Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

REVIEWS

Targeting macrophages:
therapeutic approaches in cancer
Luca Cassetta1 and Jeffrey W. Pollard1,2*
Abstract | Infiltration of macrophages in solid tumours is associated with poor prognosis and
correlates with chemotherapy resistance in most cancers. In mouse models of cancer,
macrophages promote cancer initiation and malignant progression by stimulating angiogenesis,
increasing tumour cell migration, invasion and intravasation and suppressing antitumour
immunity. At metastatic sites, macrophages promote tumour cell extravasation, survival and
subsequent growth. Each of these pro-tumoural activities is promoted by a subpopulation of
macrophages that express canonical markers but have unique transcriptional profiles, which
makes tumour-associated macrophages (TAMs) good targets for anticancer therapy in humans
through either their ablation or their re‑differentiation away from pro-tumoural towards
antitumoural states. In this Review, we evaluate the state of the art of TAM-targeting strategies,
focusing on the limitations and potential side effects of the different therapies such as toxicity,
rebound effects and compensatory mechanisms. We provide an extensive overview of the
different types of therapy used in the clinic and their limitations in light of known macrophage
biology and propose new strategies for targeting TAMs.

The concept that has emerged over the past century However, once tumours progress past this initial state,
suggests that macrophages represent an evolutionarily the immune TME is modified to support the tumour and
ancient, dispersed, homeostatic system, on a par with promote its progression while suppressing any immune
the nervous and endocrine systems … Macrophages cell-mediated cytotoxicity 2. In this process, substantial
are essential for survival and provide an attractive clinical and experimental evidence indicates that macro­
target to manipulate the host, for both immunologic phages — present abundantly in most tumour types
and metabolic purposes. — have a major regulatory role in promoting tumour
Siamon Gordon, The macrophage, past, present, progression to malignancy 3.
future. European Journal of Immunology 37 (Suppl. 1), Tumour-associated macrophages (TAMs), at least
S9–S17 (2007). in mouse models, largely originate from bone marrow
monocytes that are recruited through inflammatory sig-
Metastases cause 90% of cancer deaths worldwide. nals released by cancer cells in the primary and meta­
In most cases, this is because metastatic tumour cells static tumour, where they differentiate into TAMs and
become resistant to therapy and develop methods to facilitate tumour progression4,5. However, in cancers
1
MRC Centre for Reproductive evade immune responses. Resistance to cancer treatment such as gliomas and those of the pancreas, TAMs can
Health, College of Medicine can be intrinsic to the tumour cells, but it is often con- also derive from erythro-myeloid progenitors (EMPs)
and Veterinary Medicine,
Queen’s Medical Research
ferred by non-malignant cells that make up the tumour developed in the yolk sac at the embryonic stage6–8
Institute, The University of microenvironment (TME). The TME is composed of (BOX 1). Nevertheless, in either case, the TME promotes
Edinburgh, Edinburgh, UK. tissue-resident cells and a large proportion of recruited differentiation of these progenitors to new tumour-
2
Department of immune cells that, in certain solid tumours such as associated phenotypes that vary depending on their
Developmental and Molecular
breast cancer, can constitute up to 50% of the tumour location in the tumour but are generally pro-tumoural.
Biology, Albert Einstein
College of Medicine, Bronx, mass. Original theories assumed that these immune Several strategies in immuno-oncology have been
NY, USA. cells were part of the body’s response to reject tumours. developed in the past few years to reactivate the adap-
*e-mail: Indeed, it is still proposed that, at the earliest stage of tive and innate immune systems to mount a robust anti-
jeff.pollard@ed.ac.uk tumour onset, the immune system reacts to the presence tumoural immune response as an alternative approach
doi:10.1038/nrd.2018.169 of cancer by activating T cells and macrophages, which to classic anticancer treatments, to which tumours gen-
Published online 26 Oct 2018 clear the tumour and reduce the incidence of cancer 1. erally develop resistance. Clinical trials with immune

NATURE REVIEWS | DRUG DISCOVERY VOLUME 17 | DECEMBER 2018 | 887


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Box 1 | Macrophage lineage and TAM origin redefined


The understanding of the origins of macrophages has recently undergone a profound shift owing to the use of modern
lineage tracing techniques. These methods use inducible Cre recombinases expressed from cell-specific promoters —
such as the inducible Csf1r‑iCre/Esr1 mouse crossed with floxed, coloured reporter mice — that permanently tag
progeny cells at specific times in development. These methods, together with single-cell and bulk RNA sequencing, have
allowed precise developmental origins of macrophages to be ascertained. A revelation in these studies is that most
tissue-resident macrophages are not derived from bone marrow progenitors, as previously thought, but instead from the
yolk sac or fetal liver270,271. Detailed lineage tracing has indicated that macrophages originate from at least three different
embryonic sources (from erythro-myeloid progenitor (EMP) in the yolk sac and fetal liver, and from macrophage/dendritic
cell progenitor cells (MDPs) in the bone marrow) and differentiate into tissue-specific macrophages according to their
origin, or to other cell types (such as common dendritic cell progenitors (CDP; see the figure) 272,273. Some tissue-specific
macrophages in adults are almost exclusively derived from one source (such as those in the brain, which are derived from
yolk sac, and those in the intestine, which are derived from bone marrow), while in other tissues — such as the skin —
different proportions of macrophages derive from one source or another. In some tissues, such as heart, the yolk sac‑
derived macrophages are replaced from fetal liver monocytes (however, see REFS269,271 for different lineage
interpretations)271,274,275. The presence of persistent embryonic populations throughout life in most tissues suggests that
these tissues harbour pre-macrophages (pMACs) that can proliferate to give rise to mature macrophages275. These data
have also revealed different regulatory mechanisms; for example, bone marrow‑derived cells require transcription factor
cMYB, which is not required for yolk sac‑derived macrophages270. Nevertheless, the dominant transmembrane receptor
controlling the differentiation and survival of almost all macrophages regardless of their origins is the colony-stimulating
factor 1 receptor (CSF1R). In its absence, almost all macrophages are dramatically depleted in mice, with some notable
exceptions such as the lung, in which they are regulated by CSF2 (REF.276). However, it should be recognized that in
addition to this requirement for CSF1R there are tissue-specific growth factors and chemokines as well as
microenvironmental cues that specify macrophage local identity277.
These observations open a number of intriguing questions, for example, about the importance of the phenotype of
macrophages according to their lineage compared with their tissue environment, whether the replacement of yolk
sac-derived or fetal liver-derived macrophages with bone marrow-derived macrophages results in identical phenotypes
and whether macrophages from different origins can be individually targeted. In the context of tumours, these questions
are important because, although in many mouse models most TAMs seem to be of bone marrow origin, there are several
exceptions such as in models of pancreatic cancer and glioma, in which the populations are a mixture of fetal
liver-derived and bone marrow‑derived macrophages 7,8. For example, in pancreatic cancer models, it is the yolk
sac‑derived macrophages but not the bone marrow‑derived macrophages that are pro-tumoural, suggesting that origin
is important57. These different origins might be of clinical relevance, as they could allow independent targeting of
subpopulations6. It also raises questions as to whether inhibition of bone marrow‑derived macrophage recruitment might
result in compensation by yolk sac-derived and/or fetal liver‑derived tissue progenitors or vice versa. Such observations
again emphasize the importance of understanding macrophage origin, heterogeneity and dynamics in the tumour
microenvironment.

Yolk sac Fetal liver Bone marrow

EMP
FLT3L

EMP
MDP CDP Dendritic cell
IL-34 CSF1

CSF1
pMACs FL monocyte
IL-34 CSF1

CSF1
Brain Pancreas Spleen Liver Langerhans • Kidney Intestine Patrolling
microglia cells • Lung
(CSF2) Classical Non-classical
• Heart monocyte monocyte

Tissue macrophages Dendritic cell


F4/80high TAM F4/80low

Figure adapted from REF.183, Springer Nature Limited.

Nature Reviews | Drug Discovery

888 | DECEMBER 2018 | VOLUME 17 www.nature.com/nrd


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

checkpoint inhibitors (such as monoclonal antibodies Although in the late 19th century the scientific
(mAbs) against cytotoxic T lymphocyte antigen 4 community accepted that tumours were a mixture of
(CTLA4), programmed cell death 1 (PD1) and PD1 malignant and normal cells, it was not until the 1970s
ligand (PDL1) that potentiate the activity of cytotoxic that the modern concept of the TME and the idea that
CD8+ T cells) have shown success in the treatment of immune cells can actually promote cancer growth were
cancers such as melanoma and lung cancer. However, in introduced20. However, even at this time, the dominant
most cases, only a small fraction of patients fully respond view was that macrophages were tumoricidal, as differ-
to immunotherapy for unknown reasons9. ent studies showed that activated macrophages could
In mouse models, TAMs promote the recovery of kill tumour cells in vitro21–23. This view began to change
tumours from biologic therapies, chemotherapies and in the early 1990s when several key studies identified a
radiotherapies through a mixture of activities that potential role of macrophages in tumour progression
include promotion of angiogenesis, maintenance of by showing correlations of high intra-tumoural density
stem cells and inhibition of immune responses10. In and increased expression of CSF1 with clinical markers
some tumours, macrophage infiltration also interferes of poor prognosis (grade, stage and so on)24. In 2001,
with efficacy of immunotherapy, neutralizing efforts to the genetic ablation of Csf1, and thereby macrophages,
reactivate CD8+ T cells. For this reason, several thera- in the polyoma middle T (PyMT) model of breast can-
peutic strategies to modulate TAM function, infiltration cer resulted in the inhibition of tumour progression and
or activation are emerging to both block resistance to metastasis, formally proving that macrophages can be
conventional therapies and promote T cell-based thera- pro-tumoural25. After this discovery, several independent
pies3,10,11. This article discusses the mechanism of action studies in different cancers confirmed the pro-tumoural
by which macrophages exert these activities and also the role of TAMs and identified several subtypes of TAM in
manner in which they may be targeted therapeutically. It mice and their roles in promoting primary tumour pro-
describes macrophage diversity and how this promotes gression and metastasis3,26–28. Consistent with these exper-
tumour progression in primary and metastatic sites. imental mouse models, a meta-analysis of 55 studies of
Finally, it also discusses ways to exploit this diversity different human cancers indicated that high infiltration of
therapeutically to create an antitumoural microenvi- TAMs correlated with poor overall survival in breast, gas-
ronment and to improve chemotherapy, radiotherapy tric, oral, ovarian, bladder and thyroid cancers but not in
or immunotherapy. colorectal cancer (CRC)29 (FIG. 1). The correlation between
TAM infiltration and progression in cancer was further
Macrophages in the TME confirmed by additional recent meta-analyses in breast
Macrophages are innate immune cells that populate cancer, gastric cancer, Hodgkin lymphoma and non-
all tissues12 and have multiple roles in development, small-cell lung cancer (NSCLC)30–33. More specifically,
homeostasis and tissue repair 13. They have different overexpression of CSF1 and the urokinase plasminogen
embryological origins (BOX 1) and expand not only in activator (uPA) in CSF1R‑positive ovarian cancer cell lines
response to infiltration of monocytes but also through improved their invasive properties34. At the clinical level,
local proliferation of tissue-resident macrophage pro- high serum levels of CSF1 are found in several cancers
genitors. Although macrophages express canonical including metastatic breast cancer 35, and they correlate
markers such as colony-stimulating factor 1 receptor with poor prognosis in ovarian and endometrial can-
(CSF1R) in humans and mice, and adhesion G pro- cers36–38. In endometrial cancer, epithelial CSF1 synthesis
tein-coupled receptor (ADGRE; also known as F4/80)14 is an independent predictor of survival39. In breast can-
only in mice, studies indicate considerable transcrip- cer, a CSF1 expression signature was described that pre-
tomic diversity between macrophage populations even dicted poor survival40. CSF1R expression combined with
within a tissue. This diversity is consistent with their CD68‑positive macrophage infiltration is considered an
diverse origin, their adaptation to different tissue niches independent predictor of short progression-free survival
and their involvement in different pathologies. CSF1R in Hodgkin lymphoma41. Normal breast and normal ovar-
is a transmembrane tyrosine kinase class III receptor ian tissues do not express CSF1R and CSF1 (REFS35,42) but,
that is required for the presence of the vast majority sometimes, breast and ovarian tumour cells can express
of macrophages15. It binds to two ligands, CSF1 and this receptor–ligand pair 43–45. Nevertheless, expression of
interleukin‑34 (IL‑34), and regulates macrophage dif- CSF1R on cancer cells is rare and is probably due to acti-
ferentiation, proliferation and survival in humans and vation as a result of demethylation of a recently acquired
mice16. IL‑34 shows overlapping functions with, but retroviral element in humans; thus, it is not found in
no sequence similarity to, CSF1 and binds to a differ- mice46. These data over the past 18 years, in many differ-
ent binding site on CSF1R with a significantly higher ent cancer contexts, have supported the conclusion that,
affinity than CSF1 (REF.17). IL‑34 regulates the develop- in general, TAMs promote tumour progression to malig-
ment of a subset of macrophages, particularly micro- nancy through their interactions with cancer cells (FIG. 1).
glia and Langerhans cells18. The role of IL‑34 in cancer
progression is still largely unknown, although a recent TAMs in solid tumours
study by Baghdadi et al.19 showed that IL‑34 produced Cancer initiation
by chemoresistant cancer cells was able to sustain the The activation of key transcriptional factors such as
immunosuppressive functions of TAMs and promote nuclear factor-κB (NF-κB), signal transducer and acti-
the survival of cancer cells. vator of transcription 3 (STAT3) and hypoxia-inducible

NATURE REVIEWS | DRUG DISCOVERY VOLUME 17 | DECEMBER 2018 | 889


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a Percentage of survival b Immune infiltrates in tumour


Bladder
Bladder
Type of cancer

Breast
Bowel
Breast
Stomach

Type of cancer
Lung
Bowel

0 20 40 60 80 100
Percentage (%) Stomach
Localized Invasive Metastasized
Lung

B cells CD8+ γδ T Macrophages Mast


c Macrophage density as a predictor of poor survival T cells cells cells
+
Plasma CD4 NK Dendritic
Macrophage density cells T cells cells cells
Patient survival Polymorphonuclear
cells

Figure 1 | Macrophage infiltration and survival in cancer. a | Five-year survival values are high if cancer is restricted to
the primary site (localized), even if it is invasive. However, survival is dramatically reduced if the cancer has metastasized to
Naturethat
distant sites. Data are expressed as 5‑year survival values (expressed in percentages) in tumours Reviews | Drug Discovery
are localized (blue
dots), invasive (grey dots) or metastasized (red dots). b | CIBERSORT analysis of tissue microarray data sets of solid human
tumours revealed the average immune cell composition of bladder, breast, bowel, stomach and lung cancers. Data are
expressed as estimated fractions of leukocyte RNA. As shown, immune infiltrates vary according to cancer type; however,
macrophages represent the major population infiltrating most human cancers. c | Increased human macrophage density
(red wedge) correlates with markers of poor survival (grey wedge) in most cancer types. NK, natural killer. Part a
republished with permission of The Economist, from Targeting tumours, Loder, N., 2017; permission conveyed through
Copyright Clearance Center, Inc. (REF.278). Part b is adapted from REF.279, Springer Nature Limited.

factor 1α (HIF1α) by chronic inflammation (caused by cancers)6,25,56 and, at least in mammary tumours, pre-
persistent infection, exposure to irritants or autoimmune mature recruitment of macrophages by overexpression
disease) or by oncogene activation results in the pro- of CSF1 promotes the transition to malignancy 25. It is
duction of cytokines and chemokines that engage the thought that in the first stages of tumour formation mac-
innate immune system and especially macrophages47. rophages are mainly tumoricidal, as they reflect an acti-
Macrophages, in turn, can contribute to cancer initia- vated state, although the evidence for this conclusion is
tion by producing pro-inflammatory mediators such limited. What is certain, however, is that, as the tumour
as IL‑6, tumour necrosis factor (TNF) and interferon-γ grows, T helper 2 (TH2) cells in the microenvironment
(IFNγ), growth factors including epidermal growth factor that ‘educate’ macrophages to become pro-tumoural and
(EGF) and WNTs, proteases and reactive oxygen species bias the immune response from a cytotoxic to a support-
(ROS) and nitrogen species that may create a mutagenic ive role begin to dominate. In mouse models of breast
microenvironment 48,49. Thus, chronic inflammation, cancer, this state is driven by IL‑4 secreted from CD4+
infection or irritation is associated with the initiation of T cells57,58 recruited to the tumour through unknown
many cancer types such as those in the colon or stom- mechanisms.
ach50. Genetic ablation of Stat3 in macrophages causes The transition to malignancy is also exacerbated by
exuberant inflammation and the formation of invasive macrophages through their production of angiogenic
tumours in the colon in a mouse model of inflammatory factors, secretion of growth factors and production of
bowel disease51. Similarly, the deletion of the anti-inflam- proteases.
matory cytokine IL‑10, which signals through STAT3, in Tumour progression requires the creation of a vascu-
macrophages is also associated with tumour initiation and lar network for oxygenation, nutrition and waste disposal.
promotion52,53. By contrast, removal of ROS production in This process, known as the ‘angiogenic switch’, is mainly
myeloid cells, including macrophages, inhibits carcino- characterized by a dramatic increase in new vessel forma-
genesis in an intestinal cancer model49. These chronic tion that often involves vessel dilatation and recruitment
inflammatory responses are involved with complicated of perivascular cells59–61. TAMs support these processes by
interactions with the microbiome, changes in dominance the production of pro-angiogenic factors such as vascular
of bacterial species and breakdown of mucosal barriers endothelial growth factor A (VEGFA)62 and angiogenic
that allow infiltration of pathogenic types54. Thus, in ani- CXC chemokines (CXCL8 and CXCL12)63. Additional
mal models, in some cases, the initiation of inflamma- factors such as WNT7B, transforming growth factor-β
tion-induced cancer can be suppressed by prophylactic (TGFβ), TNF and thymidine phosphorylase contribute
treatment with broad-spectrum antibiotics55. to the angiogenic process by recruitment and activation
The role of macrophages in the transition from a of endothelial cells or other cells such as fibroblasts or
benign to a malignant tumour has been studied in only pericytes that further support the generation of vascular
a few cancer models (mammary, skin and pancreatic networks in the microenvironment61,64. The identification

890 | DECEMBER 2018 | VOLUME 17 www.nature.com/nrd


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

of a subpopulation of TAMs that are characterized by which are able to negatively modulate the activation
the expression of the angiopoietin 1 receptor (TIE2; also of NK cells and T cells by interaction with CD94 and
known as TEK) and that lie in close association with ves- leukocyte immunoglobulin-like receptor subfamily B
sels confirmed the central role of TAMs in tumour angio­ member 1 (LIR1, also know as ILT2), respectively 74.
genesis65, as their depletion inhibited tumour growth and TAMs express T cell immune checkpoint ligands, such
metastasis66. as PDL1, PDL2, B7‑1 (also known as CD80) and B7‑2
Highly invasive tumour cells are able to move from (also known as CD86), which directly inhibit T cell
the primary tumour and intravasate into the blood or functions75,76, while also secreting several cytokines,
lymphatic vessels, where they migrate to distant sites such as IL‑10 and TGFβ, that contribute to the main-
to establish micro-metastases 28. TAMs are in part tenance of a strong immunosuppressive microenvi-
responsible for the creation of this invasive TME. TAMs ronment by inhibiting CD4+ (TH1 cells and TH2 cells)
directly help tumour cells to migrate via a paracrine loop and CD8+ T cells and by inducing regulatory T (Treg)
between macrophages and tumour cells that involves cell expansion. TAM-mediated release of chemokines
secretion of EGF family ligands from macrophages such as CCL2, CCL3, CCL4, CCL5 and CCL20 fur-
and CSF1 from tumour cells, which improves tumour ther contributes to the recruitment of Treg cells in the
cell invasive properties. TAMs also produce cathepsins TME3. TAMs also directly inhibit T cell cytotoxicity
and matrix-remodelling enzymes that stimulate this pro- through depletion of l‑arginine (which is essential for
cess and increase intravasation by the upregulation of the re‑expression of the T cell receptor (TCR) after anti-
metalloproteinases such as matrix metalloproteinase 9 gen engagement on T cells) via the release of arginase 1,
(MMP9)67–69. Tumour cells are attracted to vessels, where which metabolizes l‑arginine to urea and l‑ornithine3.
they engage with perivascular TIE2+ macrophages that Similarly, depletion of tryptophan or production of tryp-
act as a conduit for the escape of tumour cells in part tophan metabolites by indoleamine 2,3‑dioxygenase
through expression of VEGFA, which allows bursts of (IDO) expressed by macrophages can inhibit cytotoxic
vessel permeability 70. This tripartite structure consist- T cells77,78.
ing of perivascular TAMs, cancer cells that have height-
ened motility owing to the expression of the invasive TAMs in metastasis
isoform of the actin-binding protein mammalian ena- To establish metastasis, invasive cancer cells need to
bled (MENA) and endothelial cells has been called the avoid eradication by the immune system, survive in
tumour microenvironment for metastasis (TMEM). the blood or lymphatic circulation, arrest at a distant
Similar structures identified in clinical samples led to the site and survive and grow in these often hostile envi-
identification of a prognostic clinical score that predicts ronments. In mouse models of breast cancer and CRC,
the chance of metastasis in breast cancer regardless of metastasis-associated macrophages (MAMs) promote
tumour stage or clinical subtype71. these latter steps by increasing extravasation, sending
Transcriptional profiling of invasive TAMs also survival and growth signals to tumour cells and inhib-
revealed upregulation of key components of the WNT iting anti-cytotoxic T cells5,79,80,81. Importantly, ablation
and Hedgehog gene families, which are mainly involved of these MAMs or blockade of their recruitment results
in tissue patterning and development in homeostatic in inhibition of metastasis and in some cases prolonged
conditions72,73. The importance of WNT signalling was survival of mice5,79,82, suggesting that they represent
confirmed by the identification of WNT7B as one of thera­peutic targets.
the key factors secreted by TAMs to promote tumour Mechanistically, metastatic cells attract bone marrow-
progression and metastasis, in part through effects on derived classical monocytes (that express lymphocyte
angiogenesis but also through promotion of tumour cell antigen 6C2 (LY6C)) in mice and CD14highCD16negative
invasion61. in humans) through a CCL2–CCR2 mechanism5. Once
extravasated, these monocytes (called MAM precursor
TAM-mediated immune suppression cells (MAMPCs)) differentiate into MAMs79 through
Macrophages can potentially mount a robust anti­ engagement of a chemokine cascade that involves
tumoural response, as they are able to directly eliminate CCR1–CCL3 autocrine signalling 80. MAMs found
cancer cells if properly activated by IFNγ. They also in the lungs of mice with metastatic disease are tran-
can support the adaptive immune response through scriptionally different from resident macrophages and
presentation of tumour antigens and the production of MAMPCs83 and are characterized by the expression of
chemokines and cytokines to recruit and activate cyto- the markers CD11b, VEGF receptor 1 (VEGFR1; also
toxic CD8+ T cells and natural killer (NK) cells. However, known as FLT1), CXCR3 and CCR2 (REFS79,80). Moreover,
these macrophage activities are restricted by the TH2 cell lung intravital and ex vivo experiments showed that
dominance in the TME, which profoundly affects mac- macrophages in the lung interact physically with meta­
rophage functions such that their phenotypes resemble static cells84 and support their survival through a vas-
those involved in tissue development and repair, with cular cell adhesion protein 1 (VCAM1)-dependent
a consequent suppression of antitumoural response3,13. and AKT-dependent mechanism as well as by induc-
TAM-induced immune suppression is mediated ing epithelial to mesenchymal transition by producing
by the expression of inhibitory receptors, including TGFβ85,86. Moreover, MAMs and metastatic cancer cells
non-classical major histocompatibility complex (MHC) engage in a crosstalk that improves MAM retention
class I (MHC‑I) molecules such as HLA‑E and HLA‑G, in the metastatic foci, which further supports tumour

NATURE REVIEWS | DRUG DISCOVERY VOLUME 17 | DECEMBER 2018 | 891


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

• VEGFR1+
• CCR2+
Metastasis • CXCR4–
• TIE2–
• Arginase+
• MARCO+
TAM • IL-10+
• Wnt signaling • CCL22+ Immune
Invasion • CTSS+ • MRC1+ regulation
and/or • CTSB+ • MHC-IIhigh
intravasation • EGF+ • CD11b+
• CSF1R+
• F4/80+ • IL-12
• MHC-IIhigh Inflammation
• iNOS+
• CXCR4 + • TNF+
Resistance • B7.1 and/or
to therapy • MRC1+
• TIE2+ B7.2

• VEGFR1+
• VEGF+
• TIE2+ Angiogenesis
• CXCR4+
Stem cell • CTSB+
maintenance WNT proteins • CTSS+

Figure 2 | Macrophage diversity drives tumour progression to metastasis and resistance to therapy.
Tumour-associated macrophages (TAMs) that express canonical macrophage markers in the Nature Reviews
mouse (CD11b,| Drug Discovery
colony-stimulating factor 1 receptor (CSF1R) and adhesion G protein-coupled receptor (ADGRE; also known as F4/80))
can be polarized to adopt different pro-tumoural functions depending on the environment, as indicated. These activities
promote tumour initiation through inflammation, tumour progression to malignancy via increasing angiogenesis,
immunosuppression, invasion, intravasation and at distant sites tumour cell extravasation and persistent growth. Each of
these functions is performed by a subpopulation of macrophages with a different transcriptome and cell surface markers,
as indicated. EGF, epidermal growth factor; MARCO, macrophage receptor with collagenous structure; MHC-II, major
histocompatibility complex class II; TIE2, angiopoietin 1 receptor; TNF, tumour necrosis factor; VEGF, vascular endothelial
growth factor; VEGFR1, vascular endothelial growth factor receptor 1.

growth through expression of unidentified survival and TAMs in conventional therapies


growth factors80. MAMs and their precursors inhibit Radiotherapy. Ionizing radiation is designed to specifically
cytotoxic T cells, suggesting that they are also involved in target cancer cells by inducing DNA damage in cells that
the maintenance of an immunosuppressive environment usually have impaired DNA repair mechanisms. However,
that further promotes metastasis83. Similar mechanisms the effect of radiotherapy on the TME, and especially on
have been observed in bone metastasis, in which another TAMs, is only partially understood88. After ablative radio-
class of macrophages — the bone-degrading osteoclasts therapy (single dose of 10 Gy), the innate immune system
— are activated by metastatic cells to engage a vicious is activated by inflammatory cytokines, such as IL‑1 and
cycle because the bone resorption liberates entrapped TNF, and pro-fibrotic factors such as TGFβ that recruit
growth factors that further promote metastatic growth87. macrophages with a tissue-reparative phenotype and con-
There are currently limited data available on tumoural tribute to tumour recurrence and progression89–93. A recent
infiltration in human metastasis, although MAMs that study by Pinto et al. showed that fractionated cumulative
express high levels of VEGFR1 have been identified in radiation dose regimens similar to those used during can-
lymph node metastases82. cer treatment induced a pro-inflammatory phenotype
in macrophages in vitro but did not alter their ability to
Targeting TAMs in cancer therapy promote cancer invasion and cancer angiogenesis94. It is
The subpopulations of macrophages with identifia- becoming a priority to tailor dose and fractionation of
ble markers, transcriptomes and phenotypes (FIG. 2) radiotherapy in different cancers, as the response elicited
described above are therefore attractive therapeutic by macrophages and the stroma in general can influence
targets for combination therapies including standard of the outcome95. Consequently, macrophage targeting in
care and immunotherapy. In mouse models and clin- combination with radiotherapy is a potential therapeutic
ical contexts, TAMs can synergize with the anticancer strategy to modulate the stroma and allow better tumour
therapy or, alternatively, induce pro-tumoural functions killing but, as yet, there are no clinical trials reported.
by the activation of tissue repair mechanisms. Here, we
describe the latest studies reporting dichotomous behav- Chemotherapy. The TME and, in particular, macrophages
iours of TAMs after standard therapy, and strategies to play an important part in chemotherapy response and
improve anti-macrophage therapeutics (FIGS. 3,4). resistance96–98. The first observation that suggested a

892 | DECEMBER 2018 | VOLUME 17 www.nature.com/nrd


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

role of TAMs in mediating resistance to chemotherapy and myeloid-derived suppressor cells) and that they
was the demonstration that CSF1 inhibition was able are exposed to an immunosuppressive cytokine milieu
to reverse chemoresistance of breast cancer cell lines in (IL‑10 and TGFβ).
xenograft mouse models99. DeNardo100 et al. confirmed With this increasing understanding, the immuno-
and extended this initial observation and showed that therapy field has had a rapid expansion particularly with
tumour biopsy samples from cancer patients who the discovery of immune checkpoint inhibitors, such as
received neoadjuvant therapy had a much larger infil- mAbs against CTLA4 and PD1, whose main function is
trate of CD45+CD11b+CD14+ macrophages than those to remove the ‘brakes’ on T cells, allowing an effective
from patients who received only surgery. They also antitumoural immune response106. One anti‑CTLA4
demonstrated that treatment of PyMT mice with pacl- (ipilimumab) and two anti‑PD1 (pembrolizumab and
itaxel in combination with anti‑CSF1R antibodies signif- nivolumab) neutralizing antibodies are currently US
icantly reduced tumour burden and vessel density and Food and Drug Administration-approved for the treat-
increased cytotoxic T cell infiltration compared with ment of several cancers including melanoma, advanced
treatment with paclitaxel alone. renal carcinoma, gastric cancer, NSCLC and CRC;
TAMs are responsible for the increased produc- hundreds of clinical trials on multiple solid cancers are
tion of cathepsins that leads to increased lymphangio- ongoing to evaluate their efficacy. Although efficacy in
genesis and metastasis after paclitaxel treatment 101,102. melanoma and some other cancers is high, especially
Upon 5‑fluorouracil treatment of CRC, TAMs release when anti‑CTLA4 and anti‑PD1 therapies are combined,
the diamine putrescine, which confers on cancer cells in many other cancers, only a small fraction of treated
resistance to apoptosis in a JNK-dependent and caspase patients respond. One hypothesis is that the efficacy of
3‑dependent manner 103. Doxorubicin, another chemo- checkpoint inhibitors could be improved by the modu-
therapeutic agent, can also induce the selective accumu- lation of immunosuppressive cells such as TAMs. TAMs
lation of perivascular MRC1+ TAMs around the blood express high levels of PDL1 and PDL2, as well as PD1.
vessels. These perivascular TAMs promoted recurrence PD1 expression in TAMs also negatively correlates with
after therapy partly through the expression of VEGF and the ability of these cells to phagocytose cancer cells, and
subsequent increased angiogenesis. Selective targeting of therefore, TAM-specific PD1 inhibition reduces tumour
the recruitment of this perivascular TAM population by growth107.
CXCR4 blockade showed reduced tumour relapse after Moreover, CSF1 expression correlates with accumula-
chemotherapy, suggesting that this targeting strategy is tion of CD8+ T cells and CD163+ TAMs in melanoma, and
of clinical utility 63. anti‑PD1 and anti‑CSF1R combination therapy induced
regression of melanoma in preclinical studies108. Clinical
Immunotherapy. Mutated tumour cells can express trials combining checkpoint inhibitors and anti-TAM
tumour antigens as a product of oncogenic viruses, agents (such anti‑CSF1R antibodies; see below) are cur-
differentiation antigens or tumour-specific mutations rently ongoing in different solid tumour contexts109–119.
(tumour neo-antigens). The production of neo-antigens
is not equal across tumour types, with tumours such Methods of TAM targeting
as melanoma and lung cancers expressing the highest TAM depletion. The dependence of macrophages on
rate of neo-antigens, and haematological malignancies CSF1R signalling makes this an attractive target to selec-
(acute myeloid leukaemia (AML), acute lymphoblas- tively deplete macrophages. Consequently, different anti-
tic leukaemia and chronic myelogenous leukaemia) bodies and small molecules mainly targeting CSF1R are
expressing the lowest 104. T cells are able to recognize being studied in different clinical trials both as mono­
tumour antigens loaded to the MHC on the cancer therapies or in combination with standard therapy or
cell through binding to the TCR; however, to get com- immunotherapies.
pletely activated, they require interaction of CD28 The small molecules under clinical development and
with co‑stimulatory B7 molecules (CD80 and CD86) study are PLX3397, JNJ‑40346527, PLX7486, ARRY‑382
expressed on the antigen-presenting cell (APC). Cancer and BLZ945. Preclinical studies showed that PLX3397, or
cells do not express B7 molecules, and so, without a sec- pexidartinib, reduced the number of tumour-associated
ond stimulatory signal provided by other APCs (such as microglia and glioblastoma invasion120,121; in glioma
dendritic cells and macrophages) recruited by inflam- mouse models, the tyrosine kinase inhibitors dovitinib
matory signals, the antitumoural T cell response will and vatalanib increased sensitivity to PLX3397. PLX3397
not start. was tested in a phase I dose-escalation clinical trial fol-
The identification of tumour antigens led to the lowed by a phase II extension study in patients with
development of several tumour vaccination strategies in advanced tenosynovial giant cell tumours122, a tumour
the 1980s, in which tumour-derived antigens (DNA or type characterized by the high expression of CSF1 and
peptides) were injected together with cytokines in order CSF1R123,124. Results from the phase I and phase II stud-
to enhance the immunological response. Results from ies indicated that PLX3397 was tolerated at a dose of
these trials, however, were not as striking as expected105, 1,000 mg, and in the extension study, 12 out of 23 patients
suggesting that the regulation of T cell activation in the (52%) showed antitumour responses after treatment122.
tumour is complex. Both preclinical and clinical stud- In a phase II study in patients with recurrent glioblas-
ies indicated that tumours are infiltrated by immuno- toma, PLX3397 treatment was tolerated, and PLX3397 was
suppressive cells (Treg cells, TAMs, cancer fibroblasts able to pass the blood–tumour barrier; however, PLX3397

NATURE REVIEWS | DRUG DISCOVERY VOLUME 17 | DECEMBER 2018 | 893


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

1 • Metastasis 2
Trabectedin Phagocytosis
• Dissemination
Blood 6 • Angiogenesis
vessel Monocyte
recruitment Anti-VEGFR SIRP1α inhibitors

• Anti-CCR2 TRAIL • Anti-MUC1 Anti-CD47


Monocyte VEGF
• Anti-CSF1R • Anti-EGFR
SIRPα
Bisphosphonates
CD47
CCR2
Caspase 8
CSF1R
DICER
Survival Apoptosis DICER
CCL2 inhibitor
Tumour 5ʹ
cells Anti-CCL2 3ʹ
Pre-miRNA

CSF1 TLR8 TLR7


Anti-CSF1 TLR9

3 TLR agonists
IL-10 Checkpoint and
B7-1 arginase inhibitors
and/or
B7-2 PDL1
and/or FcγRIIB MARCO
Dendritic cell TGFβ PDL2
PI3Kγ CD40

CD28 HDAC
PD1 inhibitors
CD40
T cell or agonists
NK cell Arginase PI3Kγ
inhibitors Anti-MARCO

Treg cell
4
5
Immune suppression Reprogramming

Pro-tumour macrophage Anti-tumour macrophage

Figure 3 | Targeting and reprogramming TAM pro-tumoural activities. killing or by T cell activation; (5) inhibiting the tumour suppressive
The figure shows pro-tumoural tumour-associated macrophage (TAM) microenvironment; and (6) inhibitingNature Reviews
recruitment Drugmonocytic
of| the Discovery
activities (angiogenesis, immune escape, dissemination, and so on) progenitors of TAMs; CSF1, colony-stimulating factor 1; CSF1R, CSF1
that have been targeted in either preclinical models or therapeutic trials receptor; EGFR, epidermal growth factor receptor; HDAC, histone
in humans. The strategies fall into six main groups: (1) blocking deacetylase; MARCO, macrophage receptor with collagenous structure;
pro-tumoural functions such as those that promote intravasation, miRNA, microRNA; NK, natural killer; PD1, programmed cell death 1;
angiogenesis or metastatic cell extravasation and/or persistent growth; PDL1, PD1 ligand 1; PI3Kγ, phosphoinositide 3‑kinase-γ; SIRPα, signal
(2) promoting phagocytosis of tumour cells by macrophages; (3) using regulatory protein-α; TGFβ, transforming growth factor-β; TLR, Toll-like
checkpoint or anti-immunosuppressive cytokine and/or protein receptor; TRAIL, tumour necrosis factor-related apoptosis-inducing
inhibitors to allow cytotoxic T cell activity; (4) reprogramming TAMs to ligand; Treg cell, regulatory T cell; VEGF, vascular endothelial growth
become antitumour macrophages either directly through tumour cell factor; VEGFR, VEGF receptor.

treatment did not show improvement in 6‑month pro- with activating mutations in brain-derived neurotrophic
gression-free survival values when compared with those factor (BDNF))127, and ARRY‑382 is currently involved in
of the group receiving radiotherapy and temozolomide125. two phase I clinical trials128,111 in patients with metastatic
JNJ‑40346527, another kinase inhibitor, was tested in a disease and advanced-stage solid tumours. BLZ945 was
phase I/II study on 21 patients with relapsed or refrac- reported to alter macrophage polarization and to block
tory Hodgkin lymphoma. One patient showed complete glioma progression129 with promising results when used
remission, and eleven patients showed stable disease126. in combination with inhibitors of insulin-like growth
Several phase I clinical trials are ongoing with additional factor 1 receptor (IGF1R) and phosphoinositide 3‑kinase
CSF1R inhibitors: PLX7486 is being tested as a single (PI3K)130 and is currently under assessment in a trial in
agent in patients with advanced-stage solid tumours (ten- advanced-stage solid tumours as a single agent or in com-
osynovial giant cell tumour and tumours of any histology bination with the anti‑PD1 antibody PDR001 (REF.112).

894 | DECEMBER 2018 | VOLUME 17 www.nature.com/nrd


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

There are three anti‑CSF1R mAbs under clinical eval- A more directed approach when using bisphospho-
uation, RG7155, IMC‑CS4 and FPA008. RG7155 (emac- nates is to encapsulate clodronate, for example, in lipo­
tuzumab) is a humanized mAb that binds to CSF1R and somes (clodrolip), which are preferentially taken up by
blocks its dimerization. Preclinical studies showed that macrophages owing to their phagocytic activities. This
RG7155 depletes CSF1R+CD163+ macrophages in vitro treatment reduced macrophage tumour infiltration
and in vivo. In mouse models of CRC and fibrosarcoma, in experimental models of bone metastasis from lung
treatment with a chimeric version of RG7155 reduced cancer 142 and lung metastasis from breast cancer 79 and
the number of infiltrating TAMs and increased the thereby limited metastatic outgrowth. Similar results
CD8+:CD4+ T cell ratio124; moreover, RG7155 showed sig- were obtained in mice injected with human melanoma
nificant reduction of CSF1R+CD163+ macrophages and cells, in which clodrolip reduced tumour mass and angio­
T cell TME composition in patients with advanced-stage genesis143. Clodrolip in combination with anti-VEGF
diffuse-type giant cell tumours treated with RG7155 as mAbs showed antitumour properties in mice injected
monotherapy or in combination with paclitaxel124. These with teratocarcinoma and rhabdomyosarcoma cells, with
promising results led to a dose-escalation phase I clinical a significant reduction of TAM infiltration144. Moreover,
trial in 12 patients with tenosynovial giant cell tumours to in mouse models of metastatic hepatocellular carcinoma,
investigate the clinical benefit of RG7155; the compound the combination treatment with clodrolip and sorafenib
showed no dose toxicity, and common adverse effects caused decreased tumour burden, angiogenesis and
reported were facial oedema, asthenia and pruritus. In metastasis145. Clodrolip was also tested in dogs with
the dose-expansion phase, out of the 28 patients tested, spontaneous soft tissue sarcomas, in which it depleted
24 patients (86%) showed an objective response, and CD11b+ macrophages in tumours and decreased IL‑8
2 (7%) achieved a complete response131. There are two serum levels, although the antitumour properties of
other antibodies currently under phase I clinical trials: the treatment were not significant 146. Zoledronic acid,
FPA008 — currently under clinical investigation in three another bisphosphonate, reduced tumour burden in a
ongoing clinical trials116,132,133 in diffuse-type tenosynovial mouse model of bone metastases from breast cancer 147
giant cell tumour and advanced-stage solid tumours — and to modulate the TME by reducing the number of
and IMC‑CS4, which is also under clinical investigation TAMs and their polarization status148. Recently, Comito
in three clinical trials134–136 on solid tumours including et al.149 demonstrated that treatment with zoledronic
pancreatic, prostate and breast cancers. acid impairs macrophage polarization, reduces macro­
Overall, these preliminary results suggest that target- phage-induced angiogenesis and decreases tumour
ing the CSF1–CSF1R axis could be a promising strategy. invasion in prostate cancer. Current studies using
Indeed, in tumours overexpressing CSF1, such as the nanotechnology are trying to optimize the delivery of
synovial giant cell tumours, it is likely that this approach bisphosphonates by their encapsulation in stealth lipo­
will be advanced as an effective treatment. Nevertheless, somes or in PEGylated nanoparticles; preclinical tests were
reported toxicity has limited dose escalation, as it is promising, showing better antitumoural activity and
problematic to deplete all macrophages from the body lower TAM numbers than those seen with treatment
for a long period of time. with free bisphosphonates150–152.
Another therapeutic strategy is to selectively deplete At the clinical level, clodronate and zoledronic acid
TAMs and not the other components of the stroma. An were tested in several clinical trials on a variety of dif-
example of this strategy is the use of bisphosphonates. ferent cancers with inconsistent results that suggest the
These inorganic compounds are stable, and their struc- need to optimize better combination treatments and
ture is identical to pyrophosphatases of the bone matrix, for longer clinical trials, as discussed in several meta-
so they can be metabolized rapidly by osteoclasts and analyses153,139,154. There are currently two ongoing clin-
inhibit their resorption. Moreover, they are already in ical trials evaluating the effect of zoledronic acid in
use as anticancer agents for the treatment of haemato- triple-negative breast cancer 155 and stage IIIb and IV
logical and solid malignancies137,138. Bisphosphonates lung cancer 156. Clodronate is also being tested in differ-
are mainly subdivided into two classes on the basis of ent clinical trials as a neoadjuvant agent in patients with
their structure and mechanism of action: clodronate, breast cancer 157 and in combination with chemother-
etidronate and tiludronate belong to the first group, and apy and hormonal therapy 158; additional trials include
alendronate, ibandronate, pamidronate, risedronate and a clodronate–chemotherapy combination treatment in
zolenodrate belong to the second group. At the preclin- patients with metastatic refractory prostate cancer 159.
ical level, bisphosphonates exhibited direct and indirect Trabectedin is a tetrahydroisoquinoline alkaloid
antitumour properties. They are reported to inhibit can- that was initially isolated from the Caribbean tuni-
cer cell proliferation, induce tumour cell apoptosis, block cate Ecteinascidia turbinata160. It is an anti-neoplastic
angiogenesis, inhibit cell adhesion and invasion and drug approved in Europe, Russia and South Korea for
PEGylated nanoparticles interfere with immune surveillance through activation the treatment of advanced-stage tissue sarcoma and plat-
A type of therapeutic delivery of γδ T cells139. Different studies showed that bisphos- inum-sensitive relapsed ovarian cancer in combination
system that results from phonates are also able to inhibit proliferation, migration with PEGylated liposomal doxorubicin161–163. Trabectedin,
PEGylation (a chemical and invasion of macrophages, causing apoptosis140,141. in addition to targeting tumour cells, specifically induces
modification that involves the
conjugation of polyethylene
They, however, mainly affect osteoclasts that share the apoptosis of monocytes and macrophages in the tumour
glycol (PEG) to different same lineage with macrophages and thus have been used by the activation of caspase 8 through a TNF-related
molecules) of nanoparticles. in preclinical bone metastasis models. apoptosis-inducing ligand (TRAIL; also known as

NATURE REVIEWS | DRUG DISCOVERY VOLUME 17 | DECEMBER 2018 | 895


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Recruitment Survival Reprogramming

Monocyte Cancer cell TAM TAM Apoptosis Pro-tumoural Tumoricidal


macrophage macrophage
Drug name Target Inhibitor type Drug name Target Inhibitor type Drug name Target Inhibitor type
Carlumab CCL2 mAb Clodronate NA Small molecule Hu5F9-G4 CD47 mAb
PF04136309 CCR2 Small molecule Zoledronic acid NA Small molecule CC-90002 CD47 mAb
PLX3397 CSF1R Small molecule Trabectedin Caspase 8 Small molecule TTI-621 SIRPα Fusion protein
PLX7486 CSF1R Small molecule PLX7486 CSF1R Small molecule CP-870,893 CD40 Agonistic antibody
JNJ-40346527 CSF1R Small molecule JNJ-40346527 CSF1R Small molecule RO7009789 CD40 Agonistic antibody
ARRY-382 CSF1R Small molecule ARRY-382 CSF1R Small molecule Imiquimod TLR7 Small molecule
BLZ945 CSF1R Small molecule BLZ945 CSF1R Small molecule 852A TLR7 Small molecule
IMC-CS4 CSF1R mAb IMC-CS4 CSF1R mAb IMO-2055 TLR9 Small molecule
R05509554 CSF1R mAb R05509554 CSF1R mAb BLZ945 CSF1R Small molecule
RG7155 CSF1R mAb RG7155 CSF1R mAb
FPA008 CSF1R mAb FPA008 CSF1R mAb

Figure 4 | Selective examples of anti-TAM drugs currently under clinical trial investigation. For each of the strategies
targeting tumour-associated macrophages (TAMs) (recruitment, survival and reprogramming),
Nature there are drugs
Reviews currently
| Drug Discovery
being tested in clinical trials as monotherapies or in combination with chemotherapy and immunotherapy. In many cases,
one anti-TAM drug can affect more than one process, such as colony-stimulating factor 1 receptor (CSF1R) inhibitors that
inhibit recruitment, survival and function. mAb, monoclonal antibody; NA, not applicable; TLR, Toll-like receptor.

TNFSF10)-dependent mechanism164. These results sug- CCL2 in serum, as well in the tumour, are associated
gested that the apoptotic receptor family TRAIL could with poor prognosis in different types of tumours,
be a therapeutic target to selectively kill immune cells such as breast cancer 175,176. For these reasons, several
and, especially, macrophages. A recent report by Liguori CCL2‑neutralizing antibodies are now being tested
et al.165 explored this hypothesis and demonstrated that in clinical trials. The two main drugs currently tested
monocytes and macrophages express the functional are carlumab (CNTO 888), an anti‑CCL2 mAb, and
TRAIL receptors TRAILR1 (also known as TNFSF10A) PF‑04136309, a small molecule inhibitor that targets
and TRAILR2 (also known as TNFRSF10B), whereas CCR2.
neutrophils and lymphocytes express the non-functional Carlumab is a human immunoglobin G1κ antibody
decoy receptor TRAILR3 (also known as TNFRSF10C). that binds to CCL2. In prostate cancer mouse models,
Interestingly, human TAMs in mammary, hepatic and systemic injection of the antibody reduced tumour
colon carcinoma, but not resident tissue macrophages, growth, infiltration of CD68+ macrophages and vascular
express functional TRAILRs165, making these receptors density 177,178. CCL2 inhibition was also able to improve
interesting targets for therapy 166. the effect of paclitaxel and carboplatin therapies in
mouse models of ovarian cancer 179. A phase I clinical
Inhibition of TAM recruitment. TAM expansion in trial was carried out in 2013 to assess tolerance to differ-
the tumour is often mediated by monocytic recruit- ent doses of carlumab in 44 patients with different solid
ment through the CCL2–CCR2 axis. CCL2 is a potent tumours180. The results indicated that CCL2 levels were
chemo­attractant for monocytes, T cells and NK cells167, only partially suppressed by carlumab, with an increase of
and several mouse studies have demonstrated a role for free CCL2 after treatment over the pretreatment baseline
it and other chemokines in macrophage accumulation of more than 1,000‑fold173. A phase II clinical trial was
in the tumours168–171. CCL2 released by tumour cells then carried out in 46 patients with castration-resistant
recruits classical monocytes that express the receptor metastatic prostate cancer, with no reported therapeutic
CCR2 to the tumour sites, and inhibition of CCL2 cor- efficacy of carlumab181.
relates with reduced tumour burden and metastasis in The CCR2 small molecule inhibitor PF‑04136309
different experimental models of prostate, breast, lung was recently assessed in a phase Ib non-randomized
and liver cancers and melanoma172. However, withdrawal trial in patients with locally advanced pancreatic can-
of anti‑CCL2 treatment accelerated lung metastasis in cer in combination with FOLFIRINOX chemotherapy
mouse models of breast cancer and resulted in death (oxaliplatin and irinotecan plus leucovorin and fluoro-
of mice owing to a rebound in monocyte recruitment, uracil); 47 patients were treated and 8 patients received
which raised important concerns about the long-term only FOLFIRINOX, while the remaining patients
efficacy of this approach173,174. However, high levels of received FOLFIRINOX and PF‑04136309. Results

896 | DECEMBER 2018 | VOLUME 17 www.nature.com/nrd


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

showed that PF‑04136309 treatment in combination macrophages189. In the latter case, CD47 inhibits phago-
with FOLFIRINOX was safe and well tolerated com- cytosis by the prevention of myosin IIA accumulation at
pared with chemotherapy alone. Patients treated only the phagocytic synapse190. The result of this interaction
with FOLFIRINOX did not show an objective response is a ‘do not eat me’ signal that prevents phagocytosis of
to the treatment. By contrast, in the PF‑04136309 plus autologous cells in homeostatic conditions. This mecha­
FOLFIRINOX group, 16 of the 33 patients, who under- nism is tightly regulated, and it is mainly activated in
went repeated imaging assessments, had an objective pro-inflammatory conditions, as CD47‑null mutant
tumour response, and 32 of those patients achieved local mice show a phenotype against self only during inflam-
tumour control182. matory conditions191. CD47 is overexpressed in a variety
These trials, however, were largely disappointing of tumours192–195 through the activation of CD47‑specific
and indicated that much more biological knowledge super-enhancers196. It is involved in tumour invasion,
is needed to achieve effective inhibition of monocyte metastasis and, more importantly, inhibition of phago-
recruitment. For example, CCL2–CCR2 signalling is cytosis by the innate immune system by interacting with
required for monocyte egress from the bone into the SIRPα195 expressed on phagocytes.
blood, and CCL2 inhibition results in a severe deple- Several preclinical studies in xenograft mouse models
tion of monocytes. This depletion is recognized, and demonstrated that CD47 inhibition is an effective strat-
the organism attempts to overcome this deficiency with egy for tumour therapy 197–199, as it enables killing and
a dramatic elevation in CCL2 concentration, which in phagocytosis of tumour cells by macrophages. The role
turn prevents efficacy of the reagents. In our own stud- of CD47 in the inhibition of phagocyte-mediated killing
ies using genetic ablation, we have demonstrated the was confirmed recently in human small-cell lung and
requirement for CCR2 to recruit monocytes to the pri- ovarian cancer cell lines200,201. Highly phagocytic bone
mary tumour in the PyMT model but that in the absence marrow-derived macrophages, in which SIRPα had been
of CCR2 in monocytes this recruitment is completely inhibited by injection of antitumour antibodies against
overcome by unknown redundancy mechanisms4. In MUC1 and EGF receptor (EGFR) (cetuximab), could
addition, there may be compensatory proliferation of effectively reach the tumour and engulf human lung car-
tissue-resident macrophages if recruitment is blocked6. cinoma A549 cancer cells, causing tumour regression.
Therefore, much more biological understanding is However, the antitumoural effect observed was then lost
required before this approach to selectively inhibit owing to their differentiation to TAMs202.
monocyte recruitment to the tumour or its metastatic At the moment, there are two anti‑CD47 mAbs
derivatives is likely to be effective. Indeed, it may be (Hu5F9‑G4 and CC‑90002) and one soluble recombi-
better to explore inhibition of molecules required for nant SIRPα–crystallizable fragment (Fc) fusion protein
retention of monocytes, such as CCL3, or molecules that (TTI‑621) currently being tested in phase I clinical trials.
induce their differentiation4,80. Hu5F9‑G4 showed promising results at the preclinical
level in human AML203 and in paediatric brain tumours204.
Reprogramming of TAMs There are currently four ongoing clinical trials on different
Despite generally being pro-tumoural, TAMs can, solid and haematological malignancies205–208 to study the
depending on context, be tumoricidal and also suppress safety of Hu5F9‑G4 in patients. Preliminary results from
tumour growth by activating immune responses183. the NCT02216409 trial indicated that Hu5F9‑G4 was tol-
This suggests that macrophage plasticity can be thera­ erated with reversible side effects observed such as anae-
peutically exploited to restore antitumour properties mia, headache, nausea and retinal toxicity 209. Two trials
to TAMs184. Indeed, it might indicate the paucity of the assessing CC‑90002210,211 have been initiated in patients
approach of targeting all macrophages, as both pro- with haematological malignancies, but results have not
tumoural and antitumoural macrophages will be been published yet.
depleted. Instead, macrophage reprogramming is a tar- TTI‑621 is a fully human recombinant protein that
geting strategy that provides an opportunity to rebalance blocks the CD47–SIRPα axis and improves killing of
the microenvironment immune infiltrate therapeutically cancer cells. A recent study investigated the efficacy of
from a pro-tumoural one to one that actively rejects the TTI‑621 in aggressive AML and B cell lymphoma xeno­
tumour in synergy with T cell-enhancing drugs such as grafts; TTI‑621 successfully improved macrophage-
checkpoint inhibitors. It also eliminates the drawbacks mediated phagocytosis of cancer cells but not normal
and long-term toxicity of ablation of all macrophages, cells. Moreover, in vivo data suggested that TTI‑621
such as that seen in the case of anti‑CSF1R therapeu- treatment was able to control the growth of haemato-
tics discussed above. Different methods are currently logical and solid tumours in mouse xenografts models212.
being tested at the preclinical and clinical levels, as TTI‑621 is currently being tested at the clinical level in
discussed below. two ongoing clinical trials on haematological and mul-
tiple solid tumours213,214.
Anti‑CD47 antibodies. CD47 is a ubiquitous protein
that regulates cell migration, axon extension, cytokine Toll-like receptor agonist. Toll-like receptors (TLRs)
production and T cell activation185–188. CD47 interacts are innate immunity pattern recognition receptors that
with thrombospondin 1 and signal regulatory protein-α have fundamental roles in the activation of the innate
(SIRPα; also known as SHPS1), which are mainly immune response215. Activation of TLR by bacterial
expressed by myeloid cells, including dendritic cells and particles (such as lipopolysaccharide) and viral nucleic

NATURE REVIEWS | DRUG DISCOVERY VOLUME 17 | DECEMBER 2018 | 897


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

acids (RNA or DNA) polarizes macrophages towards a tumours were treated with a single intravenous dose of
pro-inflammatory phenotype. For this reason, different CP‑870,893; common adverse effects included cytokine
TLR synthetic ligands have been tested in cancer models release syndrome and alterations of immune cell num-
in order to assess their efficacy in the phenotypic switch ber but, in general, the treatment was well tolerated, and
of TAMs to tumoricidal macrophages in the TME216. CP‑870,893 led to an objective response and antitumour
In an orthotropic mammary tumour mouse model, activity 239. CP‑870,893 treatment in combination with
intra-tumoural delivery of TLR7 and TLR9 agonists carboplatin and paclitaxel was tested in 32 patients with
caused increased monocyte infiltration in the tumour advanced-stage solid tumours in another phase I trial; 6
and macrophage repolarization217; similar results were out of 30 evaluable patients showed partial response to
obtained with an agonist of both TLR7 and TLR8 treatment and depletion of B cells together with upreg-
(3M‑052) that induced macrophage repolarization and ulation of immune co‑stimulatory molecules240. Similar
improved tumoricidal activity in melanoma218. In pre- results were obtained in patients with malignant pleural
clinical models, the TLR7 ligand imiquimod, the only mesothelioma treated with CP‑870,893 in combination
TLR agonist approved for clinical use, showed anti­ with cisplatin and pemetrexed241 and in patients with
tumoural activity in basal cell carcinoma, melanoma advanced-stage pancreatic ductal adenocarcinoma242.
and breast cancer skin metastases219–222. RO7009789 is being studied in four ongoing clinical
Two TLR7 ligands (imiquimod and 852A) and one trials on advanced-stage solid tumours114,243–245.
TLR9 ligand (IMO‑2055) are being tested for their
antitumoural properties in clinical trials. Imiquimod Histone deacetylase inhibitors. Histone deacetylases
has been tested on several cancers: in a prospective (HDACs), of which there are 18 in mammals, are
clinical trial, topical treatment of skin metastases from divided into four classes246,247. HDACs are responsible
breast cancers with imiquimod was well tolerated, and for removing the acetyl groups on histones, a crucial
responders showed histological tumour regression and process in epigenetic regulation of gene expression.
increased lymphoid immune infiltration221. 852A has TMP195, a specific inhibitor of class IIA HDACs248,
been tested in five clinical trials in melanoma, leukae- can modify the epigenomic profile of monocytes and
mia and gynaecological cancers223–227. A phase I clinical macrophages, resulting in, for example, altered CCL1
trial on patients with advanced-stage cancer showed and CCL2 expression in monocytes and a pro-inflam-
that treatment with 852A three times per week for matory phenotype. In a model of luminal B‑type breast
2 weeks was well tolerated, with reversible side effects228. cancer, intraperitoneal injection of TMP195 increased
IMO‑2055 has been tested in CRCs and head and neck, infiltration of CD11b+ myeloid cells from blood into the
lung and renal cancers229–233. Results from a clinical trial tumour, where they differentiated into antitumoural
in patients with advanced metastatic NSCLC showed macrophages249. This resulted in reduced vessel permea­
good tolerability and potential antitumoural activity of bility as well as in reduced vasculature and tumour cell
IMO‑2055 when used in combination with erlotinib and proliferation. Moreover, the antitumour macrophage
bevacizumab234. phenotype induced by TMP195 treatment increased the
efficacy and durability of both standard chemotherapeu-
Anti‑CD40 antibodies. CD40 is a receptor that belongs tic regimens (carboplatin and paclitaxel) and immuno-
to the TNF receptor superfamily, and it is expressed by therapy (anti‑PD1 antibodies). These findings suggest
APCs such as monocytes, macrophages, dendritic cells that class IIA HDAC inhibitors can selectively repro-
and B cells235, but it can be expressed by endothelial gramme monocytes and macrophages in the tumour and
and epithelial cells as well. The natural ligand of CD40 open interesting therapeutic opportunities, although it
is CD40L, which is mainly expressed by CD4+ T cells, remains to be seen whether targeting this HDAC sub-
basophils and mast cells236. The CD40–CD40L inter­ class will be sufficiently specific if given systemically
action upregulates the expression of MHC molecules to patients.
and the production of pro-inflammatory cytokines, such
as IL‑12, which primes naive CD4+ and CD8+ T cells Anti-MARCO antibody therapy. The macrophage
into T helper and cytotoxic cells, respectively. Agonistic receptor with collagenous structure (MARCO) is a
anti‑CD40 antibodies exert tumour inhibitory effects in pattern recognition receptor that belongs to the class A
several tumour mouse models, an observation that has scavenger receptor family. MARCO is mainly expressed
opened the way for the development of clinically relevant by macrophages250, and its expression was linked to
anti‑CD40 antibodies. poor prognosis in breast cancer 251. A recent report by
Interestingly, TAM treatment with CD40 agonists Georgoudaki et al.252 showed that MARCO is expressed
in combination with anti‑CSF1R antibodies results in a in TAMs of patients with breast cancer and metastatic
profound TAM reprogramming before their depletion; melanoma. MARCO neutralization with antibodies
these reprogrammed TAMs create a pro-inflammatory inhibits tumour growth and metastasis in the 4T1 mam-
environment that elicits effective T cell responses even mary carcinoma model. Similarly, using a B16 melanoma
in tumours that were non-responsive to immune check- mouse model, treatment with an anti-MARCO antibody
points inhibitors237,238. inhibited tumour growth and improved the effects of
Two agonistic anti‑CD40 antibodies are being anti‑CTLA4 immunotherapy 252. The antitumour activity
tested in clinical trials: CP‑870,893 and RO7009789. In of anti-MARCO therapy was dependent on the ability of
a phase I dose-escalation study, 29 patients with solid the Fc portion of the anti-MARCO antibody to engage

898 | DECEMBER 2018 | VOLUME 17 www.nature.com/nrd


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

with the inhibitory Fc receptor FcγRIIB, as previously reprogramming (such as PI3Kγ and HDAC inhibitors)
demonstrated with other mAb-mediated reprogram- and targeting of functional molecules (such as argin-
ming strategies253. This study highlights the feasibility of ase 1 and Fc receptors) are in preclinical and clinical
antibody-mediated reprogramming of macrophages by trials (FIG. 4). Despite these advances, each strategy
using TAM-derived targets and stresses the importance needs further investigation, as they all have limitations.
of the correct design of the antibodies, especially the Fc For example, the general depletion of monocytes and
region, for future clinical interventions. macrophages exerted by CSF1R inhibitors is not TAM-
specific and thus has substantial toxicity over time259.
PI3Kγ inhibitors. PI3Ks are involved in almost all types Furthermore, given the complexity of TAM popula-
of signalling in cells254. There are several subclasses of tions, there is growing awareness that the functioning of
PI3K, of which class 1B PI3Kγ is mainly expressed in macro­phages and dendritic cells is required for anti‑PD1
haematopoietic cells. Mice that lack PI3Kγ expression (REF.260 ) and anti‑CTLA4 therapies261, respectively. Thus,
show impaired recruitment of inflammatory cells, mainly potential alternative strategies would be to ablate TAMs
macrophages and neutrophils255. Kaneda et al.256 showed transiently, followed by recovery periods during which
that PI3Kγ is a key regulator of the tumour immune sup- monocytes could be recruited into the tissue to promote
pression exerted by TAMs; genetic and pharmacological antitumour immune reactions before differentiating into
inhibition of this target induced the expression of MHC pro-tumoural TAMs. This strategy is attractive, but it will
class II (MHC‑II) molecules together with upregulation require a careful timing and a better knowledge of the
of IL‑12 and decreased secretion of IL‑10. As a result, immune interactions ongoing in all phases of tumour
the inhibition of PI3Kγ in TAMs caused recruitment of formation262.
cells associated with antitumour adaptive immunity and Therefore, going forward, a better strategy would be
tumour growth inhibition. At the clinical level, patients to specifically target pro-tumoural macrophages and
with head and neck and lung cancers with low PI3Kγ enhance the activity of antitumoural ones or to repo-
activity had a better prognosis and longer overall sur- larize existing ones to have antitumoural activities.
vival, suggesting that PI3Kγ could be a potential future In this context, TAM therapy aimed at the functional
therapeutic target. modulation of TAM subpopulations through the use of
mAbs showed promising preclinical results. This strat-
Inhibition of microRNA activity. MicroRNAs (mi­RNAs) egy, combined with the use of Fc receptor inhibitors,
are small non-coding RNAs that regulate transcription seems to be the most promising one for the modulation
and translation in a sequence-specific manner, and of the TME, as macrophages are able to rapidly take up
their maturation is regulated by the RNase-III enzyme anti‑PD1 antibodies through their Fc receptors, limiting
DICER257. A recent study showed that inhibition of the efficacy of the checkpoint inhibitors. Thus, it will be
DICER in macrophages affects TAM programming fundamental to identify TAM-specific targets (such as
and is associated with tumour regression and altered MARCO) to improve therapy specificity. Other meth-
infiltration of immune cells258. DICER inhibition repro- ods of macrophage manipulation have been introduced,
grammed TAMs to express an IFNγ–STAT1 signature including macrophages that express chimeric antigen
and to become antitumoural. DICER inhibition in TAMs receptors (CAR-macrophages) and designer supramo-
was also associated with a better response to immunos- lecules — for example, against CD47 and CSF1R — that
timulatory antibodies258. These data raise the possibili- home to SIRPα-expressing macrophages and amplify
ties of identification and targeting mi­RNAs to repolarize immune responses, limiting tumour growth263,264. These
TAMs. exciting strategies will need considerable refinement to
reach human applications but may well be the wave of
Conclusions and perspectives the future.
TAMs represent a heterogeneous population with Another interesting approach might also be to under-
defined functions that differ between primary and stand the interactions between the microbiome and
metastatic tumours. Immune cell infiltrates also vary macrophages. Such a concept is based on recent studies
according to tumour type and, thus, dynamic inter­ suggesting that manipulation of the microbiome alters
actions between TAMs and other immune cells are cancer incidence as well as responses to therapy 265,266.
likely to vary according to cancer type and stage of pro- As macrophages are sentinel cells for changes in the
gression. In addition to pro-tumoural effects, TAMs can microbiota, usually through TLR engagement, under-
also have antitumour effects that in some cases might be standing the exact nature of these interactions and their
dominant. Consequently, it is necessary to understand consequences could potentially help tailor an anticancer
tumour heterogeneity and how it evolves during the pro- microbial cocktail267.
gression to malignancy and also following therapy. It is Another strategy is to target cancer-associated mye-
also judicious to realize that much of these data derive loid immature progenitors268, which are cells that are
from mouse models, and that there is scant knowledge mainly found in the blood of cancer patients and that
except at the most descriptive level about TAMs in are thought to be the progenitors of tumour-infiltrating
human cancers. myeloid cells. Several studies indicated that these imma-
Despite these caveats, given the consensus that ture cells have intrinsic immunosuppressive functions
TAMs are overall pro-tumoural, several anti-TAM strat- in vitro and therefore could represent a very promising
egies aimed at depletion (such as CSF1R inhibitors), target in combination with immunotherapy 269. However,

NATURE REVIEWS | DRUG DISCOVERY VOLUME 17 | DECEMBER 2018 | 899


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

again, the challenge is to identify specific markers that specific populations of cells, compensatory mechanisms
will allow the selective targeting of these abnormally between cells of the innate system and their mechanisms
expanded immature populations, as there are currently of immunosuppression and tissue repair. In this context,
not enough markers available to distinguish them from the extensive use of single-cell RNA sequencing, multi­
mature myeloid cells. plex immunohistochemistry and mass cytometry will
To improve all therapeutic modalities, a thorough considerably increase our knowledge, and it will allow
understanding of the TME is required in humans and the selection of novel TAM targets for the modulation
in different cancer types, including the identification of of the TME to improve therapy.

1. Dunn, G. P., Old, L. J. & Schreiber, R. D. The three Es 19. Baghdadi, M. et al. Chemotherapy-induced IL34 37. Price, F. V. et al. Colony-stimulating factor‑1 in primary
of cancer immunoediting. Annu. Rev. Immunol. 22, enhances immunosuppression by tumor-associated ascites of ovarian cancer is a significant predictor of
329–360 (2004). macrophages and mediates survival of chemoresistant survival. Am. J. Obstet. Gynecol. 168, 520–527
2. Gajewski, T. F., Schreiber, H. & Fu, Y. X. Innate and lung cancer cells. Cancer Res. 76, 6030–6042 (2016). (1993).
adaptive immune cells in the tumor microenvironment. 20. Prehn, R. T. The immune reaction as a stimulator of 38. Smith, H. O. et al. The role of colony-stimulating factor
Nat. Immunol. 14, 1014–1022 (2013). tumor growth. Science 176, 170–171 (1972). 1 and its receptor in the etiopathogenesis of
3. Noy, R. & Pollard, J. W. Tumor-associated This is a study that introduces the idea that endometrial adenocarcinoma. Clin. Cancer Res. 1,
macrophages: from mechanisms to therapy. Immunity immune cells promote tumour growth. 313–325 (1995).
41, 49–61 (2014). 21. Fidler, I. J. & Schroit, A. J. Recognition and destruction This is the first prospective study to show that
4. Arwert, E. N. et al. A unidirectional transition from of neoplastic cells by activated macrophages: patients with endometrial cancer have elevated
migratory to perivascular macrophage is required for discrimination of altered self. Biochim. Biophys. Acta levels of serum CSF1 and high expression of CSF1
tumor cell intravasation. Cell Rep. 23, 1239–1248 948, 151–173 (1988). and its receptor at the tissue level compared with
(2018). 22. Mantovani, A., Ming, W. J., Balotta, C., Abdeljalil, B. & control groups.
This paper shows both the recruitment of Bottazzi, B. Origin and regulation of tumor-associated 39. Smith, H. O. et al. The clinical significance of
monocytes and their ultimate differentiation to macrophages: the role of tumor-derived chemotactic inflammatory cytokines in primary cell culture in
perivascular macrophages that promote tumour factor. Biochim. Biophys. Acta 865, 59–67 (1986). endometrial carcinoma. Mol. Oncol. 7, 41–54
cell intravasation. 23. Hibbs, J. B., Vavrin, Z. & Taintor, R. R. l‑Arginine is (2013).
5. Qian, B. Z. et al. CCL2 recruits inflammatory required for expression of the activated macrophage 40. West, R. B. et al. Determination of stromal signatures
monocytes to facilitate breast-tumour metastasis. effector mechanism causing selective metabolic in breast carcinoma. PLOS Biol. 3, e187 (2005).
Nature 475, 222–225 (2011). inhibition in target cells. J. Immunol. 138, 550–565 41. Steidl, C. et al. Tumor-associated macrophages and
This is the first paper to show that classical (1987). survival in classic Hodgkin’s lymphoma. N. Engl.
monocytes are recruited to lung metastases from 24. Lewis, C. E. & Pollard, J. W. Distinct role of J. Med. 362, 875–885 (2010).
breast tumours through a CCL2–CCR2‑dependent macrophages in different tumor microenvironments. 42. Sapi, E. The role of CSF‑1 in normal physiology of
mechanism and that inhibition of this recruitment Cancer Res. 66, 605–612 (2006). mammary gland and breast cancer: an update. Exp.
reduces metastasis. 25. Lin, E. Y., Nguyen, A. V., Russell, R. G. & Pollard, J. W. Biol. Med. 229, 1–11 (2004).
6. Zhu, Y. et al. Tissue-resident macrophages in Colony-stimulating factor 1 promotes progression of 43. Tang, R. et al. M‑CSF (monocyte colony stimulating
pancreatic ductal adenocarcinoma originate from mammary tumors to malignancy. J. Exp. Med. 193, factor) and M‑CSF receptor expression by breast
embryonic hematopoiesis and promote tumor 727–740 (2001). tumour cells: M‑CSF mediated recruitment of
progression. Immunity 47, 597 (2017). This is the first study to show that CSF1 and tumour infiltrating monocytes? J. Cell. Biochem. 50,
This paper shows that TAMs in pancreatic ductal macrophages promote tumour progression and 350–356 (1992).
adenocarcinoma originate from both bone metastasis of mammary tumours. 44. Scholl, S. M. et al. Expression of colony-stimulating
marrow-derived monocytes and embryonic-derived 26. Hanahan, D. & Coussens, L. M. Accessories to the factor‑1 and its receptor (the protein product of c‑fms)
tissue-resident macrophages and that the latter crime: functions of cells recruited to the tumor in invasive breast tumor cells: induction of urokinase
support tumour progression. microenvironment. Cancer Cell 21, 309–322 (2012). production via this pathway? Ann. NY Acad. Sci. 698,
7. Chen, Z. et al. Cellular and molecular identity of 27. Quail, D. F. & Joyce, J. A. Microenvironmental 131–135 (1993).
tumor-associated macrophages in glioblastoma. regulation of tumor progression and metastasis. Nat. 45. Chambers, S. K., Kacinski, B. M., Ivins, C. M. &
Cancer Res. 77, 2266–2278 (2017). Med. 19, 1423–1437 (2013). Carcangiu, M. L. Overexpression of epithelial
8. Bowman, R. L. et al. Macrophage ontogeny underlies 28. Kitamura, T., Qian, B. Z. & Pollard, J. W. Immune cell macrophage colony-stimulating factor (CSF‑1) and
differences in tumor-specific education in brain promotion of metastasis. Nat. Rev. Immunol. 15, CSF‑1 receptor: a poor prognostic factor in epithelial
malignancies. Cell Rep. 17, 2445–2459 (2016). 73–86 (2015). ovarian cancer, contrasted with a protective effect of
9. Topalian, S. L., Drake, C. G. & Pardoll, D. M. Immune 29. Zhang, Q. W. et al. Prognostic significance of tumor- stromal CSF‑1. Clin. Cancer Res. 3, 999–1007
checkpoint blockade: a common denominator associated macrophages in solid tumor: a meta-analysis (1997).
approach to cancer therapy. Cancer Cell 27, 450–461 of the literature. PLOS ONE 7, e50946 (2012). 46. Lamprecht, B. et al. Derepression of an endogenous
(2015). 30. Yin, S. et al. The prognostic and clinicopathological long terminal repeat activates the CSF1R proto-
10. Ruffell, B. & Coussens, L. M. Macrophages and significance of tumor-associated macrophages in oncogene in human lymphoma. Nat. Med. 16, 571–
therapeutic resistance in cancer. Cancer Cell 27, patients with gastric cancer: a meta-analysis. PLOS 579 (2010).
462–472 (2015). ONE 12, e0170042 (2017). 47. Crusz, S. M. & Balkwill, F. R. Inflammation and cancer:
11. Mantovani, A., Marchesi, F., Malesci, A., Laghi, L. & 31. Zhao, X. et al. Prognostic significance of tumor- advances and new agents. Nat. Rev. Clin. Oncol. 12,
Allavena, P. Tumour-associated macrophages as associated macrophages in breast cancer: a meta- 584–596 (2015).
treatment targets in oncology. Nat. Rev. Clin. Oncol. analysis of the literature. Oncotarget 8, 30576–30586 48. Moore, R. J. et al. Mice deficient in tumor necrosis
14, 399–416 (2017). (2017). factor-alpha are resistant to skin carcinogenesis. Nat.
12. Gordon, S., Plüddemann, A. & Martinez Estrada, F. 32. Guo, B., Cen, H., Tan, X. & Ke, Q. Meta-analysis of the Med. 5, 828–831 (1999).
Macrophage heterogeneity in tissues: phenotypic prognostic and clinical value of tumor-associated 49. Canli, O. et al. Myeloid cell-derived reactive oxygen
diversity and functions. Immunol. Rev. 262, 36–55 macrophages in adult classical Hodgkin lymphoma. species induce epithelial mutagenesis. Cancer Cell 32,
(2014). BMC Med. 14, 159 (2016). 869–883 (2017).
13. Pollard, J. W. Trophic macrophages in development 33. Mei, J. et al. Prognostic impact of tumor-associated 50. Balkwill, F. R. & Mantovani, A. Cancer-related
and disease. Nat. Rev. Immunol. 9, 259–270 macrophage infiltration in non-small cell lung cancer: inflammation: common themes and therapeutic
(2009). A systemic review and meta-analysis. Oncotarget 7, opportunities. Semin. Cancer Biol. 22, 33–40
14. Gautier, E. L. et al. Gene-expression profiles and 34217–34228 (2016). (2012).
transcriptional regulatory pathways that underlie the 34. Chambers, S. K., Wang, Y., Gertz, R. E. & 51. Deng, L. et al. A novel mouse model of inflammatory
identity and diversity of mouse tissue macrophages. Kacinski, B. M. Macrophage colony-stimulating factor bowel disease links mammalian target of rapamycin-
Nat. Immunol. 13, 1118–1128 (2012). mediates invasion of ovarian cancer cells through dependent hyperproliferation of colonic epithelium to
15. Chitu, V. & Stanley, E. R. Colony-stimulating factor‑1 in urokinase. Cancer Res. 55, 1578–1585 (1995). inflammation-associated tumorigenesis. Am. J. Pathol.
immunity and inflammation. Curr. Opin. Immunol. 18, 35. Kacinski, B. M. et al. FMS (CSF‑1 receptor) and CSF‑1 176, 952–967 (2010).
39–48 (2006). transcripts and protein are expressed by human 52. Kong, L. et al. Deletion of interleukin‑6 in monocytes/
16. Stanley, E. R. & Chitu, V. CSF‑1 receptor signaling in breast carcinomas in vivo and in vitro. Oncogene 6, macrophages suppresses the initiation of
myeloid cells. Cold Spring Harb. Perspect. Biol. 6, 941–952 (1991). hepatocellular carcinoma in mice. J. Exp. Clin. Cancer
a021857 (2014). 36. Scholl, S. M. et al. Circulating levels of colony- Res. 35, 131 (2016).
17. Lin, H. et al. Discovery of a cytokine and its receptor stimulating factor 1 as a prognostic indicator in 82 53. Oft, M. IL‑10: master switch from tumor-promoting
by functional screening of the extracellular proteome. patients with epithelial ovarian cancer. Br. J. Cancer inflammation to antitumor immunity. Cancer Immunol.
Science 320, 807–811 (2008). 69, 342–346 (1994). Res. 2, 194–199 (2014).
18. Wang, Y. et al. IL‑34 is a tissue-restricted ligand of This is the first study to show that elevated levels 54. Jobin, C. Precision medicine using microbiota. Science
CSF1R required for the development of Langerhans of CSF1 are an independent prognostic factor 359, 32–34 (2018).
cells and microglia. Nat. Immunol. 13, 753–760 associated with poor survival in patients with 55. Jobin, C. Colorectal cancer: looking for answers in the
(2012). ovarian cancer. microbiota. Cancer Discov. 3, 384–387 (2013).

900 | DECEMBER 2018 | VOLUME 17 www.nature.com/nrd


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

56. de Visser, K. E., Eichten, A. & Coussens, L. M. 77. Mellor, A. L., Keskin, D. B., Johnson, T., Chandler, P. 100. DeNardo, D. G. et al. Leukocyte complexity predicts
Paradoxical roles of the immune system during & Munn, D. H. Cells expressing indoleamine breast cancer survival and functionally regulates
cancer development. Nat. Rev. Cancer 6, 24–37 2,3‑dioxygenase inhibit T cell responses. J. Immunol. response to chemotherapy. Cancer Discov. 1, 54–67
(2006). 168, 3771–3776 (2002). (2011).
57. DeNardo, D. G. et al. CD4(+) T cells regulate 78. Mbongue, J. C. et al. The role of indoleamine 2, This study demonstrates that inhibition of
pulmonary metastasis of mammary carcinomas by 3‑dioxygenase in immune suppression and macrophage recruitment in the tumour can
enhancing protumor properties of macrophages. autoimmunity. Vaccines 3, 703–729 (2015). influence response to chemotherapy, suggesting
Cancer Cell 16, 91–102 (2009). 79. Qian, B. et al. A distinct macrophage population that macrophage targeting is a therapeutic strategy
This study elucidates a pro-tumoural role of mediates metastatic breast cancer cell extravasation, for cancer treatment.
IL‑4‑expressing CD4+ TH2 cells, which regulate the establishment and growth. PLOS ONE 4, e6562 101. Alishekevitz, D. et al. Macrophage-induced
phenotype of macrophages in mammary tumours (2009). lymphangiogenesis and metastasis following paclitaxel
and promote tumour progression. This is the first study to identify a novel chemotherapy is regulated by VEGFR3. Cell Rep. 17,
58. Ruffell, B., Affara, N. I. & Coussens, L. M. Differential subpopulation of TAMs that accumulate in metastatic 1344–1356 (2016).
macrophage programming in the tumor lesions and promote tumour dissemination. 102. Shree, T. et al. Macrophages and cathepsin proteases
microenvironment. Trends Immunol. 33, 119–126 80. Kitamura, T. et al. CCL2‑induced chemokine cascade blunt chemotherapeutic response in breast cancer.
(2012). promotes breast cancer metastasis by enhancing Genes Dev. 25, 2465–2479 (2011).
59. Bruno, A. et al. Orchestration of angiogenesis by retention of metastasis-associated macrophages. 103. Zhang, X. et al. Macrophages induce resistance to
immune cells. Front. Oncol. 4, 131 (2014). J. Exp. Med. 212, 1043–1059 (2015). 5‑fluorouracil chemotherapy in colorectal cancer
60. Bergers, G. & Benjamin, L. E. Tumorigenesis and the 81. Zhao, L. et al. Recruitment of a myeloid cell subset through the release of putrescine. Cancer Lett. 381,
angiogenic switch. Nat. Rev. Cancer 3, 401–410 (CD11b/Gr1 mid) via CCL2/CCR2 promotes the 305–313 (2016).
(2003). development of colorectal cancer liver metastasis. 104. Schumacher, T. N. & Schreiber, R. D. Neoantigens
61. Yeo, E. J. et al. Myeloid WNT7b mediates the Hepatology 57, 829–839 (2013). in cancer immunotherapy. Science 348, 69–74
angiogenic switch and metastasis in breast cancer. 82. Qian, B. Z. et al. FLT1 signaling in metastasis- (2015).
Cancer Res. 74, 2962–2973 (2014). associated macrophages activates an inflammatory 105. Rosenberg, S. A., Yang, J. C. & Restifo, N. P. Cancer
62. Lin, E. Y. & Pollard, J. W. Tumor-associated signature that promotes breast cancer metastasis. immunotherapy: moving beyond current vaccines. Nat.
macrophages press the angiogenic switch in breast J. Exp. Med. 212, 1433–1448 (2015). Med. 10, 909–915 (2004).
cancer. Cancer Res. 67, 5064–5066 (2007). 83. Kitamura, T. et al. Monocytes differentiate to immune 106. Sharma, P. & Allison, J. P. Immune checkpoint
63. Hughes, R. et al. Perivascular M2 macrophages suppressive precursors of metastasis-associated targeting in cancer therapy: toward combination
stimulate tumor relapse after chemotherapy. Cancer macrophages in mouse models of breast cancer. strategies with curative potential. Cell 161, 205–214
Res. 75, 3479–3491 (2015). Front. Immunol. 8, 2004 (2018). (2015).
64. Hirano, H., Tanioka, K., Yokoyama, S., Akiyama, S. & 84. Entenberg, D. et al. In vivo subcellular resolution 107. Gordon, S. R. et al. PD‑1 expression by tumour-
Kuratsu, J. Angiogenic effect of thymidine optical imaging in the lung reveals early metastatic associated macrophages inhibits phagocytosis
phosphorylase on macrophages in glioblastoma proliferation and motility. Intravital 4, e1086613 and tumour immunity. Nature 545, 495–499
multiforme. J. Neurosurg. 95, 89–95 (2001). (2015). (2017).
65. De Palma, M. et al. Tie2 identifies a hematopoietic 85. Chen, Q., Zhang, X. H. & Massague, J. Macrophage This study shows that human and mouse TAMs
lineage of proangiogenic monocytes required for binding to receptor VCAM‑1 transmits survival express PD1, which inhibits tumour phagocytosis.
tumor vessel formation and a mesenchymal signals in breast cancer cells that invade the lungs. Inhibition of PD1 on TAMs reduced tumour growth,
population of pericyte progenitors. Cancer Cell 8, Cancer Cell 20, 538–549 (2011). suggesting that checkpoint inhibitor therapy is also
211–226 (2005). 86. Gao, D. et al. Myeloid progenitor cells in the effective on TAMs.
This is the first study to identify a novel population premetastatic lung promote metastases by inducing 108. Neubert, N. J. et al. T cell-induced CSF1 promotes
of pro-angiogenic monocytes expressing TIE2, mesenchymal to epithelial transition. Cancer Res. melanoma resistance to PD1 blockade. Sci. Transl
which are recruited to the tumour and account for 72, 1384–1394 (2012). Med. 10, eaan3311 (2018).
most of the pro-angiogenic activity among myeloid 87. Camacho, D. F. & Pienta, K. J. A multi-targeted 109. US National Library of Medicine. ClinicalTrials.gov
cells in the tumour. approach to treating bone metastases. Cancer https://clinicaltrials.gov/ct2/show/NCT02452424
66. Mazzieri, R. et al. Targeting the ANG2/TIE2 axis Metastasis Rev. 33, 545–553 (2014). (2018).
inhibits tumor growth and metastasis by impairing 88. Madani, I., De Neve, W. & Mareel, M. Does ionizing 110. US National Library of Medicine. ClinicalTrials.gov
angiogenesis and disabling rebounds of radiation stimulate cancer invasion and metastasis? https://clinicaltrials.gov/ct2/show/NCT02777710
proangiogenic myeloid cells. Cancer Cell 19, Bull. Cancer 95, 292–300 (2008). (2017).
512–526 (2011). 89. Moding, E. J., Kastan, M. B. & Kirsch, D. G. 111. US National Library of Medicine. ClinicalTrials.gov
67. Wyckoff, J. et al. A paracrine loop between tumor cells Strategies for optimizing the response of cancer and https://clinicaltrials.gov/ct2/show/NCT02880371
and macrophages is required for tumor cell migration normal tissues to radiation. Nat. Rev. Drug Discov. (2018).
in mammary tumors. Cancer Res. 64, 7022–7029 12, 526–542 (2013). 112. US National Library of Medicine. ClinicalTrials.gov
(2004). 90. Russell, J. S. & Brown, J. M. The irradiated tumor https://clinicaltrials.gov/ct2/show/NCT02829723
This study identifies a direct crosstalk between microenvironment: role of tumor-associated (2018).
tumour cells and macrophages that involves EGF macrophages in vascular recovery. Front. Physiol. 4, 113. US National Library of Medicine. ClinicalTrials.gov
and CSF1 and is required for tumour cell migration. 157 (2013). https://clinicaltrials.gov/ct2/show/NCT02323191
68. Goswami, S. et al. Macrophages promote the invasion 91. Kaur, P. & Asea, A. Radiation-induced effects and the (2018).
of breast carcinoma cells via a colony-stimulating immune system in cancer. Front. Oncol. 2, 191 (2012). 114. US National Library of Medicine. ClinicalTrials.gov
factor‑1/epidermal growth factor paracrine loop. 92. Shiao, S. L. & Coussens, L. M. The tumor-immune https://clinicaltrials.gov/ct2/show/NCT02760797
Cancer Res. 65, 5278–5283 (2005). microenvironment and response to radiation therapy. (2018).
69. Hernandez, L. et al. The EGF/CSF‑1 paracrine invasion J. Mammary Gland Biol. Neoplasia 15, 411–421 115. US National Library of Medicine. ClinicalTrials.gov
loop can be triggered by heregulin beta1 and CXCL12. (2010). https://clinicaltrials.gov/ct2/show/NCT02713529
Cancer Res. 69, 3221–3227 (2009). 93. Barker, H. E., Paget, J. T., Khan, A. A. & (2018).
70. Harney, A. S. et al. Real-time imaging reveals local, Harrington, K. J. The tumour microenvironment after 116. US National Library of Medicine. ClinicalTrials.gov
transient vascular permeability, and tumor cell radiotherapy: mechanisms of resistance and https://clinicaltrials.gov/ct2/show/NCT02526017
intravasation stimulated by TIE2hi macrophage- recurrence. Nat. Rev. Cancer 15, 409–425 (2015). (2018).
derived VEGFA. Cancer Discov. 5, 932–943 (2015). 94. Teresa Pinto, A. et al. Ionizing radiation modulates 117. US National Library of Medicine. ClinicalTrials.gov
71. Rohan, T. E. et al. Tumor microenvironment of human macrophages towards a pro-inflammatory https://clinicaltrials.gov/ct2/show/NCT02718911
metastasis and risk of distant metastasis of breast phenotype preserving their pro-invasive and pro- (2018).
cancer. J. Natl Cancer Inst. 106, dju136 (2014). angiogenic capacities. Sci. Rep. 6, 18765 (2016). 118. US National Library of Medicine. ClinicalTrials.gov
72. Ojalvo, L. S., King, W., Cox, D. & Pollard, J. W. High- 95. Hellevik, T. & Martinez-Zubiaurre, I. Radiotherapy https://clinicaltrials.gov/ct2/show/NCT02807844
density gene expression analysis of tumor-associated and the tumor stroma: the importance of dose and (2018).
macrophages from mouse mammary tumors. Am. fractionation. Front. Oncol. 4, 1 (2014). 119. US National Library of Medicine. ClinicalTrials.gov
J. Pathol. 174, 1048–1064 (2009). 96. Correia, A. L. & Bissell, M. J. The tumor https://clinicaltrials.gov/ct2/show/NCT02554812
73. Ojalvo, L. S., Whittaker, C. A., Condeelis, J. S. & microenvironment is a dominant force in multidrug (2018).
Pollard, J. W. Gene expression analysis of resistance. Drug Resist. Updat. 15, 39–49 (2012). 120. Coniglio, S. J. et al. Microglial stimulation of
macrophages that facilitate tumor invasion 97. Nakasone, E. S. et al. Imaging tumor-stroma glioblastoma invasion involves epidermal growth factor
supports a role for Wnt-signaling in mediating their interactions during chemotherapy reveals receptor (EGFR) and colony stimulating factor 1
activity in primary mammary tumors. J. Immunol. contributions of the microenvironment to resistance. receptor (CSF‑1R) signaling. Mol. Med. 18, 519–527
184, 702–712 (2010). Cancer Cell 21, 488–503 (2012). (2012).
74. Morandi, F. & Pistoia, V. Interactions between HLA‑G 98. Shaked, Y. Balancing efficacy of and host immune 121. Yan, D. et al. Inhibition of colony stimulating factor‑1
and HLA‑E in physiological and pathological responses to cancer therapy: the yin and yang receptor abrogates microenvironment-mediated
conditions. Front. Immunol. 5, 394 (2014). effects. Nat. Rev. Clin. Oncol. 13, 611–626 (2016). therapeutic resistance in gliomas. Oncogene 36,
75. Santarpia, M. & Karachaliou, N. Tumor immune 99. Paulus, P., Stanley, E. R., Schafer, R., Abraham, D. & 6049–6058 (2017).
microenvironment characterization and response to Aharinejad, S. Colony-stimulating factor‑1 antibody 122. Tap, W. D. et al. Structure-guided blockade of CSF1R
anti‑PD‑1 therapy. Cancer Biol. Med. 12, 74–78 reverses chemoresistance in human MCF‑7 breast kinase in tenosynovial giant-cell tumor. N. Engl.
(2015). cancer xenografts. Cancer Res. 66, 4349–4356 J. Med. 373, 428–437 (2015).
76. Buchbinder, E. I. & Desai, A. CTLA‑4 and PD‑1 (2006). 123. Molena, B. et al. Synovial colony-stimulating factor‑1
pathways: similarities, differences, and implications of This study shows the antitumoural effects of mRNA expression in diffuse pigmented villonodular
their inhibition. Am. J. Clin. Oncol. 39, 98–106 anti‑CSF1 therapy in MCF‑7 breast cancer synovitis. Clin. Exp. Rheumatol 29, 547–550
(2016). xenografts. (2011).

NATURE REVIEWS | DRUG DISCOVERY VOLUME 17 | DECEMBER 2018 | 901


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

124. Ries, C. H. et al. Targeting tumor-associated 147. Daubiné, F., Le Gall, C., Gasser, J., Green, J. & 170. Fang, W. B. et al. Targeted gene silencing of CCL2
macrophages with anti-CSF‑1R antibody reveals a Clézardin, P. Antitumor effects of clinical dosing inhibits triple negative breast cancer progression
strategy for cancer therapy. Cancer Cell 25, 846–859 regimens of bisphosphonates in experimental breast by blocking cancer stem cell renewal and M2
(2014). cancer bone metastasis. J. Natl Cancer Inst. 99, macrophage recruitment. Oncotarget 7,
This is the first clinical report showing activity of an 322–330 (2007). 49349–49367 (2016).
anti‑CSF1R antibody in diffuse-type giant cell 148. Coscia, M. et al. Zoledronic acid repolarizes tumour- 171. Peña, C. G. et al. LKB1 loss promotes endometrial
tumours. associated macrophages and inhibits mammary cancer progression via CCL2‑dependent macrophage
125. Butowski, N. et al. Orally administered colony carcinogenesis by targeting the mevalonate pathway. recruitment. J. Clin. Invest. 125, 4063–4076 (2015).
stimulating factor 1 receptor inhibitor PLX3397 in J. Cell. Mol. Med. 14, 2803–2815 (2010). This is an important paper showing how loss of a
recurrent glioblastoma: an Ivy Foundation Early Phase 149. Comito, G. et al. Zoledronic acid impairs stromal tumour suppressor gene can induce CCL2
Clinical Trials Consortium phase II study. Neuro Oncol. reactivity by inhibiting M2‑macrophages polarization production that results in TAM recruitment.
18, 557–564 (2016). and prostate cancer-associated fibroblasts. 172. Li, M., Knight, D. A., A. Snyder, L., Smyth, M. J. &
126. von Tresckow, B. et al. An open-label, multicenter, Oncotarget 8, 118–132 (2017). Stewart, T. J. A role for CCL2 in both tumor
phase I/II Study of JNJ‑40346527, a CSF‑1R 150. Marra, M. et al. Cutting the limits of progression and immunosurveillance.
inhibitor, in patients with relapsed or refractory aminobisphosphonates: new strategies for the Oncoimmunology 2, e25474 (2013).
Hodgkin lymphoma. Clin. Cancer Res. 21, potentiation of their anti-tumour effects. Curr. Cancer 173. Bonapace, L. et al. Cessation of CCL2 inhibition
1843–1850 (2015). Drug Targets 9, 791–800 (2009). accelerates breast cancer metastasis by promoting
127. US National Library of Medicine. ClinicalTrials.gov 151. Marra, M. et al. Nanotechnologies to use angiogenesis. Nature 515, 130–133 (2014).
https://clinicaltrials.gov/ct2/show/NCT01804530 bisphosphonates as potent anticancer agents: the This study reports the paradoxical pro-metastatic
(2018). effects of zoledronic acid encapsulated into liposomes. effect of the cessation CCL2 inhibition in different
128. US National Library of Medicine. ClinicalTrials.gov Nanomedicine 7, 955–964 (2011). tumour models.
https://clinicaltrials.gov/ct2/show/NCT01316822 152. Marra, M. et al. New self-assembly nanoparticles and 174. Hitchcock, J. R. & Watson, C. J. Anti‑CCL2: building a
(2018). stealth liposomes for the delivery of zoledronic acid: a reservoir or opening the floodgates to metastasis?
129. Pyonteck, S. M. et al. CSF‑1R inhibition alters comparative study. Biotechnol. Adv. 30, 302–309 Breast Cancer Res. 17, 68 (2015).
macrophage polarization and blocks glioma (2012). 175. Ueno, T. et al. Significance of macrophage
progression. Nat. Med. 19, 1264–1272 (2013). 153. Ben-Aharon, I. et al. Bisphosphonates in the adjuvant chemoattractant protein‑1 in macrophage
This is an important paper in mouse models of setting of breast cancer therapy—effect on survival: a recruitment, angiogenesis, and survival in human
glioblastoma that shows that macrophages can be systematic review and meta-analysis. PLOS ONE 8, breast cancer. Clin. Cancer Res. 6, 3282–3289
repolarized to become antitumoural. e70044 (2013). (2000).
130. Quail, D. F. et al. The tumor microenvironment 154. Gnant, M. et al. Endocrine therapy plus zoledronic 176. Lebrecht, A. et al. Monocyte chemoattractant
underlies acquired resistance to CSF‑1R inhibition in acid in premenopausal breast cancer. N. Engl. J. Med. protein‑1 serum levels in patients with breast cancer.
gliomas. Science 352, aad3018 (2016). 360, 679–691 (2009). Tumour Biol. 25, 14–17 (2004).
131. Cassier, P. A. et al. CSF1R inhibition with 155. US National Library of Medicine. ClinicalTrials.gov 177. Loberg, R. D. et al. CCL2 as an important mediator of
emactuzumab in locally advanced diffuse-type https://clinicaltrials.gov/ct2/show/NCT02347163 prostate cancer growth in vivo through the regulation
tenosynovial giant cell tumours of the soft tissue: a (2017). of macrophage infiltration. Neoplasia 9, 556–562
dose-escalation and dose-expansion phase 1 study. 156. US National Library of Medicine. ClinicalTrials.gov (2007).
Lancet Oncol. 16, 949–956 (2015). https://clinicaltrials.gov/ct2/show/NCT02622607 178. Loberg, R. D. et al. Targeting CCL2 with systemic
132. US National Library of Medicine. ClinicalTrials.gov (2017). delivery of neutralizing antibodies induces prostate
https://clinicaltrials.gov/ct2/show/NCT02471716 157. US National Library of Medicine. ClinicalTrials.gov cancer tumor regression in vivo. Cancer Res. 67,
(2018). https://clinicaltrials.gov/ct2/show/NCT00127205 9417–9424 (2007).
133. US National Library of Medicine. ClinicalTrials.gov (2017). 179. Moisan, F. et al. Enhancement of paclitaxel and
https://clinicaltrials.gov/ct2/show/NCT03158272 158. US National Library of Medicine. ClinicalTrials.gov carboplatin therapies by CCL2 blockade in ovarian
(2018). https://clinicaltrials.gov/ct2/show/NCT00009945 cancers. Mol. Oncol. 8, 1231–1239 (2014).
134. US National Library of Medicine. ClinicalTrials.gov (2017). 180. Sandhu, S. K. et al. A first‑in‑human, first‑in‑class,
https://clinicaltrials.gov/ct2/show/NCT01346358 159. US National Library of Medicine. ClinicalTrials.gov phase I study of carlumab (CNTO 888), a human
(2018). https://clinicaltrials.gov/ct2/show/NCT00003232 monoclonal antibody against CC‑chemokine ligand 2
135. US National Library of Medicine. ClinicalTrials.gov (2010). in patients with solid tumors. Cancer Chemother.
https://clinicaltrials.gov/ct2/show/NCT02265536 160. Cuevas, C. & Francesch, A. Development of Yondelis Pharmacol. 71, 1041–1050 (2013).
(2018). (trabectedin, ET‑743). A semisynthetic process solves 181. Pienta, K. J. et al. Phase 2 study of carlumab (CNTO
136. US National Library of Medicine. ClinicalTrials.gov the supply problem. Nat. Prod. Rep. 26, 322–337 888), a human monoclonal antibody against
https://clinicaltrials.gov/ct2/show/NCT03153410 (2009). CC‑chemokine ligand 2 (CCL2), in metastatic
(2018). 161. Le Cesne, A. et al. A retrospective analysis of antitumour castration-resistant prostate cancer. Invest. New
137. Rodan, G. A. & Fleisch, H. A. Bisphosphonates: activity with trabectedin in translocation-related Drugs 31, 760–768 (2013).
mechanisms of action. J. Clin. Invest. 97, 2692–2696 sarcomas. Eur. J. Cancer 48, 3036–3044 (2012). 182. Nywening, T. M. et al. Targeting tumour-associated
(1996). 162. Grosso, F. et al. Efficacy of trabectedin macrophages with CCR2 inhibition in combination
138. Stresing, V., Daubiné, F., Benzaid, I., Mönkkönen, H. & (ecteinascidin‑743) in advanced pretreated myxoid with FOLFIRINOX in patients with borderline
Clézardin, P. Bisphosphonates in cancer therapy. liposarcomas: a retrospective study. Lancet Oncol. 8, resectable and locally advanced pancreatic cancer: a
Cancer Lett. 257, 16–35 (2007). 595–602 (2007). single-centre, open-label, dose-finding, non-
139. Van Acker, H. H., Anguille, S., Willemen, Y., 163. Monk, B. J. et al. Trabectedin plus pegylated randomised, phase 1b trial. Lancet Oncol. 17,
Smits, E. L. & Van Tendeloo, V. F. Bisphosphonates for liposomal doxorubicin (PLD) versus PLD in recurrent 651–662 (2016).
cancer treatment: mechanisms of action and lessons ovarian cancer: overall survival analysis. Eur. J. Cancer 183. Wynn, T. A., Chawla, A. & Pollard, J. W. Macrophage
from clinical trials. Pharmacol. Ther. 158, 24–40 48, 2361–2368 (2012). biology in development, homeostasis and disease.
(2016). 164. Germano, G. et al. Role of macrophage targeting in Nature 496, 445 (2013).
140. Moreau, M. F. et al. Comparative effects of five the antitumor activity of trabectedin. Cancer Cell 23, 184. Williams, C. B., Yeh, E. S. & Soloff, A. C. Tumor-
bisphosphonates on apoptosis of macrophage cells 249–262 (2013). associated macrophages: unwitting accomplices in
in vitro. Biochem. Pharmacol. 73, 718–723 (2007). This study shows that the antitumoural drug breast cancer malignancy. NPJ Breast Cancer 2,
141. Rogers, T. L. & Holen, I. Tumour macrophages as trabectedin induces monocyte and macrophage 15025 (2016).
potential targets of bisphosphonates. J. Transl Med. apoptosis through the activation of caspase 8 in a 185. Brown, E. J. & Frazier, W. A. Integrin-associated
9, 177 (2011). TRAIL-dependent mechanism. protein (CD47) and its ligands. Trends Cell Biol. 11,
142. Hiraoka, K. et al. Inhibition of bone and muscle 165. Liguori, M. et al. Functional TRAIL receptors in 130–135 (2001).
metastases of lung cancer cells by a decrease in the monocytes and tumor-associated macrophages: a 186. Gao, A. G. et al. Integrin-associated protein is a
number of monocytes/macrophages. Cancer Sci. 99, possible targeting pathway in the tumor receptor for the C‑terminal domain of
1595–1602 (2008). microenvironment. Oncotarget 7, 41662–41676 thrombospondin. J. Biol. Chem. 271, 21–24 (1996).
143. Gazzaniga, S. et al. Targeting tumor-associated (2016). 187. Liu, Y. et al. The role of CD47 in neutrophil
macrophages and inhibition of MCP‑1 reduce 166. Amarante-Mendes, G. P. & Griffith, T. S. Therapeutic transmigration. Increased rate of migration correlates
angiogenesis and tumor growth in a human melanoma applications of TRAIL receptor agonists in cancer and with increased cell surface expression of CD47. J. Biol.
xenograft. J. Invest. Dermatol. 127, 2031–2041 beyond. Pharmacol. Ther. 155, 117–131 (2015). Chem. 276, 40156–40166 (2001).
(2007). 167. Deshmane, S. L., Kremlev, S., Amini, S. & 188. Lindberg, F. P. et al. Decreased resistance to bacterial
144. Zeisberger, S. M. et al. Clodronate-liposome-mediated Sawaya, B. E. Monocyte chemoattractant protein‑1 infection and granulocyte defects in IAP-deficient mice.
depletion of tumour-associated macrophages: a new (MCP‑1): an overview. J. Interferon Cytokine Res. 29, Science 274, 795–798 (1996).
and highly effective antiangiogenic therapy approach. 313–326 (2009). 189. Okazawa, H. et al. Negative regulation of phagocytosis
Br. J. Cancer 95, 272–281 (2006). 168. Chun, E. et al. CCL2 promotes colorectal in macrophages by the CD47‑SHPS‑1 system.
145. Zhang, W. et al. Depletion of tumor-associated carcinogenesis by enhancing polymorphonuclear J. Immunol. 174, 2004–2011 (2005).
macrophages enhances the effect of sorafenib in myeloid-derived suppressor cell population and 190. Barclay, A. N. & Van den Berg, T. K. The interaction
metastatic liver cancer models by antimetastatic and function. Cell Rep. 12, 244–257 (2015). between signal regulatory protein alpha (SIRPα) and
antiangiogenic effects. Clin. Cancer Res. 16, 169. Fang, W. B. et al. CCL2/CCR2 chemokine signaling CD47: structure, function, and therapeutic target.
3420–3430 (2010). coordinates survival and motility of breast cancer cells Annu. Rev. Immunol. 32, 25–50 (2014).
146. Guth, A. M., Hafeman, S. D., Elmslie, R. E. & through Smad3 protein- and p42/44 mitogen- 191. Bian, Z. et al. Cd47‑Sirpα interaction and IL‑10
Dow, S. W. Liposomal clodronate treatment for activated protein kinase (MAPK)-dependent constrain inflammation-induced macrophage
tumour macrophage depletion in dogs with soft-tissue mechanisms. J. Biol. Chem. 287, 36593–36608 phagocytosis of healthy self-cells. Proc. Natl Acad. Sci.
sarcoma. Vet. Comp. Oncol. 11, 296–305 (2013). (2012). USA 113, E5434–E5443 (2016).

902 | DECEMBER 2018 | VOLUME 17 www.nature.com/nrd


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

192. Jaiswal, S. et al. CD47 is upregulated on circulating 218. Singh, M. et al. Effective innate and adaptive 244. US National Library of Medicine. ClinicalTrials.gov
hematopoietic stem cells and leukemia cells to avoid antimelanoma immunity through localized TLR7/8 https://clinicaltrials.gov/ct2/show/NCT02665416
phagocytosis. Cell 138, 271–285 (2009). activation. J. Immunol. 193, 4722–4731 (2014). (2018).
This study shows that CD47 expression is 219. Kobold, S., Wiedemann, G., Rothenfußer, S. & 245. US National Library of Medicine. ClinicalTrials.gov
upregulated by leukaemic cells to escape Endres, S. Modes of action of TLR7 agonists in cancer https://clinicaltrials.gov/ct2/show/NCT02588443
phagocytosis by macrophages. therapy. Immunotherapy 6, 1085–1095 (2014). (2017).
193. Chao, M. P. et al. Anti‑CD47 antibody synergizes with 220. Vacchelli, E. et al. Trial watch: immunotherapy plus 246. Arrowsmith, C. H., Bountra, C., Fish, P. V., Lee, K. &
rituximab to promote phagocytosis and eradicate radiation therapy for oncological indications. Schapira, M. Epigenetic protein families: a new
non-Hodgkin lymphoma. Cell 142, 699–713 (2010). Oncoimmunology 5, e1214790 (2016). frontier for drug discovery. Nat. Rev. Drug Discov. 11,
194. Zhao, H. et al. CD47 promotes tumor invasion and 221. Adams, S. et al. Topical TLR7 agonist imiquimod can 384–400 (2012).
metastasis in non-small cell lung cancer. Sci. Rep. 6, induce immune-mediated rejection of skin metastases 247. Choudhary, C., Weinert, B. T., Nishida, Y., Verdin, E. &
29719 (2016). in patients with breast cancer. Clin. Cancer Res. 18, Mann, M. The growing landscape of lysine acetylation
195. Zhang, H. et al. HIF‑1 regulates CD47 expression in 6748–6757 (2012). links metabolism and cell signalling. Nat. Rev. Mol.
breast cancer cells to promote evasion of phagocytosis 222. Menzies, S., Mc Menamin, M. & Barry, R. Lentigo Cell Biol. 15, 536–550 (2014).
and maintenance of cancer stem cells. Proc. Natl maligna successfully treated with combination therapy 248. Lobera, M. et al. Selective class IIa histone
Acad. Sci. USA 112, E6215–E6223 (2015). of topical tazarotene and imiquimod. Clin. Exp. deacetylase inhibition via a nonchelating zinc-binding
196. Betancur, P. A. et al. A CD47‑associated super- Dermatol. 42, 468–470 (2017). group. Nat. Chem. Biol. 9, 319–325 (2013).
enhancer links pro-inflammatory signalling to CD47 223. US National Library of Medicine. ClinicalTrials.gov 249. Guerriero, J. L. et al. Class IIa HDAC inhibition reduces
upregulation in breast cancer. Nat. Commun. 8, https://clinicaltrials.gov/ct2/show/NCT00319748 breast tumours and metastases through anti-tumour
14802 (2017). (2017). macrophages. Nature 543, 428–432 (2017).
197. Willingham, S. B. et al. The CD47‑signal regulatory 224. US National Library of Medicine. ClinicalTrials.gov This paper shows that class IIa HDAC inhibitors can
protein alpha (SIRPa) interaction is a therapeutic https://clinicaltrials.gov/ct2/show/NCT00095160 reprogramme TAMs to antitumoural macrophages
target for human solid tumors. Proc. Natl Acad. Sci. (2017). and increase the efficacy of chemotherapy and
USA 109, 6662–6667 (2012). 225. US National Library of Medicine. ClinicalTrials.gov immunotherapy in preclinical models of cancer.
198. Edris, B. et al. Antibody therapy targeting the CD47 https://clinicaltrials.gov/ct2/show/NCT00276159 250. Elomaa, O. et al. Cloning of a novel bacteria-binding
protein is effective in a model of aggressive metastatic (2017). receptor structurally related to scavenger receptors
leiomyosarcoma. Proc. Natl Acad. Sci. USA 109, 226. US National Library of Medicine. ClinicalTrials.gov and expressed in a subset of macrophages. Cell 80,
6656–6661 (2012). https://clinicaltrials.gov/ct2/show/NCT00189332 603–609 (1995).
199. Xiao, Z. et al. Antibody mediated therapy targeting (2008). 251. Bergamaschi, A. et al. Extracellular matrix signature
CD47 inhibits tumor progression of hepatocellular 227. US National Library of Medicine. ClinicalTrials.gov identifies breast cancer subgroups with different
carcinoma. Cancer Lett. 360, 302–309 (2015). https://clinicaltrials.gov/ct2/show/NCT00091689 clinical outcome. J. Pathol. 214, 357–367 (2008).
200. Liu, R. et al. CD47 promotes ovarian cancer (2008). 252. Georgoudaki, A. M. et al. Reprogramming tumor-
progression by inhibiting macrophage phagocytosis. 228. Dudek, A. Z. et al. First in human phase I trial of associated macrophages by antibody targeting
Oncotarget 8, 39021–39032 (2017). 852A, a novel systemic toll-like receptor 7 agonist, to inhibits cancer progression and metastasis. Cell Rep.
201. Weiskopf, K. et al. CD47‑blocking immunotherapies activate innate immune responses in patients with 15, 2000–2011 (2016).
stimulate macrophage-mediated destruction of small- advanced cancer. Clin. Cancer Res. 13, 7119–7125 253. Li, F. & Ravetch, J. V. Inhibitory Fcγ receptor
cell lung cancer. J. Clin. Invest. 126, 2610–2620 (2007). engagement drives adjuvant and anti-tumor activities of
(2016). 229. US National Library of Medicine. ClinicalTrials.gov agonistic CD40 antibodies. Science 333, 1030–1034
202. Alvey, C. M. et al. SIRPA-inhibited, marrow-derived https://clinicaltrials.gov/ct2/show/NCT00719199 (2011).
macrophages engorge, accumulate, and differentiate (2013). 254. Sotsios, Y. & Ward, S. G. Phosphoinositide 3‑kinase: a
in antibody-targeted regression of solid tumors. Curr. 230. US National Library of Medicine. ClinicalTrials.gov key biochemical signal for cell migration in response to
Biol. 27, 2065–2077 (2017). https://clinicaltrials.gov/ct2/show/NCT00633529 chemokines. Immunol. Rev. 177, 217–235 (2000).
203. Liu, J. et al. Pre-clinical development of a humanized (2013). 255. Hirsch, E. et al. Central role for G protein-coupled
anti‑CD47 antibody with anti-cancer therapeutic 231. US National Library of Medicine. ClinicalTrials.gov phosphoinositide 3‑kinase gamma in inflammation.
potential. PLOS ONE 10, e0137345 (2015). https://clinicaltrials.gov/ct2/show/NCT01040832 Science 287, 1049–1053 (2000).
204. Gholamin, S. et al. Disrupting the CD47‑SIRPα anti- (2017). 256. Kaneda, M. M. et al. PI3Kgamma is a molecular
phagocytic axis by a humanized anti‑CD47 antibody is 232. US National Library of Medicine. ClinicalTrials.gov switch that controls immune suppression. Nature
an efficacious treatment for malignant pediatric brain https://clinicaltrials.gov/ct2/show/NCT00729053 539, 437–442 (2016).
tumors. Sci. Transl Med. 9, eaaf2968 (2017). (2018). This study shows that PI3Kγ is a key regulator of
205. US National Library of Medicine. ClinicalTrials.gov 233. US National Library of Medicine. ClinicalTrials.gov TAM pro-tumoural activities and that its inhibition
https://clinicaltrials.gov/ct2/show/NCT02216409 https://clinicaltrials.gov/ct2/show/NCT01360827 contributes to TAM reprogramming and tumour
(2018). (2014). inhibition.
206. US National Library of Medicine. ClinicalTrials.gov 234. Smith, D. A. et al. Antitumor activity and safety of 257. Ha, M. & Kim, V. N. Regulation of microRNA bio-
https://clinicaltrials.gov/ct2/show/NCT02678338 combination therapy with the Toll-like receptor 9 genesis. Nat. Rev. Mol. Cell Biol. 15, 509–524 (2014).
(2018). agonist IMO‑2055, erlotinib, and bevacizumab in 258. Baer, C. et al. Suppression of microRNA activity
207. US National Library of Medicine. ClinicalTrials.gov advanced or metastatic non-small cell lung cancer amplifies IFN‑γ‑induced macrophage activation and
https://clinicaltrials.gov/ct2/show/NCT02953782 patients who have progressed following promotes anti-tumour immunity. Nat. Cell Biol. 18,
(2018). chemotherapy. Cancer Immunol. Immunother. 63, 790–802 (2016).
208. US National Library of Medicine. ClinicalTrials.gov 787–796 (2014). This is the first study showing reprogramming of
https://clinicaltrials.gov/ct2/show/NCT02953509 235. Khalil, M. & Vonderheide, R. H. Anti‑CD40 agonist TAMs to tumoricidal macrophages by the
(2018). antibodies: preclinical and clinical experience. Update modulation of miRNA activity.
209. Sikic, B. I. et al. A first‑in‑human, first‑in‑class phase I Cancer Ther. 2, 61–65 (2007). 259. Cannarile, M. A. et al. Colony-stimulating factor 1
trial of the anti‑CD47 antibody Hu5F9‑G4 in patients 236. van Kooten, C. & Banchereau, J. CD40‑CD40 ligand. receptor (CSF1R) inhibitors in cancer therapy.
with advanced cancers. J. Clin. Oncol. 34 (Suppl. 15), J. Leukoc. Biol. 67, 2–17 (2000). J. Immunother. Cancer 5, 53 (2017).
3019 (2016). 237. Hoves, S. et al. Rapid activation of tumor-associated 260. Krieg, C. et al. High-dimensional single-cell analysis
210. US National Library of Medicine. ClinicalTrials.gov macrophages boosts preexisting tumor immunity. predicts response to anti‑PD‑1 immunotherapy. Nat.
https://clinicaltrials.gov/ct2/show/NCT02367196 J. Exp. Med. 215, 859–876 (2018). Med. 24, 144–153 (2018).
(2018). 238. Perry, C. J. et al. Myeloid-targeted immunotherapies This study shows that the frequency of
211. US National Library of Medicine. ClinicalTrials.gov act in synergy to induce inflammation and antitumor CD14+CD16–HLA-DRhigh monocytes is a strong
https://clinicaltrials.gov/ct2/show/NCT02641002 immunity. J. Exp. Med. 215, 877–893 (2018). predictor of response to PD1 immunotherapy in
(2018). 239. Vonderheide, R. H. et al. Clinical activity and immune patients with melanoma.
212. Petrova, P. S. et al. TTI‑621 (SIRPαFc): a modulation in cancer patients treated with 261. Romano, E. et al. Ipilimumab-dependent cell-mediated
CD47‑blocking innate immune checkpoint inhibitor CP‑870,893, a novel CD40 agonist monoclonal cytotoxicity of regulatory T cells ex vivo by
with broad antitumor activity and minimal erythrocyte antibody. J. Clin. Oncol. 25, 876–883 (2007). nonclassical monocytes in melanoma patients. Proc.
binding. Clin. Cancer Res. 23, 1068–1079 (2017). 240. Vonderheide, R. H. et al. Phase I study of the CD40 Natl Acad. Sci. USA 112, 6140–6145 (2015).
213. US National Library of Medicine. ClinicalTrials.gov agonist antibody CP‑870,893 combined with 262. Gil Del Alcazar, C. R. et al. Immune escape in breast
https://clinicaltrials.gov/ct2/show/NCT02663518 carboplatin and paclitaxel in patients with advanced cancer during in situ to invasive carcinoma transition.
(2018). solid tumors. Oncoimmunology 2, e23033 (2013). Cancer Discov. 7, 1098–1115 (2017).
214. US National Library of Medicine. ClinicalTrials.gov 241. Nowak, A. K. et al. A phase 1b clinical trial of the 263. Harrer, D. C., Dörrie, J. & Schaft, N. Chimeric antigen
https://clinicaltrials.gov/ct2/show/NCT02890368 CD40‑activating antibody CP‑870,893 in receptors in different cell types: new vehicles join the
(2018). combination with cisplatin and pemetrexed in race. Hum. Gene Ther. 29, 547–558 (2018).
215. Medzhitov, R. & Janeway, C. The Toll receptor family malignant pleural mesothelioma. Ann. Oncol. 26, 264. Kulkarni, A. et al. A designer self-assembled
and microbial recognition. Trends Microbiol. 8, 2483–2490 (2015). supramolecule amplifies macrophage immune
452–456 (2000). 242. Beatty, G. L. et al. A phase I study of an agonist CD40 responses against aggressive cancer. Nat. Biomed.
216. Kaczanowska, S., Joseph, A. M. & Davila, E. TLR monoclonal antibody (CP‑870,893) in combination Eng. 2, 589–599 (2018).
agonists: our best frenemy in cancer immunotherapy. with gemcitabine in patients with advanced In this paper, the authors design a supramolecule
J. Leukoc. Biol. 93, 847–863 (2013). pancreatic ductal adenocarcinoma. Clin. Cancer Res. that inhibits CSF1R signalling and blocks the
217. Le Mercier, I. et al. Tumor promotion by intratumoral 19, 6286–6295 (2013). CD47–SIRPα pathway in macrophages; this
plasmacytoid dendritic cells is reversed by TLR7 243. US National Library of Medicine. ClinicalTrials.gov compound can reprogramme TAMs and exert
ligand treatment. Cancer Res. 73, 4629–4640 https://clinicaltrials.gov/ct2/show/NCT02304393 potent antitumour effects in aggressive models of
(2013). (2018). breast cancer and melanoma.

NATURE REVIEWS | DRUG DISCOVERY VOLUME 17 | DECEMBER 2018 | 903


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

265. Gopalakrishnan, V. et al. Gut microbiome modulates 272. Perdiguero, E. G. & Geissmann, F. The development 278. [No authors listed]. Technology quarterly: targeting
response to anti‑PD‑1 immunotherapy in melanoma and maintenance of resident macrophages. Nat. tumours. Treating cancer: progress on many fronts.
patients. Science 359, 97–103 (2018). Immunol. 17, 2–8 (2016). The Economist (London) https://www.economist.com/
266. Iida, N. et al. Commensal bacteria control cancer 273. Lavin, Y., Mortha, A., Rahman, A. & Merad, M. technology-quarterly/2017-09-16/treating-cancer (2017).
response to therapy by modulating the tumor Regulation of macrophage development and function 279. Gentles, A. J. et al. The prognostic landscape of genes
microenvironment. Science 342, 967–970 (2013). in peripheral tissues. Nat. Rev. Immunol. 15, 731–744 and infiltrating immune cells across human cancers.
267. Roy, S. & Trinchieri, G. Microbiota: a key orchestrator (2015). Nat. Med. 21, 938–945 (2015).
of cancer therapy. Nat. Rev. Cancer 17, 271–285 274. Ginhoux, F. & Jung, S. Monocytes and
(2017). macrophages: developmental pathways and tissue Acknowledgements
268. Veglia, F., Perego, M. & Gabrilovich, D. Myeloid- homeostasis. Nat. Rev. Immunol. 14, 392–404 The authors apologize to the many authors whose work they
derived suppressor cells coming of age. Nat. Immunol. (2014). could not cite owing to space constraints. Research in the
19, 108–119 (2018). 275. Mass, E. et al. Specification of tissue-resident authors’ laboratories is supported by the Wellcome Trust
269. de Haas, N., de Koning, C., Spilgies, L., de Vries, I. J. & macrophages during organogenesis. Science 353, (101067/Z/13/Z) and Medical Research Council (MRC) Centre
Hato, S. V. Improving cancer immunotherapy by aaf4238 (2016). grant MR/N022556/1 to J.W.P.
targeting the STATe of MDSCs. Oncoimmunology 5, 276. Cecchini, M. G. et al. Role of colony stimulating
e1196312 (2016). factor‑1 in the establishment and regulation of Author contributions
270. Schulz, C. et al. A lineage of myeloid cells independent tissue macrophages during postnatal development The authors contributed equally to all aspects of the article.
of Myb and hematopoietic stem cells. Science 336, of the mouse. Development 120, 1357–1372 Competing interests
86–90 (2012). (1994). The authors declare no competing interests.
271. Hoeffel, G. et al. C‑Myb(+) erythro-myeloid 277. Okabe, Y. & Medzhitov, R. Tissue-specific signals
progenitor-derived fetal monocytes give rise to adult control reversible program of localization and Publisher’s note
tissue-resident macrophages. Immunity 42, 665–678 functional polarization of macrophages. Cell 157, Springer Nature remains neutral with regard to jurisdictional
(2015). 832–844 (2014). claims in published maps and institutional affiliations.

904 | DECEMBER 2018 | VOLUME 17 www.nature.com/nrd


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like