ArticlePDF Available

Macrophages Regulate the Angiogenic Switch in a Mouse Model of Breast Cancer

Authors:

Abstract and Figures

The development of a tumor vasculature or access to the host vasculature is a crucial step for the survival and metastasis of malignant tumors. Although therapeutic strategies attempting to inhibit this step during tumor development are being developed, the biological regulation of this process is still largely unknown. Using a transgenic mouse susceptible to mammary cancer, PyMT mice, we have characterized the development of the vasculature in mammary tumors during their progression to malignancy. We show that the onset of the angiogenic switch, identified as the formation of a high-density vessel network, is closely associated with the transition to malignancy. More importantly, both the angiogenic switch and the progression to malignancy are regulated by infiltrated macrophages in the primary mammary tumors. Inhibition of the macrophage infiltration into the tumor delayed the angiogenic switch and malignant transition whereas genetic restoration of the macrophage population specifically in these tumors rescued the vessel phenotype. Furthermore, premature induction of macrophage infiltration into premalignant lesions promoted an early onset of the angiogenic switch independent of tumor progression. Taken together, this study shows that tumor-associated macrophages play a key role in promoting tumor angiogenesis, an essential step in the tumor progression to malignancy.
Content may be subject to copyright.
2006;66:11238-11246. Published OnlineFirst November 17, 2006.Cancer Res
Elaine Y. Lin, Jiu-Feng Li, Leoid Gnatovskiy, et al.
Model of Breast Cancer
Macrophages Regulate the Angiogenic Switch in a Mouse
Updated version
10.1158/0008-5472.CAN-06-1278doi:
Access the most recent version of this article at:
Cited Articles
http://cancerres.aacrjournals.org/content/66/23/11238.full.html#ref-list-1
This article cites by 34 articles, 16 of which you can access for free at:
Citing articles
http://cancerres.aacrjournals.org/content/66/23/11238.full.html#related-urls
This article has been cited by 79 HighWire-hosted articles. Access the articles at:
E-mail alerts
related to this article or journal.Sign up to receive free email-alerts
Subscriptions
Reprints and
.pubs@aacr.orgDepartment at
To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Permissions
.permissions@aacr.orgDepartment at
To request permission to re-use all or part of this article, contact the AACR Publications
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
Published OnlineFirst November 17, 2006; DOI: 10.1158/0008-5472.CAN-06-1278
Macrophages Regulate the Angiogenic Switch in a Mouse
Model of Breast Cancer
Elaine Y. Lin,
1
Jiu-Feng Li,
1
Leoid Gnatovskiy,
1
Yan Deng,
2
Liyin Zhu,
1
Dustin A. Grzesik,
2
Hong Qian,
3
Xiao-nan Xue,
3
and Jeffrey W. Pollard
1
1
Department of Developmental and Molecular Biology, Center of Reproductive Biology and Women’s Health,
2
Analytical Imaging
Facility, and
3
Department of Epidemiology and Population Health, Albert Einstein Cancer Center, Albert Einstein
College of Medicine, Bronx, New York
Abstract
The development of a tumor vasculature or access to the host
vasculature is a crucial step for the survival and metastasis of
malignant tumors. Although therapeutic strategies attempting
to inhibit this step during tumor development are being
developed, the biological regulation of this process is still
largely unknown. Using a transgenic mouse susceptible to
mammary cancer, PyMT mice, we have characterized the
development of the vasculature in mammary tumors during
their progression to malignancy. We show that the onset of the
angiogenic switch, identified as the formation of a high-
density vessel network, is closely associated with the transition
to malignancy. More importantly, both the angiogenic switch
and the progression to malignancy are regulated by infiltrated
macrophages in the primary mammary tumors. Inhibition of
the macrophage infiltration into the tumor delayed the
angiogenic switch and malignant transition whereas genetic
restoration of the macrophage population specifically in these
tumors rescued the vessel phenotype. Furthermore, premature
induction of macrophage infiltration into premalignant
lesions promoted an early onset of the angiogenic switch
independent of tumor progression. Taken together, this study
shows that tumor-associated macrophages play a key role in
promoting tumor angiogenesis, an essential step in the tumor
progression to malignancy. (Cancer Res 2006; 66(23): 11238-46)
Introduction
Tumor progression is characterized by an initial ‘‘avascular
phase’’ when the tumors are small and usually dormant (1) with
diffusion being the major way to support their metabolic needs (2).
In the subsequent ‘‘vascular phase,’’ the development of a unique
tumor vasculature is required for the increased metabolic demand
of tumors that have grown beyond a certain size. The induction of
this vasculature, termed the ‘‘angiogenic switch’’ (1, 3, 4), can occur
at various stages of tumor progression, depending on the tumor
type and the environment (1). However, it is clear that malignant
tumors require its development as it has been shown that the
initiation of revascularization in dormant lesions allows them to
progress (5, 6).
The stroma of solid tumors are replete with many leukocytic
cells of which macrophages represent a major component (7).
Recent clinical and experimental studies have indicated that these
tumor-associated macrophages promote the progression to
malignancy (7, 8). In human breast cancers, macrophages cluster
in ‘‘hotspots’’ in avascular areas in human breast cancer samples
(9), which correlates with a high level of angiogenesis and with
decreased relapse-free and overall survival of the patients (10).
Macrophages play a crucial role in regulating angiogenesis in
wound healing (11). They produce many proangiogenic factors
including vascular endothelial growth factor (VEGF), tumor
necrosis factor a, granulocyte macrophage colony-stimulating
factor, interleukin (IL)-1, IL-6 (11), and other factors including
matrix metalloproteinases (MMP) and nitric oxide (12, 13) that also
have the potential to regulate angiogenesis (7, 11). Parallels have
been drawn between the microenvironment of wound-induced
inflammation and that of tumors, as proposed in the hypothesis
that tumors are ‘wounds that never heal’’ (14). However, whether
tumor-associated macrophages are able to promote angiogenesis is
still not clear.
We have reported in the mouse model of breast cancer caused by
the mammary epithelial cell restricted expression of the Polyoma
middle T oncoprotein (PyMT mice) that the infiltration of
macrophages in primary mammary tumors was positively asso-
ciated with tumor progression to malignancy (8). Depletion of
macrophages in this model severely delayed tumor progression and
dramatically reduced metastasis whereas an increase in macro-
phage infiltration by transgenic means remarkably accelerated
these processes (8). To identify the mechanism(s) that macro-
phages use to promote tumor progression, we have tested the
hypothesis that tumor-associated macrophages stimulate the deve-
lopment of tumor vasculature. Our results indicate that tumor-
associated macrophages were actively involved in promoting the
angiogenic switch during the malignant transition as well as in the
maintenance and/or remodeling of an established vessel network
in malignant tumors.
Materials and Methods
Mice. All procedures involving mice were conducted in accordance with
NIH regulations about the use and care of experimental animals. The study
of mice was approved by the Albert Einstein College of Medicine animal use
committee. The PyMT transgenic mice and mice carrying the CSF-1R-GFP
transgene were kindly provided by Drs. W.J. Muller (McGill University,
Montreal, Quebec, Canada) and David Hume (University of Brisbane,
Brisbane, Australia), respectively. The origin, care, and identification of
CSF-1 null mutant (Csf1
op
/Csf1
op
) mice have previously been described (8).
Because +/Csf1
op
mice have normal serum concentration of CSF-1, normal
tissue population of macrophages, and are in all aspects tested equivalent
to wild-type (+/+) mice (15), these +/Csf1
op
are used as controls. The
preparation of CSF-1-expressing transgenic mice has previously been
described (8). The mice used were in a mixed genetic background of C3H/
B6/FVB. The genotype of the CSF-1R-GFP mice was determined by directly
Requests for reprints: Jeffrey W. Pollard, Albert Einstein College of Medicine, 607
Chanin Building, 1300 Morris Park Avenue, Bronx, NY 10461. Phone: 718-430-2090;
Fax: 718-430-8663; E-mail: pollard@aecom.yu.edu.
I2006 American Association for Cancer Research.
doi:10.1158/0008-5472.CAN-06-1278
Cancer Res 2006; 66: (23). December 1, 2006
11238
www.aacrjournals.org
Research Article
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
Published OnlineFirst November 17, 2006; DOI: 10.1158/0008-5472.CAN-06-1278
visualizing the green fluorescent protein (GFP) expression in blood
monocytes.
In vivo vessel labeling methods. Texas red–conjugated dextran (mol wt
70,000; Molecular Probes, Eugene, OR) was prepared to 6.2 mg/mL in PBS;
21 Ag/g of mouse body weight was i.v. injected. The mice were killed for
tissue preparation 5 minutes after the injection. For the Lycopersicon
esculentum lectin (LEL)/dextran colabeling, 1.3 Ag FITC-conjugated LEL
(Sigma)/g body weight were mixed with dextran solution in the
concentration described above and injected into the tail vein. For analysis,
tumors were isolated and fixed with formalin, paraffin embedded, and
sectioned following standard procedures.
Macrophage labeling and quantitative analysis. Immunohistochem-
istry of macrophage with rat anti-mouse F4/80 antibody was previously
described (8). To measure the macrophage density in tumors, immuno-
histochemically stained sections were photographed to TIFF Images using
an Olympus IX70 microscope and a Sensicam QE cooled CCD camera.
Macrophages in the vicinity of a lesion were measured and normalized by
the circumference of the lesion using ImageJ program. To prepare tissue
sections from CSF-1R-GFP transgenic mice, tissues were fixed in 5%
formalin in 20% sucrose/PBS solution at 4jC for 24 hours followed by
freezing and sectioning. To label macrophages using dextran, 21 Ag/g mouse
body weight of FITC-conjugated dextran (Molecular Probes) were i.v.
injected and tumors were isolated 1 hour after the injection. Using
transgenic mice expressing GFP specifically in macrophages (CSF-1R-GFP
mice), we have observed that within the tumors, only macrophages took up
the injected dextran (data not shown). The standard procedure was used for
immunohistochemistry of anti-VEGF antibody (Santa Cruz Biotechnology,
Santa Cruz, CA) and anti–von Willebrand factor (vWF) antibody (DAKO,
Glostrup, Denmark).
Histologic analysis and vessel density measurements. For quantita-
tive analysis of vessel distribution in the tumor, the entire midline section
from a lesion was photographed into TIFF images and the area in the lesion
marked by dextran was measured using the ImageJ program. For the
comparison of vessel density at different stages of progression classified as
previously described (16), such dextran-marked areas were normalized by
the number of slides for each section and the mean vessel density at
different stages was compared. As previously described (8), a large
percentage of PyMT mice analyzed at 5 to 6, 7 to 8, 9 to 10, or >12 weeks
of age carry mammary lesions at hyperplasia, adenoma/mammary intra-
epithelial neoplasia (MIN), early carcinoma (EC), and late carcinoma (LC)
stages, respectively. To determine the density of the vessels in tumors
(Fig. 5B, iii), the dextran-marked vasculature was skeletonized using ImageJ
program. At least four mice per group were analyzed.
Results
Increased vessel density is associated with the transition to
malignancy. Mammary lesions in PyMT mice progress through
four stages from a benign hyperplasia to an adenoma/MIN and
then to the malignant EC and LC stages (Fig. 1A; ref. 8). A high
frequency of pulmonary metastasis was detected when the primary
mammary tumors progressed to the malignant stages (8, 17). This
stereotypic passage through defined stages provides an excellent
model to study the relationship of the development of the
vasculature according to stage and to test the hypothesis that
tumor-associated macrophages play a key role in angiogenesis.
To characterize the development of the vessels in tumors, PyMT
mice were i.v. injected with Texas red–conjugated, lysine-fixable
dextran 5 minutes before killing. The conjugation of the dextran
with lysine allows cross-linking of the florescent dye–labeled
dextran onto the vessel wall during fixation to permanently mark
the vessels during histologic analysis. This i.v. injection method
gives an accurate and quantitative measurement of vessel density
and architecture in tumors. It also allows to label functional blood
vessels in the tumor (18).
In the passage from hyperplasia to the adenoma/MIN stage
(Fig. 1A), the tumor mass was obviously increased. However, the
density of dextran-marked vessels remained constant (Fig. 1B,
H versus A/M; P = 0.259) and was comparable to the density of the
vessels surrounded the normal ducts (Fig. 1B, duct).
A dramatic change in vessel distribution and density was
observed during the transition from the premalignant to the EC
malignant stage (Fig. 1A; ref. 16). The characteristic feature of the
EC-stage lesion is that in the center of the lesion, the acina-like
structure is replaced by a small solid nodule in which tumor cells
have an increased nuclear pleomorphism and cytologic atypia. The
lesion was classified as the transition stage of malignancy because
such solid nodule(s) with malignant features were only observed in
limited region(s) of the lesion (Fig. 1A, EC, inset), whereas the
majority of the acini were still at the premalignant stages (Fig. 1A,
EC, arrows). The high-density vessel network was only found in the
solid nodular area (Fig. 1A, EC) and not in the surrounding
premalignant acini, in which the vessel density was similar to that
found in lesions at the premalignant stages (Fig. 1A, H and A/M).
Consistent with these histologic observations, a quantitative
analysis indicated a significant increase of vessel density in the
solid nodule compared with the lesions at adenoma/MIN stage
(Fig. 1B, A/M versus EC; P = 0.006). These observations suggest
that such an increase of vessel density is closely associated with
the malignant transition in the tumor.
Further changes in vessel distribution and density were observed
in primary tumors as they progressed to the most advanced
malignant LC stage. A LC-stage lesion consists of multiple large
solid nodules that contain layers of tumor cells with little or no
remaining acina structure and stroma (Fig. 1A, LC). These solid
nodules had both high- and low-density vessel networks but the
overall density of the vessels was similar to that of the EC-stage
lesions (Fig. 1B, EC versus LC).
To confirm that the dextran injection method could precisely
mark blood vessels in tumors, we did a coinjection experiment with
the Texas red–conjugated dextran and FITC-conjugated LEL, which
specifically binds to endothelial cells (19). Using confocal
microscopy, we observed that the majority of dextran-marked
channels in the tumors were colocalized with FITC-LEL staining
(Fig. 2A and B). The representative image showed a FITC-LEL–
labeled vessel (Fig. 2B, b) in the tumor that completely overlapped
with the dextran-marked channel (Fig. 2B, c and d) and the lumen
of the channel was filled with RBC (Fig. 2B, arrows). There were
also some diffuse areas of dextran labeling in cysts or necrotic
areas in large tumors (Fig. 2A, a and c, arrows), suggesting dextran
leakage in these areas. These areas of low density and dull staining
were not included in our analysis of the vessel density.
The validity of the dextran labeling was further confirmed by
immunohistochemistry of tumor sections using antibodies recog-
nizing the endothelial cell–specific marker vWF. As shown in
Fig. 2C, the pattern of vWF staining (Fig. 2C, a) largely overlapped
with the dextran labeling in the tumor section (Fig. 2C, b and c),
indicating that dextran-marked channels are lined by vWF-
expressing endothelial cells. In fact, there are more vWF-positive
areas than dextran-labeled regions in the tumor section (Fig. 2C, c,
arrows), suggesting that injected dextran only labeled vessels with
open lumens or functional blood vessels (18). Higher magnification
confirmed that the fluorescently labeled dextran was in the lumen
formed by vWF-positive cells (Fig. 2C, d, arrow). In addition to the
tissue staining, we have also observed the flow of the RBC through
all of the dextran-filled channels in tumors of living PyMT mice
Macrophages Promote Tumor Angiogenesis
www.aacrjournals.org
11239
Cancer Res 2006; 66: (23). December 1, 2006
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
Published OnlineFirst November 17, 2006; DOI: 10.1158/0008-5472.CAN-06-1278
using multiphoton microscopy (ref. 20; data not shown). This
indicates that all these marked vessels are part of the circulatory
system. We can therefore conclude that the dextran injection
marked the great majority, if not all, of the active vessels in tumors.
Macrophage infiltration precedes angiogenesis and vascular
remodeling in tumors. As a first step in testing the hypothesis
that macrophages play a causal role in angiogenesis, we examined
the distribution of macrophages at different stages of tumor
progression. Few macrophages were found in the hyperplasia and
adenoma/MIN tumors in mice between 4 and 6 weeks of age, as
determined by immunohistochemistry with antibodies to the
macrophage-specific marker F4/80 (Fig. 3A, white arrows). In
fact, the density of macrophages in these premalignant lesions
was similar to that found in the adjacent normal adipose-rich
stroma but was significantly lower than macrophages surround-
ing the developing terminal end buds of normal ducts (data not
Figure 1. Vessel development in
PyMT model correlates with the
malignant transition. A, H&E
staining (top) and dark-field
images showing the dextran
labeling in adjacent sections
corresponding to the boxed area in
the H&E sections displayed above
of normal mammary gland (NM ;
black arrows, mammary ducts)
and premalignant lesions at
hyperplasia (H), adenoma/MIN
(A/M) and malignant EC and
LC stages isolated from +/Csf1
op
PyMT mice at 8, 5, 8, 9, and 16
weeks of age, respectively. EC,
arrows, regions consisted of
premalignant acini. Bar, 500 Am
(H&E); 100 Am (dark field). White
arrows, some of the
dextran-marked vessels.
B, quantitative analysis of the
vessel density in tumors at
different stage of progression.
Columns, density of dextran-
labeled vessels. Duct, normal
mammary ducts; H, hyperplasia;
A/M, adenoma/MIN; EC and LC ,
early and late carcinoma.
Statistical analysis, Welch’s
unpaired t test.
Cancer Research
Cancer Res 2006; 66: (23). December 1, 2006
11240
www.aacrjournals.org
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
Published OnlineFirst November 17, 2006; DOI: 10.1158/0008-5472.CAN-06-1278
shown; ref. 21). However, a remarkable increase in macrophage
infiltration occurred in a large percentage of the adenoma/MIN
tumors at 7 to 8 weeks of age. Macrophages in these more advanced
lesions formed dense clusters in the stroma surrounding the
enlarged tumor acini (Fig. 3A, A/M, right, white arrows). This
infiltration preceded the formation of the dense vessel network.
A large percentage of primary tumors examined at 9 to 10 weeks
of age have progressed to the EC stage. These tumors had a further
increase in macrophage infiltration. Clusters of high-density
infiltrates were found in the vicinity of tumor acini that still were
at the adenoma/MIN stage (Fig. 3B, EC left, white arrows). These
macrophages normally had a rounded morphology. The distribu-
tion of the vessels in these areas was also similar to that found in
adenoma/MIN lesion (Fig. 3A). In contrast, fewer macrophages
were found in the center of the tumor where the dense vessel
network had developed. In these areas, macrophages were attached
to the vessel network and were stretched out along the vessels
(Fig. 3B, EC right, arrows). Thus, both the morphology and
distribution of macrophages were altered depending on the tumor
stage and context.
A further increase of macrophage infiltration was observed as
tumors progressed to the LC stage. A high density of macrophages
was found in a stroma-rich ring surrounding the solid tumor
nodules with the characteristics of LC (Fig. 3B, LC). In this ring,
which also contained a high-density vessel network, a large per-
centage of macrophages seemed to stretch out and attach to the
vessels (Fig. 3B, LC, arrows). Similar to the EC, high-density clusters
of macrophages were also found in the areas that were still at the
adenoma/MIN stage with a corresponding low density of vessels
(data not shown).
To confirm the observations of F4/80 staining in tumors, LC-
stage lesions from CSF-1R-GFP.PyMT mice were used because the
CSF-1R promoter is specifically expressed in macrophages (22).
Consistent with the F4/80 staining, a high density of GFP-positive
stromal cells was found at the vicinity of the lesion (Fig. 3C) and, as
seen from the higher power image in the area containing the vessel
network, GFP-positive cells were physically associated with the
vessel (Fig. 3C).
Macrophage depletion inhibits the angiogenic switch and
the malignant transition. The data above show a remarkable
increase in macrophage infiltration into the nonmalignant
primary tumors followed by the formation of a vessel network
and the transition to malignancy. This suggests that macro-
phages may promote these two processes. We next tested
whether macrophages play a causal role in angiogenesis by de-
pleting macrophages in tumors using mice carrying the homo-
zygous null allele (Csf1
op
) for the mononuclear phagocyte growth
factor CSF-1.
In these studies, we observed a correlation between a low density
of macrophages in the primary tumors of CSF-1 null mutant mice
Figure 2. Dextran-marked channels are lined by endothelial cells. A and B, confocal images of tumor sections from Texas red-dextran and FITC-LEL coinjected
PyMT mice at 18 weeks of age. a, 4,6-diamidino-2-phenylindole (DAPI) counterstaining; b, FITC-LEL; c, Texas red-dextran; d, merged image. A, arrows, cysts
and necrotic areas in the tumor. B, blow-up of a vessel in A. Arrows, RBC in the lumen of the dextran- or LEL-labeled vessel. Bar, 80 Am(A); 20 Am(B ). C, images
of tumor sections from a Texas red-dextran–injected PyMT mouse at 16 weeks of age stained with anti-vWF antibody. a, vWF staining; b, dextran labeling;
c, overlaid image of a, b, and DAPI. c, arrows, vessels that are positively stained by vWF antibody but are dextran negative. d, a higher magnification image of
the staining. Arrow, a dextran-filled vessel lumen formed by vWF-positive cells (green). Bar, 100 Am(a-c); 20 Am(d).
Macrophages Promote Tumor Angiogenesis
www.aacrjournals.org
11241
Cancer Res 2006; 66: (23). December 1, 2006
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
Published OnlineFirst November 17, 2006; DOI: 10.1158/0008-5472.CAN-06-1278
and a marked delay in the angiogenic switch that was associated
with a significant delay in the malignant transition (8). In contrast
to the tumors in heterozygous controls, in which the formation of
dense vessel networks was found in the mammary lesions as early
as 8 weeks of age (Fig. 4A, a), a large percentage of CSF-1 null
mutant mice did not develop such networks in their primary
tumors even when they reached 18 weeks of age (Fig. 4A, b). This
was the case even though most of these lesions were larger than
8mm
3
in size. In addition, a ‘‘sponge’’ like structure was often seen
in these CSF-1 null tumors, indicating that these lesions were
formed mainly by densely packed hyperplastic duct-like structure
instead of the solid nodules that are the characteristic component
of malignant lesions (Fig. 4A, b).
We next compared tumor progression and angiogenesis in the
CSF-1 null mutant mice to their heterozygous littermates at 8 to 9
weeks of age in which f60% of the +/Csf1
op
PyMT mice have
developed malignant stage tumors. As we have previously
observed, a lower percentage of CSF-1 null tumors had progressed
to the malignant stages compared with the +/Csf1
op
controls at the
same age (36% versus 60%). Notably, in both genotypes, only those
tumors that had progressed to malignant stages developed a vessel
network, although still different between genotypes (see below).
However, as previously reported (8, 23), despite the complete
lack of CSF-1, tumors in mice homozygous for the Csf1
op
mutation
are not completely depleted of macrophages. Thus, we hypothe-
sized that the reason that CSF-1 null mutant mice examined
had tumors that had progressed to malignancy at this age was due
to the partial recovery of the macrophage population in these
tumors.
To test this, we compared the macrophage density in tumors that
either progressed or failed to progress to malignancy at this age. A
quantitative analysis was done by measuring the density of
macrophages in the surrounding stroma relative to the circumfer-
ence of the tumor section. As commented on above but now
presented numerically, in +/Csf1
op
tumors, a significantly higher
density of macrophages was found in tumors as they progressed
from the hyperplastic to adenoma to the malignant stages (Fig. 4B,
Pre, 6w versus 9w). This resulted in an almost 50-fold increase in
macrophage density in malignant tumors compared with the
hyperplastic ones. Although the macrophage density in CSF-1 null
Figure 3. Macrophage infiltration in primary mammary tumors correlates with vessel network development and tumor progression. Immunohistochemistry using
anti-F4/80 antibodies of tumor sections stained with DAPI from Texas red-dextran–injected +/Csf
op
PyMT mice. The staining from immunohistochemistry was converted
to green using Photoshop. Tumors were isolated from +/Csf1
op
PyMT mice at 5 weeks (H), 6 weeks (A/M, left), 9 weeks (A/M, right), 9 weeks (EC), and
17 weeks (LC) of age, respectively. A, lesions are at the hyperplasia (H ) and adenoma/MIN (A/M). Yellow size bars in H and A/M sections, 100 Am. B, lesions are
at EC and LC stages. White arrows, some of the F4/80+ macrophages; yellow arrows, dextran-labeled vessels. White size bar in EC and LC sections, 50 Am.
C, LC-stage tumor sections from a Texas red-dextran–injected CSF1R-GFP transgenic mouse. The inset on the left (bar, 100 Am) is shown at higher magnification on
the right (bar, 20 Am). Arrows, GFP-positive macrophages.
Cancer Research
Cancer Res 2006; 66: (23). December 1, 2006
11242
www.aacrjournals.org
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
Published OnlineFirst November 17, 2006; DOI: 10.1158/0008-5472.CAN-06-1278
tumors at the premalignant stage was f25% of that found in the
+/Csf1
op
lesions at the same stage, this only bordered on
significance probably because of the wide variation in wild-type
mice as they were dynamically transitioning into the malignant
state (Fig. 4B). This similarity in density is consistent with the
similar rates of progression and growth rates of these premalignant
tumors in both genotypes. However, a significantly higher level of
macrophage density was also found in the CSF-1 null tumors as they
progressed to the malignant stages compared with the CSF-1 null
tumors at the premalignant stages (Fig. 4B ). This positive
correlation of macrophage density and tumor progression to
malignancy in both genotypes argues that tumor-associated
macrophages have a direct effect on the malignant transition and
the angiogenic switch because this is always associated with the
malignant transition. Nevertheless, when comparing the macro-
phage density in CSF-1 null tumors to that in +/Csf1
op
tumors at the
malignant stages, there was a significantly lower density of
macrophages in CSF-1 null tumors at the same stage (Fig. 4B).
This result indicates that a relatively low density of macrophage
in these CSF-1 null tumors is sufficient for the malignant transi-
tion and angiogenic switch but that it might be the cause of a
slower rate of progression and inhibition of metastasis as pre-
viously observed (8).
As shown above, even in malignant tumors, the CSF-1 null
mice had lower numbers of macrophages. Thus, we tested
whether such a low density had any effect on the development of
tumor vasculature in tumors that had progressed to late-stage
malignancy in age-matched mice. This selection of equivalently
matched tumors prevented bias that could be caused by
comparing tumors at different stages of progression. We found
that, despite the formation of a vessel network in the CSF-1 null
tumors (Fig. 5A, / ), a much lower density of such networks
was observed in these lesions compared with their +/Csf1
op
counterparts (Fig. 5A, +/ ). This was directly due to the absence
of macrophages in the tumors because restoration of macrophage
numbers in the tumors of Csf1
op
/Csf1
op
mice by the transgenic
expression of CSF-1 specifically in the mammary epithelium (8)
resulted in the vessel density returning to the +/Csf1
op
level (Fig.
5A, Trans).
To confirm these histologic observations, a quantitative analysis
of the tumor was done to compare vessel area and density. Sections
of the entire LC-stage tumors of all three genotypes were imaged
and the area covered with vessels in each section was highlighted
and the total area occupied per section was measured as described
in Materials and Methods (Fig. 5B, ii). Afterwards, the images were
skeletonized to analyze the vessel density (Fig. 5B, iii). This method
allows vessels with either big or small lumens to be represented as
lines with similar thickness (pointed by blue or red arrows,
respectively) such that total vessel length could be measured
(Fig. 5B, arrows).
Based on the analysis of more than 300 images, the mean areas
in LC-stage tumors covered by the vessels was 40% lower in CSF-1
null tumors compared with their +/Csf1
op
counterparts (Fig. 5C, OP
versus WT; P = 0.014). Similarly, the vessel density in CSF-1 null LC-
stage tumors was significantly lower than +/Csf1
op
controls (Fig.
5D, OP versus WT; P = 0.03). Restoration of the infiltration of
macrophages by expression of CSF-1 in the CSF1 null tumors
increased this vessel area to a level similar to the +/Csf1
op
tumors
(Fig. 5C, WT versus TRANS; P = 0.35) and also restored the vessel
density to the +/Csf1
op
level (Fig. 5D, WT versus TRANS; P = 0.29). A
similar difference (f1.8-fold) was found when comparing the areas
covered by vessels and vessel densities between +/Csf1
op
and CSF-1 null tumors, suggesting that the sizes of the vessel
lumens in these two genotypes were similar. Taken together,
these data indicated that the infiltration of macrophages in tumors
regulates the density of vessels in LC-stage tumors with a high
level of macrophage infiltration correlated with a high density of
vessels.
Figure 4. The relationship between macrophage infiltration and malignant
transition. A, delayed angiogenic switch and malignant transition in macrophage
depleted tumors. Tumor sections from a +/Csf1
op
mouse at 8 weeks of age (a )
and from a Csf1
op
/Csf1
op
mouse at 18 weeks of age (b ) injected with Texas
red–conjugated dextran. The tumor sections were stained with DAPI. Bar,
100 Am. B, a quantitative analysis of macrophage density in +/Csf1
op
and CSF-1
null tumors, Csf1
op
/Csf1
op
.6wor 9w, mice were at 5 to 6 or 8 to 9 weeks of age,
respectively. Pre, tumors were at premalignant stages; Mag, tumors were at
malignant stages. The data were prepared as described in Materials and
Methods and analyzed using Welch’s unpaired t test.
Macrophages Promote Tumor Angiogenesis
www.aacrjournals.org
11243
Cancer Res 2006; 66: (23). December 1, 2006
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
Published OnlineFirst November 17, 2006; DOI: 10.1158/0008-5472.CAN-06-1278
Premature macrophage infiltration enhances angiogenesis.
The results presented above suggest that macrophages induce both
the formation of the dense vessel network and the malignant
transition. However, based only on these data, it was difficult to
determine whether the vasculature development during the
malignant transition was directly regulated by macrophages or
whether it was a secondary effect induced by this malignant
transition consequent to other effects of macrophages. To
distinguish between these possibilities, we examined vasculature
development in +/Csf1
op
PyMT mice carrying the MMTV LTR-CSF-
1 transgene. Because the MMTV LTR in these transgenic PyMT
mice is active early in mammary gland development, the
expression of the transgenic CSF-1 is induced in young mice
independent of tumor progression. We have previously reported
that a striking increase in macrophage infiltration was observed in
the tumors of these transgenic mice at a young age (8), and that
this correlated with accelerated tumor progression and a doubling
of the metastatic rate compared with the nontransgenic +/Csf1
op
PyMT mice (8).
In the CSF-1-overexpressing transgenic mice, we found that a
dense vessel network developed prematurely in premalignant
tumors from mice at 5 weeks of age (Fig. 6A, top, white arrows).
This dramatic enhancement of the vessel network appeared several
weeks earlier than in the nontransgenic controls. Importantly, in
the nontransgenic mice, such dense vessel networks were found
exclusively in tumors at the malignant stages (Fig. 6A, +/op-9wk)
whereas in the CSF-1 transgenic mice, the network was already
formed in tumors still at premalignant adenoma/MIN (Fig. 6A,
top right) and even at the earliest premalignant stage, hyperplasia
(Fig. 6A, top left). No such dense vessel network was ever seen in
non-CSF-1 transgenic PyMT mice at the hyperplasia stage (Fig. 6A,
+/op-5wk). In parallel with the elevated angiogenesis, a high level of
macrophage infiltration was observed in the stroma of the CSF-1
transgenic premalignant tumors (Fig. 6A, top, yellow arrows)
compared with tumors from nontransgenic mice at similar ages
(Fig. 6A, +/op-5wk) or at the EC stage (Fig. 6A, +/op-9wk, yellow
arrows). A quantitative analysis of macrophage density in these
tumors confirmed a 6-fold increase of macrophage infiltration in
the tumors of CSF-1 transgenic +/Csf1
op
PyMT mice compared with
the nontransgenic controls (Fig. 6B; P = 0.045). This observation
shows that macrophages have a direct effect on the formation of
the vessel network independent of the transition to malignancy,
and that the formation of such a network enables tumors to
advance to malignancy. A possible mechanism for this is the
observation that the major angiogenic regulator, VEGF, was
depleted in stromal cells of tumors of CSF-1 null mutant mice
Figure 5. Vessel density in advanced carcinoma correlates with the degree of macrophage infiltration. A, vessel distribution in LC-stage tumors from a +/Csf1
op
(+/ ),
a CSF-1 null (/), or a CSF-1 transgenic Csf1
op
/Csf1
op
PyMT mouse (Trans ) at 16 weeks of age. Bar, 100 Am. B, processed images for the vessel density
comparison of a tumor section from a Texas red-dextran–injected mouse counterstained with DAPI (i). The area marked by dextran was highlighted using ImageJ
program and skeletonized to indicate the length of individual vessels (ii and iii). Blue arrows, thick vessels that were represented as thin lines after the image was
skeletonized. Red arrows, thin vessels in both images. C, a comparison of areas in tumors covered by dextran-marked vessels. D, comparison of vessel density.
WT, +/Csf1
op
; OP, CSF-1 null or Csf1
op
/Csf1
op
; TRANS, CSF-1 transgenic Csf1
op
/Csf1
op
mice. Significance following analysis by the Mann-Whitney test.
Cancer Research
Cancer Res 2006; 66: (23). December 1, 2006
11244
www.aacrjournals.org
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
Published OnlineFirst November 17, 2006; DOI: 10.1158/0008-5472.CAN-06-1278
compared with +/Csf1
op
mice, suggesting that this may be a
significant part of the reason for this reduced angiogenesis
(Fig. 6C). Thus, overexpression of macrophage VEGF may have
caused the premature vessel formation.
Discussion
Studies in dormant and malignant tumors have suggested that
malignant tumors require the development of a unique vasculature
(5, 6). In this article, using a mouse model where the mammary
tumors undergo a natural progression through premalignant to
malignant stages, we show that the angiogenic switch is required
for the malignant transition.
At the malignant transition, although the lesions mostly
consist of premalignant acini, small solid nodules form in the
center that are the site of the initial malignant transition (16).
The angiogenic switch, identified by the formation of a dense
vascular network, occurred exclusively in this malignant transition
region. This is consistent with other studies using transgenic mouse
models of cancers in which a transition stage, termed angiogenic
dysplasia, occured before the malignant carcinoma was identified
(24–28). However, the close association between the angiogenic
switch and malignant transition was not reported in these studies.
Furthermore, by manipulating the formation of the vasculature, we
were able to show that this angiogenic switch was required for the
malignant transition. In support of this conclusion was the delay in
the malignant transition following the inhibition of the formation of
such vascular networks in CSF-1 null mutant mice. Even more
decisive was the accelerated progression of the tumor to malig-
nancy following premature induction of the angiogenic switch in
premalignant lesions in the CSF-1 transgenic +/Csf1
op
mice.
Tumors are highly populated with macrophages, and in
clinical studies of breast cancer their density correlates with
areas of angiogenesis and with poor prognosis (9). Despite these
clinical correlations, the role(s) of macrophages in promoting
tumor angiogenesis is largely unexplored. In this study, we show
that macrophages promote the development of the tumor
vasculature. We observed an increase of macrophage infiltration
in the stroma of primary tumors shortly before the development
of a dense vessel network in the area. Depletion of this
macrophage infiltration inhibited the angiogenic switch. Further-
more, increased macrophage infiltration in premalignant lesions
through the transgenic expression of CSF-1, of which the only
target is CSF-1 receptor–bearing macrophages (8, 29), induced
premature formation of the vessel network and accelerated the
malignant transition (8). These data indicate that macrophages
have a direct effect on angiogenesis and regulate the angiogenic
switch.
This study identified a unique feature of tumor vasculature in
the PyMT model that reflects the evolution of the tumor through
benign to malignant stages. We found that a dense vessel network
is initiated in the center of the PyMT lesion only where the
malignant transition occurs. This contrasts with that observed in
transplanted tumors that are uniform in malignant progression
and that need to gain vascular support from the host for the
survival of the graft. Therefore the vasculature development that
occurs at the periphery of the tumor (30) does not reflect the
angiogenic switch in naturally evolving cancers. This is different
from PyMT tumors that develop spontaneously and establish a
vascular system in premalignant stages that is similar to that
observed in the surrounding normal tissues. Therefore, the
angiogenic switch identified in this model more closely reflects
that seen during human cancer progression.
Our studies here indicated that the angiogenic switch was not
simply induced by the expansion of tumor size. We found that
the formation of the dense vessel network was initiated in the
malignant transition region of EC-stage lesion that was often
<1 mm
3
in sizes. This indicates that the event is not induced by
the overall growth of the lesion but is stimulated by a regional
factor(s) associated with the progression of the tumor cells to
malignancy. Furthermore, the angiogenic switch was not detected
in more than 40% of CSF-1 null mutant mice that reached 16
weeks of age whereas all of the wild-type mice at this age had
tumors developed to the most advanced malignant stage with
vessel network formation. Interestingly, although the angiogenic
switch was not detected in these CSF-1 null tumors, most of the
Figure 6. Premature expression of CSF-1 induces vessel network formation
in premalignant lesions. A, +/opT sections from a hyperplasia (left ) and adenoma
(right) from CSF-1 transgenic +/Csf1
op
PyMT mouse at 5 weeks of age. Blood
vessels in the tumor were labeled by i.v. injection of Texas red-dextran
(white arrows ) and macrophages were labeled by FITC-conjugated dextran
(yellow arrows ). Bar, 100 Am. +/op-5wk, representative image of F4/80
immunohistochemistry of a tumor lesion at hyperplasia stage isolated from
a+/Csf1
op
PyMT mouse at 5 weeks of age. Note that the densities of both
vessels (white arrows ) and macrophages (yellow arrows ) are much lower than
in the +/opT sections. Bar, 100 Am. +/op-9wk, representative F4/80
immunohistochemistry image of a lesion at the EC stage isolated from a +/Csf1
op
PyMT mouse at 9 weeks of age. Note that macrophages (yellow arrow) seem
to attach to the vessels network. Bar, 50 Am. B, quantitative analysis of
macrophage infiltration in CSF-1 transgenic (Trans ) and nontransgenic (Control )
+/Csf1
op
PyMT mice at 5 to 6 weeks of age. Columns, mean; bars, SD.
C, immunohistochemistry for VEGF in LC-stage tumor sections from a +/Csf1
op
or a Csf1
op
/Csf1
op
PyMT mouse at 18 weeks of age. VEGF-positive cells
were mainly in stroma as indicated by arrows. Representative of those
obtained from six +/Csf1
op
and five Csf1
op
/Csf1
op
PyMT mice with tumors at
LC stage.
Macrophages Promote Tumor Angiogenesis
www.aacrjournals.org
11245
Cancer Res 2006; 66: (23). December 1, 2006
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
Published OnlineFirst November 17, 2006; DOI: 10.1158/0008-5472.CAN-06-1278
tumors were often >8 mm
3
, further confirming this lack of
association with size.
This study has shown that macrophages play a causal role in
promoting tumor angiogenesis and the induction of macrophage
infiltration in tumors of PyMT model occurred over a narrow time
frame when lesions are still at premalignant stage and small in size.
These observations suggest that the tumor microenvironment at
the premalignant stage produces local factors that attract the
infiltration of macrophages that promote tumor angiogenesis.
Studies have shown that tumor cells produce various chemo-
attractants for leukocytes including CSF-1, CCL2, CCL5, VEGF, and
IL-8 (11), suggesting that such infiltration might be manipulated by
tumor cells. Clearly, overexpression of CSF-1 is sufficient to recruit
abundant macrophages. This is consistent with the clinical
observation that overexpression of CSF-1 correlates with leukocytic
infiltration, which in turn correlates with poor prognosis (31, 32).
This raises the question of what tumor and/or environmental
factor(s) triggers the release of these leukocytic chemoattractants.
Numerous studies suggest that hypoxia might be the factor that
induces the up-regulation of chemoattractants in tumor cells (33).
However, we did not see any obvious colocalization of macrophage
infiltration and hypoxia in tumors when Hypoxyprobe-1 (pimoni-
dazole hydrochloride) was used to map the hypoxia regions in the
PyMT model (data not shown).
The mechanisms macrophages use to promote angiogenesis in
tumor are unclear. Macrophages can produce angiogenesis
regulators (7, 11) and may also induce tissue remodeling by
producing various proteinase activators and inhibitors that may
destroy the integrity of the basement membrane and extracellular
matrix, liberating matrix-bound factors. Indeed, a recent study
using the human papillomavirus-16–induced model of cervical
carcinoma showed that macrophages produce MMP-9 that seems
to be required for release of VEGF locally in the tumor, and that
inhibition of this by bisphosphonate inhibited angiogenesis and
reduced tumor incidence and growth (25). Furthermore, inhibition
of CSF-1 function in human tumors xenografted into immuno-
compromized mice reduced their growth and this was correlated
with poor macrophage recruitment and reduced angiogenesis due
to a depletion of VEGF (34). Indeed, we showed that most of the
VEGF-expressing cells in the PyMT model were in stroma and that
that they were reduced in the CSF-1 null tumors and presumably,
therefore, are macrophages.
In summary, this study uses genetic means to show that
macrophages regulate the angiogenic switch, an essential step for
the progression of mammary tumors to malignancy. Such
information will be very important for the development of novel
and effective therapeutic strategies against tumors.
Acknowledgments
Received 4/7/2006; revised 8/2/2006; accepted 9/1/2006.
Grant support: Albert Einstein Cancer Center Analytical Imaging Facility and
histopathology facilities, National Cancer Institute grants CA 94173 and CA 100324,
and Albert Einstein Cancer Center Core grant P30 CA 13330.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
We thank Dr. J.E. Segall for critical comments on the manuscript; Drs. S. Patan,
A.W. Ashton, R.N. Kitsis, B. Terman, P. Davies, A. Orlofsky, and V. Gouon-Evans for
helpful suggestions; and Jim Lee for technical support.
Cancer Research
Cancer Res 2006; 66: (23). December 1, 2006
11246
www.aacrjournals.org
References
1. Bergers G, Benjamin LE. Tumorigenesis and the
angiogenic switch. Nat Rev Cancer 2003;3:401–10.
2. Folkman J. How is blood vessel growth regulated in
normal and neoplastic tissue? G.H.A. Clowes Memorial
Award lecture. Cancer Res 1986;46:467–73.
3. Coussens LM, Tinkle CL, Hanahan D, Werb Z. MMP-9
supplied by bone marrow-derived cells contributes to
skin carcinogenesis. Cell 2000;103:481–90.
4. Folkman J. Incipient angiogenesis. J Natl Cancer Inst
2000;92:94–5.
5. Folkman J, Hahnfeldt P, Hlatky L. Cancer: looking
outside the genome. Nat Rev Mol Cell Biol 2000;1:76–9.
6. Hanahan D, Folkman J. Patterns and emerging
mechanisms of the angiogenic switch during tumori-
genesis. Cell 1996;86:353–64.
7. Pollard JW. Tumour-educated macrophages promote
tumour progression and metastasis. Nat Rev Cancer
2004;4:71–8.
8. Lin EY, Nguyen AV, Russell RG, Pollard JW. Colony-
stimulating factor 1 promotes progression of mammary
tumors to malignancy. J Exp Med 2001;193:727–40.
9. Leek RD, Lewis CE, Whitehouse R, Greenall M, Clarke
J, Harris AL. Association of macrophage infiltration with
angiogenesis and prognosis in invasive breast carcino-
ma. Cancer Res 1996;56:4625–9.
10. Leek RD, Harris AL. Tumor-associated macrophages
in breast cancer. J Mammary Gland Biol Neoplasia 2002;
7:177–89.
11. Crowther M, Brown NJ, Bishop ET, Lewis CE.
Microenvironmental influence on macrophage regula-
tion of angiogenesis in wounds and malignant tumors.
J Leukoc Biol 2001;70:478–90.
12. Lechner M, Lirk P, Rieder J. Inducible nitric oxide
synthase (iNOS) in tumor biology: the two sides of the
same coin. Semin Cancer Biol 2005;15:277–89.
13. Lijnen HR. Plasmin and matrix metalloproteinases in
vascular remodeling. Thromb Haemost 2001;86:324–33.
14. Dvorak HF. Tumors: wounds that do not heal.
Similarities between tumor stroma generation and
wound healing. N Engl J Med 1986;315:1650–9.
15. Pollard JW, Stanley ER. Pleiotropic roles for CSF-1 in
development defined by the mouse mutation osteope-
trotic (op). Advances in Developmental Biochemistry
1996;4:153–93.
16. Lin EY, Jones JG, Li P, et al. Progression to malignancy
in the polyoma middle T oncoprotein mouse breast
cancer model provides a reliable model for human
diseases. Am J Pathol 2003;163:2113–26.
17. Guy CT, Cardiff RD, Muller WJ. Induction of
mammary tumors by expression of polyomovirus
middle T oncogenes: a transgenic mouse mode of a
metastatic disease. Mol Cell Biol 1992;12:954–61.
18. McDonald DM, Choyke PL. Imaging of angiogenesis:
from microscope to clinic. Nat Med 2003;9:713–25.
19. Ezaki T, Baluk P, Thurston G, La Barbara A, Woo C,
McDonald DM. Time course of endothelial cell prolif-
eration and microvascular remodeling in chronic
inflammation. Am J Pathol 2001;158:2043–55.
20. Condeelis J, Segall JE. Intravital imaging of cell
movement in tumours. Nat Rev Cancer 2003;3:921–30.
21. Gouon-Evans V, Rothenberg ME, Pollard JW.
Postnatal mammary gland development requires
macrophages and eosinophils. Development 2000;127:
2269–82.
22. Sasmono RT, Oceandy D, Pollard JW, et al. A
macrophage colony-stimulating factor receptor-green
fluorescent protein transgene is expressed throughout
the mononuclear phagocyte system of the mouse. Blood
2003;101:1155–63.
23. Cecchini MG, Dominguez MG, Mocci S, et al. Role of
colony stimulating factor-1 in the establishment and
regulation of tissue macrophages during postnatal
development of the mouse. Development 1994;120:
1357–72.
24. Calvo A, Yokoyama Y, Smith LE, et al. Inhibition of
the mammary carcinoma angiogenic switch in C3(1)/
SV40 transgenic mice by a mutated form of human
endostatin. Int J Cancer 2002;101:224–34.
25. Giraudo E, Inoue M, Hanahan D. An amino-
bisphosphonate targets MMP-9-expressing macropha-
ges and angiogenesis to impair cervical carcinogenesis.
J Clin Invest 2004;114:623–33.
26. Parangi S, O’Reilly M, Christofori G, et al. Antiangio-
genic therapy of transgenic mice impairs de novo tumor
growth. Proc Natl Acad Sci U S A 1996;93:2002–7.
27. Bergers G, Javaherian K, Lo KM, Folkman J, Hanahan
D. Effects of angiogenesis inhibitors on multistage
carcinogenesis in mice. Science 1999;284:808–12.
28. Coussens LM, Raymond WW, Bergers G, et al.
Inflammatory mast cells up-regulate angiogenesis dur-
ing squamous epithelial carcinogenesis. Genes Dev 1999;
13:1382–97.
29. Wyckoff J, Wang W, Lin EY, et al. A paracrine loop
between tumor cells and macrophages is required for
tumor cell migration in mammary tumors. Cancer Res
2004;64:7022–9.
30. Gilead A, Meir G, Neeman M. The role of angiogenesis,
vascular maturation, regression and stroma infiltration
in dormancy and growth of implanted MLS ovarian
carcinoma spheroids. Int J Cancer 2004;108:524–31.
31. Scholl SM, Pallud C, Beuvon F, et al. Anti-colony-
stimulating factor-1 antibody staining in primary breast
adenocarcinomas correlates with marked inflammatory
cell infiltrates and prognosis. J Natl Cancer Inst 1994;86:
120–6.
32. Tang RP, Kackinski B, Validire P, et al. Oncogene
amplification correlates with dense lymphocyte infiltra-
tion in human breast cancers: a role for hematopoietic
growth factor release by tumor cells? J Cell Biochem
1990;44:189–98.
33. Murdoch C, Giannoudis A, Lewis CE. Mechanisms
regulating the recruitment of macrophages into hypoxic
areas of tumors and other ischemic tissues. Blood 2004;
104:2224–34.
34. Aharinejad S, Abraham D, Paulus P, et al. Colony-
stimulating factor-1 antisense treatment suppresses
growth of human tumor xenografts in mice. Cancer
Res 2002;62:5317–24.
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
Published OnlineFirst November 17, 2006; DOI: 10.1158/0008-5472.CAN-06-1278
... Myelomonocytic cells, including monocytes, monocyte-derived macrophages and tissue-resident macrophages-collectively referred to as tumor-associated macrophages (TAMs)-have long been implicated as positive regulators of tumor angiogenesis 100,101 . TAMs undergo proangiogenic programming in response to hypoxia and tumor-derived factors 39,102,103 , acting as a source of VEGFA, other proangiogenic factors and ECM-remodeling proteases 29,39,40,42,[104][105][106] . They closely interact with nascent TBVs to promote angiogenesis 79,107-109 and facilitate vascular co-option by invasive cancer cells through their ECM-remodeling capacity 110 . ...
Article
Research into the mechanisms and manifestations of solid tumor vascularization was launched more than 50 years ago with the proposition and experimental demonstrations that angiogenesis is instrumental for tumor growth and was, therefore, a promising therapeutic target. The biological knowledge and therapeutic insights forthcoming have been remarkable, punctuated by new concepts, many of which were not foreseen in the early decades. This article presents a perspective on tumor vascularization and its therapeutic targeting but does not portray a historical timeline. Rather, we highlight eight conceptual milestones, integrating initial discoveries and recent progress and posing open questions for the future.
... Lymphocytes play a vital role in the human immune system and are crucial for the immune response against cancer. When the lymphocyte count decreases, the effectiveness of the immune system in fighting tumors is reduced, leading to accelerated tumor growth (Lin et al. 2006). This analysis helps to explain why a decrease in neutrophils or an increase in lymphocytes can improve the LC rate of lung tumors. ...
Article
Full-text available
Purpose The study aims to investigate whether including the inflammation-related parameters would enhance the accuracy of a nomogram for local control (LC) prediction in lung cancer patients undergoing stereotactic body radiation therapy (SBRT). Methods 158 primary or metastatic lung cancer patients treated with SBRT were retrospectively analyzed. The clinical, dosimetric and inflammation-related parameters were collected for the Cox regression analysis. The ACPB model was constructed by employing the clinical and dosimetric factors. And the ACPBLN model was established by adding the inflammation-related factors to the ACPB model. The two models were compared in terms of ROC, Akaike Information Criterion (AIC), C-index, time-dependent AUC, continuous net reclassification index (NRI), integrated discrimination improvement (IDI), calibration plots and decision curve analysis (DCA). Results Multivariate Cox regression analysis revealed that six prognostic factors were independently associated with LC, including age, clinical stage, planning target volume (PTV) volume, BED of the prescribed dose (BEDPD), the lymphocyte count and neutrocyte count. The ACPBLN model performed better in AIC, bootstrap-corrected C-index, time-dependent AUC, NRI and IDI than the ACPB model. The calibration plots showed good consistency between the probabilities and observed values in the two models. The DCA curves showed that the ACPBLN nomogram had higher overall net benefit than the ACPB model across a majority of threshold probabilities. Conclusion The inflammation-related parameters were associated with LC for lung cancer patients treated with SBRT. The inclusion of the inflammation-related parameters improved the predictive performance of the nomogram for LC prediction.
... Tumor-infiltrated myeloid cells also suppress antitumoral T cells by inducing T cell 'exhaustion' through the upregulation of immune checkpoint molecules within the tumor [180], by enhancing inhibitory Treg responses [181] or by secreting immunomodulatory mediators [180,182]. Furthermore, tumor-infiltrated innate immune cells promote angiogenesis or neovascularization which is essential for tumor sustenance and growth [183,184]. Those innate immune cells provide multiple proangiogenic substances inducing and facilitating endothelial cell proliferation and behaviors. ...
Article
Full-text available
Immune cell migration is required for the development of an effective and robust immune response. This elegant process is regulated by both cellular and environmental factors, with variables such as immune cell state, anatomical location, and disease state that govern differences in migration patterns. In all cases, a major factor is the expression of cell surface receptors and their cognate ligands. Rapid adaptation to environmental conditions partly depends on intrinsic cellular immune factors that affect a cell’s ability to adjust to new environment. In this review, we discuss both myeloid and lymphoid cells and outline key determinants that govern immune cell migration, including molecules required for immune cell adhesion, modes of migration, chemotaxis, and specific chemokine signaling. Furthermore, we summarize tumor-specific elements that contribute to immune cell trafficking to cancer, while also exploring microenvironment factors that can alter these cellular dynamics within the tumor in both a pro and antitumor fashion. Specifically, we highlight the importance of the secretome in these later aspects. This review considers a myriad of factors that impact immune cell trajectory in cancer. We aim to highlight the immunotherapeutic targets that can be harnessed to achieve controlled immune trafficking to and within tumors.
... TAMs have also been known to modify the ECM by creating a profibrotic microenvironment and degrading collagen in the TME (16,17). Thus, TAMs have been shown to promote tumorigenesis by leading to senescence and to promote increased invasion and metastasis (18)(19)(20). ...
Article
Full-text available
Lung cancer is the leading cause of cancer-related deaths in the world, and non-small cell lung cancer (NSCLC) is the most common subset. We previously found that infiltration of tumor inflammatory monocytes (TIMs) into lung squamous carcinoma (LUSC) tumors is associated with increased metastases and poor survival. To further understand how TIMs promote metastases, we compared RNA-Seq profiles of TIMs from several LUSC metastatic models with inflammatory monocytes (IMs) of non-tumor-bearing controls. We identified Spon1 as upregulated in TIMs and found that Spon1 expression in LUSC tumors corresponded with poor survival and enrichment of collagen extracellular matrix signatures. We observed SPON1+ TIMs mediate their effects directly through LRP8 on NSCLC cells, which resulted in TGF-β1 activation and robust production of fibrillar collagens. Using several orthogonal approaches, we demonstrated that SPON1+ TIMs were sufficient to promote NSCLC metastases. Additionally, we found that Spon1 loss in the host, or Lrp8 loss in cancer cells, resulted in a significant decrease of both high-density collagen matrices and metastases. Finally, we confirmed the relevance of the SPON1/LRP8/TGF-β1 axis with collagen production and survival in patients with NSCLC. Taken together, our study describes how SPON1+ TIMs promote collagen remodeling and NSCLC metastases through an LRP8/TGF-β1 signaling axis.
... However, it should be noted that, in a mouse model of breast cancer caused by expression of the polyoma middle T oncoprotein, macrophages produce VEGF, which induces neoangiogenesis, leading to malignancy in mammary neoplasia. 22 Similar processes may occur in ORF. ...
Article
ORF virus (ORFV) causes contagious ecthyma (“ORF”), a disease of sheep and goats characterized by lesions ranging from vesicles and pustules to atypical papilloma-like and angiomatous lesions in the skin and mucosae. The authors investigated the molecular factors leading to the ORF-associated atypical tumor-like changes. Fifteen lambs, 15 kids, and an adult ram clinically affected by natural ORFV infection were enrolled in the study and examined by several methods. ORFV was detected by viral culture or real-time polymerase chain reaction (RT-PCR) in the lesioned tissues and in the blood of the clinically affected sheep and goats. Surprisingly, ORFV was also detected in the blood of healthy goats from an affected herd. Microscopically, they found a pseudo-papillomatous proliferation of the epithelium, while the dermis and lamina propria were expanded by a proliferating neovascular component that highly expressed the viral vascular endothelial growth factor (vVEGF) and its host receptor vascular endothelial growth factor receptor 2 (VEGFR2). Immunohistochemistry, immunofluorescence, and in situ hybridization for mRNA showed that epidermal growth factor receptor (EGFR) was expressed in the fibrovascular component, in the infiltrating CD163+ macrophages, and in the basal stratum of the epidermis. Confocal immunofluorescence microscopy demonstrated that CD163+ macrophages were associated with VEGF and VEGFR2. Finally, they found by quantitative RT-PCR the overexpression of the interleukin-6 and VEGFR2 genes in the lesioned tissues. These findings suggest that ORFV activates an inflammatory reaction characterized by CD163+ macrophages expressing EGFR and VEGFR2, which might play an oncogenic role through synergistic action with vVEGF signaling.
Article
Full-text available
The majority of cancer patients receive radiotherapy during the course of treatment, delivered with curative intent for local tumor control or as part of a multimodality regimen aimed at eliminating distant metastasis. A major focus of research has been DNA damage; however, in the past two decades, emphasis has shifted to the important role the immune system plays in radiotherapy-induced anti-tumor effects. Radiotherapy reprograms the tumor microenvironment, triggering DNA and RNA sensing cascades that activate innate immunity and ultimately enhance adaptive immunity. In opposition, radiotherapy also induces suppression of anti-tumor immunity, including recruitment of regulatory T cells, myeloid-derived suppressor cells, and suppressive macrophages. The balance of pro- and anti-tumor immunity is regulated in part by radiotherapy-induced chemokines and cytokines. Microbiota can also influence radiotherapy outcomes and is under clinical investigation. Blockade of the PD-1/PD-L1 axis and CTLA-4 has been extensively investigated in combination with radiotherapy; we include a review of clinical trials involving inhibition of these immune checkpoints and radiotherapy.
Chapter
The increasing the colorectal cancer (CRC) prevalence and rate of metastasation stimulates the research efforts to identify potential targets for therapy, such as those found in tumor microenvironment (TME). TME consists both of cancer cells and cancer-associated cells of different types with their products, including tumor-associated macrophages (TAMs). The aim of our review is to present the literature available data regarding TAMs and their products, their interactions with different TME constituents in CRC development and progression, along with available therapies directed against TAMs. Our search was done in PubMed, Web of Science, and Google Scholar databases, using the following keywords “tumor associated-macrophages,” “tumor associated-cells,” “colorectal cancer,” and “tumor microenvironment.” According to available data, macrophages can adopt two distinct polarization states, by specific differentiation: proinflammatory or classically activated M1 macrophages and anti-inflammatory or alternatively activated M2 macrophages, with opposite involvement in cancer progression. The metastatic spread of CRC is based on the invasion-metastasis cascade, with multiple steps and the involvement of two significant mechanisms: epithelial-mesenchymal transition (EMT) and angiogenesis. TAMs are strongly involved in CRC development and progression, as M2 macrophages promote angiogenesis and immunosuppression. Considering TAMs complex roles, current therapies are tested in order to prevent monocyte infiltration, to repolarize macrophages phenotype, and to target them in metastatic CRC. As a consequence, multiple therapies have strong associations with TAMs and their re-education from M2 to M1 phenotype. Current studies may identify other TAMs targets for exploitation in future personalized therapies, such as TAMs products, TAMs metabolic substrates, and TME products involved in TAMs survival and function. The exploitation of TAMs attack and manipulation arsenal may be validated in future studies, while their efficiency might be tested in clinical application, contributing to CRC patients’ improved prognosis.
Article
Tumor‑associated macrophages (TAMs) are essential components of the tumor microenvironment (TME) and display phenotypic heterogeneity and plasticity associated with the stimulation of bioactive molecules within the TME. TAMs predominantly exhibit tumor‑promoting phenotypes involved in tumor progression, such as tumor angiogenesis, metastasis, immunosuppression and resistance to therapies. In addition, TAMs have the potential to regulate the cytotoxic elimination and phagocytosis of cancer cells and interact with other immune cells to engage in the innate and adaptive immune systems. In this context, targeting TAMs has been a popular area of research in cancer therapy, and a comprehensive understanding of the complex role of TAMs in tumor progression and exploration of macrophage‑based therapeutic approaches are essential for future therapeutics against cancers. The present review provided a comprehensive and updated overview of the function of TAMs in tumor progression, summarized recent advances in TAM‑targeting therapeutic strategies and discussed the obstacles and perspectives of TAM‑targeting therapies for cancers.
Article
Full-text available
The mechanisms responsible for recruiting monocytes from the bloodstream into solid tumors are now well characterized. However, recent evidence has shown that these cells then differentiate into macrophages and accumulate in large numbers in avascular and necrotic areas where they are exposed to hypoxia. This parallels their tendency to congregate in ischemic areas of other diseased tissues such as atherosclerotic plaques and arthritic joints. In tumors, macrophages appear to undergo marked phenotypic changes when exposed to hypoxia and to switch on their expression of a number of mitogenic and proangiogenic cytokines and enzymes. This then promotes tumor growth, angiogenesis, and metastasis. Here, we compare the various mechanisms responsible for monocyte recruitment into tumors with those regulating the accumulation of macrophages in hypoxic/necrotic areas. Because the latter are best characterized in human tumors, we focus mainly on these but also discuss their relevance to macrophage migration in ischemic areas of other diseased tissues. Finally, we discuss the relevance of these mechanisms to the development of novel cancer therapies, both in providing targets to reduce the proangiogenic contribution made by hypoxic macrophages in tumors and in developing the use of macrophages to deliver therapeutic gene constructs to hypoxic areas of diseased tissues.
Article
Full-text available
In human breast carcinomas, overexpression of the macrophage colony–stimulating factor (CSF-1) and its receptor (CSF-1R) correlates with poor prognosis. To establish if there is a causal relationship between CSF-1 and breast cancer progression, we crossed a transgenic mouse susceptible to mammary cancer with mice containing a recessive null mutation in the CSF-1 gene (Csf1op) and followed tumor progression in wild-type and null mutant mice. The absence of CSF-1 affects neither the incidence nor the growth of the primary tumors but delayed their development to invasive, metastatic carcinomas. Transgenic expression of CSF-1 in the mammary epithelium of both Csf1op/Csf1op and wild-type tumor-prone mice led to an acceleration to the late stages of carcinoma and to a significant increase in pulmonary metastasis. This was associated with an enhanced infiltration of macrophages into the primary tumor. These studies demonstrate that the growth of mammary tumors and the development to malignancy are separate processes and that CSF-1 selectively promotes the latter process. CSF-1 may promote metastatic potential by regulating the infiltration and function of tumor-associated macrophages as, at the tumor site, CSF-1R expression was restricted to macrophages. Our data suggest that agents directed at CSF-1/CSF-1R activity could have important therapeutic effects.
Article
Full-text available
The effect of mammary gland-specific expression of the polyomavirus middle T antigen was examined by establishing lines of transgenic mice that carry the middle T oncogene under the transcriptional control of the mouse mammary tumor virus promoter/enhancer. By contrast to most transgenic strains carrying activated oncogenes, expression of polyomavirus middle T antigen resulted in the widespread transformation of the mammary epithelium and the rapid production of multifocal mammary adenocarcinomas. Interestingly, the majority of the tumor-bearing transgenic mice developed secondary metastatic tumors in the lung. Taken together, these results suggest that middle T antigen acts as a potent oncogene in the mammary epithelium and that cells that express it possess an enhanced metastatic potential.
Article
Angiogenesis is the development of blood vessels from an existing vasculature. This process is fundamental to both physiological wound healing and the growth of malignant tumors, as it restores or creates a blood supply to growing tissue. In both cases, the release of angiogenic molecules by macrophages recruited to the wound or tumor site is central to the formation of these neovessels. Reduced vascular perfusion in tissues generates tissue ischemia and a marked reduction in local levels of oxygen (hypoxia) and glucose. Cells adapt by switching to anaerobic metabolic pathways, with a concomitant increase in lactate production and reduction in extracellular pH. In tumors, these microenvironmental “stress” factors stimulate tumor cells to secrete a wide array of proangiogenic cytokines and enzymes, promoting the re-establishment of a local vascular supply. Here we review the evidence that these stress factors, in particular hypoxia and high lactate levels, stimulate macrophages to perform similar proangiogenic functions in both tumors and wounds. The resolution of wounds results in restoration of tissue integrity and perfusion, and macrophage presence is reduced to preinjury levels. However, in tumors a high number of macrophages persists and might contribute to the ongoing growth, neovascularization, and metastasis of malignant cells.
Article
A mouse model involving the human papillomavirus type-16 oncogenes develops cervical cancers by lesional stages analogous to those in humans. In this study the angiogenic-phenotype was characterized, revealing intense angiogenesis in high-grade cervical intraepithelial neoplasias (CIN-3) and carcinomas. MMP-9, a proangiogenic protease implicated in mobilization of VEGF, appeared in the stroma concomitant with the angiogenic switch, expressed by infiltrating macrophages, similar to what has been observed in humans. Preclinical trials sought to target MMP-9 and angiogenesis with a prototypical MMP inhibitor and with a bisphosphonate, zoledronic acid (ZA), revealing both to be antiangiogenic, producing effects comparable to a Mmp9 gene KO in impairing angiogenic switching, progression of premalignant lesions, and tumor growth. ZA therapy increased neoplastic epithelial and endothelial cell apoptosis without affecting hyperproliferation, indicating that ZA was not antimitotic. The analyses implicated cellular and molecular targets of ZA's actions: ZA suppressed MMP-9 expression by infiltrating macrophages and inhibited metalloprotease activity, reducing association of VEGF with its receptor on angiogenic endothelial cells. Given its track record in clinical use with limited toxicity, ZA holds promise as an "unconventional" MMP-9 inhibitor for antiangiogenic therapy of cervical cancer and potentially for additional cancers and other diseases where MMP-9 expression by infiltrating macrophages is evident.
Chapter
This chapter discusses the pleiotropic roles for colony-stimulating factor-1 (CSF-1) in development defined by the mouse mutation osteopetrotic (op). The mouse mutation op is an inactivating mutation in the CSF-1 gene. Thus, op/op mice have been invaluable in establishing pleiotropic roles for CSF-1 in vivo. Studies with the op/op mouse have established the central role for CSF-1 in osteoclastogenesis and confirmed its central roles in the regulation of mononuclear phagocyte proliferation, survival, and differentiation. Colony stimulating factors (CSFs) induce the proliferation and differentiation of hematopoietic progenitor cells to granulocytes and/or macrophages. Apart from the CSF that specifically stimulates the production of granulocytes, there are three which cause differentiation to macrophages. All of the effects of CSF-1 are mediated by a high affinity cell surface receptor tyrosine kinase that is encoded by the c-fms proto-oncogene product.
Article
Animal models are powerful tools to analyze the mechanism of the induction of human breast cancer. Here we report a detailed analysis of mammary tumor progression in one mouse model of breast cancer caused by expression of the polyoma middle T oncoprotein (PyMT) in the mammary epithelium, and its comparison to human breast tumors. In PyMT mice, four distinctly identifiable stages of tumor progression from premalignant to malignant stages occur in a single primary tumor focus and this malignant transition is followed by a high frequency of distant metastasis. These stages are comparable to human breast diseases classified as benign or in situ proliferative lesions to invasive carcinomas. In addition to the morphological similarities with human breast cancer, the expression of biomarkers in PyMT-induced tumors is also consistent with those associated with poor outcome in humans. These include a loss of estrogen and progesterone receptors as well as integrin-beta1 expression and the persistent expression of ErbB2/Neu and cyclinD1 in PyMT-induced tumors as they progress to the malignant stage. An increased leukocytic infiltration was also closely associated with the malignant transition. This study demonstrates that the PyMT mouse model is an excellent one to understand the biology of tumor progression in humans.
Article
One hundred six primary breast cancer samples were analysed for c-erbB2, int-2, and c-myc gene amplification. Surgically confirmed nodal involvement was observed in 42%. Level of gene amplification was studied by Southern and/or slot blottechniques. Amplified c-erbB2 gene sequences were present in 21.5% of all samples. Int-2 was amplified in 13.1% and c-myc was amplified in 10.3%. In a non-parametric test (Kruskal-Wallis) a strong negative association was found between high levels of c-erbB2 amplification and absence of estrogen receptor (ER) (P = .0009) or progesterone receptor (PR) (P = .011) expression. No correlations were found between all or high levels of amplification of each oncogene separately or combined with T, N, grade, multifocality of tumor, or associated carcinoma in situ. There was a trend approaching statistical significance for patients with c-erbB2 amplifications to have positive lymph nodes at surgery (P = 0.09). A somewhat surprising finding however was a very strong association between oncogene amplification and dense lymphocyte infiltration of the tumor (P = .05). This correlation is even stronger when only high levels of amplification are considered, either for each oncogene separately (P = .0048) or in combination (P = .007). We propose that malignant cell cytokine production may help explain this observation.