Download The Role of Macrophages in Murine Intraocular - Ti

Document related concepts

Major urinary proteins wikipedia , lookup

Molecular mimicry wikipedia , lookup

Lymphopoiesis wikipedia , lookup

Phagocyte wikipedia , lookup

Immunomics wikipedia , lookup

Innate immune system wikipedia , lookup

Macrophage wikipedia , lookup

Cancer immunotherapy wikipedia , lookup

Adoptive cell transfer wikipedia , lookup

Transcript
University of Veterinary Medicine Hannover
The Role of Macrophages
in Murine Intraocular Melanoma
THESIS
Submitted in partial fulfilment of the requirements for the degree
Doctor of Veterinary Medicine
Doctor medicinae veterinariae
(Dr. med. vet.)
by
Marta M. Kilian
Dramburg
Hannover 2015
Academic supervision:
Prof. C. Pfarrer
Department of Anatomy
University of Veterinary Medicine Hannover
Prof. F. G. Holz
Department of Ophthalmology
University Hospital Bonn
1. Referee:
Prof. C. Pfarrer / Prof. F. G. Holz
2. Referee:
Prof. H.-J. Schuberth
Day of oral examination:
26.11.2015
Contents
List of Publications.....................................................................................................................
List of Abbreviations..................................................................................................................
I Introduction.......................................................................................................................... 11
Human uveal melanoma....................................................................................................... 11
The tumour microenvironment and tumour associated macrophages.................................. 14
Tumour vascularisation ........................................................................................................ 17
Animal models ..................................................................................................................... 20
Aims of the study ................................................................................................................. 21
II Materials and Methods ...................................................................................................... 23
Animals ................................................................................................................................ 23
HCmel12 melanoma cell line............................................................................................... 24
Murine macrophage cell line................................................................................................ 25
Macrophage polarisation in vitro ......................................................................................... 25
Macrophage depletion .......................................................................................................... 27
Intraocular tumour cell injection .......................................................................................... 28
Histology and immunohistochemistry ................................................................................. 29
Flow cytometry analysis....................................................................................................... 31
Ex vivo imaging.................................................................................................................... 31
Statistical analysis ................................................................................................................ 32
Histology and immunohistochemistry of Hgf-Cdk4R24C mice eyes..................................... 32
III Intravitreally injected HCmel12 melanoma cells serve as a mouse model of tumour
biology of intraocular melanoma .......................................................................................... 33
Abstract ................................................................................................................................ 34
Introduction .......................................................................................................................... 35
Materials and Methods ......................................................................................................... 37
Animals ............................................................................................................................ 37
HCmel12 melanoma cell line........................................................................................... 38
Clinical procedures........................................................................................................... 39
Histology .......................................................................................................................... 39
Results .................................................................................................................................. 41
Histology of intraocular HCmel12 melanoma ................................................................. 41
Immunohistochemistry of intraocular HCmel12 melanoma............................................ 45
Metastases of HCmel12 melanoma.................................................................................. 46
Discussion ............................................................................................................................ 47
References ............................................................................................................................ 50
IV Impact of macrophages on tumour growth characteristics in a murine ocular tumour
model ....................................................................................................................................... 53
Abstract ................................................................................................................................ 54
Introduction .......................................................................................................................... 56
Materials and Methods ......................................................................................................... 58
Animals ............................................................................................................................ 58
Mouse model of intraocular melanoma............................................................................ 59
Macrophage depletion (experiment 1) ............................................................................. 60
Macrophage polarisation (experiment 2) ......................................................................... 60
Histology and immunohistochemistry ............................................................................. 61
Flow cytometry analysis................................................................................................... 62
Statistical analysis ............................................................................................................ 63
Results .................................................................................................................................. 64
Macrophage depletion model (experiment 1) .................................................................. 64
Macrophage polarisation model (experiment 2) .............................................................. 67
Flow cytometry analysis................................................................................................... 72
Discussion ............................................................................................................................ 74
The role of TAM .............................................................................................................. 75
The impact of M2 macrophages....................................................................................... 76
The impact of age............................................................................................................. 78
Summary .......................................................................................................................... 79
Acknowledgements .............................................................................................................. 80
References ............................................................................................................................ 81
VI Origin of intratumoural vascular structures in a murine ocular tumour model........ 84
Abstract ................................................................................................................................ 85
Introduction .......................................................................................................................... 86
Materials and Methods ......................................................................................................... 88
Animals ............................................................................................................................ 88
Mouse model of intraocular melanoma............................................................................ 88
Ex vivo imaging................................................................................................................ 89
Histology and immunohistochemistry ............................................................................. 89
Statistical analysis ............................................................................................................ 90
Results .................................................................................................................................. 91
Ex vivo imaging................................................................................................................ 91
Histology and immunohistochemistry ............................................................................. 92
Discussion ............................................................................................................................ 95
Acknowledgements ............................................................................................................ 100
V Overexpression of Hepatocyte Growth Factor and an oncogenic CDK4 variant in mice
alters corneal stroma morphology but does not lead to spontaneous ocular melanoma103
Letter to the Editor ............................................................................................................. 104
References .......................................................................................................................... 108
VII Discussion....................................................................................................................... 109
Animal models ................................................................................................................... 109
Tumour associated macrophages in intraocular melanoma ............................................... 111
Impact of macrophage polarisation .................................................................................... 112
Impact of advanced age...................................................................................................... 114
Tumour associated macrophages and angiogenesis ........................................................... 116
The origin of tumour vessels.............................................................................................. 117
Outlook............................................................................................................................... 120
Summary ............................................................................................................................ 120
VIII References..................................................................................................................... 122
IX Summary.......................................................................................................................... 129
X Zusammenfassung ............................................................................................................ 132
XI Acknowledgements ......................................................................................................... 136
List of publications
List of Publications
Parts of this thesis are published or are in revision in following journals:
Original Papers
1. Marta M. Kilian, Karin U. Loeffler, Christiane Pfarrer, Frank G. Holz, Christian
Kurts, Martina C. Herwig
Intravitreally injected HCmel12 melanoma cells serve as a mouse model of tumour
biology of intraocular melanoma
Current Eye Research, ISSN: 0271-3683 print / 1460-2202 online DOI:
10.3109/02713683.2015.1004721
2. Marta M. Kilian, Karin U. Loeffler, Christian Kurts, Tobias Hoeller, Christiane
Pfarrer, Frank G. Holz, Martina C. Herwig
Impact of macrophages on tumour growth characteristics in a murine ocular tumour
model
Experimental Eye Research
manuscript submitted
3. Marta M. Kilian, Karin U. Löffler, Christiane Pfarrer, Tobias Höller, Frank G. Holz,
Daniela Wenzel, Martina C. Herwig
Origin of intratumoural vascular structures in a murine ocular tumour model
Journal
manuscript in preparation
Letter to the Editor (Own data)
1. Marta M. Kilian, Karin U. Löffler, Frank G. Holz, Thomas Tüting, Martina C.
Herwig
Hgf-Cdk4 mice fail as a spontaneous ocular melanoma model but exhibit unusual
histomorphologic ocular features
Melanoma Research
Accepted
List of publications
Contributions to Congresses
1. Marta M. Kilian, Karin U. Loeffler, Frank G. Holz, Christiane Pfarrer3, Christian
Kurts, Martina C. Herwig
Impact of macrophages and age on tumour growth characteristics in a murine ocular
tumour model
Poster at the 18th ECCO - 40th ESMO European Cancer Congress, Vienna, Austria
Abstract Number 172
2. Marta M. Kilian, Karin U. Loeffler, Frank G. Holz, Christiane Pfarrer3, Christian
Kurts, Martina C. Herwig
The effects of polarised macrophages on melanoma growth characteristics in vitro and
in vivo
Poster at the ARVO 2015 Annual Meeting in Denver, Colorado, USA
Kilian MM, et al. IOVS 2015,56:ARVO E-Abstract 5321
3. Marta M. Kilian, Karin U. Löffler, Frank G. Holz, Christiane Pfarrer, Tobias Höller,
Christian Kurts, Martina C. Herwig
Der Einfluss von Makrophagen auf das Tumorwachstum im Mausmodel für das
intraokuläre Melanom
Talk at the DOG 2014 in Leipzig, Germany; Abstract Number Fr06-05
4. Martina C. Herwig, Marta M. Kilian, Frank G. Holz, Daniela Wenzel, Karin U.
Löffler
Origin of Tumour-Associated Vessels and Vasculogenic Mimicry in an Intraocular
Murine Melanoma Model
Poster at the ARVO 2014 Annual Meeting in Orlando, Florida, USA
Kilian MM, et al. IOVS 2014,55:ARVO E-Abstract 5066
5. Karin U. Löffler, Marta M. Kilian, Frank G. Holz, Thomas Tüting, Martina C.
Herwig
Morphologic and immunohistochemical features in the ageing Hgf-Cdk4 mouse eye
Poster at the ARVO 2014 Annual Meeting in Orlando, Florida, USA
Kilian MM, et al. IOVS 2014,55:ARVO E-Abstract 5071
6. Marta M. Kilian, Karin U. Loeffler, Hans E. Grossniklaus, Frank G. Holz, Christiane
Pfarrer, Christian Kurts, Martina C. Herwig
The effects of macrophages on tumour growth characteristics in a mouse-model of
intraocular melanoma
Poster at the ARVO 2014 Annual Meeting in Orlando, Florida, USA
Kilian MM, et al. IOVS 2014,55:ARVO E-Abstract 5070
List of publications
7. Marta M. Kilian, Karin U. Löffler, Hans E. Grossniklaus, Frank G. Holz, Christian
Kurts, Marina C. Herwig
Evaluation einer neuen murinen Hautmelanomzelllinie als potentielles Mausmodell
für das intraokuläre Melanom
Poster at the DOG 2013 Annual Meeting in Berlin, Germany; Poster award winner
Abstract Number A-572-0008-00718
8. Marta M. Kilian, Karin U. Loeffler, Hans E. Grossniklaus, Frank G. Holz, Christiane
Pfarrer, Christian Kurts, Martina C. Herwig
Comparison of Two Melanoma Cell Lines as Mouse-Models of Uveal Melanoma
Poster at the ARVO 2013 Annual Meeting in Seattle, Washington, USA;
Kilian MM, et al. IOVS 2013,54:ARVO E-Abstract 4203
List of Abbreviations
Ab
AEC
BAP1
BC199
CDK4
CSC
DMEM/F12
DT101
ECM
ELISA
FACS
FCS
GFP
GNA11
GNAQ
H&E
HGF
HMB45
HRP
IFNγ
IHC
IL
LPS
MФ
M1
M2
MVD
NEAA
NIH
NK
PAS
PBS
PEDF
RAS
RPE
RPMI 1640
SCID
TAM
TEC
TBS
TGFβ
TNFα
UM
VEGF
VM
Antibody
3-amino-9-ethylcarbazole
BRCA1 associated protein-1
Melanoma antigen clone
Cyclin-dependent kinase 4
Cancer stem cells
Dulbeccos Modified Eagle Medium /F12, cell culture medium
Melanoma antigen clone
Extracellular matrix
Enzyme-linked immunosorbent assay
Fluorescence-activated cell sorting
Fetal calve serum
Green fluorescent protein
Guanine nucleotide binding protein (G protein), alpha 11
Guanine nucleotide binding protein (G protein), q polypeptide
Hematoxylin and eosin
Hepatic growth factor
Human melanoma black 45 antigen
Horseradish peroxidase
Interferon γ
Immunohistochemistry
Interleukin
Lipopolysaccharide
Macrophages
Macrophages with a functional M1 polarisation
Macrophages with a functional M2 polarisation
Microvascular density
Non-essential amino acid
National Insitutes of Health
Natural killer (cells)
Periodic acid-Schiff
Phosphate buffered saline
Pigment epithelium-derived factor
Rat sarcoma, ongcogene
Retinal pigment epithelium
Cell culture medium Roswell Park Memorial Institute
Severe combined immunodeficiency
Tumour associated macrophages
Tumour endothelial cells
Tris buffered saline
Transforming growth factor β
Tumour necrosis factor α
Uveal melanoma
Vascular endothelial growth factor
Vasculogenic mimicry
11
I Introduction
Human uveal melanoma
Uveal melanoma is the most common primary intraocular tumour in the adult Caucasian
population. It occurs usually in the elderly with an incidence of ~ 2-8 cases per million/year
(Virgili, Gatta et al. 2007; Singh, Turell et al. 2011; Mallone, De Vries et al. 2012). Other
ethnities exhibit a far lower incidence rate (Eagle 2013). Uveal melanoma arises from
melanocytes of the choroid, the stroma of the ciliary body and/or the iris. It can develop from
a pre-existing nevus or denovo. However, intraocular tumours may be of primary or of
metastatic origin. Uveal metastases represent the most frequent ocular malignancy. They most
commonly derive from breast (in female) or lung (in male) carcinoma but may also originate
from prostate, skin, colon or other carcinomas (reviewed in (Arepalli, Kaliki et al. 2015)).
Clinical characteristics of primary uveal melanoma are defined as tumour growth with a
thickness of ≥ 3 mm, lipofuscin accumulation, subretinal fluid/serous retinal detachment and
visual symptoms (Fig. 1) (Shields, Manalac et al. 2014).
Figure 1. Cross section of an enucleated human eye with a primary uveal melanoma (left) and
schematic drawing of an eye with uveal melanoma (right).
12
I Introduction
Local treatment of the primary tumour is very promising and depends on tumour
characteristics such as size and localisation (reviewed in (Singh and Singh 2012)). It ranges
from enucleation of the globe to eye-saving strategies like local radiation therapy
(brachytherapy), thermotherapy or proton beam therapy of the tumour.
In about 40% of cases of uveal melanoma metastases develop within 10 years (Singh, Shields
et al. 2001). A special characteristic of intraocular melanomas is their hematogenous pathway
of metastasis due to the absence of lymphatic vessels in the eye. The metastases show a
predisposition to the liver and are rarely detected in the lungs or other sites (Singh, Shields et
al. 2001). At the time of initial diagnosis the ocular tumour often already has developed
micrometastases which may remain dormant for decades (Bakalian, Marshall et al. 2008).
However, once micrometastases undergo an angiogenic switch and start to grow, metastatic
disease develops. Prognosis worsens dramatically upon metastatic disease due to lack of
effective systemic treatment options (Kujala, Makitie et al. 2003).
Many studies have focused on prognostic parameters of uveal melanoma to estimate the risk
of metastasis and patient’s outcome. Such clinical and histopathological prognostic factors
include (advanced) patient age, primary tumour size (diameter and prominence), cell type,
ciliary
body
involvement,
mean
vascular
density
(MVD),
extravascular
matrix
patterns/vasculogenic mimicry (VM) and infiltrating inflammatory cell type, in particular
macrophages (Fig. 2) (Foss, Alexander et al. 1996; Folberg, Mehaffey et al. 1997; Shields,
Kaliki et al. 2013; Kaliki, Shields et al. 2015). Therefore, histologic examination of
enucleated eyes harbouring an uveal melanoma may provide prognostic information. In
addition, molecular gene analysis of uveal melanoma biopsies and enucleation specimens
became available recently, providing relevant prognostic information with regard to
development of metastases and patient’s outcome. Such analysis can distinguish two
I Introduction
13
prognostic molecular subsets of primary uveal melanoma: class 1 tumours with low metastatic
risk and class 2 tumours with high metastatic risk (Onken, Worley et al. 2004). Chromosomal
analysis and genetic profiling were found to be more reliable indicators for patient’s
prognosis than histopathologic characteristics (Onken, Worley et al. 2012; Field and Harbour
2014). However, the histologic parameters reflect uveal melanoma growth characteristics and
allow for their evaluation in animal models, which may deliver important insights into tumour
biology, progression and metastasis.
Figure 2: Prognostic histological features of uveal melanoma
(A+B) An epitheloid cell type (A) has a worse prognosis than a spindle cell type (B, H&E); (C) The
occurrence of vasculogenic mimicry and extravascular matrix patterns is associated with death from
metastatic disease in uveal melanoma (PAS w/o hematoxylin); (D) A high mitotic activity is
considered as a prognostic factor in many solid tumours including uveal melanoma (IHC Ser-10
antibody); (E) Tumour associated macrophages play an important role in the pathogenesis of uveal
melanoma and their occurrence is associated with patients’ prognosis (IHC, CD68 [left] and CD68 +
CD163 antibody[right]); (modified from (Damato, Eleuteri et al. 2011) and (Herwig, Bergstrom et al.
2013)).
I Introduction
14
Several genetic anomalies (e.g. monosomy 3, GNAQ, GNA11, Bap1, class 2 gene expression
profile) influencing tumour progression and metastasis have been identified (Sisley, Rennie et
al. 1990; Prescher, Bornfeld et al. 1996; Onken, Worley et al. 2004; Ehlers, Worley et al.
2008). But unlike cutaneous or conjunctival melanoma, mutations in B-RAF, RAS or KIT
genes occur rarely in uveal melanoma (Griewank, Westekemper et al. 2013). Characteristic
mutations differ between uveal and cutaneous melanoma and even among tumours itself,
accounting for different progression and metastatic behaviour (Mehnert and Kluger 2012).
Metastatic dissemination is a complex multistep process with local invasion, intravasation,
colonisation of distant tissues and adaptation to a foreign microenvironment at the secondary
site. As metastatic potential is linked to many biological features of the primary tumour, basic
research on the primary tumour microenvironment may offer new implications for strategies
against metastasis and identify new therapeutic targets.
The tumour microenvironment and tumour associated macrophages
Each tumour has its own microenvironment. It is composed of neoplastic and non-neoplastic
cells as well as the stroma and it involves their functional interactions through different
cytokines, chemokines or growth factors. Such interactions may withhold tumour growth or
create a synergistic interplay and enhance tumour progression as well as metastasis (Catalano,
Turdo et al. 2013). Recent data showed that hypoxia is a crucial factor of the
microenvironment. It can induce changes in gene expression of tumour cells and/or nonneoplastic cells and it thereby may influence apoptosis, invasion and angiogenesis via
different mechanisms (Bronkhorst, Jehs et al. 2014). Hypoxia-induced secretion of
chemotactic cytokines or growth factors recruits blood leucocytes into the tumour which in
turn further orchestrate the microenvironment. Tumour associated inflammation is one of the
I Introduction
15
hallmarks of cancer and includes the presence of infiltrating leucocytes. Main players of the
inflammatory infiltrate in many tumours - like uveal melanoma - are tumour associated
lymphocytes and macrophages (Sica and Mantovani 2012). To overcome hypoxia tumour
cells and tumour associated macrophages (TAM) secrete pro-angiogenic factors in order to
enhance angiogenesis and other strategies of tumour vascularisation (e.g. vasculogenic
mimicry (Fig. 2C)). An inflammatory and pro-angiogenic microenvironment favours tumour
growth and is thus correlated with a poor prognosis. Further, the direct crosstalk between
melanoma cells and TAM may either enhance or reduce tumour growth (Hussein 2006).
TAM are an important component of the leukocytic infiltrate in tumours and are believed to
play a major role in malignant neoplasms (Fig. 3). In most solid tumours, including uveal
melanoma, high numbers of TAM correlate statistically significant with poor prognosis
(Allavena, Sica et al. 2008). In uveal melanoma, they are further associated with monosomy
3, several histopathologic features such as a high mean vascular density (MVD) or certain
extravascular matrix patterns, and thus with an aggressive tumour phenotype (Folberg,
Mehaffey et al. 1997; Makitie, Summanen et al. 2001; Hendrix, Seftor et al. 2003; Toivonen,
Makitie et al. 2004; Maat, Ly et al. 2008). Different functional macrophage phenotypes
(polarisation) exist, which are influenced by the present microenvironmental stimuli (Arnold,
Henry et al. 2007). The TAM polarisation phenotype is determined by cytokines produced by
cells in the tumour microenvironment (Arnold, Henry et al. 2007). It may range within a
broad spectrum from M1 to M2 macrophages (Jager, Ly et al. 2011). M1-polarised
macrophages exhibit pro-inflammatory, tumouricidal and anti-angiogenic qualities with the
potential to suppress tumour growth and dissemination (Sica and Mantovani 2012). In nonprogressing tumours, TAM mainly resemble the M1 type and exhibit anti-tumour activity. In
malignant and advanced tumours, TAM are rather polarised toward the M2 phenotype that
I Introduction
16
instead favours tumour malignancy (Biswas and Mantovani 2010). Tumour driven
macrophage polarisation tends to specialise TAM into the M2 phenotype with pro-angiogenic,
immunosuppressive and tumour-favouring properties. TAM are a source and target for
various cytokines and chemokines within the tumour microenvironment (Fig. 3).
Figure 3: Role of tumour associated macrophages (TAM) within the tumour microenvironment.
TAM are a source and target for various cytokines and chemokines which regulate tumour growth,
progression and invasion; they further interact with other components like the stroma as well as with
other players of the adaptive immunity (modified from (Mantovani, Sozzani et al. 2002)).
M2-polarised TAM exhibit characteristic cytokine expression levels of IL-10high, IL-12low,
TNFαlow, IFNγlow and TGF-βhigh (Mantovani, Sozzani et al. 2002). A high M2/M1 ratio in
uveal melanomas was shown to be related to worse prognosis and to histologic negative
prognostic parameters (Herwig, Bergstrom et al. 2013).
I Introduction
17
With increasing age, changes in the capability of the innate immunity induce a shift of the
macrophage secretory profile towards a M2 profile (Gomez, Nomellini et al. 2008).
Accordingly, macrophages from geriatric mice are particularly sensitive to signals that
promote their M2-polarisation, which in turn may promote tumour development (Jackaman,
Radley-Crabb et al. 2013). A M2-dominated tumour microenvironment comprises proangiogenic factors such as vascular endothelial growth factor (VEGF) and other growth
factors, angiopoietins, certain interleukins and matrix metalloproteinases (Schmid and Varner
2010). Such a microenvironment is essential for tumour growth, tumour vascularisation and
subsequently a basis for metastasis in uveal melanoma. Thus, TAM may promote a
microenvironment which enhances angiogenesis, extracellular matrix remodelling and
immune suppression (Mantovani, Sozzani et al. 2002).
Tumour vascularisation
Angiogenesis is a crucial point in the development of most solid tumours, including uveal
melanoma. Solid tumours are dependent on constant blood supply to ensure tumour growth
beyond diffusion limits and to satisfy the subsequent need for oxygen and nutrients. To
address this demand tumours use different forms of vascularisation which represents one of
the hallmarks of cancer (Hanahan and Weinberg 2011). One source of initial blood supply
may be angiotropism and the incorporation of pre-existing vessels from surrounding
structures (co-option) - in case of intraocular melanoma this includes structures such as the
uvea. Second, the tumour may induce growth of new vessels by stimulating sprouting or
intussusception from pre-existing host vessels (angiogenesis) or de novo formation of vessels
by recruiting endothelial progenitor cells (vasculogenesis) (Fig. 4).
I Introduction
18
Figure 4: Strategies of tumour vascularisation
Co-option – colonisation of existing vessels; Angiogenesis – branching and colonisation of existing
host vessels; vasculogenesis – formation of new vessels (de novo) from circulating endothelial
progenitor cells (migrating from the bone marrow); vasculogenic mimicry - tumour cells lining
channels of extracellular matrix forming vasculogenic networks (modified from (Burrell and Zadeh
2012)).
Recently, a slowly increasing number of studies in different tumour entities reported vessel
formation out of cancer cells (Ricci-Vitiani, Pallini et al. 2010; Wang, Chadalavada et al.
2010; Ohmura-Kakutani, Akiyama et al. 2014; Chroscinski, Sampey et al. 2015).
Hypothetically, such tumour-derived blood vessel cells originated from multipotent cancer
stem cells. However, their existence and function in tumour vascularisation remain subject to
ongoing discussion.
The essential role of sprouting angiogenesis in solid tumours and its potential therapeutic
implications were first described by Folkman and coworkers (Folkman 1971). Many
preclinical experiments indicated so far that solid tumours were profoundly angiogenesisdependant. However, the long-held belief that cancer patients can be effectively treated with
I Introduction
19
anti-angiogenic therapy to prevent tumour progression and metastasis diminished after
disappointing clinical results. Heterogeneity and complexity of tumours as well as alternative
vascularisation strategies apart from angiogenesis may be a reason for that (Verheul, Voest et
al. 2004). However, anti-angiogenic compounds such as bevacizumab (anti-VEGF antibody)
still represent an adjuvant therapeutic option in patients with certain metastatic cancers such
as breast carcinomas (Keating 2014). Alternative forms of primary tumour vascularisation in
aggressive tumours may include mosaic vessels (tumour- and endothelial-lined vasculature)
(Chang, di Tomaso et al. 2000), tumour-derived endothelial-like vessels, tumour-lined
vascular channels (Timar and Toth 2000), and vasculogenic mimicry (Maniotis, Folberg et al.
1999).
Vasculogenic mimicry (VM) - which was initially observed in uveal melanoma as well as in
metastatic cutaneous melanoma (Maniotis, Folberg et al. 1999) - may represent an alternative
perfusion to angiogenesis. VM is characterised by vasculogenic-like non-endothelial channels
rich of PAS-positive extracellular matrix (ECM). It exhibits high expression levels of ECM
related molecules (e.g. matrix-metalloproteinases, VE-cadherin, VEGF-A) and is lined by
tumour cells (Fig. 4) (Seftor, Hess et al. 2012). VM is mainly found in aggressive and fast
growing tumours in which tumour cell plasticity and the hypoxic microenvironment
contribute to formation of this alternative perfusion pathway. Aggressive melanoma cells are
potentially able to modify their surrounding microenvironment towards VM and thereby
adapt to rapid tumour growth and overcome hypoxia (Hendrix, Seftor et al. 2003). The
formation of microcirculatory patterns of VM, like back-to-back loops or parallel vessels with
cross-linkings, are associated with death from metastatic disease in uveal as well as in
cutaneous melanoma (Folberg, Mehaffey et al. 1997).
I Introduction
20
Animal models
Considering aforementioned characteristic features of uveal melanoma like hematogenous
spread, different strategies of vascularisation, the inflammatory microenvironment and
genetic attributes, a meaningful mouse model should offer similar properties.
Since no spontaneous primary uveal melanomas in wild type mice have been described yet,
all existing mouse models have restrictions or limitations. Different models have been
developed including transgenic mice, intraocular inoculation with cutaneous melanoma cells
into wild type mice, injection of human uveal melanoma cells into mice with severe combined
immunodeficiency (SCID) or induction of uveal proliferations by chemicals, radiation or
viruses (Dithmar, Albert et al. 2000). Pigmented intraocular tumours arising in transgenic
mice were identified to be either of retinal pigment epithelium origin (Syed, Windle et al.
1998; Albert, Kumar et al. 2004) or as small uveal tumours which failed to metastasise to the
liver (Kramer, Powell et al. 1998; Tolleson, Doss et al. 2005; Latendresse, Muskhelishvili et
al. 2007). Injection of human uveal melanoma cells into immunodeficient mice also resulted
in non-metastatic disease (Mueller, Maniotis et al. 2002) and thus does not allow to study
influences and immunologic interactions between the tumour and the host. The best
reasonable compromise and an established mouse model so far was created and established by
injecting murine, cutaneous B16LS9 melanoma cells into the vitreous of C57Bl/6 mice,
generating an ocular tumour with metastatic dissemination to the lungs and the liver (Fig. 5)
(Diaz, Rusciano et al. 1999; Dithmar, Albert et al. 2000).
21
I Introduction
C
C
C
I
I
I
I
I
L
L
M
R
R
R
A
B
C
Figure 5: Mouse model of intraocular melanoma; H&E stained sections through the right mouse
eye, R: retina, L: lens, I: iris, C: cornea, M: melanoma, 40x magnification.
(A) Injection of murine cutaneous melanoma cells into the vitreous by a schematic needle; (B)
Injected melanoma cells distribute throughout the vitreous after injection, arrow: point of previous
injection; (C) Solid intraocular melanoma, nine days after tumour cell injection.
For most of the above mentioned mouse models young mice (aged 8-12 weeks) were used.
Thus, age-related changes in the tumour microenvironment and in immune cells were not
considered. Differences in mice age, different definitions of “elderly mice” and thus a lack of
standardised examination methods may lead to non-uniform results among study groups who
examine tumour biology (Jackaman, Radley-Crabb et al. 2013). However, such age-related
influences should be considered when investigating immunological aspects of tumour biology
since old mice have different immune properties compared to young mice.
Aims of the study
1. As macrophages play a pivotal role in the pathology of uveal melanoma and orchestration
of the tumour microenvironment, we wanted to further investigate their influence on tumour
growth characteristics, vascularisation as well as metastasis. Therefore, we modified an
I Introduction
22
established B16 model in CX3CR1+/GFP mice (chapter III, Intravitreally injected HCmel12
melanoma cells serve as a mouse model of tumour biology of intraocular melanoma).
2. To determine the role of macrophages and their polarisation, we examined
histopathological tumour characteristics of macrophage-depleted mice in comparison to
untreated mice as well as of M1- and M2-conditioned tumours with regard to age (chapter IV,
Impact of macrophages on tumour growth characteristics in a murine ocular tumour model).
3. Further, we intended to study the macrophage-dependent origin and composition of
intratumoural vessels in transgenic flt-1/eGFP BAC mice (chapter V, Origin of intratumoural
vascular structures in a murine ocular tumour model).
4. In permanent search of a mouse model of spontaneous uveal melanoma, we evaluated
ocular findings of elderly Hgf-Cdk4(R24C) mice (chapter VI, Overexpression of hepatocyte
growth factor and an oncogenic CDK4 variant in mice alters corneal stroma morphology but
does not lead to spontaneous ocular melanoma).
23
II Materials and Methods
Animals
Breeding and housing of C57Bl/6, CX3CR1+/GFP, flt-1/eGFP BAC and Hgf-Cdk4R24C mice
mice was carried out and supervised according to the Association of Research in Vision and
Ophthalmology statement for the Use of Animals in Ophthalmic and Vision Research. All
experiments were approved by the Landesamt für Natur, Umwelt und. Verbraucherschutz
NRW (Recklinghausen, Germany). In total, 143 mice of four different strains were used for
the studies (Table 1). Of these, six C57Bl/6 mice and 58 CX3CR1+/GFP mice were examined at
the age of 8-12 weeks (young), whereas three C57Bl/6 mice, 43 CX3CR1+/GFP mice and 15 flt1/eGFP BAC mice examined at the age of 8-12 months (old). Four C57Bl/6 mice and two
CX3CR1+/GFP mice were kept as negative controls. The eyes of 12 Hgf-Cdk4R24C mice aged 11
months were examined.
In CX3CR1+/GFP mice which are based on a C57Bl/6 strain, the transmembrane-receptor
CX3CR1 for CX3C chemokine fractalkine had been replaced by green fluorescent protein
(GFP) reporter gene. Hence, these mice exhibit an in vivo labelling of peripheral blood
monocytes, macrophages, subsets of NK and dendritic cells, as well as of retinal microglia by
fluorescence at 488nm (Jung, Aliberti et al. 2000). This innate fluorescent signal of monocytic
cells may allow their direct visualisation by immunofluorescence microscopy. Homozygous
male CX3CR1GFP/GFP mice for breeding were kindly provided by Prof. C. Kurts, Institute of
Experimental Immunology, University of Bonn, Bonn, Germany. Female C57Bl/6 mice were
crossbred with homozygous male CX3CR1GFP/GFP mice generating a heterozygous
CX3CR1+/GFP mice generation.
24
II Materials and Methods
Flt-1/eGFP BAC mice exhibit a persistent eGFP expression in small vessels which is driven
by the flt-1 promoter (VEGF receptor 1, VEGFR-1) (Herz, Heinemann et al. 2012). Thus, all
ocular vessels originating from the host (mouse) exhibit a constant eGFP expression.
In Hgf-Cdk4R24C mice overexpression of hepatocyte growth factor (HGF) promotes cutaneous
melanogenesis by activating RAS signal transduction pathways via its receptor c-MET
(Landsberg, Gaffal et al. 2010). Additionally, the oncogenic germline mutation in the cyclindependent kinase 4 (CDK4 R24C) further contributes to melanoma development in HgfCdk4R24C mice because it functionally inactivates the tumour suppressor p16⁄INK4a, which is
of critical importance for oncogene-induced senescence (Landsberg, Kohlmeyer et al. 2012).
Table 1: Total animal numbers (all experiments), *=negative controls
C57Bl/6
CX3CR1+/GFP
flt-1/eGFP BAC
Hgf-Cdk4R24C
Young
(8-12 weeks)
Old
(8-12 months)
6 (+4*)
58 (+2*)
-
3
43
15
12
HCmel12 Melanoma Cell Line
HCmel12 melanoma cells were generously provided by Prof. T. Tüting (Laboratory of
Experimental Dermatology, University of Bonn, Bonn, Germany). This murine cutaneous
melanoma cell line had been derived from an autochthonous cutaneous melanoma of an HgfCdk4 R24C mouse which spontaneously develops a spectrum of primary cutaneous melanomas
(Landsberg, Gaffal et al. 2010). These cells exhibit particular characteristics that may qualify
them as a cell line for a potent model of metastasising intraocular melanoma. HCmel12
II Materials and Methods
25
melanoma cells were obtained at cell culture passage seven. Cell lines at such an early
passage number still exhibit “original” growth characteristics because mutations which may
change their phenotype might have not established, yet. HCmel12 melanoma cells were
cultured in RPMI 1640 + Hepes medium supplemented with 10% FCS, 1%
Penicillin/Streptomycin, 1% NEAA, 1% Sodium-Pyruvate and 0.75% β-Mercaptoethanol.
Cells were incubated at 37°C in a humidified atmosphere containing 5% CO2 and grown to
approx. 70% confluence. HCmel12 melanoma cells were cryo-conserved at cell culture
passage 8. After thawing, cells were cultured for another passage, trypsinised and washed in
PBS buffer before aliquots suspended in PBS buffer were injected into the eye.
Murine macrophage cell line
A bone marrow derived and immortalised macrophage cell line from wild type C57Bl/6 mice
was generated (Hornung, Bauernfeind et al. 2008) and kindly provided by Prof. Eicke Latz
(Institute of Innate Immunity, University of Bonn, Bonn, Germany). The macrophage cell line
was cultured in DMEM/F12 medium at 37°C in a humidified atmosphere containing 5% CO2,
grown to max. 70% confluence and passaged every second to third day as described above.
Cells were cryo-conserved at a density of approx. 1000 cells per µl.
Macrophage polarisation and proliferation in vitro
To investigate the impact of polarised macrophages on intraocular melanoma, HCmel12
melanoma cells were exposed in vitro to the supernatant of murine, polarised macrophages
prior to intravitreal injection. Murine macrophages were cultured in medium without fetal calf
serum (FCS) for two hours prior to in vitro polarisation to avoid any impact of cytokines or
chemokines within the FCS. Macrophages were treated with specific cytokines and
II Materials and Methods
26
lipopolysaccharide (LPS) to obtain different polarised phenotypes (according to a modified
protocol by (Mosser and Zhang 2008)). IFNγ (200ng/ml) for M1-macrophages and
Interleukin 4 (IL-4) (20ng/ml) for M2 macrophages were added to the serum-free medium in
order to polarise macrophages. After 10 hours of incubation, LPS (100ng/ml) was
supplemented for each macrophage type for a total incubation time of 24 hours. Polarisation
status of unfixed macrophages was verified functionally by using a multiple cytokine ELISA
covering the inflammatory cytokines IL-6, IL-10. IL-12, IL-23, TNFα and IFNγ (MultiAnalyte ELISArrayTM Kit, Mouse Mix-N-Match, Qiagen, Venlo, Netherlands). M1-specific
expression of these cytokines comprises levels of IL-6high, IL-10low, IL-12high, IL-23high,
TNFαhigh and IFNγhigh (M2-specific expression levels vice versa) (Mantovani, Sica et al.
2004). Phenotypical macrophage polarisation status of 4% PFA-fixed cells was examined by
immunocytology with a double staining kit (MultiVision Polymer Detection System, Thermo
Scientific, Waltham, MA, USA) using a F4/80- (for all macrophages) and CD163- (for M2
macrophages only; CD163, M-96, Santa Cruz Biotechnology, Santa Cruz, CA, USA) primary
antibody. To examine the impact of a M1- or M2-conditioned tumour microenvironment,
HCmel12 melanoma cells were treated with serum-free medium for two hours and afterwards
exposed to the supernatant of M1- or M2-polarised macrophages for 20 hours, respectively
(Figure 6). By this means, HCmel12 melanoma cells were incubated in a M1- or M2dominated microenvironment. These M1- or M2-conditioned HCmel12 melanoma cells were
then injected intravitreally as described below.
Cell proliferation rates of unpolarised and of M1- or M2-conditioned HCmel12 melanoma
cells were compared in vitro using a BrdU proliferation assay (BrdU Cell Proliferation Assay,
Milipore, Billerica, MA; USA).
II Materials and Methods
27
Figure 6. Experimental design and workflow of in vitro polarisation of macrophages, its verification,
conditioning of HCmel12 melanoma cells, and examination of the impact of polarised macrophages on
melanoma growth.
Macrophage depletion
To study the impact of macrophages in intraocular melanoma in CX3CR1+/GFP and flt-1/eGFP
BAC mice, macrophage-depleted mice were studied in comparison to untreated mice.
Liposome-encapsulated dichloromethylene diphosphonate (Cl2MDP; Clodronate, ordered
through clodronateliposomes.org) was used to systemically deplete blood monocytes and
macrophages at a dosage of 0.1mg/g body weight (van Rooijen and van Kesteren-Hendrikx
2003). Mice received repeated intraperitoneal injections of Clodronate liposomes on days -6, 3, 0 (day of intravitreal tumour cell injection) and day 3. To evaluate macrophage depletion,
liver sections of Clodronate-treated and untreated mice were stained immunohistochemically
with a GFP-antibody (GFP Rabbit IgG, Life Technologies, Carlsbad, CA, USA; dilution
1:100) or F4/80-antibody (F4/80 Antibody, Cl:A3-1, AbD Serotec, Oxford, U.K.; dilution
1:100) as described below (Histology).
II Materials and Methods
28
Intraocular tumour cell injection
Experimental groups which received intraocular tumour cell injections consisted of 12 mice in
the animal model experiment (chapter III), 95 mice in the depletion and polarisation
experiment (chapter IV), and 15 mice in the vascularisation experiment (chapter V). General
anaesthesia comprised Isofluran induction followed by intraperitoneal Ketamin/Xylazin
injection. On day 0. in general anaesthesia, 1x105 HCmel12 melanoma cells suspended in 1µl
PBS were injected intravitreally into the right eye of each mouse. An automatic microsyringe
injector (UltraMicroPump III, World Precision Instruments, Sarasota, USA) with a 10µl
microinjection syringe (701 ASRN, Hamilton, Reno, USA) equipped with a 34-gauge needle
(RN Needle, Hamilton, Reno, USA) was used for injection. The needle was inserted approx. 1
mm posterior to the limbus through the sclera into the vitreous. Negative controls in the
animal model experiment (chapter III) received 1µl of PBS, a sole needle stitch with no
injecting agent (n=2) or no treatment at all (n=1). Intraocular injections were performed very
carefully to avoid punctuation of the lens. Negative controls may show whether a noteworthy
influx of immune cells was caused by the injection process of PBS or the stitch alone.
Tumour bearing eyes of all mice were enucleated in general anaesthesia on day 9 and 12
CX3CR1+/GFP and C57Bl/6 mice were sacrificed on day 42 in the animal model experiment.
Then tissue samples from lungs, liver and spleen, as well as the contralateral eye and
mandibular lymph nodes were collected for further examination to verify metastatic routes
and sites (chapter III). In all other experiments, mice were euthanised on day 9 with
immediate enucleation of the tumour bearing eye (chapter IV+V).
All but 20 eyes of CX3CR1+/GFP, all C57Bl/6 mice and 12 eyes of flt-1/eGFP BAC mice were
processed for histology. 20 eyes of CX3CR1+/GFP mice were investigated by flow cytometry
analysis and three eyes of flt-1/eGFP BAC mice were investigated by ex vivo imaging.
II Materials and Methods
29
Histology and immunohistochemistry
Specimens for histologic examination were submitted to 4% paraformaldehyde for 24 hours,
dehydrated with alcohol in ascending order and cleared with xylene. After embedding in
paraffin, 5 µm thick serial sections of the eye and five sections through the center of the other
tissues were cut using a manual rotary microtome (Thermo Scientific Shandon Finesse 325
Microtome). They were routinely processed for light microscopic examination and stained
with hematoxylin and eosin (H&E) or periodic acid-Schiff (PAS) without hematoxylin. H&E
stains were investigated at 40-100x magnification to evaluate histologic characteristics of the
tumour as well as strict intraocular tumour localisation versus tumour growth with extraocular
extension. H&E stained sections of liver, lung, spleen, sentinel lymph nodes and the
contralateral eye were scanned for (micro-) metastases at 100-400x magnification. PAS
stained sections without hematoxylin were investigated for vasculogenic mimicry and
extravascular matrix patterns using a polarised filter at 100-400x magnification.
For immunohistochemical melanoma detection we used a melanoma marker (Anti-Melanoma
antibody, HMB45+DT101+BC199, Abcam, Cambridge, UK; dilution 1:50). After
deparaffinisation and rehydration, heat-mediated antigen retrieval was performed with 0.01M
citrate buffer for 15 minutes. Washing steps were performed with TBS buffer (Tris buffer
[TBS: 50mM, pH 7.6]). After blocking with hydrogen peroxide the sections were incubated
with the primary antibody at 4°C overnight. The antigen-antibody-binding reaction was
visualised with an antigen detection system using horseradish peroxidase (HRP) and 3-amino9-ethylcarbazole (AEC) as the chromogen (MaxHomo Mouse on Mouse Polymer HRP
Detection Kit, Max Vision Biosciences Inc., Washington, USA).
Macrophage labelling in the tumour bearing eye was achieved using a F4/80 primary antibody
(Rat Anti-Mouse F4/80 Antibody, clone Cl:A3-1, AbD Serotec, Oxford, UK; dilution 1:100)
II Materials and Methods
30
and additionally with a GFP-antibody in CX3CR1+/GFP mice (GFP Rabbit IgG Polyclonal
Antibody Fraction, Life Technologies, Carlsbad, CA, USA; dilution 1:100). Tumour
vasculature and mean vascular density (MVD) were determined in sections stained with a
CD31-antibody (CD31, SZ31, Dianova, Hamburg, Germany, dilution 1:10). At 200x
magnification, vessels were counted within a 0.25mm2 grid in 1-3 areas according to Foss et
al. (Foss, Alexander et al. 1996). Staining of extracellular matrix (ECM) was additionally
performed with a collagen IV antibody (Collagen IV Antibody, Novus Biologicals, Littleton,
CO, USA dilution 1:100) and a laminin antibody (Laminin Antibody, Novus Biologicals,
Littleton, CO, USA, dilution 1:100). In flt-1/eGFP BAC mice all smaller host vessels were
additionally labelled by a GFP-antibody. Dual labelling in immunofluorescence microscopy
of CD31 and GFP allows for determination of the origin of endothelial-lined vessels. Dual
labeling of CD31 + Ki67 (Anti-Ki67, SP6, Abcam, Cambridge, UK, dilution 1:50) is
supposed to reveal active angiogenesis in immunofluorescence microscopy.
Tumour microvasculature was additionally labelled by Vascular Endothelial Cadherin (VECadherin (Phospho Tyr731) Antibody, Assaybiotech, Sunnywale, CA, USA; dilution 1:200)
with antigen retrieval by pepsin (3 minutes), overnight incubation of the primary antibody at
4°C, blocking with hydrogen peroxide, a secondary antibody incubation for one hour at room
temperature (Polyclonal Swine Anti-Rabbit Immunoglobulins, DakoCytomation, Glostrup,
Denkmark; dilution 1:200), and streptavidin one hour at room temperature (Streptavidinbiotinylated horseradish peroxidase complex, GE Healthcare, Amersham, Buckinghamshire,
U.K.; dilution 1:100). To distinguish the genuine ocular pigment from the immunoreaction we
visualised the latter with the red chromogen AEC (15 minutes at room temperature) instead of
a commonly used brown chromogen.
II Materials and Methods
31
Flow cytometry analysis
Ocular tumours of 20 CX3CR1+/GFP mice (young n=10 and old n=10) were dissected
microsurgically, pooled, respectively, and digested in 0.5mg/ml collagenase and 100µg/mg
DNAase I in RPMI medium for 40 minutes. The washed cells were filtered through a 100µm
nylon mesh to obtain a single cell suspension. To evaluate macrophage infiltration within
intraocular tumours, surface staining was performed with monoclonal antibodies against
characteristic leucocyte and macrophage receptors which were conjugated to different
fluorochromes: CD45 (Biolegend Inc., San Diego, CA, USA), CD11b (Biolegend), F4/80
(Biolegend), CD80 (Biolegend), CD163 (M-96, Santa Cruz Biotechnology, Santa Cruz, CA,
USA + conjugation kit, Abcam, Cambridge, UK), and Gr-1 (Biolegend). Controls were run as
single stains and unstained probes. Measurements were carried out on a flow cytometer
(LSRFortessa, BD, Franklin Lakes, NJ, USA) and results were analysed by using FlowJo
software (Tristar Inc. San Carlos, CA, USA).
Ex vivo imaging
In three flt-1/eGFP BAC mice, tumour-bearing and contralateral control eyes were enucleated
immediately after euthanasia. Eyes were kept in warm buffer (PBS) and shortly after
investigated using an Axio Zoom microscope (Carl Zeiss Microscopy GmbH, Jena,
Germany). Specimens were examined for GFP-positive vessels in areas of extraocular tumour
extension at x25 power magnification. Afterwards, the anterior parts of the eye were removed
and intraocular tumour parts in the posterior compartment of the eye were examined for GFPpositive vessels.
II Materials and Methods
32
Statistical analysis
Statistical analysis was performed with IBM SPSS Statistics 22.0 (IBM Corp, Armonk, NY,
USA). Evaluation of histological sections was performed by light microscopy by two
independent investigators. Number of infiltrating macrophages as well as the presence and
amount of collagen IV- and laminin-positive structures were analysed semi-quantitatively
from 0 to +++ by two independent examiners and a p-value of (MCH, MMK).
Tumour size was measured in mm2 at 40x magnification using ImageJ 1.45s (W Rasband,
NIH, USA). Tumour size and MVD of different study groups were evaluated by an analysis
of variance and a Post Hoc Test. Chi2 test and Pearson's correlation coefficient were used to
determine the relationship between different study groups and tumour growth characteristics.
Student’s t-test and ROC analysis was applied for interpretation of cytokine levels of in vitro
polarised macrophages. A p≤0.05 was considered as statistically significant and significance
level was indicated by using * for p ≤ 0.05.
Histology and immunohistochemistry of Hgf-Cdk4R24C mice eyes
12 eyes from 11 month old Hgf-Cdk4R24C mice were investigated and were compared with
three eyes from 10 months old C57Bl/6 mice. Step sections were stained with H&E and PAS,
and sections in-between were investigated by immunohistochemical staining. Primary
antibodies against Ki67 (proliferation marker), MART1 (melanocyte/melanoma marker),
CKpan (epithelial cell differentiation) and F4/80 (macrophages) were used. Prior to
immunohistochemistry, sections of these highly pigmented mice were bleached for better
visualisation of the red chromogen AEC.
33
III
Intravitreally injected HCmel12 melanoma cells serve as a
mouse model of tumour biology of intraocular melanoma
Marta M. Kilian1, Karin U. Loeffler1, Christiane Pfarrer2, Frank G. Holz1, Christian Kurts3,
Martina C. Herwig1
1
Department of Ophthalmology, University of Bonn, Bonn, Germany
2
Department of Anatomy, University of Veterinary Medicine Hannover, Hannover, Germany
3
Institute of Experimental Immunology, University of Bonn, Bonn, Germany
Current Eye Research
Informa Healthcare USA, Inc.
ISSN: 0271-3683 print / 1460-2202 online
DOI: 10.3109/02713683.2015.1004721
III Intravitreally injected HCmel12 melanoma cells serve as a mouse model
of tumor biology of intraocular melanoma
34
Abstract
Purpose: To establish a mouse model with histologic characteristics of uveal melanoma for
investigation of intraocular tumour biology of melanoma.
Methods: After injection of 1x105 of HCmel12 melanoma cells, a cutaneous melanoma cell
line, into the vitreous of CX3CR1+/GFP or C57Bl/6 mice (n=12), tumour growth patterns,
clinicopathological features, angiogenesis and metastatic behaviour were analysed by
histology (H&E, PAS without hematoxylin) and immunohistochemistry (HMB45/MART-1Ab, F4/80-Ab, GFP-Ab, VE-cadherin-Ab).
Results: HCmel12 cells formed intraocularly growing tumour masses which showed
histologic features of intraocular melanoma such as angiotropism, intratumoural endotheliallined vasculature, vasculogenic mimicry including prognostic significant extravascular matrix
patterns, and invasion by inflammatory cells, in particular macrophages. There was no
difference in tumour growth characteristics between CX3CR1+/GFP and C57Bl/6 mice. Five out
of ten mice proceeded to extrascleral tumour growth, and three of these developed metastases.
Conclusions: Intraocularly injected HCmel12 cells developed tumour masses with histologic
characteristics of aggressive melanoma similar to human uveal melanoma. Since
hematogenous dissemination to the liver was not observed, intravitreally injected HCmel12
cells do not qualify as a model for metastasizing intraocular melanoma. However, since the
eye represents a semi-closed compartment with access to constant blood supply, these
intraocular tumours represent a model for studies of isolated parameters in general tumour
biology of intraocular melanoma.
III Intravitreally injected HCmel12 melanoma cells serve as a mouse model
of tumor biology of intraocular melanoma
35
Introduction
Uveal melanoma is the most common primary intraocular tumour in the adult Caucasian
population. While local treatment of the primary tumour is very promising, the outcome
worsens dramatically when metastatic disease appears (Kujala, Makitie et al. 2003). Uveal
melanoma metastasises predominantly hematogenously to the liver and rarely to the lungs or
other organs. Metastases develop in up to 40% of patients within 10 years of initial diagnosis
(Singh, Shields et al. 2001).
In order to sustain solid growth the tumour has to ensure sufficient blood supply. By releasing
certain factors or chemokines and thereby influencing the microenvironment the tumour
utilises angiogenesis and other sources like co-option of pre-existing vessels or formation of
mosaic vessels (Hanahan and Weinberg 2011). Many growth characteristics concerning the
tumour’s blood supply are related to metastasis and thus to prognosis. Microvascular density
(MVD) has been found to be of prognostic value for uveal melanoma (Foss, Alexander et al.
1996; Makitie, Summanen et al. 1999). Intravascular ingrowth of uveal melanoma cells and
extraocular growth are also contributing factors to metastasis formation (Ly, Odish et al.
2010). Metastasis is further associated with the primary tumour exhibiting certain patterned
meshworks of fluid conducting non-endothelial channels described as extravascular matrix
patterns or vasculogenic mimicry (VM) (Folberg, Mehaffey et al. 1997). VM is rich of PASpositive extracellular matrix (ECM) with high expression levels of ECM related molecules
(e.g. matrix-metalloproteinases, VE-cadherin, VEGF-A) (Seftor, Hess et al. 2012).
Apparently, tumour microcirculation consists of a combination of angiogenic vessels, mosaic
vessels as well as ECM-rich VM meshworks (Hendrix, Seftor et al. 2003). Angiogenesis and
the tumour microenvironment are further influenced by factors released by tumour infiltrating
III Intravitreally injected HCmel12 melanoma cells serve as a mouse model
of tumor biology of intraocular melanoma
36
leucocytes (Sica and Mantovani 2012), in particular lymphocytes and macrophages. Tumour
growth favoring phenotypes of macrophages are preferably recruited into the tumour area
(Bronkhorst and Jager 2012). Increased numbers of tumour associated macrophages (TAM)
correlate - among other tumour characteristics - with high MVD and with patient’s death from
metastatic disease (Toivonen, Makitie et al. 2004). In turn, the microenvironment has a major
impact on primary tumour cells with regard to tumour progression and metastasis. Although
each tumour may be genetically predetermined to enter lymphatic and/or blood vessels for
dissemination (Nathanson 2003), successful metastases development is also affected by
molecular interactions between circulating tumour cells and the pre-metastatic site (Mendoza
and Khanna 2009). For human uveal melanoma certain receptors and factors accounting for
liver-specific metastases have been determined, including c-met, a receptor for hepatocyte
growth factor (HGF) /scatter factor (Bakalian, Marshall et al. 2008). Furthermore, several
genetic anomalies (e.g. monosomy 3, class 2 gene expression profile, GNAQ, GNA11, Bap1)
of uveal melanoma influencing tumour progression and metastasis have been identified
(Sisley, Rennie et al. 1990; Onken, Worley et al. 2004; Ehlers, Worley et al. 2008).
Nonetheless, characteristic mutations differ between uveal and cutaneous melanoma and even
among tumours themselves (Mehnert and Kluger 2012). However, uveal and cutaneous
melanomas do share fundamental characteristics in terms of tumour growth, inflammatory
cell
infiltration,
microenvironmental
stimuli
and
vascularisation.
Considering
the
aforementioned characteristics of uveal melanoma like hematogenous spread, characteristic
microcirculation, the inflammatory microenvironment and genetic attributes, a meaningful
mouse model should offer similar characteristics.
The aim of this study was to evaluate histopathologic growth and metastatic characteristics of
intravitreally injected HCmel12 melanoma cells. Evaluation emphasised the applicability of
III Intravitreally injected HCmel12 melanoma cells serve as a mouse model
of tumor biology of intraocular melanoma
37
HCmel12 melanoma cells as a mouse model for intraocular melanoma in terms of tumour
biology and metastasis.
Materials and Methods
Animals
Breeding and housing of CX3CR1+/GFP mice and C57Bl/6 mice was carried out and supervised
according to the Association of Research in Vision and Ophthalmology statement for the Use
of Animals in Ophthalmic and Vision Research. Six CX3CR1+/GFP mice and six C57Bl/6 mice
were examined at the age of 8-12 weeks, whereas two CX3CR1+/GFP mice and four C57Bl/6
mice of the same age were kept as negative controls (Table 1).
In CX3CR1+/GFP mice which are based on a C57Bl/6 strain, the transmembrane-receptor
CX3CR1 for CX3C chemokine fractalkine had been replaced by green fluorescent protein
(GFP) reporter gene. Hence, these mice exhibit an in vivo labelling of peripheral blood
monocytes, macrophages, subsets of NK and dendritic cells, as well as of retinal microglia by
fluorescence at 488nm (Jung, Aliberti et al. 2000). This innate fluorescent signal of monocytic
cells may allow their direct visualization by immunofluorescence microscopy.
Homozygous male CX3CR1GFP/GFP mice for breeding were kindly provided by Prof. C. Kurts,
Institute of Experimental Immunology, University of Bonn, Bonn, Germany. Female C57Bl/6
mice were crossbred with homozygous male CX3CR1GFP/GFP mice generating a heterozygous
CX3CR1+/GFP mice generation. C57Bl/6 mice were also used as controls since the original
III Intravitreally injected HCmel12 melanoma cells serve as a mouse model
of tumor biology of intraocular melanoma
38
B16LS9 mouse model for metastasizing intraocular melanoma was established in this mouse
strain (Diaz, Rusciano et al. 1999).
HCmel12 melanoma cell line
HCmel12 melanoma cells were generously provided by Prof. T. Tüting, Laboratory of
Experimental Dermatology, University of Bonn, Bonn, Germany. This murine cutaneous
melanoma cell line had been derived from an autochthonous cutaneous melanoma of an HgfCdk4 R24C mouse which spontaneously develops a spectrum of primary cutaneous melanomas
(Landsberg, Gaffal et al. 2010). HCmel12 cells of an early cell culture passage exhibit
particular characteristics like increased c-met expression, angiotropism and spontaneous
metastatic properties that may qualify them as a cell line for a potent model of metastasizing
intraocular melanoma. Furthermore, HCmel12 cells’ growth characteristics resemble those of
human uveal melanoma regarding angiotropism and VM, as well as the attraction of a high
number of macrophages (unpublished data).
The HCmel12 melanoma cell line was cultured in RPMI 1640 + Hepes medium supplemented
with 10% FCS, 1% Penicillin/Streptomycin, 1% NEAA, 1% Sodium-Pyruvate and 0.75% βMercaptoethanol. Cells were incubated at 37°C in a humidified atmosphere containing 5%
CO2 and grown to approx. 70% confluence. HCmel12 melanoma cells were cryo-conserved at
cell culture passage 8. After thawing, cells were cultured for another passage, trypsinised and
washed in PBS buffer before aliquots suspended in PBS buffer were injected into the eye.
III Intravitreally injected HCmel12 melanoma cells serve as a mouse model
of tumor biology of intraocular melanoma
39
Clinical procedures
The experimental group consisted of 12 mice; six mice were used as negative controls.
General
anaesthesia
comprised
Isofluran
induction
followed
by
intraperitoneal
Ketamin/Xylazin injection.
On day 0. in general anaesthesia, 1x105 HCmel12 melanoma cells suspended in 1µl PBS were
injected intravitreally into the right eye of both mouse strains, CX3CR1+/GFP mice and
C57Bl/6 mice. An automatic microsyringe injector (UltraMicroPump III, World Precision
Instruments, Sarasota, USA) with a 10µl microinjection syringe (701 ASRN, Hamilton, Reno,
USA) equipped with a 34-gauge needle (RN Needle, Hamilton, Reno, USA) was used for
injection. The needle was inserted approx. 1 mm posteriour to the limbus through the sclera
into the vitreous. Negative controls received 1µl of PBS (n=3, 1 CX3CR1+/GFP mouse, 2
C57Bl/6 mice), a sole needle stitch with no injecting agent (n=2, 1 CX3CR1+/GFP and 1
C57Bl/6 mouse) or no treatment at all (n=1, C57Bl/6 mouse). Negative controls may show
whether a noteworthy influx of immune cells was caused by the injection process of PBS or
the stitch alone. Tumour bearing eyes of CX3CR1+/GFP mice and C57Bl/6 mice were
enucleated in general anaesthesia on day 9 and the mice were sacrificed on day 42. Then
tissue samples from lungs, liver and spleen, as well as the contralateral eye and mandibular
lymph nodes were collected for further examination to verify metastatic routes and sites.
Histology
Specimens were submitted to 4% paraformaldehyde for 24 hours, dehydrated with alcohol in
ascending order and cleared with xylene. After embedding in paraffin, 5 µm thick serial
sections of the eye and five sections through the center of the other tissues were cut using a
manual rotary microtome (Thermo Scientific Shandon Finesse 325 Microtome). They were
III Intravitreally injected HCmel12 melanoma cells serve as a mouse model
of tumor biology of intraocular melanoma
40
routinely processed for light microscopic examination and stained with hematoxylin and eosin
(H&E) or periodic acid-Schiff (PAS) without hematoxylin. H&E stains were investigated at
40-100x magnification to evaluate histologic characteristics of the tumour as well as strict
intraocular tumour localization versus tumour growth with extraocular extension. Tumour
size was measured at 40x magnification using ImageJ 1.45s. H&E stained sections of liver,
lung, spleen, sentinel lymph nodes and the contralateral eye were scanned for metastases at
100-400x magnification. PAS stained sections without hematoxylin were investigated for VM
and extravascular matrix patterns using a polarised filter at 100-400x magnification.
For immunohistochemical melanoma detection we used a commercially available melanoma
marker (Anti-Melanoma antibody, HMB45+DT101+BC199, Abcam, Cambridge, UK;
dilution 1:50). After deparaffinization and rehydration, heat-mediated antigen retrieval was
performed with 0.01M citrate buffer for 15 minutes. Washing steps were performed with TBS
buffer (Tris buffer [TBS: 50mM, pH 7.6]). After blocking with hydrogen peroxide the
sections were incubated with the primary antibody overnight at 4°C. The antigen-antibodybinding reaction was visualised with an antigen detection system using horseradish
peroxidase (HRP) and 3-amino-9-ethylcarbazole (AEC) as the chromogen (MaxHomo Mouse
on Mouse Polymer HRP Detection Kit, Max Vision Biosciences Inc., Washington, USA).
Macrophage labelling was achieved using a F4/80 primary antibody (Rat Anti-Mouse F4/80
Antibody, clone Cl:A3-1, AbD Serotec, Oxford, UK; dilution 1:100) in both mouse strains
and additionally with a GFP-antibody in CX3CR1+/GFP mice (GFP Rabbit IgG Polyclonal
Antibody Fraction, Life Technologies, Carlsbad, CA, USA; dilution 1:100); tumour
microvasculature was demonstrated by Vascular Endothelial Cadherin (VE-Cadherin
(Phospho Tyr731) Antibody, Assaybiotech, Sunnywale, CA, USA; dilution 1:200) with
antigen retrieval by pepsin (3 minutes), overnight incubation of the primary antibody at 4°C,
III Intravitreally injected HCmel12 melanoma cells serve as a mouse model
of tumor biology of intraocular melanoma
41
blocking with hydrogen peroxide, a secondary antibody incubation for one hour at room
temperature (Polyclonal Swine Anti-Rabbit Immunoglobulins, DakoCytomation, Glostrup,
Denkmark; dilution 1:200), streptavidin one hour at room temperature (Streptavidinbiotinylated horseradish peroxidase complex, GE Healthcare, Amersham, Buckinghamshire,
U.K.; dilution 1:100) and visualization by AEC (15 minutes at room temperature).
The analysis was performed by light microscopic evaluation by two independent
investigators.
Results
We intended to evaluate intraocular growth and metastatic characteristics of HCmel12 cells
which represent a new murine cutaneous cell line originating from an autochthonous
melanoma in mice with a C57Bl/6 background. Therefore, six CX3CR1+/GFP mice and six
C57Bl/6 mice received an intravitreal tumour cell injection at the age of 8-12 weeks.
Enucleated eyes and potential organs for metastatic tumour localization were processed for
histological examination.
Histology of intraocular HCmel12 melanoma
Of twelve mice two mice (1 CX3CR1+/GFP and 1 C57Bl/6 mouse) deceased early in the
experiment and were excluded from statistics. The eyes of the remaining ten mice which had
been injected with HCmel12 cells comprised five specimens with intraocular tumour growth
III Intravitreally injected HCmel12 melanoma cells serve as a mouse model
of tumor biology of intraocular melanoma
42
strictly confined to the eye (Fig. 1a), while the other five specimens exhibited additionally
extraocular extension (Fig. 1b) (Table 1).
Table 1: Experimental groups of animals
Mouse
1
2
3
4
5
6
7
8
9
10
11
12
Genotype
Tumour size[mm2]
Extraocular Metastases
C57 Bl/6
C57 Bl/6
C57 Bl/6
C57 Bl/6
C57 Bl/6
C57 Bl/6†
CX3CR1+/GFP
CX3CR1+/GFP
CX3CR1+/GFP
CX3CR1+/GFP
CX3CR1+/GFP
CX3CR1+/GFP†
0.1805
1.4929
0.3169
0.2049
0.142
No
Yes
No
No
Yes
No
Yes
No
No
Yes
0.1353
0.0608
0.1179
0.6604
0.2727
Yes
No
Yes
Yes
No
No
No
No
Yes
No
†=deceased
All mice received an intravitreal injection of 1x105 HCmel12 melanoma cells.
Tumour bearing eyes were enucleated on day 9 and mice were euthanised on day
42.
One mouse (C57Bl/6) deceased shortly after intravitreal injection allowing for histological
evaluation of intraocular conditions immediately after tumour cell injection. In this mouse
HCmel12 cells were distributed uniformly within the posteriour compartment of the eye.
However, a few cells remained along the injection channel and outside intraocular structures
(Fig. 1c). In ten mice with tumour bearing eyes H&E sections revealed tumour growth mostly
around the optic nerve with weak pigmentation and occasional necrotic areas. Also,
intraocular tumours exhibited invasive properties with destruction of surrounding ocular
structures such as the retina and the choroid (n=7) (Fig. 1d+f). They eventually (n=5)
proceeded to extraocular growth, most likely along the injection channel by following the
weakest point along the sclera.
III Intravitreally injected HCmel12 melanoma cells serve as a mouse model
of tumor biology of intraocular melanoma
43
Figure 1: Ocular HCmel12 tumours, H&E
(a) Intraocular tumour growth (arrow), 40x magnification, CX3CR1+/GFP mouse; (b) Intraocular tumour
growth (arrow) with extraocular extension (arrowhead), 40x, C57Bl/6 mouse; (c) Intravitreally
injected HCmel12 cells distribute in the vitreous and along the injecting channel on day of injection,
100x, C57Bl/6 mouse; (d) Intraocular tumour with destructive growth characteristics and evenly
distributed microvasculature, 200x, C57Bl/6 mouse; (e) Intraocular tumour exhibiting angiotropism
(arrow), blood channels and lakes (arrowhead), 400x, CX3CR1+/GFP mouse; (f) Intraocular tumour
penetrates the retina and invades into the choroid, 200x, C57Bl/6 mouse.
Tumour cells grew towards and around retinal blood vessels (n=8) indicating angiotropism
(Fig. 1e). Solid tumours exhibited channels and lakes filled with blood (n=8) (Fig. 1e) which
were enclosed by melanoma cells. Intratumoural endothelial lined vasculature was present
(Fig. 1d) as well as co-option of retinal vessels (Fig. 2a). Further, H&E stains showed a
scattered tumour infiltration by inflammatory cells. By PAS without hematoxylin, PASpositive ECM-rich channels enclosed by melanoma cells were detected (Fig. 2b).
Extravascular matrix patterns were identified and ranged from straights (n=5), parallel vessels
III Intravitreally injected HCmel12 melanoma cells serve as a mouse model
of tumor biology of intraocular melanoma
44
(n=4), in some cases with interconnecting branches (n=2) to arcs (n=7), arcs with branches
(n=7) and loops (n=4) (Fig. 2c). However, interconnecting loops creating networks were not
observed.
Figure 2: Ocular HCmel12 tumours, sections stained with PAS without hematoxylin viewed with a
polarising microscope showing ECM-rich structures, CX3CR1+/GFP mouse
(a) Co-option of retinal vessels (arrow), 200x magnification; (b+c) ECM-rich meshwork considered as
vasculogenic mimicry with patterns like arcs with branches, parallels with interconnections or loops
(arrows) 100-200x.
Histologic characteristics of intraocular HCmel12 tumours showed no evident differences
between CX3CR1+/GFP mice and C57Bl/6 mice. Also, measured intraocular tumour size did
not differ statistically between CX3CR1+/GFP mice and C57Bl/6 mice (p=0.46) (Tab. 1).
Tumour size was statistically not correlated with extraocular extension (p=0.078).
In H&E sections, control mice injected with PBS or nothing, did not show any immunological
reactions to the injection process. The removed contralateral eye without tumour was
unremarkable in all investigated mice.
III Intravitreally injected HCmel12 melanoma cells serve as a mouse model
of tumor biology of intraocular melanoma
45
Immunohistochemistry of intraocular HCmel12 melanoma
In CX3CR1+/GFP mice the innate GFP signal was too weak for allowing direct evaluation by
immunofluorescence microscopy. However, labelling of macrophages using a F4/80- and a
GFP-antibody revealed positively stained cells within the retina, most likely representing
retinal microglia. Labelled cells were also found evenly distributed throughout intra- and
extraocular parts of the tumour; in intraocular parts especially near the retina or the choroid
(Fig. 3a). Some appeared as large cells with vacuoles or granula, others as cells with a small
cell body and protruding dendrites (Fig. 3b).
The microvasculature was positively stained by the VE-Cadherin antibody throughout the
tumours (Fig. 3c) but did not correlate with the characteristic ECM-rich patterns as seen in
PAS stains without hematoxylin.
Figure 3 Immunohistochemical staining of ocular HCmel12 melanoma and remaining retina
(asterisk), CX3CR1+/GFP mouse
(a) GFP-antibody labelling of retinal microglia and infiltrating tumour associated macrophages, 100x
magnification; (b) High power area indicated in box of Fig. 3a, 200x; (c) VE-cadherin-antibody
labelling of microvasculature, 400x
III Intravitreally injected HCmel12 melanoma cells serve as a mouse model
of tumor biology of intraocular melanoma
46
Metastases of HCmel12 melanoma
Distant micro- and macrometastases were diagnosed in H&E sections (Fig. 4a) as well as by
an immunohistochemical melanoma marker (HMB45/MART-1) in lungs (Fig. 4b) in three out
of ten examined mice. All of these mice exhibited previous extraocular tumour growth. No
mice with tumours confined strictly to the eye showed any metastatic colonization in distant
tissue. Statistically, the incidence of metastases was not correlated with tumour size (p=0.16)
nor with the presence of extravascular matrix patterns (p=0.33). However, because of the
small number of animals with metastases, this data must be interpreted carefully.
Metastatic involvement of the lymphnodes could not be unequivocally detected due to the
high cellularity of the lymph nodes and the unspecific immunohistochemical signal in this
tissue. However, the immunohistochemical labelling in the lungs was confined to the
metastases and interpreted as reliable. Metastases into the liver and spleen could not be
detected.
Figure 4: Pulmonary metastasis of HCmel12 melanoma
(a) Lung metastasis, H&E, 100x magnification, C57Bl/6 mouse; (b) Immunohistochemical staining for
HMB45/MART-1, 200x, CX3CR1+/GFP mouse
III Intravitreally injected HCmel12 melanoma cells serve as a mouse model
of tumor biology of intraocular melanoma
47
Discussion
In order to study the pathogenesis of human uveal melanoma several mouse models have been
developed including transgenic mice, intraocular inoculation with cutaneous melanoma cells
into wildtype mice, injection of human uveal melanoma cells into SCID mice or induction of
uveal proliferations by chemicals, radiation or viruses (Dithmar, Albert et al. 2000). Since
spontaneous primary uveal melanoma has not been described in wildtype mice yet, existing
mouse models are still subject to restrictions. Pigmented intraocular tumours arising in
transgenic mice were identified to be either of retinal pigment epithelium origin (Syed,
Windle et al. 1998; Albert, Kumar et al. 2004) or were small uveal tumours which failed to
metastasise to the liver (Kramer, Powell et al. 1998; Tolleson, Doss et al. 2005; Latendresse,
Muskhelishvili et al. 2007). Injection of human uveal melanoma cells into immunodeficient
mice also resulted in lacking metastatic disease (Mueller, Maniotis et al. 2002) and does not
allow the study of influences on tumour and host immunology. The best reasonable
compromise, and an established mouse model thus far, was created by injecting murine,
cutaneous B16LS9 melanoma cells into the vitreous of C57Bl/6 mice, generating an ocular
tumour with metastatic dissemination to the lungs and the liver. According to Diaz and
coworkers intraocular tumours were restricted to the eye at the time of enucleation, hence
metastatic dissemination to the lungs and liver occurred hematogenously since the eye lacks
lymphatic vessels (Diaz, Rusciano et al. 1999). Focussing further on hepatic metastases a
mouse model with PEDF null mice has been generated (Lattier, Yang et al. 2013). Another
group focussed on intraocular tumour growth of intraocularly injected B16 cells to investigate
the distribution of extravascular matrix patterns in intraocular melanoma. VM was found
along with endothelium-dependant vessels and with connection to the host’s circulation
III Intravitreally injected HCmel12 melanoma cells serve as a mouse model
of tumor biology of intraocular melanoma
48
(Chen, Zhang et al. 2009). These findings also show that the stroma compartment of the
choroid is not mandatory for ECM and VM since the tumour cells were directly injected into
the vitreous and not all tumours gained access to the murine choroid (Grossniklaus 1998).
In our model, we investigated ocular tumours and mestastatic behaviour of HCmel12
melanoma cells, which represent a new murine cutaneous cell line with certain characteristics
that may qualify them as a promising cell line for a metastasizing intraocular melanoma
model. HCmel12 melanoma cells succeeded in solid intraocular tumour growth and showed
properties of an aggressive melanoma, including metastatic spread to the lungs. There were
no differences in tumour growth, size or metastatic characteristics between inoculated
C57Bl/6 and CX3CR1+/GFP mice. Tumour size did not have an impact on extraocular
extension and metastasis. However, as this statistical data is based on a small number of
animals, further experiments are necessary to confirm this finding in a larger cohort.
Interestingly - despite angiotropic growth, VM and an increased c-met expression of
HCmel12 cells - none of the exclusively intraocular growing HCmel12 tumours succeeded in
colonization of distant tissue. On the other hand, tumours with extraocular extension
metastasised in 60% of cases preferably to the lungs, probably under participation of the
conjunctival lymphatic system. This inability of forming hematogenously dispersed
metastases may be due to failed vascular ingrowth of tumour cells or a lack of effective
molecular interactions between trafficking primary tumour cells within the blood system and
a potential pre-metastatic site like the liver. Thus, HCmel12 melanoma cells qualify very
restrictedly as a meaningful mouse model for metastasizing uveal melanoma. However,
focusing on intraocular tumour biology, HCmel12 melanoma cells show apparent similarities
to human uveal melanoma. Intraocular solid tumours revealed aggressive growth and
infiltration by inflammatory cells, particularly macrophages. Further, the tumour exhibited
III Intravitreally injected HCmel12 melanoma cells serve as a mouse model
of tumor biology of intraocular melanoma
49
angiotropic growth, angiogenesis and induced the formation of ECM-rich, PAS-positive
channels considered as VM. Microcirculation of HCmel12 tumours revealed formation of
prognostic significant ECM patterns which are known to be correlated with a class 2 gene
expression profile of uveal melanoma (Onken, Worley et al. 2004; Onken, Lin et al. 2005).
Isolated histopathologic factors like MVD, inflammatory cell infiltration, VM (including
extravascular matrix patterns) are no longer crucial for evaluating prognosis in human uveal
melanoma as gene expression profiling has been found to be a more meaningful indicator for
patient’s prognosis (Onken, Worley et al. 2012). However, these histologic prognostic
parameters are reflecting uveal melanoma growth characteristics and allow for their
evaluation.
Overall, intravitreally injected HCmel12 cells still represent murine melanomas of cutaneous
origin. Even though they may share a number of genetic and histopathologic properties with
human uveal melanoma, they still differ significantly in specific mutations (Hurst, Harbour et
al. 2003). Animal models are rarely capable of imitating an entire human pathologic
condition, but are able to reflect certain characteristics. Because the eye is lacking lymphatic
drainage but comprises a constant blood supply, intraocular tumours exhibit a
microenvironment of a virtual semi-closed system which can be used to study isolated
parameters in tumour biology.
As HCmel12 melanoma cells are preserved at a low cell culture passage, alterations in
genetics and growth characteristics - which may be caused by ongoing cell culture procedures
- are less probable. Intraocular HCmel12 growth resembles human intraocular melanoma in
many ways. Thus, tumours of intravitreally injected HCmel12 melanoma cells qualify as a
model for intraocular melanoma and allow studies on specific questions related to the
tumour’s microcirculation, including VM patterns.
III Intravitreally injected HCmel12 melanoma cells serve as a mouse model
of tumor biology of intraocular melanoma
50
With regard to intraocular melanoma, this study shows that intraocularly injected HCmel12
cells are a potent model for intraocular tumour biology allowing for analysis of tumour
angiogenesis, extravascular matrix patterns and microenvironmental influences of
inflammatory cells like macrophages.
References
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
Kujala, E., T. Makitie, and T. Kivela, Very long-term prognosis of patients with
malignant uveal melanoma. Invest Ophthalmol Vis Sci, 2003. 44(11): p. 4651-9.
Singh, A.D., C.L. Shields, and J.A. Shields, Prognostic factors in uveal melanoma.
Melanoma Res, 2001. 11(3): p. 255-63.
Hanahan, D. and R.A. Weinberg, Hallmarks of cancer: the next generation. Cell,
2011. 144(5): p. 646-74.
Foss, A.J., et al., Microvessel count predicts survival in uveal melanoma. Cancer Res,
1996. 56(13): p. 2900-3.
Makitie, T., et al., Microvascular density in predicting survival of patients with
choroidal and ciliary body melanoma. Invest Ophthalmol Vis Sci, 1999. 40(11): p.
2471-80.
Ly, L.V., et al., Intravascular presence of tumor cells as prognostic parameter in
uveal melanoma: a 35-year survey. Invest Ophthalmol Vis Sci, 2010. 51(2): p. 65865.
Folberg, R., et al., The microcirculation of choroidal and ciliary body melanomas. Eye
(Lond), 1997. 11 ( Pt 2): p. 227-38.
Seftor, R.E., et al., Tumor cell vasculogenic mimicry: from controversy to therapeutic
promise. Am J Pathol, 2012. 181(4): p. 1115-25.
Hendrix, M.J., et al., Vasculogenic mimicry and tumour-cell plasticity: lessons from
melanoma. Nat Rev Cancer, 2003. 3(6): p. 411-21.
Sica, A. and A. Mantovani, Macrophage plasticity and polarization: in vivo veritas. J
Clin Invest, 2012. 122(3): p. 787-95.
Bronkhorst, I.H. and M.J. Jager, Uveal Melanoma: The Inflammatory
Microenvironment. J Innate Immun, 2012.
Toivonen, P., et al., Microcirculation and tumor-infiltrating macrophages in choroidal
and ciliary body melanoma and corresponding metastases. Invest Ophthalmol Vis Sci,
2004. 45(1): p. 1-6.
Nathanson, S.D., Insights into the mechanisms of lymph node metastasis. Cancer,
2003. 98(2): p. 413-23.
III Intravitreally injected HCmel12 melanoma cells serve as a mouse model
of tumor biology of intraocular melanoma
14.
15.
16.
17.
18.
19.
20.
21.
22.
23.
24.
25.
26.
27.
28.
29.
30.
31.
32.
33.
51
Mendoza, M. and C. Khanna, Revisiting the seed and soil in cancer metastasis. Int J
Biochem Cell Biol, 2009. 41(7): p. 1452-62.
Bakalian, S., et al., Molecular pathways mediating liver metastasis in patients with
uveal melanoma. Clin Cancer Res, 2008. 14(4): p. 951-6.
Sisley, K., et al., Cytogenetic findings in six posteriour uveal melanomas: involvement
of chromosomes 3, 6, and 8. Genes Chromosomes Cancer, 1990. 2(3): p. 205-9.
Onken, M.D., et al., Gene expression profiling in uveal melanoma reveals two
molecular classes and predicts metastatic death. Cancer Res, 2004. 64(20): p. 7205-9.
Ehlers, J.P., et al., Integrative genomic analysis of aneuploidy in uveal melanoma.
Clin Cancer Res, 2008. 14(1): p. 115-22.
Mehnert, J.M. and H.M. Kluger, Driver mutations in melanoma: lessons learned from
bench-to-bedside studies. Curr Oncol Rep, 2012. 14(5): p. 449-57.
Jung, S., et al., Analysis of fractalkine receptor CX(3)CR1 function by targeted
deletion and green fluorescent protein reporter gene insertion. Mol Cell Biol, 2000.
20(11): p. 4106-14.
Diaz, C.E., et al., B16LS9 melanoma cells spread to the liver from the murine ocular
posteriour compartment (PC). Curr Eye Res, 1999. 18(2): p. 125-9.
Landsberg, J., et al., Autochthonous primary and metastatic melanomas in Hgf-Cdk4
R24C mice evade T-cell-mediated immune surveillance. Pigment Cell Melanoma Res,
2010. 23(5): p. 649-60.
Dithmar, S., D.M. Albert, and H.E. Grossniklaus, Animal models of uveal melanoma.
Melanoma Res, 2000. 10(3): p. 195-211.
Syed, N.A., et al., Transgenic mice with pigmented intraocular tumors: tissue of
origin and treatment. Invest Ophthalmol Vis Sci, 1998. 39(13): p. 2800-5.
Albert, D.M., et al., Effectiveness of 1alpha-hydroxyvitamin D2 in inhibiting tumor
growth in a murine transgenic pigmented ocular tumor model. Arch Ophthalmol,
2004. 122(9): p. 1365-9.
Kramer, T.R., et al., Pigmented uveal tumours in a transgenic mouse model. Br J
Ophthalmol, 1998. 82(8): p. 953-60.
Latendresse, J.R., et al., Two cases of uveal amelanotic melanoma in transgenic TyrHRAS+ Ink4a/Arf heterozygous mice. Toxicol Pathol, 2007. 35(6): p. 827-32.
Tolleson, W.H., et al., Spontaneous uveal amelanotic melanoma in transgenic TyrRAS+ Ink4a/Arf-/- mice. Arch Ophthalmol, 2005. 123(8): p. 1088-94.
Mueller, A.J., et al., An orthotopic model for human uveal melanoma in SCID mice.
Microvasc Res, 2002. 64(2): p. 207-13.
Lattier, J.M., et al., Host pigment epithelium-derived factor (PEDF) prevents
progression of liver metastasis in a mouse model of uveal melanoma. Clin Exp
Metastasis, 2013.
Chen, L., et al., A pilot study of vasculogenic mimicry immunohistochemical
expression in intraocular melanoma model. Oncol Rep, 2009. 21(4): p. 989-94.
Grossniklaus, H.E., Tumor vascularity and hematogenous metastasis in experimental
murine intraocular melanoma. Trans Am Ophthalmol Soc, 1998. 96: p. 721-52.
Onken, M.D., et al., Association between microarray gene expression signature and
extravascular matrix patterns in primary uveal melanomas. Am J Ophthalmol, 2005.
140(4): p. 748-9.
III Intravitreally injected HCmel12 melanoma cells serve as a mouse model
of tumor biology of intraocular melanoma
34.
35.
52
Onken, M.D., et al., Collaborative ocular oncology group report number 1:
prospective validation of a multi-gene prognostic assay in uveal melanoma.
Ophthalmology, 2012. 119(8): p. 1596-603.
Hurst, E.A., J.W. Harbour, and L.A. Cornelius, Ocular melanoma: a review and the
relationship to cutaneous melanoma. Arch Dermatol, 2003. 139(8): p. 1067-73.
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
53
IV
Impact of macrophages on tumour growth characteristics in a
murine ocular tumour model
Marta M. Kilian1, Karin U. Loeffler1, Christian Kurts2, T. Hoeller3 Christiane Pfarrer4, Frank
G. Holz1, Martina C. Herwig1
1
Department of Ophthalmology, University of Bonn, Bonn Germany
2
Institute of Experimental Immunology, University of Bonn, Bonn, Germany
3
Institute for Medical Biometry, Informatics and Epidemiology, University of Bonn, Bonn,
Germany
4
Department of Anatomy, University of Veterinary Medicine Hannover, Hannover, Germany
Manuscript in preparation
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
54
Abstract
Purpose: Tumour associated macrophages (TAM), extravascular matrix patterns, and
advanced patients’ age are associated with a poor prognosis in e.g. uveal melanoma. We
concentrated on specific mechanisms of TAM under the influence of age to study the relation
between the tumour microenvironment and tumour growth characteristics.
Methods: Ninety five CX3CR1+/GFP mice (young 8-12 weeks, old 10-12 months) received an
intravitreal injection of 1x105 HCmel12 melanoma cells. Subgroups were either systemically
macrophage-depleted by Clodronate liposomes (n=23) or received melanoma cells, which
were pre-incubated with the supernatant of M1- or M2-polarised macrophages (n=26). Eyes
were processed histologically/immunohistochemically (n=75), or for flow cytometry (n=20)
to analyse tumour size, mean vascular density (MVD), extravascular matrix patterns,
extracellular matrix (ECM) and the presence/polarisation of TAM.
Results: Extravascular matrix patterns were more frequently present in untreated old
compared to young mice (p=0.024) as well as in untreated mice of both age groups compared
to macrophage-depleted mice (p=0.014). Independent from age, M2-conditioned tumours
showed more TAM (p=0.001), increased ECM levels (p=0.024) and a higher MVD (p=0.02)
than M1-conditioned tumours. Flow cytometry showed a larger proportion of M2
macrophages in old than in young mice.
Conclusions: The results indicate that old mice represent a more suitable tumour model
compared with young mice since the histologic tumour pattern better resembles human
tumours. TAM appear to be responsible for a more aggressive tumour phenotype. Tumour
favouring and pro-angiogenic effects can directly be attributed to a M2-dominated
microenvironment rather than to age-dependent factors alone. However, an aged
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
55
immunoprofile with an increased number of M2 macrophages may provide a tumourfavouring basis.
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
56
Introduction
Histologic tumour characteristics such as angiogenesis, vascular-like structures which are rich
of PAS positive extracellular matrix (ECM), organised into fluid conducting, but nonendothelial channels and termed extravascular matrix patterns or vasculogenic mimicry as
wells as inflammatory cells, in particular tumour associated macrophages (TAM) can be
found in various tumours, such as uveal melanoma, cutaneous melanoma, breast cancer and
others (Hendrix, Seftor et al. 2003; Mantovani, Marchesi et al. 2007; Allavena, Sica et al.
2008). In UM, outcome is associated with these characteristics and also correlates with
patients’ age, tumour size, and extraocular extension (Foss, Alexander et al. 1996; Folberg,
Mehaffey et al. 1997; Shields, Kaliki et al. 2013). Histopathologic factors are no longer
crucial for prognosis, since chromosomal analysis and genetic profiling were found to be
reliable indicators for patients’ outcome (Onken, Worley et al. 2012; Field and Harbour
2014). However, those histologic parameters reflect tumour growth characteristics and allow
for their evaluation in animal models.
The tumour microenvironment is characterised by a synergistic interplay between the tumour
cells, the stroma and ECM, different inflammatory cell types, and soluble factors such as
cytokines. Interactions between the tumour and the microenvironment may orchestrate
tumour progression and metastasis (Catalano, Turdo et al. 2013) and a more detailed
understanding of these interactions might help to interfere or even prevent these events. TAM
are a major component of the inflammatory infiltrate in solid tumours. High numbers of TAM
and their specific microenvironment correlate statistically with poor prognosis in different
tumours (Allavena, Sica et al. 2008). In uveal melanoma, they are further associated with
monosomy 3 and several histopathologic factors such as high mean vascular density (MVD)
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
57
and extravascular matrix patterns/ VM, and thus with a prognostically aggressive tumour
phenotype (Folberg, Mehaffey et al. 1997; Makitie, Summanen et al. 2001; Hendrix, Seftor et
al. 2003; Toivonen, Makitie et al. 2004; Maat, Ly et al. 2008). TAM functional polarisation
may range within a broad spectrum from pro-inflammatory, tumouricidal M1 macrophages to
anti-inflammatory, tumour-favoring M2 macrophages (Jager, Ly et al. 2011). Cytokines
produced by tumour cells and other cells in the tumour microenvironment determine whether
TAM become M1 or M2 (Arnold, Henry et al. 2007). In response to the tumour
microenvironment TAM tend to specialise predominantly into the M2-phenotype with proangiogenic and tumour-favoring properties (Mantovani, Sozzani et al. 2002). Also, a high
M2/M1 ratio in UM was shown to be related to worse prognosis and to histologic negative
prognostic parameters (Herwig, Bergstrom et al. 2013). As the microenvironment and the
capability of innate immunity changes with increasing age macrophages shift their secretory
profile towards a M2-profile (Gomez, Nomellini et al. 2008). Macrophages from geriatric
mice are also particularly sensitive to signals that promote their M2-polarisation, which in
turn may promote tumour development (Jackaman, Radley-Crabb et al. 2013). Thus, the
tumour microenvironment and advanced age have an influence on TAM polarisation.
As macrophages play a pivotal role in the pathogenesis of several tumours and orchestrate the
tumour microenvironment, we investigated their influence on tumour biology in a murine
ocular tumour model. Herein, the tumour exhibits histologic features such as angiotropism,
angiogenesis, extravascular matrix patterns/ VM and inflammatory cell infiltration which
allow to study the intraocular tumour microenvironment. The latter can be further influenced
by systemic drug application or intraocular injections (Kilian, Loeffler et al. 2015). In our
study, we focussed on histopathologic factors with regard to tumour size and different forms
of vascularisation. The general role of macrophages was examined in mice depleted of
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
58
macrophages in comparison to untreated mice. In addition, in order to address the impact of
M1- or M2-polarised macrophages, we investigated intraocular tumours of M1- or M2conditioned melanoma cells. To study the influence of age on tumour growth patterns and
macrophage invasion, the experiments were conducted both in young and in old mice.
Materials and Methods
Animals
Breeding, housing and investigations of CX3CR1+/GFP mice were carried out and supervised
according to accepted standards of the Policy on Humane Care and Use of Laboratory
Animals of the National Institutes of Health (NIH), Bethesda, MD, USA.
In CX3CR1+/GFP mice the transmembrane-receptor CX3 CR1 for CX3C chemokine fractalkine
is replaced by green fluorescent protein (GFP) reporter gene. Hence, these mice exhibit GFP
expression in peripheral blood monocytes, subsets of NK, dendritic cells and retinal microglia
as well as macrophages, in which we were particularly interested (Jung, Aliberti et al. 2000).
We studied intraocular tumours of 95 CX3CR1+/GFP mice in two different age groups. Young
mice were examined at the age of 8-12 weeks; old mice were 10-12 months of age. In the first
experiment, we investigated the age-dependent influence of TAM in 49 CX3CR1+/GFP mice.
Macrophages were depleted systemically by repeated intraperitoneal injection of Clodronate
liposomes in young (n=13) and old (n=10) mice and compared to untreated mice (young
n=15, old n=11) (Tab. 1). In the second experiment, the impact of a micromilieu dominated
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
59
by M1 or M2 macrophages on tumour growth patterns was investigated in another cohort of
26 CX3CR1+/GFP mice (young M1: n=7, M2: n=7; old M1: n=6, M2: n=6) (Tab. 1).
Tumours of 10 young and 10 old CX3CR1+/GFP mice were examined in regard to number of
inflammatory cells, macrophages and their expression of the M2 marker CD163 by flow
cytometry analysis (Tab. 1).
Table 1: Number of animals for macrophage (MФ) depletion and polarisation experiment as well as
flow cytometry analysis
Experiment
Young mice
(8-12 weeks)
Old mice
(10-12 months)
MФ depletion
MФ polarisation
FACS
MФ
Presence
MФ
Depletion
M1conditioned
tumours
M2conditioned
tumours
n= 15
n= 13
n= 7
n= 7
n= 10
n= 11
n= 10
n= 6
n= 6
n= 10
Mouse model of intraocular melanoma
According to a previously established mouse model of metastasizing ocular melanoma (Diaz,
Rusciano et al. 1999), we created a modified mouse model of intraocular melanoma to study
tumour biology (Kilian, Loeffler et al. 2015). In our model, 1x105 HCmel12 cells (a murine
cutaneous melanoma cell line) were injected intravitreally into the right eye of each mouse in
all study groups on day 0 under general anaesthesia. Mice were sacrificed on day 9 post
intravitreal injection, the right eye was enucleated and processed for histologic or
immunohistochemical examination or for flow cytometry analysis.
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
60
Macrophage depletion (experiment 1)
For the first experiment, macrophage-depleted mice were studied in comparison to untreated
mice. Liposome-encapsulated dichloromethylene diphosphonate (Cl2MDP; Clodronate,
ordered through clodronateliposomes.org) was used to systemically deplete blood monocytes
and macrophages at a dosage of 0.1mg/g body weight (van Rooijen and van KesterenHendrikx 2003). Mice received repeated intraperitoneal injections of Clodronate liposomes on
days -6, -3, 0 (day of intravitreal tumour cell injection) and day 3. To evaluate macrophage
depletion, liver sections of Clodronate-treated and untreated mice were stained
immunohistochemically with a GFP-antibody (GFP Rabbit IgG, Life Technologies, Carlsbad,
CA, USA; dilution 1:100) or F4/80-antibody (F4/80 Antibody, Cl:A3-1, AbD Serotec,
Oxford, U.K.; dilution 1:100). Accordingly, HCmel12 melanoma cells were cultured in vitro
in standard medium or in supernatant of unpolarized macrophages for 20 hours. Then, cell
proliferation rates were compared by a BrdU cell proliferation assay (BrdU Cell Proliferation
Assay, Milipore, Billerica, MA; USA).
Macrophage polarisation (experiment 2)
For the second experiment, HCmel12 melanoma cells were exposed in vitro to the supernatant
of polarised macrophages prior to intravitreal injection. Murine macrophages were cultured in
medium without fetal calf serum (FCS) for two hours prior to in vitro polarisation to avoid
any impact of cytokines or chemokines within the FCS. Macrophages were treated with
different cytokines and lipopolysaccharide (LPS) to obtain different polarised phenotypes
according to a modified protocol (Mosser and Zhang 2008). IFNγ (200ng/ml) for M1macrophages and Interleukin 4 (IL-4) (20ng/ml) for M2 macrophages were added to the
serum-free medium in order to polarise macrophages. After 10 hours of incubation, LPS
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
61
(100ng/ml) was supplemented for each macrophage type for a total incubation time of 24
hours. Polarisation status of unfixed macrophages was verified by using a multiple cytokine
ELISA comprising the inflammatory cytokines IL-6, IL-10. IL-12, IL-23, TNFα and IFNγ
(Multi-Analyte ELISArrayTM Kit, Mouse Mix-N-Match, Qiagen, Venlo, Netherlands). M1specific expression of inflammatory cytokines comprises characteristic levels of IL-6high, IL10low, IL-12high, IL-23high, TNFαhigh and IFNγhigh (M2-specific expression levels vice versa)
(Mantovani, Sica et al. 2004). Phenotypical macrophage polarisation status of 4% PFA-fixed
cells was examined by immunocytology with a double staining kit (MultiVision Polymer
Detection System, Thermo Scientific, Waltham, MA, USA) using a F4/80- (for all
macrophages) and CD163- (for M2 macrophages only; CD163, M-96, Santa Cruz
Biotechnology, Santa Cruz, CA, USA) primary antibody. To examine the impact of a M1- or
M2-conditioned tumour microenvironment, HCmel12 melanoma cells were treated with
serum-free medium for two hours and afterwards exposed to the supernatant of M1- or M2polarised macrophages for 20 hours, respectively. These M1- or M2-conditioned HCmel12
melanoma cells were then injected intravitreally as described above. Cell proliferation rates
and growth characteristics of these M1- or M2-conditioned HCmel12 melanoma cells were
then compared in vitro using a BrdU proliferation assay and in vivo after intravitreal injection.
Histology and immunohistochemistry
Processing of histologic specimens of both experiments was carried out as described before
(Kilian, Loeffler et al. 2015). Briefly, in hematoxylin-eosin (H&E) stained paraffin sections,
tumour size was measured in mm² at 40x magnification using ImageJ 1.45s (W Rasband,
National Institute of Health (NIH), USA); vascular characteristics like angiotropism, cooption of retinal vessels and intratumoural angiogenesis were evaluated at 200x
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
62
magnification. Sections stained with periodic acid-Schiff reaction (PAS) without hematoxylin
were investigated for analysis of ECM-rich channels/vasculogenic mimicry along with their
prognostic significant extravascular matrix patterns. For determination of the MVD, sections
were stained with a CD31-antibody (CD31, SZ31, Dianova, Hamburg, Germany, dilution
1:10). At 200x magnification, vessels were counted within a 0.25mm2 grid in 1-3 areas
according to Foss et al. (Foss, Alexander et al. 1996). Staining of ECM was additionally
performed with a collagen IV antibody (Collagen IV Antibody, Novus Biologicals, Littleton,
CO, USA dilution 1:100) and a laminin antibody (Laminin Antibody, Novus Biologicals,
Littleton, CO, USA, dilution 1:100). To distinguish the genuine ocular pigment from the
immunoreaction we visualised the latter with the red chromogen AEC. Macrophage
infiltration of the eye and of ocular tumours was verified by a GFP-antibody. Number of
infiltrating macrophages as well as the presence and amount of collagen IV- and lamininpositive structures were analysed semi-quantitatively from 0 to +++ by two independent
examiners (MCH, MMK).
Flow cytometry analysis
Ocular tumours of 10 young and 10 old mice were dissected microsurgically, pooled,
respectively, and digested in 0.5mg/ml collagenase and 100µg/mg DNAase I in RPMI
medium for 40 minutes. The washed cells were filtered through a 100µm nylon mesh. Surface
staining was performed with monoclonal antibodies for CD45 (Biolegend Inc., San Diego,
CA, USA), CD11b (Biolegend), F4/80 (Biolegend), CD80 (Biolegend), CD163 (M-96, Santa
Cruz Biotechnology, Santa Cruz, CA, USA + conjugation kit, Abcam, Cambridge, UK), and
Gr-1 (Biolegend). Controls were run as single stains and unstained probes. Measurements
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
63
were carried out on a flow cytometer (LSRFortessa, BD, Franklin Lakes, NJ, USA) and
results were analysed by using FlowJo software (Tristar Inc. San Carlos, CA, USA).
Statistical analysis
Statistical analysis was performed with IBM SPSS Statistics 22.0 (IBM Corp, Armonk, NY,
USA). Tumour size in mm2 and MVD of different study groups were evaluated by an analysis
of variance and a Post Hoc Test. Chi2 test and Pearson's correlation coefficient were used to
determine the relationship between different study groups and tumour growth characteristics.
Student’s t-test and ROC analysis was applied for interpretation of cytokine levels of in vitro
polarised macrophages. Comparison of in vitro melanoma cell proliferation between
differently treated cells was verified by student’s t-test. A p≤0.05 was considered as
statistically significant and significance level was indicated by using * for p ≤ 0.05.
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
64
Results
Macrophage depletion model (experiment 1)
All 49 CX3CR1+/GFP mice that received an intravitreal HCmel12 tumour cell injection
developed an intraocular tumour. Inflammatory cell infiltration was examined on H&E
stained sections as well as immunohistochemically. Intraocular tumours of untreated mice
exhibited high macrophage recruitment at an even distribution throughout the tumour and
within the retina, often associated with vascular structures. Apart from two very small
tumours, all tumours exhibited angiotropism, co-option of retinal vessels and formed own
blood conducting structures. Immunohistochemical staining for CD31 indicated endotheliumlined vasculature in the tumours. MVD revealed no significant differences between the study
groups of different age and macrophage status (p=0.542). In addition to endothelium-lined
vessels, specific extravascular matrix patterns were present in all tumours, including areas
with PAS-positive straights, parallels with and without cross-links, arcs with and without
branching, and loops. Since Folberg et al. showed PAS-positive parallels with cross-links,
loops and networks to be of prognostic value in choroidal melanoma (Folberg, Mehaffey et al.
1997), we focused on these three extravascular matrix patterns (see below).
Intraperitoneal injection of Clodronate liposomes led to systemic macrophage depletion which
could be verified in immunohistochemical stains of the liver: while untreated mice exhibited
frequent F4/80-posititive staining of Kupffer cells, Clodronate liposome-treated mice of both
age groups lacked positively stained cells (Supplementary Fig. S1).
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
65
Supplementary Figure S1: Experiment 1, systemic macrophage status confirmed by
immunohistochemical staining for F4/80 (red), x200 magnification. (A) Liver section of untreated
mouse; (B) Liver section of macrophage-depleted mouse.
Intraocular tumours of untreated old mice exhibited more frequently extravascular matrix
patterns than tumours of untreated young mice (p=0.024) when analysed in PAS stained
sections without hematoxylin (Fig. 1). In tumours of untreated mice of both ages these
patterns were significantly more frequent than in systemically macrophage-depleted mice of
both age groups (p=0.014). When subdivided into young and old mouse eyes, this effect was
much more pronounced in old mice (pyoung=0.34, pold=0.012).
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
66
Figure 1: Experiment 1, ocular tumours of young (A-B), old (C-D) and old macrophage-depleted
(E-F) mice, x100 magnification; (A,C,E) Sections stained with H&E giving an overview of the
tumour with solid tumour growth, angiotropism and angiogenesis; (B,D,F) sections stained with PAS
without hematoxylin showing extravascular matrix pattern (arrows); x100 magnification.
Prognostically significant extravascular matrix pattern were more frequently found in untreated old
mice in comparison to untreated young (p=0.024) as well as between untreated mice of both age
groups in comparison with macrophage-depleted mice (p=0.014).
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
67
Collagen IV staining labelled endothelium dependent vessels, extravascular matrix patterns as
well as other ECM-rich structures in the tumour. However, these collagen IV-positive patterns
did not exactly represent the patterns seen with PAS w/o hematoxylin. There was no
significant difference in collagen IV expression between the four study groups (p=0.213).
However, aged mice showed a tendency of having more collagen IV positive structures
compared to the young (p=0.074). Intraocular tumour size [in mm2] was statistically not
correlated with macrophage status or age (p=0.529). The presence of prognostic significant
extravascular matrix patterns was also not correlated with tumour size (p=0.611). However, in
vitro melanoma cell proliferation was increased significantly in cells which were incubated in
macrophage supernatant in comparison to cells cultured in standard medium (p=0.005) (Fig.
2A).
Macrophage polarisation model (experiment 2)
Macrophage polarisation in vitro
Macrophage polarisation was functionally verified and confirmed by a multiple cytokine
ELISA as described in methods. Levels of IL-12 and IL-23 were significantly higher in M1than in M2-polarised macrophages (pIL-12=0.043, pIL-23=0.012) and IL-10 levels were
significantly higher in M2 macrophages (pIL-10=0.008) (Supplementary Fig. S2). Levels of IL6, TNFα and IFNγ levels were exceedingly high and could not be used for statistical analysis.
IFNγ levels were not evaluated for M1 macrophages due to their initial polarisation with this
cytokine. However, IFNγ level within the M2-polarised culture was evidently low, in
agreement with reported M2 characteristics. Further, immunocytological dual labelling with
the M2 marker CD163 and the murine macrophage marker F4/80 revealed increased numbers
of CD163 labelled cells in the M2-polarised macrophage cell culture compared to the M1-
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
68
polarised cell culture (ratio CD163:F4/80 in M1-polarised culture 4:10; in M2-polarised
culture 7:10) (Supplementary Fig. S3). The combination of the functional characterization by
the multiple cytokine ELISA and phenotypical staining suggests a reliable in vitro
polarisation of macrophages.
Supplementary Figure S2: Experiment 2, multiple cytokine ELISA of macrophage polarisation
in vitro; M0: unpolarised macrophages (MФ), M1: MФ polarised with IFNγ+LPS, M2: MФ polarised
with IL-4+LPS, MM: HCmel12 melanoma cells, MM+M0 Co: HCmel12 melanoma cells cultured in
M0 supernatant, Neg: negative; characteristic cytokine levels for M1- and M2-polarised MФ with
significant differences in IL-10. IL-12, IL-23 expression, IL-6 and TNFα levels were exceeding
measurable dimension for M1- and M2-polarised MФ, IFNγ was also exceeding measurable
dimension for M1 MФ, but was evidently low for M2 MФ. HCmel12 melanoma cells expressed no
reasonable levels of investigated cytokines, however, after co-incubation with M0 supernatant
elevated levels of IL-6 and TNFα were measured.
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
69
Supplementary Figure S3: Experiment 2, immunocytochemistry of in vitro polarised
macrophages. Ratio M2 macrophage marker CD163 (blue) / pan-macrophage marker F4/80 (red).
(A) Macrophages after M1 polarisation (ratio 0.42 with predominantly round morphology);
(B) Macrophages after M2 polarisation (ratio 0.74 with predominantly dendritic morphology).
Macrophage conditioned melanoma cells in vivo and in vitro
Mice having received an intravitreal injection of M2-conditioned HCmel12 melanoma cells
exhibited a higher degree of inflammatory cell infiltration in ocular (retina and uvea) and
tumoural structures than mice with M1-conditioned tumours. Accordingly, increased levels of
GFP-labelled macrophages were found in tumours and retinas of mice with M2-conditioned
tumours compared to M1-conditioned tumours (p=0.001) (Fig. 3). Comparing M1- and M2conditioned tumours, age did not have an impact on the degree of inflammatory cell
infiltration of the tumour bearing eye (p=0.747).
In all mice, angiotropism, co-option of retinal vessels and intratumoural blood conducting
structures were identified immunohistochemically and by H&E staining. Mice which had
received an intravitreal injection of M2-conditioned HCmel12 melanoma cells exhibited a
statistically higher MVD compared to mice with M1-conditioned tumours (p=0.02). Age did
not have an impact on the MVD (p=0.132). Immunohistochemically, collagen IV-staining
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
70
was detected in all tumours. The staining reaction was found predominantly at tumour borders
and mostly sparing the central area of tumours (Fig. 3). Collagen IV-positive structures within
the tumour were increased in M2-conditioned tumours compared to M1-conditioned tumours
(p=0.024) but were not age dependent (p=0.84). In PAS sections without hematoxylin,
extravascular matrix patterns were not unequivocally evaluable and were thus not statistically
evaluated. Tumour size was not correlated with previous M1- or M2-conditioning of
HCmel12 cells, nor with age (p=0.179). ). However, in vitro cell proliferation was
significantly increased in melanoma cells which were previously incubated in M2-polarised
supernatant in comparison to cells cultured in supernatant of M1-polarised macrophages
(p=0.005) (Fig. 2B).
Figure 2: BrdU cell proliferation assay of HCmel12 melanoma cells (MM) which were cultured in
standard medium (standard), supernatant of unpolarized macrophages (M0) or of polarized
macrophages (M1, M2). Melanoma cells cultured in unpolarized macrophage supernatant (M0)
showed a significantly higher proliferation rate (p=0.005) than cells cultured in standard medium
(standard) (A). Melanoma cells cultured in supernatant of M2-polarized macrophages (M2) exhibited
a significantly higher proliferation rate compared to cells cultured in supernatant of M1-polarized
macrophages (M1) (p=0.005).
IV Impact of macrophages on tumour growth characteristics in a murine ocular tumour model
71
Figure 3: Experiment 2, ocular tumours of M1- (A-D) and M2-conditioned tumour cells (E-H), H&E and immunohistochemical
staining, x100 magnification; significant differences between M1- and M2-conditioned tumours regarding microvessel density (MVD,
p=0.02), collagen IV expression (extracellular matrix (ECM), p=0.024) and level of macrophage infiltration (GFP, p=0.001).
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
72
Flow cytometry analysis
Pooled ocular tumours of young (n=10) and old (n=10) CX3CR1+/GFP mice were investigated
for infiltrating leucocytes (CD45). The fraction of CX3CR1 GFP positive and F4/80 positive
macrophages was subclassified into bone marrow derived infiltrating macrophages (F4/80
positive, GFPhigh, Gr-1high) and resident macrophages (microglia) (F4/80 positive, GFPhigh, Gr1low) as well as M1 macrophages (CD80high, CD163low) and M2 macrophages (CD80intermediate,
CD163high). In pooled intraocular tumours 7.44% of cells in young and 7.1% of cells in old
mice were CD45 positive leucocytes. The majority of these (70.43% in young, 72.97% in old
mice) were CD11b positive phagocytes of which 79.13% (in young) and 74% (in old) were
found to be GFP positive cells (CX3CR1). Almost all GFP positive cells were macrophages
(F4/80 positive). Old mice showed a slight majority of CD163 positive cells (M2 marker)
when compared to young mice. However, noteworthy differences were found between bone
marrow recruited macrophages (Gr-1 positive) infiltrating the tumour and resident
macrophages (Gr-1 negative) with regard to age. Young mice exhibited increased levels of
Gr-1 positive macrophages when compared to the old (young 73.27%, old 62%, p=0.004)
(Supplementary Fig. S4). Monocytes which are recruited into the tumour via the blood
circulation migrate into the tumour as unpolarised phenotypes and proliferate into
macrophages. Depending on the existing tumour microenvironment, they are polarised into
tumouricidal M1 or tumour-favoring M2 macrophages. In our study, bone marrow derived
and infiltrating macrophages (Gr-1 positive) exhibited more frequently M2 markers (CD163)
in old mice than in their younger counterparts (p=0.08) (Fig. 4). This suggests a M2dominated tumour microenvironment in old mice (Tab. 2).
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
CD45
Young mice
Old mice
7.44
7.10
70.43
72.97
79.13
74.00
35.77
33.90
98.67
97.70
30.20
31.90
73.27
62.00
99.83
99.50
32.47
37.73
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
CD45 CD11b
CD45 CD11b GFP
CD45 CD11b GFP F4/80
CD45 CD11b GFP F4/80 CD80
high
CD45 CD11b GFP F4/80 CD163
CD45 CD11b GFP F4/80 Gr-1
high
CD45 CD11b GFP F4/80 Gr-1 CD80
high
CD45 CD11b GFP F4/80 Gr-1 CD163
high
73
Supplementary Figure S4: Flow cytometry analysis of surface markers of tumour associated
inflammatory cells [in%], in particular macrophages, in young (n=10) and old (n=10) mice.
45,00
+
Percentage of CD45 CD11b GFP Gr-1 cells
M2 Marker CD163
+
+
40,00
35,00
+
37,73
30,00
32,47
25,00
20,00
Young mice
Old mice
Figure 4: Flow cytometry analysis of tumour associated inflammatory macrophages
(CD45+CD11b+GFP+Gr-1+) in intraocular melanoma regarding their expression of the M2 marker
CD163. In old mice, these bone marrow derived macrophages exhibited more frequently a M2
phenotype with elevated levels of CD163high macrophages than their younger counterparts (p=0.08).
74
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
Table 2: Overview of tumour characteristics comparing untreated young and untreated old mice.
Young mice
Old mice
Measurement
p-value
0.68 ± 0.048
0.88 ± 0.035
Vessel/mm2
0.138
46.66 %
90.9 %
Presence yes/no
0.024
1.65
2.26
φ score (0-3)
0.074
Intratumoural CD45+ cells
7.44 %
7.1 %
Percentage
0.11
Inflammatory Gr-1+ cells
73.27 %
62.0 %
Percentage
0.025
CD163+ M2 macrophages
32.47 %
37.73 %
Percentage
0.087
MVD (mean)
Extravascular matrix pattern
Collagen IV
Tumours in old mice have a more aggressive phenotype and better resemble human malignancies in
terms of higher MVD, presence of prognostically significant extravascular matrix pattern, amount of
collagen IV, intratumoural leucocytes, bone marrow derived and tumour infiltrating macrophages and
M2 polarised macrophages.
Discussion
Certain tumour characteristics which are part of the tumour microenvironment appear to
represent general features of tumour biology. Mouse models allow to study such features,
including immunologic aspects or different forms of vascularisation (Chen, Zhang et al. 2009;
Kilian, Loeffler et al. 2015). In our mouse model, cutaneous melanoma cells are injected into
the mouse eye. Resulting tumours exhibit histologic features such as angiotropism, VM/
extravascular matrix patterns, and inflammatory cell infiltration. Since the eye represents a
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
75
semi-closed system with access to blood vessels (but no lymphatic vessels), the intraocularly
injected tumour cells may grow inside an immune privileged compartment towards a tumour
mass and develop a microenvironment which can be iatrogenically manipulated by systemic
or intraocular injections (Kilian, Loeffler et al. 2015). Thus, our model provides the
opportunity to address hypotheses on general tumour biology including vascularisation and
matrix remodelling. As particularly macrophages play a pivotal role in various tumours
including UM, we used our established mouse model to specifically study their influence on
intraocular growth characteristics.
The role of TAM
To determine the impact of macrophages (regardless of their polarisation) we compared
intraocular melanoma growth characteristics in vivo in systemically macrophage- depleted
mice in comparison to untreated mice. Depletion of macrophages can result in tumour growth
inhibition and reduced angiogenesis as shown in an athymic mouse model of cutaneous
melanoma (Gazzaniga, Bravo et al. 2007). Another animal study of an intraocular melanoma
model also showed a tumour promoting role of M2 macrophages for tumour outgrowth in old
mice (Ly, Baghat et al. 2010). In our model, tumour progression in terms of tumour size did
not correlate with macrophage status or age. However, in vitro results showed increased cell
proliferation rates in melanoma cells which were grown in an environment (supernatant)
shaped by unpolarised macrophages. In vivo, advanced age and the presence of macrophages
were individually associated with an increased occurrence of prognostically significant
extravascular matrix patterns and thus, with an aggressive phenotype. Melanoma cells are
able to actively remodel the ECM in order to arrange stromal patterns such as vascular
networks (Daniels, Boldt et al. 1996). Remodelling of the microcirculation to form such
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
76
vascular networks appears a precondition for metastasis in UM und thereby a determinant for
patient’s outcome. Besides tumour cells themselves, macrophages are also potentially able to
enhance extravascular matrix pattern formation, tumour remodelling and angiogenesis by
producing specific inflammatory cytokines or enzymes (Seftor, Seftor et al. 2001). The effect
of an increased frequency of prognostically significant extravascular matrix patterns in aged
and untreated mice might be directly related to the crosstalk between melanoma cells and
TAM.
The impact of M2 macrophages
A tumour may take advantage of an inflammatory microenvironment that is dominated by M2
macrophages. In many tumours including uveal melanoma infiltrating TAM are indeed of a
M2 polarisation phenotype and are statistically correlated with tumour progression
(Mantovani, Sozzani et al. 2002; Gordon and Martinez 2010; Mantovani and Sica 2010). Our
results from the polarisation experiment display tumour characteristics under iatrogenically
induced extreme tumour microenvironments of M1 or M2 dominance. They do not model
actual conditions in intraocular tumours but demonstrate in which ways polarised
macrophages are capable of influencing tumour characteristics (and thus prognosis). In vitro
as well as in vivo, our results point to far more aggressive tumour growth patterns in M2conditioned tumours in terms of tumour cell proliferation rates and histologic growth
characteristics. Infiltration with leucocytes was much more pronounced in M2- than in M1conditioned tumours whereby increased numbers of lymphocytes and macrophages were
found throughout the tumour but also throughout adjacent ocular structures, particularly in
vicinity to vessels. In uveal melanoma, high lymphocytic infiltration of the tumour is often
associated with an increased number of M2 macrophages which in turn is correlated with
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
77
monosomy 3 and thus with a poor prognosis (Bronkhorst, Vu et al. 2012). Furthermore, in our
M2-conditioned tumours collagen IV-positive matrix patterns were more frequently present,
indicating an increased tissue remodelling compared to M1-conditioned tumours. Collagen IV
positive ECM is correlated with tumour size and/ or a worse prognosis in different tumour
entities (Krecicki, Zalesska-Krecicka et al. 2001; Ioachim, Charchanti et al. 2002; Pierard,
Pierard-Franchimont et al. 2012). Changes in the composition and content of ECM influence
tumour and stromal properties and may promote metastatic spread (Gilkes, Semenza et al.).
Further, stromal cells are important contributors to the tumour’s microenvironment as
released mediators such as metalloproteinases may promote tissue remodelling and cell
migration. In cutaneous melanoma, among other factors, collagen IV was found to influence
tumour cell mobility and thus may contribute to tumour progression and may possibly be a
prognostic indicator (Pierard, Pierard-Franchimont et al. 2012). A synergistic interplay
between the host and the tumour aiming for increased tissue remodelling and angiogenesis is
crucial for the tumour’s circulation and progression (Catalano, Turdo et al. 2013). In line with
this, co-option of host vessels and increased collagen IV rates were found predominantly
adjacent to the retina (i.e. at the edge of the tumour) in our model. As these effects were
pronounced in tumours with M2-conditioned melanoma cells, such a remodelling effect might
be attributed to a microenvironment dominated by M2 macrophages.
In addition, MVD was also significantly increased in M2-conditioned tumours. MVD is a
histopathological indicator associated with tumour microvessels that has independent
prognostic significance in uveal melanoma and other tumour entities. It is further correlated
with largest basal diameter and abundance of TAM and thus with a worse prognosis (Makitie,
Summanen et al. 1999; Makitie, Summanen et al. 2001). Enhanced angiogenesis in terms of
an increased MVD in M2-conditioned tumours, again, points to a more aggressive phenotype,
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
78
which supports the paradigm of pro-angiogenic, tumour-favoring effects of M2-polarised
macrophages.
In our study, advanced age did not have an impact on growth characteristics of the
conditioned ocular tumours while M2-conditioned tumours exhibited a worse phenotype than
M1-conditioned tumours. This suggests that the impact on poor prognosis can be attributed
more directly to an increased M2 influence rather than to other age-dependent factors alone.
The impact of age
Since advanced age was associated with an increased occurrence of M2 macrophages and
more frequent extravascular matrix patterns, we concluded that old mice represent a better
tumour model than young mice. Thus, for questions related to the tumour microenvironment
old mice represent a better animal model than their younger counterparts and should be used
in studies. This is in accordance with the situation in human tumours where age and
extravascular matrix patterns are associated with a worse prognosis.
Apparently, advanced age has an impact on the recruitment and functional phenotype of
macrophages (Gomez, Nomellini et al. 2008). In our model, bone marrow derived TAM (Gr-1
positive) exhibited increased M2 phenotypical characteristics in old mice when compared to
the young, as shown by flow cytometry analysis. This effect was slightly beneath statistical
significance (p=0.08). However, since macrophage polarisation is a continuum and other
studies are in accordance with our observation, the tumour microenvironment in old mice
appears to be M2-dominated. This is in line with the fact that macrophages from geriatric
mice are particularly sensitive to signals that promote their M2 polarisation (Jackaman,
Radley-Crabb et al. 2013). Further, in an anterior chamber melanoma model tumour
progression and outgrowth in old mice depended on the presence of macrophages which had a
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
79
predisposition for the M2 phenotype (Ly, Baghat et al. 2010). This suggests a M2-driven
progression and thus an influence on the patient’s prognosis. However, in our polarisation
experiment we could not document a significant influence of advanced age on histopathologic
growth factors when compared to the young. This might be attributed to the dominant effect
of the iatrogenically manipulated M2 microenvironment so that the age’s impact diminished.
Overall, as old mice exhibited a M2-dominated tumour microenvironment an aged
immunoprofile may provide a basis for more aggressive tumours while a progressing
aggressive tumour phenotype is depending on infiltrating macrophages and their M2
phenotype.
Summary
Old mice exhibited more aggressive tumour phenotypes and thereby mimicking human
intraocular melanomas more appropriately. Pro-angiogenic and tumour promoting factors
such as extravascular matrix patterns, stroma remodelling and MVD could be directly
attributed to the existence of TAM in vivo, in particular to M2 macrophages. In vitro results
confirmed increased tumour malignancy in M2-dominated conditions. Since old mice also
exhibited increased numbers of M2 macrophages in the tumour, an aged immunoprofile
provides a basis for tumour growth with M2 macrophages being directly involved into a more
aggressive phenotype.
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
80
Acknowledgements
We gratefully acknowledge research funding from BONFOR (University of Bonn) and the
“Verein der Rheinisch-Westfälischen Augenärzte (RWA)”. We have no financial disclosures
to report. We would like to thank Hans E. Grossniklaus (Emory University, Atlanta, GA,
USA) for scientific guidance, Thomas Tüting (Laboratory of Experimental Dermatology,
University of Bonn, Bonn, Germany) for kindly providing the HCmel12 murine melanoma
cell line and Eicke Latz (Institute of Innate Immunity, University of Bonn, Bonn, Germany)
for kindly providing the murine macrophages for in vitro experiments. Further we are
thankful to André Tittel (Institute of Experimental Immunology, University of Bonn, Bonn,
Germany) and Tobias Bald (Laboratory for Experimental Dermatology, University of Bonn,
Bonn, Germany) for their assistance in flow cytometry and management of the melanoma cell
line, respectively. Especially, we want to thank Parand Widmar and Claudine Strack
(Department of Ophthalmology, University of Bonn, Bonn, Germany) for their assistance in
histology.
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
81
References
1. Mantovani A, Marchesi F, Porta C, Sica A, Allavena P. Inflammation and cancer:
breast cancer as a prototype. Breast 2007;16 Suppl 2:S27-33.
2. Hendrix MJ, Seftor EA, Hess AR, Seftor RE. Vasculogenic mimicry and tumor-cell
plasticity: lessons from melanoma. Nat Rev Cancer 2003;3:411-21.
3. Allavena P, Sica A, Solinas G, Porta C, Mantovani A. The inflammatory microenvironment in tumor progression: the role of tumor-associated macrophages. Crit Rev Oncol
Hematol 2008;66:1-9.
4. Shields CL, Kaliki S, Furuta M, Fulco E, Alarcon C, Shields JA. American Joint
Committee on Cancer classification of posteriour uveal melanoma (tumor size category)
predicts prognosis in 7731 patients. Ophthalmology 2013;120:2066-71.
5. Folberg R, Mehaffey M, Gardner LM, Meyer M, Rummelt V, Pe'er J. The
microcirculation of choroidal and ciliary body melanomas. Eye (Lond) 1997;11 ( Pt 2):22738.
6. Foss AJ, Alexander RA, Jefferies LW, Hungerford JL, Harris AL, Lightman S.
Microvessel count predicts survival in uveal melanoma. Cancer Res 1996;56:2900-3.
7. Onken MD, Worley LA, Char DH, Augsburger JJ, Correa ZM, Nudleman E,
Aaberg TM, Jr., Altaweel MM, Bardenstein DS, Finger PT, Gallie BL, Harocopos GJ, et al.
Collaborative ocular oncology group report number 1: prospective validation of a multi-gene
prognostic assay in uveal melanoma. Ophthalmology 2012;119:1596-603.
8. Field MG, Harbour JW. Recent developments in prognostic and predictive testing in
uveal melanoma. Curr Opin Ophthalmol 2014;25:234-9.
9. Catalano V, Turdo A, Di Franco S, Dieli F, Todaro M, Stassi G. Tumor and its
microenvironment: a synergistic interplay. Semin Cancer Biol 2013;23:522-32.
10. Makitie T, Summanen P, Tarkkanen A, Kivela T. Tumor-infiltrating macrophages
(CD68(+) cells) and prognosis in malignant uveal melanoma. Invest Ophthalmol Vis Sci
2001;42:1414-21.
11. Toivonen P, Makitie T, Kujala E, Kivela T. Microcirculation and tumor-infiltrating
macrophages in choroidal and ciliary body melanoma and corresponding metastases. Invest
Ophthalmol Vis Sci 2004;45:1-6.
12. Maat W, Ly LV, Jordanova ES, de Wolff-Rouendaal D, Schalij-Delfos NE, Jager
MJ. Monosomy of chromosome 3 and an inflammatory phenotype occur together in uveal
melanoma. Invest Ophthalmol Vis Sci 2008;49:505-10.
13. Jager MJ, Ly LV, El Filali M, Madigan MC. Macrophages in uveal melanoma and
in experimental ocular tumor models: Friends or foes? Prog Retin Eye Res 2011;30:129-46.
14. Arnold L, Henry A, Poron F, Baba-Amer Y, van Rooijen N, Plonquet A, Gherardi
RK, Chazaud B. Inflammatory monocytes recruited after skeletal muscle injury switch into
antiinflammatory macrophages to support myogenesis. J Exp Med 2007;204:1057-69.
15. Mantovani A, Sozzani S, Locati M, Allavena P, Sica A. Macrophage polarization:
tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes.
Trends Immunol 2002;23:549-55.
16. Herwig MC, Bergstrom C, Wells JR, Holler T, Grossniklaus HE. M2/M1 ratio of
tumor associated macrophages and PPAR-gamma expression in uveal melanomas with class 1
and class 2 molecular profiles. Exp Eye Res 2013;107:52-8.
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
82
17. Gomez CR, Nomellini V, Faunce DE, Kovacs EJ. Innate immunity and aging. Exp
Gerontol 2008;43:718-28.
18. Jackaman C, Radley-Crabb HG, Soffe Z, Shavlakadze T, Grounds MD, Nelson
DJ. Targeting macrophages rescues age-related immune deficiencies in C57BL/6J geriatric
mice. Aging Cell 2013;12:345-57.
19. Kilian MM, Loeffler KU, Pfarrer C, Holz FG, Kurts C, Herwig MC. Intravitreally
Injected HCmel12 Melanoma Cells Serve as a Mouse Model of Tumor Biology of Intraocular
Melanoma. Curr Eye Res 2015:1-8.
20. Jung S, Aliberti J, Graemmel P, Sunshine MJ, Kreutzberg GW, Sher A, Littman
DR. Analysis of fractalkine receptor CX(3)CR1 function by targeted deletion and green
fluorescent protein reporter gene insertion. Mol Cell Biol 2000;20:4106-14.
21. Diaz CE, Rusciano D, Dithmar S, Grossniklaus HE. B16LS9 melanoma cells
spread to the liver from the murine ocular posteriour compartment (PC). Curr Eye Res
1999;18:125-9.
22. van Rooijen N, van Kesteren-Hendrikx E. "In vivo" depletion of macrophages by
liposome-mediated "suicide". Methods Enzymol 2003;373:3-16.
23. Mosser DM, Zhang X. Activation of murine macrophages. Curr Protoc Immunol
2008;Chapter 14:Unit 14 2.
24. Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine
system in diverse forms of macrophage activation and polarization. Trends Immunol
2004;25:677-86.
25. Chen L, Zhang S, Li X, Sun B, Zhao X, Zhang D, Zhao S. A pilot study of
vasculogenic mimicry immunohistochemical expression in intraocular melanoma model.
Oncol Rep 2009;21:989-94.
26. Gazzaniga S, Bravo AI, Guglielmotti A, van Rooijen N, Maschi F, Vecchi A,
Mantovani A, Mordoh J, Wainstok R. Targeting tumor-associated macrophages and inhibition
of MCP-1 reduce angiogenesis and tumor growth in a human melanoma xenograft. J Invest
Dermatol 2007;127:2031-41.
27. Ly LV, Baghat A, Versluis M, Jordanova ES, Luyten GP, van Rooijen N, van Hall
T, van der Velden PA, Jager MJ. In aged mice, outgrowth of intraocular melanoma depends
on proangiogenic M2-type macrophages. J Immunol 2010;185:3481-8.
28. Daniels KJ, Boldt HC, Martin JA, Gardner LM, Meyer M, Folberg R. Expression
of type VI collagen in uveal melanoma: its role in pattern formation and tumor progression.
Lab Invest 1996;75:55-66.
29. Seftor RE, Seftor EA, Koshikawa N, Meltzer PS, Gardner LM, Bilban M, StetlerStevenson WG, Quaranta V, Hendrix MJ. Cooperative interactions of laminin 5 gamma2
chain, matrix metalloproteinase-2, and membrane type-1-matrix/metalloproteinase are
required for mimicry of embryonic vasculogenesis by aggressive melanoma. Cancer Res
2001;61:6322-7.
30. Mantovani A, Sica A. Macrophages, innate immunity and cancer: balance,
tolerance, and diversity. Curr Opin Immunol 2010;22:231-7.
31. Gordon S, Martinez FO. Alternative activation of macrophages: mechanism and
functions. Immunity 2010;32:593-604.
32. Bronkhorst IH, Vu TH, Jordanova ES, Luyten GP, Burg SH, Jager MJ. Different
subsets of tumor-infiltrating lymphocytes correlate with macrophage influx and monosomy 3
in uveal melanoma. Invest Ophthalmol Vis Sci 2012;53:5370-8.
IV Impact of macrophages on tumour growth characteristics in a murine
ocular tumour model
83
33. Ioachim E, Charchanti A, Briasoulis E, Karavasilis V, Tsanou H, Arvanitis DL,
Agnantis NJ, Pavlidis N. Immunohistochemical expression of extracellular matrix
components tenascin, fibronectin, collagen type IV and laminin in breast cancer: their
prognostic value and role in tumor invasion and progression. Eur J Cancer 2002;38:2362-70.
34. Pierard GE, Pierard-Franchimont C, Delvenne P. Malignant melanoma and its
stromal nonimmune microecosystem. J Oncol 2012;2012:584219.
35. Krecicki T, Zalesska-Krecicka M, Jelen M, Szkudlarek T, Horobiowska M.
Expression of type IV collagen and matrix metalloproteinase-2 (type IV collagenase) in
relation to nodal status in laryngeal cancer. Clin Otolaryngol Allied Sci 2001;26:469-72.
36. Gilkes DM, Semenza GL, Wirtz D. Hypoxia and the extracellular matrix: drivers
of tumor metastasis. Nat Rev Cancer;14:430-9.
37. Makitie T, Summanen P, Tarkkanen A, Kivela T. Microvascular density in
predicting survival of patients with choroidal and ciliary body melanoma. Invest Ophthalmol
Vis Sci 1999;40:2471-80.
V Origin of intratumoral vascular structures in a murine ocular tumour
model
84
V
Origin of intratumoural vascular structures in a murine ocular
tumour model
Marta M. Kilian1, Karin U. Loeffler1, T. Hoeller2, Frank G. Holz1, Daniela Wenzel3, Martina
C. Herwig1
1
Department of Ophthalmology, University of Bonn, Bonn Germany
2
Institute for Medical Biometry, Informatics and Epidemiology, University of Bonn, Bonn,
Germany
3
Institute of Physiology I, University of Bonn, Bonn, Germany
V Origin of intratumoral vascular structures in a murine ocular tumour
model
85
Abstract
Purpose: Angiogenesis represents the main pathway of tumour vascularisation. In our study,
we investigated characteristics of tumour vascularisation with respect to the origin of tumour
vessels and the impact of tumour associated macrophages (TAM) in a murine model of
intraocular melanoma.
Materials and Methods: 15 flt-1/eGFP BAC mice which express GFP in all smaller vessels,
received an intravitreal melanoma cell injection. Ocular tumours were examined by ex vivo
imaging (n=3) or by histology (H&E, PAS without hematoxylin) and immunohistochemistry
(GFP, CD31, collagen IV, laminin, Ki67) (n=12). One group of mice was macrophagedepleted by systemic Clodronate injections prior to tumour cell inoculation (n=7).
Results: Dual labelling of GFP (murine host-vessels) and CD31 (all vessels) revealed that
almost all tumour vessels co-expressed GFP and CD31; very few vessels expressed only
CD31. Active angiogenesis (dual labelling CD31+Ki67) was found predominantly at tumour
borders adjacent to the retina and was more pronounced in untreated mice when compared to
macrophage-depleted mice.
Conclusion: Rapidly growing tumours are dependent on a highly pro-angiogenic
microenvironment in which they are able to adopt host vessels and initiate angiogenic
sprouting from incorporated vessels. TAM exhibited pro-angiogenic properties which
underlines their important role in tumour progression and circulation.
V Origin of intratumoral vascular structures in a murine ocular tumour
model
86
Introduction
Angiogenesis is a crucial point in the development of most solid tumours, including
melanomas. A specific feature of uveal melanomas is their hematogenous pathway of
metastasis due to the absence of lymphatic vessels in the eye. This underlines the importance
of angiotropism and angiogenesis for metastatic dissemination and represents a potential
target for therapeutic intervention.
For enduring growth, solid tumours are dependent on constant blood supply and use different
forms of vascularisation (Hanahan and Weinberg 2011). One source of initial blood supply
may be angiotropic growth and the incorporation of pre-existing vessels from surrounding
structures. Secondly, the tumour may induce growth of new vessels by stimulating sprouting
from pre-existing host vessels, known as angiogenesis. Further, alternative forms of tumour
vascularisation in aggressive tumours have been proposed such as mosaic vessels (tumourand endothelial-lined vasculature) (Chang, di Tomaso et al. 2000), vasculogenesis
(recruitment of endothelial progenitor cells) (Asahara, Murohara et al. 1997), tumour-lined
vascular channels (Timar and Toth 2000), and vasculogenic mimicry (Maniotis, Folberg et al.
1999; Hillen and Griffioen 2007). The latter does not represent true vessels but is composed
of matrix meshworks of fluid conducting channels, which are lined by tumour cells instead of
endothelial cells and which share anastomoses with “true” blood vessels (Chen, Zhang et al.
2009; Seftor, Hess et al. 2012).
However, angiogenesis still represents the main pathway of perfusion in most primary
tumours and is initiated by the “angiogenic switch”. It depends on a pro-angiogenic tumour
microenvironment and is regulated by pro- and anti-angiogenic factors, which are produced
by both malignant cells and the host cells recruited to the tumour site including tumour
V Origin of intratumoral vascular structures in a murine ocular tumour
model
87
infiltrating macrophages (reviewed in (Burrell and Zadeh 2012)). A pro-angiogenic
microenvironment comprises several growth factors such as VEGF, as well as angiopoietins,
interleukins, matrix metalloproteinases and others. Tumour associated macrophages (TAM)
orchestrate the tumour microenvironment in response to secreted factors from tumour cells
and other players of the micromilieu in uveal melanoma. TAM tend to specialise into a M2
phenotype with pro-angiogenic and tumour promoting properties (Mantovani, Sozzani et al.
2002). Subsequently, M2-polarised TAM are able to directly enhance tumour angiogenesis
and vasculogenic mimicry as shown in our intraocular melanoma model (unpublished data,
under review). In uveal melanoma and in other ocular and non-ocular tumours an increased
angiogenesis along with a high mean vascular density (MVD) as well as vasculogenic
mimicry were associated with an increased infiltration with TAM and with patient’s outcome
(Makitie, Summanen et al. 1999; Makitie, Summanen et al. 2001).
In order to better understand different strategies of intraocular tumour perfusion we studied
tumour vascularisation in our intraocular melanoma model. We investigated vascular patterns
and the origin of intratumoural endothelial-lined vasculature in flt-1/eGFP BAC mice which
express GFP in all smaller blood vessels (Herz, Heinemann et al. 2012). Intratumoural GFPexpressing vessels might be visualised by in vivo and ex vivo imaging at different time stages.
Discrimination between angiogenic and vasculogenic as well as between host- and tumourderived vessels can be performed by immunohistochemical dual-labelling.
As macrophages play a pivotal role in tumour angiogenesis, we investigated their influence on
different forms of vascularisation strategies and on tumour circulation patterns, in particular
on active angiogenesis and on vasculogenic mimicry.
V Origin of intratumoral vascular structures in a murine ocular tumour
model
88
Materials and Methods
Animals
Breeding and housing of flt-1/eGFP BAC mice was carried out and supervised according to
the Association of Research in Vision and Ophthalmology statement for the Use of Animals
in Ophthalmic and Vision Research. These transgenic mice exhibit a persistent eGFP
expression in small vessels which is driven by the flt-1 promoter (VEGF receptor 1, VEGFR1) (Herz, Heinemann et al. 2012). Thus, all ocular vessels originating from the host (mouse)
exhibit a constant eGFP expression.
15 flt-1/eGFP BAC mice were used for the study. Three mice were investigated at the age of
eight months for ex vivo imaging, 12 mice were investigated at the age of 10 months for
histology and immunohistochemistry, as intraocular melanomas in such old mice better
resemble human tumours with regard to histologic features of the tumour microenvironment
and vascularisation.
Mouse model of intraocular melanoma
As previously described, 1x105 of murine cutaneous HCmel12 melanoma cells were
intravitreally injected into the right eye of each mouse of all study groups at day 0 in general
anaesthesia (Kilian, Loeffler et al. 2015). Mice were sacrificed at day 10 post intravitreal
injection, right eyes were enucleated and processed for histologic or immunohistochemical
examination.
Macrophage-depleted mice (n=7) were studied in comparison to untreated mice (n=5).
Liposome-encapsulated dichloromethylene diphosphonate (Cl2MDP; clodronate, ordered
through clodronateliposomes.org) was used to systemically deplete blood monocytes and
V Origin of intratumoral vascular structures in a murine ocular tumour
model
89
macrophages at a dosage of 0.1mg/g body weight (van Rooijen and van Kesteren-Hendrikx
2003). Seven mice received repeated intraperitoneal injections of Clodronate liposomes at
days -6, -3, 0 and 3 and 6 of study. To evaluate actual macrophage depletion, liver sections of
Clodronate-treated and untreated mice were stained immunohistochemically with a F4/80
antibody (F4/80 Antibody, Cl:A3-1, AbD Serotec, Oxford, U.K.; dilution 1:100).
Ex vivo imaging
In three mice, tumour-bearing and contralateral control eyes were enucleated immediately
after euthanasia. Eyes were kept in PBS and shortly after investigated using an Axio Zoom
microscope (Carl Zeiss Microscopy GmbH, Jena, Germany). Specimens were examined for
GFP-positive vessels in areas of extraocular tumour extension and after removing the
anteriour parts of the eye for GFP-positive vessels in the intraocular tumour.
Histology and immunohistochemistry
Processing of histologic specimens of 12 mice was carried out as described before (Kilian,
Loeffler et al. 2015). Briefly, in hematoxylin eosin (H&E) stains, tumour size was measured
in mm² at 40x magnification using ImageJ 1.45s (W Rasband, NIH, USA); vascular
characteristics like angiotropism, co-option of retinal vessels and intratumoural endothelial
vessels were evaluated at 200x magnification. PAS (periodic acid-Schiff reaction) stains
without hematoxylin were investigated for ECM-rich channels/VM along with their
prognostic significant extravascular matrix patterns (parallels with cross-linkings, loops,
networks). For analysis of the MVD, sections were stained with a GFP- and CD31-antibody
(GFP Rabbit IgG, Life Technologies, Carlsbad, CA, USA; dilution 1:100; CD31, SZ31,
Dianova, Hamburg, Germany, dilution 1:10). Under x200 power, counts of positive staining
V Origin of intratumoral vascular structures in a murine ocular tumour
model
90
vessels inside a 0.25mm2 graticule were performed in 1-3 areas according to introduced
standards (Foss, Alexander et al. 1996). Staining of ECM was performed with a collagen IV
antibody (Collagen IV Antibody, Novus Biologicals, Littleton, CO, USA dilution 1:100) and
a laminin antibody (Laminin Antibody, Novus Biologicals, Littleton, CO, USA, dilution
1:100). Dual labelling of CD31 + GFP, CD31 + Collagen IV, and CD31 + Ki67 (Anti-Ki67,
SP6, Abcam, Cambridge, UK, dilution 1:50) was performed and investigated by
immunofluorescence microscopy. Dual labelling of CD31 and GFP allows for determination
of the origin of endothelial-lined vessels. Vessels, which are positive for CD31 and
simultaneously for GFP are of host-origin (mouse). On the other hand, exclusively CD31positive vessels are derived from other origins e.g. vasculogenesis of from the tumour itself.
Dual labelling for CD31 and the proliferation marker Ki67 is supposed to reveal active
angiogenesis.
Statistical analysis
Statistical analysis was performed with IBM SPSS Statistics 22.0 (IBM Corp, Armonk, NY,
USA). Tumour sizes in mm2 and MVD of different study groups were evaluated by an
analysis of variance and a Post Hoc Test. Chi2 test and Pearson's correlation coefficient were
used to determine the relationship between different study groups and tumour growth
characteristics. A p≤0.05 was considered as statistically significant and significance level was
indicated by using * for p ≤ 0.05.
V Origin of intratumoral vascular structures in a murine ocular tumour
model
91
Results
Ex vivo imaging
At investigation by the Axio Zoom microscope at x25 magnification, networks of GFPexpressing small vessels were identified on the ocular surface and in the anterior chamber. In
detail, a compact network was found along the limbus which was drained by a major vein.
Fine vessels were observed within the pigmented iris as well as in adherent orbital structures
such as the conjunctiva and muscle parts (Fig.1A).
Two of three investigated mouse eyes exhibited extraocular tumour extension which
presented as dark masses near the ocular limbus (Fig.1B). The location of the extraocular
tumour apparently represented the site of previous intravitreal injection as the tumour
probably took the path of least resistance for outgrowth. GFP-positive vessels were
surrounding the extraocular tumour basis but no obvious vessels were found on the tumour
surface at this early stage of tumour growth (day 10).
Figure 1.: Eyes of flt-1/eGFP BAC mice viewed by Axio Zoom microscope (x25 magnification)
P: pupil I: iris V: vein, T: extraocular tumour part
(A) Eye of control mouse; (B) Eye of tumour inoculated mouse with extraocular tumour extension
V Origin of intratumoral vascular structures in a murine ocular tumour
model
92
Histology and immunohistochemistry
All mice which received an intravitreal HCmel12 tumour cell injection developed solid
intraocular tumours, which gained access to the host’s blood circulation. In H&E stains, these
tumours presented with angiotropic growth, co-option of retinal or choroidal vessels,
networks of endothelial vessels as well as of vasculogenic mimicry.
Dual-labelling of CD31 and GFP revealed that the majority of tumour vessels were positive
for both markers and were thus of mouse origin (Fig.2A+B). Endothelial vessels labelled
exclusively by the CD31-antibody were very sparse and represented only small vessels with a
narrow lumen. A vessel sequence representing mosaic vessels (CD31+GFP+ vessels in linear
arrangement with CD31+GFP- vessels) was not unequivocally observed in any of the tumours.
A
B
Figure 2: Vascular structures in intraocular tumours ((A) x100 and (B) x200 magnification)
(A+B) Dual labelling for host vessels’ GFP (green) and CD31 (red) revealed most intratumoural
vessels being positive for both markers and thus of angiogenic host origin. Only few vessels were
exclusively CD31 positive (arrows).
V Origin of intratumoral vascular structures in a murine ocular tumour
model
93
Active proliferation of endothelial vessels was regarded as a positive nuclear Ki67 staining in
a CD31 positive endothelial cell (cytoplasm). Such a dual-labelling was found predominantly
at tumour margins which were adjacent to host structures. This suggests active sprouting of
blood vessels from surrounding ocular structures (Fig.3A+B).
Intraperitoneal injection of Clodronate liposomes led to systemic macrophage depletion as
examined in immunohistochemical stains of the liver. While untreated mice exhibited
frequent F4/80-posititive staining of Kupffer cells throughout the liver, Clodronate liposometreated mice were lacking positively staining cells. In untreated mice 39.1% of endothelial
vessels showed proliferative activity compared to 30.9% of proliferating vessels in
macrophage-depleted mice (p=0.114).
A
B
Figure 3: Active angiogenesis in intraocular tumours ( (A)x100 and (B) x200 magnification)
(A+B) Dual labelling for Ki67 (green) and CD31 (red) revealed active angiogenesis (arrows) which
was predominantly found at tumour margins adjacent to host structures.
Collagen IV expression revealed no statistical significant difference between the two study
groups (p=0.364). However, staining for collagen IV suggested a slightly pronounced
expression in untreated mice compared to macrophage-depleted mice. Collagen IV-positive
V Origin of intratumoral vascular structures in a murine ocular tumour
model
94
structures formed arcing and looping patterns, exhibited a patchy distribution and they were
mainly found in close vicinity to endothelial vessels. Such tumour areas with increased tissue
remodelling were also positive for laminin and were located predominantly at tumour margins
(Fig.4A). Intraocular tumour size did not differ statistically between both study groups (mean
size: untreated mice 0.7mm2, macrophage-depleted mice 0.73mm2, p=0.752). MVD did not
differ statistically between untreated and macrophage-depleted mice (p=0.513). In PAS stains
without hematoxylin, ECM-rich structures of untreated mice exhibited more frequently
prognostic extravascular matrix patterns. However, this effect was statistically not significant
(p=0.197) in contrast to another experiment with an analogous tumour model. This might be
attributed to the minor number of animals in this study.
A
B
Figure 4: Extracellular matrix (ECM) in intraocular tumours ((A) x100 and (B) x200
magnification)
(A+B) Dual labelling for the ECM component collagen IV (green) and CD31 (red) showed abundant
ECM structures adjacent to endothelial vessels and of arcing and looping patterns.
V Origin of intratumoral vascular structures in a murine ocular tumour
model
95
Discussion
In many tumours different strategies of vascularisation and correlation to tumour progression
have been well described (reviewed in (Hillen and Griffioen 2007)). Generally, angiogenesis
is thought to be the main pathway of tumour vascularisation. The essential role of
angiogenesis in solid tumours and its potential therapeutic implications were first described
by Folkman and coworkers (Folkman 1971). However, the long-held belief that cancer
patients can be effectively treated with anti-angiogenic therapy diminished after disappointing
clinical results. Heterogeneity and complexity of tumours and of their vascular architecture
may be a reason for that (Verheul, Voest et al. 2004). Co-option of host vessels, mosaic
vessels, vasculogenesis and VM might be of more importance than previously thought. In
uveal, cutaneous and oral melanoma, numerous studies have focused on angiogenesis and
other mechanisms by which melanomas ensure their vascularisation. Besides sprouting
angiogenesis, also patterns of co-option of host vessels, mosaic vessels, tumour-lined
vasculature, and networks of vessel-like structures not lined by endothelial cells (vasculogenic
mimicry) have been described (reviewed in (Pastushenko, Vermeulen et al. 2014), (Hendrix,
Seftor et al. 2002; Lee, Nagai et al. 2002; Zhang, Zhang et al. 2006). These studies used PAS
and immunohistochemical staining of endothelial vessels, extracellular matrix (ECM) and
other structures (e.g. CD31, collagen IV) to visualise and investigate the characteristics of
melanoma vascularisation. In our study, we were able to further address the origin of tumour
vessels in an intraocular melanoma model with immunocompetent mice.
V Origin of intratumoral vascular structures in a murine ocular tumour
model
96
Origin of tumour vessels
Almost all tumour vessels in flt-1/eGFP BAC mice exhibited a co-expression of GFP and
CD31 and were thus of angiogenic host origin. Apparently, the tumour adopted host vessels
including their capacity for angiogenesis
and benefited from
a pro-angiogenic
microenvironment. Thus, angiogenesis represented the main vascularisation strategy of the
tumour. However, a minor number of intratumoural vessels were found to be negative for
GFP and solely marked by the CD31 antibody and must, subsequently, be of other origin than
sprouting angiogenesis from host vessels. These vessels might have risen from two other
potential origins: from bone marrow derived endothelial progenitor cells in terms of
vasculogenesis or from tumour-derived endothelial vessels.
Circulating endothelial precursors, shed from the vessel wall or mobilised from the bone
marrow, can also contribute to tumour vascularisation (vasculogenesis) (Rafii 2000). Such
endothelial precursor cells may line intratumoural vessels and express characteristic markers
(CD31, CD34, VEGFR-2) along with the stem cell marker CD133 (Timmermans, Plum et al.
2009). To identify such vasculogenic vessels formed by endothelial progenitor cells we
(intend to) perform dual labelling of CD31 and CD133. Shall we be able to confirm that these
vessels have originated from vasculogenesis by being built of endothelial progenitor cells
(CD31+CD133+GFP-), this would proof heterogeneity of tumour vascularisation in our model.
Further, flt-1/eGFP BAC mice would represent a potent model to distinguish between
vasculogenesis (CD31+CD133+GFP- vessels) and angiogenesis (CD31+CD133+GFP+ vessels).
This might offer to study these different forms of vascularisation in several tumour entities
and other pathologies like injury or inflammatory processes.
On the other hand, if CD31+GFP- vessels in flt-1/eGFP BAC mice reveal not to have
originated from vasculogenesis (CD133-), another hypothesis of their origin might be a
V Origin of intratumoral vascular structures in a murine ocular tumour
model
97
derivation from cancer stem cells. This hypothesis might further be supported by dual
labelling of CD31 and a melanoma marker (e.g. HMB45/MART-1). Two studies in
glioblastoma found that blood vessel cells within the tumours contained genetic markers
characteristic of cancer cells (with stem cell like properties), suggesting that these blood
vessels were of tumour origin (Ricci-Vitiani, Pallini et al. 2010; Wang, Chadalavada et al.
2010). Another group describes „tumour endothelial cells (TEC)“ as intratumoural vessels of
tumour origin with highly angiogenic phenotypes and higher proliferative and migratory
abilities than „normal endothelial cells“ (=host-derived, angiogenic endothelial cells)
(Ohmura-Kakutani, Akiyama et al. 2014). However, these conclusions were based on in vitro
studies and their transferability into in vivo conditions remains unanswered. In an ischemic,
nude mouse model with inoculated human aggressive melanoma cells, however, newly
formed vasculature within the tumour showed endothelial cells of human and of mouse origin
in a linear arrangement. Thus, besides angiogenic murine vessels, human melanoma cells may
acquire endothelial-like morphology and are incorporated in capillaries, and thus, may
actively contribute to neovascularisation (Mihic-Probst, Ikenberg et al. 2012).
Our ongoing studies may reveal the origin of CD31+GFP- intratumoural vessels in flt-1/eGFP
BAC mice and either offer a new model for distinguishing vasculogenesis from angiogenesis
or contribute to recent discussions regarding tumour-derived endothelial vessels. This would
represent the first orthotopic animal model of such tumour-derived endothelial cells in
melanoma.
Intratumoural vascularisation and TAM
In our study, angiogenesis represented the primary mechanism of tumour vascularisation.
However, other vascularisation strategies were also observed, like co-option of host vessels
V Origin of intratumoral vascular structures in a murine ocular tumour
model
98
and VM. Studies in murine and human cutaneous melanoma revealed that vascularisation
strategies such as VM and mosaic vessels were not uniformly distributed throughout the
tumour but appeared in patches. Blood supply was sustained simultaneously by VM and
endothelial lined vasculature (Zhang, Zhang et al. 2006). In our model, true mosaic vessels
which are lined by host endothelial cells (CD31+GFP+) with a transient change to tumour
endothelial cells (HMB45/MART-1+CD31+GFP-) were not yet unequivocally observed.
However, in accordance with the study by Zhang (vide supra), we observed a patchy
distribution of VM and collagen IV positive ECM structures. This underlines how the
intraocular tumour ensures constant blood supply by diverse strategies in our model.
In order to effectively inhibit tumour vascularisation – as a potential therapeutic interventionall different strategies of vascularisation should be addressed. One point of action might be
manipulation of the pro-angiogenic tumour microenvironment which in turn is mainly
orchestrated by TAM (in case of ocular melanoma and many other tumours). Manipulating
TAM and thus altering the tumour microenvironment might be a promising approach to
control tumour progression and vascularisation (Bronkhorst and Jager 2013).
In our study, the impact of macrophages on different tumour vascularisation strategies was
determined to a minor extend. We examined intraocular tumours of macrophage-depleted
mice in comparison to untreated mice. Dual labelling of CD31 and Ki67 (proliferation
marker) revealed active angiogenesis which was predominantly found at tumour margins
located near host structures. This is in accordance with a study which found high numbers of
proliferating endothelial cells at tumour margins of poor differentiated (and thus aggressive)
tumours (Raluca, Cimpean et al. 2015). In these regions the tumour might have orchestrated
the microenvironment towards pro-angiogenic conditions in order to induce sprouting
angiogenesis from surrounding host structures. Studies investigating MVD and the proportion
V Origin of intratumoral vascular structures in a murine ocular tumour
model
99
of active angiogenesis in cutaneous melanoma and other tumour entities showed that the
number of proliferating endothelial cells (CD31+/CD34+/Nestin+ and Ki67+) but not MVD
was correlated with tumour stage (Baeten, Castermans et al. 2006; Hillen, van de Winkel et
al. 2006; Kruger, Stefansson et al. 2013). Thus, the proportion of proliferating endothelial
cells might be also an indicator for tumour progression and possibly also for patient’s
outcome. In our study, in untreated mice 39.1% of endothelial vessels showed proliferative
activity compared to 30.9% of proliferating vessels in macrophage depleted mice (p=0.114).
As tumours in untreated mice showed a tendency of an increased rate of active angiogenesis
in comparison to macrophage-depleted mice, TAM might have directly shaped the tumour
microenvironment by secreting pro-angiogenic factors – probably in response to a crosstalk
with melanoma cells due to hypoxic conditions. TAM are generally able to stimulate
proliferation and migration of endothelial cells (reviewed in (Lamagna, Aurrand-Lions et al.
2006)) and may thereby influence the rate of active angiogenesis in aggressive tumours. This
points to another pathway by which TAM directly affect prognostically significant
histopathologic tumour characteristics.
Further, untreated mice more frequently showed prognostically significant extravascular
matrix patterns than macrophage-depleted mice. This underlines the pro-angiogenic properties
of TAM and their important role in tumour progression and vascularisation. In another study,
we were able to better retrace the role of macrophages in intraocular tumour biology by using
an increased number of animals (unpublished, under review). Thus, understanding the role of
TAM in tumour vascularisation, controlling or re-polarising TAM into tumouricidal
phenotypes might offer new strategies to manipulate the tumour microenvironment and thus
control tumour progression (Catalano, Turdo et al. 2013).
V Origin of intratumoral vascular structures in a murine ocular tumour
model
100
Summary
Intraocular tumours exhibited different forms of vascularisation including angiogenesis and
vasculogenic mimicry and potentionally also vasculogenesis or tumour-derived endothelial
vessels. Angiogenesis represented the main vascularisation strategy, whereas only very few
vessels derived from other origins. Active angiogenesis was found throughout the tumour but
mainly at tumour borders and was pronounced in untreated mice when compared to
macrophage-depleted mice. TAM had an impact on the frequency of extravascular matrix
pattern and on active angiogenesis. Targeting the tumour microenvironment which is
orchestrated by pro-angiogenic TAM might offer new therapeutic targets to control tumour
vascularisation, progression and metastasis.
Acknowledgements
We gratefully acknowledge research funding from BONFOR (University of Bonn) and the
“Verein der Rheinisch-Westfälischen Augenärzte (RWA)”. We would like to thank Thomas
Tüting (Laboratory of Experimental Dermatology, University of Bonn, Bonn, Germany) for
kindly providing the HCmel12 murine melanoma cell line and Katia Herz (Institute of
Physiology I, University of Bonn, Bonn, Germany) for the assistance ex vivo imaging.
Especially, we want to thank Parand Widmar and Claudine Strack (Department of
Ophthalmology, University of Bonn, Bonn, Germany) for their assistance in histology.
V Origin of intratumoral vascular structures in a murine ocular tumour
model
101
References
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell
2011;144(5):646-74.
Chang YS, di Tomaso E, McDonald DM, Jones R, Jain RK, Munn LL. Mosaic blood
vessels in tumors: frequency of cancer cells in contact with flowing blood. Proc Natl
Acad Sci U S A 2000;97(26):14608-13.
Timar J, Toth J. Tumor sinuses- vascular channels. Pathol Oncol Res 2000;6(2):83-6.
Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, et al. Isolation of
putative progenitor endothelial cells for angiogenesis. Science 1997;275(5302):964-7.
Maniotis AJ, Folberg R, Hess A, Seftor EA, Gardner LM, Pe'er J, et al. Vascular
channel formation by human melanoma cells in vivo and in vitro: vasculogenic
mimicry. Am J Pathol 1999;155(3):739-52.
Hillen F, Griffioen AW. Tumour vascularization: sprouting angiogenesis and beyond.
Cancer Metastasis Rev 2007;26(3-4):489-502.
Seftor RE, Hess AR, Seftor EA, Kirschmann DA, Hardy KM, Margaryan NV, et al.
Tumor cell vasculogenic mimicry: from controversy to therapeutic promise. Am J
Pathol 2012;181(4):1115-25.
Chen L, Zhang S, Li X, Sun B, Zhao X, Zhang D, et al. A pilot study of vasculogenic
mimicry immunohistochemical expression in intraocular melanoma model. Oncol Rep
2009;21(4):989-94.
Mantovani A, Sozzani S, Locati M, Allavena P, Sica A. Macrophage polarization:
tumor-associated macrophages as a paradigm for polarized M2 mononuclear
phagocytes. Trends Immunol 2002;23(11):549-55.
Makitie T, Summanen P, Tarkkanen A, Kivela T. Microvascular density in predicting
survival of patients with choroidal and ciliary body melanoma. Invest Ophthalmol Vis
Sci 1999;40(11):2471-80.
Makitie T, Summanen P, Tarkkanen A, Kivela T. Tumor-infiltrating macrophages
(CD68(+) cells) and prognosis in malignant uveal melanoma. Invest Ophthalmol Vis
Sci 2001;42(7):1414-21.
Herz K, Heinemann JC, Hesse M, Ottersbach A, Geisen C, Fuegemann CJ, et al. Live
monitoring of small vessels during development and disease using the flt-1 promoter
element. Basic Res Cardiol 2012;107(2):257.
Kilian MM, Loeffler KU, Pfarrer C, Holz FG, Kurts C, Herwig MC. Intravitreally
Injected HCmel12 Melanoma Cells Serve as a Mouse Model of Tumor Biology of
Intraocular Melanoma. Curr Eye Res 2015:1-8.
van Rooijen N, van Kesteren-Hendrikx E. "In vivo" depletion of macrophages by
liposome-mediated "suicide". Methods Enzymol 2003;373:3-16.
Foss AJ, Alexander RA, Jefferies LW, Hungerford JL, Harris AL, Lightman S.
Microvessel count predicts survival in uveal melanoma. Cancer Res
1996;56(13):2900-3.
Pastushenko I, Vermeulen PB, Van den Eynden GG, Rutten A, Carapeto FJ, Dirix LY,
et al. Mechanisms of tumour vascularization in cutaneous malignant melanoma:
clinical implications. Br J Dermatol 2014;171(2):220-33.
V Origin of intratumoral vascular structures in a murine ocular tumour
model
17.
18.
19.
20.
21.
22.
23.
24.
102
Lee YJ, Nagai N, Siar CH, Nakano K, Nagatsuka H, Tsujigiwa H, et al.
Angioarchitecture of primary oral malignant melanomas. J Histochem Cytochem
2002;50(11):1555-62.
Zhang S, Zhang D, Wang Y, Zhao W, Guo H, Zhao X, et al. Morphologic research of
microcirculation patterns in human and animal melanoma. Med Oncol
2006;23(3):403-9.
Hendrix MJ, Seftor RE, Seftor EA, Gruman LM, Lee LM, Nickoloff BJ, et al.
Transendothelial function of human metastatic melanoma cells: role of the
microenvironment in cell-fate determination. Cancer Res 2002;62(3):665-8.
Mihic-Probst D, Ikenberg K, Tinguely M, Schraml P, Behnke S, Seifert B, et al.
Tumor cell plasticity and angiogenesis in human melanomas. PLoS One
2012;7(3):e33571.
Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med
1971;285(21):1182-6.
Verheul HM, Voest EE, Schlingemann RO. Are tumours angiogenesis-dependent? J
Pathol 2004;202(1):5-13.
Bronkhorst IH, Jager MJ. Inflammation in uveal melanoma. Eye (Lond)
2013;27(2):217-23.
Catalano V, Turdo A, Di Franco S, Dieli F, Todaro M, Stassi G. Tumor and its
microenvironment: a synergistic interplay. Semin Cancer Biol 2013;23(6 Pt B):52232.
103
VI
Overexpression of Hepatocyte Growth Factor and an oncogenic
CDK4 variant in mice alters corneal stroma morphology but does
not lead to spontaneous ocular melanoma
Marta M. Kilian1, Martina C. Herwig1, Frank G. Holz1, Thomas Tüting2, Karin U. Loeffler1
1
Department of Ophthalmology, University of Bonn, Bonn, Germany
2
Experimental Dermatology, University of Bonn, Bonn, Germany
Letter to the Editor
Accepted at Melanoma Research
VI Overexpression of HGF and an oncogenic CDK4 variant in mice alters corneal
stroma morphology but does not lead to spontaneous ocular melanoma
104
Editor,
Searching for an animal model of spontaneous uveal melanoma, we evaluated the ocular
findings in Hgf-Cdk4R24C mice, an established mouse model of spontaneous skin melanoma.
In humans and in Hgf-Cdk4R24C mice overexpression of hepatocyte growth factor (HGF) may
promote cutaneous melanogenesis by activating RAS signal transduction pathways via its
receptor c-MET (Landsberg, Gaffal et al. 2010). In uveal melanoma, high expression of cMET correlates with an aggressive tumour phenotype (Mallikarjuna, Pushparaj et al. 2007).
Additionally, the oncogenic germline mutation in the cyclin-dependent kinase 4 (CDK4
R24C) further contributes to melanoma development in Hgf-Cdk4R24C mice and human skin
because it functionally inactivates the tumour suppressor p16⁄INK4a, which is of critical
importance for oncogene-induced senescence (Landsberg, Kohlmeyer et al. 2012). This study
should also further clarify the role and impact of HGF and its receptor tyrosine kinase c-MET
as well as cyclin-dependent kinase 4 (CDK4) on ocular melanocytes in comparison with
dermal melanocytes. The majority of these highly pigmented mice spontaneously develops a
spectrum of nevi and primary melanomas without prior carcinogen treatment within the first
year of life (Landsberg, Gaffal et al. 2010). These melanomas are predominantly spreading to
regional lymph nodes and the lungs (Tormo, Ferrer et al. 2006). As Hgf-Cdk4R24C mice
represent an established model of spontaneous cutaneous melanoma, we questioned whether
this mouse strain concurrently shows prolifferations and tumourigenesis in ocular melanocytic
structures.
12 eyes from 11 month old Hgf-Cdk4R24C mice (C57Bl/6 background) were investigated and
were compared with three eyes from 10 months old C57Bl/6 mice. Step sections were stained
with H&E and PAS, and sections in-between were labelled with antibodies to Ki67
VI Overexpression of HGF and an oncogenic CDK4 variant in mice alters corneal
stroma morphology but does not lead to spontaneous ocular melanoma
105
(proliferation marker), MART1 (melanocyte/melanoma marker), CKpan (epithelial cell
differentiation) and F4/80 (macrophages). Prior to immunohistochemistry (IHC), sections
were bleached for better visualisation of the red chromogen AEC (3-amino-9-ethylcarbazole).
At the age of 11 months all Hgf-Cdk4R24C mice exhibited apparent dermal melanocytic
proliferations, partly exceeding 2 mm and were - according to (Landsberg, Gaffal et al. 2010)
- therefore considered as melanomas. By gross examination of the eyes, ocular alterations
were not observed (Figure 1A+B). No significant increase in epithelial pigmentation of either
corneal or conjunctival epithelium was noted (Figure 1C+D). Proliferation of uveal
melanocytes or cells of the retinal pigment epithelium (RPE) was also not observed and the
choroid appeared even somewhat less pigmented with some amelanotic cellular areas
compared to C57Bl/6 mice (n=3) (Figure 1G+H). IHC so far showed no significant difference
in labelling between Hgf-Cdk4R24C and C57Bl/6 mouse eyes. However, histologically, the
most marked difference between Hgf-Cdk4R24C and wildtype mice was the incidental finding
of heavily pigmented cells in the anterior corneal stroma (and conjunctiva to a lesser degree)
and along the chamber angle (Figure 1C-F). This was associated with inflammation,
vascularisation and a marked proliferation of these pigmented cells in three of twelve eyes
(Figure 1C). Histological and immunohistochemical staining was unable to further
characterise the cellular corneal infiltrate.
In Hgf-Cdk4R24C mice of 11 months of age, despite of apparent dermal melanocytic
proliferations progressing toward cutaneous melanomas, no major difference in pigmentation
or cellular components was observed in the ocular epithelium or in the posterior segment.
Thus, Hgf-Cdk4R24C mice up to this age do not qualify as a model for spontaneous
conjunctival or uveal melanoma. A delayed melanomagenesis at an even more advanced age
may not completely be ruled out. Apparently, genetic alterations regarding Hgf and Cdk4 and
VI Overexpression of HGF and an oncogenic CDK4 variant in mice alters corneal
stroma morphology but does not lead to spontaneous ocular melanoma
106
the subsequent deregulation of the c-MET mediated RAS signal transduction pathways as
well as the p16/Ink4a-dependent cell cycle regulation apparently lead to cutaneous primary
and metastatic melanomas. However, uveal or conjunctival cell cycles do not seem to be
profoundly affected. To date, several characteristic genetic anomalies for tumour progression
and metastasis of cutaneous as well as uveal melanoma have been identified (van den Bosch,
Kilic et al. 2010). But unlike in cutaneous melanoma, B-Raf, Ras or Kit mutations occur
rarely in uveal melanoma and characteristic mutations differ between uveal and cutaneous
melanoma (van den Bosch, Kilic et al. 2010). Although mutations similar to skin melanoma
such as B-RAF V600 have been found in conjunctival melanomas (Lake, Jmor et al. 2011),
our findings underline genetic differences also between conjunctival and skin melanomas.
However, since remarkable changes regarding pigmentation were noted in the cornea and
chamber angle in these animals, further investigation of the anterior segment might allow
studying mechanisms of corneal neovascularisation and possibly also glaucoma.
VI Overexpression of HGF and an oncogenic CDK4 variant in mice alters corneal
stroma morphology but does not lead to spontaneous ocular melanoma
107
Figure 1: Hgf-Cdk4R24C (A,C,E,G) and C57Bl/6 (B,D,F,H) mouse eyes, H&E; arrows
indicating corneal intrastromal pigment.
(A+B) Overview of mouse eyes; (C+D) Cornea: (C) cornea with a deeply pigmented cellular
lesion (stars) within the anterior stroma and diffuse neovascularisation (arrowheads) and (D)
cornea without evidence of corneal pigment; (E+F) Chamber angle: (E) chamber angle with
increased pigmentation of the trabecular meshwork and cornea with intrastromal pigment and
(F) unremarkable chamber angle; (G+H) Unremarkable choroid and retina of the HgfCdk4R24C mouse and C57Bl/6 mouse.
VI Overexpression of HGF and an oncogenic CDK4 variant in mice alters corneal
stroma morphology but does not lead to spontaneous ocular melanoma
108
References
1.
2.
3.
4.
5.
6.
Landsberg J, Gaffal E, Cron M, Kohlmeyer J, Renn M, Tuting T: Autochthonous
primary and metastatic melanomas in Hgf-Cdk4 R24C mice evade T-cell-mediated
immune surveillance. Pigment Cell Melanoma Res 2010;23:649-660.
Mallikarjuna K, Pushparaj V, Biswas J, Krishnakumar S: Expression of epidermal
growth factor receptor, ezrin, hepatocyte growth factor, and c-Met in uveal melanoma:
an immunohistochemical study. Curr Eye Res 2007;32:281-290.
Landsberg J, Kohlmeyer J, Renn M, Bald T, Rogava M, Cron M, Fatho M, Lennerz V,
Wolfel T, Holzel M, Tuting T: Melanomas resist T-cell therapy through inflammationinduced reversible dedifferentiation. Nature 2012;490:412-416.
Tormo D, Ferrer A, Gaffal E, Wenzel J, Basner-Tschakarjan E, Steitz J, Heukamp LC,
Gutgemann I, Buettner R, Malumbres M, Barbacid M, Merlino G, Tuting T: Rapid
growth of invasive metastatic melanoma in carcinogen-treated hepatocyte growth
factor/scatter factor-transgenic mice carrying an oncogenic CDK4 mutation. Am J
Pathol 2006;169:665-672.
van den Bosch T, Kilic E, Paridaens D, de Klein A: Genetics of uveal melanoma and
cutaneous melanoma: two of a kind? Dermatol Res Pract 2010;2010:360136.
Lake SL, Jmor F, Dopierala J, Taktak AF, Coupland SE, Damato BE: Multiplex
ligation-dependent probe amplification of conjunctival melanoma reveals common
BRAF V600E gene mutation and gene copy number changes. Invest Ophthalmol Vis
Sci 2011;52:5598-5604.
109
VII Discussion
Animal models
Experimental animal models are valuable tools for a better understanding of the
pathophysiology of human disease and for identifying novel therapeutic targets. Based on
genetic alterations found in human tumours, multiple models have been generated in
genetically modified or drug-induced animals. As no mouse model of spontaneous and
metastasising uveal melanoma has been described yet, there is an unmet need for such a
model for basic and pre-clinical research. Many groups examined ocular findings of
transgenic mice which mostly represented models of spontaneous skin melanoma or intended
to induce intraocular melanomas with radiation or other compounds (Dithmar, Albert et al.
2000). Although several studies described ocular melanocytic proliferations, most pigmented
intraocular tumours however, revealed to originate from the RPE or failed to metastasise
(Kramer, Powell et al. 1998; Syed, Windle et al. 1998; Albert, Kumar et al. 2004; Tolleson,
Doss et al. 2005; Latendresse, Muskhelishvili et al. 2007). Recently, Schiffner and co-workers
showed in another transgenic mouse breed - with spontaneous skin melanoma - that
pigmented choroidal proliferations mimicked spontaneous uveal melanoma (Schiffner,
Braunger et al. 2014). However, applicability as a model for studying intraocular melanomas
remains questionable. In constant search for a potential mouse model, we evaluated ocular
findings in Hgf-Cdk4R24C mice. These represent a mouse model of spontaneous and
metastasising skin melanoma and their eyes have not yet been examined for ocular
melanomas. (Landsberg, Gaffal et al. 2010). Apart from incidental findings of unusual heavily
pigmented cells in the cornea and conjunctiva, however, no evidence for melanocytic
VII Discussion
110
proliferations in the uvea was found (Kilian, Loeffler et al. 2015, in print). These negative
findings underline genetic differences between uveal, conjunctival and cutaneous melanoma
which result in different pathways of tumourigenesis, pathology and potential therapeutic
targets of these tumours (van den Bosch, Kilic et al. 2010) (chapter VI).
To date, all existing mouse models of uveal melanoma exhibit limitations and one need to
carefully select a model which best qualifies for the specific research questions. To
investigate intraocular tumour biology the best compromise appears to inject intravitreally
cutaneous melanoma cells in order to induce an intraocular melanoma. Despite genetic
differences between cutaneous and uveal melanoma, such a model allows investigations on
intraocular tumour characteristics and immunology in an immunocompetent animal. In our
study on the role of tumour associated macrophages (TAM) in intraocular melanoma we
modified an established murine model. Following the intravitreal injection of murine
cutaneous HCmel12 melanoma cells into the eyes of CX3CR1+/GFP mice, intraocular tumours
showed solid tumour growth, angiotropism, angiogenesis, vasculogenic mimicry with
extravascular matrix patterns as well as inflammatory cell infiltration including macrophages.
In addition, metastases were observed in regional lymph nodes and lungs. However,
metastasis occurred only in mice which previously exhibited extraocular tumour extension.
Thereby, these tumours might have gained access to the conjunctival lymphatics and
metastasised via the lymphatic system. Haematogenously dispersed metastases of tumours
strictly confined to the eye were not observed. Thus, this mouse model does not qualify as a
model for metastasising intraocular melanoma. However, although being of cutaneous origin
intraocular tumours of HCmel12 melanoma cells showed apparent similarities to human uveal
melanoma such as solid tumour growth, angiogenesis, macrophage infiltration and other (vide
supra). These characteristics appear to represent general features of tumour biology since they
VII Discussion
111
can be observed in many solid tumour entities. Thus, tumours of intravitreally injected
HCmel12 melanoma cells qualify as a model for intraocular melanoma and allow studies on
questions related to the tumour’s microenvironment, immunologic aspects as well as
angiogenesis and other vascularisation strategies (chapter III).
Tumour associated macrophages in intraocular melanoma
The eye represents a semi-closed system with access to blood vessels but no lymphatic
vessels. Thereby, the intraocularly injected tumour cells may grow inside an immune
privileged compartment and may develop a microenvironment which can be manipulated
iatrogenically by systemic or intraocular injections. In uveal melanoma and other tumours,
TAM represent a major player within the tumour microenvironment and may contribute to
tumour progression (Allavena, Sica et al. 2008; Mantovani and Sica 2010). As we were
interested in the role of macrophages in intraocular melanoma, we applied our described
intraocular melanoma model to mice with systemically depleted macrophages in comparison
to untreated mice of two different ages (young 8-12 weeks; old 10-12 months). By repeated
intraperitoneal injections of liposome-encapsulated clodronate macrophages were effectively
depleted. Systemic depletion of macrophages can result in tumour growth inhibition and
reduced angiogenesis in cutaneous melanoma (Gazzaniga, Bravo et al. 2007). In our animal
model, TAM did not have a statistical significant impact on tumour size. However, in vitro
results showed increased cell proliferation rates in melanoma cells which were grown in an
environment (supernatant) shaped by unpolarised macrophages (chapter IV). In vivo,
advanced age and the presence of macrophages were individually associated with an
increased occurrence of prognostically significant extravascular matrix patterns (untreated
young vs. untreated old mice: p=0.024; untreated vs. macrophage depleted mice: p=0.014;
VII Discussion
112
chapter IV). This patterned microcirculation characteristic of vasculogenic mimicry was first
described in uveal melanoma (Folberg, Pe'er et al. 1992; Folberg, Rummelt et al. 1993;
Maniotis, Folberg et al. 1999; Folberg, Hendrix et al. 2000). These structures are part of the
extracellular matrix (ECM), share anastomoses with “true” blood vessels and may contribute
to perfusion in aggressive tumours (Maniotis, Folberg et al. 1999; Chen, Zhang et al. 2009;
Seftor, Hess et al. 2012). Aggressive melanomas which need to gain access to oxygen and
nutrients are able to actively remodel the ECM in order to arrange stromal patterns such as
vascular networks (Daniels, Boldt et al. 1996). Besides tumour cells themselves, TAM are
also potentially able to enhance tissue remodelling and extravascular matrix pattern formation
(Seftor, Seftor et al. 2001). Thus, the frequency of prognostically significant extravascular
matrix pattern in aged and untreated mice might be directly related to the crosstalk between
melanoma cells and TAM.
Impact of macrophage polarisation
TAM obtain their functional polarisation phenotype in response to stimuli present in the
tumour microenvironment. Polarised macrophages differ in terms of receptor expression,
effector function as well as cytokine and chemokine production (Mantovani, Sozzani et al.
2002). In many malignant tumours - including uveal melanoma - infiltrating TAM exhibit
predominantly a M2 polarisation phenotype (Mantovani, Sozzani et al. 2002; Pierard, PierardFranchimont et al. 2012). Presence and a higher number of such a M2 phenotype are
associated with tumour favouring properties and are statistically correlated to poor prognosis
(Herwig, Bergstrom et al. 2013).
Our results from the polarisation experiment (chapter IV) display tumour characteristics under
iatrogenically induced tumour microenvironments of extreme M1- or M2 dominance. This
VII Discussion
113
experimental design does not model actual conditions in intraocular tumours but demonstrates
in which ways polarised macrophages may influence tumour characteristics (and thus
prognosis). In vitro as well as in vivo, our results indicate a far more aggressive tumour
phenotype in M2-conditioned tumours. Intraocular tumours of HCmel12 melanoma cells which were previously incubated in M2-dominated conditions - showed an increased
infiltration of inflammatory cells (lymphocytes and macrophages) compared to M1conditioned tumours (p=0.001) (chapter IV). In uveal melanoma, a high infiltration with
lymphocytes and M2 macrophages is statistically correlated with negative prognostic factors
such as monosomy 3 and thus with a poor prognosis (Bronkhorst, Vu et al. 2012). Generally,
M2-polarised TAM are regarded as a paradigm for cancer promoting inflammation. They may
orchestrate tumour growth as well as matrix deposition and stroma remodelling in order to
construct a metastatic niche (Sica 2010). Accordingly, our M2-conditioned tumours exhibited
higher levels of ECM collagen IV positive structures which resulted in an increased tissue
remodelling compared to M1-conditioned tumours (p=0.024). This effect of increased levels
of collagen IV positive structures was also suggestively observed in untreated mice when
compared to systemically macrophage depleted mice (in CX3CR1+/GFP mice chapter IV, in flt1/eGFP BAC mice chapter V), but did not reach statistical significance. In cutaneous
melanoma, collagen IV was found to influence tumour cell mobility and thus contributes to
tumour progression and may possibly represent a prognostic indicator (Pierard, PierardFranchimont et al. 2012). In our model with M2-conditioned tumours, we were able to
directly address increased tissue remodelling rates to a M2-dominated microenvironment. In
addition, MVD was also significantly increased in M2-conditioned tumours compared to M1conditioned tumours (p=0.01). In uveal melanoma and other solid tumours, MVD represents a
well-known histopathological prognostic indicator and is further statistically correlated with
VII Discussion
114
tumour size, number of infiltrating TAM and finally with metastasis (Folberg, Mehaffey et al.
1997; Makitie, Summanen et al. 1999). However, results from the depletion experiment in
CX3CR1+/GFP mice (chapter IV) and flt-1/eGFP BAC mice (chapter V) did not show
significant differences in MVD between macrophage-depleted and untreated mice. This might
be explained by either an insufficient majority of M2 macrophages over M1 macrophages in
untreated mice or by a short time (nine days) of tumour growth in our model. Thus, a M2associated pro-angiogenic influence on the MVD could not emerge. However, in
iatrogenically M2-conditioned tumours such a pro-angiogenic effect on the MVD could be
demonstrated (polarisation experiment chapter IV). This underlines functionally statistical
data of previously reported correlations between the level of TAM infiltration and
histopathological prognostic factors such as extravascular matrix patterns or MVD. Thus in
our model, pro-angiogenic and other tumour-promoting factors could be attributed directly
and functionally to a tumour microenvironment dominated by M2 macrophages.
Impact of advanced age
Advanced age is associated with defects in both the innate and the adaptive arm of the
immune system. The complex process of immunosenescence affects numbers, function, and
early stages of immune cell activation (Gomez, Boehmer et al. 2005; Gomez, Nomellini et al.
2008). A dysfunctional innate immune system represents a contributing factor for patients’
outcomes after injury, inflammation or neoplastic disease in the elderly. Aging may have
different effects on macrophages (as a part of the innate immune system) depending on the
type of tissue (Stout and Suttles 2005; Gomez, Nomellini et al. 2008). In tumours of aged
mice, TAM exhibit more often a M2 polarisation, since macrophages from geriatric mice are
particularly sensitive to M2 phenotype promoting signals (Jackaman, Radley-Crabb et al.
VII Discussion
115
2013). To address and evaluate age-dependent variations and changes in tumour growth
characteristics and macrophage abundance/polarisation we conducted our studies in two
different age groups (young 8-12 weeks; old 10-12 months). We identified differences in
histopathologic features as well as in macrophage polarisation (chapter IV). In our model,
TAM of old mice showed increased expression levels of a M2 marker (CD163), as shown in
flow cytometry analysis. Particularly, tumour infiltrating Gr-1 positive macrophages - which
are derived from the bone marrow, enter the tumour as circulating monocytes and then
proliferate and polarise into macrophages - showed an increased rate of the M2 marker when
compared to their younger counterparts (p=0.08). This effect was slightly below statistical
significance. However, as macrophage polarisation is a continuum and other studies are in
accordance with our observation, the tumour microenvironment in old mice appears to be M2dominated (generally as well as in our experimental design). Such a M2-dominated tumour
microenvironment features a pro-angiogenic basis for the tumour to induce angiogenesis or
other vascularisation strategies. In physiologic and neoplastic conditions, angiogenesis is
altered and delayed in aging due to different impairments in pathways regarding matrix
composition, growth factor expression and inflammatory response (Yamaura and Matsuzawa
1980; Cohen 1994; Sadoun and Reed 2003). In our experiments, we could not retrace an
impact of age on the MVD comparing young and old animals (p=0.542). However, since mice
were terminated at day nine post intraocular tumour cell injection, differences in MVD might
have not reached statistical significance in this early tumour stages (vide supra).
Alternative vascularisation strategies - such as vasculogenic mimicry - gain importance for
tumours in aged mice (and presumably also in humans). In our macrophage-depletion and
macrophage-polarisation studies, advanced age was individually associated with more
frequent extravascular matrix patterns (p=0.024) and a tendency of increased collagen IV
VII Discussion
116
positive ECM levels (p=0.074). Further, an increased appearance of M2 macrophages was
found in aged mice in comparison to young mice (flow cytometry analysis, p=0.08, chapter
IV). This is in accordance with the situation in human uveal and cutaneous melanoma where
age, extravascular matrix patterns, increased collagen IV levels and an increased amount of
M2 macrophages are correlated statistically with a worse prognosis (Folberg, Mehaffey et al.
1997; Makitie, Summanen et al. 2001; Pierard, Pierard-Franchimont et al. 2012).
In our study, these tumour characteristics in old mice are associated with a more aggressive
tumour phenotype and better resemble human tumours. However, most described mouse
models of tumour biology are still conducted in young mice. A study design comprising old
mice is costly in terms of time and expenses. However, considering our results, we concluded
that old mice represent a better tumour model than young mice and should be used for studies
on the tumour microenvironment, vascularisation or tumour associated macrophages.
Tumour associated macrophages and angiogenesis
To investigate the role of macrophages in tumour angiogenesis we applied the same
experimental design (macrophage-depleted mice in comparison to untreated mice) in flt1/eGFP BAC mice, which exhibit GFP expression in all murine smaller vessels. Dual
labelling of CD31 (endothelial vessel marker) and Ki67 (proliferation marker) revealed active
angiogenesis which was predominantly found at tumour margins located near host structures.
This finding is in accordance with a study which found high numbers of proliferating
endothelial cells at tumour margins of poorly differentiated (and thus aggressive) tumours
(Raluca, Cimpean et al. 2015). In these regions the tumour might have orchestrated the
microenvironment towards pro-angiogenic conditions in order to induce angiogenesis from
surrounding host structures. Studies investigating MVD and the proportion of active
VII Discussion
117
angiogenesis in cutaneous melanoma and other tumour entities showed that the number of
proliferating endothelial cells (CD31+/CD34+/Nestin+ and Ki67+) but not MVD was correlated
with tumour stage (Baeten, Castermans et al. 2006; Hillen, van de Winkel et al. 2006; Kruger,
Stefansson et al. 2013). Thus, the proportion of proliferating endothelial cells might be also an
indicator for tumour progression and possibly also for the patient’s outcome. In our study, in
untreated mice 39.1% of endothelial vessels showed proliferation activity compared to 30.9%
of proliferating vessels in macrophage-depleted mice (p=0.114). As tumours in untreated
mice showed a tendency of an increased rate of active angiogenesis in comparison to
macrophage-depleted mice, TAM might have directly shaped the tumour microenvironment
by secreting pro-angiogenic factors – probably in response to a crosstalk with melanoma cells
due to hypoxic conditions. TAM are in general able to stimulate proliferation and migration
of endothelial cells (reviewed in (Lamagna, Aurrand-Lions et al. 2006)) and may thereby
influence the rate of active angiogenesis in aggressive tumours. This indicates another
pathway by which TAM may directly affect histopathologic tumour characteristics.
The origin of tumour vessels
When tumours grow beyond the size which can be supplied by diffusion from surrounding
structures, they are forced to angiotropic growth and/or to attract blood vessels from the
surrounding stroma and/or to initiate other vascularisation strategies. This process is regulated
by a variety of pro- and anti-angiogenic factors, and it is an indispensable prerequisite for
further growth of the tumour (Carmeliet and Jain 2000). In uveal, cutaneous and oral
melanoma, numerous studies have focused on angiogenesis and other mechanisms of tumour
vascularisation. Besides sprouting angiogenesis, also patterns of mosaic vessels, tumour-lined
vasculature, co-option of host vessels and networks of vessel-like structures not lined by
VII Discussion
118
endothelial cells (vasculogenic mimicry) have been described (reviewed in (Pastushenko,
Vermeulen et al. 2014), (Hendrix, Seftor et al. 2002; Lee, Nagai et al. 2002; Zhang, Zhang et
al. 2006). These studies used PAS and immunohistochemical staining of endothelial vessels,
ECM and other structures (e.g. CD31, collagen IV) to visualise and investigate the
characteristics of melanoma vascularisation. In our study, we further addressed the origin of
tumour vessels by using the flt-1/eGFP BAC mouse with endogenous endothelial GFP
expression. Almost all intratumoural vessels exhibited a co-expression of CD31 and GFP and
were thus of host origin (chapter V). Apparently, rapidly growing tumours are dependent on a
highly pro-angiogenic microenvironment in which they are able to adopt host vessels and
initiate angiogenic sprouting from incorporated vessels as a main vascularisation strategy.
This is in accordance with the general doctrine that angiogenesis represents the main pathway
of tumour vascularisation. However, a minor number of intratumoural vessels in the described
flt-1/eGFP BAC mouse model were found to be solely marked by the CD31 antibody and
must, subsequently, be of other origin than angiogenesis. These vessels might have originated
from two other vascularisation strategies: vasculogenesis (endothelial precursor cells) or
tumour-derived vessels. To distinguish its origin further investigation of their surface markers
are needed. Circulating endothelial precursors - shed from the vessel wall or mobilised from
the bone marrow - can also contribute to tumour vascularisation in terms of vasculogenesis
(Rafii 2000). Such endothelial precursor cells express characteristic vessel markers (e.g.
CD31, CD34, VEGFR-2) along with the stem cell marker CD133 (Timmermans, Plum et al.
2009). To identify such vessels formed by endothelial progenitor cells we (intend to) perform
dual staining of CD31 and CD133.
Should we be able to demonstrate that these vessels originated from vasculogenesis by being
built of endothelial progenitor cells (CD31+CD133+GFP-), this would explain heterogeneity of
VII Discussion
119
tumour vascularisation in our model. Further, flt-1/eGFP BAC mice would represent a potent
model to distinguish between vasculogenesis (CD31+CD133+GFP- vessels) and angiogenesis
(CD31+CD133+GFP+ vessels). This might offer a new model to study these two different
forms of vascularisation in further pathologies like injury, inflammatory processes or other
tumour entities.
On the other hand, if CD31+GFP- vessels in flt-1/eGFP BAC mice reveal not to have
originated from vasculogenesis (CD133-), another hypothesis of their origin might be a
derivation from cancer stem cells. This hypothesis might further be supported by dual
labelling of CD31 and a melanoma marker (e.g. HMB45/MART-1). Two studies in
glioblastoma showed that intratumoural blood vessels expressed genetic markers of cancer
cells (with stem cell like properties). This suggests that these blood vessels were of tumour
origin (Ricci-Vitiani, Pallini et al. 2010; Wang, Chadalavada et al. 2010). Another group
describes „tumour endothelial cells (TEC)“ as intratumoural vessels of tumour origin with
highly angiogenic phenotypes and higher proliferative and migratory abilities than „normal
endothelial cells“ (=host-derived, angiogenic endothelial cells) (Ohmura-Kakutani, Akiyama
et al. 2014). However, these conclusions were based on in vitro studies and their translation
into in vivo conditions remains unknown. However, in an ischemic nude mouse model with
inoculated human aggressive melanoma cells, newly formed vasculature within the tumour
showed endothelial cells of human and of mouse origin in a linear arrangement. Thus, besides
angiogenic murine vessels, human melanoma cells may have acquired endothelial-like
morphology and were incorporated in capillaries. Along these lines, the tumour may actively
contribute to neovascularisation (Mihic-Probst, Ikenberg et al. 2012).
Our ongoing studies may reveal the origin of CD31+GFP- intratumoural vessels in flt-1/eGFP
BAC mice and either offer a new model for distinguishing vasculogenesis from angiogenesis
VII Discussion
120
or contribute to recent discussions regarding tumour-derived endothelial vessels. This would
represent the first orthotopic animal model of such tumour-derived endothelial cells in
melanoma.
Outlook
Manipulating the tumour microenvironment may offer an approach for novel therapeutic
strategies. As TAM represent major orchestrators of the tumour microenvironment, targeting
TAM and limiting their pro-angiogenic and tumour-favouring properties may be promising.
This could be achieved by blocking the polarisation cascade into an M2 phenotype by e.g. a
Stat6 inhibitor (Binnemars- Postma et al. 2015, Poster at the ECC 2015) or by repolarising
TAM from the M2 into a tumouricidal M1 phenotype (e.g. by zoledronic acid) and thereby
moving the balance toward a tumouricidal immune function. However, when considering
future systemic therapies, which alter macrophage properties, other important macrophage
functions in resistance against infections or wound healing need to be respected. Further
(animal) studies on pathways of secreted cytokines or chemokines are needed which promote
such a pro-angiogenic and tumour-favouring microenvironment. Altering these pathways
might potentially enable us to help control tumour progression and metastasis. As there are
several promising therapeutic options for the primary intraocular tumour, such novel
therapeutic strategies will be of interest regarding the prevention of metastasis of uveal
melanoma and improving patients’ outcome.
Summary
No spontaneous melanoma mouse model has been described yet, thus, all murine models are
subject to certain limitation. To investigate tumour biology and the role of macrophages in
VII Discussion
121
intraocular melanoma, we modified an established mouse model by injecting murine
cutaneous HCmel12 melanoma cells into the vitreous of mouse eyes. Intraocular tumours
exhibited characteristic histopathologic features similar to human uveal melanoma, which
were influenced by advanced age of mice and by the presence of tumour associated
macrophages (TAM). The frequency of prognostically significant extravascular matrix
patterns and level of active angiogenesis were directly correlated with the presence of TAM.
Mean vascular density, collagen IV-positive extracellular matrix and the amount of
infiltrating immune cells were significantly increased in tumours with a M2-dominated
microenvironment. Old mice exhibited histopathologic and immunologic features, which
better resembled human tumours. Thus, we suggest to use mice of advanced age in future
studies on the tumour microenvironment. Angiogenesis represented the main vascularisation
strategy, however, the origin of few CD31+GFP- vessels – originated from either
vasculogenesis or tumour-derived endothelial vessels - remains subject to ongoing
investigations.
Taken together, an aged immunoprofile provides a basis for tumour growth with M2
macrophages being directly involved into a more aggressive tumour phenotype.
122
VIII References
Albert, D. M., A. Kumar, et al. (2004). "Effectiveness of 1alpha-hydroxyvitamin D2 in
inhibiting tumor growth in a murine transgenic pigmented ocular tumor model." Arch
Ophthalmol 122(9): 1365-9.
Allavena, P., A. Sica, et al. (2008). "The Yin-Yang of tumor-associated macrophages in
neoplastic progression and immune surveillance." Immunol Rev 222: 155-61.
Allavena, P., A. Sica, et al. (2008). "The inflammatory micro-environment in tumor
progression: the role of tumor-associated macrophages." Crit Rev Oncol Hematol
66(1): 1-9.
Arepalli, S., S. Kaliki, et al. (2015). "Choroidal metastases: origin, features, and therapy."
Indian J Ophthalmol 63(2): 122-7.
Arnold, L., A. Henry, et al. (2007). "Inflammatory monocytes recruited after skeletal muscle
injury switch into antiinflammatory macrophages to support myogenesis." J Exp Med
204(5): 1057-69.
Asahara, T., T. Murohara, et al. (1997). "Isolation of putative progenitor endothelial cells for
angiogenesis." Science 275(5302): 964-7.
Baeten, C. I., K. Castermans, et al. (2006). "Proliferating endothelial cells and leukocyte
infiltration as prognostic markers in colorectal cancer." Clin Gastroenterol Hepatol
4(11): 1351-7.
Bakalian, S., J. C. Marshall, et al. (2008). "Molecular pathways mediating liver metastasis in
patients with uveal melanoma." Clin Cancer Res 14(4): 951-6.
Biswas, S. K. and A. Mantovani (2010). "Macrophage plasticity and interaction with
lymphocyte subsets: cancer as a paradigm." Nat Immunol 11(10): 889-96.
Bronkhorst, I. H. and M. J. Jager (2012). "Uveal Melanoma: The Inflammatory
Microenvironment." J Innate Immun.
Bronkhorst, I. H. and M. J. Jager (2013). "Inflammation in uveal melanoma." Eye (Lond)
27(2): 217-23.
Bronkhorst, I. H., T. M. Jehs, et al. (2014). "Effect of hypoxic stress on migration and
characteristics of monocytes in uveal melanoma." JAMA Ophthalmol 132(5): 614-21.
Bronkhorst, I. H., T. H. Vu, et al. (2012). "Different subsets of tumor-infiltrating lymphocytes
correlate with macrophage influx and monosomy 3 in uveal melanoma." Invest
Ophthalmol Vis Sci 53(9): 5370-8.
Burrell, K. and G. Zadeh (2012). Molecular Mechanisms of Tumor Angiogenesis.
Carmeliet, P. and R. K. Jain (2000). "Angiogenesis in cancer and other diseases." Nature
407(6801): 249-57.
Catalano, V., A. Turdo, et al. (2013). "Tumor and its microenvironment: a synergistic
interplay." Semin Cancer Biol 23(6 Pt B): 522-32.
Chang, Y. S., E. di Tomaso, et al. (2000). "Mosaic blood vessels in tumors: frequency of
cancer cells in contact with flowing blood." Proc Natl Acad Sci U S A 97(26): 1460813.
Chen, L., S. Zhang, et al. (2009). "A pilot study of vasculogenic mimicry
immunohistochemical expression in intraocular melanoma model." Oncol Rep 21(4):
989-94.
VIII References
123
Chroscinski, D., D. Sampey, et al. (2015). "Registered report: tumour vascularization via
endothelial differentiation of glioblastoma stem-like cells." Elife 4.
Cohen, H. J. (1994). "Biology of aging as related to cancer." Cancer 74(7 Suppl): 2092-100.
Damato, B., A. Eleuteri, et al. (2011). "Estimating prognosis for survival after treatment of
choroidal melanoma." Prog Retin Eye Res 30(5): 285-95.
Daniels, K. J., H. C. Boldt, et al. (1996). "Expression of type VI collagen in uveal melanoma:
its role in pattern formation and tumor progression." Lab Invest 75(1): 55-66.
Diaz, C. E., D. Rusciano, et al. (1999). "B16LS9 melanoma cells spread to the liver from the
murine ocular posterior compartment (PC)." Curr Eye Res 18(2): 125-9.
Dithmar, S., D. M. Albert, et al. (2000). "Animal models of uveal melanoma." Melanoma Res
10(3): 195-211.
Eagle, R. C., Jr. (2013). "The pathology of ocular cancer." Eye (Lond) 27(2): 128-36.
Ehlers, J. P., L. Worley, et al. (2008). "Integrative genomic analysis of aneuploidy in uveal
melanoma." Clin Cancer Res 14(1): 115-22.
Field, M. G. and J. W. Harbour (2014). "Recent developments in prognostic and predictive
testing in uveal melanoma." Curr Opin Ophthalmol 25(3): 234-9.
Folberg, R., M. J. Hendrix, et al. (2000). "Vasculogenic mimicry and tumor angiogenesis."
Am J Pathol 156(2): 361-81.
Folberg, R., M. Mehaffey, et al. (1997). "The microcirculation of choroidal and ciliary body
melanomas." Eye (Lond) 11 ( Pt 2): 227-38.
Folberg, R., J. Pe'er, et al. (1992). "The morphologic characteristics of tumor blood vessels as
a marker of tumor progression in primary human uveal melanoma: a matched casecontrol study." Hum Pathol 23(11): 1298-305.
Folberg, R., V. Rummelt, et al. (1993). "The prognostic value of tumor blood vessel
morphology in primary uveal melanoma." Ophthalmology 100(9): 1389-98.
Folkman, J. (1971). "Tumor angiogenesis: therapeutic implications." N Engl J Med 285(21):
1182-6.
Foss, A. J., R. A. Alexander, et al. (1996). "Microvessel count predicts survival in uveal
melanoma." Cancer Res 56(13): 2900-3.
Gazzaniga, S., A. I. Bravo, et al. (2007). "Targeting tumor-associated macrophages and
inhibition of MCP-1 reduce angiogenesis and tumor growth in a human melanoma
xenograft." J Invest Dermatol 127(8): 2031-41.
Gilkes, D. M., G. L. Semenza, et al. "Hypoxia and the extracellular matrix: drivers of tumour
metastasis." Nat Rev Cancer 14(6): 430-9.
Gomez, C. R., E. D. Boehmer, et al. (2005). "The aging innate immune system." Curr Opin
Immunol 17(5): 457-62.
Gomez, C. R., V. Nomellini, et al. (2008). "Innate immunity and aging." Exp Gerontol 43(8):
718-28.
Gordon, S. and F. O. Martinez (2010). "Alternative activation of macrophages: mechanism
and functions." Immunity 32(5): 593-604.
Griewank, K. G., H. Westekemper, et al. (2013). "Conjunctival melanomas harbor BRAF and
NRAS mutations and copy number changes similar to cutaneous and mucosal
melanomas." Clin Cancer Res 19(12): 3143-52.
Grossniklaus, H. E. (1998). "Tumor vascularity and hematogenous metastasis in experimental
murine intraocular melanoma." Trans Am Ophthalmol Soc 96: 721-52.
Hanahan, D. and R. A. Weinberg (2011). "Hallmarks of cancer: the next generation." Cell
144(5): 646-74.
VIII References
124
Hendrix, M. J., E. A. Seftor, et al. (2003). "Vasculogenic mimicry and tumour-cell plasticity:
lessons from melanoma." Nat Rev Cancer 3(6): 411-21.
Hendrix, M. J., R. E. Seftor, et al. (2002). "Transendothelial function of human metastatic
melanoma cells: role of the microenvironment in cell-fate determination." Cancer Res
62(3): 665-8.
Herwig, M. C., C. Bergstrom, et al. (2013). "M2/M1 ratio of tumor associated macrophages
and PPAR-gamma expression in uveal melanomas with class 1 and class 2 molecular
profiles." Exp Eye Res 107: 52-8.
Herz, K., J. C. Heinemann, et al. (2012). "Live monitoring of small vessels during
development and disease using the flt-1 promoter element." Basic Res Cardiol 107(2):
257.
Hillen, F. and A. W. Griffioen (2007). "Tumour vascularization: sprouting angiogenesis and
beyond." Cancer Metastasis Rev 26(3-4): 489-502.
Hillen, F., A. van de Winkel, et al. (2006). "Proliferating endothelial cells, but not microvessel
density, are a prognostic parameter in human cutaneous melanoma." Melanoma Res
16(5): 453-7.
Hornung, V., F. Bauernfeind, et al. (2008). "Silica crystals and aluminum salts activate the
NALP3 inflammasome through phagosomal destabilization." Nat Immunol 9(8): 84756.
Hurst, E. A., J. W. Harbour, et al. (2003). "Ocular melanoma: a review and the relationship to
cutaneous melanoma." Arch Dermatol 139(8): 1067-73.
Hussein, M. R. (2006). "Tumour-associated macrophages and melanoma tumourigenesis:
integrating the complexity." Int J Exp Pathol 87(3): 163-76.
Ioachim, E., A. Charchanti, et al. (2002). "Immunohistochemical expression of extracellular
matrix components tenascin, fibronectin, collagen type IV and laminin in breast
cancer: their prognostic value and role in tumour invasion and progression." Eur J
Cancer 38(18): 2362-70.
Jackaman, C., H. G. Radley-Crabb, et al. (2013). "Targeting macrophages rescues age-related
immune deficiencies in C57BL/6J geriatric mice." Aging Cell 12(3): 345-57.
Jager, M. J., L. V. Ly, et al. (2011). "Macrophages in uveal melanoma and in experimental
ocular tumor models: Friends or foes?" Prog Retin Eye Res 30(2): 129-46.
Jung, S., J. Aliberti, et al. (2000). "Analysis of fractalkine receptor CX(3)CR1 function by
targeted deletion and green fluorescent protein reporter gene insertion." Mol Cell Biol
20(11): 4106-14.
Kaliki, S., C. L. Shields, et al. (2015). "Uveal melanoma: estimating prognosis." Indian J
Ophthalmol 63(2): 93-102.
Keating, G. M. (2014). "Bevacizumab: a review of its use in advanced cancer." Drugs 74(16):
1891-925.
Kilian, M. M., K. U. Loeffler, et al. (2015). "Intravitreally Injected HCmel12 Melanoma Cells
Serve as a Mouse Model of Tumor Biology of Intraocular Melanoma." Curr Eye Res:
1-8.
Kramer, T. R., M. B. Powell, et al. (1998). "Pigmented uveal tumours in a transgenic mouse
model." Br J Ophthalmol 82(8): 953-60.
Krecicki, T., M. Zalesska-Krecicka, et al. (2001). "Expression of type IV collagen and matrix
metalloproteinase-2 (type IV collagenase) in relation to nodal status in laryngeal
cancer." Clin Otolaryngol Allied Sci 26(6): 469-72.
VIII References
125
Kruger, K., I. M. Stefansson, et al. (2013). "Microvessel proliferation by co-expression of
endothelial nestin and Ki-67 is associated with a basal-like phenotype and aggressive
features in breast cancer." Breast 22(3): 282-8.
Kujala, E., T. Makitie, et al. (2003). "Very long-term prognosis of patients with malignant
uveal melanoma." Invest Ophthalmol Vis Sci 44(11): 4651-9.
Lake, S. L., F. Jmor, et al. (2011). "Multiplex ligation-dependent probe amplification of
conjunctival melanoma reveals common BRAF V600E gene mutation and gene copy
number changes." Invest Ophthalmol Vis Sci 52(8): 5598-604.
Lamagna, C., M. Aurrand-Lions, et al. (2006). "Dual role of macrophages in tumor growth
and angiogenesis." J Leukoc Biol 80(4): 705-13.
Landsberg, J., E. Gaffal, et al. (2010). "Autochthonous primary and metastatic melanomas in
Hgf-Cdk4 R24C mice evade T-cell-mediated immune surveillance." Pigment Cell
Melanoma Res 23(5): 649-60.
Landsberg, J., J. Kohlmeyer, et al. (2012). "Melanomas resist T-cell therapy through
inflammation-induced reversible dedifferentiation." Nature 490(7420): 412-6.
Latendresse, J. R., L. Muskhelishvili, et al. (2007). "Two cases of uveal amelanotic melanoma
in transgenic Tyr-HRAS+ Ink4a/Arf heterozygous mice." Toxicol Pathol 35(6): 82732.
Lattier, J. M., H. Yang, et al. (2013). "Host pigment epithelium-derived factor (PEDF)
prevents progression of liver metastasis in a mouse model of uveal melanoma." Clin
Exp Metastasis.
Lee, Y. J., N. Nagai, et al. (2002). "Angioarchitecture of primary oral malignant melanomas."
J Histochem Cytochem 50(11): 1555-62.
Ly, L. V., A. Baghat, et al. (2010). "In aged mice, outgrowth of intraocular melanoma
depends on proangiogenic M2-type macrophages." J Immunol 185(6): 3481-8.
Ly, L. V., O. F. Odish, et al. (2010). "Intravascular presence of tumor cells as prognostic
parameter in uveal melanoma: a 35-year survey." Invest Ophthalmol Vis Sci 51(2):
658-65.
Maat, W., L. V. Ly, et al. (2008). "Monosomy of chromosome 3 and an inflammatory
phenotype occur together in uveal melanoma." Invest Ophthalmol Vis Sci 49(2): 50510.
Makitie, T., P. Summanen, et al. (1999). "Microvascular density in predicting survival of
patients with choroidal and ciliary body melanoma." Invest Ophthalmol Vis Sci
40(11): 2471-80.
Makitie, T., P. Summanen, et al. (2001). "Tumor-infiltrating macrophages (CD68(+) cells)
and prognosis in malignant uveal melanoma." Invest Ophthalmol Vis Sci 42(7): 141421.
Mallikarjuna, K., V. Pushparaj, et al. (2007). "Expression of epidermal growth factor receptor,
ezrin, hepatocyte growth factor, and c-Met in uveal melanoma: an
immunohistochemical study." Curr Eye Res 32(3): 281-90.
Mallone, S., E. De Vries, et al. (2012). "Descriptive epidemiology of malignant mucosal and
uveal melanomas and adnexal skin carcinomas in Europe." Eur J Cancer 48(8): 116775.
Maniotis, A. J., R. Folberg, et al. (1999). "Vascular channel formation by human melanoma
cells in vivo and in vitro: vasculogenic mimicry." Am J Pathol 155(3): 739-52.
Mantovani, A., F. Marchesi, et al. (2007). "Inflammation and cancer: breast cancer as a
prototype." Breast 16 Suppl 2: S27-33.
VIII References
126
Mantovani, A. and A. Sica (2010). "Macrophages, innate immunity and cancer: balance,
tolerance, and diversity." Curr Opin Immunol 22(2): 231-7.
Mantovani, A., A. Sica, et al. (2004). "The chemokine system in diverse forms of macrophage
activation and polarization." Trends Immunol 25(12): 677-86.
Mantovani, A., S. Sozzani, et al. (2002). "Macrophage polarization: tumor-associated
macrophages as a paradigm for polarized M2 mononuclear phagocytes." Trends
Immunol 23(11): 549-55.
Mehnert, J. M. and H. M. Kluger (2012). "Driver mutations in melanoma: lessons learned
from bench-to-bedside studies." Curr Oncol Rep 14(5): 449-57.
Mendoza, M. and C. Khanna (2009). "Revisiting the seed and soil in cancer metastasis." Int J
Biochem Cell Biol 41(7): 1452-62.
Mihic-Probst, D., K. Ikenberg, et al. (2012). "Tumor cell plasticity and angiogenesis in human
melanomas." PLoS One 7(3): e33571.
Mosser, D. M. and X. Zhang (2008). "Activation of murine macrophages." Curr Protoc
Immunol Chapter 14: Unit 14 2.
Mueller, A. J., A. J. Maniotis, et al. (2002). "An orthotopic model for human uveal melanoma
in SCID mice." Microvasc Res 64(2): 207-13.
Nathanson, S. D. (2003). "Insights into the mechanisms of lymph node metastasis." Cancer
98(2): 413-23.
Ohmura-Kakutani, H., K. Akiyama, et al. (2014). "Identification of tumor endothelial cells
with high aldehyde dehydrogenase activity and a highly angiogenic phenotype." PLoS
One 9(12): e113910.
Onken, M. D., A. Y. Lin, et al. (2005). "Association between microarray gene expression
signature and extravascular matrix patterns in primary uveal melanomas." Am J
Ophthalmol 140(4): 748-9.
Onken, M. D., L. A. Worley, et al. (2012). "Collaborative ocular oncology group report
number 1: prospective validation of a multi-gene prognostic assay in uveal
melanoma." Ophthalmology 119(8): 1596-603.
Onken, M. D., L. A. Worley, et al. (2004). "Gene expression profiling in uveal melanoma
reveals two molecular classes and predicts metastatic death." Cancer Res 64(20):
7205-9.
Pastushenko, I., P. B. Vermeulen, et al. (2014). "Mechanisms of tumour vascularization in
cutaneous malignant melanoma: clinical implications." Br J Dermatol 171(2): 220-33.
Pierard, G. E., C. Pierard-Franchimont, et al. (2012). "Malignant melanoma and its stromal
nonimmune microecosystem." J Oncol 2012: 584219.
Prescher, G., N. Bornfeld, et al. (1996). "Prognostic implications of monosomy 3 in uveal
melanoma." Lancet 347(9010): 1222-5.
Rafii, S. (2000). "Circulating endothelial precursors: mystery, reality, and promise." J Clin
Invest 105(1): 17-9.
Raluca, B. A., A. M. Cimpean, et al. (2015). "Endothelial Cell Proliferation and Vascular
Endothelial Growth Factor Expression in Primary Colorectal Cancer and
Corresponding Liver Metastases." Asian Pac J Cancer Prev 16(11): 4549-53.
Ricci-Vitiani, L., R. Pallini, et al. (2010). "Tumour vascularization via endothelial
differentiation of glioblastoma stem-like cells." Nature 468(7325): 824-8.
Sadoun, E. and M. J. Reed (2003). "Impaired angiogenesis in aging is associated with
alterations in vessel density, matrix composition, inflammatory response, and growth
factor expression." J Histochem Cytochem 51(9): 1119-30.
VIII References
127
Schiffner, S., B. M. Braunger, et al. (2014). "Tg(Grm1) transgenic mice: a murine model that
mimics spontaneous uveal melanoma in humans?" Exp Eye Res 127: 59-68.
Schmid, M. C. and J. A. Varner (2010). "Myeloid cells in the tumor microenvironment:
modulation of tumor angiogenesis and tumor inflammation." J Oncol 2010: 201026.
Seftor, R. E., A. R. Hess, et al. (2012). "Tumor cell vasculogenic mimicry: from controversy
to therapeutic promise." Am J Pathol 181(4): 1115-25.
Seftor, R. E., E. A. Seftor, et al. (2001). "Cooperative interactions of laminin 5 gamma2
chain, matrix metalloproteinase-2, and membrane type-1-matrix/metalloproteinase are
required for mimicry of embryonic vasculogenesis by aggressive melanoma." Cancer
Res 61(17): 6322-7.
Shields, C. L., S. Kaliki, et al. (2013). "American Joint Committee on Cancer classification of
posterior uveal melanoma (tumor size category) predicts prognosis in 7731 patients."
Ophthalmology 120(10): 2066-71.
Shields, C. L., J. Manalac, et al. (2014). "Choroidal melanoma: clinical features,
classification, and top 10 pseudomelanomas." Curr Opin Ophthalmol 25(3): 177-85.
Sica, A. (2010). "Role of tumour-associated macrophages in cancer-related inflammation."
Exp Oncol 32(3): 153-8.
Sica, A. and A. Mantovani (2012). "Macrophage plasticity and polarization: in vivo veritas." J
Clin Invest 122(3): 787-95.
Singh, A. D., C. L. Shields, et al. (2001). "Prognostic factors in uveal melanoma." Melanoma
Res 11(3): 255-63.
Singh, A. D., M. E. Turell, et al. (2011). "Uveal melanoma: trends in incidence, treatment,
and survival." Ophthalmology 118(9): 1881-5.
Singh, P. and A. Singh (2012). "Choroidal melanoma." Oman J Ophthalmol 5(1): 3-9.
Sisley, K., I. G. Rennie, et al. (1990). "Cytogenetic findings in six posterior uveal melanomas:
involvement of chromosomes 3, 6, and 8." Genes Chromosomes Cancer 2(3): 205-9.
Stout, R. D. and J. Suttles (2005). "Immunosenescence and macrophage functional plasticity:
dysregulation of macrophage function by age-associated microenvironmental
changes." Immunol Rev 205: 60-71.
Syed, N. A., J. J. Windle, et al. (1998). "Transgenic mice with pigmented intraocular tumors:
tissue of origin and treatment." Invest Ophthalmol Vis Sci 39(13): 2800-5.
Timar, J. and J. Toth (2000). "Tumor sinuses- vascular channels." Pathol Oncol Res 6(2): 836.
Timmermans, F., J. Plum, et al. (2009). "Endothelial progenitor cells: identity defined?" J Cell
Mol Med 13(1): 87-102.
Toivonen, P., T. Makitie, et al. (2004). "Microcirculation and tumor-infiltrating macrophages
in choroidal and ciliary body melanoma and corresponding metastases." Invest
Ophthalmol Vis Sci 45(1): 1-6.
Tolleson, W. H., J. C. Doss, et al. (2005). "Spontaneous uveal amelanotic melanoma in
transgenic Tyr-RAS+ Ink4a/Arf-/- mice." Arch Ophthalmol 123(8): 1088-94.
Tormo, D., A. Ferrer, et al. (2006). "Rapid growth of invasive metastatic melanoma in
carcinogen-treated hepatocyte growth factor/scatter factor-transgenic mice carrying an
oncogenic CDK4 mutation." Am J Pathol 169(2): 665-72.
van den Bosch, T., E. Kilic, et al. (2010). "Genetics of uveal melanoma and cutaneous
melanoma: two of a kind?" Dermatol Res Pract 2010: 360136.
van Rooijen, N. and E. van Kesteren-Hendrikx (2003). ""In vivo" depletion of macrophages
by liposome-mediated "suicide"." Methods Enzymol 373: 3-16.
VIII References
128
Verheul, H. M., E. E. Voest, et al. (2004). "Are tumours angiogenesis-dependent?" J Pathol
202(1): 5-13.
Virgili, G., G. Gatta, et al. (2007). "Incidence of uveal melanoma in Europe." Ophthalmology
114(12): 2309-15.
Wang, R., K. Chadalavada, et al. (2010). "Glioblastoma stem-like cells give rise to tumour
endothelium." Nature 468(7325): 829-33.
Yamaura, H. and T. Matsuzawa (1980). "Decrease in capillary growth during aging." Exp
Gerontol 15(2): 145-50.
Zhang, S., D. Zhang, et al. (2006). "Morphologic research of microcirculation patterns in
human and animal melanoma." Med Oncol 23(3): 403-9.
129
IX Summary
Marta Kilian
The Role of Macrophages in Murine Intraocular Melanoma
Uveal melanoma is the most common intraocular tumour in the adult Caucasian population.
While local treatment of the primary ocular tumour is very effective, prognosis worsens
dramatically when metastatic disease appears. Hepatic micrometastases may establish early
and remain dormant for decades. Understanding its pathways and preventing hematogeneous
metastasis is a major goal uveal melanoma research. Several prognostic factors have been
determined so far, including clinical (e.g. age), histopathologic, (e.g. tumour size, mean
vascular density [MVD], vasculogenic mimicry/ extravascular matrix patterns, tumour
associated macrophages [TAM]) and genetic characteristics. However, in vivo studies in mice
remain challenging. All murine models are subject to restrictions as spontaneous uveal
melanoma has not been described in wild type mice, yet. In search for spontaneous ocular
melanomas, among other groups, we examined the eyes of transgenic mice which exhibit
spontaneous skin melanoma. Apart from an incidental and unusual finding of heavily
pigmented cells in the cornea, Hgf-Cdk4 mice did not show any melanocytic proliferations in
the uvea (chapter VI). However, this underlines genetic and clinical differences between uveal
and skin melanoma.
Besides mouse models in which human uveal melanoma cells are injected into the eye of
immunodeficient mice, there are also other well established mouse models like intraocular
inoculation with murine cutaneous melanoma cells. Despite genetic differences, such a model
IX Summary
130
allows investigations on intraocular tumour biology in an immunocompetent animal. We
modified such an established mouse model by injecting HCmel12 murine cutaneous
melanoma cells into the ocular vitreous of CX3CR1+/GFP mice and flt-1/eGFP BAC mice of
different age groups. We were particularly interested in the role of macrophages in intraocular
melanoma of aged mice (10-12 months). Intraocular tumours of HCmel12 melanoma cells
showed solid and aggressive growth with angiotropism, angiogenesis, vasculogenic mimicry/
extravascular matrix patterns, inflammatory cell infiltration with pronounced macrophage
proportions, but no hematogenously dispersed metastases (chapter III). In our studies, old
mice exhibited more aggressive tumours (histologically) and an increased number of M2
macrophages (flow cytometry analysis) compared to young mice. Thus, aged mice better
resembled uveal melanomas growth characteristics. Hence, our intraocular tumour model in
mice of advanced age offers the opportunity to study the tumour microenvironment in a semiclosed system (eye, blood but no lymphatic vessels), which can be manipulated iatrogenically
by systemic or intraocular injections.
To investigate the general role of macrophages in intraocular melanoma we examined
histologically tumour growth characteristics of systemically macrophage-depleted mice in
comparison to untreated mice. The frequency of prognostically significant extravascular
matrix patterns (chapter IV) and active angiogenesis (chapter V) was increased in untreated
mice compared to macrophage-depleted mice. These results suggest that TAM promote
different tumour vascularisation strategies in intraocular melanoma and thereby contribute to
tumour progression. Further, we studied the role of polarised macrophages which can be
functionally subdivided into a M1 phenotype with tumouricidal and a M2 phenotype with
tumour-favouring properties. HCmel12 cells were incubated in vitro in a M1- or M2dominated environment and then injected into the mouse eye. Tumours of M2-conditioned
IX Summary
131
melanoma cells showed an increased TAM infiltration, more collagen IV stroma patterns and
a higher MVD compared to M1-conditioned tumours. Thus, aggressive, pro-angiogenic and
tumour promoting factors such as inflammatory cell infiltration, stroma remodelling and
MVD could be directly attributed to the existence of TAM, in particular to M2 macrophages
(chapter IV). In flt-1/eGFP BAC mice we further found that the majority of tumour vessels
were CD31+GFP+ and thus of angiogenic host origin (chapter V). Hence, the main
vascularisation strategy in our tumour model was co-option of host vasculature followed by
induction of angiogenesis from existing host vessels. However, some vessels were observed
to be CD31+GFP- and thus of vasculogenic or tumour-derived origin. Ongoing studies using
stem cell and melanoma markers might reveal their final origin and offer potentially new
animal models to distinguish between angiogenesis and vasculogenesis in tumours or other
pathologies.
Taken together, an aged immunoprofile with an increased number of M2 macrophages
provides a tumour-favouring microenvironment and might be a basis for tumour progression
and metastasis. These findings could be a target for future therapeutic interventions in
different tumour entities.
132
X Zusammenfassung
Marta Kilian
Rolle der Makrophagen beim intraokularen Melanom der Maus
Das uveale Melanom ist der häufigste intraokulare Tumor bei der erwachsenen kaukasischen
Bevölkerung. Die Behandlung des primären okularen Tumors bietet vielversprechende
Therapiemöglichkeiten, jedoch verschlechtert sich die Prognose des Patienten enorm, wenn
Metastasen klinisch in Erscheinung treten. Mikrometastasen in der Leber können sich sehr
früh etablieren, zugleich aber für mehrere Jahrzehnte “stumm” verbleiben. Das Augenmerk
vieler Studien über das uveale Melanom richtet sich daher auf die Signalwege und die
Prävention
der
hämatogenen
Metastasierung.
Zahlreiche
Eigenschaften
von
Aderhautmelanomen konnten bislang statistisch mit der Prognose des Patienten korreliert
werden. Hierzu gehören klinische (z.B. Alter des Patienten), histopathologische (z.B.
Tumorgröße, Gefäßdichte, vasculogenic mimicry, tumor-assoziierte Makrophagen) und
genetische Charakteristiken. In vivo Studien in Mäusen gestalten sich jedoch nach wie vor
schwierig: Alle bisherigen Mausmodelle besitzen gewisse Einschränkungen, da spontane
uveale Melanome in Wildtyp-Mäusen noch nicht beschrieben wurden und demnach als
adäquates Modell nicht zur Verfügung stehen.
Neben zahlreichen anderen Arbeitsgruppen untersuchten auch wir eine neue transgene (HgfCdk4) Mauslinie, welche spontan Hautmelanome entwickelt und deren Augen bisher noch
nicht auf die Entstehung von spontan auftretenden uvealen Melanomen überprüft wurde.
Abgesehen vom Zufallsbefund stark pigmentierter Zellen in der Cornea der Hgf-Cdk4 Mäuse,
X Zusammenfassung
133
konnten wir keinerlei Hinweise auf melanozytäre Proliferationen in der Uvea finden (Kapitel
VI). Dies unterstreicht nochmals die Unterschiede in der Pathogenese zwischen kutanen und
uvealen Melanomen, die sich sowohl genetisch als auch durch ihr klinisches Verhalten
unterscheiden. Neben Mausmodellen, bei denen humane Aderhautmelanomzellen in das Auge
immunsupprimierter Mäuse injiziert werden, ist die intraokulare Injektion von murinen
Hautmelanomzellen gut etabliert. Trotz gewisser genetischer Unterschiede zwischen
Aderhaut- und Hautmelanomzellen, ermöglicht diese Methode Untersuchungen zur
intraokularen Tumorbiologie in einem immunologisch funktionsfähigen Organismus. Wir
modifizierten daher ein bisher etabliertes Mausmodell und injizierten HCmel12 Zellen
(murine, kutane Melanomzellen) in den Glaskörperraum des Auges von CX3CR1+/GFP und flt1/eGFP BAC Mäusen zweier Altersklassen, um insbesondere den Einfluss tumor-assoziierter
Makrophagen auf das Tumormikromilieu unter Berücksichtigung des Lebensalters zu
untersuchen. Intraokulare Tumore von HCmel12 Zellen zeichneten sich aus durch angiotropes
Wachstum, Angiogenese, Vasculogenic Mimicry/ Mikrozirkulationsmuster und eine
Immunzellinfiltration mit hohem Makrophagenanteil. Diese Eigenschaften sind allesamt für
das humane Aderhautmelanom charakteristisch. Eine rein hämatogene Metastasierung konnte
jedoch nicht nachgewiesen werden (Kapitel II).
Im Vergleich zu jungen Mäusen zeigten alte Mäuse in unseren Studien ein aggressiveres
Wachstumsmuster (histologisch) sowie das vermehrte Auftreten von M2 Makrophagen
(durchflusszytometrische Analyse) und wiesen somit eine größere Ähnlichkeit zu humanen
uvealen Melanomen auf (Kapitel IV). Demzufolge bietet das von uns etablierte Tumormodell
in alten Mäusen die Möglichkeit, das intraokulare Tumormikromilieu in dem semigeschlossenem System des Auges (Blut- aber keine Lymphgefäße) zu untersuchen, welches
iatrogen durch systemische oder intraokulare Injektionen manipuliert werden kann.
X Zusammenfassung
134
Um die allgemeine Rolle der Makrophagen beim intraokularen Melanom untersuchen zu
können, wurden histologische Tumorwachstumseigenschaften von systemisch Makrophagendepletierten Mäusen mit denen unbehandelter Tiere verglichen. Bei unbehandelten Mäusen
war die Häufigkeit prognostisch signifikanter Mikrozirkulationsmuster (Kapitel IV) sowie der
Anteil aktiver Angiogenese (Kapitel V) erhöht. Diese Ergebnisse lassen vermuten, dass
tumor-assoziierte Makrophagen unterschiedliche Vaskularisationsstrategien des Tumors
begünstigen und damit zur Tumorprogression beitragen können. Weiterhin untersuchten wir
die Rolle von funktionell polarisierten Makrophagen, welche einen tumoriziden M1 Phänotyp
oder einen Tumor-begünstigenden M2 Phänotyp annehmen können. Hierfür wurden
HCmel12 Zellen in vitro in einem M1- oder M2-dominierten Mikromilieu inkubiert und
anschließend ins Mausauge injiziert. Tumore aus M2-konditionierten Melanomzellen zeigten
histologisch eine erhöhte Infiltration mit Makrophagen, ein verstärktes Auftreten von
Kollagen 4-positiven Strukturen und eine höhere mittlere Gefäßdichte verglichen zu M1konditionierte Tumoren. Demnach konnten wir zeigen, dass tumor-assoziierte Makrophagen und hier insbesondere M2 Makrophagen – durch tumor-begünstigende Faktoren (wie stromale
Umbaumechanismen im Sinne von Mikrozirkulationsmustern, Entzündungszellinfiltration
und vermehrter Angiogenese) direkt zu einem aggressiveren Tumorphänotyp beitragen
können (Kapitel IV). Zudem konnten wir in flt-1/eGFP BAC Mäusen feststellen, dass der
Großteil der Tumorgefäße CD31+GFP+ war und demnach murinen Strukturen entstammte.
Dies deutet auf “Co-option” von Wirtsgefäßen mit anschließender Angiogenese aus bereits
existierenden Mausgefäßen als Hauptvaskularisationsstrategie des Tumors hin (Kapitel V).
Allerdings zeigten sich auch vereinzelt CD31+GFP- Gefäße, welche folglich einer anderen
Vaskularisierungsstrategie - Vaskulogenese oder aus Tumorzellendothelien - entstammt sein
müssen. Laufende Untersuchungen mit Stammzell- und Melanommarkern sollen deren
X Zusammenfassung
135
endgültigen Ursprung aufklären. Dies könnte dazu beitragen potentiell ein neues Modell zur
Unterscheidung von Angiogenese und Vaskulogenese zu etablieren.
Zusammenfassend, ein “gealtertes” Immunsystem mit einem erhöhten Anteil an M2
Makrophagen bietet eine tumor-begünstigende Grundlage. Diese Erkenntnisse können
potentielle Ansatzpunkte für zukünftige therapeutische Interventionen darstellen, da sie
möglicherweise auf andere Tumoren übertragbar sind.
XI Acknowledgements
136
XI Danksagung
Forschung: ein buntes Konvolut aus springenden Ideen, geduldigem Feilen an scheinbar zum
Scheitern vorbestimmten Methoden, Momenten der unerwarteten Freude wie auch
befürchteter Enttäuschungen; Spontanität und Kreativität als auch langwierige
Literaturrecherche; finanzielle, bauliche sowie erbauliche Rahmenbedingungen. Mit jedem
einzelnen Punkt bin ich mittlerweile sehr vertraut, manchmal meines Widerwollens zum
Trotz. Doch schließlich ist es die Freude am Hinterfragen und Entdecken sowie eine
erfolgreiche Teamarbeit, die mich an der Wissenschaft begeisterte. Ich weiß mich sehr
glücklich zu schätzen, dass ich Teil eines großartigen Labors der Augenklinik Bonn sein
durfte, in dem ich diese Erfahrungen machen und meine Promotion abschließen konnte.
Mein besonderer Dank gilt Frau Dr. M. Herwig, zum einen für die Möglichkeit an diesem
spannenden Thema arbeiten zu dürfen, zum anderen für die wissenschaftliche Anleitung und
insbesondere dafür, dass sie mich im Laufe dieses Projektes unentwegt betreut, beraten,
kritisiert, motiviert und - vor allem - unterstützt hat. Mein ganz spezieller Dank geht an Frau
Prof. K. Löffler, die durch ihre Expertise sowie immerwährende wertvolle Unterstützung das
Projekt maßgeblich mitgestaltet und betreut hat.
Sehr dankbar bin ich ebenfalls Frau Prof. C. Pfarrer, dass sie mir als „Doktormutter“ eine
Promotion an der Tierärztlichen Hochschule Hannover ermöglichte und – trotz der Ferne –
mich stets engagiert betreut und freundlich beraten hat. Damit hat sie meinen tierärztlichen
Weg in die Forschung geebnet und mir die Möglichkeit gegeben meine Arbeit im
tiermedizinischen Rahmen vorzustellen und zu verteidigen.
Ebenso gilt großer Dank Herrn Prof. F. Holz, der mit Enthusiasmus und Nachhaltigkeit
Forschungsarbeiten an der Universitäts-Augenklink Bonn unterstützt und stets motivierend
die Möglichkeiten bot, die eigenen Forschungsfortschritte im Bonner Hause sowie auf
nationalen und internationalen Kongressen präsentieren und diskutieren zu dürfen.
Des Weiteren möchte ich unseren zahlreichen Kooperationspartnern aus anderen Instituten
und Kliniken danken, die unser Projekt ermöglicht und mit Fachkompetenz unterstützt sowie
beraten haben: Prof. C. Kurts (Institute of Experimental Immunology, Universität Bonn),
Prof. T. Tüting (Labor für Experimentelle Dermatologie, Universität Bonn), Prof. D. Wenzel
(Institut für Physiologie I, Universität Bonn), Prof. E. Latz (Institut für angeborene
Immunität, Universität Bonn) und Prof. H. Grossniklaus (Emory Eye Center, Emory
University, Atlanta, GA, USA), Tobias Höller (Institut für Medizinische Biometrie,
Informatik und Epidemiologie, Universität Bonn). Besonderer Dank gilt nicht minder den
Wissenschaftlern, technischen Angestellten und Mitarbeitern der genannten Institute, die mir
stets helfend und beratend zur Seite standen: Dr. Andre Tittel, Dr. Daniel Engel, Andreas
Dolf, Peter Wurst, Tobias Bald, Katia Herz, Anne Alfter, Florian Hoß.
XI Acknowledgements
137
Herzlich möchte ich P. Widmar und C. Strack danken für ihre geduldigen Einweisungen,
hilfreichen Ratschläge sowie tatkräftige Zusammenarbeit und die damit verbundene große
Unterstützung bei der Verwirklichung des Projektes.
Ein ganz besonderer Dank geht allgemein an das gesamte Laborteam der Augenklinik Bonn.
Wenn auch verschiedenen AGs zugeordnet, so waren wir doch immer eine Gemeinschaft, die
sich gegenseitig unterstützt, motiviert und geholfen hat, die zusammen geforscht und gelacht
sowie auf Kongresse gefahren und Nachlesen gehalten hat. Und wenn man manchmal das
Gefühl hatte, dass es um Leben und Toad, Gelingen oder Scheitern eines Versuches oder
Vortrages ging, konnte man sich stets auf fachliche sowie menschliche Unterstützung
verlassen, sodass es einen nicht all zu sehr aus der Kurve geworfen hat. Herzlichen Dank
Karin Löffler, Alfred Wegener, Tim Krohne, Martina Herwig, Parand Widmar, Claudine
Strack, Johanna Meyer, Carolina Brandstetter, Franziska Lörch, Wolf Harmening, Michael
Linden, Niklas Domdei…