease in miCSCs (CD133+ CXCR4+) (compared to CXCL12 treated sh_SCR).
66
Results
Figure 5.2.7. CXCL12 – CXCR4 maintains CSCs and miCSCs population via BMI1: (A)
Experimental scheme to evaluate CD133 and CXCR4 surface expression using flow cytometry for
Panc354 and MetPO1 (sh_SCR, sh1_BMI1 and sh2_BMI1) with (or without) CXCL12. (B) Flow
cytometry analysis of CD133+ cells in Panc354 and MetPO1 (sh_SCR, sh1_BMI1 and sh2_BMI1)
treated with (or without) CXCL12. (C) Flow cytometry analysis of CD133+ CXCR4+ cells in Panc354
and MetPO1 (sh_SCR, sh1_BMI1 and sh2_BMI1) treated with (or without) CXCL12 and (D)
representative cytometry plots for MetPO1 cell line. Error bars represent the standard deviation. n≥3
for all experiments. *p < 0.05, ns = not significant.
67
Results
In order to uncover the potency of EPI-X4 and its derivatives to functionally target
CXCR4 in human pancreatic cancer cells, we utilized 3D transwell migration assays
to determine effects on the CXCR4-mediated migratory and metastatic activity. We
used the CXCR4 ligand CXCL12 as a chemoattractant to test the inhibitory capacity
of four peptides: EPI-X4, the modified EPI-X4 derivates WSCO2, JM#21 and an
inactive variant as control on Panc354 cells (Fig. 5.3.1A, B). Although 1µM and
10µM concentrations of EPI-X4 and WSCO2 significantly downregulated migration
capacity of Panc354 to approximately 50%. In case of JM#21 even lower
concentrations (0.1μM and 0.5μM) were sufficient to strongly abrogate migration.
As expected, the inactive peptide showed no effect on the migration of Panc354
cells towards CXCL12. We further confirmed the effect of JM#21 on migration in
other PDX-derived primary human pancreatic cancer cell lines, namely MetPO1,
Bo80 and Gö13 (Fig. 5.3.1C). Concentrations of 5μM and 10μM JM#21 significantly
downregulated the migratory capacity of all the cell lines. Taken together these
results confirmed that JM#21 is one of the most potent EPI-X4 derivatives regarding
the effect on migration of primary PDAC cells in vitro.
68
Results
The IC50 was calculated for each time point (0, 2, 8 and 24 hours). JM#21 incubated
in 10% FCS was found to be inactivated (by the proteases contained in FCS) over
time, whereas there was no such inactivation observed in PBS or B27 serum
supplement (Fig. 5.3.1D), which is critical for the potential use of these peptides for
extended time periods in vitro. Next, dose-dependent inhibition of antibody binding
by JM#21 was measured. JM#21 was active up to 24 hours in culture media
supplemented with B27 instead of FCS (where more than 50% JM#21activity was
lost within the first 10 hours) (Fig. 5.3.1E). As a consequence of these peptide
stability assays and recognizing the low stability of these peptides in serum-
containing culture conditions234, we used B27 as serum supplement for the next
experiments. JM#21 was used at 10μM concentration for further experiments to
achieve effects with the same drug dose for all cell lines.
69
Results
Figure 5.3.1. JM#21, most potent EPI-X4 derivative to target CXCR4: (A) Migration assays
towards CXCL12 using Panc354 for EPI-X4 and its derivatives at depicted concentrations. Pre-
treatment with EPI-X4, WSCO2, JM#21 and the inactive peptide was applied for 30 minutes. (B)
Representative micrographs (10x, DAPI staining) of transwell migration assays in Panc354 cells for
the indicated conditions and concentrations. (C) Migration assays towards CXCL12 for indicated cell
lines using JM#21 and the inactive peptide at depicted concentrations. (D) Peptide stability assays
for JM#21 in PBS, 10% FCS and B27 to detect activity up to 24 hours. n≥3 for all experiments. *p <
0.05, ns = not significant.
70
Results
71
Results
Figure 5.3.2. JM#21 inhibits CXCL12 induced EMT and stemness: (A) Quantifications of percent
mesenchymal structures after 15 minutes and 6 hours of CXCL12 treatment in Panc354 and (B)
MetPO1 cells. JM#21 pre-treatment was applied for 30 minutes. (C) Representative micrographs for
72
Results
indicated cell lines with white arrowheads marking mesenchymal structures of actin filaments stained
with Phalloidin (pink) and nuclear staining using DAPI (blue). (D) Gene expression analysis for
indicated cell lines with genes involved in EMT and SHH pathway. (E) Gene expression analysis for
indicated cell lines with genes involved in stemness. (F) Western blot analysis of CADHERIN-1,
VIMENTIN, CADHERIN-2, NANOG and BMI1 for indicated cell lines. GAPDH was used as a loading
control. (G) Sphere formation assays for 1st and 2nd generation of sphere formation and (H)
representative micrographs of sphere cultures after 7 days in Panc354 cells after the indicated
treatments. JM#21 and inactive peptide pre-treatment was applied for 30 minutes, respectively. Error
bars represent the standard deviation. n≥3 for all experiments. *p < 0.05, ns = not significant.
In order to elucidate the underlying role of CSCs in the success of the above-
mentioned combination treatment, we used flow cytometry to quantify CD133+
CSCs in Panc354 and MetPO1 cells after day 12 for control, JM#21, gemcitabine
(1μM) and JM#21 + gem (1μM) treatments (Fig. 5.3.3G, H). We found that
73
Results
74
Results
Figure 5.3.3. JM#21 sensitizes PDAC cells towards chemotherapy: (A) Quantification of cell
viability and representative pictures for clonogenic assays after 48 hours of JM#21 treatment for
CXCL12 pretreated Panc354 cell line. (B) Experimental design for combination therapy analyzing
relapse using JM#21, gemcitabine (labelled as G) and paclitaxel (labelled as P). Quantification of
cell viability and representative pictures for clonogenic assays after treatment with JM#21 (10μM),
gemcitabine (Gem) or paclitaxel (Pac) for indicated concentrations as depicted in experimental
design in (C) Panc354, (D) MetPO1, (E) Gö13 and (F) Bo80 cells. (G) Flow cytometry for CD133 in
Panc354 and cells for the indicated treatments shown as fold change and (H) representative
cytometry plots for Panc354 cells. Error bars represent the standard deviation. n≥3 for all
experiments. *p < 0.05, ns = not significant.
75
Results
76
Results
Figure 5.3.4. Stabilization of JM#21 in serum conditions: (A) Sequence and molecular weight of
fatty acid (palmitic) conjugated JM#21, namely JM#143 and JM#194 (B) TEM micrographs of silica
nanoparticles (MSN and DMSN). Migration assays towards CXCL12 in FBS-containing medium with
the indicated compounds, tested in (C) Panc354, (D) MetPO1, (E) Gö13 and (F) Bo80 cells. (G)
Representative micrographs (10x, DAPI staining) of transwell migration assays in MetPO1 cells for
the indicated conditions and concentrations. Error bars represent the standard deviation. n≥3 for all
experiments. *p < 0.05, ns = not significant.
We next wanted to delineate the capacity of serum stable JM#21 to target migrating
cancer stem cells. We have shown previously that co-cultures with PSCs increase
the miCSC subpopulation (Fig. 5.2.3C). We next investigated the effect of fatty acid
conjugated JM#21 (JM#143 and JM#194) and Si-NP encapsulated JM#21 pre-
treatment on PDX/O cell lines in co-culture with PSCs (Fig. 5.3.5A). In order to
characterize the effects, we performed pathway-focused gene expression analysis
for key genes involved in EMT and stemness. This indicated that JM#143 and
JM#194 significantly reduced expression of both stemness- (NANOG, ALDH1a1)
and EMT- (VIM, SNAI2) associated factors in Panc354 and MetPO1 cells.
Additionally, a substantial increase in CDH1 was also observed in both cell lines
after fatty acid conjugated JM#21 treatment (JM#143 and JM#194) (Fig. 5.3.5 B).
Genes involved in imparting chemoresistance exhibited a stark decrease in ABCC5
expression after JM#143 treatment. Furthermore, both JM#143 and JM#194
significantly upregulated the expression of DCK1 compared to vehicle-treated
control in both cell lines tested. Antioxidant enzymes (SOD1, GPX1, GPX2), on the
77
Results
other hand, were found to be downregulated (Fig. 5.3.5 C). Flow cytometry analysis
revealed no changes in CD133+ expression after fatty acid conjugated JM#21
treatment in both Panc354 and MetPO1 cells, indicating no specific effect on the
general CSC population. In contrast, CD133+ CXCR4+ miCSCs and CXCR4+ cells
were practically abrogated (Fig. 5.3.5 D, H), indicating a strong effect on migratory
cells.
78
Results
Figure 5.3.5. Serum stable JM#21 reduces miCSCs in co-cultures with PSCs: (A) Experimental
design for treatment with fatty acid conjugated JM#21 and Si-NP encapsulated JM#21 in PDX/O –
PSCs co-culture. (B) Gene expression analysis for indicated cell lines with genes involved in EMT
and stemness after treatment with DMSO (vehicle control), JM#143 and JM#194. (C) Gene
expression analysis for indicated cell lines with genes involved in chemoresistance and antioxidant
signaling after treatment with DMSO, JM#143 and JM#194. (D) Flow cytometry analysis performed
79
Results
for CD133+, CD133+CXCR4+ and CXCR4+ cells for depicted cell lines after treatment with DMSO,
JM#143 and JM#194. (E) Gene expression analysis for indicated cell lines with genes involved in
EMT and stemness after treatment with DMSN and MSN (Empty & JM#21). (F) Gene expression
analysis for indicated cell lines with genes involved in chemoresistance and antioxidant signaling
after treatment with DMSN and MSN (Empty & JM#21). (G) Flow cytometry analysis performed for
CD133+, CD133+CXCR4+ and CXCR4+ cells for depicted cell lines after treatment with DMSN and
MSN (Empty & JM#21). (H) Representative cytometry plots for MetPO1 cell line after treatment with
DMSO, JM#143 and JM#194 for CD133+, CD133+CXCR4+ and CXCR4+ cells. (I) Representative
cytometry plots for MetPO1 cells after treatment with DMSN and MSN (Empty & JM#21) for CD133+,
CD133+CXCR4+ and CXCR4+ cells. n≥3 for all experiments. *p < 0.05, ns = not significant.
80
Discussion
6 Discussion
Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive malignancy,
characterized by an unusually high metastatic burden already at the time of
diagnosis270. Furthermore, PDAC typically displays a high degree of resistance to
chemotherapy and radiation271. All this has been associated with the presence of
CSCs272. Considering that CSCs are essential for metastatic spread62, they might
be the population that gives rise to CTCs, and ultimately to metastasis. CTCs have
been demonstrated to have both tumor-initiating and EMT traits98. Therefore,
understanding the nuances of the regulatory mechanisms and the prerequisites for
the maintenance and expansion of migrating CSCs (and CTCs) will result in the
discovery and exploration of novel therapeutics, which in turn could result in
significantly improved therapy.
81
Discussion
more MEK-dependent than the general cell population, since unspecific cytotoxicity
was not observed. This is further corroborated by the observation that stemness-
and pluripotency-associated markers are significantly downregulated upon MEK
inhibitor treatment. However, studies in colorectal cancer have shown that MEK
inhibition could lead to aberrant activation of Wnt/β-catenin signalling, which could
further modulate cancer stem cell homeostasis277. Additionally, signals from other
(non-cancer cells) cells within the tumor microenvironment can foster and potentiate
CSCs population278,279. It would therefore be interesting to further elucidate the
aspect that MEK inhibition has on other signaling pathways or in conjunction with
them in PDAC.
3D organoid cultures are a more physiological cell culture model than 2D monolayer
cultures that better reflects in vivo conditions by maintaining cell-to-cell signaling259.
Therefore, the investigation of MEK inhibition on primary organoid cultures is a very
promising way to contemplate treatment efficacy in a “physiological” in vitro
setting. We demonstrated that the capacity of primary murine PDAC tumor cells to
form organoids is hindered under MEK inhibition. Taken together with diminished
sphere forming capacity this suggests a decrease in their tumor-initiating potential.
We also observed similar results with the treatment performed in pre-formed
organoids, additionally validating our in vitro dataset.
The MAPK pathway has also been reported to promote the expression of EMT-
related transcription factors, in particular that of the Snail superfamily members
during development280,281, fibrosis282, and cancer progression and
metastasis283. The decrease in wound healing capacity of murine PDAC cells after
MEK inhibition was therefore in line with the current understanding of the role of the
MAPK pathway. Although we performed these experiments with matching cell
numbers after pretreatment, it still might be difficult to negate the effects of
proliferation on wound healing ability after scratch application. We therefore
performed 3D transwell migration assays towards serum-containing media and
found a strong dose-dependent (PD0325901-mediated) decrease in the migration
of murine PDAC cells. Additionally, MEK inhibitors can decrease tumor formation in
animal models, particularly in pancreatic cancer247. We demonstrated for the first
time that MEK inhibition additionally causes significant reduction in the number of
82
Discussion
CSCs, organoids, and circulating tumor cells (CTCs) in vivo. Although, CTC
numbers do not naturally correlate directly with the metastatic load in patients, they
are indeed a strong indicator of prognosis284. Refametinib treatment significantly
247
reduced the size of primary tumors (previously published in Ref. No. ), the
observed effects on CTC numbers might (at least partially) also be as a result of a
general reduction in tumor size. However, the significant reduction of Slug-
expressing cells in vitro propose a strong inhibition of TGFβ-induced EMT by MEK
inhibition, which ultimately could result in the observed decrease in CTCs. This
offers a possible mechanism by which MEK can exert its function: promoting
survival, migration, stemness, and CTC initiation, and in turn supporting the
relevance of MEK signaling in PDAC.
The primary tumor cell lines we used in this study are derived from a mouse model
which spontaneously develops metastatic PDAC (KPC mice), and was expanded
using an RFP reporter system to report cells of pancreatic origin, and a YFP reporter
system to indicate Slug activity. TGFβ promotes tumor progression in advanced
cancer stages by inducing tumor growth in itself. Most importantly it strongly induces
EMT, resulting in increased invasion and metastasis285 via upregulation of
transcription factors such as the zinc finger proteins Snail and Slug286. We therefore
utilized the RFP – YFP dual reporter system described above and TGFβ treatment
to delineate the role of MEK inhibition on an active EMT program. On the one hand
we found that increased mesenchymal differentiation (i.e., increased Slug activity)
after TGFβ pre-treatment was indeed diminished by MEK inhibition. A continuous
TGFβ treatment on the other hand displayed no effect on MEK inhibition. This
indicated that these two pathways, although capable of cooperating, act through
different downstream effectors or that compensatory feedback loop mechanisms
play a relevant role.
Taken together, these results also imply a possible interference of other pathways
(apart from TGFβ) involving cytokines and chemokines within the tumor
microenvironment which would hinder MEK inhibition efficacy. The TME is a
complex system containing several cell types such as immune cells, stellate cells,
83
Discussion
and stromal fibroblasts as well as extracellular matrix, blood and lymphatic vessels,
all of which contribute to the growth and aggressiveness of a tumor287. The TME
provides necessary growth factors and signaling mediators like cytokines to the
tumor cells288. The extracellular matrix (ECM) acts as a scaffold and demonstrates
complex biochemical interactions inside the defined microenvironment. ECM
components constantly interact with integrin receptors from the cell surface which
forms a two-directional signaling regulated by GFs289. The concurrent response of
integrin–ECM–GF interactions inside the tumor microenvironment promotes the
activation as well as localization of RAS to the inner cell membrane290, promoting
resistance to MEK inhibitors. Studies have also shown that the TME is an important
root of resistance to MAPK pathway inhibitors by secretion of the macrophage-
derived mediator TNFα (Tumor necrosis factor alpha)291. Furthermore, inhibition of
MAPK interferes with the transactivation activity of p300/CREB (cAMP responsive
element binding protein 1)-binding protein (CBP) by HIF-1α and HIF-2α that
undergo oxygen-dependent degradation292. MAPK signaling hence promotes HIF-
1α activation which in turn is known to potentiate CSCs293 and metastasis in
PDAC294. These studies further strengthen the importance of TME components in
providing resistance towards MEK inhibitors.
84
Discussion
CXCL12 (or SDF1) is critical for the homing of a variety of cell lineages during
development, tissue repair and metastasis298. All pancreatic tumor tissues express
CXCR4 and CXCL12 at higher expression levels compared to non-tumor tissue299.
CXCR4 is activated by CXCL12, which is abundantly present in the pancreatic tumor
85
Discussion
PSCs are also known to play a role in PDAC chemoresistance through various
mechanisms, among which inducing hypoxic conditions is highly prevalent301-303.
Extensive fibrosis produced by the PSCs and the ECM results in significant
intratumoral hypoxia and a self-perpetuating hypoxia-fibrosis cycle303. PDX/O –
PSC (vs. PDX/O – PDX/O) co-culture elevated levels of ABCC5 expression, which
results in reduced drug uptake by the tumor cells. This is a somewhat controversial
result, since blocking ABCC5 has been shown to enhance effects of
chemotherapies like gemcitabine, but ABCC5 blocking has also been implicated in
a positive correlation to PDAC patient survival304. DCK1, encoding deoxycytidine
kinase, is downregulated, which further benefits the tumor cells against gemcitabine
as it catalyzes the reaction to convert gemcitabine to its active metabolite261.
Reactive oxygen species (ROS) are generally increased in pancreatic cancer cells
compared to normal cells. ROS play a vital role in the orchestration of the TME:
ROS can facilitate carcinogenesis and cancer progression with mild to moderate
elevation levels, while excessive ROS damage cancer cells dramatically and may
induce cell death305. The first line of defense against ROS-induced damage is the
conversion of superoxide to hydrogen peroxide by the antioxidant enzyme
superoxide dismutase (SOD)306. These hydrogen peroxide molecules are then
86
Discussion
87
Discussion
In head and neck squamous cell carcinoma (HNSCC), TWIST1 and BMI1 mutually
promote EMT by repressing both E-cadherin and p16INK4a322. Utilizing sh-RNA
mediated knockdown of BMI1, we showed that BMI1 and EMT factors are strongly
correlated. BMI1 knockdown causes significant downregulation of SNAI1 and
TWST1 mRNA, as well as N-CADHERIN protein. Functionally, migration assays
revealed a diminished migratory capacity of BMI1 knockdown cells compared to
scrambled controls. During EMT, tumor cells undergo a genetic and morphological
transformation, manifested by cell elongation, migration, and invasion, coordinated
by a reorganization of actin fibers in the cytoskeleton323. Immunofluorescent staining
in MetPO1 cells showed that BMI1 knockdown severely reduces these
morphological changes by reducing mesenchymal structures and actin stress fibers.
In intestinal tumors and HNSCC, BMI1 has been recognized as a cancer stem cell
marker324,325. Additionally, Dimri et al. have shown that BMI1 induces telomerase
activity and immortalizes human mammary epithelial cells326. Telomerase activity
has been shown in our laboratory to be a highly relevant factor in CSC
maintenance327. Although we observed no differences in sphere forming capacity
after BMI1 knockdown, there was a strong downregulation in key stemness-
associated genes (PROM1, ALDH1a1, POU5F1, NANOG, KLF4 and SOX2). BMI1
also had a pronounced effect on the CSC population, decreasing the expression of
88
Discussion
Paranjape et al. describe that BMI1 can modulate Nanog and affect both self-
renewal and EMT in breast cancer329. In recognition of the overlapping role of BMI1
in both CSC features and migration, deciphering the importance of BMI1 in miCSC
maintenance downstream of CXCR4-CXCL12 in such a highly metastatic disease
as PDAC is imperative. BMI1 knockdown led to a reduction of miCSCs in untreated
cell lines, and CXCL12 treatment was incapable of increasing the CSC or miCSC
population in BMI1 knockdown cells. However, the reverse effects of BMI1
knockdown on Panc354 miCSCs and EMT-associated genes could be due to an
aberrant CD133 upregulation after lentiviral transduction. CD133 overexpression
has been linked to SNAI2 upregulation330 and it would (at least partially) explain the
contradicting results in miCSC population, EMT-associated gene expression (VIM
and SNAI2), and the lack of changes in mesenchymal structures in Panc354 BMI1
knockdown cells. Nevertheless, BMI1 can potentiate PI3K/AKT and SHH/GLI1
signaling in a direct manner by induction of Nanog/NF-κB, and in an indirect manner
by hyperactivation of the PI3K/Akt/NF-κΒ axis331-333. In this context, we show
decreased ERK1/2 phosphorylation after BMI1 knockdown. Since we have
previously described the MEK/ERK axis to be highly relevant for both EMT and
stemness, this further underscores the role of BMI1 in maintaining (mi)CSCs.
From a therapeutic standpoint, Yin et al. have shown that BMI1 inhibition sensitizes
PDAC cells towards gemcitabine334. BMI1 is upregulated in PDX/PDO-derived cell
lines in PSC co-culture (with PSCs secreting high levels of CXCL12) or upon
treatment with CXCL12. On the other hand, CXCR4 knockdown cells show strongly
89
Discussion
reduced BMI1 expression, which even CXCL12 or PSCs co-culture was not able to
upregulate. This establishes BMI1 as another possible regulator through which
CXCL12 exerts its effects on chemoresistance. In conclusion, we here demonstrate
that BMI1 is regulated by CXCL12 downstream CXCR4. CXCL12 conceivably
utilizes BMI1 to impart (at least partially) its distinct effects on EMT, stemness and
chemoresistance. BMI1 has diverse roles downstream of CXCR4 – CXCL12
signaling and represents a possible molecular link between EMT and stemness.
90
Discussion
91
Discussion
92
Discussion
93
Summary
7 Summary
MEK is downstream of the oncogenic driver mutation KRAS and has been reported
to show outstanding relevance in PDAC biology. Thus, our first aim was to establish
the role of RAS/MEK/ERK signaling to promote EMT and support stem-like
phenotypes in disseminating cells. Using primary murine cancer cell lines with a
Slug – YFP reporter system and MEK inhibitors, we established the contribution of
MEK signaling on survival, migration and self-renewal capacity in PDAC. In brief,
TGFβ-induced EMT can be abrogated by MEK inhibition, evidenced by strong
decrease of Slug-expressing cells, which translates into a significant reduction in
CTCs in mice.
94
Summary
cells secrete elevated levels of CXCL12, enabling tumor cells to self-renew and to
migrate, and ultimately to metastasize. Mechanistically, CXCL12 acts through
CXCR4 to activate multiple pathways that include sonic hedgehog, AKT and NF-κΒ,
which further promotes NANOG and BMI1 expression. In addition, BMI1 plays the
molecular link between inducing stemness and activating different EMT programs.
We established CXCR4 as a prominent target for therapeutic interventions to reduce
CSCs and abrogate miCSCs.
In conclusion, the data in the present study (i) demonstrate the importance of
RAS/MEK/ERK signaling on CTC initiation, (ii) provide novel molecular insights on
CSC and miCSC maintenance and (iii) explore human endogenous peptides as
innovative therapy for targeting miCSCs and migration / metastasis. In addition,
tumor-educated PSCs were shown to secrete elevated levels of CXCL12, which
promotes EMT, stemness and chemoresistance via CXCR4. CXCL12 was also
revealed to activate BMI1, which we describe as a downstream molecular link
between EMT and stemness.
95
List of references
8 List of references
1 Walter, K. et al. MEK Inhibition Targets Cancer Stem Cells and Impedes
Migration of Pancreatic Cancer Cells In Vitro and In Vivo. Stem Cells Int
2019, 8475389, doi:10.1155/2019/8475389 (2019).
2 Board, P. D. Q. A. T. E. in PDQ Cancer Information Summaries (National
Cancer Institute (US), 2002).
3 Kleeff, J. et al. Pancreatic cancer. Nat Rev Dis Primers 2, 16022,
doi:10.1038/nrdp.2016.22 (2016).
4 Owens, D. K. et al. Screening for Pancreatic Cancer: US Preventive Services
Task Force Reaffirmation Recommendation Statement. Jama 322, 438-444,
doi:10.1001/jama.2019.10232 (2019).
5 Luo, W., Tao, J., Zheng, L. & Zhang, T. Current epidemiology of pancreatic
cancer: Challenges and opportunities. Chin J Cancer Res 32, 705-719,
doi:10.21147/j.issn.1000-9604.2020.06.04 (2020).
6 Rahib, L. et al. Projecting cancer incidence and deaths to 2030: the
unexpected burden of thyroid, liver, and pancreas cancers in the United
States. Cancer Res 74, 2913-2921, doi:10.1158/0008-5472.Can-14-0155
(2014).
7 Hu, J. X. et al. Pancreatic cancer: A review of epidemiology, trend, and risk
factors. World J Gastroenterol 27, 4298-4321, doi:10.3748/wjg.v27.i27.4298
(2021).
8 Aier, I., Semwal, R., Sharma, A. & Varadwaj, P. K. A systematic assessment
of statistics, risk factors, and underlying features involved in pancreatic
cancer. Cancer Epidemiol 58, 104-110, doi:10.1016/j.canep.2018.12.001
(2019).
9 Sperti, C., Pasquali, C., Piccoli, A. & Pedrazzoli, S. Recurrence after
resection for ductal adenocarcinoma of the pancreas. World J Surg 21, 195-
200, doi:10.1007/s002689900215 (1997).
10 Thomas, S. K., Lee, J. & Beatty, G. L. Paracrine and cell autonomous
signalling in pancreatic cancer progression and metastasis. EBioMedicine
53, 102662, doi:10.1016/j.ebiom.2020.102662 (2020).
11 Siegel, R. L., Miller, K. D. & Jemal, A. Cancer statistics, 2019. CA Cancer J
Clin 69, 7-34, doi:10.3322/caac.21551 (2019).
96
List of references
97
List of references
98
List of references
99
List of references
100
List of references
101
List of references
69 Alcalá, S., Martinelli, P., Hermann, P. C., Heeschen, C. & Sainz, B., Jr. The
Anthrax Toxin Receptor 1 (ANTXR1) Is Enriched in Pancreatic Cancer Stem
Cells Derived from Primary Tumor Cultures. Stem Cells Int 2019, 1378639,
doi:10.1155/2019/1378639 (2019).
70 Boivin, D. et al. The stem cell marker CD133 (prominin-1) is phosphorylated
on cytoplasmic tyrosine-828 and tyrosine-852 by Src and Fyn tyrosine
kinases. Biochemistry 48, 3998-4007, doi:10.1021/bi900159d (2009).
71 Taïeb, N. et al. The first extracellular domain of the tumour stem cell marker
CD133 contains an antigenic ganglioside-binding motif. Cancer Lett 278,
164-173, doi:10.1016/j.canlet.2009.01.013 (2009).
72 Roper, K., Corbeil, D. & Huttner, W. B. Retention of prominin in microvilli
reveals distinct cholesterol-based lipid micro-domains in the apical plasma
membrane. Nat Cell Biol 2, 582-592, doi:10.1038/35023524 (2000).
73 Giebel, B. et al. Segregation of lipid raft markers including CD133 in polarized
human hematopoietic stem and progenitor cells. Blood 104, 2332-2338,
doi:10.1182/blood-2004-02-0511 (2004).
74 Ohnishi, S. et al. hypoxia-inducible factors activate CD133 promoter through
ETS family transcription factors. PLoS One 8, e66255,
doi:10.1371/journal.pone.0066255 (2013).
75 Jiang, X. et al. Inactivating mutations of RNF43 confer Wnt dependency in
pancreatic ductal adenocarcinoma. Proc. Natl. Acad. Sci. U. S. A. 110,
12649-12654, doi:10.1073/pnas.1307218110 (2013).
76 Zhan, T., Rindtorff, N. & Boutros, M. Wnt signaling in cancer. Oncogene 36,
1461-1473, doi:10.1038/onc.2016.304 (2017).
77 Nagathihalli, N. S. et al. Inverse Correlation of STAT3 and MEK Signaling
Mediates Resistance to RAS Pathway Inhibition in Pancreatic Cancer.
Cancer Res 78, 6235-6246, doi:10.1158/0008-5472.Can-18-0634 (2018).
78 Xin, B. et al. Nerve growth factor regulates CD133 function to promote tumor
cell migration and invasion via activating ERK1/2 signaling in pancreatic
cancer. Pancreatology 16, 1005-1014, doi:10.1016/j.pan.2016.09.005
(2016).
79 Chen, Y. S. et al. CD133/Src axis mediates tumor initiating property and
epithelial-mesenchymal transition of head and neck cancer. PLoS One 6,
e28053, doi:10.1371/journal.pone.0028053 (2011).
102
List of references
103
List of references
104
List of references
101 Tu, Z. et al. CXCR4 is involved in CD133-induced EMT in non-small cell lung
cancer. Int J Oncol 50, 505-514, doi:10.3892/ijo.2016.3812 (2017).
102 Chaffer, C. L. & Weinberg, R. A. A perspective on cancer cell metastasis.
Science 331, 1559-1564, doi:10.1126/science.1203543 (2011).
103 Ayala, G. et al. Reactive stroma as a predictor of biochemical-free recurrence
in prostate cancer. Clin Cancer Res 9, 4792-4801 (2003).
104 Ligorio, M. et al. Stromal Microenvironment Shapes the Intratumoral
Architecture of Pancreatic Cancer. Cell 178, 160-175 e127,
doi:10.1016/j.cell.2019.05.012 (2019).
105 Carr, R. M. & Fernandez-Zapico, M. E. Pancreatic cancer microenvironment,
to target or not to target? EMBO Mol Med 8, 80-82,
doi:10.15252/emmm.201505948 (2016).
106 Fares, J., Fares, M. Y., Khachfe, H. H., Salhab, H. A. & Fares, Y. Molecular
principles of metastasis: a hallmark of cancer revisited. Signal Transduct
Target Ther 5, 28, doi:10.1038/s41392-020-0134-x (2020).
107 Haque, M. R. et al. Patient-derived pancreatic cancer-on-a-chip recapitulates
the tumor microenvironment. Microsyst Nanoeng 8, 36, doi:10.1038/s41378-
022-00370-6 (2022).
108 Rucki, A. A. et al. Heterogeneous Stromal Signaling within the Tumor
Microenvironment Controls the Metastasis of Pancreatic Cancer. Cancer Res
77, 41-52, doi:10.1158/0008-5472.Can-16-1383 (2017).
109 Chen, Q. et al. Tumour cell-derived debris and IgG synergistically promote
metastasis of pancreatic cancer by inducing inflammation via tumour-
associated macrophages. Br J Cancer 121, 786-795, doi:10.1038/s41416-
019-0595-2 (2019).
110 Sung, G. H., Chang, H., Lee, J. Y., Song, S. Y. & Kim, H. S. Pancreatic-
cancer-cell-derived trefoil factor 2 impairs maturation and migration of human
monocyte-derived dendritic cells in vitro. Anim Cells Syst (Seoul) 22, 368-
381, doi:10.1080/19768354.2018.1527721 (2018).
111 Sun, X. et al. Inflammatory cell-derived CXCL3 promotes pancreatic cancer
metastasis through a novel myofibroblast-hijacked cancer escape
mechanism. Gut 71, 129-147, doi:10.1136/gutjnl-2020-322744 (2022).
105
List of references
106
List of references
107
List of references
133 Zhan, H. X., Xu, J. W., Wu, D., Zhang, T. P. & Hu, S. Y. Pancreatic cancer
stem cells: new insight into a stubborn disease. Cancer Lett 357, 429-437,
doi:10.1016/j.canlet.2014.12.004 (2015).
134 Lonardo, E., Frias-Aldeguer, J., Hermann, P. C. & Heeschen, C. Pancreatic
stellate cells form a niche for cancer stem cells and promote their self-
renewal and invasiveness. Cell Cycle 11, 1282-1290, doi:10.4161/cc.19679
(2012).
135 Sainz, B., Jr. et al. Microenvironmental hCAP-18/LL-37 promotes pancreatic
ductal adenocarcinoma by activating its cancer stem cell compartment. Gut
64, 1921-1935, doi:10.1136/gutjnl-2014-308935 (2015).
136 Zhang, Y. et al. Canonical wnt signaling is required for pancreatic
carcinogenesis. Cancer Res 73, 4909-4922, doi:10.1158/0008-5472.Can-
12-4384 (2013).
137 Chen, K., Huang, Y. H. & Chen, J. L. Understanding and targeting cancer
stem cells: therapeutic implications and challenges. Acta Pharmacol Sin 34,
732-740, doi:10.1038/aps.2013.27 (2013).
138 Lee, J. Y., Song, S. Y. & Park, J. Y. Notch pathway activation is associated
with pancreatic cancer treatment failure. Pancreatology 14, 48-53,
doi:10.1016/j.pan.2013.11.011 (2014).
139 Abel, E. V. et al. The Notch pathway is important in maintaining the cancer
stem cell population in pancreatic cancer. PLoS One 9, e91983,
doi:10.1371/journal.pone.0091983 (2014).
140 Onishi, H. & Katano, M. Hedgehog signaling pathway as a new therapeutic
target in pancreatic cancer. World J Gastroenterol 20, 2335-2342,
doi:10.3748/wjg.v20.i9.2335 (2014).
141 Sainz, B., Jr., Martín, B., Tatari, M., Heeschen, C. & Guerra, S. ISG15 is a
critical microenvironmental factor for pancreatic cancer stem cells. Cancer
Res 74, 7309-7320, doi:10.1158/0008-5472.Can-14-1354 (2014).
142 Szczerba, B. M. et al. Neutrophils escort circulating tumour cells to enable
cell cycle progression. Nature 566, 553-557, doi:10.1038/s41586-019-0915-
y (2019).
143 Bramhall, S. R. et al. A double-blind placebo-controlled, randomised study
comparing gemcitabine and marimastat with gemcitabine and placebo as first
108
List of references
109
List of references
110
List of references
111
List of references
112
List of references
113
List of references
114
List of references
204 Balic, A., Dorado, J., Alonso-Gómez, M. & Heeschen, C. Stem cells as the
root of pancreatic ductal adenocarcinoma. Exp Cell Res 318, 691-704,
doi:10.1016/j.yexcr.2011.11.007 (2012).
205 Ma, Y., Hwang, R. F., Logsdon, C. D. & Ullrich, S. E. Dynamic mast cell-
stromal cell interactions promote growth of pancreatic cancer. Cancer Res
73, 3927-3937, doi:10.1158/0008-5472.Can-12-4479 (2013).
206 Park, B., Prasad, S., Yadav, V., Sung, B. & Aggarwal, B. B. Boswellic acid
suppresses growth and metastasis of human pancreatic tumors in an
orthotopic nude mouse model through modulation of multiple targets. PLoS
One 6, e26943, doi:10.1371/journal.pone.0026943 (2011).
207 Zhou, P. et al. The epithelial to mesenchymal transition (EMT) and cancer
stem cells: implication for treatment resistance in pancreatic cancer. Mol
Cancer 16, 52, doi:10.1186/s12943-017-0624-9 (2017).
208 Hirai, I. et al. Perineural invasion in pancreatic cancer. Pancreas 24, 15-25,
doi:10.1097/00006676-200201000-00003 (2002).
209 Maréchal, R. et al. High expression of CXCR4 may predict poor survival in
resected pancreatic adenocarcinoma. Br J Cancer 100, 1444-1451,
doi:10.1038/sj.bjc.6605020 (2009).
210 Wang, Z. et al. Aberrant expression of CXCR4 and β-catenin in pancreatic
cancer. Anticancer Res 33, 4103-4110 (2013).
211 Mizukami, T. et al. Immunohistochemical analysis of cancer stem cell
markers in pancreatic adenocarcinoma patients after neoadjuvant
chemoradiotherapy. BMC Cancer 14, 687, doi:10.1186/1471-2407-14-687
(2014).
212 Cui, K. et al. The CXCR4-CXCL12 pathway facilitates the progression of
pancreatic cancer via induction of angiogenesis and lymphangiogenesis. J
Surg Res 171, 143-150, doi:10.1016/j.jss.2010.03.001 (2011).
213 Bachet, J. B. et al. Contribution of CXCR4 and SMAD4 in predicting disease
progression pattern and benefit from adjuvant chemotherapy in resected
pancreatic adenocarcinoma. Ann Oncol 23, 2327-2335,
doi:10.1093/annonc/mdr617 (2012).
214 Xu, Q. et al. Stromal-derived factor-1α/CXCL12-CXCR4 chemotactic
pathway promotes perineural invasion in pancreatic cancer. Oncotarget 6,
4717-4732, doi:10.18632/oncotarget.3069 (2015).
115
List of references
215 Wu, H. et al. Coexpression of EGFR and CXCR4 predicts poor prognosis in
resected pancreatic ductal adenocarcinoma. PLoS One 10, e0116803,
doi:10.1371/journal.pone.0116803 (2015).
216 Demir, I. E. et al. Early pancreatic cancer lesions suppress pain through
CXCL12-mediated chemoattraction of Schwann cells. Proc. Natl. Acad. Sci.
U. S. A. 114, E85-e94, doi:10.1073/pnas.1606909114 (2017).
217 Lee, Y. et al. PAUF functions in the metastasis of human pancreatic cancer
cells and upregulates CXCR4 expression. Oncogene 29, 56-67,
doi:10.1038/onc.2009.298 (2010).
218 Kim, J. et al. Identification of anti-malarial compounds as novel antagonists
to chemokine receptor CXCR4 in pancreatic cancer cells. PLoS One 7,
e31004, doi:10.1371/journal.pone.0031004 (2012).
219 Eberl, M. et al. Hedgehog-EGFR cooperation response genes determine the
oncogenic phenotype of basal cell carcinoma and tumour-initiating
pancreatic cancer cells. EMBO Mol Med 4, 218-233,
doi:10.1002/emmm.201100201 (2012).
220 Singh, S., Srivastava, S. K., Bhardwaj, A., Owen, L. B. & Singh, A. P.
CXCL12-CXCR4 signalling axis confers gemcitabine resistance to pancreatic
cancer cells: a novel target for therapy. Br J Cancer 103, 1671-1679,
doi:10.1038/sj.bjc.6605968 (2010).
221 Zlotnik, A., Burkhardt, A. M. & Homey, B. Homeostatic chemokine receptors
and organ-specific metastasis. Nat Rev Immunol 11, 597-606,
doi:10.1038/nri3049 (2011).
222 Arora, S. et al. An undesired effect of chemotherapy: gemcitabine promotes
pancreatic cancer cell invasiveness through reactive oxygen species-
dependent, nuclear factor κB- and hypoxia-inducible factor 1α-mediated up-
regulation of CXCR4. J Biol Chem 288, 21197-21207,
doi:10.1074/jbc.M113.484576 (2013).
223 Brave, M. et al. FDA review summary: Mozobil in combination with
granulocyte colony-stimulating factor to mobilize hematopoietic stem cells to
the peripheral blood for collection and subsequent autologous
transplantation. Oncology 78, 282-288, doi:10.1159/000315736 (2010).
116
List of references
117
List of references
118
List of references
119
List of references
120
List of references
266 Gilg, A. et al. Absence of the CXCR4 antagonist EPI-X4 from pharmaceutical
human serum albumin preparations. J Transl Med 19, 190,
doi:10.1186/s12967-021-02859-6 (2021).
267 Xu, X. et al. Genome-wide screening reveals an EMT molecular network
mediated by Sonic hedgehog-Gli1 signaling in pancreatic cancer cells. PLoS
One 7, e43119, doi:10.1371/journal.pone.0043119 (2012).
268 Xu, X. et al. Sonic hedgehog-Gli1 signaling pathway regulates the epithelial
mesenchymal transition (EMT) by mediating a new target gene, S100A4, in
pancreatic cancer cells. PLoS One 9, e96441,
doi:10.1371/journal.pone.0096441 (2014).
269 Smigiel, J. M., Parameswaran, N. & Jackson, M. W. Potent EMT and CSC
Phenotypes Are Induced By Oncostatin-M in Pancreatic Cancer. Mol Cancer
Res 15, 478-488, doi:10.1158/1541-7786.Mcr-16-0337 (2017).
270 Le Large, T. Y. S. et al. Key biological processes driving metastatic spread
of pancreatic cancer as identified by multi-omics studies. Semin Cancer Biol
44, 153-169, doi:10.1016/j.semcancer.2017.03.008 (2017).
271 Capurso, G. & Sette, C. Drug resistance in pancreatic cancer: New player
caught in act. EBioMedicine 40, 39-40, doi:10.1016/j.ebiom.2019.02.008
(2019).
272 Hermann, P. C. & Sainz, B., Jr. Pancreatic cancer stem cells: A state or an
entity? Semin Cancer Biol 53, 223-231,
doi:10.1016/j.semcancer.2018.08.007 (2018).
273 Chen, D., Wei, L., Yu, J. & Zhang, L. Regorafenib inhibits colorectal tumor
growth through PUMA-mediated apoptosis. Clin Cancer Res 20, 3472-3484,
doi:10.1158/1078-0432.Ccr-13-2944 (2014).
274 de la Puente, P. et al. MEK inhibitor, TAK-733 reduces proliferation, affects
cell cycle and apoptosis, and synergizes with other targeted therapies in
multiple myeloma. Blood Cancer J 6, e399, doi:10.1038/bcj.2016.7 (2016).
275 Hoshino, R., Tanimura, S., Watanabe, K., Kataoka, T. & Kohno, M. Blockade
of the extracellular signal-regulated kinase pathway induces marked G1 cell
cycle arrest and apoptosis in tumor cells in which the pathway is constitutively
activated: up-regulation of p27(Kip1). J Biol Chem 276, 2686-2692,
doi:10.1074/jbc.M006132200 (2001).
121
List of references
122
List of references
287 Zhang, Y., Yang, J., Bronze, M. S., Houchen, C. W. & Li, M. Interplay of
Tumor Microenvironment Factors and Cancer Stem Cell Enrichment in
Pancreatic Ductal Adenocarcinoma. Gastroenterology 161, 1800-1802,
doi:10.1053/j.gastro.2021.09.024 (2021).
288 Barker, H. E., Paget, J. T., Khan, A. A. & Harrington, K. J. The tumour
microenvironment after radiotherapy: mechanisms of resistance and
recurrence. Nat Rev Cancer 15, 409-425, doi:10.1038/nrc3958 (2015).
289 Braicu, C. et al. A Comprehensive Review on MAPK: A Promising
Therapeutic Target in Cancer. Cancers (Basel) 11,
doi:10.3390/cancers11101618 (2019).
290 Ionescu, C. et al. TIMP-1 expression in human colorectal cancer is
associated with SMAD3 gene expression levels: a pilot study. J
Gastrointestin Liver Dis 23, 413-418, doi:10.15403/jgld.2014.1121.234.smad
(2014).
291 Smith, M. P. et al. The immune microenvironment confers resistance to
MAPK pathway inhibitors through macrophage-derived TNFα. Cancer
Discov 4, 1214-1229, doi:10.1158/2159-8290.Cd-13-1007 (2014).
292 Sang, N. et al. MAPK signaling up-regulates the activity of hypoxia-inducible
factors by its effects on p300. J Biol Chem 278, 14013-14019,
doi:10.1074/jbc.M209702200 (2003).
293 Zhang, Q., Han, Z., Zhu, Y., Chen, J. & Li, W. Role of hypoxia inducible factor-
1 in cancer stem cells (Review). Mol Med Rep 23,
doi:10.3892/mmr.2020.11655 (2021).
294 Tiwari, A. et al. Loss of HIF1A From Pancreatic Cancer Cells Increases
Expression of PPP1R1B and Degradation of p53 to Promote Invasion and
Metastasis. Gastroenterology 159, 1882-1897.e1885,
doi:10.1053/j.gastro.2020.07.046 (2020).
295 Pedersen, K. et al. Pancreatic cancer heterogeneity and response to Mek
inhibition. Oncogene 36, 5639-5647, doi:10.1038/onc.2017.174 (2017).
296 Brabletz, T., Jung, A., Spaderna, S., Hlubek, F. & Kirchner, T. Opinion:
migrating cancer stem cells - an integrated concept of malignant tumour
progression. Nat Rev Cancer 5, 744-749, doi:10.1038/nrc1694 (2005).
123
List of references
124
List of references
308 Young, B., Purcell, C., Kuang, Y. Q., Charette, N. & Dupré, D. J. Superoxide
Dismutase 1 Regulation of CXCR4-Mediated Signaling in Prostate Cancer
Cells is Dependent on Cellular Oxidative State. Cell Physiol Biochem 37,
2071-2084, doi:10.1159/000438566 (2015).
309 Meng, Q. et al. GPx1 is involved in the induction of protective autophagy in
pancreatic cancer cells in response to glucose deprivation. Cell Death Dis 9,
1187, doi:10.1038/s41419-018-1244-z (2018).
310 Aughton, K. et al. hENT1 Predicts Benefit from Gemcitabine in Pancreatic
Cancer but Only with Low CDA mRNA. Cancers (Basel) 13,
doi:10.3390/cancers13225758 (2021).
311 Zhi, Y. et al. Involvement of the nuclear factor-kappaB signaling pathway in
the regulation of CXC chemokine receptor-4 expression in neuroblastoma
cells induced by tumor necrosis factor-alpha. Int J Mol Med 35, 349-357,
doi:10.3892/ijmm.2014.2032 (2015).
312 Valk-Lingbeek, M. E., Bruggeman, S. W. & van Lohuizen, M. Stem cells and
cancer; the polycomb connection. Cell 118, 409-418,
doi:10.1016/j.cell.2004.08.005 (2004).
313 Widschwendter, M. et al. Epigenetic stem cell signature in cancer. Nat Genet
39, 157-158, doi:10.1038/ng1941 (2007).
314 Molofsky, A. V. et al. Bmi-1 dependence distinguishes neural stem cell self-
renewal from progenitor proliferation. Nature 425, 962-967,
doi:10.1038/nature02060 (2003).
315 Iwama, A. et al. Enhanced self-renewal of hematopoietic stem cells mediated
by the polycomb gene product Bmi-1. Immunity 21, 843-851,
doi:10.1016/j.immuni.2004.11.004 (2004).
316 Sangiorgi, E. & Capecchi, M. R. Bmi1 is expressed in vivo in intestinal stem
cells. Nat Genet 40, 915-920, doi:10.1038/ng.165 (2008).
317 Jacobs, J. J., Kieboom, K., Marino, S., DePinho, R. A. & van Lohuizen, M.
The oncogene and Polycomb-group gene bmi-1 regulates cell proliferation
and senescence through the ink4a locus. Nature 397, 164-168,
doi:10.1038/16476 (1999).
318 Czermin, B. et al. Drosophila enhancer of Zeste/ESC complexes have a
histone H3 methyltransferase activity that marks chromosomal Polycomb
sites. Cell 111, 185-196, doi:10.1016/s0092-8674(02)00975-3 (2002).
125
List of references
126
List of references
127
List of references
128
Acknowledgements
9 Acknowledgements
I would like to first thank my supervisor PD Dr. Dr. Patrick Christian Hermann (Dept.
of Internal Medicine I, University of Ulm) for providing me with an excellent
opportunity to perform my doctoral studies under his guidance. I express my sincere
gratitude for his continuous support and also thank him for allowing me to contribute
to different intriguing research projects. His supervision and motivation modelled my
learning capabilities to autonomously design projects, collaborate, guide other
students in the lab as well as for writing this thesis. I am convinced that his input
and encouragement will help me further in my scientific career.
Beside my supervisor, I would also like to thank the whole Hermann Lab. In
particular, I would express my earnest gratitude to Dr. Karolin Walter for her
exceptional support, cooperation, remarkable patience and kind words for the last
four years. Additionally, I would like to thank Dr. Eva Rodriguez-Aznar for critiquing
together numerous publications and inculcating in me the scientific urge to read.
Moreover, I would also like to thank all the students I got the pleasure to supervise
and/or work together, Arijan, Tabea, Inaas, Anton, Selina and Steffi. Even though it
was my responsibility to help and guide them, I did learn a lot from them. They made
some of the most stressful moments within the lab very joyous. In addition, I like to
acknowledge Andrea for her support and technical assistance. Without all of your
help, I wouldn’t have been able to achieve such great aims during my PhD period.
I would also like to thank people from the Dept. of Internal Medicine I, specially,
Akshaya, Sabine, Jessica and Florian for lengthy discussions of both scientific and
non-scientific manner. It made my time working in the department cheerful and
memorable.
129
Acknowledgements
Further, I would like to acknowledge Prof. Jan Münch and Mirja Harms (Institute of
Molecular Virology, University of Ulm) as well as Prof. Mika Lindèn, Bastian
Beitzinger and Roman Schmid (Institute of Inorganic Chemistry II, University of Ulm)
for their invaluable collaboration and continuous support.
Furthermore, I would like to thank CRC 1279 for an excellent collaborative research
program and funding of my position and research work. I further express my
gratitude to the International Graduate School in Molecular Medicine (IGradU) from
the University of Ulm for providing funding for attendances on conferences and
workshops. I thank the IGradU for the activities that allowed the opportunity to
develop my skills for my future career.
Moreover, I would like to fondly thank my family and all of my friends who
continuously motivated me and more importantly listened to me throughout my
whole study time.
And last, I would like to share my utmost love and heartfelt gratitude with Frank
Arnold. He, for the past 4.5 years, not only comforted, encouraged and consoled
me but also challenged me to become the best version of myself. I cannot thank you
enough.
130
Statutory declaration
10 Statutory declaration
I hereby declare that I wrote the present dissertation with the topic:
Novel molecular insights to discern and target migrating cancer stem cells
in pancreatic ductal adenocarcinomas
independently and used no other aids that those cited. In each individual case, I
have clearly identified the source of the passages that are taken word for word or
paraphrased from other works.
I also hereby declare that I have carried out my scientific work according to the
principles of good scientific practice in accordance with the current „Satzung der
Universität Ulm zur Sicherung guter wissenschaftlicher Praxis“ [Rules of the
University of Ulm for Assuring Good Scientific Practice].
Ulm, 30.05.2022
Kanishka Tiwary
131