Download as pdf or txt
Download as pdf or txt
You are on page 1of 24

Downloaded from http://perspectivesinmedicine.cshlp.

org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

The Immune Microenvironment and Cancer


Metastasis

Asmaa El-Kenawi,1,3 Kay Hänggi,1,3 and Brian Ruffell1,2


1
Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
2
Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa,
Florida 33612, USA
Correspondence: Brian.Ruffell@moffitt.org

The dynamic interplay between neoplastic cells and the immune microenvironment regu-
lates every step of the metastatic process. Immune cells contribute to invasion by secreting a
cornucopia of inflammatory factors that promote epithelial-to-mesenchymal transition and
remodeling of the stroma. Cancer cells then intravasate to the circulatory system assisted by
macrophages and use several pathways to avoid recognition by cytotoxtic lymphocytes and
phagocytes. Circulating tumor cells that manage to adhere to the vasculature and encounter
premetastic niches are able to use the associated myeloid cells to extravasate into ectopic
organs and establish a dormant microscopic colony. If successful at avoiding repetitive
immune attack, dormant cells can subsequently grow into overt, clinically detectable meta-
www.perspectivesinmedicine.org

static lesions, which ultimately account to most cancer-related deaths. Understanding how
disseminated tumor cells evade and corrupt the immune system during the final stages of
metastasis will be pivotal in developing new therapeutic modalities that combat metastasis.

M etastasis is a multistep process character-


ized by the spread of malignant cells to
distant organs. This is evolutionary driven by
or vascular system (survival, arrest at distant
site, extravasation), and survive and adjust to a
new microenvironment (colonization and out-
genetic and/or epigenetic alteration within can- growth) (Lambert et al. 2017). For cancer cells to
cer cells but is also dependent on an intricate migrate through tissue, they acquire several abil-
interaction with stromal cells at a local and sys- ities that act in concert to promote invasion,
temic level (Fig. 1). Tumor cells must overcome including cytoskeletal reorganization, secretion
several hurdles to establish macroscopic metas- of proteases, and altered adhesion receptor-
tasis, collectively referred to as the invasion– ligand interactions (Kessenbrock et al. 2010;
metastatic cascade (Valastyan and Weinberg Quail and Joyce 2013). In addition, cancer
2011). During this process, carcinoma cells cells can undergo a reversible program termed
deviate from their primary growth site (local epithelial to mesenchymal transition (EMT)
invasion and intravasation), disseminate sys- through which loss of epithelial polarization
temically via circulation through the lymphatic and intercellular adhesion allows for motility

3
These authors contributed equally to this work.
Editors: Jeffrey W. Pollard and Yibin Kang
Additional Perspectives on Metastasis: Mechanism to Therapy available at www.perspectivesinmedicine.org
Copyright © 2020 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a037424
Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424

1
Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

A. El-Kenawi et al.

(1) (2) (3) (4)

Angiogenesis

ROS
Mechanisms

Extracellular modelling
Oxygen
EMT

T cell Monocyte Macrophage Neutrophil DC ECM

Figure 1. Establishment of the tumor immune microenvironment: Environmental insults or spontaneous genetic
alterations in epithelial cells can lead to immune surveillance (1) or the growth of premalignant cells that evade
clearance by innate and adaptive immune cells (2). In addition to driving cell proliferation, certain oncogenic
alterations are associated with the release of cytokines that recruit additional immune cells to aid neoplastic
progression by providing mitogenic factors or producing reactive oxygen species that result in the accumulation
www.perspectivesinmedicine.org

of additional genetic mutations (3). Uncontrolled cellular growth at this stage causes environmental changes such
as reduced oxygen levels (hypoxia), extracellular matrix (ECM) turnover, and stromal disruption (4). These
signals of tissue damage result in a pathological healing response that includes the creation of a disordered and
permeable vascular network (angiogenic switch), as well as morphological and transcriptional changes in
epithelial cells that reflect a mesenchymal phenotype (EMT). Malignant cells are thus able to migrate toward
and intravasate into the vascular lumen, becoming circulating tumor cells.

and invasiveness (Thiery et al. 2009). Neoplastic ESTABLISHMENT OF THE TUMOR IMMUNE
cells leaving the primary tumor and intravasat- MICROENVIRONMENT
ing into circulation must then survive severe
physical shear stress, attack by the immune sys- The tumor immune microenvironment (TIME)
tem, and oxidative stress (Labelle et al. 2011; Le promotes every aspect of carcinogenesis, includ-
Gal et al. 2015). Once cells arrest at a distant site ing initiation, survival, growth, metastasis, and
or are physically trapped in minute capillaries, immune evasion. Depending on the tissue of
they can proliferate within the intravascular origin, the TIME shows a unique immune rep-
space or extravasate by modifying the endothe- ertoire, with different proportions and function-
lium (Al-Mehdi et al. 2000; Deneve et al. 2013; al states of lymphocytes, granulocytes, mono-
Strilic and Offermanns 2017). Only a small frac- cytes/macrophages, and dendritic cells (DCs)
tion of cells that disseminate from the primary (Gentles et al. 2015). Although several of these
tumor will manage to invade and adapt to their immune populations have the potential to erad-
new surrounding (Nagrath et al. 2007). Thus, icate malignant cells, numerous factors in the
the ability of neoplastic cells to co-opt immune tumor act to blunt this activity or redirect the
processes to aid in these steps and evade im- cells toward promoting tumorigenesis. In addi-
mune recognition are necessary for successful tion to relative proportions, the locations of
metastasis (Sosa et al. 2014; Massagué and Obe- immune cells are associated with disease pro-
nauf 2016). gression. For example, sites of focal myoepithe-

2 Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

Immune Microenvironment and Cancer Metastasis

lial cell layer dysregulation in ductal carcinoma sion of lymphocytes (Kortlever et al. 2017).
in situ (DCIS) display an enrichment of leuko- Collectively, these data establish an important
cytes, suggesting a role in driving or sustaining role for tumor genotype in driving the immune
local invasion (Gil Del Alcazar et al. 2017), repertoire in cancer.
whereas in late stage disease the exclusion of T There are also several studies showing that
lymphocytes from tumor beds promotes resis- cells undergoing EMT modify the TIME through
tance to immune checkpoint blockade (Jayapra- chemokine secretion (Dongre and Weinberg
kash et al. 2018; Jerby-Arnon et al. 2018). The 2019). The EMT master regulator SNAIL pro-
degree to which parallel programs are used to motes expression of CCL2 and CCL5, leading
establish the metastatic immune microenviron- to macrophage recruitment (Hsu et al. 2014),
ment (MIME) are largely unknown, with the and expression of CXCR2 ligands leading to re-
exception of myeloid cell recruitment to estab- cruitment of suppressive myeloid cells (Taki
lish the premetastatic niche (Liu and Cao 2016). et al. 2018). Other mediators of epithelial mes-
enchymal transition, such as SRC-1, promote
macrophage recruitment via expression of CSF-
Tumor Intrinsic Signaling
1 (Qin et al. 2009; Wang et al. 2009), and mesen-
In addition to the type of cancer, immune com- chymal tumors display prominent macrophage
position correlates with the underlying genetic infiltration (Dongre et al. 2017). The ability of
alterations driving carcinogenesis (Rooney et al. EMT to promote an immunosuppressive envi-
2015), as oncogenic signals in transformed cells ronment is likely further augmented by high pro-
are associated with the release of chemokines duction of transforming growth factor (TGF)-β,
that attract immune cells to the tumor (Wellen- which can lead to the exclusion of lymphocytes
stein and de Visser 2018). As one of the most from tumor beds (Mariathasan et al. 2018; Taur-
www.perspectivesinmedicine.org

frequently mutated genes in cancer, loss of iello et al. 2018) and regulate the activation state
Trp53 activates the NF-κB pathway, stimulates of most immune cells. This has been shown in
cytokines release from cancer cells, which one study with overexpression of SNAIL, which
through paracrine interactions modify the im- leads to production of TGF-β and thrombospon-
mune landscape of cancer (Meylan et al. 2009; din-1, and increased conversion of regulatory T
Kastenhuber and Lowe 2017). Similarly, the ac- cells (Tregs) (Kudo-Saito et al. 2009). Given the
tivation of the transcription factor MYC endows critical role of EMT in the metastatic process it
cancer cells with the ability to overproduce po- will be important to determine if these pathways
tent proinflammatory cytokines that facilitate are also involved in establishing the MIME.
tumor angiogenesis and recruitment of protu-
moral mast cells, macrophages, and neutrophils
Homeostatic Imbalance
(Soucek et al. 2007), whereas activation of the
oncogenic EGFR family of receptor tyrosine Solid tumors show large regions with low oxy-
kinases results in secretion of CSF-1, a chemo- gen levels (hypoxia) and nutrient insufficiency
tactic and survival factor for macrophages. (Gatenby and Gillies 2004). Hypoxia has the
Interestingly, oncogenic Kras G12D drives unique ability to modulate immune functions directly
programs in different cancer types, although by activating HIF-1α-dependent transcriptional
each cumulates in the recruitment of an im- changes within the immune cells, or indirectly
munosuppressive myeloid population. This in- by rewiring tumor cell secreted metabolites and
cludes expression of GM-CSF and CXCL3 in cytokine repertoire (Cairns et al. 2007). For
pancreatic cancer and colon carcinoma, respec- example, hypoxia was shown to trigger HIF-
tively (Pylayeva-Gupta et al. 2012; Liao et al. 1α-dependent immunosuppressive activity in
2019), as well as CCL9 expression in lung cancer macrophages (Doedens et al. 2010). Through
in the context of Myc activation (Kortlever et al. indirect effects on the tumor and the stroma,
2017). Oncogenic Kras and Myc also cooperate hypoxia also causes the overproduction of
to induce interleukin (IL)-23-dependent exclu- monocyte recruitment factors including CCL2,

Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424 3


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

A. El-Kenawi et al.

CCL5, CXC-chemokine ligand 12 (CXCL12), blasts can suppress a T-cell response through a
colony-stimulating factor (CSF-1), and vascular myriad of pathways, including direct suppres-
endothelial growth factor (VEGF) (Murdoch sion via PD-L2 and FAS ligand expression (La-
et al. 2004). As an indirect consequence of hyp- kins et al. 2018), expression of cytokines (Kato
oxia, tumor cells up-regulate glycolysis and se- et al. 2018), and recruitment of immuno-
crete lactic acid, which induces phenotypic suppressive myeloid cells (Kumar et al. 2017).
changes and immunosuppressive function in Cancer cells can even create gap junctions with
macrophages (Colegio et al. 2014). Lactate-acti- astrocytes to transfer the second messenger
vated macrophages in turn enhance the glyco- cGAMP to astrocytes, activating the STING
lytic phenotype of breast cancer cells through the pathway and production of type I interferons
release of extracellular vesicles containing a HIF- (Chen et al. 2016). It should also be noted that
1α-stabilizing long noncoding RNA (Chen et al. changes in the physical properties of the stroma
2019). The acidic microenvironment of tumors further modulate the TIME (DeNardo and Ruf-
also directly regulates macrophage phenotype, fell 2019). For example, higher levels of collagen
as seen following the genetic ablation of the (Wesley et al. 1998; Stahl et al. 2013; Pickup et al.
macrophage pH sensor, Gpr132, or the buffer- 2014) and collagen cross-linking (Van Goethem
ing of tumor secreted acids, which reduces the et al. 2010; McWhorter et al. 2015) enhance the
spontaneous lung metastasis of breast cancer protumoral phenotype of macrophages, and in-
cells and prostate cancer invasiveness, respec- hibiting the hyperactivation of focal adhesion
tively (Chen et al. 2017a; El-Kenawi et al. kinase (FAK) is sufficient to alter macrophage
2019). Finally, areas of hypoxia frequently coin- polarization and increase response to immune
cide with regions of cell death, leading to recog- checkpoint blockade (Jiang et al. 2016).
nition of apoptotic cells by phagocytes and/or There is also an elaborate interplay between
www.perspectivesinmedicine.org

the release of damage-associated molecular pat- immune cells in the tumor that controls the phe-
terns (DAMPs) during secondary necrosis. notype and functional role of the cells. Most
Phosphatidylserine recognition receptors thus well established is the ability of macrophages,
promote myeloid-dependent immune suppres- monocytes, neutrophils, and other myeloid popu-
sion in the TIME (Cook et al. 2013; Crittenden lations to suppress the cytotoxic function of lym-
et al. 2016; Ubil et al. 2018); whereas several phocytes, through mechanisms that have been
DAMPs have been shown to activate DCs and reviewed elsewhere (Veglia et al. 2018; DeNardo
elicit a cytotoxic T-cell response (Kroemer et al. and Ruffell 2019). As will be discussed, myeloid
2013). cells are a critical component of the immuno-
suppressive TIME and MIME, and depleting
these cells can reduce metastasis by unleashing
Immune-Stroma Crosstalk
T and natural killer (NK) cell cytotoxicity. Sim-
In addition to regulating the immune compart- ilarly, Tregs promote pulmonary metastasis by
ment directly, tumor cells drive phenotypic blocking NK cell function (Olkhanud et al.
changes in the nonimmune stromal cells, in- 2009), with Treg conversion occurring through
cluding fibroblasts, endothelial cells, and peri- the activity of Bregs (Olkhanud et al. 2011). T and
cytes. This “reactive” stroma possesses a unique B lymphocytes can also promote the protumori-
gene/protein expression profile that differs from genic properties of macrophages (Gunderson
the signature of the corresponding normal tissue and Coussens 2013), including IL-4-producing
(Finak et al. 2008; Nonn et al. 2009) and is cre- CD4+ T cells enhancing the ability of macro-
ated, in part, by the influence of inflammatory phages to foster invasion (DeNardo et al. 2009).
mediators and cytokines. For example, TGF-β
cooperates with SDF-1 (CXCL12) to generate
INVASION AND INTRAVASATION
myofibroblasts with enhanced tumor-promot-
ing capability through autocrine signaling loops Cancer cells must acquire phenotypic changes
(Kojima et al. 2010). Cancer-associated fibro- that endow them with ability to invade sur-

4 Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

Immune Microenvironment and Cancer Metastasis

rounding stroma as a precursor to intravasation. whereas in a model of Her2+ breast cancer


These invasive characteristics include alter- Tie2+ macrophages initiate EMT through ex-
ations in expression of adhesion molecules and pression of Wnt (Linde et al. 2018). This asso-
expression of proteases to degrade the extracel- ciation in breast cancer drives dissemination in
lular matrix (ECM). Each of these characteristics the absence of a palpable lesion (Linde et al.
is fuelled by the presence of immune cells and 2018), and thus may also explain the impact of
can potentially cross-regulate each other. For macrophage ablation on the invasive phenotype
example, EMT leads to the recruitment of mac- in a model of Trp53 null breast cancer (Carron
rophages, which can secrete and activate TGF-β, et al. 2017). Intriguingly, macrophage expres-
further supporting the induction of EMT (Don- sion of IL-35 at the site of metastasis has recently
gre and Weinberg 2019). Similar feedback loops been shown to promote mesenchymal-to-epi-
are required for intravasation, with tumor cells thelial transition (Lee et al. 2018), highlighting
stimulating the chemotactic and angiogenic the critical role of macrophages at multiple stag-
properties of macrophages (Lewis et al. 2016). es of the metastatic process.
Interestingly, recent studies have shown that Immature monocytes and neutrophils have
cells egressing the primary site early during can- also been shown to contribute to EMT through
cer progression are more metastasis-efficient the production of TGF-β, epidermal growth
than cells leaving the tumor at later stages (Hü- factor (EGF), and hepatocyte growth factor
semann et al. 2008; Ghajar and Bissell 2016; (HGF), and these cells are found around the
Harper et al. 2016; Hosseini et al. 2016). The invasive edge of tumors in which markers of
metastatic competency of early disseminated EMT are most prevalent (Toh et al. 2011; San-
cells—which would harbor fewer mutations— galetti et al. 2016; Ouzounova et al. 2017; Wang
reiterates that microenvironmental factors con- et al. 2019), with neutrophil levels at the inva-
www.perspectivesinmedicine.org

tributes to the acquisition of metastatic traits sive margin of gastric cancer being an indepen-
(Hendry et al. 2017; Linde et al. 2018). In sup- dent, negative predictor of disease-free survival
port of this, although the molecular profiles of (Li et al. 2019). Neutrophils have also been
invasive breast cancer and preinvasive was in- shown to contribute to EMT by creating abnor-
distinguishable, aggressive lesions displayed mal vasculature in lung tumors, resulting in
high infiltration of macrophages and Tregs hypoxia and induction of a Snail-dependent
(Abba et al. 2015; Nelson et al. 2018). EMT program (Faget et al. 2017). Given an
analogous role for macrophages in breast can-
cer (Stockmann et al. 2008), it may be that
Epithelial-to-Mesenchymal Transition
macrophages promote EMT through the same
An established TIME provides several cytokines mechanism.
and chemokines with the potential to activate Although it has not been extensively evalu-
latent EMT programming in epithelial cells, ated, there are also a few reports suggesting that
causing decreased E-cadherin expression, loss T cells can activate EMT. In vitro, coculturing
of cell–cell junction and, morphological chang- premalignant cells with CD4+ T cells decreased
es that enable cells to invade and migrate to dis- E-cadherin expression and elevated expression
tant sites (Dongre and Weinberg 2019). This can of mesenchymal proteins (Goebel et al. 2015;
be mediated through cooperation with the TGF- Chen et al. 2017b). Whether this is mediated
β signaling pathway, one of the primary induces by TGF-β production by Tregs is not clear (Li
of EMT (Lu et al. 2014; Su et al. 2014; Chockley et al. 2007; Worthington et al. 2012). One study
and Keshamouni 2016), although independent also observed an in vivo role for CD8+ T cells
mechanisms have also been described. For ex- involving selection of Her2/neu escape variants
ample, in a model of gastric cancer, macrophage (Santisteban et al. 2009). However, this may be
production of CXCL1 and CXCL5 increase Snail an artifact of this particular model system, and a
transcription through activation of a CXCR2/ role for T cells in vessel normalization (Motz
STAT3 feed-forward loop (Zhou et al. 2019), et al. 2014; Schmittnaegel et al. 2017; Tian

Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424 5


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

A. El-Kenawi et al.

et al. 2017) suggests these cells could actually structure of the ECM to facilitate the creation of
reduce hypoxia and EMT in other settings. “tracks,” or act through some combination
thereof (Condeelis and Segall 2003).
Extracellular Matrix Remodeling
Intravasation
The ECM is a dynamic, acellular, three-dimen-
sional structure that serves as scaffold, on-de- Angiogenesis is a hallmark of cancer, and angio-
mand reservoir for growth factors, and ligand genic factors such as VEGF, fibroblast growth
for cellular adhesion. The ECM undergoes a factor (FGF), and placental-derived growth fac-
continuous process of remodeling that is tightly tor (PlGF) are produced by immune cells in the
regulated by the MMP and cathepsin family of tumor, especially macrophages (Rivera and
proteases (Bonnans et al. 2014). Not surpris- Bergers 2015), which are critical for the angio-
ingly, ECM remodeling is highly abnormal genic switch during tumor development (Lin
in tumors, with increased deposition of key et al. 2006). Macrophage production of these
components such as collagen and fibronectin, factors does not necessarily regulate the extent
increased degradation and turnover, and an ab- of angiogenesis in late stage tumors, as VEGF
normal structure marked by higher density and overexpression can bypass a requirement for
tensile force (Northey et al. 2017). Navigating macrophages (Lin et al. 2007). However, macro-
the ECM is thus critical for cancer cells to invade phages retain their importance as regulators of
tissue and gain access to the vasculature. vascular structure and permeability (Stockmann
Studies documenting a role for immune cells et al. 2008). Thus ablating macrophages produc-
in ECM deposition are limited, but production tion of VEGF increases pericyte coverage, im-
of SPARC (secreted protein, acidic rich in cys- proves tissue perfusion, and increases tumor
www.perspectivesinmedicine.org

teine) by macrophages leads to increases in growth (Stockmann et al. 2008), while simulta-
collagen and fibronectin, along with higher lev- neously increasing susceptibility to chemother-
els of metastasis (Sangaletti et al. 2008). Inflam- apy (Stockmann et al. 2008; Hughes et al. 2015)
matory monocytes have also been shown to elic- and reducing intravsation and metastasis (Har-
it FXIIIA-mediated fibrin cross-linking, which ney et al. 2015). This phenotype is driven largely
provides a scaffold for local tumor cell invasion by a population of Tie2+CXCR4+ macrophages
(Porrello et al. 2018). Conversely, production of that reside in the perivascular space around ves-
interferon (IFN)-γ by NK cells restricts metas- sels (De Palma et al. 2005; Welford et al. 2011),
tasis by inducing expression of fibronectin allowing for local production of VEGF to in-
(Glasner et al. 2018), highlighting the complex crease vessel permeability and ease transendo-
association of ECM location and structure with thelial migration (Harney et al. 2015). Targeting
the invasive process. A number of stromal pro- these cells by blocking the ligand for Tie2, an-
teases are also known to promote local tumor giopoietin (Ang)-2, thus also results in reduced
invasion, including urokinase/plasminogen ac- metastasis (Mazzieri et al. 2011). Although not
tivator (uPA), MMP-9 and cathepsin B, S, or Z necessarily related to the metastatic cascade, it is
(Almholt et al. 2005; Vasiljeva et al. 2006; Yang worth noting that vessel normalization pro-
et al. 2008; Gocheva et al. 2010; Yan et al. 2010; motes T-cell infiltration, which then maintains
Bekes et al. 2011; Akkari et al. 2014). Most vessel normalization by producing IFN-γ (Motz
of these proteases are expressed primarily by et al. 2014; Schmittnaegel et al. 2017; Tian et al.
tumor macrophages, with the exception of 2017).
MMP-9, which is also expressed by neutrophils Macrophages are also responsible for re-
and mast cells (Coussens et al. 2000; Bekes et al. cruiting tumor cells to these sites of vascular
2011). However, it is currently unclear whether permeability through a well-described paracrine
these proteases act by clearing out the ECM to loop involving epidermal growth factor (EGF).
permit invasion, release factors that promote an As mentioned, tumor cells are an important
invasive phenotype (e.g., active TGF-β), alter the source of CSF-1, and this induces the produc-

6 Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

Immune Microenvironment and Cancer Metastasis

tion of EGF in macrophages, resulting in tumor es that are linked to the recruitment of neutro-
cell chemotaxis toward the vasculature (Wyck- phils and monocytes/macrophages.
off et al. 2004, 2007; Wang et al. 2009; Ishihara
et al. 2013). This process is not mediated by
Avoiding Cytotoxic Cells
Tie2+ perivascular macrophages, but rather the
recently recruited population of monocyte- Increased NK cell cytotoxicity or enhanced ex-
derived macrophages that forms the dominant pression of NK cell activation receptors are as-
population within tumors (Arwert et al. sociated with good prognosis for patients at risk
2018). After entering the tumor parenchyma, of metastatic disease (López-Soto et al. 2017).
these macrophages up-regulate expression of For example, the infiltration of NK cells express-
CXCR4 and are slowly recruited back to the ing high levels of the activating receptors NCR1/
vasculature via CXCL12-expressing fibroblasts, NKp46 (natural cytotoxicity triggering receptor
creating an EGF chemotactic gradient and path- 1) and NKG2D (killer cell lectin-like receptor
way of altered ECM that directs cancer cell mi- K1) was positively associated with recurrence-
gration to the vasculature (Arwert et al. 2018). free survival in prostate cancer following surgi-
Finally, endothelial cell interactions promote cal resection (Pasero et al. 2016). Several studies
differentiation into the Tie2+CXCR4+ perivas- have shown that NKp46 expression in particular
cular macrophage population that facilitates is critical for NK cell recognition of metastatic
transendothelial migration. Thus, interfering cells (Halfteck et al. 2009; Lakshmikanth et al.
with monocyte recruitment (CCR2-CCL2), sur- 2009; Elboim et al. 2010; Glasner et al. 2012) and
vival and differentiation (CSF1R-CSF1), migra- loss of NK cells increases metastasis (Smyth
tion to the vasculature (CXCR4-CXCL12), or et al. 1999; Bidwell et al. 2012). Although it is
maturation to perivascular cells (Tie2-Ang2) difficult to distinguish between a role for NK
www.perspectivesinmedicine.org

functions to severely limit the ability of tumor cells within the primary tumor, circulation, or
cells to intravasate into the blood stream (Qian metastatic sites, at least some experimental evi-
and Pollard 2010; Kitamura et al. 2015). As with dence exists for the direct killing of CTCs (Pal-
vascular normalization, the ability of macro- umbo et al. 2005; Hanna et al. 2015), and poor
phages to promote tumor cell chemotaxis is infiltration by NK cells in the absence of treat-
controlled by infiltrating T cells, with IL-4-ex- ment likely limits their role in most primary
pressing CD4+ T cells augmenting EGF expres- tumors (Böttcher et al. 2018).
sion by macrophages (DeNardo et al. 2009). Not surprisingly, tumors use multiple mech-
anisms to avoid recognition by NK cells (Box 1).
First and foremost, down-regulated protein or
surface expression of NK activating ligands, es-
SURVIVAL AND EXTRAVASATION
pecially MHC I polypeptide-related sequence A
Most primary tumors release millions of cells (MICA), and MICB, can reduce NK cell recog-
into the blood stream, but only a small number nition and killing of tumor cells (Nausch and
of metastatic lesions usually develop, indicating Cerwenka 2008; Wang et al. 2014). Conversely,
the inefficiency of tumor cell dissemination tumor cells can secrete soluble versions of these
(Nagrath et al. 2007). In particular, circulating same molecules to suppress NK cell immuno-
tumor cells (CTCs) have to resist physical shear surveillance (Schlecker et al. 2014; Zhang et al.
stress caused by blood flow, death by anoikis, 2015). This occurs within the primary tumor,
and avoid recognition by cytotoxic lymphocytes but is sufficient to drive a systemic reduction
and phagocytic macrophages (Fig. 2) (Mohme in the cognate receptors on NK cells (Pasero
et al. 2017). With this limited capacity to survive et al. 2016), and high levels of these soluble li-
within the vasculature, the capacity of CTCs to gands in the circulation correlate with disease
adhere to the vasculature and migrate into the progression (Paschen et al. 2009; Yamaguchi
extravascular space is also central to their met- et al. 2012). That said, a high-affinity NKG2D
astatic potential (Reymond et al. 2013), process- ligand (MULT1) has also been shown to stimu-

Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424 7


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

A. El-Kenawi et al.

Survival in the blood stream


Endothelial cell (EC) layer

Cancer cell
1
3
Apoptotic
cancer cell

4
2 Neutrophil

Platelets

Adherence extravasation
Monocyte

Macrophage

Activating
receptor
5 6 7 8
NK cell
Inhibitory
receptor

Figure 2. Immune evasion and assistance during cancer cell dissemination. During circulation within the
www.perspectivesinmedicine.org

intravascular space, major histocompatibility complex (MHC) I expression protects by circulating tumor cells
(CTCs) from natural killer (NK) cell recognition (1), whereas down-regulation of MHC I and up-regulation of
NK cell activating ligands can lead to cytotoxicity (2). Conversely, recognition can be antagonized via up-
regulation of NK cell inhibitory ligands (3) or physical shielding using platelet/fibrin coagulates (4). To extrav-
asate into the tissue, CTCs interact with endothelial cells via selectins, cadherins, integrins, CD44, junctional
adhesion molecules (5). Neutrophils are able to assist this process via ICAM-1 and VCAM-1 interactions, as
well as through the release of NETs that trap CTCs and hide the cells from lympohcytes (6). Recruitment of
CCR2+Ly6C+ monocytes (7) results in local production of VEGF, thereby increasing vascular permeability to
facilitate CTC extravasation (8).

late NK cell-mediated killing of tumor cells after and this can promote survival, EMT, and adhe-
shedding (Deng et al. 2015), so there may be a sion to the endothelium (Gay and Felding-Hab-
cost to this approach in some instances. Second, ermann 2011; Labelle et al. 2011). This also has
although HLA down-regulation is a common the effect of physically masking CTCs from NK
mechanism of evading T-cell recognition (Cam- cells (Camerer et al. 2004; Palumbo et al. 2005)
poli and Ferrone 2008), HLA expression is never and/or transferring platelet-derived membrane
completely lost, likely because of the selective vesicles containing MHC I molecules that act to
pressure of NK cells. Instead, HLA loss of het- “normalize” the tumor cells (Placke et al. 2012).
erozygosity is commonly observed as an alter- Additionally, platelet-derived TGF-β has been
native to loss of neoantigen expression, allowing shown to induce down-regulation of NKG2D
tumors to limit detection by both of the cytotox- to prevent NK cell reactivity (Kopp et al.
ic lymphocyte populations simultaneously 2009). Cumulatively, platelets and their associ-
(McGranahan et al. 2017; Rosenthal et al. 2019). ated factors promote metastatic spread, a phe-
There is also a complex interaction between notype that is dependent on the presence of NK
tumor cells, platelets, and NK cells that regulates cells in most models (Camerer et al. 2004; Pal-
cytoxicity in the blood. Tumor cells become umbo et al. 2005; Palumbo 2008; Turpin et al.
coated with platelets on entering the circulation, 2014; Adams et al. 2015).

8 Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

Immune Microenvironment and Cancer Metastasis

BOX 1. NONCLASSICAL MHC SIGNALING

Histocompatibility antigen class I G (HLA-G) is a human leukocyte antigen that belongs to the
“nonclassical” HLA-class Ib molecules (Choo 2007). HLA-Ib molecules have minimal polymor-
phisms compared with the HLA-Ia molecules and are involved in immune modulation rather than
antigen presentation (Kievits et al. 1987; Braud et al. 1997). HLA-G has been described to contribute
to immune evasion in cancer (Lin and Yan 2015) and expression correlates with poor patient survival
in melanoma, colorectal, hepatocellular, breast, glioma, ovarian, and lung cancers (Cai et al. 2009;
de Kruijf et al. 2010; He et al. 2010; Guo et al. 2015). Moreover, soluble HLA-G is secreted by the
primary tumor were identified and negatively correlate with patient outcome (Contini et al. 2003;
Zheng et al. 2014). HLA-G can bind to a variety of receptors, such as CD8, leukocyte immunoglob-
ulin-like receptor subfamily B member 1 (LIR-1), and killer cell immunoglobulin-like receptor (KIR),
all of which protect cancer cells from CD8 T cell- or NK cell-induced killing (Wiendl et al. 2002;
Kochan et al. 2013; Loumagne et al. 2014; Lin and Yan 2015). HLA-G polymorphisms also impact
graft-versus-tumor responses in renal cell carcinoma (Crocchiolo et al. 2018) and correlate with
outcome in epithelial ovarian carcinoma (Schwich et al. 2019).

Avoiding Phagocytosis metastasis and decreased survival (Baccelli


et al. 2013, 2014). Taken together, the evidence
CD47 is a surface protein belonging to the im-
favors CD47 expression reducing clearance by
munoglobulin superfamily that regulates a
monocytes/macrophages. The question remains
broad range of cellular functions, including
whether this clearance is relevant within the in-
adhesion, migration, proliferation, apoptosis
www.perspectivesinmedicine.org

travascular or extravascular space. It seems un-


and—most relevant for metastasis—recogni-
likely that monocytes and CTCs are interacting
tion by phagocytes via signal regulatory protein
while they are both in circulation. However,
α (SIRPα) (Reinhold et al. 1995; Jiang et al.
nonclassical Ly6C− monocytes actively patrol
1999). SIRPα is expressed on monocytes, mac-
the endothelial lumen (Auffray et al. 2007; Car-
rophages, and CD11b+ classical dendritic cells
lin et al. 2013) and rapidly take up cellular debris
(cDCs), and on binding to CD47 the phagocyt-
(Headley et al. 2016), which induces chemokine
ic activity of these cells is blocked, thereby act-
expression and recruitment of NK cells to limit
ing as a “don’t eat me” signal (Jaiswal et al.
metastasis (Hanna et al. 2015). Whether these
2009). CD47 is ubiquitously expressed on
patrolling monocytes also phagocytose live cells
mouse and human myeloid leukemias, thereby
is unclear, and it may be that phagocytosis in the
allowing them to evade clearance and populate
extravascular space or tissue parenchyma plays a
organs with an abundance of macrophages
more critical role in restricting metastasis.
(Jaiswal et al. 2009) and hinting that the same
process may allow for CTCs to colonize ectopic
organs.
Adhesion to the Endothelium
Certainly, CD47 is overexpressed in many
solid tumors (Liu et al. 2017), and it can protect Circulating neutrophils correlate with poor
cancer stem cells from elimination during con- prognosis (Teramukai et al. 2009; Lee et al.
ventional antitumor therapies, which in turn 2012; Gondo et al. 2013), and although this
increases the probability of recurrence (Solta- may partially reflect altered hematopoiesis and
nian and Matin 2011). Gene expression profil- the establishment of an immunosuppressive mi-
ing of primary tumors and CTCs from patients croenvironment (Coffelt et al. 2015), it has also
with colorectal cancers also found pronounced become increasingly clear that circulating neu-
expression of CD47 (Steinert et al. 2014), where- trophils promote the conversion of CTCs to
as the presence of CD47+MET+CTCs in breast disseminated tumor cells (DTCs) through two
cancer patients was associated with increased distinct mechanisms. The first involves binding

Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424 9


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

A. El-Kenawi et al.

between integrins expressed by neutrophils (i.e., in circulation (Szczerba et al. 2019) and the abil-
CD11b/CD18, VLA-4) and immunoglobulin ity of tumor cells to induce the release of
superfamily ligands expressed by endothelial NETs109, it will be interesting to determine if
cells and CTCs (ICAM-1, VCAM-1). This al- this alters the dynamics of CTC retention and/
lows neutrophils to form a cellular bridge for or allows CTCs to create their own protective
CTCs to adhere to the endothelium (Box 2) shell.
(Huh et al. 2010; Spicer et al. 2012). These inter-
actions also induce “outside-in” signaling that
Extravasation
promotes the proliferative capacity of CTCs, en-
hancing their metastatic potential (Szczerba Ly6C+CCR2HI monocytes are recruited to the
et al. 2019). As the majority of CTCs are associ- site of CTC arrest via a classical multistep ad-
ated with neutrophils—at least in breast cancer hesion cascade involving selectins, chemokines,
patients—this appears to be an important mech- and integrin ligands (Qian et al. 2011; Ferjančič
anism underlying metastatic spread (Szczerba et al. 2013; Häuselmann et al. 2016). In addition
et al. 2019). to CCL2 emanating from the tissue, cancer cell
The other major mechanism by which neu- production of CCL2 is necessary for this pro-
trophils promote metastasis is through the re- cess (Qian et al. 2011; Häuselmann et al. 2016),
lease of neutrophil extracellular traps (NETs) suggesting that it initiates the inflammatory
containing their nuclear DNA (Park et al. cascade. Reminiscent of the role of macrophag-
2016). Originally described to trap CTCs during es in tumor cell intravasation, monocytes re-
injury or infection (Tohme et al. 2016), it has also cruited to the endothelial lumen via CCL2
been shown that release of NETs can be induced secrete VEGF and increase endothelial perme-
by the presence of CTCs in the lungs, leading to ability, thereby enhancing transmigration of tu-
www.perspectivesinmedicine.org

an increase in micrometastasis (Cools-Lartigue mor cells (Qian et al. 2011). Interfering with the
et al. 2013; Najmeh et al. 2017). Beyond simply CCR2/CCL2 chemokine axis thereby reduces
sequestering CTCs, NETs inhibit NK cell-medi- liver and lung metastasis in multiple murine
ated cytotoxicity, and the release of factors such model systems (Qian et al. 2011; Sanford et al.
as IL-1B, MMP-9, and HMGB1 promotes ex- 2013), provided that the mice are left on treat-
travasation (Spiegel et al. 2016; Tohme et al. ment for the duration of the study (Bonapace
2016). Importantly, the formation of NETs in et al. 2014). Platelet-bound CTCs can also re-
postoperative patients was linked to a reduction cruit monocytes to promote metastasis, but the
in survival, highlighting the potential clinical rel- degree to which this process involves CCL2
evance of this mechanism (Tohme et al. 2016). and/or VEGF is unknown (Gil-Bernabe et al.
Given the prevalence of CTC-neutrophil clusters 2012).

BOX 2. TUMOR–ENDOTHELIAL CELL INTERACTIONS

CTCs becoming trapped in the capillaries is thought to be one of the major mechanisms of arrest in the
circulation, but the process of adhesion to the endothelial layer and migration into the tissue is an
active process (Chambers et al. 2002; Wirz et al. 2008; Strilic and Offermanns 2017). The vascular
endothelium differs structurally depending on the organ, and as a result, leukocyte extravasation in
lung and liver occurs in the microvasculature, whereas it occurs in the postcapillary venules in the
skin, muscle, and mesentery (Aird 2007; Strell and Entschladen 2008). However, although the pro-
cesses of adhesion and transmigration (diapedesis) are at least partially shared between leukocytes
and tumor cells (Miles et al. 2008), the molecules described for tumor cell transmigration vary and are
often tumor-specific (Bendas and Borsig 2012; Strilic and Offermanns 2017). However, it is still not
entirely clear the degree to which all of these molecules are involved in tumor cell adhesion/trans-
migration in vivo, or whether different tumor subclones may use unique strategies to colonize different
tissues (Caswell and Swanton 2017).

10 Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

Immune Microenvironment and Cancer Metastasis

ECTOPIC GROWTH used to achieve a similar goal, but presumably


there are subtleties in the activation state of the
Outgrowth of metastatic colonies represents the
recruited myeloid cells, as well as alternations in
final stage of disease and is responsible for the
the functions of other stromal cells that play a
majority of cancer-related deaths, and unfortu-
complementary and supportive role in estab-
nately, there is evidence that most patients have
lishing the niche (McAllister and Weinberg
DTCs at the time of diagnosis (Friberg and Ny-
2014; Peinado et al. 2017).
strom 2015; Massagué and Obenauf 2016).
These DTCs are largely in a dormant state and
thus resistant to conventional cytotoxic chemo- Escaping Dormancy
therapies, resulting in metastatic recurrence for
There is only limited evidence that immune cells
many patients long after the primary tumor has
can promote the survival and growth of DTCs
been removed. Addressing the issue of DTCs, as
(Qian and Pollard 2010), with macrophages
well as the treatment of overt metastasis, are
shown to provide survival signals via binding
therefore the two most critical areas of cancer
to VCAM-1 (Chen et al. 2011) and neutrophils
research (Sosa et al. 2014; Massagué and Obe-
enhancing tumorigenicity via leukotriene pro-
nauf 2016). Thankfully, it is becoming clear that
duction (Wculek and Malanchi 2015). However,
modulating the immune response has the
macrophages in primary tumors promote ther-
potential to address both of these issues, with
apeutic resistance by producing factors such as
the approval of immune checkpoint blockade
TNF-α and IL-6 (Ruffell and Coussens 2015),
agents for metastatic disease being the most sig-
which activate signaling pathways known to
nificant advancement.
promote the proliferation of dormant cells
(Massagué and Obenauf 2016). Thus, it is likely
www.perspectivesinmedicine.org

that these and other pathways are important at


Formation of the Premetastatic Niche
the metastatic site, and that our limited knowl-
The premetastatic niche was originally de- edge in this area reflects experimental difficulties
scribed as VEGFR1-expressing myeloid cells ac- evaluating the early stages of colonization. This
cumulating at sites of distant metastasis before includes the use of murine cancer cell lines that
the arrival of tumor cells (Kaplan et al. 2005), do not display dormancy or extended periods of
and is usually defined by the recruitment of latency, and evaluation of metastasis primarily
macrophages and neutrophils (Fig. 3) (Liu and to the lung even though there is evidence of a
Cao 2016). The formation of the niche is driven tissue-specific role for critical molecules such as
by a range of tumor and stroma-derived soluble TGF-β (Massagué and Obenauf 2016). Indeed,
factors (e.g., G-CSF, VEGF, CCL2, TNF-α, TGF- using a model of metastatic dormancy, it was
β, S100A8/A9) in addition to the release of recently shown that NETs promote metastasis
tumor-derived extracellular vesicles (usually via release of neutrophil elastase and MMP9,
containing microRNAs [mRNAs]) (Liu and resulting in cleavage of the ECM component
Cao 2016). Regardless of the molecules identi- laminin (Albrengues et al. 2018). Rather than
fied or the mechanism proposed, each appears activating myeloid cells via TLRs, as has been
to act primarily by inducing the recruitment of described for versican in the premetastatic niche
myeloid cells. For example, S100A8/A9 pro- (Kim et al. 2009), cleaved laminin triggered tu-
motes the release of serum amyloid A3, which mor cell proliferation via integrin activation and
activates an inflammatory program in macro- FAK signaling (Albrengues et al. 2018).
phages via toll-like receptor (TLR)4 (Hiratsuka Other studies that have addressed these
et al. 2008). These cells then promote the ability experimental issues have noted a clear role for
of DTCs to become established by generating an lymphocytes in maintaining tumor dormancy.
immunosuppressive and/or permissive micro- CD8+ T-cell-depletion dramatically increased
environment, as we will discuss below. It is un- the rate of metastasis in a model of spontaneous
clear why so many divergent approaches are uveal melanoma (Eyles et al. 2010) and following

Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424 11


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

A. El-Kenawi et al.

Preparation of the pre-


1
metastatic niche Cancer cell

Apoptotic
cancer cell

Extracellular vesicles/
exosomes
Growth factors, cytokines
chemokines and MMPs

Neutrophil
2 Colonization

Monocyte

3 Outgrowth Macrophage

NK cell

T lymphocyte
www.perspectivesinmedicine.org

Figure 3. Establishment of the metastatic immune microenvironment: The primary tumor can actively prime
organs by forming a metastasis-permissive microenvironment before the arrival of disseminated tumor cells
(DTCs) (1). This premetastatic niche includes recruited monocytes, macrophages, and neutrophils, as well as
tumor-secreted factors such as cytokines, chemokines, growth factors, and proteases. Enhanced vascular leak-
iness, reorganization of the stroma and extracellular matrix, and the establishment of an immunosuppressive
environment are hallmarks of the metastatic microenvironment (2). These allow DTCs to evade natural killer
(NK) cell immunity and colonize the ectopic organ as single cells or micrometastatic lesions. Micrometastatic
lesions often enter a period of dormancy, as they are ill-adapted to the new microenvironment, encounter growth
suppressive factors such as TGF-β, and are subjected to NK cell-mediated immune surveillance. Outgrowth
involves tumor-intrinsic changes alongside the induction of angiogenesis and the creation of a mature immu-
nosuppressive microenvironment to evade recognition by cytotoxic lymphocytes (3).

the surgical resection of implanted fibrosarcoma via suppression of angiogenic pathways (Müller-
(Romero et al. 2014). Interestingly, although Hermelink et al. 2008; Andreu et al. 2010).
CD8+ T-cell-depletion resulted in a 100% rate Critically, a functional immune system prevents
of metastasis, NK cell-depletion led to a rate of disease recurrence in humans, as seen from the
87%, and CD4+ T-cell-depletion resulted in a unfortunate development of metastasis follow-
rate of 23% (Romero et al. 2014). This may reflect ing kidney transplantations from donors with a
a complex interplay between these cell types in previous history of melanoma or lung cancer
maintaining dormancy, or simply the produc- (MacKie et al. 2003; Xiao et al. 2013).
tion of a common set of cytokines such as
IFN-γ and TNF-α. Certainly, the incidence of
Evading Immunity
primary tumors is suppressed by these cytokines
(Müller-Hermelink et al. 2008), either through Leaving the immunosuppressive microenviron-
cytostatic/cytotoxic effects on carcinoma cells or ment of the primary tumor exposes metastatic

12 Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

Immune Microenvironment and Cancer Metastasis

cells to recognition by cytotoxic lymphocytes, as SUMMARY


seen from the ability of NK cells to protect
The mechanisms underlying early steps of the
against experimental and spontaneous metasta-
metastatic process have been relatively well stud-
sis but not primary tumor growth (Smyth et al.
ied compared with colonization and immune
1999, 2001; Takeda et al. 2001; Olkhanud et al.
evasion, despite early dissemination of tumor
2009; Milsom et al. 2013; Paolino et al. 2014).
cells making these later steps more clinically
Thus, it is not surprising that successful metas-
relevant. The critical role for T cell- and NK
tasis is linked to the formation of a myeloid-rich
cell-mediated surveillance in restraining the
environment (de Mingo Pulido and Ruffell
growth of metastatic lesions represents a unique
2016). What remains largely unexplored is the
therapeutic opportunity as agents that promote
degree to which immunosuppressive mecha-
the activity of these cells become available. In
nisms used by metastatic cells recapitulate those
addition to approved antagonist antibodies
found in the primary tumor, represent the
against CTLA-4 and PD-1/PD-L1, agents block-
unique biology of the ectopic organ, and/or re-
ing LAG-3, TIGIT, and TIM-3 are in early phase
flect an incipient microenvironment. For exam-
clinical trials (Anderson et al. 2016), whereas
ple, in a model of lobular breast cancer, primary
several new targets have been identified, includ-
tumors drive neutrophil expansion and an im-
ing those that unleash NK cell activity (André
munosuppressive phenotype via initiation of an
et al. 2018). Combining these agents with those
IL-1β/IL-17/G-CSF cascade, which acts to blunt
that block the inhibitory function of myeloid
CD8+ T-cell-dependent rejection of early meta-
cells, or activate their immunostimulatory po-
static lesions (Coffelt et al. 2015). Interfering
tential, have already shown potential in murine
with any component of this pathway reversed
models of primary tumors (DeNardo and Ruf-
this immune suppression, but had no impact
www.perspectivesinmedicine.org

fell 2019).
on any measured parameter within primary tu-
Beyond the treatment of overt metastatic le-
mors. This discrepancy may relate to the size of
sions, the role of lymphocytes in maintaining
the lesion, as the complexity of an advanced
dormancy suggests the possibility of developing
microenvironment would make it resistant to
immunotherapies in an adjuvant setting. Al-
perturbations in a single pathway and late stage
though long-term treatment is difficult and
metastatic lesions were not impacted by neutro-
expensive, the potential benefit of preventing
phil depletion (Coffelt et al. 2015). However, it
recurrence would be immense, and the use of a
has also been described that macrophages in pri-
minimal effective dose could improve the viabil-
mary tumors indirectly suppress a CD8+ T-cell-
ity of this approach. Alternatively, understand-
response through production of IL-10, whereas
ing how the immune system promotes escape
their ability to limit T-cell activity in metastatic
from dormancy could allow for the develop-
tumors occurs through a different mechanism of
ment of therapies targeting systemic inflam-
action (Ruffell et al. 2014), such as the produc-
mation. Recent technological developments
tion of reactive oxygen species or expression of
including CRISPR/Cas9 screens, single cell ge-
CTLA4 ligands (Kitamura et al. 2018). Con-
nomics, and mass cytometry will allow progress
versely, the clinical success of immune check-
to be made in this area, and dissecting how life
point blockade in metastatic and primary
events such as inflammatory diseases, infec-
tumors, including in relatively immune privi-
tions, and injury relate to recurrence-free sur-
leged sites, highlights the importance of tumor
vival should allow findings in murine models to
immunogenicity (Hodi et al. 2014; Topalian
be correlated with patient data.
et al. 2014; Overman et al. 2018). Thus, intrinsic,
extrinsic, and temporal factors appear to regulate
the vulnerability of metastatic lesions to immune
ACKNOWLEDGMENTS
surveillance, and understanding these factors
should assist in the development of immuno- K.H. was supported by a Postdoctoral Fellow-
therapies for patients with metastatic disease. ship from the Swiss National Science Founda-

Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424 13


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

A. El-Kenawi et al.

tion (P400PM_183881). B.R. was supported by tion from migratory to perivascular macrophage is re-
quired for tumor cell intravasation. Cell Rep 23: 1239–
the Florida Breast Cancer Foundation, the Flo- 1248. doi:10.1016/j.celrep.2018.04.007
rida Department of Health Bankhead-Coley Auffray C, Fogg D, Garfa M, Elain G, Join-Lambert O, Kayal
Cancer Research Program (8BC02), and spon- S, Sarnacki S, Cumano A, Lauvau G, Geissmann F. 2007.
sored research agreements with Tesaro, Inc. Monitoring of blood vessels and tissues by a population of
monocytes with patrolling behavior. Science 317: 666–
670. doi:10.1126/science.1142883
REFERENCES Baccelli I, Schneeweiss A, Riethdorf S, Stenzinger A, Schillert
A, Vogel V, Klein C, Saini M, Bäuerle T, Wallwiener M, et
Abba MC, Gong T, Lu Y, Lee J, Zhong Y, Lacunza E, Butti M, al. 2013. Identification of a population of blood circulat-
Takata Y, Gaddis S, Shen J, et al. 2015. A molecular por- ing tumor cells from breast cancer patients that initiates
trait of high-grade ductal carcinoma in situ. Cancer Res metastasis in a xenograft assay. Nat Biotechnol 31: 539–
75: 3980–3990. doi:10.1158/0008-5472.CAN-15-0506 544. doi:10.1038/nbt.2576
Adams GN, Rosenfeldt L, Frederick M, Miller W, Waltz D, Baccelli I, Stenzinger A, Vogel V, Pfitzner BM, Klein C, Wall-
Kombrinck K, McElhinney KE, Flick MJ, Monia BP, Re- wiener M, Scharpff M, Saini M, Holland-Letz T, Sinn HP,
venko AS, et al. 2015. Colon cancer growth and dissem- et al. 2014. Co-expression of MET and CD47 is a novel
ination relies upon thrombin, stromal PAR-1, and fibrin- prognosticator for survival of luminal breast cancer pa-
ogen. Cancer Res 75: 4235–4243. doi:10.1158/0008-5472 tients. Oncotarget 5: 8147–8160. doi:10.18632/oncotarget
.CAN-15-0964 .2385
Aird WC. 2007. Phenotypic heterogeneity of the endotheli- Bekes EM, Schweighofer B, Kupriyanova TA, Zajac E, Ardi
um: I. Structure, function, and mechanisms. Circ Res 100: VC, Quigley JP, Deryugina EI. 2011. Tumor-recruited
158–173. doi:10.1161/01.RES.0000255691.76142.4a neutrophils and neutrophil TIMP-free MMP-9 regulate
coordinately the levels of tumor angiogenesis and effi-
Akkari L, Gocheva V, Kester JC, Hunter KE, Quick ML,
ciency of malignant cell intravasation. Am J Pathol 179:
Sevenich L, Wang HW, Peters C, Tang LH, Klimstra
1455–1470. doi:10.1016/j.ajpath.2011.05.031
DS, et al. 2014. Distinct functions of macrophage-derived
and cancer cell-derived cathepsin Z combine to promote Bendas G, Borsig L. 2012. Cancer cell adhesion and metas-
tumor malignancy via interactions with the extracellular tasis: Selectins, integrins, and the inhibitory potential of
matrix. Genes Dev 28: 2134–2150. doi:10.1101/gad heparins. Int J Cell Biol 2012: 676731. doi:10.1155/2012/
www.perspectivesinmedicine.org

.249599.114 676731
Albrengues J, Shields MA, Ng D, Park CG, Ambrico A, Bidwell BN, Slaney CY, Withana NP, Forster S, Cao Y, Loi S,
Poindexter ME, Upadhyay P, Uyeminami DL, Pommier Andrews D, Mikeska T, Mangan NE, Samarajiwa SA, et al.
A, Kuttner V, et al. 2018. Neutrophil extracellular traps 2012. Silencing of Irf7 pathways in breast cancer cells
produced during inflammation awaken dormant cancer promotes bone metastasis through immune escape. Nat
cells in mice. Science 361: eaao4227. doi:10.1126/science Med 18: 1224–1231. doi:10.1038/nm.2830
.aao4227 Bonapace L, Coissieux MM, Wyckoff J, Mertz KD, Varga Z,
Al-Mehdi AB, Tozawa K, Fisher AB, Shientag L, Lee A, Junt T, Bentires-Alj M. 2014. Cessation of CCL2 inhibi-
Muschel RJ. 2000. Intravascular origin of metastasis tion accelerates breast cancer metastasis by promoting
from the proliferation of endothelium-attached tumor angiogenesis. Nature 515: 130–133. doi:10.1038/na
cells: A new model for metastasis. Nat Med 6: 100–102. ture13862
doi:10.1038/71429 Bonnans C, Chou J, Werb Z. 2014. Remodelling the extra-
Almholt K, Lund LR, Rygaard J, Nielsen BS, Danø K, Rømer cellular matrix in development and disease. Nat Rev Mol
J, Johnsen M. 2005. Reduced metastasis of transgenic Cell Biol 15: 786–801. doi:10.1038/nrm3904
mammary cancer in urokinase-deficient mice. Int J Can- Böttcher JP, Bonavita E, Chakravarty P, Blees H, Cabeza-
cer 113: 525–532. doi:10.1002/ijc.20631 Cabrerizo M, Sammicheli S, Rogers NC, Sahai E, Zelenay
Anderson AC, Joller N, Kuchroo VK. 2016. Lag-3, Tim-3, S, Reis e Sousa C. 2018. NK cells stimulate recruitment of
and TIGIT: Co-inhibitory receptors with specialized cDC1 into the tumor microenvironment promoting can-
functions in immune regulation. Immunity 44: 989– cer immune control. Cell 172: 1022–1037.e14. doi:10
1004. doi:10.1016/j.immuni.2016.05.001 .1016/j.cell.2018.01.004
André P, Denis C, Soulas C, Bourbon-Caillet C, Lopez J, Braud V, Yvonne Jones E, McMichael A. 1997. The human
Arnoux T, Bléry M, Bonnafous C, Gauthier L, Morel A, major histocompatibility complex class Ib molecule
et al. 2018. Anti-NKG2A mAb is a checkpoint inhibitor HLA-E binds signal sequence-derived peptides with pri-
that promotes anti-tumor immunity by unleashing both mary anchor residues at positions 2 and 9. Eur J Immunol
T and NK cells. Cell 175: 1731–1743.e13. doi:10.1016/j 27: 1164–1169. doi:10.1002/eji.1830270517
.cell.2018.10.014 Cai MY, Xu YF, Qiu SJ, Ju MJ, Gao Q, Li YW, Zhang BH,
Andreu P, Johansson M, Affara NI, Pucci F, Tan T, Junankar Zhou J, Fan J. 2009. Human leukocyte antigen-G protein
S, Korets L, Lam J, Tawfik D, DeNardo DG, et al. 2010. expression is an unfavorable prognostic predictor of he-
FcRγ activation regulates inflammation-associated squa- patocellular carcinoma following curative resection. Clin
mous carcinogenesis. Cancer Cell 17: 121–134. doi:10 Cancer Res 15: 4686–4693. doi:10.1158/1078-0432.CCR-
.1016/j.ccr.2009.12.019 09-0463
Arwert EN, Harney AS, Entenberg D, Wang Y, Sahai E, Cairns RA, Papandreou I, Sutphin PD, Denko NC. 2007.
Pollard JW, Condeelis JS. 2018. A unidirectional transi- Metabolic targeting of hypoxia and HIF1 in solid tumors

14 Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

Immune Microenvironment and Cancer Metastasis

can enhance cytotoxic chemotherapy. Proc Natl Acad Sci LJAC, Jonkers J, et al. 2015. IL-17-producing γδ T cells
104: 9445–9450. doi:10.1073/pnas.0611662104 and neutrophils conspire to promote breast cancer me-
Camerer E, Qazi AA, Duong DN, Cornelissen I, Advincula tastasis. Nature 522: 345–348. doi:10.1038/nature14282
R, Coughlin SR. 2004. Platelets, protease-activated recep- Colegio OR, Chu NQ, Szabo AL, Chu T, Rhebergen AM,
tors, and fibrinogen in hematogenous metastasis. Blood Jairam V, Cyrus N, Brokowski CE, Eisenbarth SC, Phillips
104: 397–401. doi:10.1182/blood-2004-02-0434 GM, et al. 2014. Functional polarization of tumour-asso-
Campoli M, Ferrone S. 2008. HLA antigen changes in ma- ciated macrophages by tumour-derived lactic acid. Nature
lignant cells: Epigenetic mechanisms and biologic signifi- 513: 559–563. doi:10.1038/nature13490
cance. Oncogene 27: 5869–5885. doi:10.1038/onc.2008 Condeelis J, Segall JE. 2003. Intravital imaging of cell move-
.273 ment in tumours. Nat Rev Cancer 3: 921–930. doi:10
Carlin LM, Stamatiades EG, Auffray C, Hanna RN, Glover L, .1038/nrc1231
Vizcay-Barrena G, Hedrick CC, Cook HT, Diebold S, Contini P, Ghio M, Poggi A, Filaci G, Indiveri F, Ferrone S,
Geissmann F. 2013. Nr4a1-dependent Ly6Clow mono- Puppo F. 2003. Soluble HLA-A,-B,-C and -G molecules
cytes monitor endothelial cells and orchestrate their dis- induce apoptosis in T and NK CD8+ cells and inhibit
posal. Cell 153: 362–375. doi:10.1016/j.cell.2013.03.010 cytotoxic T cell activity through CD8 ligation. Eur J Im-
Carron EC, Homra S, Rosenberg J, Coffelt SB, Kittrell F, munol 33: 125–134. doi:10.1002/immu.200390015
Zhang Y, Creighton CJ, Fuqua SA, Medina D, Machado Cook RS, Jacobsen KM, Wofford AM, DeRyckere D, Stan-
HL. 2017. Macrophages promote the progression of pre- ford J, Prieto AL, Redente E, Sandahl M, Hunter DM,
malignant mammary lesions to invasive cancer. Oncotar- Strunk KE, et al. 2013. MerTK inhibition in tumor leu-
get 8: 50731–50746. doi:10.18632/oncotarget.14913 kocytes decreases tumor growth and metastasis. J Clin
Caswell DR, Swanton C. 2017. The role of tumour hetero- Invest 123: 3231–3242. doi:10.1172/JCI67655
geneity and clonal cooperativity in metastasis, immune Cools-Lartigue J, Spicer J, McDonald B, Gowing S, Chow S,
evasion and clinical outcome. BMC Med 15: 133. doi:10 Giannias B, Bourdeau F, Kubes P, Ferri L. 2013. Neutro-
.1186/s12916-017-0900-y phil extracellular traps sequester circulating tumor cells
Chambers AF, Groom AC, MacDonald IC. 2002. Dissemi- and promote metastasis. J Clin Invest 123: 3446–3458.
nation and growth of cancer cells in metastatic sites. Nat doi:10.1172/JCI67484
Rev Cancer 2: 563–572. doi:10.1038/nrc865 Coussens LM, Tinkle CL, Hanahan D, Werb Z. 2000. MMP-
Chen Q, Zhang XH, Massagué J. 2011. Macrophage binding 9 supplied by bone marrow-derived cells contributes to
skin carcinogenesis. Cell 103: 481–490. doi:10.1016/
www.perspectivesinmedicine.org

to receptor VCAM-1 transmits survival signals in breast


cancer cells that invade the lungs. Cancer Cell 20: 538– S0092-8674(00)00139-2
549. doi:10.1016/j.ccr.2011.08.025 Crittenden MR, Baird J, Friedman D, Savage T, Uhde L,
Chen Q, Boire A, Jin X, Valiente M, Er EE, Lopez-Soto A, Alice A, Cottam B, Young K, Newell P, Nguyen C, et al.
Jacob L, Patwa R, Shah H, Xu K, et al. 2016. Carcinoma- 2016. Mertk on tumor macrophages is a therapeutic target
astrocyte gap junctions promote brain metastasis by to prevent tumor recurrence following radiation therapy.
cGAMP transfer. Nature 533: 493–498. doi:10.1038/na Oncotarget 7: 78653–78666. doi:10.18632/oncotarget
ture18268 .11823
Chen P, Zuo H, Xiong H, Kolar MJ, Chu Q, Saghatelian A, Crocchiolo R, Ringden O, Bay JO, Blaise D, Omasic B, Mazzi
Siegwart DJ, Wan Y. 2017a. Gpr132 sensing of lactate B, Picard C, Trinca S, Barkholt L, Peccatori J, et al. 2018.
mediates tumor–macrophage interplay to promote breast Impact of HLA-G polymorphism on the outcome of al-
cancer metastasis. Proc Natl Acad Sci 114: 580–585. logeneic hematopoietic stem cell transplantation for met-
doi:10.1073/pnas.1614035114 astatic renal cell carcinoma. Bone Marrow Transplant 53:
Chen Q, Yang D, Zong H, Zhu L, Wang L, Wang X, Zhu X, 213–218. doi:10.1038/bmt.2017.243
Song X, Wang J. 2017b. Growth-induced stress enhances de Kruijf EM, Sajet A, van Nes JGH, Natanov R, Putter
epithelial-mesenchymal transition induced by IL-6 in H, Smit VTHBM, Liefers GJ, van den Elsen PJ, van de
clear cell renal cell carcinoma via the Akt/GSK-3β/β-cat- Velde CJH, Kuppen PJK. 2010. HLA-E and HLA-G ex-
enin signaling pathway. Oncogenesis 6: e375. doi:10.1038/ pression in classical HLA class I-negative tumors is of
oncsis.2017.74 prognostic value for clinical outcome of early breast can-
Chen F, Chen J, Yang L, Liu J, Zhang X, Zhang Y, Tu Q, Yin cer patients. J Immunol 185: 7452–7459. doi:10.4049/jim
D, Lin D, Wong PP, et al. 2019. Extracellular vesicle-pack- munol.1002629
aged HIF-1α-stabilizing lncRNA from tumour-associat- de Mingo Pulido A, Ruffell B. 2016. Immune regulation of
ed macrophages regulates aerobic glycolysis of breast can- the metastatic process: Implications for therapy. Adv
cer cells. Nat Cell Biol 21: 498–510. doi:10.1038/s41556- Cancer Res 132: 139–163. doi:10.1016/bs.acr.2016.05.004
019-0299-0 DeNardo DG, Ruffell B. 2019. Macrophages as regulators of
Chockley PJ, Keshamouni VG. 2016. Immunological conse- tumour immunity and immunotherapy. Nat Rev Immu-
quences of epithelial–mesenchymal transition in tumor nol 19: 369–382. doi:10.1038/s41577-019-0127-6
progression. J Immunol 197: 691–698. doi:10.4049/jim DeNardo DG, Barreto JB, Andreu P, Vasquez L, Tawfik D,
munol.1600458 Kolhatkar N, Coussens LM. 2009. CD4+ T cells regulate
Choo SY. 2007. The HLA system: Genetics, immunology, pulmonary metastasis of mammary carcinomas by en-
clinical testing, and clinical implications. Yonsei Med J hancing protumor properties of macrophages. Cancer
48: 11–23. doi:10.3349/ymj.2007.48.1.11 Cell 16: 91–102. doi:10.1016/j.ccr.2009.06.018
Coffelt SB, Kersten K, Doornebal CW, Weiden J, Vrijland K, Deneve E, Riethdorf S, Ramos J, Nocca D, Coffy A, Daures
Hau C-S, Verstegen NJM, Ciampricotti M, Hawinkels JP, Maudelonde T, Fabre JM, Pantel K, Alix-Panabieres C.

Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424 15


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

A. El-Kenawi et al.

2013. Capture of viable circulating tumor cells in the liver Gatenby RA, Gillies RJ. 2004. Why do cancers have high
of colorectal cancer patients. Clin Chem 59: 1384–1392. aerobic glycolysis? Nat Rev Cancer 4: 891–899. doi:10
doi:10.1373/clinchem.2013.202846 .1038/nrc1478
Deng W, Gowen BG, Zhang L, Wang L, Lau S, Iannello A, Xu Gay LJ, Felding-Habermann B. 2011. Contribution of plate-
J, Rovis TL, Xiong N, Raulet DH. 2015. Antitumor im- lets to tumour metastasis. Nat Rev Cancer 11: 123–134.
munity. A shed NKG2D ligand that promotes natural doi:10.1038/nrc3004
killer cell activation and tumor rejection. Science 348: Gentles AJ, Newman AM, Liu CL, Bratman SV, Feng W, Kim
136–139. doi:10.1126/science.1258867 D, Nair VS, Xu Y, Khuong A, Hoang CD, et al. 2015. The
De Palma M, Venneri MA, Galli R, Sergi Sergi L, Politi LS, prognostic landscape of genes and infiltrating immune
Sampaolesi M, Naldini L. 2005. Tie2 identifies a hemato- cells across human cancers. Nat Med 21: 938–945.
poietic lineage of proangiogenic monocytes required for doi:10.1038/nm.3909
tumor vessel formation and a mesenchymal population of Ghajar CM, Bissell MJ. 2016. Pathways of parallel progres-
pericyte progenitors. Cancer Cell 8: 211–226. doi:10.1016/ sion. Nature 540: 528–529. doi:10.1038/nature21104
j.ccr.2005.08.002
Gil-Bernabe AM, Ferjancic S, Tlalka M, Zhao L, Allen PD,
Doedens AL, Stockmann C, Rubinstein MP, Liao D, Zhang Im JH, Watson K, Hill SA, Amirkhosravi A, Francis JL, et
N, DeNardo DG, Coussens LM, Karin M, Goldrath AW, al. 2012. Recruitment of monocytes/macrophages by tis-
Johnson RS. 2010. Macrophage expression of hypoxia- sue factor-mediated coagulation is essential for metastatic
inducible factor-1α suppresses T-cell function and pro- cell survival and premetastatic niche establishment in
motes tumor progression. Cancer Res 70: 7465–7475. mice. Blood 119: 3164–3175. doi:10.1182/blood-2011-
doi:10.1158/0008-5472.CAN-10-1439 08-376426
Dongre A, Weinberg RA. 2019. New insights into the mech- Gil Del Alcazar CR, Huh SJ, Ekram MB, Trinh A, Liu LL,
anisms of epithelial-mesenchymal transition and impli- Beca F, Zi X, Kwak M, Bergholtz H, Su Y, et al. 2017.
cations for cancer. Nat Rev Mol Cell Biol 20: 69–84. doi:10 Immune escape in breast cancer during in situ to invasive
.1038/s41580-018-0080-4 carcinoma transition. Cancer Discov 7: 1098–1115. doi:10
Dongre A, Rashidian M, Reinhardt F, Bagnato A, Keckesova .1158/2159-8290.CD-17-0222
Z, Ploegh HL, Weinberg RA. 2017. Epithelial-to-mesen- Glasner A, Ghadially H, Gur C, Stanietsky N, Tsukerman
chymal transition contributes to immunosuppression in P, Enk J, Mandelboim O. 2012. Recognition and preven-
breast carcinomas. Cancer Res 77: 3982–3989. doi:10 tion of tumor metastasis by the NK receptor NKp46/
.1158/0008-5472.CAN-16-3292 NCR1. J Immunol 188: 2509–2515. doi:10.4049/jimmu
www.perspectivesinmedicine.org

Elboim M, Gazit R, Gur C, Ghadially H, Betser-Cohen G, nol.1102461


Mandelboim O. 2010. Tumor immunoediting by NKp46. Glasner A, Levi A, Enk J, Isaacson B, Viukov S, Orlanski S,
J Immunol 184: 5637–5644. doi:10.4049/jimmunol Scope A, Neuman T, Enk CD, Hanna JH, et al. 2018.
.0901644 NKp46 receptor-mediated interferon-γ production by
El-Kenawi A, Gatenbee C, Robertson-Tessi M, Bravo R, natural killer cells increases fibronectin 1 to alter tumor
Dhillon J, Balagurunathan Y, Berglund A, Visvakarma architecture and control metastasis. Immunity 48: 107–
N, Ibrahim-Hashim A, Choi J, et al. 2019. Acidity pro- 119.e4. doi:10.1016/j.immuni.2017.12.007
motes tumor progression by altering macrophage pheno- Gocheva V, Wang HW, Gadea BB, Shree T, Hunter KE,
type in prostate cancer. Br J Cancer. doi:10.1038/s41416- Garfall AL, Berman T, Joyce JA. 2010. IL-4 induces ca-
019-0542-2 thepsin protease activity in tumor-associated macrophag-
Eyles J, Puaux AL, Wang X, Toh B, Prakash C, Hong M, Tan es to promote cancer growth and invasion. Genes Dev 24:
TG, Zheng L, Ong LC, Jin Y, et al. 2010. Tumor cells 241–255. doi:10.1101/gad.1874010
disseminate early, but immunosurveillance limits meta- Goebel L, Grage-Griebenow E, Gorys A, Helm O, Genrich G,
static outgrowth, in a mouse model of melanoma. J Clin Lenk L, Wesch D, Ungefroren H, Freitag-Wolf S, Sipos B,
Invest 120: 2030–2039. doi:10.1172/JCI42002 et al. 2015. CD4+ T cells potently induce epithelial-mes-
Faget J, Groeneveld S, Boivin G, Sankar M, Zangger N, Gar- enchymal-transition in premalignant and malignant pan-
cia M, Guex N, Zlobec I, Steiner L, Piersigilli A, et al. 2017. creatic ductal epithelial cells-novel implications of CD4+
Neutrophils and Snail orchestrate the establishment of a T cells in pancreatic cancer development. Oncoimmunol-
pro-tumor microenvironment in lung cancer. Cell Rep 21: ogy 4: e1000083. doi:10.1080/2162402X.2014.1000083
3190–3204. doi:10.1016/j.celrep.2017.11.052 Gondo T, Nakashima J, Ohno Y, Hashimoto T, Takizawa I,
Ferjančič Š, Gil-Bernabé AM, Hill SA, Allen PD, Richardson Sakamoto N, Horiguchi Y, Aoyagi T, Ohori M, Tachibana
P, Sparey T, Savory E, McGuffog J, Muschel RJ. 2013. M. 2013. Preoperative prediction of malignant involve-
VCAM-1 and VAP-1 recruit myeloid cells that promote ment of resected ureters in patients undergoing radical
pulmonary metastasis in mice. Blood 121: 3289–3297. cystectomy for bladder cancer. Int J Urol 20: 501–506.
doi:10.1182/blood-2012-08-449819 doi:10.1111/j.1442-2042.2012.03203.x
Finak G, Bertos N, Pepin F, Sadekova S, Souleimanova M, Gunderson AJ, Coussens LM. 2013. B cells and their medi-
Zhao H, Chen H, Omeroglu G, Meterissian S, Omeroglu ators as targets for therapy in solid tumors. Exp Cell Res
A, et al. 2008. Stromal gene expression predicts clinical 319: 1644–1649. doi:10.1016/j.yexcr.2013.03.005
outcome in breast cancer. Nat Med 14: 518–527. doi:10 Guo ZY, Lv YG, Wang L, Shi SJ, Yang F, Zheng GX, Wen
.1038/nm1764 WH, Yang AG. 2015. Predictive value of HLA-G and
Friberg S, Nystrom A. 2015. Cancer metastases: Early dis- HLA-E in the prognosis of colorectal cancer patients.
semination and late recurrences. Cancer Growth Metas- Cell Immunol 293: 10–16. doi:10.1016/j.cellimm.2014
tasis 8: 43–49. doi:10.4137/CGM.S31244 .10.003

16 Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

Immune Microenvironment and Cancer Metastasis

Halfteck GG, Elboim M, Gur C, Achdout H, Ghadially H, of macrophages. Cancer Cell 26: 534–548. doi:10.1016/j
Mandelboim O. 2009. Enhanced in vivo growth of lym- .ccell.2014.09.002
phoma tumors in the absence of the NK-activating recep- Hughes R, Qian BZ, Rowan C, Muthana M, Keklikoglou I,
tor NKp46/NCR1. J Immunol 182: 2221–2230. doi:10 Olson OC, Tazzyman S, Danson S, Addison C, Clemons
.4049/jimmunol.0801878 M, et al. 2015. Perivascular M2 macrophages stimulate
Hanna RN, Cekic C, Sag D, Tacke R, Thomas GD, Nowyhed tumor relapse after chemotherapy. Cancer Res 75:
H, Herrley E, Rasquinha N, McArdle S, Wu R, et al. 2015. 3479–3491. doi:10.1158/0008-5472.CAN-14-3587
Patrolling monocytes control tumor metastasis to the Huh SJ, Liang S, Sharma A, Dong C, Robertson GP. 2010.
lung. Science 350: 985–990. doi:10.1126/science.aac9407 Transiently entrapped circulating tumor cells interact
Harney AS, Arwert EN, Entenberg D, Wang Y, Guo P, Qian with neutrophils to facilitate lung metastasis develop-
BZ, Oktay MH, Pollard JW, Jones JG, Condeelis JS. 2015. ment. Cancer Res 70: 6071–6082. doi:10.1158/0008-
Real-time imaging reveals local, transient vascular perme- 5472.CAN-09-4442
ability, and tumor cell intravasation stimulated by Tie2Hi Hüsemann Y, Geigl JB, Schubert F, Musiani P, Meyer M,
macrophage-derived VEGFA. Cancer Discov 5: 932–943. Burghart E, Forni G, Eils R, Fehm T, Riethmuller G, et
doi:10.1158/2159-8290.CD-15-0012 al. 2008. Systemic spread is an early step in breast cancer.
Harper KL, Sosa MS, Entenberg D, Hosseini H, Cheung JF, Cancer Cell 13: 58–68. doi:10.1016/j.ccr.2007.12.003
Nobre R, Avivar-Valderas A, Nagi C, Girnius N, Davis RJ, Ishihara D, Dovas A, Hernandez L, Pozzuto M, Wyckoff J,
et al. 2016. Mechanism of early dissemination and metas- Segall JE, Condeelis JS, Bresnick AR, Cox D. 2013. Wis-
tasis in Her2+ mammary cancer. Nature 540: 588–592. kott-Aldrich syndrome protein regulates leukocyte-de-
doi:10.1038/nature20609 pendent breast cancer metastasis. Cell Rep 4: 429–436.
Häuselmann I, Roblek M, Protsyuk D, Huck V, Knopfova L, doi:10.1016/j.celrep.2013.07.007
Grässle S, Bauer AT, Schneider SW, Borsig L. 2016. Jaiswal S, Jamieson CHM, Pang WW, Park CY, Chao MP,
Monocyte induction of E-selectin-mediated endothelial Majeti R, Traver D, Van Rooijen N, Weissman IL. 2009.
activation releases VE–cadherin junctions to promote tu- CD47 is upregulated on circulating hematopoietic stem
mor cell extravasation in the metastasis cascade. Cancer
cells and leukemia cells to avoid phagocytosis. Cell 138:
Res 76: 5302–5312. doi:10.1158/0008-5472.CAN-16-
271–285. doi:10.1016/j.cell.2009.05.046
0784
Jayaprakash P, Ai M, Liu A, Budhani P, Bartkowiak T, Sheng
He X, Dong DD, Yie SM, Yang H, Cao M, Ye SR, Li K, Liu J,
J, Ager C, Nicholas C, Jaiswal AR, Sun Y, et al. 2018.
Chen J. 2010. HLA-G expression in human breast cancer:
www.perspectivesinmedicine.org

Targeted hypoxia reduction restores T cell infiltration


Implications for diagnosis and prognosis, and effect on
and sensitizes prostate cancer to immunotherapy. J Clin
allocytotoxic lymphocyte response after hormone treat-
Invest 128: 5137–5149. doi:10.1172/JCI96268
ment in vitro. Ann Surg Oncol 17: 1459–1469. doi:10
.1245/s10434-009-0891-9 Jerby-Arnon L, Shah P, Cuoco MS, Rodman C, Su MJ,
Melms JC, Leeson R, Kanodia A, Mei S, Lin JR, et al.
Headley MB, Bins A, Nip A, Roberts EW, Looney MR, Ge-
2018. A cancer cell program promotes T cell exclusion
rard A, Krummel MF. 2016. Visualization of immediate
and resistance to checkpoint blockade. Cell 175: 984–997.
immune responses to pioneer metastatic cells in the lung.
e24. doi:10.1016/j.cell.2018.09.006
Nature 531: 513–517. doi:10.1038/nature16985
Jiang P, Lagenaur CF, Narayanan V. 1999. Integrin-associ-
Hendry S, Pang JB, Byrne DJ, Lakhani SR, Cummings MC,
Campbell IG, Mann GB, Gorringe KL, Fox SB. 2017. ated protein is a ligand for the P84 neural adhesion mol-
Relationship of the breast ductal carcinoma in situ im- ecule. J Biol Chem 274: 559–562. doi:10.1074/jbc.274.2
mune microenvironment with clinicopathological and .559
genetic features. Clin Cancer Res 23: 5210–5217. doi:10 Jiang H, Hegde S, Knolhoff BL, Zhu Y, Herndon JM, Meyer
.1158/1078-0432.CCR-17-0743 MA, Nywening TM, Hawkins WG, Shapiro IM, Weaver
Hiratsuka S, Watanabe A, Sakurai Y, Akashi-Takamura S, DT, et al. 2016. Targeting focal adhesion kinase renders
Ishibashi S, Miyake K, Shibuya M, Akira S, Aburatani H, pancreatic cancers responsive to checkpoint immuno-
Maru Y. 2008. The S100A8-serum amyloid A3-TLR4 therapy. Nat Med 22: 851–860. doi:10.1038/nm.4123
paracrine cascade establishes a pre-metastatic phase. Kaplan RN, Riba RD, Zacharoulis S, Bramley AH, Vincent L,
Nat Cell Biol 10: 1349–1355. doi:10.1038/ncb1794 Costa C, MacDonald DD, Jin DK, Shido K, Kerns SA, et
Hodi FS, Lee S, McDermott DF, Rao UN, Butterfield LH, al. 2005. VEGFR1-positive haematopoietic bone marrow
Tarhini AA, Leming P, Puzanov I, Shin D, Kirkwood JM. progenitors initiate the pre-metastatic niche. Nature 438:
2014. Ipilimumab plus sargramostim vs ipilimumab 820–827. doi:10.1038/nature04186
alone for treatment of metastatic melanoma: A random- Kastenhuber ER, Lowe SW. 2017. Putting p53 in context. Cell
ized clinical trial. JAMA 312: 1744–1753. doi:10.1001/ 170: 1062–1078. doi:10.1016/j.cell.2017.08.028
jama.2014.13943 Kato T, Noma K, Ohara T, Kashima H, Katsura Y, Sato H,
Hosseini H, Obradović MMS, Hoffmann M, Harper KL, Komoto S, Katsube R, Ninomiya T, Tazawa H, et al. 2018.
Sosa MS, Werner-Klein M, Nanduri LK, Werno C, Ehrl Cancer-associated fibroblasts affect intratumoral CD8+
C, Maneck M, et al. 2016. Early dissemination seeds me- and FoxP3+ T cells via IL6 in the tumor microenviron-
tastasis in breast cancer. Nature 540: 552–558. doi:10 ment. Clin Cancer Res 24: 4820–4833. doi:10.1158/1078-
.1038/nature20785 0432.CCR-18-0205
Hsu DS, Wang HJ, Tai SK, Chou CH, Hsieh CH, Chiu PH, Kessenbrock K, Plaks V, Werb Z. 2010. Matrix metallopro-
Chen NJ, Yang MH. 2014. Acetylation of Snail modulates teinases: Regulators of the tumor microenvironment. Cell
the cytokinome of cancer cells to enhance the recruitment 141: 52–67. doi:10.1016/j.cell.2010.03.015

Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424 17


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

A. El-Kenawi et al.

Kievits F, Ivanyi P, Krimpenfort P, Berns A, Ploegh HL. 1987. ognition and lysis of human and mouse melanoma cell
HLA-restricted recognition of viral antigens in HLA lines in vitro and in vivo. J Clin Invest 119: 1251–1263.
transgenic mice. Nature 329: 447–449. doi:10.1038/ doi:10.1172/JCI36022
329447a0 Lambert AW, Pattabiraman DR, Weinberg RA. 2017.
Kim S, Takahashi H, Lin WW, Descargues P, Grivennikov S, Emerging biological principles of metastasis. Cell 168:
Kim Y, Luo JL, Karin M. 2009. Carcinoma-produced fac- 670–691. doi:10.1016/j.cell.2016.11.037
tors activate myeloid cells through TLR2 to stimulate me- Lee YY, Choi CH, Kim HJ, Kim TJ, Lee JW, Lee JH, Bae DS,
tastasis. Nature 457: 102–106. doi:10.1038/nature07623 Kim BG. 2012. Pretreatment neutrophil:lymphocyte ratio
Kitamura T, Qian BZ, Pollard JW. 2015. Immune cell pro- as a prognostic factor in cervical carcinoma. Anticancer
motion of metastasis. Nat Rev Immunol 15: 73–86. doi:10 Res 32: 1555–1561.
.1038/nri3789 Lee CC, Lin JC, Hwang WL, Kuo YJ, Chen HK, Tai SK, Lin
Kitamura T, Doughty-Shenton D, Cassetta L, Fragkogianni CC, Yang MH. 2018. Macrophage-secreted interleukin-
S, Brownlie D, Kato Y, Carragher N, Pollard JW. 2018. 35 regulates cancer cell plasticity to facilitate metastatic
Monocytes differentiate to immune suppressive precur- colonization. Nat Commun 9: 3763. doi:10.1038/s41467-
sors of metastasis-associated macrophages in mouse 018-06268-0
models of metastatic breast cancer. Front Immunol 8: Le Gal K, Ibrahim MX, Wiel C, Sayin VI, Akula MK, Karls-
2004. doi:10.3389/fimmu.2017.02004 son C, Dalin MG, Akyürek LM, Lindahl P, Nilsson J.
Kochan G, Escors D, Breckpot K, Guerrero-Setas D. 2013. 2015. Antioxidants can increase melanoma metastasis
Role of non-classical MHC class I molecules in cancer in mice. Sci Transl Med 7: 308re308. doi:10.1126/sci
immunosuppression. Oncoimmunology 2: e26491. translmed.aad3740
doi:10.4161/onci.26491 Lewis CE, Harney AS, Pollard JW. 2016. The multifaceted
Kojima Y, Acar A, Eaton EN, Mellody KT, Scheel C, Ben- role of perivascular macrophages in tumors. Cancer Cell
Porath I, Onder TT, Wang ZC, Richardson AL, Weinberg 30: 18–25. doi:10.1016/j.ccell.2016.05.017
RA, et al. 2010. Autocrine TGF-β and stromal cell-derived Li MO, Wan YY, Flavell RA. 2007. T cell-produced trans-
factor-1 (SDF-1) signaling drives the evolution of forming growth factor-β1 controls T cell tolerance and
tumor-promoting mammary stromal myofibroblasts. regulates Th1- and Th17-cell differentiation. Immunity
Proc Natl Acad Sci 107: 20009–20014. doi:10.1073/pnas 26: 579–591. doi:10.1016/j.immuni.2007.03.014
.1013805107 Li S, Cong X, Gao H, Lan X, Li Z, Wang W, Song S, Wang Y,
Kopp HG, Placke T, Salih HR. 2009. Platelet-derived trans-
www.perspectivesinmedicine.org

Li C, Zhang H, et al. 2019. Tumor-associated neutrophils


forming growth factor-β down-regulates NKG2D thereby induce EMT by IL-17a to promote migration and inva-
inhibiting natural killer cell antitumor reactivity. Cancer sion in gastric cancer cells. J Exp Clin Cancer Res 38: 6.
Res 69: 7775–7783. doi:10.1158/0008-5472.CAN-09- doi:10.1186/s13046-018-1003-0
2123 Liao W, Overman MJ, Boutin AT, Shang X, Zhao D, Dey P, Li
Kortlever RM, Sodir NM, Wilson CH, Burkhart DL, Pelle- J, Wang G, Lan Z, Li J, et al. 2019. KRAS-IRF2 axis drives
grinet L, Brown Swigart L, Littlewood TD, Evan GI. 2017. immune suppression and immune therapy resistance in
Myc cooperates with Ras by programming inflammation colorectal cancer. Cancer Cell 35: 559–572.e7. doi:10
and immune suppression. Cell 171: 1301–1315.e14. .1016/j.ccell.2019.02.008
doi:10.1016/j.cell.2017.11.013 Lin A, Yan WH. 2015. Human leukocyte antigen-G (HLA-
Kroemer G, Galluzzi L, Kepp O, Zitvogel L. 2013. Immuno- G) expression in cancers: Roles in immune evasion, me-
genic cell death in cancer therapy. Annu Rev Immunol 31: tastasis and target for therapy. Mol Med 21: 782–791.
51–72. doi:10.1146/annurev-immunol-032712-100008 doi:10.2119/molmed.2015.00083
Kudo-Saito C, Shirako H, Takeuchi T, Kawakami Y. 2009. Lin EY, Li JF, Gnatovskiy L, Deng Y, Zhu L, Grzesik DA,
Cancer metastasis is accelerated through immunosup- Qian H, Xue XN, Pollard JW. 2006. Macrophages regulate
pression during Snail-induced EMT of cancer cells. Can- the angiogenic switch in a mouse model of breast cancer.
cer Cell 15: 195–206. doi:10.1016/j.ccr.2009.01.023 Cancer Res 66: 11238–11246. doi:10.1158/0008-5472
Kumar V, Donthireddy L, Marvel D, Condamine T, Wang F, .CAN-06-1278
Lavilla-Alonso S, Hashimoto A, Vonteddu P, Behera R, Lin EY, Li JF, Bricard G, Wang W, Deng Y, Sellers R, Porcelli
Goins MA, et al. 2017. Cancer-associated fibroblasts neu- SA, Pollard JW. 2007. Vascular endothelial growth factor
tralize the anti-tumor effect of CSF1 receptor blockade by restores delayed tumor progression in tumors depleted of
inducing PMN-MDSC infiltration of tumors. Cancer Cell macrophages. Mol Oncol 1: 288–302. doi:10.1016/j
32: 654–668.e5. doi:10.1016/j.ccell.2017.10.005 .molonc.2007.10.003
Labelle M, Begum S, Hynes RO. 2011. Direct signaling be- Linde N, Casanova-Acebes M, Sosa MS, Mortha A, Rahman
tween platelets and cancer cells induces an epithelial– A, Farias E, Harper K, Tardio E, Reyes Torres I, Jones J, et
mesenchymal-like transition and promotes metastasis. al. 2018. Macrophages orchestrate breast cancer early dis-
Cancer Cell 20: 576–590. doi:10.1016/j.ccr.2011.09.009 semination and metastasis. Nat Commun 9: 21. doi:10
Lakins MA, Ghorani E, Munir H, Martins CP, Shields JD. .1038/s41467-017-02481-5
2018. Cancer-associated fibroblasts induce antigen-spe- Liu Y, Cao X. 2016. Characteristics and significance of the
cific deletion of CD8+ T cells to protect tumour cells. Nat pre-metastatic niche. Cancer Cell 30: 668–681. doi:10
Commun 9: 948. doi:10.1038/s41467-018-03347-0 .1016/j.ccell.2016.09.011
Lakshmikanth T, Burke S, Ali TH, Kimpfler S, Ursini F, Liu X, Kwon H, Li Z, Fu YX. 2017. Is CD47 an innate im-
Ruggeri L, Capanni M, Umansky V, Paschen A, Sucker mune checkpoint for tumor evasion? J Hematol Oncol 10:
A, et al. 2009. NCRs and DNAM-1 mediate NK cell rec- 12. doi:10.1186/s13045-016-0381-z

18 Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

Immune Microenvironment and Cancer Metastasis

López-Soto A, Gonzalez S, Smyth MJ, Galluzzi L. 2017. Con- veillance and escape. Nat Rev Clin Oncol 14: 155–167.
trol of metastasis by NK cells. Cancer Cell 32: 135–154. doi:10.1038/nrclinonc.2016.144
doi:10.1016/j.ccell.2017.06.009 Motz GT, Santoro SP, Wang L-P, Garrabrant T, Lastra RR,
Loumagne L, Baudhuin J, Favier B, Montespan F, Carosella Hagemann IS, Lal P, Feldman MD, Benencia F, Coukos G.
ED, Rouas-Freiss N. 2014. In vivo evidence that secretion 2014. Tumor endothelium FasL establishes a selective
of HLA-G by immunogenic tumor cells allows their eva- immune barrier promoting tolerance in tumors. Nat
sion from immunosurveillance. Int J Cancer 135: 2107– Med 20: 607–615. doi:10.1038/nm.3541
2117. doi:10.1002/ijc.28845 Müller-Hermelink N, Braumüller H, Pichler B, Wieder T,
Lu H, Clauser KR, Tam WL, Fröse J, Ye X, Eaton EN, Rein- Mailhammer R, Schaak K, Ghoreschi K, Yazdi A, Haub-
hardt F, Donnenberg VS, Bhargava R, Carr SA, et al. 2014. ner R, Sander CA, et al. 2008. TNFR1 signaling and IFN-γ
A breast cancer stem cell niche supported by juxtacrine signaling determine whether T cells induce tumor dor-
signalling from monocytes and macrophages. Nat Cell mancy or promote multistage carcinogenesis. Cancer Cell
Biol 16: 1105–1117. doi:10.1038/ncb3041 13: 507–518. doi:10.1016/j.ccr.2008.04.001
MacKie RM, Reid R, Junor B. 2003. Fatal melanoma trans- Murdoch C, Giannoudis A, Lewis CE. 2004. Mechanisms
ferred in a donated kidney 16 years after melanoma sur- regulating the recruitment of macrophages into hypoxic
gery. N Engl J Med 348: 567–568. doi:10.1056/ areas of tumors and other ischemic tissues. Blood 104:
NEJM200302063480620 2224–2234. doi:10.1182/blood-2004-03-1109
Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Nagrath S, Sequist LV, Maheswaran S, Bell DW, Irimia D,
Wang Y, Kadel III EE, Koeppen H, Astarita JL, Cubas R, et Ulkus L, Smith MR, Kwak EL, Digumarthy S, Muzikan-
al. 2018. TGFβ attenuates tumour response to PD-L1 sky A, et al. 2007. Isolation of rare circulating tumour cells
blockade by contributing to exclusion of T cells. Nature in cancer patients by microchip technology. Nature 450:
554: 544–548. doi:10.1038/nature25501 1235–1239. doi:10.1038/nature06385
Massagué J, Obenauf AC. 2016. Metastatic colonization by Najmeh S, Cools-Lartigue J, Rayes RF, Gowing S, Vourtzou-
circulating tumour cells. Nature 529: 298–306. doi:10 mis P, Bourdeau F, Giannias B, Berube J, Rousseau S, Ferri
.1038/nature17038 LE, et al. 2017. Neutrophil extracellular traps sequester
circulating tumor cells via β1-integrin mediated interac-
Mazzieri R, Pucci F, Moi D, Zonari E, Ranghetti A, Berti A, tions. Int J Cancer 140: 2321–2330. doi:10.1002/ijc.30635
Politi LS, Gentner B, Brown JL, Naldini L, et al. 2011.
Nausch N, Cerwenka A. 2008. NKG2D ligands in tumor
Targeting the ANG2/TIE2 axis inhibits tumor growth
immunity. Oncogene 27: 5944–5958. doi:10.1038/onc
www.perspectivesinmedicine.org

and metastasis by impairing angiogenesis and disabling


.2008.272
rebounds of proangiogenic myeloid cells. Cancer Cell 19:
512–526. doi:10.1016/j.ccr.2011.02.005 Nelson AC, Machado HL, Schwertfeger KL. 2018. Breaking
through to the other side: Microenvironment contribu-
McAllister SS, Weinberg RA. 2014. The tumour-induced
tions to DCIS initiation and progression. J Mammary
systemic environment as a critical regulator of cancer
Gland Biol Neoplasia 23: 207–221. doi:10.1007/s10911-
progression and metastasis. Nat Cell Biol 16: 717–727.
018-9409-z
doi:10.1038/ncb3015
Nonn L, Ananthanarayanan V, Gann PH. 2009. Evidence for
McGranahan N, Rosenthal R, Hiley CT, Rowan AJ, Watkins field cancerization of the prostate. Prostate 69: 1470–
TBK, Wilson GA, Birkbak NJ, Veeriah S, Van Loo P, 1479. doi:10.1002/pros.20983
Herrero J, et al. 2017. Allele-specific HLA loss and im-
mune escape in lung cancer evolution. Cell 171: 1259– Northey JJ, Przybyla L, Weaver VM. 2017. Tissue force pro-
1271.e11. doi:10.1016/j.cell.2017.10.001 grams cell fate and tumor aggression. Cancer Discov 7:
1224–1237. doi:10.1158/2159-8290.CD-16-0733
McWhorter FY, Davis CT, Liu WF. 2015. Physical and me-
Olkhanud PB, Baatar D, Bodogai M, Hakim F, Gress R,
chanical regulation of macrophage phenotype and func-
Anderson RL, Deng J, Xu M, Briest S, Biragyn A. 2009.
tion. Cell Mol Life Sci 72: 1303–1316. doi:10.1007/s00018-
Breast cancer lung metastasis requires expression of che-
014-1796-8
mokine receptor CCR4 and regulatory T cells. Cancer Res
Meylan E, Dooley AL, Feldser DM, Shen L, Turk E, Ouyang 69: 5996–6004. doi:10.1158/0008-5472.CAN-08-4619
C, Jacks T. 2009. Requirement for NF-κB signalling in a
Olkhanud PB, Damdinsuren B, Bodogai M, Gress RE, Sen R,
mouse model of lung adenocarcinoma. Nature 462: 104–
Wejksza K, Malchinkhuu E, Wersto RP, Biragyn A. 2011.
107. doi:10.1038/nature08462 Tumor-evoked regulatory B cells promote breast cancer
Miles FL, Pruitt FL, van Golen KL, Cooper CR. 2008. Step- metastasis by converting resting CD4+ T cells to T-regu-
ping out of the flow: Capillary extravasation in cancer latory cells. Cancer Res 71: 3505–3515. doi:10.1158/0008-
metastasis. Clin Exp Metastasis 25: 305–324. doi:10 5472.CAN-10-4316
.1007/s10585-007-9098-2 Ouzounova M, Lee E, Piranlioglu R, El Andaloussi A, Kolhe
Milsom CC, Lee CR, Hackl C, Man S, Kerbel RS. 2013. R, Demirci MF, Marasco D, Asm I, Chadli A, Hassan KA,
Differential post-surgical metastasis and survival in et al. 2017. Monocytic and granulocytic myeloid derived
SCID, NOD-SCID and NOD-SCID-IL-2Rγnull mice suppressor cells differentially regulate spatiotemporal tu-
with parental and subline variants of human breast can- mour plasticity during metastatic cascade. Nat Commun
cer: Implications for host defense mechanisms regulating 8: 14979. doi:10.1038/ncomms14979
metastasis. PLoS ONE 8: e71270. doi:10.1371/journal Overman MJ, Lonardi S, Wong KYM, Lenz H-J, Gelsomino
.pone.0071270 F, Aglietta M, Morse MA, Van Cutsem E, McDermott R,
Mohme M, Riethdorf S, Pantel K. 2017. Circulating and Hill A. 2018. Durable clinical benefit with nivolumab plus
disseminated tumour cells—Mechanisms of immune sur- ipilimumab in DNA mismatch repair-deficient/microsat-

Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424 19


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

A. El-Kenawi et al.

ellite instability-high metastatic colorectal cancer. J Clin Qian BZ, Pollard JW. 2010. Macrophage diversity enhances
Oncol 36: 773–779. doi:10.1200/JCO.2017.76.9901 tumor progression and metastasis. Cell 141: 39–51. doi:10
Palumbo JS. 2008. Mechanisms linking tumor cell-associat- .1016/j.cell.2010.03.014
ed procoagulant function to tumor dissemination. Semin Qian BZ, Li J, Zhang H, Kitamura T, Zhang J, Campion
Thromb Hemost 34: 154–160. doi:10.1055/s-2008- LR, Kaiser EA, Snyder LA, Pollard JW. 2011. CCL2 re-
1079255 cruits inflammatory monocytes to facilitate breast-tu-
mour metastasis. Nature 475: 222–225. doi:10.1038/na
Palumbo JS, Talmage KE, Massari JV, La Jeunesse CM, Flick
ture10138
MJ, Kombrinck KW, Jirouskova M, Degen JL. 2005. Plate-
lets and fibrin(ogen) increase metastatic potential by im- Qin L, Liu Z, Chen H, Xu J. 2009. The steroid receptor
peding natural killer cell-mediated elimination of tumor coactivator-1 regulates twist expression and promotes
breast cancer metastasis. Cancer Res 69: 3819–3827.
cells. Blood 105: 178–185. doi:10.1182/blood-2004-06-
doi:10.1158/0008-5472.CAN-08-4389
2272
Quail DF, Joyce JA. 2013. Microenvironmental regulation of
Paolino M, Choidas A, Wallner S, Pranjic B, Uribesalgo I,
tumor progression and metastasis. Nat Med 19: 1423–
Loeser S, Jamieson AM, Langdon WY, Ikeda F, Fededa JP, 1437. doi:10.1038/nm.3394
et al. 2014. The E3 ligase Cbl-b and TAM receptors reg-
Reinhold MI, Lindberg FP, Plas D, Reynolds S, Peters MG,
ulate cancer metastasis via natural killer cells. Nature 507:
Brown EJ. 1995. In vivo expression of alternatively spliced
508–512. doi:10.1038/nature12998
forms of integrin-associated protein (CD47). J Cell Sci
Park J, Wysocki RW, Amoozgar Z, Maiorino L, Fein MR, 108: 3419–3425.
Jorns J, Schott AF, Kinugasa-Katayama Y, Lee Y, Won Reymond N, d’Água BB, Ridley AJ. 2013. Crossing the en-
NH, et al. 2016. Cancer cells induce metastasis-support- dothelial barrier during metastasis. Nat Rev Cancer 13:
ing neutrophil extracellular DNA traps. Sci Transl Med 8: 858–870. doi:10.1038/nrc3628
361ra138. doi:10.1126/scitranslmed.aag1711
Rivera LB, Bergers G. 2015. Intertwined regulation of angio-
Paschen A, Sucker A, Hill B, Moll I, Zapatka M, Nguyen genesis and immunity by myeloid cells. Trends Immunol
XD, Sim GC, Gutmann I, Hassel J, Becker JC, et al. 36: 240–249. doi:10.1016/j.it.2015.02.005
2009. Differential clinical significance of individual Romero I, Garrido C, Algarra I, Collado A, Garrido F, Gar-
NKG2D ligands in melanoma: Soluble ULBP2 as an cia-Lora AM. 2014. T lymphocytes restrain spontaneous
indicator of poor prognosis superior to S100B. Clin Can- metastases in permanent dormancy. Cancer Res 74:
cer Res 15: 5208–5215. doi:10.1158/1078-0432.CCR-09-
www.perspectivesinmedicine.org

1958–1968. doi:10.1158/0008-5472.CAN-13-2084
0886 Rooney MS, Shukla SA, Wu CJ, Getz G, Hacohen N. 2015.
Pasero C, Gravis G, Guerin M, Granjeaud S, Thomassin- Molecular and genetic properties of tumors associated
Piana J, Rocchi P, Paciencia-Gros M, Poizat F, Bentobji with local immune cytolytic activity. Cell 160: 48–61.
M, Azario-Cheillan F, et al. 2016. Inherent and tumor- doi:10.1016/j.cell.2014.12.033
driven immune tolerance in the prostate microenviron- Rosenthal R, Cadieux EL, Salgado R, Bakir MA, Moore DA,
ment impairs natural killer cell antitumor activity. Cancer Hiley CT, Lund T, Tanić M, Reading JL, Joshi K, et al.
Res 76: 2153–2165. doi:10.1158/0008-5472.CAN-15- 2019. Neoantigen-directed immune escape in lung cancer
1965 evolution. Nature 567: 479–485. doi:10.1038/s41586-
Peinado H, Zhang H, Matei IR, Costa-Silva B, Hoshino A, 019-1032-7
Rodrigues G, Psaila B, Kaplan RN, Bromberg JF, Kang Y, Ruffell B, Coussens LM. 2015. Macrophages and therapeutic
et al. 2017. Pre-metastatic niches: Organ-specific homes resistance in cancer. Cancer Cell 27: 462–472. doi:10
for metastases. Nat Rev Cancer 17: 302–317. doi:10.1038/ .1016/j.ccell.2015.02.015
nrc.2017.6 Ruffell B, Chang-Strachan D, Chan V, Rosenbusch A, Ho
Pickup MW, Mouw JK, Weaver VM. 2014. The extracellular CM, Pryer N, Daniel D, Hwang ES, Rugo HS, Coussens
matrix modulates the hallmarks of cancer. EMBO Rep 15: LM. 2014. Macrophage IL-10 blocks CD8+ T cell-depen-
1243–1253. doi:10.15252/embr.201439246 dent responses to chemotherapy by suppressing IL-12
expression in intratumoral dendritic cells. Cancer Cell
Placke T, Orgel M, Schaller M, Jung G, Rammensee HG,
26: 623–637. doi:10.1016/j.ccell.2014.09.006
Kopp HG, Salih HR. 2012. Platelet-derived MHC class I
confers a pseudonormal phenotype to cancer cells that Sanford DE, Belt BA, Panni RZ, Mayer A, Deshpande
AD, Carpenter D, Mitchem JB, Plambeck-Suess SM,
subverts the antitumor reactivity of natural killer immune
Worley LA, Goetz BD, et al. 2013. Inflammatory mono-
cells. Cancer Res 72: 440–448. doi:10.1158/0008-5472
cyte mobilization decreases patient survival in pancreatic
.CAN-11-1872
cancer: A role for targeting the CCL2/CCR2 axis. Clin
Porrello A, Leslie PL, Harrison EB, Gorentla BK, Kattula S, Cancer Res 19: 3404–3415. doi:10.1158/1078-0432
Ghosh SK, Azam SH, Holtzhausen A, Chao YL, Hayward .CCR-13-0525
MC, et al. 2018. Factor XIIIA–expressing inflammatory Sangaletti S, Di Carlo E, Gariboldi S, Miotti S, Cappetti B,
monocytes promote lung squamous cancer through fi- Parenza M, Rumio C, Brekken RA, Chiodoni C, Colombo
brin cross-linking. Nat Commun 9: 1988. doi:10.1038/ MP. 2008. Macrophage-derived SPARC bridges tumor
s41467-018-04355-w cell-extracellular matrix interactions toward metastasis.
Pylayeva-Gupta Y, Lee KE, Hajdu CH, Miller G, Bar-Sagi D. Cancer Res 68: 9050–9059. doi:10.1158/0008-5472
2012. Oncogenic Kras-induced GM-CSF production pro- .CAN-08-1327
motes the development of pancreatic neoplasia. Cancer Sangaletti S, Tripodo C, Santangelo A, Castioni N, Portararo
Cell 21: 836–847. doi:10.1016/j.ccr.2012.04.024 P, Gulino A, Botti L, Parenza M, Cappetti B, Orlandi R, et

20 Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

Immune Microenvironment and Cancer Metastasis

al. 2016. Mesenchymal transition of high-grade breast pulmonary fibrosis via CD204 and M2 macrophage acti-
carcinomas depends on extracellular matrix control of vation. PLoS ONE 8: e81382. doi:10.1371/journal.pone
myeloid suppressor cell activity. Cell Rep 17: 233–248. .0081382
doi:10.1016/j.celrep.2016.08.075 Steinert G, Schölch S, Niemietz T, Iwata N, García SA, Beh-
Santisteban M, Reiman JM, Asiedu MK, Behrens MD, Nas- rens B, Voigt A, Kloor M, Benner A, Bork U, et al. 2014.
sar A, Kalli KR, Haluska P, Ingle JN, Hartmann LC, Man- Immune escape and survival mechanisms in circulating
jili MH, et al. 2009. Immune-induced epithelial to mes- tumor cells of colorectal cancer. Cancer Res 74: 1694–
enchymal transition in vivo generates breast cancer stem 1704. doi:10.1158/0008-5472.CAN-13-1885
cells. Cancer Res 69: 2887–2895. doi:10.1158/0008-5472 Stockmann C, Doedens A, Weidemann A, Zhang N, Takeda
.CAN-08-3343 N, Greenberg JI, Cheresh DA, Johnson RS. 2008. Deletion
Schlecker E, Fiegler N, Arnold A, Altevogt P, Rose-John S, of vascular endothelial growth factor in myeloid cells ac-
Moldenhauer G, Sucker A, Paschen A, von Strandmann celerates tumorigenesis. Nature 456: 814–818. doi:10
EP, Textor S, et al. 2014. Metalloprotease-mediated tumor .1038/nature07445
cell shedding of B7-H6, the ligand of the natural killer Strell C, Entschladen F. 2008. Extravasation of leukocytes in
cell-activating receptor NKp30. Cancer Res 74: 3429–
comparison to tumor cells. Cell Commun Signal 4: 10.
3440. doi:10.1158/0008-5472.CAN-13-3017
doi:10.1186/1478-811X-6-10
Schmittnaegel M, Rigamonti N, Kadioglu E, Cassará A,
Strilic B, Offermanns S. 2017. Intravascular survival and
Wyser Rmili C, Kiialainen A, Kienast Y, Mueller HJ,
extravasation of tumor cells. Cancer Cell 32: 282–293.
Ooi CH, Laoui D, et al. 2017. Dual angiopoietin-2 and
doi:10.1016/j.ccell.2017.07.001
VEGFA inhibition elicits antitumor immunity that is en-
hanced by PD-1 checkpoint blockade. Sci Transl Med 9: Su S, Liu Q, Chen J, Chen J, Chen F, He C, Huang D, Wu W,
eaak9670. doi:10.1126/scitranslmed.aak9670 Lin L, Huang W, et al. 2014. A positive feedback loop
Schwich E, Rebmann V, Michita RT, Rohn H, Voncken JW, between mesenchymal-like cancer cells and macrophages
Horn PA, Kimmig R, Kasimir-Bauer S, Buderath P. 2019. is essential to breast cancer metastasis. Cancer Cell 25:
HLA-G 30 untranslated region variants +3187G/G, 605–620. doi:10.1016/j.ccr.2014.03.021
+3196G/G and +3035T define diametrical clinical status Szczerba BM, Castro-Giner F, Vetter M, Krol I, Gkountela S,
and disease outcome in epithelial ovarian cancer. Sci Rep Landin J, Scheidmann MC, Donato C, Scherrer R, Singer
9: 5407. doi:10.1038/s41598-019-41900-z J, et al. 2019. Neutrophils escort circulating tumour cells
Smyth MJ, Thia KY, Cretney E, Kelly JM, Snook MB, Forbes to enable cell cycle progression. Nature 566: 553–557.
www.perspectivesinmedicine.org

CA, Scalzo AA. 1999. Perforin is a major contributor to doi:10.1038/s41586-019-0915-y


NK cell control of tumor metastasis. J Immunol 162: Takeda K, Hayakawa Y, Smyth MJ, Kayagaki N, Yamaguchi
6658–6662. N, Kakuta S, Iwakura Y, Yagita H, Okumura K. 2001.
Smyth MJ, Cretney E, Takeda K, Wiltrout RH, Sedger LM, Involvement of tumor necrosis factor-related apoptosis-
Kayagaki N, Yagita H, Okumura K. 2001. Tumor necrosis inducing ligand in surveillance of tumor metastasis by
factor-related apoptosis-inducing ligand (TRAIL) con- liver natural killer cells. Nat Med 7: 94–100. doi:10
tributes to interferon γ-dependent natural killer cell pro- .1038/83416
tection from tumor metastasis. J Exp Med 193: 661–670. Taki M, Abiko K, Baba T, Hamanishi J, Yamaguchi K, Mur-
doi:10.1084/jem.193.6.661 akami R, Yamanoi K, Horikawa N, Hosoe Y, Nakamura E,
Soltanian S, Matin MM. 2011. Cancer stem cells and cancer et al. 2018. Snail promotes ovarian cancer progression by
therapy. Tumor Biol 32: 425–440. doi:10.1007/s13277- recruiting myeloid-derived suppressor cells via CXCR2
011-0155-8 ligand upregulation. Nat Commun 9: 1685. doi:10.1038/
Sosa MS, Bragado P, Aguirre-Ghiso JA. 2014. Mechanisms s41467-018-03966-7
of disseminated cancer cell dormancy: An awakening Tauriello DVF, Palomo-Ponce S, Stork D, Berenguer-Llergo
field. Nat Rev Cancer 14: 611–622. doi:10.1038/nrc3793 A, Badia-Ramentol J, Iglesias M, Sevillano M, Ibiza S,
Soucek L, Lawlor ER, Soto D, Shchors K, Swigart LB, Cañellas A, Hernando-Momblona X, et al. 2018. TGFβ
Evan GI. 2007. Mast cells are required for angiogenesis drives immune evasion in genetically reconstituted colon
and macroscopic expansion of Myc-induced pancreatic cancer metastasis. Nature 554: 538–543. doi:10.1038/na
islet tumors. Nat Med 13: 1211–1218. doi:10.1038/ ture25492
nm1649 Teramukai S, Kitano T, Kishida Y, Kawahara M, Kubota K,
Spicer JD, McDonald B, Cools-Lartigue JJ, Chow SC, Gian- Komuta K, Minato K, Mio T, Fujita Y, Yonei T, et al. 2009.
nias B, Kubes P, Ferri LE. 2012. Neutrophils promote liver Pretreatment neutrophil count as an independent prog-
metastasis via Mac-1-mediated interactions with circulat- nostic factor in advanced non-small-cell lung cancer: An
ing tumor cells. Cancer Res 72: 3919–3927. doi:10.1158/ analysis of Japan Multinational Trial Organisation LC00-
0008-5472.CAN-11-2393 03. Eur J Cancer 45: 1950–1958. doi:10.1016/j.ejca.2009
Spiegel A, Brooks MW, Houshyar S, Reinhardt F, Ardolino .01.023
M, Fessler E, Chen MB, Krall JA, DeCock J, Zervantona- Thiery JP, Acloque H, Huang RYJ, Nieto MA. 2009.
kis IK, et al. 2016. Neutrophils suppress intraluminal NK Epithelial–mesenchymal transitions in development
cell-mediated tumor cell clearance and enhance extrava- and disease. Cell 139: 871–890. doi:10.1016/j.cell.2009
sation of disseminated carcinoma cells. Cancer Discov 6: .11.007
630–649. doi:10.1158/2159-8290.CD-15-1157 Tian L, Goldstein A, Wang H, Ching Lo H, Sun Kim I, Welte
Stahl M, Schupp J, Jäger B, Schmid M, Zissel G, Müller- T, Sheng K, Dobrolecki LE, Zhang X, Putluri N, et al.
Quernheim J, Prasse A. 2013. Lung collagens perpetuate 2017. Mutual regulation of tumour vessel normalization

Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424 21


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

A. El-Kenawi et al.

and immunostimulatory reprogramming. Nature 544: Wculek SK, Malanchi I. 2015. Neutrophils support lung
250–254. doi:10.1038/nature21724 colonization of metastasis-initiating breast cancer cells.
Toh B, Wang X, Keeble J, Sim WJ, Khoo K, Wong WC, Kato Nature 528: 413–417. doi:10.1038/nature16140
M, Prevost-Blondel A, Thiery JP, Abastado JP. 2011. Mes- Welford AF, Biziato D, Coffelt SB, Nucera S, Fisher M, Pucci
enchymal transition and dissemination of cancer cells is F, Di Serio C, Naldini L, De Palma M, Tozer GM. 2011.
driven by myeloid-derived suppressor cells infiltrating the TIE2-expressing macrophages limit the therapeutic effi-
primary tumor. PLoS Biol 9: e1001162. doi:10.1371/jour cacy of the vascular-disrupting agent combretastatin A4
nal.pbio.1001162 phosphate in mice. J Clin Invest 121: 1969–1973. doi:10
Tohme S, Yazdani HO, Al-Khafaji AB, Chidi AP, Loughran .1172/JCI44562
P, Mowen K, Wang Y, Simmons RL, Huang H, Tsung A. Wellenstein MD, de Visser KE. 2018. Cancer-cell–intrinsic
2016. Neutrophil extracellular traps promote the devel- mechanisms shaping the tumor immune landscape.
opment and progression of liver metastases after surgical Immunity 48: 399–416. doi:10.1016/j.immuni.2018.03
stress. Cancer Res 76: 1367–1380. doi:10.1158/0008-5472 .004
.CAN-15-1591 Wesley RB II, Meng X, Godin D, Galis ZS. 1998. Extracellu-
Topalian SL, Sznol M, McDermott DF, Kluger HM, Carvajal lar matrix modulates macrophage functions characteristic
RD, Sharfman WH, Brahmer JR, Lawrence DP, Atkins to atheroma: Collagen type I enhances acquisition of res-
MB, Powderly JD, et al. 2014. Survival, durable tumor ident macrophage traits by human peripheral blood
remission, and long-term safety in patients with advanced monocytes in vitro. Arterioscler Thromb Vasc Biol 18:
melanoma receiving nivolumab. J Clin Oncol 32: 1020– 432–440. doi:10.1161/01.ATV.18.3.432
1030. doi:10.1200/JCO.2013.53.0105 Wiendl H, Mitsdoerffer M, Hofmeister V, Wischhusen J,
Turpin B, Miller W, Rosenfeldt L, Kombrinck K, Flick MJ, Bornemann A, Meyermann R, Weiss EH, Melms A,
Steinbrecher KA, Harmel-Laws E, Mullins ES, Shaw M, Weller M. 2002. A functional role of HLA-G expression
Witte DP, et al. 2014. Thrombin drives tumorigenesis in in human gliomas: An alternative strategy of immune
colitis-associated colon cancer. Cancer Res 74: 3020– escape. J Immunol 168: 4772–4780. doi:10.4049/jimmu
3030. doi:10.1158/0008-5472.CAN-13-3276 nol.168.9.4772
Ubil E, Caskey L, Holtzhausen A, Hunter D, Story C, Earp Wirz W, Antoine M, Tag CG, Gressner AM, Korff T, Heller-
HS. 2018. Tumor-secreted Pros1 inhibits macrophage M1 brand C, Kiefer P. 2008. Hepatic stellate cells display a
polarization to reduce antitumor immune response. J Clin functional vascular smooth muscle cell phenotype in a
Invest 128: 2356–2369. doi:10.1172/JCI97354 three-dimensional co-culture model with endothelial
www.perspectivesinmedicine.org

Valastyan S, Weinberg RA. 2011. Tumor metastasis: Molec- cells. Differentiation 76: 784–794. doi:10.1111/j.1432-
ular insights and evolving paradigms. Cell 147: 275–292. 0436.2007.00260.x
doi:10.1016/j.cell.2011.09.024 Worthington JJ, Fenton TM, Czajkowska BI, Klementowicz
Van Goethem E, Poincloux R, Gauffre F, Maridonneau-Par- JE, Travis MA. 2012. Regulation of TGFβ in the immune
ini I, Le Cabec V. 2010. Matrix architecture dictates three- system: An emerging role for integrins and dendritic cells.
dimensional migration modes of human macrophages: Immunobiology 217: 1259–1265. doi:10.1016/j.imbio
Differential involvement of proteases and podosome- .2012.06.009
like structures. J Immunol 184: 1049–1061. doi:10.4049/ Wyckoff J, Wang W, Lin EY, Wang Y, Pixley F, Stanley ER,
jimmunol.0902223 Graf T, Pollard JW, Segall J, Condeelis J. 2004. A paracrine
Vasiljeva O, Papazoglou A, Krüger A, Brodoefel H, Korovin loop between tumor cells and macrophages is required for
M, Deussing J, Augustin N, Nielsen BS, Almholt K, Bogyo tumor cell migration in mammary tumors. Cancer Res 64:
M, et al. 2006. Tumor cell-derived and macrophage-de- 7022–7029. doi:10.1158/0008-5472.CAN-04-1449
rived cathepsin B promotes progression and lung metas- Wyckoff JB, Wang Y, Lin EY, Li JF, Goswami S, Stanley ER,
tasis of mammary cancer. Cancer Res 66: 5242–5250. Segall JE, Pollard JW, Condeelis J. 2007. Direct visualiza-
doi:10.1158/0008-5472.CAN-05-4463 tion of macrophage-assisted tumor cell intravasation in
Veglia F, Perego M, Gabrilovich D. 2018. Myeloid-derived mammary tumors. Cancer Res 67: 2649–2656. doi:10
suppressor cells coming of age. Nat Immunol 19: 108– .1158/0008-5472.CAN-06-1823
119. doi:10.1038/s41590-017-0022-x Xiao D, Craig JC, Chapman JR, Dominguez-Gil B, Tong A,
Wang S, Yuan Y, Liao L, Kuang SQ, Tien JC, O’Malley BW, Wong G. 2013. Donor cancer transmission in kidney
Xu J. 2009. Disruption of the SRC-1 gene in mice sup- transplantation: A systematic review. Am J Transplant
presses breast cancer metastasis without affecting primary 13: 2645–2652. doi:10.1111/ajt.12430
tumor formation. Proc Natl Acad Sci 106: 151–156. doi:10 Yamaguchi K, Chikumi H, Shimizu A, Takata M, Kinoshita
.1073/pnas.0808703105 N, Hashimoto K, Nakamoto M, Matsunaga S, Kurai J,
Wang B, Wang Q, Wang Z, Jiang J, Yu SC, Ping YF, Yang J, Miyake N, et al. 2012. Diagnostic and prognostic impact
Xu SL, Ye XZ, Xu C, et al. 2014. Metastatic consequences of serum-soluble UL16-binding protein 2 in lung cancer
of immune escape from NK cell cytotoxicity by human patients. Cancer Sci 103: 1405–1413. doi:10.1111/j.1349-
breast cancer stem cells. Cancer Res 74: 5746–5757. doi:10 7006.2012.02330.x
.1158/0008-5472.CAN-13-2563 Yan HH, Pickup M, Pang Y, Gorska AE, Li Z, Chytil A,
Wang Y, Chen J, Yang L, Li J, Wu W, Huang M, Lin L, Su S. Geng Y, Gray JW, Moses HL, Yang L. 2010. Gr-1-
2019. Tumor-contacted neutrophils promote metastasis +CD11b+ myeloid cells tip the balance of immune pro-
by a CD90-TIMP-1 juxtacrine–paracrine loop. Clin Can- tection to tumor promotion in the premetastatic lung.
cer Res 25: 1957–1969. doi:10.1158/1078-0432.CCR-18- Cancer Res 70: 6139–6149. doi:10.1158/0008-5472
2544 .CAN-10-0706

22 Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

Immune Microenvironment and Cancer Metastasis

Yang L, Huang J, Ren X, Gorska AE, Chytil A, Aakre M, Car- associated with esophageal squamous cell carcinoma pro-
bone DP, Matrisian LM, Richmond A, Lin PC, et al. 2008. gression and poor prognosis. Immunol Lett 161: 13–19.
Abrogation of TGFβ signaling in mammary carcinomas doi:10.1016/j.imlet.2014.04.007
recruits Gr-1+CD11b+ myeloid cells that promote metas- Zhou Z, Xia G, Xiang Z, Liu M, Wei Z, Yan J, Chen W, Zhu J,
tasis. Cancer Cell 13: 23–35. doi:10.1016/j.ccr.2007.12.004 Awasthi N, Sun X, et al. 2019. A C-X-C chemokine
Zhang J, Basher F, Wu JD. 2015. NKG2D ligands in tumor receptor type 2–dominated cross-talk between tumor
immunity: Two sides of a coin. Front Immunol 6: 97. cells and macrophages drives gastric cancer metastasis.
Zheng J, Xu C, Chu D, Zhang X, Li J, Ji G, Hong L, Feng Q, Li Clin Cancer Res 25: 3317–3328. doi:10.1158/1078-0432
X, Wu G, et al. 2014. Human leukocyte antigen G is .CCR-18-3567
www.perspectivesinmedicine.org

Cite this article as Cold Spring Harb Perspect Med 2020;10:a037424 23


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 17, 2023 - Published by Cold Spring Harbor Laboratory Press

The Immune Microenvironment and Cancer Metastasis


Asmaa El-Kenawi, Kay Hänggi and Brian Ruffell

Cold Spring Harb Perspect Med 2020; doi: 10.1101/cshperspect.a037424 originally published online
September 9, 2019

Subject Collection Metastasis: Mechanism to Therapy

Cellular Plasticity during Metastasis: New Insights Premetastasis


Provided by Intravital Microscopy Yoshiro Maru
Andreia S. Margarido, Laura Bornes, Claire Vennin,
et al.
The Etiology and Impact of Muscle Wasting in Bone Tropism in Cancer Metastases
Metastatic Cancer Hai Wang, Weijie Zhang, Igor Bado, et al.
Anup K. Biswas and Swarnali Acharyya
Normal Aging and Its Role in Cancer Metastasis Impact of Immunometabolism on Cancer
Mitchell Fane and Ashani T. Weeraratna Metastasis: A Focus on T Cells and Macrophages
Nina C. Flerin, Sotiria Pinioti, Alessio Menga, et al.
Zebrafish In Vivo Models of Cancer and New Approaches on Cancer Immunotherapy
Metastasis Jong-Ho Cha, Li-Chuan Chan, Min Sup Song, et al.
Katy R. Astell and Dirk Sieger
Extracellular Vesicles and Metastasis Cancer Stem Cells and Epithelial-to-Mesenchymal
Shizhen Emily Wang Transition in Cancer Metastasis
Toni Celià-Terrassa and Mohit Kumar Jolly
The Different Facets of Liquid Biopsy: A Clinical Perspectives in Brain Metastasis
Kaleidoscopic View Krutika Deshpande, Ian Buchanan, Vahan
Zahra Eslami-S, Luis Enrique Cortés-Hernández, Martirosian, et al.
Laure Cayrefourcq, et al.
Myeloid Cells in Metastasis Brain Metastasis Organotropism
Agnieszka Swierczak and Jeffrey W. Pollard Arseniy E. Yuzhalin and Dihua Yu
Cancer Cell Dormancy in Metastasis The Immune Microenvironment and Cancer
Matthew A. Summers, Michelle M. McDonald and Metastasis
Peter I. Croucher Asmaa El-Kenawi, Kay Hänggi and Brian Ruffell

For additional articles in this collection, see http://perspectivesinmedicine.cshlp.org/cgi/collection/

Copyright © 2020 Cold Spring Harbor Laboratory Press; all rights reserved

You might also like