Liver metastases remain a major barrier to successful management of malignant disease, particularly for cancers of the gastrointestinal tract but also for other malignancies, such as breast carcinoma and melanoma. The ability of metastatic cells to survive and proliferate in the liver is determined by the outcome of complex, reciprocal interactions between tumor cells and different local resident subpopulations, including the sinusoidal endothelium, stellate, Kupffer, and inflammatory cells that are mediated through cell–cell and cell–extracellular matrix adhesion and the release of soluble factors. Cross-communication between different hepatic resident cells in response to local tissue damage and inflammation and the recruitment of bone marrow cells further enhance this intercellular communication network. Both resident and recruited cells can play opposing roles in the progression of metastasis, and the balance of these divergent effects determines whether the tumor cells will die, proliferate, and colonize the new site or enter a state of dormancy. Moreover, this delicate balance can be tilted in favor of metastasis, if factors produced by the primary tumor precondition the microenvironment to form niches of activated resident cells that promote tumor expansion. This review aims to summarize current knowledge on these diverse interactions and the impact they can have on the clinical management of hepatic metastases. Clin Cancer Res; 22(24); 5971–82. ©2016 AACR.

Cancer metastasis remains the major challenge to successful management of malignant disease. The liver is the main site of metastatic disease and a major cause of death from gastrointestinal malignancies, such as colon, gastric, and pancreatic carcinomas as well as melanoma, breast cancer, and sarcomas (1).

Circulating metastatic cells that enter the liver encounter unique cellular populations. These include the parenchymal hepatocytes and the nonparenchymal hepatocytes, including liver sinusoidal endothelial cells (LSEC), hepatic stellate cells (HSC), Kupffer cells (KC), dendritic cells, liver-associated lymphocytes, and portal fibroblasts. Circulating and bone marrow–derived immune cells are also recruited to the liver in response to, and possibly in preparation for, invading tumor cells and can affect the final outcome (reviewed in refs. 2, 3; see Table 1).

Table 1.

Cells and mediators involved in the different phases of liver metastasis

Soluble factors involved
Phase of the metastatic processCell type involvedCell surface markersRecruiting or activatingProducedReferences
Premetastatic niche 
 KCs CD11b, F4/80 M-CSF, GM-CSF, MIF TGFβ, S100P, S100A8 (11–14) 
 HSCs α-SMA, desmin, GFAP KC-derived TGFβ and TNFα Fibronectin (11, 13) 
 MDSCs CD11b, Ly6C, Ly6G Tumor-derived GM-CSF S100A8 (12, 15) 
 Neutrophils CD11b, Ly6G TIMP-1, SDF-1 S100A8 (7, 14, 67) 
Post-tumor invasion 
  • (1) Microvascular phase: Tumor cells trapped in the vasculature

 
LSECs CD31, TNFR1, TNFR2, αvβ3, E-selectin, VCAM-1, ICAM-1 TNFα, IL1β, IL18 ROS, NO, IFNγ, TNFα (5, 18–20, 23, 31) 
 KCs CD11b, F4/80 M-CSF, GM-CSF, IFNγ TNFα, IL1, IL6, IL8, IFNγ, MIP-1α, MIP-2, IP-10, MCP-1, CCL5, VEGF (5, 23, 31, 45–47, 51) 
 Neutrophils CD11b, Ly6G S100A8, S100A9, IL8, CCL2 TNFα, ROS, defensins, perforins, elastase (28, 31, 64) 
 Hepatic NK cells NK1.1, NK1.2, FcγRIII CXCL9, CXCL10, IL1, IL12, IL15, IL18 Grz, Perforin, IFNγ, TNFα (5, 31, 48) 
  • (2) Extravascular, preangiogenic phase: Tumor cells transmigrate into the space of Disse, activating a local stromal response

 
HSCs α-SMA, desmin, GFAP KC-derived TGFβ, PDGF, bFGF, SDF-1, IGF-I, CCL2 ECM deposition, MCP-1, CCL5, CCL21, TGFβ (74–77, 84) 
 Neutrophils CD11b, Ly6G; N1: ICAM-1; N2: CXCR4 S100A8, S100A9, CXCL1, CXCL2, CXCL5, TNFα, IL8, G-CSF, IFNγ, CCL2, CCL5, TGFβ TNFα, ROS, NOS, MMP-8, MMP-9, elastase, cathepsin G, VEGF (28, 60, 64, 71) 
 KCs CD11b, F4/80 M-CSF, GM-CSF, IFNγ IL6, VEGF, HGF, MMP-9, MMP-14 (5, 50, 53) 
  • (3) Angiogenic phase: Micrometastases are vascularized

 
Blood-derived monocytes CD11b, F4/80, CD68, CD163, CD206 TNFα, MCP-1 VEGF, MMPs, bFGF (57, 60) 
 HSCs α-SMA, desmin, GFAP KC-derived TGFβ, PDGF, bFGF, SDF-1, IGF-I, CCL2 VEGF, angiopoietin-1, MMP-2, -9, and -13 (74, 79–82, 88) 
 LSECs CD31, TNFR1, ICAM-1 VEGF, Ang1, inhibited by Notch1 and regulated by Ang2 Fibronectin, MIF (23, 41–44) 
 Macrophages (M2) CD11b, F4/80, Ly6C, CD163 TGFβ, IL10 NO, TNFα, IFN, VEGF, EGF, bFGF, TGFβ, IL10 (57–61) 
  • (4) Growth phase: Metastases expansion

 
Hepatocytes Albumin, tyrosine aminotransferase Adhesion to tight junction integral proteins AREG, EGF, HB-EGF, ErB2, IGF-I, HGFL, ErB3, bFGF (92–98, 101–105, 107) 
 Tregs CD4, CD25, Foxp3 TGFβ, IL10, CCL5, CCL22 VEGF, TGFβ, IL10 (35, 111, 124) 
Soluble factors involved
Phase of the metastatic processCell type involvedCell surface markersRecruiting or activatingProducedReferences
Premetastatic niche 
 KCs CD11b, F4/80 M-CSF, GM-CSF, MIF TGFβ, S100P, S100A8 (11–14) 
 HSCs α-SMA, desmin, GFAP KC-derived TGFβ and TNFα Fibronectin (11, 13) 
 MDSCs CD11b, Ly6C, Ly6G Tumor-derived GM-CSF S100A8 (12, 15) 
 Neutrophils CD11b, Ly6G TIMP-1, SDF-1 S100A8 (7, 14, 67) 
Post-tumor invasion 
  • (1) Microvascular phase: Tumor cells trapped in the vasculature

 
LSECs CD31, TNFR1, TNFR2, αvβ3, E-selectin, VCAM-1, ICAM-1 TNFα, IL1β, IL18 ROS, NO, IFNγ, TNFα (5, 18–20, 23, 31) 
 KCs CD11b, F4/80 M-CSF, GM-CSF, IFNγ TNFα, IL1, IL6, IL8, IFNγ, MIP-1α, MIP-2, IP-10, MCP-1, CCL5, VEGF (5, 23, 31, 45–47, 51) 
 Neutrophils CD11b, Ly6G S100A8, S100A9, IL8, CCL2 TNFα, ROS, defensins, perforins, elastase (28, 31, 64) 
 Hepatic NK cells NK1.1, NK1.2, FcγRIII CXCL9, CXCL10, IL1, IL12, IL15, IL18 Grz, Perforin, IFNγ, TNFα (5, 31, 48) 
  • (2) Extravascular, preangiogenic phase: Tumor cells transmigrate into the space of Disse, activating a local stromal response

 
HSCs α-SMA, desmin, GFAP KC-derived TGFβ, PDGF, bFGF, SDF-1, IGF-I, CCL2 ECM deposition, MCP-1, CCL5, CCL21, TGFβ (74–77, 84) 
 Neutrophils CD11b, Ly6G; N1: ICAM-1; N2: CXCR4 S100A8, S100A9, CXCL1, CXCL2, CXCL5, TNFα, IL8, G-CSF, IFNγ, CCL2, CCL5, TGFβ TNFα, ROS, NOS, MMP-8, MMP-9, elastase, cathepsin G, VEGF (28, 60, 64, 71) 
 KCs CD11b, F4/80 M-CSF, GM-CSF, IFNγ IL6, VEGF, HGF, MMP-9, MMP-14 (5, 50, 53) 
  • (3) Angiogenic phase: Micrometastases are vascularized

 
Blood-derived monocytes CD11b, F4/80, CD68, CD163, CD206 TNFα, MCP-1 VEGF, MMPs, bFGF (57, 60) 
 HSCs α-SMA, desmin, GFAP KC-derived TGFβ, PDGF, bFGF, SDF-1, IGF-I, CCL2 VEGF, angiopoietin-1, MMP-2, -9, and -13 (74, 79–82, 88) 
 LSECs CD31, TNFR1, ICAM-1 VEGF, Ang1, inhibited by Notch1 and regulated by Ang2 Fibronectin, MIF (23, 41–44) 
 Macrophages (M2) CD11b, F4/80, Ly6C, CD163 TGFβ, IL10 NO, TNFα, IFN, VEGF, EGF, bFGF, TGFβ, IL10 (57–61) 
  • (4) Growth phase: Metastases expansion

 
Hepatocytes Albumin, tyrosine aminotransferase Adhesion to tight junction integral proteins AREG, EGF, HB-EGF, ErB2, IGF-I, HGFL, ErB3, bFGF (92–98, 101–105, 107) 
 Tregs CD4, CD25, Foxp3 TGFβ, IL10, CCL5, CCL22 VEGF, TGFβ, IL10 (35, 111, 124) 

NOTE: Shown are the four phases of the metastatic process that follow tumor cell invasion into the liver. Listed are the hepatic cells known to be involved at each phase, the mediators regulating their recruitment and activation, and the factors they produce to block or promote the metastatic process. The process has been simplified and divided into distinct phases in the interest of clarity. However, it should be viewed as dynamic with overlapping cellular and molecular drivers at different phases. Moreover, as the metastatic process advances, the cancer and hepatic cells evolve and their phenotypes change as a consequence of their interactions. These newly acquired properties help propel the process forward (see also Figs. 1–3).

Abbreviations: ECM, extracellular matrix; MDSC, myeloid-derived suppressor cell; MMP, matrix metalloproteinase, NK, natural killer; Treg, regulatory T cell.

This review summarizes our current understanding of the role of the liver microenvironment in the growth of hepatic metastases and the reciprocal interactions between metastatic tumor cells and different liver cell populations that regulate the process.

The process of liver metastasis has previously been divided into four major phases (detailed in Table 1) that follow tumor cell entry into the liver (4, 5). Emerging evidence suggests that in addition, a premetastatic phase could set the stage for liver colonization by disseminating tumor cells.

Evidence for premetastatic niche formation in the liver

The term “premetastatic niche” was coined to describe a microenvironment in a secondary organ site that has been rendered permissive to metastatic outgrowth in advance of cancer cell entry through the activity of circulating factors released by the primary tumor (6–8). Although the dependency of metastasis on premetastatic niches remains controversial and difficult to verify in the clinical setting (9, 10), it appears that a consensus is emerging on two scores: (i) that their existence may have important implications for the clinical management of metastatic disease, and (ii) that much can be learned from their interrogation about the cellular/molecular changes required to facilitate tumor cell growth in a distant site, such as the liver. Two recent studies have confirmed their potential relevance to liver metastasis. In a mouse model of metastatic pancreatic ductal adenocarcinoma (PDAC), Costa-Silva and colleagues showed that PDAC-derived exosomes taken up by hepatic KCs, upregulate TGFβ production, leading to increased fibronectin production by HSCs and recruitment of bone marrow–derived macrophages. They identified macrophage migration inhibitory factor (MIF) as essential for premetastatic niche formation and metastasis. Moreover, in exosomes derived from patients with stage I PDAC who later developed liver metastasis, MIF levels were found to be higher than in patients whose tumors did not progress, thus identifying MIF as a potential predictor of PDAC liver metastasis (11). This group also showed that exosomes from human breast and pancreatic cancer cell lines that metastasize selectively to the lung (MDA-MB-231) or liver (BxPC-3 and HPAF-II) fused preferentially with fibroblasts and epithelial cells in the lung and KCs in the liver, and this was mediated by the exosomal integrin laminin receptors α6β1 and α6β4 and the fibronectin receptor αvβ5, respectively. In exosome-fused KCs, the proinflammatory factors S100P and S100A8 were upregulated (Table 1; Fig. 1). Significantly, in PDAC patients, a correlation was documented between the levels of circulating, αv-bearing exosomes and disease stage, suggesting that exosomes may play a role clinically and their levels and integrin cargo may predict metastases in specific organs (12). Kowanetz and colleagues reported that tumor-derived GM-CSF could mobilize Ly6C+Ly6G+ myeloid cells into metastases, enabling niches in the lung and liver, and also identified S100A8 as a driving factor (13). Tumor-derived TIMP-1 was identified as another potential inducer of increased liver susceptibility to metastasis, acting via hepatic SDF-1 and neutrophil recruitment (14). Interestingly, S100A8 was identified as a driver of liver premetastatic niche formation in several studies (12–15), suggesting that it may have clinical utility as a predictor of liver metastasis.

Figure 1.

The premetastatic niche in the liver. Shown is a diagrammatic representation of the multistep process involved in the formation of premetastatic niches in the liver based on data described in refs. 11, 12. The symbols used for depiction of different hepatic cells are listed in Fig. 3. A and B, A diagrammatic representation of a PDAC is shown in inset A, and an enlarged image of a Kupffer cell activated by PDAC-derived exosomes is shown in inset B. Encircled numbers denote cellular and molecular interactions that can potentially be targeted for liver metastasis prevention, as detailed in the right-hand corner box. BM, bone marrow.

Figure 1.

The premetastatic niche in the liver. Shown is a diagrammatic representation of the multistep process involved in the formation of premetastatic niches in the liver based on data described in refs. 11, 12. The symbols used for depiction of different hepatic cells are listed in Fig. 3. A and B, A diagrammatic representation of a PDAC is shown in inset A, and an enlarged image of a Kupffer cell activated by PDAC-derived exosomes is shown in inset B. Encircled numbers denote cellular and molecular interactions that can potentially be targeted for liver metastasis prevention, as detailed in the right-hand corner box. BM, bone marrow.

Close modal

The stage of primary tumor development at which premetastatic niches can be formed is difficult to assess in the clinical setting. Costa-Silva and colleagues found exosomal MIF upregulation in mice with pretumoral pancreatic lesions, and high plasma exosomal MIF levels were also detected in patients with stage I PDAC (11), suggesting that they could be formed at very early stages of cancer development. The potential contribution of circulating cancer cells (CTC) that may be present early in tumor development and even after resection of the primary tumor (16, 17) is also unknown.

Host innate resistance mechanism can destroy tumor cells prior to extravasation

Blood-borne cancer cells entering the liver first encounter the LSECs, KCs, and hepatic natural killer (NK) cells (pit cells) that together mount the first line of defense (18). Cancer cells entrapped in the sinusoids may undergo destruction due to mechanical stress and deformation-associated trauma or may die due to phagocytosis by KCs or cytolysis by NK cell–released perforin/granzyme (18; reviewed in ref. 4). NO, ROS, and toxic radicals released by LSECs in response to local ischemia/reperfusion can contribute to tumor cell death (19, 20). Release of NO and IFNγ by LSECs and NK cells can result in upregulation of tumor cell Fas and apoptosis (18) that can be augmented by NK and LSEC-derived TNFα (21). TNFα is one of the numerous cytokines and chemokines unleashed by KCs in response to inflammation (Table 1; ref. 22). In addition to causing cell death directly, some of these factors can mobilize and activate additional innate immune cells, such as neutrophils, thereby adding to the local tumoricidal capacity (reviewed in ref. 23). In several animal tumor models, including colon cancer, loss of NK cells increased cancer cell growth in the liver, whereas enhanced NK activity reduced liver metastasis (24–26). Indirect evidence also suggests that susceptibility to NK-mediated immune attack can affect the ability of human cancer cells to generate liver metastases (27). Circulating neutrophils can also release various factors abundant in their granules to kill tumor cells (see Table 1), and their cytokines and chemokines can activate the tumoricidal potential of resident KCs and recruit host immune T cells with antitumorigenic activities (reviewed in ref. 28).

Cancer cells can escape these cytotoxic effects by forming clusters with blood or other cancer cells that shield them from the lethal effects of shear stress or NK-mediated cytotoxicity (reviewed in ref. 29). The release of inflammatory mediators, such as IL1β, TNFα, and IL18, can also initiate a cascade that facilitates their rapid exit from the vasculature to a less “toxic” microenvironment (see below and Fig. 2).

Figure 2.

The multistep process of liver colonization by disseminated cancer cells. Shown is a diagrammatic representation of the major cell types, soluble factors, and extracellular matrix (ECM) components in the liver microenvironment that impact the progression of liver metastasis. The four major phases in the process are delineated by dashed lines. Top, major intercellular interactions that occur upon entry of the tumor cells into the sinusoidal vessels (the microvascular phase) and precede tumor extravasation; middle, events following tumor cell extravasation into the space of Disse, divided into the preangiogenic phase (middle left) that is orchestrated by stellate cell activation, ECM deposition, cytokine production, and the recruitment of innate and adaptive immune cells and culminates in neovascularization (the vascular phase; middle right); bottom, these interactions enable tumor expansion (the growth phase). The symbols used for depiction of different hepatic cells are listed in Fig. 3 or indicated in the diagram. The encircled numbers denote potential targets for therapeutic intervention, as detailed in the boxes inserted within each of the depicted phases. MDSC, myeloid-derived suppressor cell; MMP, matrix metalloproteinase; Treg, regulatory T cell.

Figure 2.

The multistep process of liver colonization by disseminated cancer cells. Shown is a diagrammatic representation of the major cell types, soluble factors, and extracellular matrix (ECM) components in the liver microenvironment that impact the progression of liver metastasis. The four major phases in the process are delineated by dashed lines. Top, major intercellular interactions that occur upon entry of the tumor cells into the sinusoidal vessels (the microvascular phase) and precede tumor extravasation; middle, events following tumor cell extravasation into the space of Disse, divided into the preangiogenic phase (middle left) that is orchestrated by stellate cell activation, ECM deposition, cytokine production, and the recruitment of innate and adaptive immune cells and culminates in neovascularization (the vascular phase; middle right); bottom, these interactions enable tumor expansion (the growth phase). The symbols used for depiction of different hepatic cells are listed in Fig. 3 or indicated in the diagram. The encircled numbers denote potential targets for therapeutic intervention, as detailed in the boxes inserted within each of the depicted phases. MDSC, myeloid-derived suppressor cell; MMP, matrix metalloproteinase; Treg, regulatory T cell.

Close modal

The sinusoidal endothelium actively participates in different phases of metastasis

Although the rapid inflammatory response initiated by cancer cell entry can lead to cell death, it may also have a tumor-protective effect by upregulating the expression of LSEC cell adhesion molecules (CAM) and in this way, enhance cancer cell adhesion and transendothelial migration into the space of Disse, where they can escape the cytotoxic effects of KCs and NK cells (30). Cancer cells can adhere to inflammation-induced E-selectin, VCAM-1, and ICAM-1, either as single cells or in association with host cells, such as KCs or neutrophils (reviewed in ref. 31). Blockade of this inflammatory cascade was shown to reduce liver metastasis (32–34), and TNFR1 signaling was identified as a major driver of this process (35).

Ultimately, the outcome of these opposing effects on tumor cell survival and progression to the next phase depends on multiple factors, including the expression on the cancer cells of the respective counter receptors, namely the E-selectin ligands sLewa and sLewx and CD44 isoforms (36; reviewed in refs. 29, 37), and VCAM-1 and ICAM-1 counter receptors integrins α4β1 and LFA-1 and Mac-1 (38), respectively. E-selectin binding triggers a signaling cascade in both tumor cells and LSECs, leading to diapedesis and transendothelial migration (39) and altering gene expression in a tumor type–specific manner (40). Clinical studies have documented increased expression of E-selectin in and around colorectal cancer liver metastases and elevated soluble E-selectin, ICAM-1, and VCAM-1 levels that correlated with disease outcome in patients with colorectal cancer (reviewed in ref. 23).

LSECs may have other metastasis-promoting functions. LSEC-secreted fibronectin can induce epithelial–mesenchymal transition (EMT) in colorectal cancer cells via integrin α9β1, enhancing ERK signaling and tumor invasion (41). Human LSECs were shown to induce colorectal cancer migration and EMT via MIF (42), thereby increasing their metastatic potential.

LSECs contribute to tumor-induced angiogenesis. A recent study identified Notch1 as a negative regulator of sinusoidal endothelial cells sprouting into micrometastases and thereby of angiogenesis and liver metastasis (43), raising questions about the advisability of Notch targeting for cancer therapy. Moreover, liver metastasis of different carcinomas, including colorectal cancer, can co-opt the sinusoidal endothelium at the tumor–liver interface, giving rise to a histologic growth pattern termed the “replacement” or “sinusoidal” growth pattern that seems to result from tumor cell invasion between LSECs and the matrix in the space of Disse and is characterized by the formation of intrametastatic vessels that appear continuous with the sinusoidal vessels (reviewed in ref. 23). The factors that drive this co-opting mechanism remain to be identified, but a recent study by Im and colleagues suggests that Ang2 may play a regulatory role (44).

Collectively, the evidence shows that LSECs are not a passive barrier to tumor cell extravasation but participate actively in the metastatic process. Cancer cell interaction with LSECs can reciprocally alter the phenotypes of both cell types, and this may lead to intravascular tumor cell destruction but can also promote metastasis through enhanced tumor cell migration and increased angiogenesis (Table 1; Figs. 2 and 3).

Figure 3.

Parenchymal, nonparenchymal, and immune cells of the liver and their role in metastasis. Listed are the parenchymal and nonparenchymal cells of the liver and their anti- and prometastatic functions. Symbols shown for each cell type were used to depict intercellular communications in Figs. 1 and 2. EMT, epithelial-mesenchymal transition; Treg, regulatory T cell.

Figure 3.

Parenchymal, nonparenchymal, and immune cells of the liver and their role in metastasis. Listed are the parenchymal and nonparenchymal cells of the liver and their anti- and prometastatic functions. Symbols shown for each cell type were used to depict intercellular communications in Figs. 1 and 2. EMT, epithelial-mesenchymal transition; Treg, regulatory T cell.

Close modal

The role of macrophages is context dependent

KCs constitute approximately 10% of all liver cells. Recent studies suggest that they may be established prenatally and maintained into adulthood, independently of replenishment from blood monocytes but in an M-CSF and GM-CSF–dependent manner (45). They reside mainly in the hepatic sinusoids and are anchored to the endothelium by long cytoplasmic processes (46; reviewed in ref. 47). In their interaction with invading cancer cells, KCs can play diverse and opposing roles, depending on factors such as the stage of the metastatic process, tumor load, and interactions with other immune cells. As discussed, KCs can promote metastasis by orchestrating the premetastatic niche. However, tumor–KC interactions can have opposing effects. Several studies have documented a rapid adhesion of tumor cells to KCs in the sinusoidal lumen, resulting mainly in tumor cell phagocytosis or apoptosis, within hours of tumor cell entry (47). This tumoricidal effect may require cooperation with local or recruited NK cells (48), and its efficiency in removing cancer cells likely depends on the tumor load (49). An intravital microscopy study (50) recently revealed that 74% of intra-arterially injected rat colon cancer cells adhered to the KCs within 6 hours, and this was associated with increased liver TNFα and IL1β levels, consistent with our own findings (51, 52) and indicative of KC activation. Extensive phagocytosis of tumor cells was observed within 2 hours postinjection. Interestingly, although elimination of KCs 2 days prior to tumor injection increased liver metastasis, it had no significant effect up to 1 week after tumor injection, suggesting that KCs exerted their most potent antitumor effect within the first 24 hours of tumor cell entry into the liver (50). A bimodal effect of KC depletion was also documented in another study of murine colorectal cancer, and it was attributed to decreased VEGF and increased iNOS levels in livers subjected to late-stage KC depletion (53). Indeed, tumor cells that survive the initial tumoricidal assault by KCs can benefit from their protumorigenic functions. Adhesion to KC can facilitate tumor cell extravasation, among others, by sequential activation of endothelial CAMs (29, 54). In addition, KCs can produce cytokines and growth factors, including IL6, hepatocyte growth factor (HGF), and VEGF, and matrix metalloproteinases (MMP), such as MMP-9 and MMP-14, that can accelerate tumor cell invasion into and within the parenchymal space as well as promote tumor cell proliferation and angiogenesis, thereby enhancing liver metastasis (see Table 1; Figs. 2 and 3).

Taken together, the evidence suggests that KC targeting could become an effective antimetastatic strategy only within a very narrow window. Limiting KC activity may be beneficial if it could prevent premetastatic niche formation, whereas enhancing the KC tumoricidal potential using agents such as IFNγ, GM-CSF, or muramyl dipeptide may be most effective if achieved before tumor cells enter the liver in large numbers (47, 55). The contribution of CTCs that may be present before liver micrometastases are established or after surgical resection of the primary tumor (16, 17) to the tumor load and KC activation is unknown. This multifaceted role of KCs in liver metastasis may explain the limited success to date of KC-targeting interventions (47).

Importantly, in addition to KCs, blood-derived monocytes can also be recruited to the tumor site in response to local injury and inflammation and differentiate locally into mature CD11bloF4/80hi macrophages in a CCR2-dependent manner (56). These macrophages can also trigger HSC activation and fibrogenesis (57), a process important in the early stages of extravascular tumor expansion (see below).

Macrophages have inherent plasticity, and their phenotypes can change within a spectrum of activation states between the polar M1 and M2 phenotypes (58, 59). M1 macrophages are tumoricidal due to high NO and TNFα production levels and produce Th1, Th17, and the NK-attracting chemokines CXCL9 and CXCL10 (60). In contrast, M2 macrophages can promote tumor growth through production of growth factors, such as VEGF, EGF, bFGF, and TGFβ (61). In addition, M1 macrophages activate Th1-type immune responses that can further amplify M1/killer-type activity through the production of IFNγ, whereas M2 macrophages induce regulatory T cells (Treg) and thereby an immunotolerant microenvironment through release of TGFβ and IL10 (Table 1; ref. 62). Evidence regarding the role of macrophage polarization in liver metastasis is scant, because macrophages within or around hepatic metastases have, with few exceptions, not been subtyped. A recent clinical study identified the M2/M1 ratio in hepatic resections as a correlate of colorectal cancer metastasis (63), implicating M2 macrophages in the clinical disease. Furthermore, F4/80, the cell surface marker frequently used to identify KCs, is also expressed on recruited monocytes, and strategies used to eliminate macrophages in vivo are not KC specific. The source and identities of macrophages in many published studies remain therefore to be verified. As immune editing of the tumor microenvironment and the conversion of tumor-associated (TAM) M2 macrophages to M1 killer macrophages are emerging as potentially relevant anticancer strategies (62), further characterization of both the source and the status of macrophage populations involved in liver metastasis will become critical.

The neutrophils: A double-edged sword

Neutrophils are part of the innate immune response to pathogens and are also rapidly activated in response to invading cancer cells (28). Bone marrow–derived neutrophils are mobilized to sites of inflammation or cancer via their cell-surface receptor CXCR2 and in response to chemokines and cytokines (Table 1) secreted by activated macrophages, endothelial cells, or the tumor cells (reviewed in ref. 28). At the tumor site, the neutrophils can exert opposing effects that depend on the inflammatory/immune context (reviewed in ref. 64). They can release cytolytic factors (detailed in Table 1) or inhibit tumor growth indirectly via recruitment of CD8+ cytotoxic T cells and macrophages. This may require direct contact with the tumor cells (64), such as can occur within the liver sinusoidal lumen (65, 66).

On the other hand, neutrophils can be mobilized into premetastatic niches in the liver in response to S100A8 and S100A9 (7) and were shown to contribute to niche formation in an orthotropic colon carcinoma model (67). Moreover, in the vascular lumen, the physical interaction with tumor cells that could lead to tumor cell kill may, alternatively, anchor circulating tumor cells to the vascular endothelium and enhance their migration into the extravascular space. Tumor-derived cytokines, such as IL8, induce expression of integrins, such as CD11b/CD18, on the neutrophils, increasing their adhesion to tumor cells via the counter receptor ICAM-1 and facilitating transendothelial migration, as shown in murine melanoma and lung carcinoma models (66, 68). Tumor cell entrapment in the sinusoids can also be mediated by neutrophil-produced extracellular (DNA) traps, resulting in increased tumor retention in the sinusoids, enhanced tumor cell adhesion, proliferation, migration and invasion, and increased liver metastasis (69, 70). Neutrophils can also release extracellular matrix (ECM)–degrading proteinases, such as MMP-8, MMP-9, elastase, and cathepsin G, thereby increasing tumor invasion (64), and can promote angiogenesis through the release of VEGF (Table 1; Figs. 2 and 3).

Moreover, a TGFβ-mediated neutrophil polarization that alters the phenotype of tumor-associated neutrophils from tumor inhibitory (N1) to tumor promoting (N2) was recently described (71). TGFβ produced by M2 macrophages may contribute to this process (reviewed in ref. 59). However, the contribution of polarized neutrophils to liver metastasis remains to be confirmed.

Clinical studies implicate neutrophils in tumor promotion. With few exceptions, elevated circulating neutrophil counts or neutrophil-to-lymphocyte ratios were associated with poorer outcomes and distant metastases in patients with various epithelial malignancies (72), including carcinomas of the gastrointestinal tract. High neutrophil counts may also be associated with increased resistance to therapy (64, 72). The 5-year survival for patients with colorectal cancer undergoing hepatic resections that had neutrophil-to-lymphocyte ratios greater than 5 was worse than for those with normal ratios (73). However, high neutrophil infiltration into the tumor site may be a consequence of advanced tumor stage (and, therefore, poorer prognosis) rather than its cause.

HSCs orchestrate a prometastatic microenvironment that is required for transition from the avascular to the vascular stage of metastasis

HSCs orchestrate the characteristic fibrogenic response of the liver to injury. Normally quiescent in the space of Disse (74), they are activated (aHSC) in response to liver damage and inflammatory stimuli, acquire a myofibroblast-like phenotype (α-SMA+), and produce ECM rich in collagen I and IV (74, 75). Chemokines and cytokines released by aHSCs (Table 1) also recruit inflammatory/immune cells (76, 77), thereby shaping the immune microenvironment.

In addition to their role in premetastatic niche formation (above), HSCs can orchestrate a prometastatic niche following tumor extravasation. In response to growth factors and inflammatory mediators (detailed in Table 1), HSCs are activated (74) and can trigger a process akin to the early events in liver repair. This was observed in animal models (4) and is also evidenced by the increased production of collagen IV in and around hepatic metastases in clinical specimens (78). Macrophages, hepatocytes, and LSECs contribute to this process by releasing TGFβ and/or TNFα (57). In turn, aHSC-derived proangiogenic factors, such as VEGF and angiopoietin-1 (79, 80), initiate angiogenesis (3), and this is enhanced by HSC-derived MMPs that facilitate endothelial cell migration and tumor invasion (23, 74; reviewed in refs. 5, 81; Table 1; Fig. 2).

Several lines of evidence confirm the essential role of HSCs in liver metastasis. Olaso and colleagues showed that myofibroblasts that infiltrated avascular micrometastases of B16 melanoma cells induced a stromal response favorable to angiogenesis, which preceded endothelial cell recruitment and was followed by colocalization of myofibroblasts and endothelial cells within angiogenic structures (82, 83). HSC and macrophage recruitment into metastatic sites and subsequent angiogenesis were shown to be CCR2/CCL2 dependent (84). More recently, Eveno and colleagues (85), using immunofluorescence, showed that 9 days postinjection of colorectal cancer LS174 cells into SCID mice, the hepatic micrometastases consisted of proliferating cancer cells, a well-organized network of aHSC and laminin deposits, but no vascular network. As the liver metastases grew, an organized vascular network appeared and laminin colocalized with CD31+ endothelial cells. Coinjection of tumor cells and aHSCs enhanced metastasis. Moreover, analysis of liver metastasis from patients with colorectal cancer revealed a surface-marker expression pattern similar to that observed in the coinjection studies.

Recently, suppression of TGFβ receptor II signaling by QGAP1 was shown to prevent HSC activation, and IQGAP1 downregulation was observed in myofibroblasts associated with human colorectal cancer liver metastases, consistent with their activation in this context (86). Furthermore, α-SMA+ cells, whose presence correlated with the degree of fibrous stroma, were identified in liver metastases of colon, gastric, and pancreatic adenocarcinomas (87).

The importance of HSCs to metastatic niche formation and their role in primary liver cancer (88) have raised interest in HSC targeting as an anticancer/antimetastatic strategy. There is little direct evidence that such an approach can succeed, due partially to the present lack of agents that specifically target HSCs without toxicity. Indirect evidence from a rat model of cholangiocarcinoma (89) and pancreatic stellate cell targeting in a PDAC model (81) suggests a potential therapeutic benefit, but this remains to be verified. Of note, portal fibroblasts and bone marrow–derived fibrocytes (90) can also contribute to the stromal response. Liver-invading cancer cells located in portal tracts and unable to activate HSCs may engage portal tract fibroblasts that produce IL8, a chemokine involved in invasion and angiogenesis (91). Specific targeting of HSCs may, therefore, not be sufficient to deprive metastatic cells of a growth-promoting stroma.

Hepatocytes can promote metastasis directly and indirectly

The role of the parenchymal hepatocytes in liver metastasis is not well understood. Tumor cell adhesion to hepatocytes was identified as one of the earliest events in liver metastasis formation and a correlate of the metastatic potential (92, 93). Desmosomes (94), integrins αv, α6, and β1 that bind to hepatocyte ECM (93), osteopontin binding via counter receptors CD44 and integrin αv (95), and claudins (96, 97) were all implicated. Adhesion of human colorectal cancer cells to hepatocyte-derived ECM was shown to upregulate the expression of genes involved in tumor cell survival, motility, and proliferation, in particular EGF family members implicated in liver metastasis (98) such as AREG, EGFR, HBEGF, and erbB2 and stem cell markers CD133 (PROM1) and LGR5 (99), suggesting that colorectal cancer adhesion to hepatocyte ECM induces autocrine growth-promoting mechanisms for tumor expansion. Tumor cell adhesion to hepatocytes can have other consequences. The FasL/FLICE-like inhibitory protein on murine MC38 cells was shown to induce apoptosis in hepatocytes by activating Fas signaling, and this destructive process created a niche for tumor expansion (100). Furthermore, hepatocytes produce several growth factors, including IGF-I (101). As we have shown, blockade of IGF-I signaling in metastatic tumor cells could inhibit liver metastasis (102–104). Other factors include the HGF-like protein/macrophage-stimulating protein (HGFL) that can enhance tumor growth, motility, and invasion via the Ron receptor (105); heregulin, a ligand of ErbB3 shown to enhance integrin αvβ5-mediated cell migration and erbB3/erbB2 signaling in human colorectal cancer cells (106); and the proangiogenic factor bFGF (107).

The clinical relevance of tumor–hepatocyte interactions is not clear. Three different histologic growth patterns have been identified in liver metastases specimens, namely the desmoplastic, pushing, and replacement growth patterns. The latter two are characterized by close tumor–hepatocyte proximity (23). Whether or not direct adhesion between metastatic cancer cells and hepatocytes influences the eventual growth pattern of colorectal cancer liver metastases is not known. Blocking tumor–hepatocyte interactions could have beneficial antimetastatic effects, as recently demonstrated by Tabaries and colleagues (108).

An immunosuppressive microenvironment facilitates metastatic expansion

Tumor cells can activate specific T-cell–mediated immune responses that curtail tumor expansion through various mechanisms (reviewed in ref. 109). However, tumor cells can evade T-cell–mediated killing, among others, by giving rise to and/or recruiting immunosuppressive cells, such as myeloid-derived suppressor cells (MDSC) and regulatory T cells (Treg) that alter the immune landscape, inducing a state of immune tolerance that is permissive to tumor expansion (reviewed in ref. 110).

The role of Tregs

Treg cells may be thymically derived (natural, nTregs) or induced from naïve CD4+ T-cell precursors (iTreg) in response to IL10 and TGFβ. They exert an immunosuppressive effect by different mechanisms that result in the inhibition of an effective, antitumorigenic T-cell response (reviewed in ref. 111). Although high Treg levels were documented in the blood and local tumor sites of patients with various epithelial cancers (e.g., ref. 112), relatively little is known about their role in liver metastasis. Connolly and colleagues, using models of early, preinvasive pancreatic neoplasia and advanced colorectal cancer showed that Tregs accelerated the development of liver metastases (113). We recently documented the accumulation of Tregs around colon carcinoma MC38 liver micrometastases and showed that this was TNFR2 dependent (35).

Intriguingly, in 57 colon cancer specimens from patients undergoing chemotherapy or chemoimmunotherapy, higher Treg infiltration scores were associated with a better prognosis and a better outcome (114). However, the relationship between Treg accumulation in the primary tumors and in the corresponding liver metastases has not been examined. Although Treg-targeting strategies are in development (115, 116; reviewed in ref. 110), their potential therapeutic benefit for liver metastases has not yet been examined.

The role of MDSCs

MDSCs are a heterogeneous population of myeloid cells that can be of the monocytic (Mo-MDSC, CD11b+Ly6C+) or granulocytic (G-MDSC, CD11b+Ly6G+) lineages (117). In humans, MDSCs are CD33+ and/or CD11b+ and HLA-DR (118). Under normal physiologic conditions, bone marrow–derived immature myeloid cells differentiate into mature granulocytes or monocytes, able to mediate host innate immune responses in the target tissue. However, in the tumor microenvironment, these precursors do not mature and exert immunosuppressive and tumor-promoting effects instead (119). Although they express granulocyte and monocyte surface markers, these cells have been characterized on the basis of functional criteria, namely their immunosuppressive capabilities (117, 120). An increase in circulating MDSCs in cancer patients and their recruitment into tumor sites have been documented (117, 121, 122).

In the liver, MDSCs can be recruited to the metastases through chemokines, such as CXCL1 and CXCL2, produced by LSECs and KCs or by activated HSCs (77). There, they can promote tumor growth by inducing immunosuppression (123), producing arginase (117), and increasing Treg numbers (Table 1; Fig. 2; ref. 124). Clinical and experimental evidence confirms their role in metastasis (125). Recruitment of tumor-promoting myeloid cells into colorectal cancer liver metastases has recently been documented, and their depletion was shown to result in a marked reduction in liver metastasis (126). We recently reported that MDSCs accumulate in hepatic micrometastases of MC38 cells within days of tumor cell injection and documented a relative preponderance of Ly6Chigh cells (the more suppressive, arginase-high subtype; ref. 127). We also identified CD33+HLA-DRTNFR2+ myeloid cells in the periphery of hepatic metastases from patients with colorectal cancer, implicating MDSCs in the clinical disease (35).

Several strategies are being developed to selectively eliminate MDSCs, including pharmacologic induction of MDSC differentiation, inhibition of MDSC expansion from bone marrow precursors, or blockade of their function (128). However, some of the drugs developed are not MDSC specific, or their long-term administration is not possible due to toxicity (128). Their translational potential remains therefore to be verified.

Cancer cells entering the liver encounter a unique and complex microenvironment. Parenchymal and nonparenchymal liver cells, as well as recruited inflammatory and immune cells, participate in the response to invading tumor cells and may inhibit or favor the progression of metastasis. The factors that determine the outcome of these opposing effects and the pattern of growth of the metastases are not yet well understood. This duality of function and the shifts in the types of cells and mediators involved at different stages of the metastatic process render the attempts to target specific cellular interactions in the microenvironment extremely challenging and may explain the slow progress to date in identifying agents that can successfully and specifically inhibit metastatic disease. With the advent of genomic profiling for personalized cancer treatment and the increased availability of surgically resected liver metastases for cellular/molecular interrogation, our understanding of the relationship between the genetic background and clinical history of the patient, the genomic/transcriptomic profile of the cancer cells, and the type of response mounted by the microenvironment may improve, opening new avenues for prevention and/or treatment of liver metastatic disease.

No potential conflicts of interest were disclosed.

The author is indebted to Dr. Janusz Rak (McGill University Health Centre) for insightful editorial comments and to Simon Milette for the superb artistic rendering of the metastatic niches of the liver and for his help with the table.

This work was made possible by support from the Canadian Institute for Health Research and by a PSR-SIIRI-843 grant from the Québec Ministère de l'Économie, de l'Innovation et des Exportations.

1.
Hess
KR
,
Varadhachary
GR
,
Taylor
SH
,
Wei
W
,
Raber
MN
,
Lenzi
R
, et al
Metastatic patterns in adenocarcinoma
.
Cancer
2006
;
106
:
1624
33
.
2.
Kmiec
Z
. 
Cooperation of liver cells in health and disease
.
Adv Anat Embryol Cell Biol
2001
;
161
:
III
XIII
.
3.
Smedsrod
B
,
Le Couteur
D
,
Ikejima
K
,
Jaeschke
H
,
Kawada
N
,
Naito
M
, et al
Hepatic sinusoidal cells in health and disease: update from the 14th International Symposium
.
Liver Int
2009
;
29
:
490
501
.
4.
Vidal-Vanaclocha
F
. 
The prometastatic microenvironment of the liver
.
Cancer Microenviron
2008
;
1
:
113
29
.
5.
Vidal-Vanaclocha
F
. 
The tumor microenvironment at different stages of hepatic metastasis
.
In
:
Brodt
P
,
editor
.
Liver metastasis: biology and clinical management
.
Dordrecht, the Netherlands
:
Springer Science+Business Media B.V.
; 
2011
.
p.
1
.
6.
Kaplan
RN
,
Riba
RD
,
Zacharoulis
S
,
Bramley
AH
,
Vincent
L
,
Costa
C
, et al
VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche
.
Nature
2005
;
438
:
820
7
.
7.
Hiratsuka
S
,
Watanabe
A
,
Aburatani
H
,
Maru
Y
. 
Tumour-mediated upregulation of chemoattractants and recruitment of myeloid cells predetermines lung metastasis
.
Nat Cell Biol
2006
;
8
:
1369
75
.
8.
Hiratsuka
S
,
Watanabe
A
,
Sakurai
Y
,
Akashi-Takamura
S
,
Ishibashi
S
,
Miyake
K
, et al
The S100A8-serum amyloid A3-TLR4 paracrine cascade establishes a pre-metastatic phase
.
Nat Cell Biol
2008
;
10
:
1349
55
.
9.
Psaila
B
,
Lyden
D
. 
The metastatic niche: adapting the foreign soil
.
Nat Rev Cancer
2009
;
9
:
285
93
.
10.
Sceneay
J
,
Smyth
MJ
,
Moller
A
. 
The pre-metastatic niche: finding common ground
.
Cancer Metastasis Rev
2013
;
32
:
449
64
.
11.
Costa-Silva
B
,
Aiello
NM
,
Ocean
AJ
,
Singh
S
,
Zhang
H
,
Thakur
BK
, et al
Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver
.
Nat Cell Biol
2015
;
17
:
816
26
.
12.
Hoshino
A
,
Costa-Silva
B
,
Shen
TL
,
Rodrigues
G
,
Hashimoto
A
,
Tesic Mark
M
, et al
Tumour exosome integrins determine organotropic metastasis
.
Nature
2015
;
527
:
329
35
.
13.
Kowanetz
M
,
Wu
X
,
Lee
J
,
Tan
M
,
Hagenbeek
T
,
Qu
X
, et al
Granulocyte-colony stimulating factor promotes lung metastasis through mobilization of Ly6G+Ly6C+ granulocytes
.
Proc Natl Acad Sci U S A
2010
;
107
:
21248
55
.
14.
Seubert
B
,
Grunwald
B
,
Kobuch
J
,
Cui
H
,
Schelter
F
,
Schaten
S
, et al
Tissue inhibitor of metalloproteinases (TIMP)-1 creates a premetastatic niche in the liver through SDF-1/CXCR4-dependent neutrophil recruitment in mice
.
Hepatology
2015
;
61
:
238
48
.
15.
Zhang
Y
,
Davis
C
,
Ryan
J
,
Janney
C
,
Peña
MMO
. 
Development and characterization of a reliable mouse model of colorectal cancer metastasis to the liver
.
Clin Exp Metastasis
2013
;
30
:
903
18
.
16.
Bork
U
,
Rahbari
NN
,
Scholch
S
,
Reissfelder
C
,
Kahlert
C
,
Buchler
MW
, et al
Circulating tumour cells and outcome in non-metastatic colorectal cancer: a prospective study
.
Br J Cancer
2015
;
112
:
1306
13
.
17.
Tsai
WS
,
Chen
JS
,
Shao
HJ
,
Wu
JC
,
Lai
JM
,
Lu
SH
, et al
Circulating tumor cell count correlates with colorectal neoplasm progression and is a prognostic marker for distant metastasis in non-metastatic patients
.
Sci Rep
2016
;
6
:
24517
.
18.
Braet
F
,
Nagatsuma
K
,
Saito
M
,
Soon
L
,
Wisse
E
,
Matsuura
T
. 
The hepatic sinusoidal endothelial lining and colorectal liver metastases
.
World J Gastroenterol
2007
;
13
:
821
5
.
19.
Wang
HH
,
McIntosh
AR
,
Hasinoff
BB
,
Rector
ES
,
Ahmed
N
,
Nance
DM
, et al
B16 melanoma cell arrest in the mouse liver induces nitric oxide release and sinusoidal cytotoxicity: a natural hepatic defense against metastasis
.
Cancer Res
2000
;
60
:
5862
9
.
20.
Yanagida
H
,
Kaibori
M
,
Yoshida
H
,
Habara
K
,
Yamada
M
,
Kamiyama
Y
, et al
Hepatic ischemia/reperfusion upregulates the susceptibility of hepatocytes to confer the induction of inducible nitric oxide synthase gene expression
.
Shock
2006
;
26
:
162
8
.
21.
Hehlgans
T
,
Pfeffer
K
. 
The intriguing biology of the tumour necrosis factor/tumour necrosis factor receptor superfamily: players, rules and the games
.
Immunology
2005
;
115
:
1
20
.
22.
Ramadori
G
,
Moriconi
F
,
Malik
I
,
Dudas
J
. 
Physiology and pathophysiology of liver inflammation, damage and repair
.
J Physiol Pharmacol
2008
;
59
Suppl 1
:
107
17
.
23.
Van den Eynden
GG
,
Majeed
AW
,
Illemann
M
,
Vermeulen
PB
,
Bird
NC
,
Hoyer-Hansen
G
, et al
The multifaceted role of the microenvironment in liver metastasis: biology and clinical implications
.
Cancer Res
2013
;
73
:
2031
43
.
24.
Bertin
S
,
Neves
S
,
Gavelli
A
,
Baque
P
,
Brossette
N
,
Simoes
S
, et al
Cellular and molecular events associated with the antitumor response induced by the cytosine deaminase/5-fluorocytosine suicide gene therapy system in a rat liver metastasis model
.
Cancer Gene Ther
2007
;
14
:
858
66
.
25.
Takehara
T
,
Uemura
A
,
Tatsumi
T
,
Suzuki
T
,
Kimura
R
,
Shiotani
A
, et al
Natural killer cell-mediated ablation of metastatic liver tumors by hydrodynamic injection of IFNalpha gene to mice
.
Int J Cancer
2007
;
120
:
1252
60
.
26.
Tatsumi
T
,
Takehara
T
,
Yamaguchi
S
,
Sasakawa
A
,
Miyagi
T
,
Jinushi
M
, et al
Injection of IL-12 gene-transduced dendritic cells into mouse liver tumor lesions activates both innate and acquired immunity
.
Gene Ther
2007
;
14
:
863
71
.
27.
Pfister
K
,
Radons
J
,
Busch
R
,
Tidball
JG
,
Pfeifer
M
,
Freitag
L
, et al
Patient survival by Hsp70 membrane phenotype: association with different routes of metastasis
.
Cancer
2007
;
110
:
926
35
.
28.
Spicer
J
,
Brodt
P
,
Ferri
LE
. 
Role of inflammation in the early stages of liver metastasis
.
In
:
Brodt
P
,
editor
.
Liver metastasis: biology and clinical management
.
New York
:
Springer
; 
2011
.
p.
155
85
.
29.
Paschos
KA
,
Majeed
AW
,
Bird
NC
. 
Natural history of hepatic metastases from colorectal cancer - pathobiological pathways with clinical significance
.
World J Gastroenterol
2014
;
20
:
3719
37
.
30.
Glinskii
OV
,
Huxley
VH
,
Glinsky
GV
,
Pienta
KJ
,
Raz
A
,
Glinsky
VV
. 
Mechanical entrapment is insufficient and intercellular adhesion is essential for metastatic cell arrest in distant organs
.
Neoplasia
2005
;
7
:
522
7
.
31.
Brodt
P
. 
Role of the host inflammatory response in colon carcinoma initiation, progression and liver metastasis
.
In
:
Beauchemin
N
,
Huot
J
,
editors
.
Metastasis of colorectal cancer
.
New York, NY
:
Springer
; 
2010
.
p.
289
319
.
32.
Jiao
S-F
,
Sun
K
,
Chen
X-J
,
Zhao
X
,
Cai
N
,
Liu
Y-J
, et al
Inhibition of tumor necrosis factor alpha reduces the outgrowth of hepatic micrometastasis of colorectal tumors in a mouse model of liver ischemia-reperfusion injury
.
J Biomed Sci
2014
;
21
:
1
.
33.
Khatib
AM
,
Fallavollita
L
,
Wancewicz
EV
,
Monia
BP
,
Brodt
P
. 
Inhibition of hepatic endothelial E-selectin expression by C-raf antisense oligonucleotides blocks colorectal carcinoma liver metastasis
.
Cancer Res
2002
;
62
:
5393
8
.
34.
Yoshimoto
K
,
Tajima
H
,
Ohta
T
,
Okamoto
K
,
Sakai
S
,
Kinoshita
J
, et al
Increased E-selectin in hepatic ischemia-reperfusion injury mediates liver metastasis of pancreatic cancer
.
Oncol Rep
2012
;
28
:
791
6
.
35.
Ham
B
,
Wang
N
,
D'Costa
Z
,
Fernandez
MC
,
Bourdeau
F
,
Auguste
P
, et al
TNF receptor-2 facilitates an immunosuppressive microenvironment in the liver to promote the colonization and growth of hepatic metastases
.
Cancer Res
2015
;
75
:
5235
47
.
36.
Elliott
VA
,
Rychahou
P
,
Zaytseva
YY
,
Evers
BM
. 
Activation of c-Met and upregulation of CD44 expression are associated with the metastatic phenotype in the colorectal cancer liver metastasis model
.
PLoS One
2014
;
9
:
e97432
.
37.
Witz
IP
. 
The selectin-selectin ligand axis in tumor progression
.
Cancer Metastasis Rev
2008
;
27
:
19
30
.
38.
Greenwald
E
,
Yuki
K
. 
A translational consideration of intercellular adhesion molecule-1 biology in the perioperative setting
.
Transl Perioper Pain Med
2016
;
1
:
17
23
.
39.
Tremblay
PL
,
Huot
J
,
Auger
FA
. 
Mechanisms by which E-selectin regulates diapedesis of colon cancer cells under flow conditions
.
Cancer Res
2008
;
68
:
5167
76
.
40.
Aychek
T
,
Miller
K
,
Sagi-Assif
O
,
Levy-Nissenbaum
O
,
Israeli-Amit
M
,
Pasmanik-Chor
M
, et al
E-selectin regulates gene expression in metastatic colorectal carcinoma cells and enhances HMGB1 release
.
Int J Cancer
2008
;
123
:
1741
50
.
41.
Ou
J
,
Peng
Y
,
Deng
J
,
Miao
H
,
Zhou
J
,
Zha
L
, et al
Endothelial cell-derived fibronectin extra domain A promotes colorectal cancer metastasis via inducing epithelial-mesenchymal transition
.
Carcinogenesis
2014
;
35
:
1661
70
.
42.
Hu
CT
,
Guo
LL
,
Feng
N
,
Zhang
L
,
Zhou
N
,
Ma
LL
, et al
MIF, secreted by human hepatic sinusoidal endothelial cells, promotes chemotaxis and outgrowth of colorectal cancer in liver prometastasis
.
Oncotarget
2015
;
6
:
22410
23
.
43.
Banerjee
D
,
Hernandez
SL
,
Garcia
A
,
Kangsamaksin
T
,
Sbiroli
E
,
Andrews
J
, et al
Notch suppresses angiogenesis and progression of hepatic metastases
.
Cancer Res
2015
;
75
:
1592
602
.
44.
Im
JH
,
Tapmeier
T
,
Balathasan
L
,
Gal
A
,
Yameen
S
,
Hill
S
, et al
G-CSF rescues tumor growth and neo-angiogenesis during liver metastasis under host angiopoietin-2 deficiency
.
Int J Cancer
2013
;
132
:
315
26
.
45.
Yona
S
,
Kim
KW
,
Wolf
Y
,
Mildner
A
,
Varol
D
,
Breker
M
, et al
Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis
.
Immunity
2013
;
38
:
79
91
.
46.
Bouwens
L
,
Baekeland
M
,
De Zanger
R
,
Wisse
E
. 
Quantitation, tissue distribution and proliferation kinetics of Kupffer cells in normal rat liver
.
Hepatology
1986
;
6
:
718
22
.
47.
van der Bij
GJ
,
Oosterling
SJ
,
Meijer
S
,
Beelen
RH
,
van Egmond
M
. 
Therapeutic potential of Kupffer cells in prevention of liver metastases outgrowth
.
Immunobiology
2005
;
210
:
259
65
.
48.
Timmers
M
,
Vekemans
K
,
Vermijlen
D
,
Asosingh
K
,
Kuppen
P
,
Bouwens
L
, et al
Interactions between rat colon carcinoma cells and Kupffer cells during the onset of hepatic metastasis
.
Int J Cancer
2004
;
112
:
793
802
.
49.
Bayon
LG
,
Izquierdo
MA
,
Sirovich
I
,
van Rooijen
N
,
Beelen
RH
,
Meijer
S
. 
Role of Kupffer cells in arresting circulating tumor cells and controlling metastatic growth in the liver
.
Hepatology
1996
;
23
:
1224
31
.
50.
Matsumura
H
,
Kondo
T
,
Ogawa
K
,
Tamura
T
,
Fukunaga
K
,
Murata
S
, et al
Kupffer cells decrease metastasis of colon cancer cells to the liver in the early stage
.
Int J Oncol
2014
;
45
:
2303
10
.
51.
Khatib
AM
,
Auguste
P
,
Fallavollita
L
,
Wang
N
,
Samani
A
,
Kontogiannea
M
, et al
Characterization of the host proinflammatory response to tumor cells during the initial stages of liver metastasis
.
Am J Pathol
2005
;
167
:
749
59
.
52.
Khatib
AM
,
Kontogiannea
M
,
Fallavollita
L
,
Jamison
B
,
Meterissian
S
,
Brodt
P
. 
Rapid induction of cytokine and E-selectin expression in the liver in response to metastatic tumor cells
.
Cancer Res
1999
;
59
:
1356
61
.
53.
Wen
SW
,
Ager
EI
,
Christophi
C
. 
Bimodal role of Kupffer cells during colorectal cancer liver metastasis
.
Cancer Biol Ther
2013
;
14
:
606
13
.
54.
Jessup
JM
,
Samara
R
,
Battle
P
,
Laguinge
LM
. 
Carcinoembryonic antigen promotes tumor cell survival in liver through an IL-10-dependent pathway
.
Clin Exp Metastasis
2004
;
21
:
709
17
.
55.
Asao
T
,
Shibata
HR
,
Batist
G
,
Brodt
P
. 
Eradication of hepatic metastases of carcinoma H-59 by combination chemoimmunotherapy with liposomal muramyl tripeptide, 5-fluorouracil, and leucovorin
.
Cancer Res
1992
;
52
:
6254
7
.
56.
Tosello-Trampont
A-C
,
Landes
SG
,
Nguyen
V
,
Novobrantseva
TI
,
Hahn
YS
. 
Kuppfer cells trigger nonalcoholic steatohepatitis development in diet-induced mouse model through tumor necrosis factor-α production
.
J Biol Chem
2012
;
287
:
40161
72
.
57.
Dey
A
,
Allen
J
,
Hankey-Giblin
PA
. 
Ontogeny and polarization of macrophages in inflammation: blood monocytes versus tissue macrophages
.
Front Immunol
2014
;
5
:
683
.
58.
Murray
PJ
,
Allen
JE
,
Biswas
SK
,
Fisher
EA
,
Gilroy
DW
,
Goerdt
S
, et al
Macrophage activation and polarization: nomenclature and experimental guidelines
.
Immunity
2014
;
41
:
14
20
.
59.
Schouppe
E
,
De Baetselier
P
,
Van Ginderachter
JA
,
Sarukhan
A
. 
Instruction of myeloid cells by the tumor microenvironment: open questions on the dynamics and plasticity of different tumor-associated myeloid cell populations
.
Oncoimmunology
2012
;
1
:
1135
45
.
60.
Galdiero
MR
,
Bonavita
E
,
Barajon
I
,
Garlanda
C
,
Mantovani
A
,
Jaillon
S
. 
Tumor associated macrophages and neutrophils in cancer
.
Immunobiology
2013
;
218
:
1402
10
.
61.
Mills
CD
. 
Anatomy of a discovery: m1 and m2 macrophages
.
Front Immunol
2015
;
6
:
212
.
62.
Mills
CD
,
Lenz
LL
,
Harris
RA
. 
A breakthrough: macrophage-directed cancer immunotherapy
.
Cancer Res
2016
;
76
:
513
6
.
63.
Cui
YL
,
Li
HK
,
Zhou
HY
,
Zhang
T
,
Li
Q
. 
Correlations of tumor-associated macrophage subtypes with liver metastases of colorectal cancer
.
Asian Pac J Cancer Prev
2013
;
14
:
1003
7
.
64.
Sionov
RV
,
Fridlender
ZG
,
Granot
Z
. 
The multifaceted roles neutrophils play in the tumor microenvironment
.
Cancer Microenviron
2015
;
8
:
125
58
.
65.
McDonald
B
,
Spicer
J
,
Giannais
B
,
Fallavollita
L
,
Brodt
P
,
Ferri
LE
. 
Systemic inflammation increases cancer cell adhesion to hepatic sinusoids by neutrophil mediated mechanisms
.
Int J Cancer
2009
;
125
:
1298
305
.
66.
Spicer
JD
,
McDonald
B
,
Cools-Lartigue
JJ
,
Chow
SC
,
Giannias
B
,
Kubes
P
, et al
Neutrophils promote liver metastasis via Mac-1-mediated interactions with circulating tumor cells
.
Cancer Res
2012
;
72
:
3919
27
.
67.
Yamamoto
M
,
Kikuchi
H
,
Ohta
M
,
Kawabata
T
,
Hiramatsu
Y
,
Kondo
K
, et al
TSU68 prevents liver metastasis of colon cancer xenografts by modulating the premetastatic niche
.
Cancer Res
2008
;
68
:
9754
62
.
68.
Huh
SJ
,
Liang
S
,
Sharma
A
,
Dong
C
,
Robertson
GP
. 
Transiently entrapped circulating tumor cells interact with neutrophils to facilitate lung metastasis development
.
Cancer Res
2010
;
70
:
6071
82
.
69.
Cools-Lartigue
J
,
Spicer
J
,
McDonald
B
,
Gowing
S
,
Chow
S
,
Giannias
B
, et al
Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis
.
J Clin Invest
2013
;
123
:
3446
58
.
70.
Tohme
S
,
Yazdani
HO
,
Al-Khafaji
AB
,
Chidi
AP
,
Loughran
P
,
Mowen
K
, et al
Neutrophil extracellular traps promote the development and progression of liver metastases after surgical stress
.
Cancer Res
2016
;
76
:
1367
80
.
71.
Fridlender
ZG
,
Sun
J
,
Kim
S
,
Kapoor
V
,
Cheng
G
,
Ling
L
, et al
Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN
.
Cancer Cell
2009
;
16
:
183
94
.
72.
Donskov
F
. 
Immunomonitoring and prognostic relevance of neutrophils in clinical trials
.
Semin Cancer Biol
2013
;
23
:
200
7
.
73.
Halazun
KJ
,
Aldoori
A
,
Malik
HZ
,
Al-Mukhtar
A
,
Prasad
KR
,
Toogood
GJ
, et al
Elevated preoperative neutrophil to lymphocyte ratio predicts survival following hepatic resection for colorectal liver metastases
.
Eur J Surg Oncol
2008
;
34
:
55
60
.
74.
Friedman
SL
. 
Hepatic stellate cells: protean, multifunctional, and enigmatic cells of the liver
.
Physiol Rev
2008
;
88
:
125
72
.
75.
Gressner
AM
,
Bachem
MG
. 
Molecular mechanisms of liver fibrogenesis–a homage to the role of activated fat-storing cells
.
Digestion
1995
;
56
:
335
46
.
76.
Muhanna
N
,
Doron
S
,
Wald
O
,
Horani
A
,
Eid
A
,
Pappo
O
, et al
Activation of hepatic stellate cells after phagocytosis of lymphocytes: a novel pathway of fibrogenesis
.
Hepatology
2008
;
48
:
963
77
.
77.
Zhao
W
,
Zhang
L
,
Xu
Y
,
Zhang
Z
,
Ren
G
,
Tang
K
, et al
Hepatic stellate cells promote tumor progression by enhancement of immunosuppressive cells in an orthotopic liver tumor mouse model
.
Lab Invest
2014
;
94
:
182
91
.
78.
Burnier
JV
,
Wang
N
,
Michel
RP
,
Hassanain
M
,
Li
S
,
Lu
Y
, et al
Type IV collagen-initiated signals provide survival and growth cues required for liver metastasis
.
Oncogene
2011
;
30
:
3766
83
.
79.
Copple
BL
,
Bai
S
,
Burgoon
LD
,
Moon
J-OK
. 
Hypoxia-inducible factor-1α regulates expression of genes in hypoxic hepatic stellate cells important for collagen deposition and angiogenesis
.
Liver Int
2011
;
31
:
230
44
.
80.
Taura
K
,
De Minicis
S
,
Seki
E
,
Hatano
E
,
Iwaisako
K
,
Osterreicher
CH
, et al
Hepatic stellate cells secrete angiopoietin 1 that induces angiogenesis in liver fibrosis
.
Gastroenterology
2008
;
135
:
1729
38
.
81.
Kang
N
,
Shah
VH
,
Urrutia
R
. 
Membrane-to-nucleus signals and epigenetic mechanisms for myofibroblastic activation and desmoplastic stroma: potential therapeutic targets for liver metastasis?
Mol Cancer Res
2015
;
13
:
604
12
.
82.
Olaso
E
,
Salado
C
,
Egilegor
E
,
Gutierrez
V
,
Santisteban
A
,
Sancho-Bru
P
, et al
Proangiogenic role of tumor-activated hepatic stellate cells in experimental melanoma metastasis
.
Hepatology
2003
;
37
:
674
85
.
83.
Badiola
I
,
Olaso
E
,
Crende
O
,
Friedman
SL
,
Vidal-Vanaclocha
F
. 
Discoidin domain receptor 2 deficiency predisposes hepatic tissue to colon carcinoma metastasis
.
Gut
2012
;
61
:
1465
72
.
84.
Yang
X
,
Lu
P
,
Ishida
Y
,
Kuziel
WA
,
Fujii
C
,
Mukaida
N
. 
Attenuated liver tumor formation in the absence of CCR2 with a concomitant reduction in the accumulation of hepatic stellate cells, macrophages and neovascularization
.
Int J Cancer
2006
;
118
:
335
45
.
85.
Eveno
C
,
Hainaud
P
,
Rampanou
A
,
Bonnin
P
,
Bakhouche
S
,
Dupuy
E
, et al
Proof of prometastatic niche induction by hepatic stellate cells
.
J Surg Res
2015
;
194
:
496
504
.
86.
Liu
C
,
Billadeau
DD
,
Abdelhakim
H
,
Leof
E
,
Kaibuchi
K
,
Bernabeu
C
, et al
IQGAP1 suppresses TbetaRII-mediated myofibroblastic activation and metastatic growth in liver
.
J Clin Invest
2013
;
123
:
1138
56
.
87.
Terada
T
,
Makimoto
K
,
Terayama
N
,
Suzuki
Y
,
Nakanuma
Y
. 
Alpha-smooth muscle actin-positive stromal cells in cholangiocarcinomas, hepatocellular carcinomas and metastatic liver carcinomas
.
J Hepatol
1996
;
24
:
706
12
.
88.
Ju
MJ
,
Qiu
SJ
,
Fan
J
,
Xiao
YS
,
Gao
Q
,
Zhou
J
, et al
Peritumoral activated hepatic stellate cells predict poor clinical outcome in hepatocellular carcinoma after curative resection
.
Am J Clin Pathol
2009
;
131
:
498
510
.
89.
Sirica
AE
,
Gores
GJ
. 
Desmoplastic stroma and cholangiocarcinoma: clinical implications and therapeutic targeting
.
Hepatology
2014
;
59
:
2397
402
.
90.
Brenner
DA
,
Kisseleva
T
,
Scholten
D
,
Paik
YH
,
Iwaisako
K
,
Inokuchi
S
, et al
Origin of myofibroblasts in liver fibrosis
.
Fibrogenesis Tissue Repair
2012
;
5
(
Suppl 1
):
S17
.
91.
Mueller
L
,
Goumas
FA
,
Affeldt
M
,
Sandtner
S
,
Gehling
UM
,
Brilloff
S
, et al
Stromal fibroblasts in colorectal liver metastases originate from resident fibroblasts and generate an inflammatory microenvironment
.
Am J Pathol
2007
;
171
:
1608
18
.
92.
Wang
J
,
Fallavollita
L
,
Brodt
P
. 
Inhibition of experimental hepatic metastasis by a monoclonal antibody that blocks tumor-hepatocyte interaction
.
J Immunother Emphasis Tumor Immunol
1994
;
16
:
294
302
.
93.
Mook
ORF
,
van Marie
J
,
Jonges
R
,
Vreeling-Sindelarova
H
,
Frederiks
WM
,
Van Noorden
CJF
. 
Interactions between colon cancer cells and hepatocytes in rats in relation to metastasis
.
J Cell Mol Med
2008
;
12
:
2052
61
.
94.
Shimizu
S
,
Yamada
N
,
Sawada
T
,
Ikeda
K
,
Nakatani
K
,
Seki
S
, et al
Ultrastructure of early phase hepatic metastasis of human colon carcinoma cells with special reference to desmosomal junctions with hepatocytes
.
Pathol Int
2000
;
50
:
953
9
.
95.
Huang
J
,
Pan
C
,
Hu
H
,
Zheng
S
,
Ding
L
. 
Osteopontin-enhanced hepatic metastasis of colorectal cancer cells
.
PLoS One
2012
;
7
:
e47901
.
96.
Tabaries
S
,
Dupuy
F
,
Dong
Z
,
Monast
A
,
Annis
MG
,
Spicer
J
, et al
Claudin-2 promotes breast cancer liver metastasis by facilitating tumor cell interactions with hepatocytes
.
Mol Cell Biol
2012
;
32
:
2979
91
.
97.
Georges
R
,
Bergmann
F
,
Hamdi
H
,
Zepp
M
,
Eyol
E
,
Hielscher
T
, et al
Sequential biphasic changes in claudin1 and claudin4 expression are correlated to colorectal cancer progression and liver metastasis
.
J Cell Mol Med
2012
;
16
:
260
72
.
98.
Radinsky
R
,
Risin
S
,
Fan
D
,
Dong
Z
,
Bielenberg
D
,
Bucana
CD
, et al
Level and function of epidermal growth factor receptor predict the metastatic potential of human colon carcinoma cells
.
Clin Cancer Res
1995
;
1
:
19
31
.
99.
Zvibel
I
,
Wagner
A
,
Pasmanik-Chor
M
,
Varol
C
,
Oron-Karni
V
,
Santo
EM
, et al
Transcriptional profiling identifies genes induced by hepatocyte-derived extracellular matrix in metastatic human colorectal cancer cell lines
.
Clin Exp Metastasis
2013
;
30
:
189
200
.
100.
Li
H
,
Fan
X
,
Stoicov
C
,
Liu
JH
,
Zubair
S
,
Tsai
E
, et al
Human and mouse colon cancer utilizes CD95 signaling for local growth and metastatic spread to liver
.
Gastroenterology
2009
;
137
:
934
44
.
101.
Long
L
,
Nip
J
,
Brodt
P
. 
Paracrine growth stimulation by hepatocyte-derived insulin-like growth factor-1: a regulatory mechanism for carcinoma cells metastatic to the liver
.
Cancer Res
1994
;
54
:
3732
7
.
102.
Long
L
,
Rubin
R
,
Baserga
R
,
Brodt
P
. 
Loss of the metastatic phenotype in murine carcinoma cells expressing an antisense RNA to the insulin-like growth factor receptor
.
Cancer Res
1995
;
55
:
1006
9
.
103.
Samani
AA
,
Chevet
E
,
Fallavollita
L
,
Galipeau
J
,
Brodt
P
. 
Loss of tumorigenicity and metastatic potential in carcinoma cells expressing the extracellular domain of the type 1 insulin-like growth factor receptor
.
Cancer Res
2004
;
64
:
3380
5
.
104.
Wang
N
,
Rayes
RF
,
Elahi
SM
,
Lu
Y
,
Hancock
MA
,
Massie
B
, et al
The IGF-Trap: novel inhibitor of carcinoma growth and metastasis
.
Mol Cancer Ther
2015
;
14
:
982
93
.
105.
Wagh
P
,
Peace
BE
,
Waltz
SE
. 
The met-related receptor tyrosine kinase Ron in tumor growth and metastasis
.
Adv Cancer Res
2008
;
100
:
1
33
.
106.
Yoshioka
T
,
Nishikawa
Y
,
Ito
R
,
Kawamata
M
,
Doi
Y
,
Yamamoto
Y
, et al
Significance of integrin alphavbeta5 and erbB3 in enhanced cell migration and liver metastasis of colon carcinomas stimulated by hepatocyte-derived heregulin
.
Cancer Sci
2010
;
101
:
2011
8
.
107.
Dome
B
,
Hendrix
MJ
,
Paku
S
,
Tovari
J
,
Timar
J
. 
Alternative vascularization mechanisms in cancer: Pathology and therapeutic implications
.
Am J Pathol
2007
;
170
:
1
15
.
108.
Tabaries
S
,
Annis
MG
,
Hsu
BE
,
Tam
CE
,
Savage
P
,
Park
M
, et al
Lyn modulates Claudin-2 expression and is a therapeutic target for breast cancer liver metastasis
.
Oncotarget
2015
;
6
:
9476
87
.
109.
Hadrup
S
,
Donia
M
,
Thor Straten
P
. 
Effector CD4 and CD8 T cells and their role in the tumor microenvironment
.
Cancer Microenviron
2013
;
6
:
123
33
.
110.
Butt
AQ
,
Mills
KH
. 
Immunosuppressive networks and checkpoints controlling antitumor immunity and their blockade in the development of cancer immunotherapeutics and vaccines
.
Oncogene
2014
;
33
:
4623
31
.
111.
Facciabene
A
,
Motz
GT
,
Coukos
G
. 
T-regulatory cells: key players in tumor immune escape and angiogenesis
.
Cancer Res
2012
;
72
:
2162
71
.
112.
Wolf
AM
,
Wolf
D
,
Steurer
M
,
Gastl
G
,
Gunsilius
E
,
Grubeck-Loebenstein
B
. 
Increase of regulatory T cells in the peripheral blood of cancer patients
.
Clin Cancer Res
2003
;
9
:
606
12
.
113.
Connolly
MK
,
Mallen-St Clair
J
,
Bedrosian
AS
,
Malhotra
A
,
Vera
V
,
Ibrahim
J
, et al
Distinct populations of metastases-enabling myeloid cells expand in the liver of mice harboring invasive and preinvasive intra-abdominal tumor
.
J Leuk Biol
2010
;
87
:
713
25
.
114.
Correale
P
,
Rotundo
MS
,
Del Vecchio
MT
,
Remondo
C
,
Migali
C
,
Ginanneschi
C
, et al
Regulatory (FoxP3+) T-cell tumor infiltration is a favorable prognostic factor in advanced colon cancer patients undergoing chemo or chemoimmunotherapy
.
J Immunother
2010
;
33
:
435
41
.
115.
Rech
AJ
,
Vonderheide
RH
. 
Clinical use of anti-CD25 antibody daclizumab to enhance immune responses to tumor antigen vaccination by targeting regulatory T cells
.
Ann N Y Acad Sci
2009
;
1174
:
99
106
.
116.
Rech
AJ
,
Mick
R
,
Martin
S
,
Recio
A
,
Aqui
NA
,
Powell
DJ
 Jr
, et al
CD25 blockade depletes and selectively reprograms regulatory T cells in concert with immunotherapy in cancer patients
.
Sci Transl Med
2012
;
4
:
134ra62
.
117.
Keskinov
AA
,
Shurin
MR
. 
Myeloid regulatory cells in tumor spreading and metastasis
.
Immunobiology
2015
;
220
:
236
42
.
118.
Almand
B
,
Clark
JI
,
Nikitina
E
,
van Beynen
J
,
English
NR
,
Knight
SC
, et al
Increased production of immature myeloid cells in cancer patients: a mechanism of immunosuppression in cancer
.
J Immunol
2001
;
166
:
678
89
.
119.
Gabrilovich
DI
,
Ostrand-Rosenberg
S
,
Bronte
V
. 
Coordinated regulation of myeloid cells by tumours
.
Nat Rev Immunol
2012
;
12
:
253
68
.
120.
Haverkamp
JM
,
Smith
AM
,
Weinlich
R
,
Dillon
CP
,
Qualls
JE
,
Neale
G
, et al
Myeloid-derived suppressor activity is mediated by monocytic lineages maintained by continuous inhibition of extrinsic and intrinsic death pathways
.
Immunity
2014
;
41
:
947
59
.
121.
Khaled
YS
,
Ammori
BJ
,
Elkord
E
. 
Increased levels of granulocytic myeloid-derived suppressor cells in peripheral blood and tumour tissue of pancreatic cancer patients
.
J Immunol Res
2014
;
2014
:
879897
.
122.
Meyer
C
,
Cagnon
L
,
Costa-Nunes
CM
,
Baumgaertner
P
,
Montandon
N
,
Leyvraz
L
, et al
Frequencies of circulating MDSC correlate with clinical outcome of melanoma patients treated with ipilimumab
.
Cancer Immunol Immunother
2014
;
63
:
247
57
.
123.
Kusmartsev
S
,
Nefedova
Y
,
Yoder
D
,
Gabrilovich
DI
. 
Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species
.
J Immunol
2004
;
172
:
989
99
.
124.
Huang
B
,
Pan
PY
,
Li
Q
,
Sato
AI
,
Levy
DE
,
Bromberg
J
, et al
Gr-1+CD115+ immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host
.
Cancer Res
2006
;
66
:
1123
31
.
125.
Diaz-Montero
CM
,
Salem
ML
,
Nishimura
MI
,
Garrett-Mayer
E
,
Cole
DJ
,
Montero
AJ
. 
Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin-cyclophosphamide chemotherapy
.
Cancer Immunol Immunother
2009
;
58
:
49
59
.
126.
Zhao
L
,
Lim
SY
,
Gordon-Weeks
AN
,
Tapmeier
TT
,
Im
JH
,
Cao
Y
, et al
Recruitment of a myeloid cell subset (CD11b/Gr1 mid) via CCL2/CCR2 promotes the development of colorectal cancer liver metastasis
.
Hepatology
2013
;
57
:
829
39
.
127.
Rodriguez
PC
,
Ernstoff
MS
,
Hernandez
C
,
Atkins
M
,
Zabaleta
J
,
Sierra
R
, et al
Arginase I-producing myeloid-derived suppressor cells in renal cell carcinoma are a subpopulation of activated granulocytes
.
Cancer Res
2009
;
69
:
1553
60
.
128.
Ugel
S
,
Delpozzo
F
,
Desantis
G
,
Papalini
F
,
Simonato
F
,
Sonda
N
, et al
Therapeutic targeting of myeloid-derived suppressor cells
.
Curr Opin Pharmacol
2009
;
9
:
470
81
.