ArticlePDF Available

Neuropilin-1 Identifies a Subset of Bone Marrow Gr1-Monocytes That Can Induce Tumor Vessel Normalization and Inhibit Tumor Growth

Authors:

Abstract and Figures

Improving tumor perfusion, thus tempering tumor-associated hypoxia, is known to impair cancer progression. Previous work from our laboratory has shown that VEGF-A165 and semaphorin 3A (Sema3A) promote vessel maturation through the recruitment of a population of circulating monocytes expressing the neuropilin-1 (Nrp1) receptor (Nrp1-expressing monocytes; NEM). Here, we define the characteristics of bone marrow NEMs and assess whether these cells might represent an exploitable tool to induce tumor vessel maturation. Gene expression signature and surface marker analysis have indicated that NEMs represent a specific subset of CD11b+ Nrp1+ Gr1- resident monocytes, distinctively recruited by Sema3A. NEMs were found to produce several factors involved in vessel maturation, including PDGFb, TGF-β, thrombospondin-1, and CXCL10; consistently, they were chemoattractive for vascular smooth muscle cells in vitro. When directly injected into growing tumors, NEMs, isolated either from the bone marrow or from Sema3A-expressing muscles, exerted antitumor activity despite having no direct effects on the proliferation of tumor cells. NEM inoculation specifically promoted mural cell coverage of tumor vessels and decreased vascular leakiness. Tumors treated with NEMs were smaller, better perfused and less hypoxic, and had a reduced level of activation of HIF-1α. We conclude that NEMs represent a novel, unique population of myeloid cells that, once inoculated into a tumor, induce tumor vessel normalization and inhibit tumor growth. Cancer Res; 72(24); 1-11. ©2012 AACR.
Content may be subject to copyright.
Published OnlineFirst December 7, 2012.Cancer Res
Alessandro Carrer, Silvia Moimas, Serena Zacchigna, et al.
and Inhibit Tumor Growth
Monocytes That Can Induce Tumor Vessel Normalization
Neuropilin-1 Identifies a Subset of Bone Marrow Gr1
Updated Version 10.1158/0008-5472.CAN-12-0762doi:
Access the most recent version of this article at:
E-mail alerts related to this article or journal.Sign up to receive free email-alerts
Subscriptions
Reprints and .pubs@aacr.orgPublications Department at
To order reprints of this article or to subscribe to the journal, contact the AACR
Permissions .permissions@aacr.orgDepartment at
To request permission to re-use all or part of this article, contact the AACR Publications
American Association for Cancer Research Copyright © 2012 on December 11, 2012cancerres.aacrjournals.orgDownloaded from
Published OnlineFirst on December 7, 2012; DOI:10.1158/0008-5472.CAN-12-0762
Microenvironment and Immunology
Neuropilin-1 Identies a Subset of Bone Marrow Gr1
Monocytes That Can Induce Tumor Vessel Normalization
and Inhibit Tumor Growth
Alessandro Carrer
1
, Silvia Moimas
1,2
, Serena Zacchigna
1
, Lucia Pattarini
1
, Lorena Zentilin
1
, Giulia Ruozi
1
,
Miguel Mano
1
, Milena Sinigaglia
1
, Federica Maione
3
, Guido Serini
3
, Enrico Giraudo
3
,
Federico Bussolino
3
, and Mauro Giacca
1,2
Abstract
Improving tumor perfusion, thus tempering tumor-associated hypoxia, is known to impair cancer
progression. Previous work from our laboratory has shown that VEGF-A165 and semaphorin 3A (Sema3A)
promote vessel maturation through the recruitment of a population of circulating monocytes expressing the
neuropilin-1 (Nrp1) receptor (Nrp1-expressing monocytes; NEM). Here, we dene the characteristics of bone
marrow NEMs and assess whether these cells might represent an exploitable tool to induce tumor vessel
maturation. Gene expression signature and surface marker analysis have indicated that NEMs represent a
specic subset of CD11bþNrp1þGr1resident monocytes, distinctively recruited by Sema3A. NEMs were
found to produce several factors involved in vessel maturation, including PDGFb, TGF-b, thrombospondin-1,
and CXCL10; consistently, they were chemoattractive for vascular smooth muscle cells in vitro.Whendirectly
injected into growing tumors, NEMs, isolated either from the bone marrow or from Sema3A-expressing
muscles, exerted antitumor activity despite having no direct effects on the proliferation of tumor cells. NEM
inoculation specically promoted mural cell coverage of tumor vessels and decreased vascular leakiness.
TumorstreatedwithNEMsweresmaller,betterperfusedandlesshypoxic,andhadareducedlevelof
activation of HIF-1a. We conclude that NEMs represent a novel, unique population of myeloid cells that, once
inoculated into a tumor, induce tumor vessel normalization and inhibit tumor growth. Cancer Res; 72(24); 1
11. 2012 AACR.
Introduction
Over the last several years, multiple evidences have indicat-
ed that different bone marrowderived myeloid cells partici-
pate in various aspects of cancer development and progression
(13). Among these are tumor-associated macrophages (TAM),
which exert proangiogenic and protumorigenic activity (4).
Other leukocyte populations recruited at the site of tumor
growth are Tie-2 expressing monocytes (TEM; ref. 5) and
myeloid-derived suppressor cells, a variegated group of
CD11bþGr1þcells able to foster tumor progression and
angiogenesis (6). Finally, tumor-derived TGF-bcan drive the
polarization of neutrophils to acquire a protumoral phenotype
(7). Of note, virtually all of the bone marrowderived cell types
that inltrate human tumors appear to possess proangiogenic
activity (1) and their ablation is detrimental to tumor growth
(8), a concept that is also currently being evaluated in clinical
trials (9).
In growing tumors, leukocytes contribute to the excessive
production of proangiogenic factors, which leads to the for-
mation of an abnormal vasculature, typically characterized by
elevated vascular density, abnormal vessel morphology, and
excessive leakiness (1012). These features seem to inuence
tumor progression positively, by inducing a constant state of
hypoxia and acidosis, which, in turn, favors the production of
tumorigenic trophic factors, the natural selection of malignant
cells and the further recruitment of protumorigenic myeloid
cells (13). "Normalizing" the tumor vasculature might thus
represent an effective strategy in cancer therapy, by exerting a
negative effect on malignant progression, blunting tumor
hypoxia, reducing vascular permeability, and enhancing drug
penetration (14, 15).
Here, we describe a novel myeloid cell population that,
when injected into growing tumors, exerts antitumoral
activity by inducing tumor vessel maturation. In recent
years, we and others have observed that the overexpression
Authors' Afliations:
1
Molecular Medicine Laboratory, International Cen-
tre for Genetic Engineering and Biotechnology (ICGEB);
2
Department of
Medical Sciences, Faculty of Medicine, University of Trieste, Trieste; and
3
Department of Oncological Sciences and Division of Vascular Biology,
Institute for Cancer Research and Treatment (IRCC), University of Torino
School of Medicine, Candiolo, Italy
Note: Supplementary data for this article are available at Cancer Research
Online (http://cancerres.aacrjournals.org/)
A. Carrer, S. Moimas, and S. Zacchigna contributed equally to this work.
Corresponding Author: Mauro Giacca, ICGEB, Padriciano 99, 34149
Trieste, Italy. Phone: 39-040-375-7324; Fax: 39-040375-7380; E-mail:
giacca@icgeb.org
doi: 10.1158/0008-5472.CAN-12-0762
2012 American Association for Cancer Research.
Cancer
Research
www.aacrjournals.org OF1
American Association for Cancer Research Copyright © 2012 on December 11, 2012cancerres.aacrjournals.orgDownloaded from
Published OnlineFirst on December 7, 2012; DOI:10.1158/0008-5472.CAN-12-0762
of the 165 amino acid isoform of VEGF-A (VEGF-A
165
)in
adult tissues determines the recruitment of cells positive for
the myeloid antigen CD11b (1618) and that these express
the neuropilin-1 (Nrp1) receptor (17). These cells turned out
to be essential in mediating muralcellcoverageofthe
vasculature formed upon VEGF stimulation. Of note, sema-
phorin-3A (Sema3A), which also binds Nrp1 similar to
VEGF-A
165
, also stimulated massive inltration of these
CD11bþcells in the expressing tissues (17). These, together
with other results, clearly pointed out that Nrp1 was an
essential receptor to mediate the recruitment of these cells.
The inltrating cells, which originate from the bone marrow
and were negative for lineage-specic leukocyte antigens,
among which CD3, CD4, CD8, and CD49b, are herein referred
to as neuropilin-1 expressing monocytes (NEM).
Here, we characterize NEMs as a population of CD11bþ
Nrp1þGr1cells, which constitutes approximately 1.0% of the
total number of bone marrow cells. We report that the intra-
tumoral injection of NEMs, puried from either the bone
marrow or Sema3A-expressing muscles, is capable of markedly
reducing tumor growth by promoting the formation of a more
mature vascular network, eventually resulting in enhanced
tumor perfusion.
Materials and Methods
Animal studies
C57BL/6 or Balb/c mice were injected subcutaneously
(1 10
6
cells/mouse) with B16.F10 melanoma [CRL-6475;
American Type Culture Collection (ATCC)] or 4T1 breast
carcinoma cells (CRL-2539; ATCC), respectively. Tumor vol-
ume was evaluated daily using calipers and expressed in mm
3
using the formula: V¼p/6 (d
max2
d
min
/2; ref. 19).
Flow cytometry and cell sorting
Bone marrow pools from 4 animals were used for ow
cytometry using the following antibodies: anti-CD45-PE,
anti-CD31-PE, anti-Flk1-PE, anti-Sca-1-PE, anti-c-kit-PE,
anti-CD11c-PE, anti-CD14-PE, anti-isotype controls (all from
Pharmingen Becton Dickinson), anti-CCR2-PE, anti-Flt-1-PE
(from R&D), anti-CXCR4-PE, anti-CD49b, anti-F4/80-PE, anti-
MHCII-PE (from eBioscience), anti-CD11b AlexaFluor647,
anti-Tie-2-PE, anti-B220-PE (all from Biolegend), anti-Ly6C-
PE, anti-CD25-PE, anti-CD16/32-PE, anti-Gr1-perCP (from
Miltenyi Biotec), and anti-Nrp1 (C-19; Santa Cruz Biotechnol-
ogy). Sorting was carried out on a FACSAria II Instrument
(Becton Dickinson Biosciences).
In vivo cell tracking
Sorted NEMs or Gr1þcells were labeled with the uorescent
dye DiD (Invitrogen) following the manufacturer's instruc-
tions. Cells were subsequently injected into B16.F10 tumors
and animals were sacriced 6 days after injection.
Migration assays
Migration assays were conducted using 5-mm pore size
Transwell permeable supports (Costar, Corning Incorporated).
Cells (2.5 10
5
) were seeded in serum-free medium in the
upper chamber, whereas recombinant Sema3A (500 ng/mL;
R&D) or FBS (10%) was placed in the lower chamber. After
overnight incubation, migrated cells on the lower chamber
were labeled with Hoechst (2 mg/mL, Invitrogen). Each assay
was carried out in triplicate.
Tissue analysis
Tissues were harvested as described (17). The following
primary antibodies were used diluted 1:200 in blocking buffer:
anti-CD11b, anti-Gr1, anti-CD31 (all from Pharmingen Becton
Dickinson), anti-Nrp1 (R&D Systems), antiproliferating cell
nuclear antigen (PCNA; F-2; Santa Cruz Biotechnology), Cy3-
coniugated anti-a-SMA (1A4; Sigma), and anti-NG2 (Chemi-
con). AlexaFluor-conjugated antibodies (1:1,000; Molecular
Probes) were used as secondary antibodies.
For hypoxia detection, mice were injected intraperitoneally
with 60 mg/kg of Hypoxyprobe-1 (HPI Inc.). After 1 hour,
animals were sacriced and tumor masses were harvested
and processed. Tissue slices were xed with PFA 4% for 30
minutes at room temperature; unmasking with citrate buffer
was carried out for 20 minutes at 98C and slices were then
incubated with the anti-Hypoxyprobe-1 uorescein isothiocy-
anateconjugated antibody (HPI Inc.) for 40 minutes in the
dark.
Fluorescence signals were quantied counting the number
of pixels that exceeded a xed background threshold.
In vitro proliferation assay
B16.F10 cells (1 10
5
) were labeled with calcein (500 ng/mL
for 30 minutes at 37C; Invitrogen), seeded onto a 24-well
plate, and counted after 6 hours; then, freshly puried NEMs
or Gr1þcells were added. After an additional 40 hours, cells
were incubated with Hoechst dye (2 mg/mL; Invitrogen)
for 15 minutes and counted. The proliferation index
was calculated as a ratio between cell count at t¼40 and
t¼0hours.
Vessel permeability assay
Vessel permeability was analyzed by an adaptation of the
Miles' assay. Mice were injected intravenously (i.v.) with 250 mL
0.5% Evans blue (Sigma) and sacriced after 30 minutes. The
tumor mass and liver were removed and weighted. The dye was
extracted by incubation in 2% formamide at 55C and spec-
trophotometrically quantied at 610 nm. For each animal,
Evans blue content was expressed as a ratio between tumor
and liver content.
Doxorubicin quantication
Doxorubicin (Sigma) was injected i.v. (10 mg/kg) in tumor-
bearing C57BL/6 mice 4 hours before sacrice. Tumors and the
liver right lobe were harvested and weighted. Tissue-bound
doxorubicin was assessed as described previously (20), nor-
malized according to tissue weight, and expressed as a ratio
between tumor and liver content.
Power Doppler imaging
Power Doppler images from subcutaneously growing tu-
mors (n¼5 per group) were obtained with a Vevo 770 system,
using a 30 MHz probe. Power Doppler signals were assessed at
Carrer et al.
Cancer Res; 72(24) December 15, 2012 Cancer Research
OF2
American Association for Cancer Research Copyright © 2012 on December 11, 2012cancerres.aacrjournals.orgDownloaded from
Published OnlineFirst on December 7, 2012; DOI:10.1158/0008-5472.CAN-12-0762
3 points along the longitudinal axis, located 0.2-cm away from
each other.
Recovery of NEMs from Sema3A-expressing muscles
Production and use of the recombinant AAV2-Sema3A
(rAAV2-Sema3A) vector was as described previously (17).
After 15 days, mice injected in the tibialis anterior of both
legs [5 10
10
viral particles (vp) per leg] were sacriced, and
muscles were removed and mechanically minced. Tissue was
digested with collagenase (0.225%; Worthington) at 37C for
60 minutes and ltered. The muscle-derived NEMs (mNEM)
were puried using anti-CD11bconjugated magnetic beads
(Miltenyi Biotec).
Proliferation and migration of vascular smooth muscle
cells
Vascular SMCs (vSMC; 1 10
5
) from coronary artery
(Clonetics, Cambrex) were seeded in a 24-well plate. After
6 hours, cells were incubated with EdU (Invitrogen) for 4
hours. Then, cells were washed, the medium was replaced,
and freshly puried NEMs or Gr1þcells (2 10
4
)were
added. Twenty hours after EdU addition, cells were washed
twice in PBS, xed in PFA 4%, and stained with a-SMA
antibody (Sigma) for 2 hours at room temperature (1:400).
EdU detection was carried out using Click-iT EdU Cell
Proliferation Assay (Invitrogen).
Migration assays were conducted using 8-mm pore size
Transwell permeable supports (Costar, Corning Inc.). vSMCs
were stained with calcein (Invitrogen). Then, 1 10
5
cells were
seeded in the upper chamber, whereas puried NEMs or Gr1þ
cells or FBS (20%) were added to the lower chamber. After
overnight incubation, migrated cells were labeled with Hoechst
(2 mg/mL) and double-positive cells were counted. Assays were
carried out in triplicate.
Real-time PCR amplication
Total RNA was extracted using TRIzol reagent (Invitrogen)
and reverse transcribed using hexameric random primers
(Invitrogen). PCR conditions are reported in Supplementary
Table S1.
Statistical analysis
Data were analyzed using a 2-tailed Student t-test or a
nonparametric MannWhitney Utest, with P<0.05 considered
signicant. Growth curves were analyzed by 2-way ANOVA for
repeated measures and post-hoc analysis. Cluster analysis was
conducted using the Genesis software (21). Data were inter-
nally normalized according to the mean centering method.
Results
Characterization of NEMs from the bone marrow
A freshly isolated bone marrow pool (n¼4) was stained
for CD11b, Gr1, and Nrp1 and analyzed by ow cytometry
and cell sorting. The monocytic fraction was discriminated
according to morphological gating and CD11b positivity
(Fig. 1A, a and b). According to the expression of the Gr1
antigen, mouse monocytes are commonly divided into
Gr1(resident) or Gr1þ(inammatory) cells (2224); this
antigen was thus used to sort apart the 2 cell populations
(Fig. 1A, c). Next, both Gr1þand Gr1cells were further
sorted according to expression of Nrp1 (Fig. 1A, d and e). On
the basis of the combined expression of Nrp1 and Gr1, 4
subsets of bone marrowmonocytic cells were thus eventu-
ally obtained (Fig. 1A, f).
Our previous ndings indicated that the cells attracted in
vivo by Sema3A were positive for Nrp1 and negative for Gr1
(17). Therefore, we denedbonemarrowNEMsasthe
Nrp1þGr1subset of CD11bþcells, which corresponded
to approximately 1% of total bone marrow cells (in green
in Fig. 1A, f).
Next, we wanted to verify the response of the 4 sorted
populations to Sema3A. Cells were seeded onto the upper
chamber in a Transwell plate and migration in response to
rSema3A was evaluated after 12 hours. Sema3A chemoat-
tracted only the Gr1subset of the Nrp1þcells (Fig. 1B).
We then determined the gene expression proles of the 4
sorted CD11bþcell subsets. In particular, we assessed the
levels of mRNAs of growth factors and receptors known to be
involved in blood vessel formation, and followed this with
multivariate analysis to detect similarities. A graphic repre-
sentation of the relative gene expression levels in the 4 cell
subsets is shown in Fig. 1C, along with the results of hierar-
chical clustering linkage (HCL). This analysis indicated that the
2 Nrp1þcell populations were more closely clustered accord-
ing to the gene panel analyzed. At the same time, this gene
expression analysis also indicated that the 4 cell subsets
possessed distinct expression signatures.
Comparative ow cytometric characterization of NEMs
and of the other puried cell populations according to
various cell surface antigens is shown in Fig. 1D. The CCR2
antigen showed a different pattern of expression in the 4 cell
subsets. Most of the Gr1þcells expressed this receptor,
however at relatively low levels; in contrast, expression was
more variable in the Gr1subset (NEMs). The CD11c
adhesion molecule was expressed at low levels, in both the
Gr1þand the Nrp1Gr1subsets, whereas NEMs showed a
bimodal distribution with about 60% of these cells expres-
sing this antigen. Moreover, approximately 30% of NEMs
expressed variable levels of MHC class II, whereas these
molecules were absent from most of the other Nrp1þand
Nrp1cells. Finally, most NEMs were negative for integrin
a2 (CD49b), which instead showed a bimodal distribution in
Gr1Nrp1cells. Supplementary Fig. S1 shows quanti-
cation of 4 independent experiments.
NEMs were further characterized for the expression of a
number of other cell surface antigens (Supplementary Fig. S2).
These cells were positive for the panleukocytic antigen CD45
and for the FcgRIII receptor CD16, but negative for B220
(expressed by B cells and plasmacytoid dendritic cells) and
for Ly6C (which is also seen by the Gr1 antibody used for
sorting). The cells were also negative for the IL-2 receptor a
chain (CD25), which is usually present on activated T and B
cells and is considered a marker of classical monocyte acti-
vation. NEMs were clearly negative for the Tie2 angiopoietin
receptor (5). Finally, both types of Nrp1þcells produced plexin
A1, A3, B2, B3, C1, and D1; plexins A1, A3, and B2 were
NEMs Induce Tumor Vessel Stabilization
www.aacrjournals.org Cancer Res; 72(24) December 15, 2012 OF3
American Association for Cancer Research Copyright © 2012 on December 11, 2012cancerres.aacrjournals.orgDownloaded from
Published OnlineFirst on December 7, 2012; DOI:10.1158/0008-5472.CAN-12-0762
differentially expressed in Gr1þand Gr1cells (Supplemen-
tary Fig. S3).
Taken together, these results dene NEMs as a subset of
Gr1, bone marrowresident monocytes specically charac-
terized by the expression of the Nrp1 receptor.
Bone marrow NEMs exert antitumor activity
We moved on to assess the effect of NEMs on established
tumors. Mice (n¼10 per group) were subcutaneously injected
with B16.F10 tumor cells and, at days 11 (when tumors became
palpable) and 13 after inoculation, the tumors were inoculated
with bone marrowpuried NEMs (CD11bþGr1Nrp1þ
cells), CD11bþGr1þNrp1þcells (cells labeled in green and
cyan in Fig. 1, respectively), or PBS. We decided to use the
CD11bþGr1þNrp1þcells (hereafter named Gr1þ)asa
control as these represent a subset of inammatory monocytes
relatively similar to NEMs, but unresponsive to Sema3A (Figs.
1C and B, respectively).
Cells (2 10
5
cells/tumor/injection; n¼10) or PBS (100 mL)
were delivered directly into the tumor mass. NEM injection
determined a signicant reduction in tumor growth (tumor
volume at 15 days: 469 84 in the NEM group vs. 1169 229 in
the Gr1þgroup vs. 806 127 in the PBS-injected mice,
expressed as percentage of pretreatment tumor size; P<
0.01; Fig. 2A). This effect was not consequent to the direct
inhibition of tumor cell proliferation, as the coculture of
puried NEMs or Gr1þcells together with B16.F10 cells did
not affect tumor cell proliferation (Fig. 2B).
A comparably powerful antitumor activity was observed
in a more aggressive, highly metastatic model of tumor
xenograft, obtained by subcutaneous injection of 4T1 cells
in Balb/c mice (n¼10 per group). Bone marrow NEMs,
Gr1þcells, or PBS were administered twice (2 10
5
cells/
tumor/injection) directly into the growing tumor masses.
Again, NEMs inhibited tumor growth (tumor volume at 18
days: 512 94 in the NEM-injected group vs. 1348 776 in
Figure 1. Isolation and characterization of NEMs from the bone marrow. A, sorting of bone marrow NEMs. a, morphological gating of lymphocytes and
monocytes; b, gating of CD11bþcells (on cells gated in a); c, gating for Gr1 expression (on cells gated in b); d, cells analyzed for Nrp1 expression
(on Gr1population in c); e, cells analyzed for Nrp1 expression (on Gr1þpopulation in c); f, schematic representation of the 4 monocyte subpopulations.
Percentages are referred to total bone marrow cells. B, migration assay with the sorted populations; color code as in A, f. FBS was a positive control
(mean SEM; ,P<0.01 over control). C, HCL on gene expression data from the 4 sorted cell populations. Data were internally normalized using the mean
centering method, according to which the mean value of a column (gene expression values of a given gene among samples) is equal to 0. Green, low
expression; red, high expression. D, ow cytometric analysis of the 4 sorted cell populations using the indicated phycoerythrin-conjugated antibodies.
Percentages of positive cells (horizontal bars) relative to total cells are reported.
Carrer et al.
Cancer Res; 72(24) December 15, 2012 Cancer Research
OF4
American Association for Cancer Research Copyright © 2012 on December 11, 2012cancerres.aacrjournals.orgDownloaded from
Published OnlineFirst on December 7, 2012; DOI:10.1158/0008-5472.CAN-12-0762
the Gr1þ-injected group vs. 1976 1422 in the PBS-injected
mice, expressed as percentage of pretreatment tumor size;
P<0.01; Fig. 2C). Similar to B16.F10 cells, in this case, neither
NEMs nor Gr1þcells affected tumor cell proliferation in
vitro (Fig. 2D).
Ex vivo labeling of NEMs and Gr1þcells with the uorescent
dye DiD before injection indicated that the cells persisted
inside the tumors at least up to 6 days after inoculation (Fig.
2E).
Staining for the PCNA antigen indicated that there were no
signicant differences in the percentage of proliferating B16.
F10 cells in the 3 groups of treated animals (Fig. 2F; quanti-
cation in Fig. 2G); similar results were also obtained for 4T1
tumors (Supplementary Fig. S4).
Figure 2. NEMs exert indirect
antitumor activity. A, growth curve of
B16.F10 melanoma tumors
implanted (day 0) subcutaneously in
C57BL/6 mice and injected at days
11 and 13 with NEMs, Gr1þcells, or
PBS. Data are expressed as
percentages over day 10, mean
SEM. B, proliferation assay of B16.
F10 cells cocultured in vitro with 2
10
4
Gr1þcells, NEMs, or PBS after a
2-day coculture (mean SEM). C,
same as in A for 4T1 tumors. D, same
as in B for 4T1 cells. E, fate of sorted
Gr1þand NEM cells injected into
B16.F10 tumors. Before injection,
cells were labeled with DiD (red);
tumors were harvested 6 days after
cell injection. Top (a), whole tumor
sections; bottom (b and c),
magnication of the indicated areas.
Scale bars in the lower panels, 100
mm. F, immunouorescence of tumor
sections from mice injected with PBS
(a), Gr1þcells (b and b'), or NEMs (c);
green, PCNA; blue, 40, 6-diamidino-
2-phenylindole (DAPI). Asterisk,
vessel lumen; arrow, perivascular,
PCNAþcells. G, percentage of
PCNAþnuclei in the tumor sections
(mean SEM; n¼8 per group).
NEMs Induce Tumor Vessel Stabilization
www.aacrjournals.org Cancer Res; 72(24) December 15, 2012 OF5
American Association for Cancer Research Copyright © 2012 on December 11, 2012cancerres.aacrjournals.orgDownloaded from
Published OnlineFirst on December 7, 2012; DOI:10.1158/0008-5472.CAN-12-0762
Together, these results are consistent with the conclusion
that NEMs markedly inhibit tumor growth when injected
intratumorally, albeit not directly affecting tumor cell
proliferation.
NEMs promote tumor vessel stabilization
The numerous clusters of PCNAþcells detected in corre-
spondence with the tumor vessels (indicated by arrows in Fig.
2F) were possibly indicative of an ongoing angiogenic process.
As these clusters were reduced in the NEM-injected tumors, we
carried out a detailed analysis of the effect of NEMs or Gr1þ
cells on vascular morphology and function. The number of
CD31-positive vessels was not different among the 3 experi-
mental groups (representative images in Fig. 3A and B; quan-
tications in Fig. 3C and D). However, the vasculature in the
NEM-treated tumors showed markedly increased positivity for
a-SMA and NG2 staining (Figs. 3A and B) and, consequently, a
marked increased in a-SMA/CD31 (P<0.01 vs. both PBS and
Gr1þ) and NG2/CD31 (P<0.05 vs. PBS and P<0.01 vs. Gr1þ)
ratios (Fig. 3E and F, respectively). Morphometric analysis of
the vasculature indicated that, in addition to pericyte coverage,
vessels in the NEM-treated tumors displayed additional mar-
kers of maturation, such as increased mural thickness (Fig. 3G)
and reduced lumen perimeter (Fig. 3H) and area (Fig. 3I).
We also wanted to exclude that the effect of NEMs on vessel
maturation might be a consequence of the reduced tumor
growth caused by their inoculation. Mice (n¼5 per group)
bearing B16.F10 tumors were injected at days 11 and 13 with 2
10
5
NEMs or Gr1þcells, or PBS, and sacriced when tumors
reached the xed volume of 1,700 mm
3
. Also in this experiment,
a signicant difference in a-SMA/CD31 ratio was evident (P<
0.01 vs. both PBS and Gr1þ; Supplementary Fig. S5).
Collectively, these results indicate that the antitumoral
effect of NEMs is concomitant with a marked increase in
vessel maturation.
NEMs enhance tumor perfusion and relieve tumor-
associated hypoxia
Recent evidence suggests that the establishment of a chronic
state of hypoxia might fuel tumor growth (25, 26). We, there-
fore, wanted to assess whether the vessel stabilization activity
of NEMs might improve tumor perfusion, thus alleviating
Figure 3. NEMs promote vessel maturation. A, immunouorescence of the vasculature of tumors injected with PBS, Gr1þcells, or NEMs. Whole tumo r
sections were stained for CD31 (green) and a-SMA (red) at 3 different magnications (a, b, c, d, g, j, m, p, and s). Split CD31 and a-SMA signals are shown in the
small panels e, f, h, i, k, l, n, o, q, r, t, and u under the respective merged pictures. Blue, DAPI. Scale bars in panel d and o, 100 mm. B, same as in A,
using antibodies against CD31 (green) and NG2 (red). CI, quantitative evaluation, in tumors injected with PBS, Gr1þcells, or NEMs, of the following
parameters: vessel number (C), area covered by CD31þendothelial cells (D), a-SMA/CD31 ratio (E), NG2/CD31 ratio (F), mural thickness (G), vessel lumen area
(H), vessel perimeter (I). Data are mean SEM. ,P<0.05; ,P<0.01 versus PBS.
Carrer et al.
Cancer Res; 72(24) December 15, 2012 Cancer Research
OF6
American Association for Cancer Research Copyright © 2012 on December 11, 2012cancerres.aacrjournals.orgDownloaded from
Published OnlineFirst on December 7, 2012; DOI:10.1158/0008-5472.CAN-12-0762
tumor-associated hypoxia. First, a set of animals injected with
B16.F10 melanoma tumors received either bone marrow NEMs
or Gr1þcells (n¼5 per group) and were then submitted to a
modied Miles assay to verify the permeability of the tumor
vasculature. The results of this assay indicated that the tumors
injected with NEMs displayed vessels that were signicantly
less leaky than those injected with Gr1þcells (Fig. 4A). Another
set of tumor-bearing animals (n¼5 per group) was injected
with either bone marrow NEMs or control Gr1þcells. After 3
days, a bolus of doxorubicin (10 mg/kg per animal) was
administered to each mouse. After an additional 4 hours, mice
were sacriced and the doxorubicin content within the tumor
was assessed. NEM-injection signicantly improved intratu-
moral drug delivery (Fig. 4B).
Consistent with the above ndings, PD echographic visual-
ization of the tumor vasculature revealed a net increase in the
number of Doppler-positive vessels, reecting an improvement
in functional tumor perfusion (sequential representative
images of NEM- and Gr1þ-injected tumors are shown
in Fig. 4C, quantication in Fig. 4D). Accordingly, by staining
the tumors by HypoxyProbe (n¼5), we found that NEMs
improved tumor oxygenation, resulting in a reduced hypoxic
tumor area (representative images and quantication in Figs.
4E and F, respectively). Finally, the levels of the hypoxia-
sensitive transcription factor HIF-1awere markedly reduced
in whole lysates from these tumors compared with those
injected with Gr1þcells (shown in Fig. 4G for 4 independent
tumors). Concomitant with reduced HIF-1 activation, there
was a signicant decrease in the levels of the mRNAs of 3 major
HIF-1a-regulated genes (VEGF-A,Angiopoietin-2, and Il-6;
ref. 27; Supplementary Fig. S6).
Taken together, these observations would indicate that the
antitumor activity of NEMs is mediated by the induction of
morphological maturation and functional normalization of the
tumor vasculature.
Sema3A-recruited NEMs inhibit tumor growth and
stabilize vessels
Recent work indicates that Sema3A exerts antitumoral
effect by affecting the tumor vasculature (2832). As our
previous work indicated that Sema3A is a powerful attractor
of NEMs in vivo (17), we wanted to assess whether the Sema3A-
recruited NEMs possessed antitumoral activity. An AAV2
vector expressing Sema3A was injected into the tibialis ante-
rior muscle of both legs in mice (n¼15). After 15 days, muscles
were inltrated by CD11bþNrp1þGr1cells (Fig. 5A). These
mNEM were puried using anti-CD11bcoupled magnetic
beads (>90% purify of CD11bþNrp1þafter recovery; Fig.
5B). Mice (n¼6 per group) injected subcutaneously with
B16.F10 cells received either mNEMs (2 10
5
cells/tumor per
injection) or PBS; cell delivery was carried out twice, at days 10
and 14 after tumor implantation. mNEM injection signicantly
Figure 4. NEMs enhance tumor perfusion and relieve tumor-associated hypoxia. A, quantication of vascular permeability in tumors injected with Gr1þcells
or NEMs (mean SEM; ,P<0.01). B, doxorubicin quantication in tumors injected with Gr1þcells or NEMs (mean SEM; ,P<0.05). C, power
Doppler imaging of 2 tumors injected with Gr1þcells or NEMs; 3 sections (frames 1 to 3) per animal are shown. Large vessels appear in orangeyellow.
D, quantication of blood ow by Doppler analysis (mean SEM; ,P<0.01). E and F, analysis of tumor hypoxia by pimonidazole (green) in sections of
tumors injected with NEMs or Gr1 þcells. E, representative pictures; F, quantication (mean SEM; ,P<0.01). G, Western blotting of whole B16.F10 tumor
lysates (WTL) from 4 different mice treated with Gr1þcells or NEMs using anti-HIF-1aand antitubulin antibodies.
NEMs Induce Tumor Vessel Stabilization
www.aacrjournals.org Cancer Res; 72(24) December 15, 2012 OF7
American Association for Cancer Research Copyright © 2012 on December 11, 2012cancerres.aacrjournals.orgDownloaded from
Published OnlineFirst on December 7, 2012; DOI:10.1158/0008-5472.CAN-12-0762
reduced tumor growth (tumor volume at 20 days: 405 80 in
the mNEM group vs. 810 180 in the controls; P<0.01; Fig. 5C).
Similar to bone marrow NEMs, the antitumoral activity of
mNEMs correlated with their capacity to promote tumor vessel
maturation, as evaluated from the extent of pericyte coverage
after staining for CD31 and a-SMA (Fig. 5D and quantied
in Fig. 5E).
Paracrine activity of NEMs toward vascular smooth
muscle cells
To understand the mechanisms by which NEMs were capa-
ble of normalizing the tumor vasculature, and leading from the
reasonable assumption that this effect was exerted in a para-
crine fashion, we analyzed the levels of expression of a series of
secreted factors and their receptors participating in blood
vessel formation. Bone marrow NEMs produced a few chemo-
kines traditionally associated with M1 macrophages and
known to generally exert antitumoral activity (33), such as
CCL2, CCL4, CCL5, CXCL9, and CXCL10, as well as other
different molecules involved in vessel stabilization (such as
PDGFb, TGF-b, SDF-1, and the antiangiogenic molecule
thrombospondin-1; ref. 34; Fig. 6A). In contrast, NEMs did not
express CCL1, CCL17, and CCL22, chemokines traditionally
associated with M2 macrophages, which have proangiogenic
properties (33). A full list of the analyzed molecules is provided
in Supplementary Table S1.
Figure 5. NEMs recovered from
Sema3A-expressing muscles
inhibit tumor growth and promote
vessel maturation. A, cells
inltrating muscles 15 days after
injection of AAV2-Sema3A.
Hematoxylin staining (a) and
immunouorescence for CD11b
(b), Nrp1 (c), Gr1 (d), or double
immunouorescence for Nrp1 and
Gr1 (e, with split signals in f and g).
Nuclei are counterstained with
DAPI; scale bar, 50 mm. B, ow
cytometry of mNEMs. Cells
puried from Sema3A-expressing
muscles were gated for
morphology (top) and analyzed for
expression of CD11b and Nrp1
(bottom). C, growth curve of B16.
F10 melanoma tumors implanted
(day 0) in C57/BL6 mice and
injected with mNEMs (mean
SEM). D, vessels in tumors treated
with mNEMs or PBS at 2 different
magnications. Green, CD31; red,
a-SMA; blue, DAPI. Scale bars, 50
mm. E, a-SMA/CD31 ratio in tumors
treated with mNEMs or PBS (mean
SEM; ,P<0.01).
Carrer et al.
Cancer Res; 72(24) December 15, 2012 Cancer Research
OF8
American Association for Cancer Research Copyright © 2012 on December 11, 2012cancerres.aacrjournals.orgDownloaded from
Published OnlineFirst on December 7, 2012; DOI:10.1158/0008-5472.CAN-12-0762
The presence of vessels covered by a-SMA and NG2-positive
cells was the most striking feature of NEM-injected tumors.
Thus, we assessed whether the factors produced by NEMs had
a direct effect on vascular smooth muscle cell (vSMC) prolif-
eration or migration. vSMCs were seeded in a 24-well plate and
cocultured with bone marrow-puried NEMs or Gr1þcells.
The percentage of proliferating cells was assessed 20 hours
after NEM addition. No signicant effect was detected on
vSMC proliferation by either cell subset (Fig. 6B). Next, we
tested the migratory capacity of vSMCs upon stimulation by
NEMs or Gr1þcells in a Transwell chamber assay. Notably,
NEMs attracted vSMCs at a signicantly greater extent than
Gr1þcells and at levels similar to the stimulation exerted by
FBS (Fig. 6C; P<0.01).
Collectively, these ndings support the conclusion that
NEMs exert their function in a paracrine manner by secreting
factors that stimulate vessel maturation and, in particular,
coverage of endothelial cells through the attraction of mural
cells.
Discussion
The existence of a population of myeloid cells that was
specically chemoattracted by Nrp1 ligands was originally
unveiled by the observations that VEGF-A
165
and Sema3A,
which both bind Nrp1, were able to induce the migration of
bone marrowderived, CD11bþcells in vitro and in vivo (16
18). We show here that the subset of CD11bþNrp1þGr1
monocytes (NEMs), puried from the normal bone marrow,
specically respond to Sema3A chemoattraction and possess
unique properties in terms of cytokine expression and func-
tional activity.
The ow cytometric prole of bone marrowderived NEMs
displayed several characteristics common with mouse resident
monocytes, including low expression of Gr1/Ly6C and CD14,
high levels of CD16, intermediate levels of F4/80, as well as
expression of low levels of CXCR4 and CD31 (23, 35, 36). Other
studies have indicated that Nrp1 is expressed by subsets of
myeloid cells both in tumors, where they exert proangiogenic
activity (37), and during development (38). In this respect,
however, Nrp1 expression is known to signicantly increase
during monocyte maturation (39), whereas this receptor is
expressed at very low levels in the bone marrow (40). Thus,
although Nrp1 expression distinguishes NEMs among bone
marrow Gr1monocytes, expression of this marker might also
be acquired by other cells in different conditions. In any case,
gene expression proling and surface marker characterization
clearly indicate that NEMs stand apart from other monocyte
populations that exert proangiogenic activity, such as TAMs,
which express a different panel of cytokines from NEMs (41), or
TEMs, which are positive for Tie2 expression (5), whereas
NEMs are clearly negative. In addition, while Sema3A acts as
a chemoattractant for NEMs, it is known to exert an apoptotic
effect for the M2 monocyte/macrophages also expressing
Nrp1 (39).
Figure 6. Effects of NEMs toward
vascular smooth muscle cells. A,
expression of genes involved in
macrophage polarization and vessel
maturation in NEMs (mean SEM
after normalization for a
housekeeping gene). B, EdU-
positive vSMCs upon coculture with
210
4
Gr1þcells or NEMs, or in the
presence of 20% FBS (positive
control; mean SEM). C, vSMCs
migrated in response to NEMs or
Gr1þcells (mean SEM;
,P<0.01).
NEMs Induce Tumor Vessel Stabilization
www.aacrjournals.org Cancer Res; 72(24) December 15, 2012 OF9
American Association for Cancer Research Copyright © 2012 on December 11, 2012cancerres.aacrjournals.orgDownloaded from
Published OnlineFirst on December 7, 2012; DOI:10.1158/0008-5472.CAN-12-0762
Among the bone marrow Nrp1þmonocytes, only the Gr1
fraction (NEMs) was able to migrate in response to Sema3A;
this is consistent with the observation that all the cells inl-
trating the muscle overexpressing Sema3A were Gr1. After
Sema3A stimulation, Nrp1 signals through heterodimerization
with 1 of the 4 type A plexins, which serve as signal-transducing
elements (42, 43). Gene expression analysis indicated that the
levels of plexin A1 and A3 were higher in NEMs compared with
Gr1þcells, possibly suggesting that these 2 receptors are
involved in the response to Sema3A. Further experiments are
clearly required to verify this possibility.
Once inoculated in vivo, NEMs persisted in the injected
tumors and markedly inhibited tumor growth in the 2 inves-
tigated tumor models. These cells were found to express
several factors involved in vessel maturation (including
PDGFb, TGF-b, and thrombospondin; ref. 44). The vasculature
of the tumors injected with NEMs showed increased pericyte
coverage and mural thickness, more regular shape, reduced
leakiness, and, most importantly, improved function. This is
also consistent with the observation that NEMs selectively
produced factors able to chemoattract vSMCs. In vivo, the
ultimate outcome of these effects was the improvement of
tumor mass perfusion and reduction of tumor hypoxia, which
is a major determinant of tumor progression (13). These
morphological and functional parameters t the concept of
vessel normalization, as originally put forward as a goal to
improve blood supply and enhance delivery of chemothera-
peutic drugs by low-dose antiangiogenic agents (14, 45).
We and others have shown that Sema3A functions as potent
antiangiogenic factors and inhibits tumor development (28
32). In particular, the ectopic delivery of an AAV vector
expressing Sema3A into RipTag2 mice signicantly reduced
vascular density and branching, inhibited tumor growth and
metastasis, and substantially extended survival (28, 29). Con-
sistent with the possibility that the recruitment of NEMs might,
at least in part, be responsible for the effects of Sema3A, we
found that the cells recruited by Sema3A in vivo actually
exerted signicant antitumoral activity. Thus, although no
evidence exists at this moment to show that NEMs might play
a physiological role during normal tumor development, they
might mediate the effects of Sema3A treatment on tumor
growth. In line with this concept, bone marrow NEM inoc-
ulation can be conceived as an experimental treatment able
to normalize the tumor vasculature. The vascular phenotype
obtained by the inoculation of NEMs, at least in the inves-
tigated tumor models, resembles that observed through a
variety of genetic approaches, including the depletion of
endothelial HIF-1a(27), the overexpression of the histidine
rich glycoprotein to skew the TAM response toward an
M1 phenotype (25), or the haploinsufency of endothelial
PHD2 (46). In addition, vascular normalization was observed
upon administration of anti-PlGF antibodies (26). All these
conditions are known to improve tumor oxygenation and
lessen tumor malignancy and invasiveness.
Disclosure of Potential Conicts of Interest
No potential conicts of interest were disclosed.
Authors' Contributions
Conception and design: A. Carrer, S. Moimas, S. Zacchigna, G. Serini,
E. Giraudo, F. Bussolino, M. Giacca
Development of methodology: A. Carrer, S. Moimas, S. Zacchigna, M.
Sinigaglia
Acquisition of data: S. Moimas, L. Pattarini, L. Zentilin, G. Ruozi, M. Mano,
M. Sinigaglia
Analysis and interpretation of data (e.g., statistical analysis, biostatistics,
computational analysis): A. Carrer, S. Moimas, S. Zacchigna
Writing, review, and/or revision of the manuscript: A. Carrer, S. Moimas,
G. Serini, E. Giraudo, F. Bussolino, M. Giacca
Administrative, technical, or material support (i.e., reporting or orga-
nizing data, constructing databases): F. Maione
Study supervision: S. Zacchigna, M. Giacca
Acknowledgments
The authors thank Mauro Sturnega for assistance in animal experimentation,
Paolo Maiuri for immunouorescence analysis, and Suzanne Kerbavcic for
editorial work.
Grant Support
Financial support for this work was provided by grants from the "Fondazione
CR Trieste," Trieste, Italy, the FIRB RBAP11Z4Z9 project, and the European
Research Council (ERC) Advanced Grant 250124 FunSel to M. Giacca, and the
Associazione Italiana per la Ricerca sul Cancro (AIRC) to S. Zacchigna, G. Serini,
E. Giraudo, and F. Bussolino.
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.
Received February 28, 2012; revised September 5, 2012; accepted September 5,
2012; published OnlineFirst December 7, 2012.
References
1. Murdoch C, Muthana M, Coffelt SB, Lewis CE. The role of myeloid cells
in the promotion of tumour angiogenesis. Nat Rev Cancer 2008;8:618
31.
2. Papaspyridonos M, Lyden D. Chapter 11. The role of bone marrow-
derived cells in tumor angiogenesis and metastatic progression.
Methods Enzymol 2008;444:25569.
3. Shojaei F, Zhong C, Wu X, Yu L, Ferrara N. Role of myeloid cells in
tumor angiogenesis and growth. Trends Cell Biol 2008;18:3728.
4. 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;
23:54955.
5. De Palma M, Venneri MA, Galli R, Sergi Sergi L, Politi LS, Sampaolesi
M, et al. Tie2 identies a hematopoietic lineage of proangiogenic
monocytes required for tumor vessel formation and a mesenchymal
population of pericyte progenitors. Cancer Cell 2005;8:21126.
6. Shojaei F, Wu X, Malik AK, Zhong C, Baldwin ME, Schanz S, et al.
Tumor refractoriness to anti-VEGF treatment is mediated by
CD11bþGr1þmyeloid cells. Nat Biotechnol 2007;25:91120.
7. Fridlender ZG, Sun J, Kim S, Kapoor V, Cheng G, Ling L, et al.
Polarization of tumor-associated neutrophil phenotype by TGF-beta:
"N1" versus "N2" TAN. Cancer Cell 2009;16:18394.
8. Gao D, Mittal V. The role of bone-marrow-derived cells in tumor
growth, metastasis initiation and progression. Trends Mol Med
2009;15:33343.
9. Loges S, Schmidt T, Carmeliet P. "Antimyeloangiogenic" therapy for
cancer by inhibiting PlGF. Clin Cancer Res 2009;15:364853.
10. Stockmann C, Doedens A, Weidemann A, Zhang N, Takeda N, Green-
berg JI, et al. Deletion of vascular endothelial growth factor in myeloid
cells accelerates tumorigenesis. Nature 2008;456:8148.
11. Carmeliet P. Angiogenesis in life, disease and medicine. Nature
2005;438:9326.
Carrer et al.
Cancer Res; 72(24) December 15, 2012 Cancer Research
OF10
American Association for Cancer Research Copyright © 2012 on December 11, 2012cancerres.aacrjournals.orgDownloaded from
Published OnlineFirst on December 7, 2012; DOI:10.1158/0008-5472.CAN-12-0762
12. McDonald DM, Choyke PL. Imaging of angiogenesis: from microscope
to clinic. Nat Med 2003;9:71325.
13. Wilson WR, Hay MP. Targeting hypoxia in cancer therapy. Nat Rev
Cancer 2011;11:393410.
14. Jain RK. Normalization of tumor vasculature: an emerging concept in
antiangiogenic therapy. Science 2005;307:5862.
15. Carmeliet P, Jain RK. Principles and mechanisms of vessel normal-
ization for cancer and other angiogenic diseases. Nat Rev Drug Discov
2011;10:41727.
16. Grunewald M, Avraham I, Dor Y, Bachar-Lustig E, Itin A, Yung S, et al.
VEGF-induced adult neovascularization: recruitment, retention, and
role of accessory cells. Cell 2006;124:17589.
17. Zacchigna S, Pattarini L, Zentilin L, Moimas S, Carrer A, Sinigaglia M,
et al. Bone marrow cells recruited through the neuropilin-1 receptor
promote arterial formation at the sites of adult neoangiogenesis. J Clin
Invest 2008;118:206275.
18. Zentilin L, Tafuro S, Zacchigna S, Arsic N, Pattarini L, Sinigaglia M, et al.
Bone marrow mononuclear cells are recruited to the sites of VEGF-
induced neovascularization but are not incorporated into the newly
formed vessels. Blood 2006;107:354654.
19. Tomayko MM, Reynolds CP. Determination of subcutaneous tumor
size in athymic (nude) mice. Cancer Chemother Pharmacol 1989;
24:14854.
20. Laginha KM, Verwoert S, Charrois GJ, Allen TM. Determination of
doxorubicin levels in whole tumor and tumor nuclei in murine breast
cancer tumors. Clin Cancer Res 2005;11:69449.
21. Sturn A, Quackenbush J, Trajanoski Z. Genesis: cluster analysis of
microarray data. Bioinformatics 2002;18:2078.
22. Ingersoll MA, Spanbroek R, Lottaz C, Gautier EL, Frankenberger M,
Hoffmann R, et al. Comparison of gene expression proles between
human and mouse monocyte subsets. Blood 2010;115:e109.
23. Auffray C, Sieweke MH, Geissmann F. Blood monocytes: develop-
ment, heterogeneity, and relationship with dendritic cells. Annu Rev
Immunol 2009;27:66992.
24. Geissmann F, Manz MG, Jung S, Sieweke MH, Merad M, Ley K.
Development of monocytes, macrophages, and dendritic cells. Sci-
ence 2010;327:65661.
25. Rolny C, Mazzone M, Tugues S, Laoui D, Johansson I, Coulon C, et al.
HRG inhibits tumor growth and metastasis by inducing macrophage
polarization and vessel normalization through downregulation of PlGF.
Cancer Cell 2011;19:3144.
26. Fischer C, Jonckx B, Mazzone M, Zacchigna S, Loges S, Pattarini L,
et al. Anti-PlGF inhibits growth of VEGF(R)-inhibitor-resistant tumors
without affecting healthy vessels. Cell 2007;131:46375.
27. Keith B, Johnson RS, Simon MC. HIF1alpha and HIF2alpha: sibling
rivalry in hypoxic tumour growth and progression. Nat Rev Cancer
2012;12:922.
28. Maione F, Molla F, Meda C, Latini R, Zentilin L, Giacca M, et al.
Semaphorin 3A is an endogenous angiogenesis inhibitor that blocks
tumor growth and normalizes tumor vasculature in transgenic mouse
models. J Clin Invest 2009;119:335672.
29. Maione F, Capano S, Regano D, Zentilin L, Giacca M, Casanovas O,
et al. Semaphorin 3A overcomes cancer hypoxia and metastatic
dissemination induced by antiangiogenic treatment in mice. J Clin
Invest 2012;122:183248.
30. Casazza A, Fu X, Johansson I, Capparuccia L, Andersson F, Gius-
tacchini A, et al. Systemic and targeted delivery of semaphorin 3A
inhibits tumor angiogenesis and progression in mouse tumor models.
Arterioscler Thromb Vasc Biol 2011;31:7419.
31.Acevedo LM, Barillas S, Weis SM, Gothert JR, Cheresh DA. Sema-
phorin 3A suppresses VEGF-mediated angiogenesis yet acts as a
vascular permeability factor. Blood 2008;111:267480.
32. Kigel B, Varshavsky A, Kessler O, Neufeld G. Successful inhibition of
tumor development by specic class-3 semaphorins is associated
with expression of appropriate semaphorin receptors by tumor cells.
PLoS One 2008;3:e3287.
33. Mantovani A, Sozzani S, Locati M, Schioppa T, Saccani A, Allavena P,
et al. Inltration of tumours by macrophages and dendritic cells:
tumour-associated macrophages as a paradigm for polarized M2
mononuclear phagocytes. Novartis Found Symp 2004;256:13745;
discussion 468, 25969.
34. Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases.
Nature 2000;407:24957.
35. Geissmann F, Jung S, Littman DR. Blood monocytes consist of two
principal subsets with distinct migratory properties. Immunity
2003;19:7182.
36. Saha P, Geissmann F. Toward a functional characterization of blood
monocytes. Immunol Cell Biol 2010;89:24.
37. Pucci F, Venneri MA, Biziato D, Nonis A, Moi D, Sica A, et al. A
distinguishing gene signature shared by tumor-inltrating Tie2-
expressing monocytes, blood "resident" monocytes, and embryonic
macrophages suggests common functions and developmental rela-
tionships. Blood 2009;114:90114.
38. Fantin A, Vieira JM, Gestri G, Denti L, Schwarz Q, Prykhozhij S, et al.
Tissue macrophages act as cellular chaperones for vascular anasto-
mosis downstream of VEGF-mediated endothelial tip cell induction.
Blood 2010;116:82940.
39. Ji JD, Park-Min KH, Ivashkiv LB. Expression and function of sema-
phorin 3A and its receptors in human monocyte-derived macro-
phages. Hum Immunol 2009;70:2117.
40. Karjalainen K, Jaalouk DE, Bueso-Ramos CE, Zurita AJ, Kuniyasu A,
Eckhardt BL, et al. Targeting neuropilin-1 in human leukemia and
lymphoma. Blood 2011;117:9207.
41. Sica A, Larghi P, Mancino A, Rubino L, Porta C, Totaro MG, et al.
Macrophage polarization in tumour progression. Semin Cancer Biol
2008;18:34955.
42. Neufeld G, Kessler O. The semaphorins: versatile regulators of tumour
progression and tumour angiogenesis. Nat Rev Cancer 2008;8:632
45.
43. Gaur P, Bielenberg DR, Samuel S, Bose D, Zhou Y, Gray MJ, et al. Role
of class 3 semaphorins and their receptors in tumor growth and
angiogenesis. Clin Cancer Res 2009;15:676370.
44. Jain RK. Molecular regulation of vessel maturation. Nat Med 2003;9:
68593.
45. Jain RK. A new target for tumor therapy. N Engl J Med 2009;
360:266971.
46. Mazzone M, Dettori D, Leite de Oliveira R, Loges S, Schmidt T, Jonckx
B, et al. Heterozygous deciency of PHD2 restores tumor oxygenation
and inhibits metastasis via endothelial normalization. Cell 2009;136:
83951.
NEMs Induce Tumor Vessel Stabilization
www.aacrjournals.org Cancer Res; 72(24) December 15, 2012 OF11
American Association for Cancer Research Copyright © 2012 on December 11, 2012cancerres.aacrjournals.orgDownloaded from
Published OnlineFirst on December 7, 2012; DOI:10.1158/0008-5472.CAN-12-0762
... A somewhat diff erent picture is observed in mice. Nrp-1 expression is registered already on early bone marrow-derived monocyte/macrophage precursor cells [60] and on circulating monocytes [61]. Moreover, this Nrp-1 + bone marrow monocyte/macrophage precursors also express PlexA1 and PlexA3 mRNAs [60]. ...
... Nrp-1 expression is registered already on early bone marrow-derived monocyte/macrophage precursor cells [60] and on circulating monocytes [61]. Moreover, this Nrp-1 + bone marrow monocyte/macrophage precursors also express PlexA1 and PlexA3 mRNAs [60]. During the maturation of bone marrow-derived macrophages treated with M-CSF, mRNAs of the following receptors can be detected: Nrp-1, PlexA1, A2, and A4, but not PlexA3 [18,61]. ...
... Semaphorin 3A is a chemoattractant for macrophages and enhances their spontaneous migration along a positive concentration gradient. This was shown in vitro with the use of transwell system for murine bone marrow-derived monocyte precursor cells [60,64], bone marrow-derived [61], and peritoneal [17] macrophages, as well as for human CD14 + macrophages isolated from the liver of patients with hepatocellular carcinoma [65]. Semaphorin 3A also stimulates in vitro migration of bone marrow-derived murine macrophages in "wound-healing" assay (migration of cells into a scratch) [63]. ...
... A somewhat diff erent picture is observed in mice. Nrp-1 expression is registered already on early bone marrow-derived monocyte/macrophage precursor cells [60] and on circulating monocytes [61]. Moreover, this Nrp-1 + bone marrow monocyte/macrophage precursors also express PlexA1 and PlexA3 mRNAs [60]. ...
... Nrp-1 expression is registered already on early bone marrow-derived monocyte/macrophage precursor cells [60] and on circulating monocytes [61]. Moreover, this Nrp-1 + bone marrow monocyte/macrophage precursors also express PlexA1 and PlexA3 mRNAs [60]. During the maturation of bone marrow-derived macrophages treated with M-CSF, mRNAs of the following receptors can be detected: Nrp-1, PlexA1, A2, and A4, but not PlexA3 [18,61]. ...
... Semaphorin 3A is a chemoattractant for macrophages and enhances their spontaneous migration along a positive concentration gradient. This was shown in vitro with the use of transwell system for murine bone marrow-derived monocyte precursor cells [60,64], bone marrow-derived [61], and peritoneal [17] macrophages, as well as for human CD14 + macrophages isolated from the liver of patients with hepatocellular carcinoma [65]. Semaphorin 3A also stimulates in vitro migration of bone marrow-derived murine macrophages in "wound-healing" assay (migration of cells into a scratch) [63]. ...
Article
Full-text available
Abstract—Semaphorin 3A is a secreted glycoprotein, which was originally identifi ed as axon guidance factor in the neuronal system, but it also possesses immunoregulatory properties. Here, the eff ect of semaphorin 3A on T-lymphocytes, myeloid dendritic cells and macrophages is systematically analyzed on the bases of all publications available in the literature for 20 years. Expression of semaphorin 3A receptors – neuropilin-1 and plexins A – in these cells is described in details. The data obtained on human and murine cells is described comparatively. A comprehensive overview of the interaction of semaphorin 3A with mononuclear phagocyte system is presented for the fi rst time. Semaphorin 3A signaling mostly results in changes of the cytoskeletal machinery and cellular morphology that regulate pathways involved in migration, adhesion, and cell–cell cooperation of immune cells. Accumulating evidence indicates that this factor is crucially involved in various phases of immune responses, including initiation phase, antigen presentation, eff ector T cell function, infl ammation phase, macrophage activation, and polarization. In recent years, interest in this fi eld has increased signifi cantly because semaphorin 3A is associated with many human diseases and therefore can be used as a target for their treatment. Its involvement in the immune responses is important to study, because semaphorin 3A and its receptors turn to be a promising new therapeutic tools to be applied in many autoimmune, allergic, and oncology diseases.
... A somewhat diff erent picture is observed in mice. Nrp-1 expression is registered already on early bone marrow-derived monocyte/macrophage precursor cells [60] and on circulating monocytes [61]. Moreover, this Nrp-1 + bone marrow monocyte/macrophage precursors also express PlexA1 and PlexA3 mRNAs [60]. ...
... Nrp-1 expression is registered already on early bone marrow-derived monocyte/macrophage precursor cells [60] and on circulating monocytes [61]. Moreover, this Nrp-1 + bone marrow monocyte/macrophage precursors also express PlexA1 and PlexA3 mRNAs [60]. During the maturation of bone marrow-derived macrophages treated with M-CSF, mRNAs of the following receptors can be detected: Nrp-1, PlexA1, A2, and A4, but not PlexA3 [18,61]. ...
... Semaphorin 3A is a chemoattractant for macrophages and enhances their spontaneous migration along a positive concentration gradient. This was shown in vitro with the use of transwell system for murine bone marrow-derived monocyte precursor cells [60,64], bone marrow-derived [61], and peritoneal [17] macrophages, as well as for human CD14 + macrophages isolated from the liver of patients with hepatocellular carcinoma [65]. Semaphorin 3A also stimulates in vitro migration of bone marrow-derived murine macrophages in "wound-healing" assay (migration of cells into a scratch) [63]. ...
Article
Semaphorin 3A is a secreted glycoprotein, which was originally identified as axon guidance factor in the neuronal system, but it also possesses immunoregulatory properties. Here, the effect of semaphorin 3A on T-lymphocytes, myeloid dendritic cells and macrophages is systematically analyzed on the bases of all publications available in the literature for 20 years. Expression of semaphorin 3A receptors - neuropilin-1 and plexins A - in these cells is described in details. The data obtained on human and murine cells is described comparatively. A comprehensive overview of the interaction of semaphorin 3A with mononuclear phagocyte system is presented for the first time. Semaphorin 3A signaling mostly results in changes of the cytoskeletal machinery and cellular morphology that regulate pathways involved in migration, adhesion, and cell-cell cooperation of immune cells. Accumulating evidence indicates that this factor is crucially involved in various phases of immune responses, including initiation phase, antigen presentation, effector T cell function, inflammation phase, macrophage activation, and polarization. In recent years, interest in this field has increased significantly because semaphorin 3A is associated with many human diseases and therefore can be used as a target for their treatment. Its involvement in the immune responses is important to study, because semaphorin 3A and its receptors turn to be a promising new therapeutic tools to be applied in many autoimmune, allergic, and oncology diseases.
... TAMs have an established protumoural function and shares features with M2-like macrophages, including the expression of Arginase 1 and of the Mannose Receptor CD206. 61 62 Carrer et al reported that SEMA3A recruits a subset of resident Nrp1+antitumoural macrophages, 63 while Casazza et al found that SEMA3A entraps protumoural macrophages in highly hypoxic areas. 64 Finally, Wallerius et al reported a differential effect of SEMA3A on the proliferation of M2 and M1-like macrophages 65 : SEMA3A favoured the expansion of antitumoural M1-like macrophages which was associated with the recruitment of cytotoxic T cells and a tumour-inhibiting effect. ...
Article
Full-text available
Objective The dysregulation of the axon guidance pathway is common in pancreatic ductal adenocarcinoma (PDAC), yet our understanding of its biological relevance is limited. Here, we investigated the functional role of the axon guidance cue SEMA3A in supporting PDAC progression. Design We integrated bulk and single-cell transcriptomic datasets of human PDAC with in situ hybridisation analyses of patients’ tissues to evaluate SEMA3A expression in molecular subtypes of PDAC. Gain and loss of function experiments in PDAC cell lines and organoids were performed to dissect how SEMA3A contributes to define a biologically aggressive phenotype. Results In PDAC tissues, SEMA3A is expressed by stromal elements and selectively enriched in basal-like/squamous epithelial cells. Accordingly, expression of SEMA3A in PDAC cells is induced by both cell-intrinsic and cell-extrinsic determinants of the basal-like phenotype. In vitro , SEMA3A promotes cell migration as well as anoikis resistance. At the molecular level, these phenotypes are associated with increased focal adhesion kinase signalling through canonical SEMA3A-NRP1 axis. SEMA3A provides mouse PDAC cells with greater metastatic competence and favours intratumoural infiltration of tumour-associated macrophages and reduced density of T cells. Mechanistically, SEMA3A functions as chemoattractant for macrophages and skews their polarisation towards an M2-like phenotype. In SEMA3A high tumours, depletion of macrophages results in greater intratumour infiltration by CD8+T cells and better control of the disease from antitumour treatment. Conclusions Here, we show that SEMA3A is a stress-sensitive locus that promotes the malignant phenotype of basal-like PDAC through both cell-intrinsic and cell-extrinsic mechanisms.
... To optimize the model for in vivo selection, we compared the growth pattern of three cancer cell lines (B16-F10 melanoma, 4T1 breast cancer, and Lewis Lung Carcinoma, LLC) upon implantation into syngeneic skeletal muscle, which is highly permissive to AAV transduction [17,18]. B16-F10 and 4T1 cells formed a compact tumor mass, squishing, but not destroying, surrounding muscle fibers. ...
Article
Full-text available
Background New drugs to tackle the next pathway or mutation fueling cancer are constantly proposed, but 97% of them are doomed to fail in clinical trials, largely because they are identified by cellular or in silico screens that cannot predict their in vivo effect. Methods We screened an Adeno-Associated Vector secretome library (> 1000 clones) directly in vivo in a mouse model of cancer and validated the therapeutic effect of the first hit, EMID2, in both orthotopic and genetic models of lung and pancreatic cancer. Results EMID2 overexpression inhibited both tumor growth and metastatic dissemination, consistent with prolonged survival of patients with high levels of EMID2 expression in the most aggressive human cancers. Mechanistically, EMID2 inhibited TGFβ maturation and activation of cancer-associated fibroblasts, resulting in more elastic ECM and reduced levels of YAP in the nuclei of cancer cells. Conclusion This is the first in vivo screening, precisely designed to identify proteins able to interfere with cancer cell invasiveness. EMID2 was selected as the most potent protein, in line with the emerging relevance of the tumor extracellular matrix in controlling cancer cell invasiveness and dissemination, which kills most of cancer patients.
... With respect to being pro-angiogenic, NRP1-expressing mononuclear phagocytes have been described as dispensable for physiological angiogenesis in the retina and elsewhere (Fantin et al, 2013;Dejda et al, 2016), yet important for vessel growth during weight gain (Wilson et al, 2018). They have also been reported to normalize tumor blood vessels (Carrer et al, 2012) and promote pathological angiogenesis in the retina (Dejda et al, 2014) and tumors (Casazza et al, 2013). ...
Article
Full-text available
Age-related macular degeneration (AMD) in its various forms is a leading cause of blindness in industrialized countries. Here, we provide evidence that ligands for neuropilin-1 (NRP1), such as Semaphorin 3A and VEGF-A, are elevated in the vitreous of patients with AMD at times of active choroidal neovascularization (CNV). We further demonstrate that NRP1-expressing myeloid cells promote and maintain CNV. Expression of NRP1 on cells of myeloid lineage is critical for mitigating production of inflammatory factors such as IL6 and IL1β. Therapeutically trapping ligands of NRP1 with an NRP1-derived trap reduces CNV. Collectively, our findings identify a role for NRP1-expressing myeloid cells in promoting pathological angiogenesis during CNV and introduce a therapeutic approach to counter neovascular AMD.
Preprint
Full-text available
Background. New drugs to tackle the next pathway or mutation fueling cancer are constantly proposed, but 97% of them are doomed to fail in clinical trials, largely because they are identified by cellular or in silico screens that cannot predict their in vivo effect. Methods. We screened an Adeno-Associated Vector secretome library (> 1000 clones) directly in vivo in a mouse model of cancer and validated the therapeutic effect of the first hit, EMID2, in both orthotopic and genetic models of lung and pancreatic cancer. Results. EMID2 overexpression inhibited both tumor growth and metastatic dissemination, consistent with prolonged survival of patients with high levels of EMID2 expression in the most aggressive human cancers. Mechanistically, EMID2 inhibited TGFβ maturation and activation of cancer-associated fibroblasts, resulting in more elastic ECM and reduced levels of YAP in the nuclei of cancer cells. Conclusions. This is the first in vivo screening, precisely designed to identify proteins able to interfere with cancer cell invasiveness. EMID2 was selected as the most potent protein, in line with the emerging relevance of the tumor extracellular matrix in controlling cancer cell invasiveness and dissemination, which kills most of cancer patients.
Article
Full-text available
Umbilical cord blood (UCB) transplantation shows proangiogenic effects and contributes to symptom amelioration in animal models of cerebral infarction. However, the effect of specific cell types within a heterogeneous UCB population are still controversial. OP9 is a stromal cell line used as feeder cells to promote the hematoendothelial differentiation of embryonic stem cells. Hence, we investigated the changes in angiogenic properties, underlying mechanisms, and impact on behavioral deficiencies caused by cerebral infarction in UCB co-cultured with OP9 for up to 24 h. In the network formation assay, only OP9 pre-conditioned UCB formed network structures. Single-cell RNA sequencing and flow cytometry analysis showed a prominent phenotypic shift toward M2 in the monocytic fraction of OP9 pre-conditioned UCB. Further, OP9 pre-conditioned UCB transplantation in mice models of cerebral infarction facilitated angiogenesis in the peri-infarct lesions and ameliorated the associated symptoms. In this study, we developed a strong, fast, and feasible method to augment the M2, tissue-protecting, pro-angiogenic features of UCB using OP9. The ameliorative effect of OP9-pre-conditioned UCB in vivo could be partly due to promotion of innate angiogenesis in peri-infarct lesions.
Chapter
Chronic inflammation is closely related to tumorigenesis. Chronic inflammatory mediators exert pleiotropic effects in the development of cancer. On the one hand, inflammation favors carcinogenesis, malignant transformation, tumor growth, invasion, and metastatic spread; on the other hand, inflammation can stimulate immune effector mechanisms that might limit tumor growth. Inflammatory cells involved include neutrophils, eosinophils, dendritic cells, lymphocytes, NK cells, platelets, mast cells, and monocytes/macrophages.
Article
Plasmacytoid dendritic cells (pDCs) are a unique subpopulation of immune cells, distinct from classical dendritic cells. pDCs are generated in the bone marrow, and following development, they typically home to secondary lymphoid tissues. Nevertheless, while peripheral tissues are generally devoid of pDCs during steady state, few tissues, including the lung, kidney, vagina, and in particular ocular tissues harbor resident pDCs. pDCs were originally appreciated for their potential to produce large quantities of type I interferons in viral immunity. Subsequent studies have now unraveled their pivotal role in mediating immune responses, in particular in the induction of tolerance. In this review, we summarize our current knowledge on pDCs in ocular tissues in both mice and humans, in particular in the cornea, limbus, conjunctiva, choroid, retina, and lacrimal gland. Further, we will review our current understanding on the significance of pDCs in ameliorating inflammatory responses during herpes simplex virus keratitis, sterile inflammations, and corneal transplantation. Moreover, we describe their novel and pivotal neuroprotective role, their key function in preserving corneal angiogenic privilege, as well as their potential application, as a cell-based therapy for ocular diseases.
Article
Full-text available
On pages e10 and e17 of the January 21, 2010, issue, there are errors in the affiliations and “Discussion” that have been corrected in the online full text and PDF. On page e10, the affiliations of the sixth and seventh authors (Reinhard Hoffmann and Roland Lang) were listed incorrectly. The correct byline and affiliations should have read as shown. Molly A. Ingersoll,¹ Rainer Spanbroek,² Claudio Lottaz,³ Emmanuel L. Gautier,¹ Marion Frankenberger,⁴ Reinhard Hoffmann,⁵ Roland Lang,⁶ Muzlifah Haniffa,⁷ Matthew Collin,⁷ Frank Tacke,¹ Andreas J. R. Habenicht,² Loems Ziegler-Heitbrock,⁴ and Gwendalyn J. Randolph¹ ¹Department of Gene and Cell Medicine and Immunology Institute, Mount Sinai School of Medicine, New York, NY; ²Institute for Vascular Medicine, Friedrich Schiller University of Jena, Jena, Germany; ³Institute of Functional Genomics, University of Regensburg, Regensburg, Germany; ⁴Clinical Cooperation Group “Inflammatory Lung Diseases,” Asklepios-Fachklinik and Helmholtz Zentrum München, German Research Center for Environmental Health, Gauting, Germany; ⁵Institute for Medical Microbiology, Immunology und Hygiene, Technische Universität München, München, Germany; ⁶Institute of Clinical Microbiology, Immunology and Hygiene, University Hospital Erlangen, Erlangen, Germany; and ⁷Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom On page e17, in the second sentence of the fourth paragraph of the “Discussion,” the figure references were incorrect in the sentence that read, “These included CXCR4, TREM-1, CD36, and CD9 (Figures 1B and 2B, and supplemental Figure 4). The sentence should have read: “These included CXCR4, TREM-1, CD36, and CD9 (Figures 2-4).” • © 2010 by The American Society of Hematology
Article
Full-text available
Cancer development, progression, and metastasis are highly dependent on angiogenesis. The use of antiangiogenic drugs has been proposed as a novel strategy to interfere with tumor growth, but cancer cells respond by developing strategies to escape these treatments. In particular, animal models show that antiangiogenic drugs currently used in clinical settings reduce tumor tissue oxygenation and trigger molecular events that foster cancer resistance to therapy. Here, we show that semaphorin 3A (Sema3A) expression overcomes the proinvasive and prometastatic resistance observed upon angiogenesis reduction by the small-molecule tyrosine inhibitor sunitinib in both pancreatic neuroendocrine tumors (PNETs) in RIP-Tag2 mice and cervical carcinomas in HPV16/E2 mice. By improving cancer tissue oxygenation and extending the normalization window, Sema3A counteracted sunitinib-induced activation of HIF-1α, Met tyrosine kinase receptor, epithelial-mesenchymal transition (EMT), and other hypoxia-dependent signaling pathways. Sema3A also reduced tumor hypoxia and halted cancer dissemination induced by DC101, a specific inhibitor of the VEGF pathway. As a result, reexpressing Sema3A in cancer cells converts metastatic PNETs and cervical carcinomas into benign lesions. We therefore suggest that this strategy could be developed to safely harnesses the therapeutic potential of the antiangiogenic treatment.
Article
Full-text available
Hypoxia-inducible factors (HIFs) are broadly expressed in human cancers, and HIF1α and HIF2α were previously suspected to promote tumour progression through largely overlapping functions. However, this relatively simple model has now been challenged in light of recent data from various approaches that reveal unique and sometimes opposing activities of these HIFα isoforms in both normal physiology and disease. These effects are mediated in part through the regulation of unique target genes, as well as through direct and indirect interactions with important oncoproteins and tumour suppressors, including MYC and p53. As HIF inhibitors are currently undergoing clinical evaluation as cancer therapeutics, a more thorough understanding of the unique roles performed by HIF1α and HIF2α in human neoplasia is warranted.
Article
Full-text available
The use of various transgenic mouse models and analysis of human tumour biopsies has shown that bone marrow-derived myeloid cells, such as macrophages, neutrophils, eosinophils, mast cells and dendritic cells, have an important role in regulating the formation and maintenance of blood vessels in tumours. In this Review the evidence for each of these cell types driving tumour angiogenesis is outlined, along with the mechanisms regulating their recruitment and activation by the tumour microenvironment. We also discuss the therapeutic implications of recent findings that specific myeloid cell populations modulate the responses of tumours to agents such as chemotherapy and some anti-angiogenic therapies.
Article
Full-text available
Hypoxia is a feature of most tumours, albeit with variable incidence and severity within a given patient population. It is a negative prognostic and predictive factor owing to its multiple contributions to chemoresistance, radioresistance, angiogenesis, vasculogenesis, invasiveness, metastasis, resistance to cell death, altered metabolism and genomic instability. Given its central role in tumour progression and resistance to therapy, tumour hypoxia might well be considered the best validated target that has yet to be exploited in oncology. However, despite an explosion of information on hypoxia, there are still major questions to be addressed if the long-standing goal of exploiting tumour hypoxia is to be realized. Here, we review the two main approaches, namely bioreductive prodrugs and inhibitors of molecular targets upon which hypoxic cell survival depends. We address the particular challenges and opportunities these overlapping strategies present, and discuss the central importance of emerging diagnostic tools for patient stratification in targeting hypoxia.
Chapter
Macrophages and dendritic cells infiltrate tumours. In the tumour microenvironment, mononuclear phagocytes acquire properties of polarized M2 (or alternatively activated) macrophages. These functionally polarized cells, and similarly oriented or immature dendritic cells present in tumours, play a key role in subversion of adaptive immunity and in inflammatory circuits which promote tumour growth and progression.
Article
Despite having an abundant number of vessels, tumours are usually hypoxic and nutrient-deprived because their vessels malfunction. Such abnormal milieu can fuel disease progression and resistance to treatment. Traditional anti-angiogenesis strategies attempt to reduce the tumour vascular supply, but their success is restricted by insufficient efficacy or development of resistance. Preclinical and initial clinical evidence reveal that normalization of the vascular abnormalities is emerging as a complementary therapeutic paradigm for cancer and other vascular disorders, which affect more than half a billion people worldwide. Here, we discuss the mechanisms, benefits, limitations and possible clinical translation of vessel normalization for cancer and other angiogenic disorders.