Abstract
Gastrointestinal cancers are still responsible for high numbers of cancer-related deaths despite advances in therapy. Tumor-associated cells play a key role in tumor biology, by supporting or halting tumor development through the production of extracellular matrix, growth factors, cytokines, and extracellular vesicles. Here, we review the roles of these tumor-associated cells in the initiation, angiogenesis, immune modulation, and resistance to therapy of gastrointestinal cancers. We also discuss novel diagnostic and therapeutic strategies directed at tumor-associated cells and their potential benefits for the survival of these patients.
1. Introduction
Gastrointestinal (GI) cancers represent the most prevalent tumors worldwide and the major cause of death related to cancer. Within this group, we can identify colon, stomach, and liver cancers as the main concerns according to their prevalence (fourth, sixth, and seventh most prevalent, respectively). As a cause of death, stomach (second higher), liver (third higher), and colon cancer (fifth higher) are the main culprits [1]. Nonetheless, this group includes other mention-worthy cancers. Although not within the ten most prevalent, pancreatic ductal adenocarcinoma (PDAC) has one of the worst prognoses and is expected to be one of the major causes of death related to cancer by 2030 [2]. Additionally, the esophagus cancer is highly prevalent in some areas of the globe [1].
The treatment outcomes are also completely different amongst GI cancers. Two of the main reasons concern the timing of diagnosis and the therapeutic approach. As an example, colon and rectum cancers are usually diagnosed at early stages and are treated with surgery (colon cancer [3]), or multimodality treatment including chemoradiotherapy and surgery (rectum cancer [4]) with high rates of success, especially in the latter. Nonetheless, other tumors such as PDAC are usually diagnosed at later stages, when surgery approaches are usually no longer feasible. In these cases, the traditional treatment is based on combined chemotherapy [3–7], with 5 years below 5% [8].
The development of a prominent desmoplastic reaction by both local and distantly recruited stromal cells has been observed in GI cancers. In addition to immune cells, bone marrow- (BM-) derived progenitor cells are recruited to the tumor microenvironment (TMEN) where they differentiate into various stromal cells, such as endothelial cells, pericytes, and fibroblasts [9]. These cells are crucial for both malignization and cancer progression [10] and are frequently associated with poor prognosis [11–14]. Indeed, the interaction of cancer cells and the host microenvironment plays a critical role in strengthening the metastatic proficiency. Thus, a better understanding of the oncological drivers of these tumors, including their interaction with the microenvironment, is of utmost importance [15, 16]. In this review, we will focus on the role of these tumor-associated cells in the tumorigenesis and progression of GI cancers, as well as on their role in treatment resistance and potential targeted therapeutic approaches.
2. BM-Derived Progenitor Cells
BM-derived cells (BMDCs) are constantly recruited to the TMEN, where they modulate tumor growth and metastasis through the regulation of angiogenesis, inflammation, and immune suppression [17]. Several studies in animal models have implicated BMDCs in the development of carcinomas of the upper GI tract [18, 19], including gastric cancer (GC) [20]. BMDCs were shown to home and repopulate the gastric mucosa in response to H. pylori chronic infection, leading to the development of metaplasia, dysplasia, and cancer over time [20]. In another study, BMDCs were found to be about 25% of H. pylori-induced dysplastic lesions in a mouse model [21]. Zhang et al. showed that highly metastatic colorectal carcinoma (CRC) cells produce elevated serum levels of OPN, MMP9, S100A8, S100A9, SAA3, and VEGFA. This promoted the setup of hepatic TMENs supportive of metastasis by BMDCs recruitment to this organ [22]. Bone marrow-derived CD34+ CD31− immature myeloid cells were also found to cluster at the invasion front of CRC in cis-Apc+/D716 Smad4± mutant mice. These immature myeloid cells expressed Metalloproteinases (MMP) 9 and MMP2 and supported tumor invasion at early stages in intestinal adenocarcinomas [23].
Bone marrow-derived mesenchymal stem cells (MSCs) constitute a nonhematopoietic stem cell subpopulation that can populate the TMEN and contribute to tumor growth and progression through paracrine signaling [24]. Data from Beckermann et al. suggested a supportive role of MSCs in angiogenesis [25]. In this study, MSCs display increased vascular endothelial growth factor (VEGF) mRNA expression and protein secretion. They were also found to migrate towards tumor blood vessels of PDAC, in vitro and in vivo, in response to tumor-secreted growth factors. This is reinforced in vivo in an orthotopic mouse model of PDAC, where siRNA directed towards VEGF induces loss of vessel density control by MSCs [25].
Hypoxia can also induce the expression of growth factors that act as chemoattractants for MSCs to the TMEN [26–29]. In fact, the expression of VEGF by MSCs was shown to increase upon stimulation by interferon- (IFN-) γ and tumor necrosis factor- (TNF-) α cytokines through a hypoxia-induced factor- (HIF-) 1α-dependent pathway. This promoted angiogenesis and tumor growth in mice bearing CRC [30].
Some reports also implicate MSCs in tumor progression and metastasis. For instance, S100 Calcium Binding Protein A4 (S100A4) secreted by MSCs isolated from patient-derived hepatocellular carcinoma (HCC) tissues upregulated the expression of miR155 in HCC cells, promoting tumor invasion through the suppressor of cytokine signaling 1- (SOCS1-) MMP9 axis [31]. Exosomes, extracellular vesicles of endosomal origin, can mediate transfer of biomolecules both locally and at distance, playing a key role in the setup of TMENs [32, 33]. For instance, MSCs-derived exosomes supported GC lymph node metastasis and venous invasion by transferring miR-214, miR-221, and miR-222, regulators of the tumor suppressor gene Phosphatase and Tensin Homolog (PTEN), to cancer cells [34].
Bone marrow-derived MSCs also have the ability to differentiate into several cell types in the stroma [24], including fibroblasts [35]. Spaeth et al. showed the propensity of MSCs to transition to a tumor-associated fibroblast-like phenotype in ovarian, breast, and PDAC-xenografted tumors. These fibroblast-like cells ultimately contributed to microvascularization and the production of tumor-stimulating paracrine factors [36]. MSCs also favor primary CRC cells metastasis to the liver [37]. Orthotopic transplantation of cancer cells mixed with MSCs (but not cancer cells on their own) into the cecal wall resulted in macroscopic liver metastasis. Interestingly, metastasized colon cancer cells recruited more MSCs to the secondary sites where these were found to differentiate into supporting fibroblast‐like cells [37]. Altogether, these results illustrate the role of MSCs in the development of tumor-supporting microenvironments.
Hematopoietic stem cells (HSC) constitute a subpopulation of BM resident cells endowed with long-lived self-renewal and multipotency that sustain the generation of all cells of the blood and immune system. The HSC niche is tightly regulated by osteoclasts and vascular cells within the BM compartment, contributing for the maintenance of a quiescent microenvironment and controlled differentiation [38, 39]. However, tumor-derived soluble factors are able to systemically induce a BM microenvironment switch, from quiescent to protumorigenic and proangiogenic, and stimulate HSC mobilization into the circulation and recruitment to the tumor [39]. These cells can indirectly modulate tumor growth through their ability to differentiate into myofibroblasts and inflammatory cells in the tumor environment.
3. Cancer-Associated Fibroblasts (CAFs)
Fibroblasts are normal components of the connective tissue. These spindle-shaped cells are the main nonepithelial and nonimmune cell elements found in the interstitial space, embedded in physiological extracellular matrix (ECM) [40]. Resident fibroblasts of healthy tissues are considered to be in a resting or quiescent state and are characterized by low metabolic and synthetic activities. In the physiological wound healing process, resting fibroblasts become activated, gaining contractile properties and becoming synthetically dynamic [41, 42]. As the wound closes and evolves into a scar, apoptosis of the activated fibroblasts (myofibroblasts) leads to a significant decrease in their numbers [43]. The inability of myofibroblasts to undergo apoptosis is the driving factor in the development of fibrotic diseases and contributes for the maintenance of other pathological conditions, such as chronic inflammation [42].
In oncologic settings, fibroblasts are frequent components of the TMEN and play an important role at all stages of cancer progression through their phenotypic plasticity and ability to secrete a wide range of signaling molecules. Next, we emphasize how these cells play a key role in generating tumor-promoting microenvironments in GI cancers.
3.1. Role of CAF in Cancer Initiation and Growth
Multiple studies have highlighted the important role of CAFs in the process of cancer initiation and progression. For example, the abundance of myofibroblasts in CRC-associated stroma is predictive of postsurgery disease recurrence [44]. It has also been suggested that the loss of transforming growth factor- (TGF-) β inhibitory effect leads to the activation of hepatocyte growth factor- (HGF-) mediated cell-cycle regulation and stimulation of epithelial proliferation, promoting invasive squamous cell carcinoma of the forestomach in the Tgfbr2fspKO knockout mice [45]. In addition, the conditional knockout of the TGF-β type II receptor gene in mouse fibroblast-specific protein 1- (FSP1-) positive fibroblasts revealed that TGF-β signaling modulates the proliferation and oncogenic potential of epithelial cells [45].
Recently, it has also been demonstrated that CAFs-secreted HGF regulates liver tumor-initiating cell plasticity through the activation of Tyrosine-Protein Kinase Met/Fos-Related Antigen 1/Hairy and Enhancer of Split-Related Protein 1 (c-Met/FRA1/HEY1) signaling. The activation of this signaling pathway was associated with fibrosis-dependent development in HCC in vivo [46]. CAFs-derived HGF was also shown to promote a stemness phenotype in CRC cells [47]. In another study, the deletion of Liver Kinase B1 (Lkb1) gene in stromal fibroblasts resulted in penetrant polyposis in mice, underscoring the involvement of these cells in the tumorigenesis of GI Peutz–Jeghers syndrome. Further analysis revealed that Lkb1 loss induces interleukin- (IL-) 11 expression in gastric fibroblasts and subsequent activation of the Janus Kinase/Signal Transducer and Activator of Transcription 3 (JAK/STAT3) pathway in tumor epithelia, promoting GI tumorigenesis [48].
Emerging data also suggest the switch from normal quiescent fibroblasts into an activated phenotype through epigenetic modifications [49–51]. Helicobacter pylori infection, one of the major causes of GC, was shown to induce the secretion of Prostaglandin E2 (PGE2) by gastric epithelial cells. The stromal PGE2 silenced miR-149 through hypermethylation, removing the suppression of its target genes, IL6 and PGE2 receptor 2. This led to elevated IL6 levels that stimulated the stem-like properties of GC cells [49].
3.2. Role of CAF in EMT, Extracellular Matrix Remodeling, and Metastasis
CAFs-mediated signaling also participates in the acquisition and maintenance of cancer cell stemness. One of the common concepts associated with metastasis initiation is epithelial-to-mesenchymal transition (EMT), that is, the process by which cells lose their epithelial characteristics (such as cell-to-cell adhesion and planar and apical polarity) to acquire mesenchymal features (such as motility and invasiveness) [52]. Paracrine signaling through TGF-β between CAFs and cancer cells leads to EMT-driven gain of stemness and metastasis initiation [53, 54]. In fact, targeting CAFs with curcumin reverted the EMT phenotypes of PDAC cells blocking their migration and metastasis [55]. In HCC, myofibroblasts can induce EMT in a TGF-β/platelet-derived growth factor- (PDGF-) dependent manner [56]. Likewise, IL-6 produced by fibroblasts can also activate JAK2/STAT3 signaling in the GC cells promoting their migration and EMT [57]. The miRNA 320a can also affect EMT by decreasing PBX Homebox 3 (PBX3), Extracellular Signal-Regulated Kinase 1 and 2 (ERK1/2) signaling, and N-cadherin expression, and simultaneously increase E-cadherin. When transferred from CAFs to HCC tumor cells via exosomes, this miRNA can inhibit tumor proliferation, migration, invasion, and metastasis. Interestingly, CAF-derived exosomes from HCC patients contain reduced levels of miR-320a, showing how the reduction of an antitumor factor in these vesicles can affect metastasis [58]. Cancer cell-derived exosomes can also reprogram normal adjacent fibroblasts into CAFs. For example, a recent study showed that exosomes derived from early- or late-stage CRC cell lines induce the activation of quiescent fibroblasts into distinct functional subtypes [59]. Specifically, the activation mediated by late-stage cancer exosomes resulted in a proinvasive profile, while fibroblasts activated by early-stage cancer exosomes presented a pro‐proliferative and proangiogenic phenotype [59].
Fibroblasts can also remodel the microenvironment and lay the tracks for cancer cell invasion through the surrounding ECM and stromal cell layers [60–62]. For instance, CAF-derived transgelin (TAGLN) induces MMP2 expression and promotes migration and invasion of GC cells [63], while CAF-derived fibroblast activation protein (FAP) remodels the ECM and promotes PDAC cell invasion [64]. In HCC, CAFs secrete different cytokines that activate the hedgehog (C-C motif chemokine 2 (CCL2) and 5 (CCL5) and TGF-β (CCL7 and C-X-C motif ligand (CXCL) 16) pathways in HCC cells, inducing their migration and invasion in vitro and metastasis in vivo [65]. In addition, Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) secreted by cancer-associated MSCs, a subpopulation of CAFs isolated from human PDAC, induced proliferation, invasion, and transendothelial migration of PDAC cells [66].
Colonization is probably the most limiting of all metastasis steps and the microenvironment at the distant sites needs to be favorable for this to happen. Paget's seed and soil theory back in 1889 was the first to suggest that metastasis to a certain organ is not random but depends on interactions with its microenvironment and that cancer cells will seed only in fertile soils [67]. The concept of “pre-metastatic niche” was introduced later in 2001 by Kaplan et al., where they showed that BM-derived cells can form clusters that home to tumor-specific premetastatic sites [67, 68]. In line with this theory, metastatic cancer cells are capable of bringing their own “soil” to the metastatic site in order to facilitate their colonization [69]. A good example is the case of IL11 production by TGF-β-stimulated CAFs, which activate Glycoprotein 130 (GP130)/STAT3 signaling in CRC cells conferring them the survival advantage for efficient organ colonization [70].
3.3. Role of CAF in Angiogenesis
CAFs tailor tumor growth and progression not only by influencing tumor cells but also by indirectly affecting other stromal cells and regulating angiogenesis, inflammation and immune modulation [10]. CAFs are capable of promoting angiogenesis by secreting VEGF and Stromal Cell-Derived Factor 1 (SDF-1) [71, 72]. For example, in a mouse model of GC, α-Smooth Muscle Actin- (α-SMA-) positive fibroblasts were the main producers of VEGF. Activation of these fibroblasts was stimulated by GC cells and shown to increase tube formation by endothelial cells in vitro [73]. CAFs are also a source of IL6 in CRC, which in turn can increase VEGF secretion by adjacent fibroblasts and induce tumor angiogenesis in xenografted cancer cells [74]. Pancreatic fibroblasts, also known as pancreatic stellate cells (PSCs), express several proangiogenic regulators such as VEGF receptors, angiopoietin-1, and Tie-2 and produce VEGF in response to hypoxia. Conditioned medium from hypoxia-induced PSCs was able to increase proliferation, migration, and angiogenesis of Human Umbilical Vein Endothelial Cells (HUVECs) both in vitro and in vivo [75]. In a similar line, hepatic stellate cells (one of the main players in HCC) can produce VEGF and angiopoietin-1 in hypoxic conditions and induce angiogenesis [76, 77].
4. Endothelial and Perivascular Cells
4.1. Role of Endothelial and Perivascular Cells in Tumor Growth
The angiogenic switch is a hallmark of cancer that allows for tumor growth by providing nutrients and oxygen and removing cellular wastes [10]. The establishment of new blood vessels is a crucial step for tumor progression, but the endothelial and perivascular cells that constitute these blood vessels are not just mere bystanders in the game. Endothelial cells (ECs) can promote a cancer stem cell phenotype in human CRC in vitro and in cocultured CRC cells ex vivo [78]. Lu et al. showed that CRC stemness is induced through paracrine activation of Notch signaling, whereby membrane-bound Jagged-1 on ECs is cleaved by ADAM metalloproteinase domain 17 (ADAM17), releasing a truncated soluble fragment that binds Notch on CRC cells. Importantly, both primary and chemotherapy-naïve liver metastatic CRC liver showed CD133+ epithelial cells located in the proximity to perivascular regions, further supporting an ECs-mediated role in the CRC stem cell phenotype in clinical specimens [78]. A subsequent study found that ECs contributed to chemoresistance in CRC cells via serine/threonine-protein kinase- (AKT-)mediated induction of Nanog Homeobox Retrogene P8 (NANOGP8) [79]. Likewise, in an in vitro model of Hepatitis B Virus- (HBV-) induced HCC, increased levels of TGF-β in the conditioned medium of HUVECs boosted the expression of mesenchymal markers, including CD133, and promoted an aggressive phenotype in stimulated Hepatitis B-X Protein- (HBx-) infected hepatoma cells [80]. Immunization of mice with glutaraldehyde-fixed HUVECs resulted in reduced expression of angiogenesis-related antigens (VEGF-2 and vascular endothelial- (VE-) cadherin), suppression of angiogenesis, and smaller esophageal squamous carcinoma (ESC) tumors [81]. This has prompted the evaluation of HUVEC vaccines promoting tumor autoimmune response targeting angiogenesis in pilot trials involving patients with metastatic CRC [82].
Lymphatic endothelial cells (LECs) are also important players in the growth of GI cancers. Expression of Growth Differentiation Factor 11 (GDF11) was increased in CRC patients and positively correlated with tumor grade. GDF11 released in LEC-derived exosomes was also identified as a key modulator of CRC growth in vitro and in vivo [83]. Moreover, increased proliferation and invasive ability of ESC cells in vitro has been demonstrated upon stimulation with conditioned medium from ESC-related lymphatic microvessel endothelial cells. This interaction leads to the upregulation of MMP9 expression and downregulation of Tissue Inhibitor of Metalloproteinases 2 (TIMP-2) expression in poorly differentiated ESC cells and promotes both lymphangiogenesis and growth of these cells in vivo [84]. Lymphatic endothelial cells also have an immunoregulatory function in GC by inhibiting the production of IL2, IL10, and IFN-γ cytokines in CD4+ T cells. Coculturing GC cells with both LECs and CD4+ T cells resulted in the upregulation of Programmed Death-Ligand 1 (PD-L1) and Indoleamine 2,3-Dioxygenase (IDO) mRNA expression. This suggests a possible mechanism of cancer immune tolerance and metastatic spread through the lymphatic vessels in GC [85].
Additionally, the development of diffuse-type GC depends on the inflammation mediated by CXCL12+ ECs and C-X-C motif receptor (CXCR) 4+ gastric innate lymphoid cells (ILCs) that form the perivascular gastric stem cell niche. Endothelial CXCL12 plays a central role recruiting Wnt Family Member 5A- (Wnt5a-) producing CXCR4+ ILCs to the stomach, which in turn activates Ras Homolog Family Member A (RhoA), inhibits anoikis in the E-cadherin-depleted cells and promotes diffuse-type GC growth [86].
4.2. Endothelial and Perivascular Cells Role in Tumor Metastasis
Blood vessels are also a gateway for distant metastasis. The lack of vascular maturation in newborn vessels facilitates cancer cell penetration and promotes distant metastasis. Immunohistochemical analysis of tissue samples from CRC patients revealed a significant correlation between lower microvessel pericyte coverage with increased hematogenous metastasis and poorer survival [87].
The importance of these vascular networks is underscored by the fact that cancer cells undergoing EMT are able to assume the identity and role of pericytes to stabilize the tumor vasculature and improve the vascular support for tumor growth [88]. Shenoy et al. found that the majority of cancer cells undergoing EMT were located preferentially in the perivascular space and were closely associated with ECs and in line with the blood vessels in tumor xenografts. It was further demonstrated that these cells expressed pericyte markers and interacted with ECs, stretching alongside Human Microvascular Endothelial Cells (HMVECs) and exhibiting tight adhesion to EC tubes in a coculture assay in vitro [88]. This phenomenon, known as vasculogenic mimicry (VM), whereby cancer cells form de novo vascular networks without the involvement of ECs, represents an alternative paradigm of tumor perfusion in HCC and GC [89–91].
Exosomes are also key mediators in this setting, as they mediate vascular permeability and angiogenesis. Zeng et al. found that CRC-derived exosomal miR-25-3p can be transferred to ECs, where it targets and silences Krüppel-Like Factor 2 (KLF2) and 4 (KLF4). In vivo, exosomal miR-25-3p elicited vascular leakiness and promoted CRC metastasis [92]. Exosome-mediated remodeling of the lymphatic network in sentinel lymph nodes was also shown to promote CRC metastasis. The uptake of CT26-derived exosomes by macrophages induced the release of VEGF-C, mediated by exosomal Interferon Regulatory Factor 2 (IRF-2), and promoted lymphangiogenesis in sentinel lymph nodes, which facilitated the development of CRC metastasis [93]. Upregulated secretion of CXCL1 by LECs can also promote migration, invasion, and adhesion of GC cells through the activation of integrin β1- (Focal Adhesion Kinase) FAK-AKT signaling. Activation of the latter induced the expression of MMP2 and MMP9 and increased lymph node metastasis in an animal model of GC [94].
When cancer cells enter the systemic circulation, they get exposed to the harsh circulating conditions. Together with the absence of cell/ECM interaction, apoptosis signaling is triggered and cancer cells undergo rapid anoikis [95]. A recent study in head and neck carcinoma showed that cancer cells bound to Bcl-2 overexpressing ECs (EC-Bcl-2) via E-selectin presented significantly higher anoikis resistance. Additionally, mice coinjected with squamous cell carcinoma cells and EC-Bcl-2 displayed significantly higher lung metastasis [96]. The described chaperoning role of ECs could, in principle, also occur in GI cancers, as increased levels of circulating ECs have also been observed in patients with colon, gastric, and esophagus cancers [97].
5. Tumor-Infiltrated Immune Cells
Infiltrating immune cells from lymphocytic and myeloid origin are also constituents of the TMEN [10]. Lymphocytes are composed by three main lineages that originate from a common precursor identified in the BM: natural killer (NK) cells, T cells, and B cells. T cells are subject to a final lineage decision in the thymus to form mature CD4 (helper) and CD8 (cytotoxic) T cells [98]. NK and CD8 T cells can rapidly degranulate and secrete IFN-γ following antigen receptor triggering, which is particularly important in antitumor responses [99]. T regulatory Cells (TRegs) can contribute to homeostasis by inducing immunotolerance and control of autoimmunity. These cells are derived from the thymus, express CD4, CD25, and Forkhead Box P3 (FOXP3, murine Foxp3) and are capable of inhibiting immune responses mediated by CD4+CD25− and CD8+ T cells [100–102].
Myeloid cells comprise polymorphonuclear cells or granulocytes, dendritic cells (DCs), monocytes, and macrophages (extravasated blood monocytes) [103, 104]. Macrophages can display a broad spectrum of activation and polarization states [105, 106]. However, in more general and simplistic models, they are frequently classified as M1, that takes part in type I T helper (Th1) cell responses when stimulated by IFN-γ and is characterized by release of radical oxygen species (ROS), nitric oxide (NO), and proinflammatory cytokines such as TNF-α and IL12; M2, involved in type II T helper (Th2) cell responses and identified by expression of arginase and release of anti-inflammatory cytokines such as IL10 when stimulated by IL4 and IL13 [107].
Myeloid-derived suppressor cells (MDSC) comprise another population of myeloid progenitor and immature myeloid cells endowed with the ability to inhibit T-cell responses [108–110]. DCs were also reported as activators of specific T cells during inflammatory responses and play a central role in protection against infection and malignancy [111, 112]. DCs can also display multiple profiles. Monocyte-derived DCs can perform marked tumor antigen uptake. cDC1s can activate CD8+ T cells, while cDC2s can reprogram protumoral macrophages when injected in mouse models. Importantly, the analysis of tumor biopsies from colorectal cancer patients revealed the presence of all the three abovementioned subsets of tumor-associated DCs [113]. For these reasons, myeloid cells play a pivotal role in the regulation of immune cell responses.
Tumor inflammation has a paradoxical role in promoting tumor growth and progression [10]. Some reports show the association between unresolved infection, chronic inflammation, and tumor initiation. Examples are the relationships between infection by Helicobater pylori and GC, chronic pancreatitis and PDAC, and ulcerative colitis and CRC [114–117]. In this section, we describe how immune cells contribute to TMENs in GI cancers.
5.1. Immune Surveillance Evasion
Although increased infiltration of tumors by immune cells has long been thought to be a consequence of failed attempts to eliminate cancer cells, recent studies show that cancer cells can frequently evade immune responses [10]. An important mechanism of immune evasion involves PD-L1/Programmed Death-1 (PD-1) that has been linked to T-cell apoptosis [118]. For example, HCC-derived IL-10 can increase expression of PD-L1 by Kupffer cells, which in turn can decrease the antitumor function and proliferation of CD8+PD-1+ cells [119]. In the same line, PD-L1+ monocytes infiltrates can suppress antitumor T-cell responses and contribute to tumor growth in vivo [120]. In both cases, PD-L1 correlated with poor survival in HCC patients and could be targeted by anti-PD-L1 antibodies [119, 120]. CXCL12 produced by FAP+CAFs has been linked to immune evasion in PDAC, and targeting this cytokine can synergize with anti-PD-L1 immunotherapy in PDAC [121, 122].
In two complementary studies, Wang et al. and Kortylewski et al. demonstrated how the constitutive activation of Stat-3 in cancer and hematopoietic-derived tumor infiltrating cells could inhibit the maturation of DCs, leading to a defective antitumor immune response. In fact, Stat-3 inhibition enhanced the antitumor function of T and NK cells, DCs, and innate immunity against tumors [123, 124]. Furthermore, in a small cohort of patients and healthy control subjects, Yanagimoto et al. have shown that the numbers and function of DCs were reduced in PDAC patients [125].
PDAC and CRC cells can also evade immune response by expressing an apoptosis-mediating surface antigen FAS (Fas) receptor, which enables these cells to resist Fas-mediated apoptosis and, at the same time, to increase the expression of Fas ligand (FasL), which mediated the killing of T cells in coculture assays [126–128]. The upregulation of FasL was also demonstrated in GC together with downregulation of caspase-3 as an immune escape mechanism [129, 130]. Another strategy by which PDAC cells could escape immune response was through the upregulation of IDO [131], a tryptophan-degrading enzyme that induces an anergic state in T cells through tryptophan starvation [132]. In addition, the presence of Th2 lymphocytes also correlates with reduced PDAC patient survival. Thymic Stromal Lymphopoietin (TSLP), which induces Th2 polarization, was found to be secreted by CAFs after stimulation by TNF-α and IL1β [133].
Immunosuppressive cells such as MDSC and TRegs are elevated in patients with PDAC, esophageal, and GC when compared with controls, being considered an independent prognostic factor for survival in all these three cancers [134]. In fact, by studying a small cohort of patients, Porembka et al. have demonstrated that human PDAC show an increased infiltrate of MDSC when compared with normal pancreatic tissue. Moreover, depletion of these cells in an animal model of PDAC resulted in delayed tumor growth [135]. MDSCs were also increased in HCC patients and could induce the formation of TReg populations, suggesting this as one of the mechanisms responsible for immunosuppression in HCC [136]. Increased populations of TRegs were also found in the blood of patients with gastric and esophageal cancers [137]. In addition, TGF-β1 produced by GC cells was shown to induce TReg development from CD4+CD25−T cells, and high levels of this factor correlated with elevated TReg numbers in GC patients [138]. Interestingly, Mizukami et al. suggested that the localization pattern rather than the numbers of TRegs might have a higher impact in survival of GC patients. They found that a diffuse pattern of TRegs accounts for poorer survival than peritumoral localization of these cells [139]. TRegs of HCC patients were also capable of impairing the function of CD8+ T cells by decreasing their proliferation, activation, degranulation, and production of enzymes such as granzymes A and B and perforin when stimulated. Not surprisingly, TRegs were associated with higher mortality in these patients [140]. Both CRC- and HCC-associated fibroblasts were also shown to impair NK-cell antitumor cytotoxicity by releasing molecules such as PGE2 [141, 142].
5.2. Immune Cells Role in EMT, Invasion. and Metastasis
In addition to immune surveillance evasion, infiltrated immune cells also promote invasive phenotypes in cancer cells through EMT. For example, PDAC cells increase the conversion of blood monocytes into monocytic MDSC, which in turn act to promote EMT features in these cancer cells [143]. Using in vitro coculture studies, Liu et al. have shown that the promotion of the M2 phenotype in macrophages induces the expression of mesenchymal markers in PDAC cell lines in vitro and that this effect was dependent on Toll-Like Receptor 4 (TLR4) and IL10 levels [144]. M2 macrophages were also capable of inducing GC invasiveness via activation of the β-catenin pathway [145].
Kim et al. suggested that myofibroblasts can induce the differentiation of myeloid cells into S100A8/9-expressing MDSC and M2 macrophages in CRC by secreting IL-6 and IL-8 [146]. In a mouse model of esophageal cancer, recruitment of MDSC was correlated with IL-6 levels and tumor invasiveness, IL-6 being shown to induce the MDSCs. In fact, IL-6 and MDSC levels predicted prognosis in patients with esophageal cancer [147]. In addition, CAFs and M2 macrophage markers predict clinical outcome in CRC, their expression being inversely correlated with survival [148]. Similarly, CAFs isolated from PDAC patients promoted M2 macrophage polarization that in turn promoted the proliferation of PDAC cells in vitro and tumor growth and invasion in vivo [149]. Polarized CD163+ (M2) macrophages were also correlated with increased angiogenesis, CXCL12 expression, and tumor progression in GC [150].
The recruitment of immune cells to secondary metastatic sites, and their role in promoting a receptive soil for metastatic growth, has also been the focus of some recent studies. For example, PDAC-derived exosomes containing macrophage migration inhibitory factor- (MIF-) induced TGF-β1 production by Kupffer cells, which induced α-SMA and fibronectin expression by hepatic stellate cells. This supported the influx of BM-derived monocytes, which constituted a liver TMEN supportive of PDAC metastasis [151]. In fact, PDAC liver metastasis depends on the early recruitment of granulin-secreting inflammatory monocytes to this organ. Granulin secretion by metastasis-associated macrophages activates resident hepatic stellate cells into myofibroblasts, which in turn secrete periostin, resulting in a fibrotic microenvironment that sustains metastatic tumor growth [152]. Seubert et al. also demonstrated an increased liver susceptibility towards metastasis through SDF-1-mediated recruitment of neutrophils to the liver. In this study, systemic TIMP-1, which was previously thought to suppress tumor metastasis, was instead driving the increased levels of hepatic neutrophil chemoattractant SDF-1 [153].
6. Stromal Signatures as Prognostic Tools
Based on the significance of stromal cells in tumor growth and progression, much effort has been done on the identification of stromal signatures of cancer prognosis. For example, tumor lymphocyte infiltrates (TIL), such as CD4+/CD8+ T and NK+ cells, have been generally associated with a good prognosis. On the other hand, infiltration by TRegs, MDSC, M2 macrophages, and CAFs has been seen as a sign of disease progression and poor prognosis, as listed in Table 1.
Table 1.
Stromal cell | Type of cancer | Levels | Clinical outcome | References |
---|---|---|---|---|
CAFs | GC | High | Metastasis | [154, 155] |
PD-L1 | PDAC | Expression | Poor prognosis | [156] |
PD-L1 | GC | Expression | Poor prognosis | [157–159] |
PD-L1 and CD8+ T cells | GC/gastroesophageal | High | Lower survival | [160] |
PD-L1 and PDL2 | Esophageal | Expression | Poor prognosis | [122, 161] |
M2 macrophages | PDAC | High | Lymphatic metastasis/poor prognosis | [162] |
CD204+ (M2) macrophages | Esophageal | High | Poor DFS | [163] |
M2 macrophage and %TRegs | PDAC | High | Lower survival | [155] |
CAFs and M2 macrophages | CRC | Expression | Reduced survival | [148] |
MDSC | PDAC | High | Progression | [164] |
MDSC | GC/PDAC/esophageal | Low | Survival | [134] |
Th2 | PDAC | Presence | Reduced survival | [149] |
TRegs | PDAC | High | Progression/poor prognosis | [165–168] |
TCD3low/TReghigh | CRC | Low/high | Lower survival | [169] |
TRegs | CRC | High | Good prognosis | [170, 171, 172] |
TRegs | HCC | High | Progression | [140, 173] |
TRegslow/CD8+ TILhigh | HCC | Low/high | High DFS | [140, 173] |
TRegshigh/CD8+ TILlow | GC | Ratio high | Lower survival | [174] |
TRegs | GC and esophageal | High | Poor survival | [175] |
DC and circulating myeloid DC | PDAC | High | Survival | [176] |
CD4+/CD8+ TILs | PDAC | High | Good prognosis after surgery | [177] |
CD4+/CD8+ TILs | Esophageal | High | Good prognosis | [178] |
CD8+/CD45RO+ TILs | CRC | High | Good prognosis | [178, 179] |
CD4+/CD8+/CD45RO+ TILs | GC | Low | Lymph node metastasis/lower survival | [180] |
CD8+ and FoxP3+ TILs | GC (microsatellite unstable) | High | Good prognosis | [181] |
M2 macrophages + CD8+ and FoxP3+ TILs | GC (microsatellite unstable) | High | Survival | [182] |
M2 macrophages | GC | High | Poor survival and tumor progression | [145, 150] |
NK+ cells | GC | High | Good prognosis | [183] |
DFS: disease-free survival.
However, conflicting evidence has shown that increased infiltration of gastric and gastroesophageal tumors by CD8+ T cells was actually associated with a worse prognosis. In fact, patients with high CD8 infiltration also presented PD-L1 expression, which was linked to immune resistance [160]. On the other hand, microsatellite unstable GC patients with high CD163+ (M2) macrophages, FOXP3+, and CD8+ TILs where those with the highest survival advantage [182]. In CRC, as opposite to other cancer types, FOXP3+ TRegs were associated with good prognosis [170, 171]. These are examples on how immune cell signatures are context-dependent and how the complexity of cell interactions and soluble factors released in the TMEN can tip the balance in opposite directions.
Given the diversity of switch mechanisms driving CAFs activation, one can expect to have CAFs with different activated phenotypes in the tumor stroma. Another question is whether all CAFs are in an activated state. Fibroblast plasticity and intratumoral heterogeneity results in an array of CAF signatures associated with different tumor types [184]. Several proteins have been used as markers for the identification of CAFs. Some of the most commonly used biomarkers include PDGFRα/β, α-SMA, and FAP. In addition, FSP1 has been suggested as a marker of fibroblasts in a quiescent state [185, 186]. Other proteins, such as vimentin, desmin, discoidin domain receptor 2 (DDR2), and podoplanin, have also been used in the identification of CAFs [185]. However, it is important to highlight that these proteins are also expressed by other cell types, and the lack of consistent and specific fibroblast molecular markers has been an important limiting factor so far [185]. Opposing actions of CAFs expressing the same protein marker can also be observed in a context-dependent way in TMENs. For example, while in CRC-associated CAFs, podoplanin was correlated with less aggressive tumors and a favorable prognosis [187, 188], its expression by CAFs in lung, breast, esophageal, and PDAC has been associated with an unfavorable prognosis [189–192]. In addition, PDAC patients with fewer myofibroblasts in the tumors had reduced survival, possibly by suppression of the immune surveillance due to increased levels of TRegs [193]. Therefore, one should be cautious when identifying CAFs and extrapolating their role in different tumors based on the analysis of the aforementioned biomarkers. It is increasingly evident that CAFs of tumors from different etiologies present different molecular biomarkers or combination of biomarkers [194].
The full spectrum of this phenotypic diversity and their functional implications in tumor growth, progression, or even therapy resistance mechanisms are yet to be fully understood. However, defining specific tumor-associated immune and CAF signatures might become a valuable prognostic tool and drive the advancement of new therapeutic strategies.
7. Impact of Stroma in Resistance to Therapies
During cancer progression, tumors become more heterogeneous due to a generation of genetically distinct tumor-cell subpopulations and to modifications in TMEN components. In this section, we will describe how tumor heterogeneity, combined with the high plasticity of tumor-associated cells, can influence resistance to therapies in GI cancers.
7.1. Desmoplasia and Tumor Resistance
A desmoplastic reaction, characterized by the formation of a dense fibrosis and increased remodeling and deposition of ECM components, is closely associated to a poor outcome in PDAC and CRC patients [195, 196]. One of the main components of the ECM is hyaluronic acid (HA), which is a high molecular mass polysaccharide. PDAC can express HA into stroma and in peritumoral connective tissue and thus impair vascularity and the delivery of chemotherapeutic drugs into tumors. In fact, gemcitabine-resistant PDAC from patients with resectable tumors showed upregulation in gene pathways related to stroma-ECM receptor interaction, focal adhesion, cell communications, gap junction, and cell adhesion molecules [197]. Enzymatic degradation of HA results in reduction of interstitial flow pressure, reexpands the microvasculature in PDAC [198], and increases the delivery of doxorubicin and gemcitabine in a mouse model of PDAC [199].
The Sonic hedgehog (Shh) pathway also promotes desmoplasia in PDAC, and its inhibition improves delivery of chemotherapy [200]. However, genetic inhibition of the Shh pathway results in more aggressive tumors in a PDAC model [201] and accelerates progression of KRAS-driven PDAC. Inhibiting VEGF receptor (VEGFR) in Shh-deficient mice increased survival and impaired tumor progression [202], suggesting that combinatory approaches could be more effective to overcome tumor resistance.
CAF heterogeneity might be responsible, at least in part, for the protumorigenic and antitumorigenic effects in cancer resistance. For example, PDAC presents a subpopulation with high expression of α-SMA adjacent to neoplastic cells, and another with low expression of α-SMA that locates distantly and secretes inflammatory mediators as IL-6 [203]. Intriguingly, depletion of CAFs based on their α-SMA expression can induce immunodepression and accelerate pancreas cancer progression [193], leading to resistance to chemotherapies. These pieces of evidence indicate that new therapeutic approaches should consider these different subpopulations when looking for effective antitumor therapies directed to CAF.
Chemotherapy can also affect stromal cells, which in turn can promote cancer resistance. A hypoxic TMEN can lead to a metabolic shift based on aerobic glycolysis and lactate production by tumor cells, leading to a low extracellular pH, which is a common feature found in solid tumors. Moreover, chemotherapy-treated CAFs change the expression of metabolic enzymes, leading to increased aerobic glycolysis and autophagy and increased energy production [204]. A recent study showed that drugs targeting mutated K-Ras force cancer cells to get energy thought autophagy in PDAC [205].
Low expression of caveolin-1 in stroma is a marker of autophagy, which occurs via oxidative stress followed by an increase in HIF-1α and NF-kappa B expression [206]. High level of HIF-1α in CAFs is related to an elevated lactate efflux and lower extracellular pH [207]. This acid microenvironment drives EMT, protecting PDAC cells from gemcitabine-induced cell death in a mechanism that involves expression of drug transporters [208]. Moreover, gemcitabine upregulates CXCR4 expression in PDAC cells and promotes their invasiveness through a reactive oxygen species-dependent mechanism [209].
7.2. Soluble Factors and Exosomes Roles in Tumor Resistance
Stromal cells produce soluble factors that play a key role in chemoresistance (Table 2). For example, expression of TGF-β1 by CAFs is frequently present in patients treated with chemoradiotherapy, its inhibition being linked to enhanced chemosensitivity of ESC cells [214]. CAFs can also release IL6, which activates the JAK-1/STAT3 signaling pathway and contributes to chemoresistance of GC cells to 5-fluorouracil (5-FU) [217]. IL-6 secreted by CAFs also plays a role in chemoresistance of ESC cells by upregulating CXCR7. In fact, ESC patients with high expression of CXCR7 and IL-6 presented worse overall survival upon receiving cisplatin after surgery [214].
Table 2.
Drugs | Tumor type | Stromal-derived mediator |
---|---|---|
Doxorubicin and gemcitabine | PDAC | Hyaluronan [199] |
Gemcitabine | PDAC | Sonic hedgehog [200] |
Gemcitabine | PDAC | Alpha-SMA [193] |
Gemcitabine | PDAC | HIF-alpha [208] |
Gemcitabine | PDAC | CXCR4 [209] |
Gemcitabine | PDAC | Snail [210] |
Oxaliplatin | CRC | LncRNA19 [211] |
5-FU | CRC | miR145, miR34-a [212] |
Bevacizumab | CRC | VEGF [213] |
Cisplatin | ESC | IL6R [214] |
5-FU and oxaliplatin | GC | AKT, p38, and survivin [215] |
Cisplatin | GC | miR-21 [216] |
Tumor-associated macrophages (TAM) release IL6, which activates the IL-6 receptor (IL6R)/STAT3 pathway in CRC cells. STAT3 inhibits the tumor suppressor miR-204-5p, leading to chemoresistance to 5-FU and to oxaliplatin [218]. This suggests that IL6 receptor inhibition in combination with chemotherapy could serve as a suitable strategy to improve chemotherapeutic efficacy through inhibition of the communication between stromal and GC cells [217]. Another example involves cisplatin resistance in GC cells by TAM-derived exosomes containing miR-21 [216]. Exosomal transfer of miR-21 led to downregulation of PTEN and activation of AKT, which resulted in less apoptosis and increased survival in GC cells treated with cisplatin [216].
Moreover, crosstalk between TAM and tumor infiltrating cells through STAT3 can improve chemotherapeutic efficacy by repressing antitumoral CD8+ T-lymphocyte activity [219].
Treatment failure can also result, at least in part, from the increase in exosome release by stromal cells. For instance, gemcitabine treatment increases fibroblast-derived exosomes containing Snail and miR146a, a Snail target, which induce resistance to chemotherapies in PDAC [210] and promote metastasis and chemotherapy resistance by enhancing cell stemness and EMT in CRC cells [220]. Upon exposure to oxaliplatin, CAFs may release exosomes containing long noncoding RNA (lncRNA) H19 to cancer cells, which has competing endogenous RNA potential for miR-141, a tumor suppressor miRNA that targets β-catenin and suppresses the Wnt/β-catenin pathway. In this way, lncRNA H19 promotes stemness of cancer stem cells and oxaliplatin resistance of CRC [211]. Similarly, exosomes secreted by gemcitabine-treated CAFs promote proliferation and gemcitabine resistance of PDAC cells by increasing Snail expression [210]. Pancreas-derived mesenchymal stromal cells treated with paclitaxel release exosomes containing paclitaxel which inhibit proliferation of PDAC [221]. Moreover, the intracellular and extracellular expression levels of miR-145 and -34a in CRC cells were associated with 5-FU resistance [212]. The resistance was in part due to the enhanced secretion of these antioncomirs in exosomes produced by resistant CRC cells after 5-FU exposure. This led to decreased intracellular levels of the antioncomirs and sustaining proliferation [212]. 5-FU and oxaliplatin treatment can also induce CAFs to release soluble factors that are taken up by CRC cells, promoting drug resistance through AKT, P38, and survivin translocation [215]. In addition, Snail-expressing fibroblasts can secrete CCL1 and contribute to 5-FU and paclitaxel chemoresistance in CRC [222]. Similarly, increased Snail expression in PDAC cells is correlated with gemcitabine resistance [223].
8. Therapies Targeting Stromal Microenvironment
8.1. Extracellular Matrix Components
In 2003, the first clinical trial of a humanized monoclonal antibody directed to human FAP, sibrotuzumab, was found clinically safe in patients with advanced solid cancers [224]. However, it showed limited clinical response in a phase II trial in patients with CRC [225]. Inspite of promising preclinical findings, therapy strategies targeting CAFs have repeatedly faced obstacles. As we pointed out above, depletion of α-SMA-expressing CAFs can accelerate pancreas cancer progression [193]. Thus, depleting CAFs based on their expression of FAP or α-SMA might not be effective, since other stromal cell types can also express these markers.
Regarding ECM remodeling, Lysyl Oxidase-like 2 (LOXL2) is upregulated in tumor-associated stroma of PDAC, ESC, and HCC [226, 227]. Simtuzumab, an antibody that inhibits LOXL2, blocks the desmoplastic reaction in CRCs in vitro [226]. However, phase II clinical trials of simtuzumab in combination with gemcitabine or FOLFIRI (folinic acid, 5-FU, irinotecan) did not improve the clinical outcome in PDAC or in CRC patients, respectively [228, 229].
Different approaches that inhibit desmoplasia in solid cancers can inhibit tumor growth and improve vascular perfusion and drug delivery. Losartan (an angiotensin I inhibitor) is an antihypertensive drug that reduces collagen and hyaluronan production by CAF through downregulation of the fibrotic signals TGF-β1, Cellular Communication Network Factor 2 (CCN2), and Protein Effector of Transcription 1 (ET-1) [230]. In fact, epidemiological studies demonstrated that gastroesophageal cancer patients presented a moderately reduced cancer-specific mortality amongst users of angiotensin receptor blockers [231]. Based on preclinical studies, a phase II study targeting TGF-β1 by using losartan in combination to FOLFIRINOX (folinic acid, 5-FU, irinotecan, oxaliplatin) in locally advanced PDAC is ongoing with an estimative to be concluded by 2025 (Table 3). Another approach is to inhibit Shh signaling, which drives stromal desmoplasia, by activating the ligand for smoothened (SMO) in CAFs [237]. The SMO inhibitor (IPI-926) reduced the abundance of myofibroblasts in the stroma in PDAC and increased tumor vasculature as well as intratumoral gemcitabine uptake. However, a phase Ib/II clinical trial using IPI-926 in combination with gemcitabine in metastatic PDAC did not show benefits in clinical outcome. Indeed, some patients receiving IPI-926 had a shorter median survival time compared with the placebo group [200].
Table 3.
Drug and association | Tumor type | Molecular and cellular target | Mechanism | Study phase | ClinicalTrial.gov identifier |
---|---|---|---|---|---|
Sibrotuzumab | CRC, PDAC | FAP (CAF) | Desmoplasia [224] | II | NCT02198274 |
Simtuzumab + FOLFIRI | CRC, PDAC | LOXL2 | Desmoplasia [229] | II | NCT01479465 |
Simtuzumab + gemcitabine | CRC, PDAC | LOXL2 | Desmoplasia [229] | II | NCT01472198 |
Losartan + F-NOX | PDAC | TGF-beta1 | Fibrosis [230] | II | NCT03563248 ∗ |
IPI-926 + gemcitabine | PDAC | SMO | Hedgehog pathway inhibition | II | NCT01130142 |
PEGPH20 + gemcitabine + nab-paclitaxel | PDAC | Hyaluronan | Desmoplasia [232, 233] | III | NCT02715804 ∗ |
PEGPH20 + FOLFIRINOX | PDAC | Hyaluronan | Desmoplasia [232, 233] | I | NCT01959139 ∗ |
Pembrolizumab + AMG820 | CRC, PDAC | PD-1 (T cells) CSF1R (macrophage) | T-cell apoptosis | II | NCT02713529 ∗ |
Durvalumab + monalizumab | CRC | PD-1 (T cells) CD94/NGK2a | T-cell apoptosis | I | NCT02671435 ∗ |
AMG820 | CRC, PDAC | CSF1R (macrophage) | M2 polarization [234] | I | NCT01444404 |
5F9 + cetuximab | CRC | CD47 (macrophage) | Restores macrophage phagocytosis | II | NCT02953782 ∗ |
Pembrolizumab + cisplatin + 5-fluorouracil | GC, GEJ | PD-1 (T cells) | T-cell apoptosis | III | NCT02494583 ∗ |
Pembrolizumab + paclitaxel | GC, GEJ | PD-1 (T cells) | T-cell apoptosis | III | NCT02370498 ∗ |
Ruxolitinib + capecitabine | PDAC | Janus 1 and Janus 2 (pancreatic stellate cells) | JAK-STAT3 pathway inhibition [235] | III | NCT02117479 # NCT02119663 # |
Nivolumab + ipilimumab | Upper GI | PD-1 (T cells) CTLA-4 (T cells) | Block T-cell inhibitory signals and activation of T cells | II | NCT02923934 ∗ |
Bevacizumab + cisplatin | GC | VEGF-A (endothelial cells) | Angiogenesis [236] | III | NCT00548548 |
Ramucirumab | Upper GI | Inhibits receptor tyrosine kinase (endothelial cells) | Angiogenesis | II | NCT02241720 |
GEJ: gastroesophageal junction. ∗ongoing; #terminated.
The tumor stroma can also play an important role in restraining tumor growth, mainly due to the heterogeneous population of fibroblast present in PDAC. Preclinical studies identified a CAF subpopulation expressing high amounts of α-SMA close to tumor cells and CAF subpopulations expressing low α-SMA and secreting IL-6 which could be responsible for the aggressiveness of PDAC [193, 201]. A phase Ib study using enzymatic ablation of hyaluronan by PEGPH20, a PEGylated recombinant hyaluronidase, in combination with gemcitabine showed a potential therapeutic benefit, especially in patients with high expression of HA [232]. In fact, a phase II clinical trial using PEGPH20 in association with gemcitabine and nab-paclitaxel showed improvement in the progression-free survival of PDAC patients [233] and it is now being evaluated in a phase III trial (Table 3). However, PEGPH20 in association with a modified FOLFIRINOX regimen presented high toxicity when compared with FOLFIRINOX alone [238].
Other strategies to inhibit FAP in PDAC showed promising results in preclinical studies [121, 239, 240]. For example, anti-FAP CAR T cells can deplete FAP+ cells in PDAC and decrease tumor growth through promotion of antitumor immunity [241]. Recently, a preclinical trial using a DNA vaccine against FAP synergized with anticancer immune therapy targeting Prostate Membrane Antigen (PMSA) in tumor-bearing mice model for prostate cancer [242]. This result suggests that therapies which target both stroma components and tumor cells might be effective for tumors expressing high amounts of FAP, such as CRC and PDAC.
8.2. Immune System
PD-1 is expressed on a large proportion of TILs from many different cancer types, while its ligand, PD-L1, is mainly expressed in antigen-presenting cells and tumor cells [243]. Since tumors can escape the T cell immune response by expressing these molecules, the blockade of this pathway has emerged as a promising anticancer strategy. This approach also showed good results as second and third line of chemotherapy in gastro-esophageal cancer [244, 245]. A clinical study evaluating nivolumab (an antibody against PD-1) monotherapy in heavily pretreated patients with advanced gastric or gastro-esophageal junction cancer showed an increased 12-month overall survival rate compared to the placebo group [244]. In another trial, both objective and complete responses were observed in patients with gastro-esophageal cancer treated with pembrolizumab (an antibody against PD-1) monotherapy, irrespective of PD-L1 tumor expression. Nonetheless, pembrolizumab conferred longer response duration in those patients with PD-L1–positive tumors [245].
CTL4-A (cytotoxic T-lymphocyte antigen 4) signaling diminishes immune response against tumor cells and the use of antibodies against CTL4-A was effective in treating tumors as melanomas [246]. However, clinical trials in PDAC using monotherapies with CTLA-4 or PD-1 inhibitors presented low response rates [247, 248], with the exception of the PDAC patient subpopulation with microsatelite instability [249]. Although the response rates from these studies remain discouraging, they could be improved by combinatory therapies. A phase II study with Nivolumab in association with Ipilimumab (an antibody against CTLA-4) in patients presenting upper GI cancers is ongoing (see Table 3). The first trials using nivolumab and pembrolizumab in HCC were encouraging [250, 251]. However, pembrolizumab as second line of treatment did not meet its coprimary endpoints of overall survival and progression-free survival [252]. A phase III trial with nivolumab in first line treatment is currently underway. Unfortunately, when a better selection of patients based on molecular characteristics from the tumor or on its etiology was performed, the data was inconclusive [250, 251]. Another study showed that a therapy targeting FAP+ cells that express CXCL12 synergized with anti-PD-L1 immunotherapy in PDAC [121], and inhibition of its receptor, CXCR4, in sorafenib-treated HCC facilitates anti-PDL-1 immunotherapy [253]. In addition, CXCR4 inhibition increased PD-1 therapy response by inducing mobilization of CD8+ T cells in PDAC [254]. Together, these studies demonstrate important systemic components that might play a role in the clinical outcome and explain, in part, the heterogeneous therapeutic response normally found in these clinical trials.
Natural Killer Cells Antigen 94 (CD94/NGK2a) is the main HLA-E receptor which mediates an inhibitory effect on CD8+ CTL and NK cells, promoting immune evasion in CRC [255]. In fact, increased levels of NGK2a-CD94+ TILs correlate with poor survival in CRC patients [256]. Although metastatic microsatellite-stable CRC patients do not respond to therapies that involve PD-1/PD-L1 blockade [257], a first phase I clinical trial studying an antibody against PD-1 (durvalumab) in combination with an antibody targeting CD94/NGK2a (monalizumab) is ongoing.
Immunotherapy checkpoints have been suggested as a good strategy to impair cancer progression [258], and strategies targeting both the innate and the adaptive immune systems show promising results in CCR [259]. CCL2, which is highly expressed in PDAC, is a chemoattractant for T cells, monocytes, and natural killer cells. CCL2 binds to its receptor, C-C Chemokine Receptor (CCR) 2, which is expressed in monocytes and controls its differentiation into TAMs [260]. CCR2 inhibition in combination with FOLFIRINOX in PDAC has been tried in phase I clinical trial, and the results showed that it was safe and well tolerated [261].
Another mention worthy molecule is CD47, an integrin-associated transmembrane protein. This integrin is overexpressed in solid cancers (e.g. CRC) and is correlated to a poor clinical outcome [262]. Both TAMs and DCs can express the CD47 receptor, signal regulatory protein alpha (SIRPα). The binding of CD47 to SIRPα inhibits phagocytosis of cancer these cells, enabling the tumor to evade immune destruction by first responder cells, such as macrophages [263]. Thus, restoring phagocytosis activity by antigen-presenting cells can enhance antigen priming of T cells. A phase I clinical trial recently described the use of the monoclonal antibody against CD47 Hu5F9-G4 in CRC and PDAC [264]. A Phase II study to evaluate Hu5F9-G4 in combination with cetuximab is ongoing in CRC [265].
As previously mentioned, the M2 macrophages are frequently found in TMEN. Since the intratumoral presence of Macrophage Colony-Stimulating Factor Receptor (CSFR)1+ macrophages correlates with the clinical aggressiveness of pancreatic neuroendocrine tumors [266], targeting CSFR1 signaling in TAMs represents an attractive strategy to eliminate these cells and block M2 polarization. A clinical trial using a monoclonal antibody against CSFR1, AMG 820, showed safety and tolerability in patients with advanced solid tumors, including CRC and PDAC [234]. However, since the study did not present significant tumor responses, it was terminated before enrollment into the dose-expansion phase. Preclinical studies have also examined the effects of CSFR1 inhibitors in combination with T-cell target therapies to improve efficacy in PDAC [267]. In fact, a clinical trial using pembrolizumab in combination with AMG 820 is ongoing in PDAC and in CRC, with an estimated date of completion in 2020 (Table 3).
8.3. Angiogenesis
Approaches focused on anti-angiogenesis cancer therapies have been studied in several clinical trials. In GC the results of trials with anti-VEGF were disappointing on the first line treatments (either with bevacizumab or with ramucirumab) [236, 268]. Interestingly, the use of ramucirumab (anti-VEGF2) in association with paclitaxel or in monotherapy showed a significant improvement on the overall survival of gastro-esophageal adenocarcinoma patients and has been approved in this setting [268, 269].
In HCC, the use of tyrosine kinases with antiangiogenic effects were the basis of systemic treatment. Since the first approved drug, sorafenib, several clinical studies showed improvement in clinical outcomes with regorafenib, ramucirumab or cabozantinib, expanding the repertoire of drugs that can be used in this particular disease [220, 270–272].
In CRC, the use of bevacizumab in association FOLFIRI (Folinic Acid, 5-FU, Irinotecan) or FOLFOX (Folinic Acid, 5-FU, oxaliplatin) showed a significant increase in overall survival, being nowadays the standard of care for patients in the metastatic stage of this disease [273]. Nonetheless, the use of bevacizumab as part of the adjuvant chemotherapy treatment in CRC patients was detrimental for survival [274]. Other drugs that change the tumoral angiogenesis, such as the VEGF 1 and 2 inhibitors ziv-aflibercept and ramucirumab, have shown an improvement in overall survival in patients with CRC when in combination with chemotherapy in second line setting after failure of a previous line of chemotherapy [275, 276].
9. Conclusions and Perspectives
Tumor masses are not cancer cells-centered entities that drive malignant progression. Instead, tumor development depends on the complex and intricate tapestry of cell-cell interactions where nontransformed cells of the TMEN play key role in cancer biology. We here summarized how stromal cells can impact tumor growth and progression as well as resistance to antitumor treatment. In fact, we show that most of these cells are important oncogenic drivers, frequently associated with poor prognosis. Therefore, the development of new therapeutic approaches directed to components of the TMEN still has a great unexplored potential. The main challenge on TMEN-directed approaches resides on the complexity of the interactions within the microenvironment, where the same cell type can have opposite effects in tumor growth and progression depending on its cell-to-cell interaction. This is not surprising considering the pleotropic diversity of all the stromal cells described here. Therefore, ideal targeted therapy is unlikely to be solely affecting a single cell type. Instead, the best therapeutic approaches should be those that are capable of tipping the whole balance in favor of tumor inhibition.
Acknowledgments
The authors thank Joana Maia and Cristian Bodo for their support in the final preparation of the manuscript. S. B. was supported by the EMBO Installation Grant 3921. A. G. was supported by the grant 2017NovPCC1058 from Breast Cancer Now's Catalyst Programme, which is supported by funding from Pfizer. This work was supported by the Champalimaud Foundation, the grant 751547 from H2020-MSCA-IF-2016 and the grant 765492 from H2020-MSCA-ITN-2017 and the grant 19/00740-0 from FAPESP.
Abbreviations
- GI:
Gastrointestinal
- PDAC:
Pancreatic ductal adenocarcinoma
- BM:
Bone marrow
- TMEN:
Tumor microenvironment
- BMDC:
Bone marrow-derived cells
- GC:
Gastric cancer
- CRC:
Colorectal carcinoma
- MMP:
Metalloproteinase
- MSC:
Mesenchymal stem cells
- VEGF:
Vascular endothelial growth factor
- IFN-γ:
Interferon gamma
- TNF-α:
Tumor necrosis factor alpha
- HIF-1α:
Hypoxia-induced factor 1 alpha
- S100A8/9:
S100 calcium binding protein A8/A9
- HCC:
Hepatocellular carcinoma
- PTEN:
Phosphatase and tensin homolog
- HSC:
Hematopoietic stem cells
- CAF:
Cancer-associated fibroblasts
- ECM:
Extracellular matrix
- TGF-β:
Transforming growth factor beta
- HGF:
Hepatocyte growth factor
- FSP1:
Fibroblast-specific protein 1
- Lkb1:
Liver kinase B1
- IL:
Interleukin
- JAK:
Janus kinase
- STAT3:
Signal transducer and activator of transcription 3
- PGE2:
Prostaglandin E2
- EMT:
Epithelial-to-mesenchymal transition
- PDGF:
Platelet-derived growth factor
- FAP:
Fibroblast activation protein
- CCL:
C-C motif chemokine
- CXCL:
C-X-C motif ligand
- CXCR:
C-X-C motif receptor
- GM-CSF:
Granulocyte-macrophage colony-stimulating factor
- SDF-1:
Stromal cell-derived factor 1
- α-SMA:
Alpha smooth muscle actin
- HUVEC:
Human umbilical vein endothelial cells
- ECs:
Endothelial cells
- AKT:
Serine/threonine-protein kinase
- ESC:
Esophageal squamous carcinoma
- LEC:
Lymphatic endothelial cells
- TIMP:
Tissue inhibitor of metalloproteinases
- PD-L1:
Programmed death-ligand 1
- IDO:
Indoleamine 2,3-dioxygenase
- NK:
Natural killer
- TRegs:
T regulatory cells
- FOXP3:
Forkhead box P3
- DC:
Dendritic cells
- Th2:
Type II helper T cells
- MDSC:
Myeloid-derived suppressor cells
- PD-1:
Programmed death-1
- Fas:
Apoptosis-mediating surface antigen FAS
- FasL:
Fas ligand
- MIF:
Migration inhibitory factor
- TIL:
Tumor lymphocyte infiltrates
- HA:
Hyaluronic acid
- Shh:
Sonic hedgehog
- 5-FU:
5-Fluorouracil
- TAM:
Tumor-associated macrophages
- LOXL2:
Lysyl oxidase-like 2
- SMO:
Smoothened
- FOLFIRINOX:
Folinic acid, 5-FU, irinotecan, oxaliplatin
- CD94/NGK2a:
Natural killer cells antigen 94
- CCR:
C-C chemokine receptor
- CSFR:
Macrophage colony-stimulating factor receptor.
Conflicts of Interest
The authors declare no conflicts of financial interest.
Authors' Contributions
S. B. and A. G. wrote the sections on BMDCs, CAF, ECs, and immune cells. A. O. and N. C. wrote the sections on stromal signatures of prognostic, tumor resistance, and therapy strategies. B. C. S. conceived and structured the review. All authors wrote and reviewed the manuscript.
References
- 1.Bray F., Ferlay J., Soerjomataram I., Siegel R. L., Torre L. A., Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians. 2018;68(6):394–424. doi: 10.3322/caac.21492. [DOI] [PubMed] [Google Scholar]
- 2.Rahib L., Smith B. D., Aizenberg R., Rosenzweig A. B., Fleshman J. M., Matrisian L. M. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Research. 2014;74(11):2913–2921. doi: 10.1158/0008-5472.can-14-0155. [DOI] [PubMed] [Google Scholar]
- 3.Benson A. B., Venook A. P., Al-Hawary M. M., et al. NCCN guidelines insights: colon cancer, version 2.2018. Journal of the National Comprehensive Cancer Network. 2018;16(4):359–369. doi: 10.6004/jnccn.2018.0021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Benson A. B., Venook A. P., Al-Hawary M. M., et al. Rectal cancer, version 2.2015. Journal of the National Comprehensive Cancer Network. 2015;13:719–728. doi: 10.6004/jnccn.2015.0100. [DOI] [PubMed] [Google Scholar]
- 5.Ilson D. H. Advances in the treatment of gastric cancer. Current Opinion in Gastroenterology. 2018;34(6):465–468. doi: 10.1097/mog.0000000000000475. [DOI] [PubMed] [Google Scholar]
- 6.Ducreux M., Cuhna A. S., Caramella C., et al. Cancer of the pancreas: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Annals of Oncology. 2015;26(5):v56–v68. doi: 10.1093/annonc/mdv295. [DOI] [PubMed] [Google Scholar]
- 7.Plentz R. R., Malek N. P. Systemic therapy of cholangiocarcinoma. Visceral Medicine. 2016;32(6):427–430. doi: 10.1159/000453084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Huang L., Jansen L., Balavarca Y., et al. Stratified survival of resected and overall pancreatic cancer patients in Europe and the USA in the early twenty-first century: a large, international population-based study. BMC Medicine. 2018;16(1):p. 125. doi: 10.1186/s12916-018-1120-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Joyce J. A., Pollard J. W. Microenvironmental regulation of metastasis. Nature Reviews Cancer. 2009;9(4):239–252. doi: 10.1038/nrc2618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Hanahan D., Weinberg R. A. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- 11.Sis B., Sarioglu S., Sokmen S., Sakar M., Kupelioglu A., Fuzun M. Desmoplasia measured by computer assisted image analysis: an independent prognostic marker in colorectal carcinoma. Journal of Clinical Pathology. 2005;58(1):32–38. doi: 10.1136/jcp.2004.018705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Ueno H., Jones A. M., Wilkinson K. H., Jass J. R., Talbot I. C. Histological categorisation of fibrotic cancer stroma in advanced rectal cancer. Gut. 2004;53(4):581–586. doi: 10.1136/gut.2003.028365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Vennin C., Murphy K. J., Morton J. P., Cox T. R., Pajic M., Timpson P. Reshaping the tumor stroma for treatment of pancreatic cancer. Gastroenterology. 2018;154(4):820–838. doi: 10.1053/j.gastro.2017.11.280. [DOI] [PubMed] [Google Scholar]
- 14.Fang M., Yuan J., Chen M., et al. The heterogenic tumor microenvironment of hepatocellular carcinoma and prognostic analysis based on tumor neo-vessels, macrophages and α-SMA. Oncology Letters. 2018;15:4805–4812. doi: 10.3892/ol.2018.7946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.McGuigan A., Kelly P., Turkington R. C., Jones C., Coleman H. G., McCain R. S. Pancreatic cancer: a review of clinical diagnosis, epidemiology, treatment and outcomes. World Journal of Gastroenterology. 2018;24(43):4846–4861. doi: 10.3748/wjg.v24.i43.4846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Waseem D., Tushar P. Intrahepatic, perihilar and distal cholangiocarcinoma: management and outcomes. Annals of Hepatology. 2017;16(1):133–139. doi: 10.5604/16652681.1226927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Gao D., Mittal V. The role of bone-marrow-derived cells in tumor growth, metastasis initiation and progression. Trends in Molecular Medicine. 2009;15(8):333–343. doi: 10.1016/j.molmed.2009.06.006. [DOI] [PubMed] [Google Scholar]
- 18.Hutchinson L., Stenstrom B., Chen D. Human Barrett’s adenocarcinoma of the esophagus, associated myofibroblasts, and endothelium can arise from bone marrow-derived cells after allogeneic stem cell transplant. Stem Cells and Development. 2010;20(1):11–17. doi: 10.1089/scd.2010.0139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Sarosi G., Brown G., Jaiswal K., et al. Bone marrow progenitor cells contribute to esophageal regeneration and metaplasia in a rat model of Barrett’s esophagus. Diseases of the Esophagus. 2008;21(1):43–50. doi: 10.1111/j.1442-2050.2007.00744.x. [DOI] [PubMed] [Google Scholar]
- 20.Houghton J., Stoicov C., Nomura S., et al. Gastric cancer originating from bone marrow-derived cells. Science. 2004;306(5701):1568–1571. doi: 10.1126/science.1099513. [DOI] [PubMed] [Google Scholar]
- 21.Varon C., Dubus P., Mazurier F., et al. Helicobacter pylori infection recruits bone marrow–derived cells that participate in gastric preneoplasia in mice. Gastroenterology. 2012;142(2):281–291. doi: 10.1053/j.gastro.2011.10.036. [DOI] [PubMed] [Google Scholar]
- 22.Zhang Y., Davis C., Ryan J., Janney C., Peña M. M. O. Development and characterization of a reliable mouse model of colorectal cancer metastasis to the liver. Clinical & Experimental Metastasis. 2013;30(7):903–918. doi: 10.1007/s10585-013-9591-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kitamura T., Kometani K., Hashida H., et al. SMAD4-deficient intestinal tumors recruit CCR1+ myeloid cells that promote invasion. Nature Genetics. 2007;39(4):467–475. doi: 10.1038/ng1997. [DOI] [PubMed] [Google Scholar]
- 24.Ridge S. M., Sullivan F. J., Glynn S. A. Mesenchymal stem cells: key players in cancer progression. Molecular Cancer. 2017;16(1):p. 31. doi: 10.1186/s12943-017-0597-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Beckermann B. M., Kallifatidis G., Groth A., et al. VEGF expression by mesenchymal stem cells contributes to angiogenesis in pancreatic carcinoma. British Journal of Cancer. 2008;99(4):622–631. doi: 10.1038/sj.bjc.6604508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Choi J. H., Lee Y. B., Jung J., Hwang S. G., Oh I.-H., Kim G. J. Hypoxia inducible factor-1α regulates the migration of bone marrow mesenchymal stem cells via integrin α4. Stem Cells International. 2016;2016:11. doi: 10.1155/2016/7932185.7932185 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Hung S.-C., Pochampally R. R., Hsu S.-C., et al. Short-term exposure of multipotent stromal cells to low oxygen increases their expression of CX3CR1 and CXCR4 and their engraftment in vivo. PLoS One. 2007;2(5) doi: 10.1371/journal.pone.0000416.e416 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Rosová I., Dao M., Capoccia B., Link D., Nolta J. A. Hypoxic preconditioning results in increased motility and improved therapeutic potential of human mesenchymal stem cells. Stem Cells. 2008;26(8):2173–2182. doi: 10.1634/stemcells.2007-1104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Yu J., Yin S., Zhang W., et al. Hypoxia preconditioned bone marrow mesenchymal stem cells promoted liver regeneration in a rat massive hepatectomy model. Stem Cell Research & Therapy. 2013;4(4):p. 83. doi: 10.1186/scrt234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Liu Y., Han Z.-P., Zhang S.-S., et al. Effects of inflammatory factors on mesenchymal stem cells and their role in the promotion of tumor angiogenesis in colon cancer. Journal of Biological Chemistry. 2011;286(28):25007–25015. doi: 10.1074/jbc.M110.213108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Yan X.-L., Jia Y.-L., Chen L., et al. Hepatocellular carcinoma-associated mesenchymal stem cells promote hepatocarcinoma progression: role of the S100A4-miR155-SOCS1-MMP9 axis. Hepatology. 2013;57(6):2274–2286. doi: 10.1002/hep.26257. [DOI] [PubMed] [Google Scholar]
- 32.Maia J., Caja S., Strano Moraes M. C., Couto N., Costa-Silva B. Exosome-based cell-cell communication in the tumor microenvironment. Frontiers in Cell and Developmental Biology. 2018;6:p. 18. doi: 10.3389/fcell.2018.00018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Couto N., Caja S., Maia J., Strano Moraes M. C., Costa-Silva B. Exosomes as emerging players in cancer biology. Biochimie. 2018;155:2–10. doi: 10.1016/j.biochi.2018.03.006. [DOI] [PubMed] [Google Scholar]
- 34.Wang M., Zhao C., Shi H., et al. Deregulated microRNAs in gastric cancer tissue-derived mesenchymal stem cells: novel biomarkers and a mechanism for gastric cancer. British Journal of Cancer. 2014;110(5):1199–1210. doi: 10.1038/bjc.2014.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Mishra P. J., Mishra P. J., Humeniuk R., et al. Carcinoma-associated fibroblast-like differentiation of human mesenchymal stem cells. Cancer Research. 2008;68(11):4331–4339. doi: 10.1158/0008-5472.CAN-08-0943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Spaeth E. L., Dembinski J. L., Sasser A. K., et al. Mesenchymal stem cell transition to tumor-associated fibroblasts contributes to fibrovascular network expansion and tumor progression. PLoS One. 2009;4(4) doi: 10.1371/journal.pone.0004992.e4992 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Shinagawa K., Kitadai Y., Tanaka M., et al. Mesenchymal stem cells enhance growth and metastasis of colon cancer. International Journal of Cancer. 2010;127(10):2323–2333. doi: 10.1002/ijc.25440. [DOI] [PubMed] [Google Scholar]
- 38.Weiskopf K., Schnorr P. J., Pang W. W., et al. Myeloid cell origins, differentiation, and clinical implications. Microbiology Spectrum. 2016;4(5) doi: 10.1128/microbiolspec.MCHD-0031-2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Yin T., Li L. The stem cell niches in bone. Journal of Clinical Investigation. 2006;116(5):1195–1201. doi: 10.1172/JCI28568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Tarin D., Croft C. B. Ultrastructural studies of wound healing in mouse skin. II. Dermo-epidermal interrelationships. Journal of Anatomy. 1970;106:79–91. [PMC free article] [PubMed] [Google Scholar]
- 41.Gabbiani G., Ryan G. B., Majno G. Presence of modified fibroblasts in granulation tissue and their possible role in wound contraction. Experientia. 1971;27(5):549–550. doi: 10.1007/bf02147594. [DOI] [PubMed] [Google Scholar]
- 42.Darby I. A., Laverdet B., Bonté F., Desmoulière A. Fibroblasts and myofibroblasts in wound healing. Clinical, Cosmetic and Investigational Dermatology. 2014;7:301–311. doi: 10.2147/CCID.S50046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Desmoulière A., Redard M., Darby I., Gabbiani G. Apoptosis mediates the decrease in cellularity during the transition between granulation tissue and scar. The American Journal of Pathology. 1995;146(1):56–66. [PMC free article] [PubMed] [Google Scholar]
- 44.Tsujino T., Seshimo I., Yamamoto H., et al. Stromal myofibroblasts predict disease recurrence for colorectal cancer. Clinical Cancer Research. 2007;13(7):2082–2090. doi: 10.1158/1078-0432.CCR-06-2191. [DOI] [PubMed] [Google Scholar]
- 45.Bhowmick N. A., Chytil A., Plieth D., et al. TGF-signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia. Science. 2004;303(5659):848–851. doi: 10.1126/science.1090922. [DOI] [PubMed] [Google Scholar]
- 46.Lau E. Y. T., Lo J., Cheng B. Y. L., et al. Cancer-associated fibroblasts regulate tumor-initiating cell plasticity in hepatocellular carcinoma through c-Met/FRA1/HEY1 signaling. Cell Reports. 2016;15(6):1175–1189. doi: 10.1016/j.celrep.2016.04.019. [DOI] [PubMed] [Google Scholar]
- 47.Vermeulen L., De Sousa E Melo F., van der Heijden M., et al. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nature Cell Biology. 2010;12(5):468–476. doi: 10.1038/ncb2048. [DOI] [PubMed] [Google Scholar]
- 48.Ollila S., Domènech-Moreno E., Laajanen K., et al. Stromal Lkb1 deficiency leads to gastrointestinal tumorigenesis involving the IL-11-JAK/STAT3 pathway. The Journal of Clinical Investigation. 2018;128(1):402–414. doi: 10.1172/JCI93597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Li P., Shan J.-X., Chen X.-H., et al. Epigenetic silencing of microRNA-149 in cancer-associated fibroblasts mediates prostaglandin E2/interleukin-6 signaling in the tumor microenvironment. Cell Research. 2015;25(5):588–603. doi: 10.1038/cr.2015.51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Mrazek A., Carmical J., Wood T., Hellmich M., Eltorky M., Chao C. Colorectal cancer-associated fibroblasts are genotypically distinct. Current Cancer Therapy Reviews. 2014;10(2):97–218. doi: 10.2174/157339471002141124123103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Albrengues J., Bertero T., Grasset E., et al. Epigenetic switch drives the conversion of fibroblasts into proinvasive cancer-associated fibroblasts. Nature Communications. 2015;6(1) doi: 10.1038/ncomms10204.10204 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Polyak K., Weinberg R. A. Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Nature Reviews Cancer. 2009;9(4):265–273. doi: 10.1038/nrc2620. [DOI] [PubMed] [Google Scholar]
- 53.Giannoni E., Bianchini F., Masieri L., et al. Reciprocal activation of prostate cancer cells and cancer-associated fibroblasts stimulates epithelial-mesenchymal transition and cancer stemness. Cancer Research. 2010;70(17):6945–6956. doi: 10.1158/0008-5472.can-10-0785. [DOI] [PubMed] [Google Scholar]
- 54.Yu Y., Xiao C.-H., Tan L.-D., Wang Q.-S., Li X.-Q., Feng Y.-M. Cancer-associated fibroblasts induce epithelial-mesenchymal transition of breast cancer cells through paracrine TGF-β signalling. British Journal of Cancer. 2014;110(3):724–732. doi: 10.1038/bjc.2013.768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Wang Q., Qu C., Xie F., et al. Curcumin suppresses epithelial-to-mesenchymal transition and metastasis of pancreatic cancer cells by inhibiting cancer-associated fibroblasts. American Journal of Cancer Research. 2017;7(1):125–133. [PMC free article] [PubMed] [Google Scholar]
- 56.van Zijl F., Mair M., Csiszar A., et al. Hepatic tumor-stroma crosstalk guides epithelial to mesenchymal transition at the tumor edge. Oncogene. 2009;28(45):4022–4033. doi: 10.1038/onc.2009.253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Wu X., Tao P., Zhou Q., et al. IL-6 secreted by cancer-associated fibroblasts promotes epithelial-mesenchymal transition and metastasis of gastric cancer via JAK2/STAT3 signaling pathway. Oncotarget. 2017;8(13):20741–20750. doi: 10.18632/oncotarget.15119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Zhang Z., Li X., Sun W., et al. Loss of exosomal miR-320a from cancer-associated fibroblasts contributes to HCC proliferation and metastasis. Cancer Letters. 2017;397:33–42. doi: 10.1016/j.canlet.2017.03.004. [DOI] [PubMed] [Google Scholar]
- 59.Rai A., Greening D. W., Chen M., Xu R., Ji H., Simpson R. J. Exosomes derived from human primary and metastatic colorectal cancer cells contribute to functional heterogeneity of activated fibroblasts by reprogramming their proteome. Proteomics. 2019;19(8) doi: 10.1002/pmic.201800148.1800148 [DOI] [PubMed] [Google Scholar]
- 60.Gaggioli C., Hooper S., Hidalgo-Carcedo C., et al. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nature Cell Biology. 2007;9(12):1392–1400. doi: 10.1038/ncb1658. [DOI] [PubMed] [Google Scholar]
- 61.Levental K. R., Yu H., Kass L., et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell. 2009;139(5):891–906. doi: 10.1016/j.cell.2009.10.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Goetz J. G., Minguet S., Navarro-Lérida I., et al. Biomechanical remodeling of the microenvironment by stromal caveolin-1 favors tumor invasion and metastasis. Cell. 2011;146(1):148–163. doi: 10.1016/j.cell.2011.05.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Yu B., Chen X., Li J., et al. Stromal fibroblasts in the microenvironment of gastric carcinomas promote tumor metastasis via upregulating TAGLN expression. BMC Cell Biology. 2013;14(1):p. 17. doi: 10.1186/1471-2121-14-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Lee H.-O., Mullins S. R., Franco-Barraza J., Valianou M., Cukierman E., Cheng J. D. FAP-overexpressing fibroblasts produce an extracellular matrix that enhances invasive velocity and directionality of pancreatic cancer cells. BMC Cancer. 2011;11(1):p. 245. doi: 10.1186/1471-2407-11-245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Liu J., Chen S., Wang W., et al. Cancer-associated fibroblasts promote hepatocellular carcinoma metastasis through chemokine-activated hedgehog and TGF-β pathways. Cancer Letters. 2016;379(1):49–59. doi: 10.1016/j.canlet.2016.05.022. [DOI] [PubMed] [Google Scholar]
- 66.Waghray M., Yalamanchili M., Dziubinski M., et al. GM-CSF mediates mesenchymal-epithelial cross-talk in pancreatic cancer. Cancer Discovery. 2016;6(8):886–899. doi: 10.1158/2159-8290.cd-15-0947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Pagat S. The distribution of secondary growths in cancer of the breast. Cancer and Metastasis Reviews. 1989;8(2):98–101. [PubMed] [Google Scholar]
- 68.Kaplan R. N., Riba R. D., Zacharoulis S., et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature. 2005;438(7069):820–827. doi: 10.1038/nature04186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Duda D. G., Duyverman A. M. M. J., Kohno M., et al. Malignant cells facilitate lung metastasis by bringing their own soil. Proceedings of the National Academy of Sciences. 2010;107(50):21677–21682. doi: 10.1073/pnas.1016234107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Calon A., Espinet E., Palomo-Ponce S., et al. Dependency of colorectal cancer on a TGF-β-driven program in stromal cells for metastasis initiation. Cancer Cell. 2012;22(5):571–584. doi: 10.1016/j.ccr.2012.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Fukumura D., Xavier R., Sugiura T., et al. Tumor induction of VEGF promoter activity in stromal cells. Cell. 1998;94(6):715–725. doi: 10.1016/s0092-8674(00)81731-6. [DOI] [PubMed] [Google Scholar]
- 72.Orimo A., Gupta P. B., Sgroi D. C., et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005;121(3):335–348. doi: 10.1016/j.cell.2005.02.034. [DOI] [PubMed] [Google Scholar]
- 73.Guo X., Oshima H., Kitmura T., Taketo M. M., Oshima M. Stromal fibroblasts activated by tumor cells promote angiogenesis in mouse gastric cancer. Journal of Biological Chemistry. 2008;283(28):19864–19871. doi: 10.1074/jbc.M800798200. [DOI] [PubMed] [Google Scholar]
- 74.Nagasaki T., Hara M., Nakanishi H., Takahashi H., Sato M., Takeyama H. Interleukin-6 released by colon cancer-associated fibroblasts is critical for tumour angiogenesis: anti-interleukin-6 receptor antibody suppressed angiogenesis and inhibited tumour-stroma interaction. British Journal of Cancer. 2014;110(2):469–478. doi: 10.1038/bjc.2013.748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Masamune A., Kikuta K., Watanabe T., Satoh K., Hirota M., Shimosegawa T. Hypoxia stimulates pancreatic stellate cells to induce fibrosis and angiogenesis in pancreatic cancer. American Journal of Physiology-Gastrointestinal and Liver Physiology. 2008;295(4):G709–G717. doi: 10.1152/ajpgi.90356.2008. [DOI] [PubMed] [Google Scholar]
- 76.Ankoma-Sey V., Wang Y., Dai Z. Hypoxic stimulation of vascular endothelial growth factor expression in activated rat hepatic stellate cells. Hepatology. 2000;31(1):141–148. doi: 10.1002/hep.510310122. [DOI] [PubMed] [Google Scholar]
- 77.Taura K., De Minicis S., Seki E., et al. Hepatic stellate cells secrete angiopoietin 1 that induces angiogenesis in liver fibrosis. Gastroenterology. 2008;135(5):1729–1738. doi: 10.1053/j.gastro.2008.07.065. [DOI] [PubMed] [Google Scholar]
- 78.Lu J., Ye X., Fan F., et al. Endothelial cells promote the colorectal cancer stem cell phenotype through a soluble form of jagged-1. Cancer Cell. 2013;23(2):171–185. doi: 10.1016/j.ccr.2012.12.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Wang R., Bhattacharya R., Ye X., et al. Endothelial cells activate the cancer stem cell-associated NANOGP8 pathway in colorectal cancer cells in a paracrine fashion. Molecular Oncology. 2017;11(8):1023–1034. doi: 10.1002/1878-0261.12071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Rawal P., Siddiqui H., Hassan M., et al. Endothelial cell-derived TGF-β promotes epithelial-mesenchymal transition via CD133 in HBx-infected hepatoma cells. Frontiers in Oncology. 2019;9:p. 308. doi: 10.3389/fonc.2019.00308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Liu H., Zhao J., Yang Y. Human umbilical vein endothelial cell vaccine suppresses the angiogenesis of esophageal squamous cell carcinoma in a humanized mouse model. Oncology Reports. 2018;40:3006–3014. doi: 10.3892/or.2018.6653. [DOI] [PubMed] [Google Scholar]
- 82.Okaji Y., Tsuno N. H., Tanaka M., et al. Pilot study of anti-angiogenic vaccine using fixed whole endothelium in patients with progressive malignancy after failure of conventional therapy. European Journal of Cancer. 2008;44(3):383–390. doi: 10.1016/j.ejca.2007.10.018. [DOI] [PubMed] [Google Scholar]
- 83.Ungaro F., Colombo P., Massimino L., et al. Lymphatic endothelium contributes to colorectal cancer growth via the soluble matrisome component GDF11. International Journal of Cancer. 2019;145(7):1913–1920. doi: 10.1002/ijc.32286. [DOI] [PubMed] [Google Scholar]
- 84.Yang X., Zhai N., Sun M., et al. Influence of lymphatic endothelial cells on proliferation and invasiveness of esophageal carcinoma cells in vitro and lymphangiogenesis in vivo. Medical Oncology. 2015;32(8):p. 222. doi: 10.1007/s12032-015-0662-3. [DOI] [PubMed] [Google Scholar]
- 85.Tokumoto M., Tanaka H., Tauchi Y., et al. Immunoregulatory function of lymphatic endothelial cells in tumor-draining lymph nodes of human gastric cancer. Anticancer Research. 2017;37:2875–2883. doi: 10.21873/anticanres.11640. [DOI] [PubMed] [Google Scholar]
- 86.Hayakawa Y., Ariyama H., Stancikova J., et al. Mist1 expressing gastric stem cells maintain the normal and neoplastic gastric epithelium and are supported by a perivascular stem cell niche. Cancer Cell. 2015;28(6):800–814. doi: 10.1016/j.ccell.2015.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Yonenaga Y., Mori A., Onodera H., et al. Absence of smooth muscle actin-positive pericyte coverage of tumor vessels correlates with hematogenous metastasis and prognosis of colorectal cancer patients. Oncology. 2005;69(2):159–166. doi: 10.1159/000087840. [DOI] [PubMed] [Google Scholar]
- 88.Shenoy A. K., Jin Y., Luo H., et al. Epithelial-to-mesenchymal transition confers pericyte properties on cancer cells. Journal of Clinical Investigation. 2016;126(11):4174–4186. doi: 10.1172/JCI86623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Sun T., Zhao N., Zhao X.-l., et al. Expression and functional significance of Twist1 in hepatocellular carcinoma: its role in vasculogenic mimicry. Hepatology. 2010;51(2):545–556. doi: 10.1002/hep.23311. [DOI] [PubMed] [Google Scholar]
- 90.Kim H. S., Won Y. J., Shim J. H., et al. Morphological characteristics of vasculogenic mimicry and its correlation with EphA2 expression in gastric adenocarcinoma. Scientific Reports. 2019;9(1):p. 3414. doi: 10.1038/s41598-019-40265-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Li M., Gu Y., Zhang Z., et al. Vasculogenic mimicry: a new prognostic sign of gastric adenocarcinoma. Pathology & Oncology Research. 2010;16(2):259–266. doi: 10.1007/s12253-009-9220-7. [DOI] [PubMed] [Google Scholar]
- 92.Zeng Z., Li Y., Pan Y., et al. Cancer-derived exosomal miR-25-3p promotes pre-metastatic niche formation by inducing vascular permeability and angiogenesis. Nature Communications. 2018;9(1) doi: 10.1038/s41467-018-07810-w.5395 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Sun B., Zhou Y., Fang Y., Li Z., Gu X., Xiang J. Colorectal cancer exosomes induce lymphatic network remodeling in lymph nodes. International Journal of Cancer. 2019;145(6):1648–1659. doi: 10.1002/ijc.32196. [DOI] [PubMed] [Google Scholar]
- 94.Wang Z., Wang Z., Li G., et al. CXCL1 from tumor-associated lymphatic endothelial cells drives gastric cancer cell into lymphatic system via activating integrin β1/FAK/AKT signaling. Cancer Letters. 2017;385:28–38. doi: 10.1016/j.canlet.2016.10.043. [DOI] [PubMed] [Google Scholar]
- 95.Frisch S. M., Francis H. Disruption of epithelial cell-matrix interactions induces apoptosis. The Journal of Cell Biology. 1994;124(4):619–626. doi: 10.1083/jcb.124.4.619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Yadav A., Kumar B., Yu J.-G., Old M., Teknos T. N., Kumar P. Tumor-associated endothelial cells promote tumor metastasis by chaperoning circulating tumor cells and protecting them from anoikis. PLoS One. 2015;10(10) doi: 10.1371/journal.pone.0141602.e0141602 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Beerepoot L. V., Mehra N., Vermaat J. S. P., Zonnenberg B. A., Gebbink M. F. G. B., Voest E. E. Increased levels of viable circulating endothelial cells are an indicator of progressive disease in cancer patients. Annals of Oncology. 2004;15(1):139–145. doi: 10.1093/annonc/mdh017. [DOI] [PubMed] [Google Scholar]
- 98.Hirose J., Kouro T., Igarashi H., Yokota T., Sakaguchi N., Kincade P. W. A developing picture of lymphopoiesis in bone marrow. Immunological Reviews. 2002;189(1):28–40. doi: 10.1034/j.1600-065x.2002.18904.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Sun J. C., Lanier L. L. NK cell development, homeostasis and function: parallels with CD8+ T cells. Nature Reviews Immunology. 2011;11(10):645–657. doi: 10.1038/nri3044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Dieckmann D., Plottner H., Berchtold S., Berger T., Schuler G. Ex vivo isolation and characterization of Cd4+Cd25+ T cells with regulatory properties from human blood. The Journal of Experimental Medicine. 2001;193(11):1303–1310. doi: 10.1084/jem.193.11.1303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Sakaguchi S. Regulatory T cells: key controllers of immunologic self-tolerance. Cell. 2000;101(5):455–458. doi: 10.1016/s0092-8674(00)80856-9. [DOI] [PubMed] [Google Scholar]
- 102.Fontenot J. D., Gavin M. A., Rudensky A. Y. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nature Immunology. 2003;4(4):330–336. doi: 10.1038/ni904. [DOI] [PubMed] [Google Scholar]
- 103.Rosmarin A. G., Yang Z., Resendes K. K. Transcriptional regulation in myelopoiesis: hematopoietic fate choice, myeloid differentiation, and leukemogenesis. Experimental Hematology. 2005;33(2):131–143. doi: 10.1016/j.exphem.2004.08.015. [DOI] [PubMed] [Google Scholar]
- 104.Fiedler K., Brunner C. The role of transcription factors in the guidance of granulopoiesis. American Journal of Blood Research. 2012;2(1):57–65. [PMC free article] [PubMed] [Google Scholar]
- 105.Murray P. J., Allen J. E., Biswas S. K., et al. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity. 2014;41(1):14–20. doi: 10.1016/j.immuni.2014.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Xue J., Schmidt S. V., Sander J., et al. Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity. 2014;40(2):274–288. doi: 10.1016/j.immuni.2014.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Qian B.-Z., Pollard J. W. Macrophage diversity enhances tumor progression and metastasis. Cell. 2010;141(1):39–51. doi: 10.1016/j.cell.2010.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Gabrilovich D. I., Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nature Reviews Immunology. 2009;9(3):162–174. doi: 10.1038/nri2506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Bronte V., Apolloni E., Cabrelle A., et al. Identification of a CD11b+/Gr-1+/CD31+ myeloid progenitor capable of activating or suppressing CD8+ T cells. Blood. 2000;96(12):3838–3846. doi: 10.1182/blood.v96.12.3838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Mazzoni A., Bronte V., Visintin A., et al. Myeloid suppressor lines inhibit T cell responses by an NO-dependent mechanism. The Journal of Immunology. 2002;168(2):689–695. doi: 10.4049/jimmunol.168.2.689. [DOI] [PubMed] [Google Scholar]
- 111.Auffray C., Sieweke M. H., Geissmann F. Blood monocytes: development, heterogeneity, and relationship with dendritic cells. Annual Review of Immunology. 2009;27(1):669–692. doi: 10.1146/annurev.immunol.021908.132557. [DOI] [PubMed] [Google Scholar]
- 112.Ueha S., Shand F. H. W., Matsushima K. Myeloid cell population dynamics in healthy and tumor-bearing mice. International Immunopharmacology. 2011;11(7):783–788. doi: 10.1016/j.intimp.2011.03.003. [DOI] [PubMed] [Google Scholar]
- 113.Laoui D., Keirsse J., Morias Y., et al. The tumour microenvironment harbours ontogenically distinct dendritic cell populations with opposing effects on tumour immunity. Nature Communications. 2016;7(1) doi: 10.1038/ncomms13720.13720 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Parsonnet J., Friedman G. D., Vandersteen D. P., et al. Helicobacter pylori infection and the risk of gastric carcinoma. New England Journal of Medicine. 1991;325(16):1127–1131. doi: 10.1056/nejm199110173251603. [DOI] [PubMed] [Google Scholar]
- 115.D’Elios M. M., Amedei A., Del Prete G. Helicobacter pylori antigen-specific T-cell responses at gastric level in chronic gastritis, peptic ulcer, gastric cancer and low-grade mucosa-associated lymphoid tissue (MALT) lymphoma. Microbes and Infection. 2003;5(8):723–730. doi: 10.1016/s1286-4579(03)00114-x. [DOI] [PubMed] [Google Scholar]
- 116.Lowenfels A. B., Maisonneuve P., Cavallini G., et al. Pancreatitis and the risk of pancreatic cancer. International pancreatitis study group. New England Journal of Medicine. 1993;328(20):1433–1437. doi: 10.1056/nejm199305203282001. [DOI] [PubMed] [Google Scholar]
- 117.Lakatos P. L., Lakatos L. Risk for colorectal cancer in ulcerative colitis: changes, causes and management strategies. World Journal of Gastroenterology. 2008;14(25):3937–3947. doi: 10.3748/wjg.14.3937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Dong H., Strome S. E., Salomao D. R., et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nature Medicine. 2002;8(8):793–800. doi: 10.1038/nm730. [DOI] [PubMed] [Google Scholar]
- 119.Wu K., Kryczek I., Chen L., Zou W., Welling T. H. Kupffer cell suppression of CD8+ T cells in human hepatocellular carcinoma is mediated by B7-H1/programmed death-1 interactions. Cancer Research. 2009;69(20):8067–8075. doi: 10.1158/0008-5472.CAN-09-0901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Kuang D. M., Zhao Q., Peng C., et al. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. The Journal of Experimental Medicine. 2009;206:1327–1337. doi: 10.1084/jem.20082173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Feig C., Jones J. O., Kraman M., et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proceedings of the National Academy of Sciences. 2013;110(50):20212–20217. doi: 10.1073/pnas.1320318110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Ohigashi Y., Sho M., Yamada Y., et al. Clinical significance of programmed death-1 ligand-1 and programmed death-1 ligand-2 expression in human esophageal cancer. Clinical Cancer Research. 2005;11(8):2947–2953. doi: 10.1158/1078-0432.ccr-04-1469. [DOI] [PubMed] [Google Scholar]
- 123.Wang T., Niu G., Kortylewski M., et al. Regulation of the innate and adaptive immune responses by Stat-3 signaling in tumor cells. Nature Medicine. 2004;10(1):48–54. doi: 10.1038/nm976. [DOI] [PubMed] [Google Scholar]
- 124.Kortylewski M., Kujawski M., Wang T., et al. Inhibiting Stat3 signaling in the hematopoietic system elicits multicomponent antitumor immunity. Nature Medicine. 2005;11(12):1314–1321. doi: 10.1038/nm1325. [DOI] [PubMed] [Google Scholar]
- 125.Yanagimoto H., Takai S., Satoi S., et al. Impaired function of circulating dendritic cells in patients with pancreatic cancer. Clinical Immunology. 2005;114(1):52–60. doi: 10.1016/j.clim.2004.09.007. [DOI] [PubMed] [Google Scholar]
- 126.von Bernstorff W., Spanjaard R. A., Chan A. K., et al. Pancreatic cancer cells can evade immune surveillance via nonfunctional Fas (APO-1/CD95) receptors and aberrant expression of functional Fas ligand. Surgery. 1999;125:73–84. doi: 10.1067/msy.2099.93570. [DOI] [PubMed] [Google Scholar]
- 127.Alderson M. R., Tough T. W., Davis-Smith T., et al. Fas ligand mediates activation-induced cell death in human T lymphocytes. Journal of Experimental Medicine. 1995;181(1):71–77. doi: 10.1084/jem.181.1.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.O’Connell J., O’Sullivan G. C., Collins J. K., Shanahan F. The Fas counterattack: Fas-mediated T cell killing by colon cancer cells expressing Fas ligand. Journal of Experimental Medicine. 1996;184(3):1075–1082. doi: 10.1084/jem.184.3.1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Bennett M. W., O’Connell J., O’Sullivan G. C., et al. Expression of Fas ligand by human gastric adenocarcinomas: a potential mechanism of immune escape in stomach cancer. Gut. 1999;44(2):156–162. doi: 10.1136/gut.44.2.156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Zheng H.-C., Sun J.-M., Wei Z.-L., Yang X.-F., Zhang Y.-C., Xin Y. Expression of Fas ligand and caspase-3 contributes to formation of immune escape in gastric cancer. World Journal of Gastroenterology. 2003;9(7):1415–1420. doi: 10.3748/wjg.v9.i7.1415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Witkiewicz A., Williams T. K., Cozzitorto J., et al. Expression of indoleamine 2,3-dioxygenase in metastatic pancreatic ductal adenocarcinoma recruits regulatory T cells to avoid immune detection. Journal of the American College of Surgeons. 2008;206(5):849–854. doi: 10.1016/j.jamcollsurg.2007.12.014. [DOI] [PubMed] [Google Scholar]
- 132.Sucher R., Kurz K., Weiss G., Margreiter R., Fuchs D., Brandacher G. IDO-mediated tryptophan degradation in the pathogenesis of malignant tumor disease. International Journal of Tryptophan Research. 2010;3:113–120. doi: 10.4137/ijtr.s4157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.De Monte L., Reni M., Tassi E., et al. Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer. The Journal of Experimental Medicine. 2011;208(3):469–478. doi: 10.1084/jem.20101876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Gabitass R. F., Annels N. E., Stocken D. D., Pandha H. A., Middleton G. W. Elevated myeloid-derived suppressor cells in pancreatic, esophageal and gastric cancer are an independent prognostic factor and are associated with significant elevation of the Th2 cytokine interleukin-13. Cancer Immunology, Immunotherapy. 2011;60(10):1419–1430. doi: 10.1007/s00262-011-1028-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Porembka M. R., Mitchem J. B., Belt B. A., et al. Pancreatic adenocarcinoma induces bone marrow mobilization of myeloid-derived suppressor cells which promote primary tumor growth. Cancer Immunology, Immunotherapy. 2012;61(9):1373–1385. doi: 10.1007/s00262-011-1178-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Hoechst B., Ormandy L. A., Ballmaier M., et al. A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4+CD25+Foxp3+ T cells. Gastroenterology. 2008;135(1):234–243. doi: 10.1053/j.gastro.2008.03.020. [DOI] [PubMed] [Google Scholar]
- 137.Ichihara F., Kono K, Takahashi A, Kawaida H, Sugai H, Fujii H. Increased populations of regulatory T cells in peripheral blood and tumor-infiltrating lymphocytes in patients with gastric and esophageal cancers. Clinical Cancer Research. 2003;9(12):4404–4408. [PubMed] [Google Scholar]
- 138.Yuan X.-L., Chen L., Zhang T.-T., et al. Gastric cancer cells induce human CD4+Foxp3+ regulatory T cells through the production of TGF-β1. World Journal of Gastroenterology. 2011;17(15):2019–2027. doi: 10.3748/wjg.v17.i15.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Mizukami Y., Kono K., Kawaguchi Y., et al. Localisation pattern of Foxp3+ regulatory T cells is associated with clinical behaviour in gastric cancer. British Journal of Cancer. 2008;98(1):148–153. doi: 10.1038/sj.bjc.6604149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Fu J., Xu D., Liu Z., et al. Increased regulatory T cells correlate with CD8 T-cell impairment and poor survival in hepatocellular carcinoma patients. Gastroenterology. 2007;132(7):2328–2339. doi: 10.1053/j.gastro.2007.03.102. [DOI] [PubMed] [Google Scholar]
- 141.Li T., Yi S., Liu W., et al. Colorectal carcinoma-derived fibroblasts modulate natural killer cell phenotype and antitumor cytotoxicity. Medical Oncology. 2013;30(3):p. 663. doi: 10.1007/s12032-013-0663-z. [DOI] [PubMed] [Google Scholar]
- 142.Li T., Yang Y., Hua X., et al. Hepatocellular carcinoma-associated fibroblasts trigger NK cell dysfunction via PGE2 and IDO. Cancer Letters. 2012;318(2):154–161. doi: 10.1016/j.canlet.2011.12.020. [DOI] [PubMed] [Google Scholar]
- 143.Panni R. Z., Sanford D. E., Belt B. A., et al. Tumor-induced STAT3 activation in monocytic myeloid-derived suppressor cells enhances stemness and mesenchymal properties in human pancreatic cancer. Cancer Immunology, Immunotherapy. 2014;63(5):513–528. doi: 10.1007/s00262-014-1527-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Liu C.-Y., Xu J.-Y., Shi X.-Y., et al. M2-polarized tumor-associated macrophages promoted epithelial-mesenchymal transition in pancreatic cancer cells, partially through TLR4/IL-10 signaling pathway. Laboratory Investigation. 2013;93(7):844–854. doi: 10.1038/labinvest.2013.69. [DOI] [PubMed] [Google Scholar]
- 145.Wu M.-H., Lee W.-J., Hua K.-T., Kuo M.-L., Lin M.-T. Macrophage infiltration induces gastric cancer invasiveness by activating the β-catenin pathway. PLoS One. 2015;10(7) doi: 10.1371/journal.pone.0134122.e0134122 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Kim J. H., Oh S.-H., Kim E.-J., et al. The role of myofibroblasts in upregulation of S100A8 and S100A9 and the differentiation of myeloid cells in the colorectal cancer microenvironment. Biochemical and Biophysical Research Communications. 2012;423(1):60–66. doi: 10.1016/j.bbrc.2012.05.081. [DOI] [PubMed] [Google Scholar]
- 147.Chen M. F., Kuan F.-C., Yen T.-C., et al. IL-6-stimulated CD11b+ CD14+ HLA-DR-myeloid-derived suppressor cells, are associated with progression and poor prognosis in squamous cell carcinoma of the esophagus. Oncotarget. 2014;5(18):8716–8728. doi: 10.18632/oncotarget.2368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Herrera M., Herrera A., Domínguez G., et al. Cancer-associated fibroblast and M2 macrophage markers together predict outcome in colorectal cancer patients. Cancer Science. 2013;104(4):437–444. doi: 10.1111/cas.12096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Zhang A., Qian Y., Ye Z., et al. Cancer-associated fibroblasts promote M2 polarization of macrophages in pancreatic ductal adenocarcinoma. Cancer Medicine. 2017;6:463–470. doi: 10.1002/cam4.993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Park J. Y., Sung J.-Y., Lee J., et al. Polarized CD163+ tumor-associated macrophages are associated with increased angiogenesis and CXCL12 expression in gastric cancer. Clinics and Research in Hepatology and Gastroenterology. 2016;40(3):357–365. doi: 10.1016/j.clinre.2015.09.005. [DOI] [PubMed] [Google Scholar]
- 151.Costa-Silva B., Aiello N. M., Ocean A. J., et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nature Cell Biology. 2015;17(6):816–826. doi: 10.1038/ncb3169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Nielsen S. R., Quaranta V., Linford A., et al. Macrophage-secreted granulin supports pancreatic cancer metastasis by inducing liver fibrosis. Nature Cell Biology. 2016;18(5):549–560. doi: 10.1038/ncb3340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Seubert B., Grünwald B., Kobuch J., 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(1):238–248. doi: 10.1002/hep.27378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Zhi K., Shen X., Zhang H., Bi J. Cancer-associated fibroblasts are positively correlated with metastatic potential of human gastric cancers. Journal of Experimental & Clinical Cancer Research. 2010;29(1):p. 66. doi: 10.1186/1756-9966-29-66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Ino Y., Yamazaki-Itoh R., Shimada K., et al. Immune cell infiltration as an indicator of the immune microenvironment of pancreatic cancer. British Journal of Cancer. 2013;108(4):914–923. doi: 10.1038/bjc.2013.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Nomi T., Sho M., Akahori T., et al. Clinical significance and therapeutic potential of the programmed death-1 ligand/programmed death-1 pathway in human pancreatic cancer. Clinical Cancer Research. 2007;13(7):2151–2157. doi: 10.1158/1078-0432.ccr-06-2746. [DOI] [PubMed] [Google Scholar]
- 157.Zhang L., Qiu M., Jin Y., et al. Programmed cell death ligand 1 (PD-L1) expression on gastric cancer and its relationship with clinicopathologic factors. International journal of clinical and experimental pathology. 2015;8(9):11084–11091. [PMC free article] [PubMed] [Google Scholar]
- 158.Wu C., Zhu Y., Jiang J., Zhao J., Zhang X.-G., Xu N. Immunohistochemical localization of programmed death-1 ligand-1 (PD-L1) in gastric carcinoma and its clinical significance. Acta Histochemica. 2006;108(1):19–24. doi: 10.1016/j.acthis.2006.01.003. [DOI] [PubMed] [Google Scholar]
- 159.Tamura T., Ohira M., Tanaka H., et al. Programmed death-1 ligand-1 (PDL1) expression is associated with the prognosis of patients with stage II/III gastric cancer. Anticancer Research. 2015;35(10):5369–5376. [PubMed] [Google Scholar]
- 160.Thompson E. D., Zahurak M., Murphy A., et al. Patterns of PD-L1 expression and CD8 T cell infiltration in gastric adenocarcinomas and associated immune stroma. Gut. 2017;66(5):794–801. doi: 10.1136/gutjnl-2015-310839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Chen L., Deng H., Lu M., et al. B7-H1 expression associates with tumor invasion and predicts patient’s survival in human esophageal cancer. International Journal of Clinical and Experimental Pathology. 2014;7(9):6015–6023. [PMC free article] [PubMed] [Google Scholar]
- 162.Kurahara H., Shinchi H., Mataki Y., et al. Significance of M2-polarized tumor-associated macrophage in pancreatic cancer. Journal of Surgical Research. 2011;167(2):e211–e219. doi: 10.1016/j.jss.2009.05.026. [DOI] [PubMed] [Google Scholar]
- 163.Shigeoka M., Urakawa N., Nakamura T., et al. Tumor associated macrophage expressing CD204 is associated with tumor aggressiveness of esophageal squamous cell carcinoma. Cancer Science. 2013;104(8):1112–1119. doi: 10.1111/cas.12188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Markowitz J., Brooks T. R., Duggan M. C., et al. Patients with pancreatic adenocarcinoma exhibit elevated levels of myeloid-derived suppressor cells upon progression of disease. Cancer Immunology, Immunotherapy. 2015;64(2):149–159. doi: 10.1007/s00262-014-1618-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Hiraoka N., Onozato K., Kosuge T., Hirohashi S. Prevalence of FOXP3+ regulatory T cells increases during the progression of pancreatic ductal adenocarcinoma and its premalignant lesions. Clinical Cancer Research. 2006;12(18):5423–5434. doi: 10.1158/1078-0432.ccr-06-0369. [DOI] [PubMed] [Google Scholar]
- 166.Ikemoto T., Yamaguchi T., Morine Y., et al. Clinical roles of increased populations of Foxp3+CD4+ T cells in peripheral blood from advanced pancreatic cancer patients. Pancreas. 2006;33(4):386–390. doi: 10.1097/01.mpa.0000240275.68279.13. [DOI] [PubMed] [Google Scholar]
- 167.Yamamoto T., Yanagimoto H., Satoi S., et al. Circulating CD4+CD25+ regulatory T cells in patients with pancreatic cancer. Pancreas. 2012;41(3):409–415. doi: 10.1097/MPA.0b013e3182373a66. [DOI] [PubMed] [Google Scholar]
- 168.Liu C., Cheng H., Luo G., et al. Circulating regulatory T cell subsets predict overall survival of patients with unresectable pancreatic cancer. International Journal of Oncology. 2017;51(2):686–694. doi: 10.3892/ijo.2017.4032. [DOI] [PubMed] [Google Scholar]
- 169.Sinicrope F. A., Rego R. L., Ansell S. M., Knutson K. L., Foster N. R., Sargent D. J. Intraepithelial effector (CD3+)/regulatory (FoxP3+) T-cell ratio predicts a clinical outcome of human colon carcinoma. Gastroenterology. 2009;137(4):1270–1279. doi: 10.1053/j.gastro.2009.06.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Salama P., Phillips M., Grieu F., et al. Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer. Journal of Clinical Oncology. 2009;27(2):186–192. doi: 10.1200/JCO.2008.18.7229. [DOI] [PubMed] [Google Scholar]
- 171.Correale P., Rotundo M. S., Del Vecchio M. T., et al. Regulatory (FoxP3+) T-cell tumor infiltration is a favorable prognostic factor in advanced colon cancer patients undergoing chemo or chemoimmunotherapy. Journal of Immunotherapy. 2010;33(4):435–441. doi: 10.1097/CJI.0b013e3181d32f01. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Frey D. M., Droeser R. A., Viehl C. T., et al. High frequency of tumor-infiltrating FOXP3+ regulatory T cells predicts improved survival in mismatch repair-proficient colorectal cancer patients. International Journal of Cancer. 2010;126:p. NA. doi: 10.1002/ijc.24989. [DOI] [PubMed] [Google Scholar]
- 173.Gao Q., Qiu S.-J., Fan J., et al. Intratumoral balance of regulatory and cytotoxic T cells is associated with prognosis of hepatocellular carcinoma after resection. Journal of Clinical Oncology. 2007;25(18):2586–2593. doi: 10.1200/jco.2006.09.4565. [DOI] [PubMed] [Google Scholar]
- 174.Shen Z., Zhou S., Wang Y., et al. Higher intratumoral infiltrated Foxp3+ Treg numbers and Foxp3+/CD8+ ratio are associated with adverse prognosis in resectable gastric cancer. Journal of Cancer Research and Clinical Oncology. 2010;136(10):1585–1595. doi: 10.1007/s00432-010-0816-9. [DOI] [PubMed] [Google Scholar]
- 175.Kono K., Kawaida H., Takahashi A., et al. CD4(+)CD25 high regulatory T cells increase with tumor stage in patients with gastric and esophageal cancers. Cancer Immunology, Immunotherapy. 2006;55(9):1064–1071. doi: 10.1007/s00262-005-0092-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Yamamoto T., Yanagimoto H., Satoi S., et al. Circulating myeloid dendritic cells as prognostic factors in patients with pancreatic cancer who have undergone surgical resection. Journal of Surgical Research. 2012;173(2):299–308. doi: 10.1016/j.jss.2010.09.027. [DOI] [PubMed] [Google Scholar]
- 177.Fukunaga A., Miyamoto M., Cho Y., et al. CD8+ tumor-infiltrating lymphocytes together with CD4+ tumor-infiltrating lymphocytes and dendritic cells improve the prognosis of patients with pancreatic adenocarcinoma. Pancreas. 2004;28(1):e26–e31. doi: 10.1097/00006676-200401000-00023. [DOI] [PubMed] [Google Scholar]
- 178.Cho Y., Miyamoto M., Kato K., et al. CD4+ and CD8+ T cells cooperate to improve prognosis of patients with esophageal squamous cell carcinoma. Cancer Research. 2003;63(7):1555–1559. [PubMed] [Google Scholar]
- 179.Pagès F., Berger A., Camus M., et al. Effector memory T cells, early metastasis, and survival in colorectal cancer. New England Journal of Medicine. 2005;353(25):2654–2666. doi: 10.1056/NEJMoa051424. [DOI] [PubMed] [Google Scholar]
- 180.Lee H. E., Chae S. W., Lee Y. J., et al. Prognostic implications of type and density of tumour-infiltrating lymphocytes in gastric cancer. British Journal of Cancer. 2008;99(10):1704–1711. doi: 10.1038/sj.bjc.6604738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Kim K.-J., Lee K. S., Cho H. J., et al. Prognostic implications of tumor-infiltrating FoxP3+ regulatory T cells and CD8+ cytotoxic T cells in microsatellite-unstable gastric cancers. Human Pathology. 2014;45(2):285–293. doi: 10.1016/j.humpath.2013.09.004. [DOI] [PubMed] [Google Scholar]
- 182.Kim K.-J., Wen X.-Y., Yang H. K., Kim W. H., Kang G. H. Prognostic implication of M2 macrophages are determined by the proportional balance of tumor associated macrophages and tumor infiltrating lymphocytes in microsatellite-unstable gastric carcinoma. PLoS One. 2015;10(12) doi: 10.1371/journal.pone.0144192.e0144192 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Ishigami S., Natsugoe S., Tokuda K., et al. Prognostic value of intratumoral natural killer cells in gastric carcinoma. Cancer. 2000;88(3):577–583. doi: 10.1002/(sici)1097-0142(20000201)88:3<577::aid-cncr13>3.0.co;2-v. [DOI] [PubMed] [Google Scholar]
- 184.Bozóky B., Savchenko A., Csermely P., et al. Novel signatures of cancer-associated fibroblasts. International Journal of Cancer. 2013;133(2):286–293. doi: 10.1002/ijc.28035. [DOI] [PubMed] [Google Scholar]
- 185.Chang Y., Li H., Guo Z. Mesenchymal stem cell-like properties in fibroblasts. Cellular Physiology and Biochemistry. 2014;34(3):703–714. doi: 10.1159/000363035. [DOI] [PubMed] [Google Scholar]
- 186.Strutz F. Identification and characterization of a fibroblast marker: FSP1. The Journal of Cell Biology. 1995;130(2):393–405. doi: 10.1083/jcb.130.2.393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Yamanashi T., Nakanishi Y., Fujii G., et al. Podoplanin expression identified in stromal fibroblasts as a favorable prognostic marker in patients with colorectal carcinoma. Oncology. 2009;77(1):53–62. doi: 10.1159/000226112. [DOI] [PubMed] [Google Scholar]
- 188.Choi S.-Y., Sung R., Lee S.-J., et al. Podoplanin, α-smooth muscle actin or S100A4 expressing cancer-associated fibroblasts are associated with different prognosis in colorectal cancers. Journal of Korean Medical Science. 2013;28(9):1293–1301. doi: 10.3346/jkms.2013.28.9.1293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Kitano H., Kageyama S., Hewitt S. M., et al. Podoplanin expression in cancerous stroma induces lymphangiogenesis and predicts lymphatic spread and patient survival. Archives of Pathology & Laboratory Medicine. 2010;134(10):1520–1527. doi: 10.1043/2009-0114-OA.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Shindo K., Aishima S., Ohuchida K., et al. Podoplanin expression in cancer-associated fibroblasts enhances tumor progression of invasive ductal carcinoma of the pancreas. Molecular Cancer. 2013;12(1):p. 168. doi: 10.1186/1476-4598-12-168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Schoppmann S. F., Jesch B., Riegler M. F., et al. Podoplanin expressing cancer associated fibroblasts are associated with unfavourable prognosis in adenocarcinoma of the esophagus. Clinical & Experimental Metastasis. 2013;30(4):441–446. doi: 10.1007/s10585-012-9549-2. [DOI] [PubMed] [Google Scholar]
- 192.Pula B., Jethon A., Piotrowska A., et al. Podoplanin expression by cancer-associated fibroblasts predicts poor outcome in invasive ductal breast carcinoma. Histopathology. 2011;59(6):1249–1260. doi: 10.1111/j.1365-2559.2011.04060.x. [DOI] [PubMed] [Google Scholar]
- 193.Özdemir B. C., Pentcheva-Hoang T., Carstens J. L., et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 2014;25(6):719–734. doi: 10.1016/j.ccr.2014.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Torres S., Bartolome R. A., Mendes M., et al. Proteome profiling of cancer-associated fibroblasts identifies novel proinflammatory signatures and prognostic markers for colorectal cancer. Clinical Cancer Research. 2013;19(21):6006–6019. doi: 10.1158/1078-0432.CCR-13-1130. [DOI] [PubMed] [Google Scholar]
- 195.Watanabe I., Hasebe T., Sasaki S., et al. Advanced pancreatic ductal cancer: fibrotic focus and β-catenin expression correlate with outcome. Pancreas. 2003;26(4):326–333. doi: 10.1097/00006676-200305000-00003. [DOI] [PubMed] [Google Scholar]
- 196.Zippi M., De Toma G., Minervini G., et al. Desmoplasia influenced recurrence of disease and mortality in stage III colorectal cancer within five years after surgery and adjuvant therapy. Saudi Journal of Gastroenterology. 2017;23(1):39–44. doi: 10.4103/1319-3767.199114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Garrido-Laguna I., Uson M., Rajeshkumar N. V., et al. Tumor engraftment in nude mice and enrichment in stroma- related gene pathways predict poor survival and resistance to gemcitabine in patients with pancreatic cancer. Clinical Cancer Research. 2011;17(17):5793–5800. doi: 10.1158/1078-0432.ccr-11-0341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Provenzano P. P., Cuevas C., Chang A. E., Goel V. K., Von Hoff D. D., Hingorani S. R. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell. 2012;21(3):418–429. doi: 10.1016/j.ccr.2012.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Jacobetz M. A., Chan D. S., Neesse A., et al. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut. 2013;62(1):112–120. doi: 10.1136/gutjnl-2012-302529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Olive K. P., Jacobetz M. A., Davidson C. J., et al. Inhibition of hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009;324(5933):1457–1461. doi: 10.1126/science.1171362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Rhim A. D., Oberstein P. E., Thomas D. H., et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 2014;25(6):735–747. doi: 10.1016/j.ccr.2014.04.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Lee J. J., Perera R. M., Wang H., et al. Stromal response to Hedgehog signaling restrains pancreatic cancer progression. Proceedings of the National Academy of Sciences. 2014;111(30):E3091–E3100. doi: 10.1073/pnas.1411679111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Öhlund D., Handly-Santana A., Biffi G., et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. The Journal of Experimental Medicine. 2017;214(3):p. 579. doi: 10.1084/jem.20162024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Peiris-Pagès M., Smith D. L., Győrffy B., Sotgia F., Lisanti M. P. Proteomic identification of prognostic tumour biomarkers, using chemotherapy-induced cancer-associated fibroblasts. Aging. 2015;7(10):816–838. doi: 10.18632/aging.100808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Bryant K. L., Stalnecker C. A., Zeitouni D., et al. Combination of ERK and autophagy inhibition as a treatment approach for pancreatic cancer. Nature Medicine. 2019;25(4):628–640. doi: 10.1038/s41591-019-0368-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Martinez-Outschoorn U. E., Trimmer C., Lin Z., et al. Autophagy in cancer associated fibroblasts promotes tumor cell survival. Cell Cycle. 2010;9(17):3515–3533. doi: 10.4161/cc.9.17.12928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Knudsen E. S., Balaji U., Freinkman E., McCue P., Witkiewicz A. K. Unique metabolic features of pancreatic cancer stroma: relevance to the tumor compartment, prognosis, and invasive potential. Oncotarget. 2016;7(48):78396–78411. doi: 10.18632/oncotarget.11893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Zheng X., Carstens J. L., Kim J., et al. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature. 2015;527(7579):525–530. doi: 10.1038/nature16064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Arora S., Bhardwaj A., Singh S., et al. An undesired effect of chemotherapy: gemcitabine promotes pancreatic cancer cell invasiveness through reactive oxygen species-dependent, nuclear factor κB- and hypoxia-inducible factor 1α-mediated up-regulation of CXCR4. Journal of Biological Chemistry. 2013;288(29):21197–21207. doi: 10.1074/jbc.M113.484576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Richards K. E., Zeleniak A. E., Fishel M. L., Wu J., Littlepage L. E., Hill R. Cancer-associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Oncogene. 2017;36(13):1770–1778. doi: 10.1038/onc.2016.353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Ren J., Ding L., Zhang D., et al. Carcinoma-associated fibroblasts promote the stemness and chemoresistance of colorectal cancer by transferring exosomal lncRNA H19. Theranostics. 2018;8(14):3932–3948. doi: 10.7150/thno.25541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Akao Y., Khoo F., Kumazaki M., Shinohara H., Miki K., Yamada N. Extracellular disposal of tumor-suppressor miRs-145 and -34a via microvesicles and 5-FU resistance of human colon cancer cells. International Journal of Molecular Sciences. 2014;15(1):1392–1401. doi: 10.3390/ijms15011392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Jiang C., Huang Y.-H., Lu J.-B., et al. Perivascular cell coverage of intratumoral vasculature is a predictor for bevacizumab efficacy in metastatic colorectal cancer. Cancer Management and Research. 2018;10:3589–3597. doi: 10.2147/cmar.s172261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Qiao Y., Zhang C., Li A., et al. IL6 derived from cancer-associated fibroblasts promotes chemoresistance via CXCR7 in esophageal squamous cell carcinoma. Oncogene. 2018;37(7):873–883. doi: 10.1038/onc.2017.387. [DOI] [PubMed] [Google Scholar]
- 215.Gonçalves-Ribeiro S., Díaz-Maroto N. G., Berdiel-Acer M., et al. Carcinoma-associated fibroblasts affect sensitivity to oxaliplatin and 5FU in colorectal cancer cells. Oncotarget. 2016;7(37):59766–59780. doi: 10.18632/oncotarget.11121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Zheng P., Chen L., Yuan X., et al. Exosomal transfer of tumor-associated macrophage-derived miR-21 confers cisplatin resistance in gastric cancer cells. Journal of Experimental & Clinical Cancer Research. 2017;36(1):p. 53. doi: 10.1186/s13046-017-0528-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Ham I.-H., Oh H. J., Jin H., et al. Targeting interleukin-6 as a strategy to overcome stroma-induced resistance to chemotherapy in gastric cancer. Molecular Cancer. 2019;18(1):p. 68. doi: 10.1186/s12943-019-0972-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Oh Y., Yao S., Hu Y., et al. The immune-microenvironment confers chemoresistance of colorectal cancer through macrophage-derived IL6. Clinical Cancer Research. 2017;23(23):7375–7387. doi: 10.1158/1078-0432.ccr-17-1283. [DOI] [PubMed] [Google Scholar]
- 219.Mitchem J. B., Brennan D. J., Knolhoff B. L., et al. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Research. 2013;73(3):1128–1141. doi: 10.1158/0008-5472.can-12-2731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Hu J. L., Wang W., Lan X. L., et al. CAFs secreted exosomes promote metastasis and chemotherapy resistance by enhancing cell stemness and epithelial-mesenchymal transition in colorectal cancer. Molecular Cancer. 2019;18(1):p. 91. doi: 10.1186/s12943-019-1019-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Pascucci L., Coccè V., Bonomi A., et al. Paclitaxel is incorporated by mesenchymal stromal cells and released in exosomes that inhibit in vitro tumor growth: a new approach for drug delivery. Journal of Controlled Release. 2014;192:262–270. doi: 10.1016/j.jconrel.2014.07.042. [DOI] [PubMed] [Google Scholar]
- 222.Li Z., Chan K., Qi Y., et al. Participation of CCL1 in snail-positive fibroblasts in colorectal cancer contribute to 5-fluorouracil/paclitaxel chemoresistance. Cancer Research and Treatment. 2018;50(3):894–907. doi: 10.4143/crt.2017.356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Namba T., Kodama R., Moritomo S., Hoshino T., Mizushima T. Zidovudine, an anti-viral drug, resensitizes gemcitabine-resistant pancreatic cancer cells to gemcitabine by inhibition of the Akt-GSK3β-snail pathway. Cell Death & Disease. 2015;6(6) doi: 10.1038/cddis.2015.172.e1795 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Scott A. M., Wiseman G., Welt S., et al. A Phase I dose-escalation study of sibrotuzumab in patients with advanced or metastatic fibroblast activation protein-positive cancer. Clinical Cancer Research. 2003;9(5):1639–1647. [PubMed] [Google Scholar]
- 225.Hofheinz R.-D., al-Batran S.-E., Hartmann F., et al. Stromal antigen targeting by a humanised monoclonal antibody: an early phase II trial of sibrotuzumab in patients with metastatic colorectal cancer. Oncology Research and Treatment. 2003;26(1):44–48. doi: 10.1159/000069863. [DOI] [PubMed] [Google Scholar]
- 226.Fong S. F. T., Dietzsch E., Fong K. S. K., et al. Lysyl oxidase-like 2 expression is increased in colon and esophageal tumors and associated with less differentiated colon tumors. Genes, Chromosomes and Cancer. 2007;46(7):644–655. doi: 10.1002/gcc.20444. [DOI] [PubMed] [Google Scholar]
- 227.Barker H. E., Cox T. R., Erler J. T. The rationale for targeting the LOX family in cancer. Nature Reviews Cancer. 2012;12(8):540–552. doi: 10.1038/nrc3319. [DOI] [PubMed] [Google Scholar]
- 228.Benson A. B., Wainberg Z. A., Hecht J. R., et al. A phase II randomized, double-blind, placebo-controlled study of simtuzumab or placebo in combination with gemcitabine for the first-line treatment of pancreatic adenocarcinoma. The Oncologist. 2017;22(3):241–e15. doi: 10.1634/theoncologist.2017-0024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Hecht J. R., Benson A. B., Vyushkov D., Yang Y., Bendell J., Verma U. A phase II, randomized, double-blind, placebo-controlled study of simtuzumab in combination with FOLFIRI for the second-line treatment of metastatic KRAS mutant colorectal adenocarcinoma. The Oncologist. 2017;22(3):243–e23. doi: 10.1634/theoncologist.2016-0479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Chauhan V. P., Martin J. D., Liu H., et al. Angiotensin inhibition enhances drug delivery and potentiates chemotherapy by decompressing tumour blood vessels. Nature Communications. 2013;4(1):p. 2516. doi: 10.1038/ncomms3516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Busby J., McMenamin Ú., Spence A., Johnston B. T., Hughes C., Cardwell C. R. Angiotensin receptor blocker use and gastro-oesophageal cancer survival: a population-based cohort study. Alimentary Pharmacology & Therapeutics. 2018;47(2):279–288. doi: 10.1111/apt.14388. [DOI] [PubMed] [Google Scholar]
- 232.Hingorani S. R., Harris W. P., Beck J. T., et al. Phase IB study of PEGylated recombinant human hyaluronidase and gemcitabine in patients with advanced pancreatic cancer. Clinical Cancer Research. 2016;22(12):2848–2854. doi: 10.1158/1078-0432.ccr-15-2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Hingorani S. R., Zheng L., Bullock A. J., et al. HALO 202: randomized phase II study of PEGPH20 plus nab-paclitaxel/gemcitabine versus nab-paclitaxel/gemcitabine in patients with untreated, metastatic pancreatic ductal adenocarcinoma. Journal of Clinical Oncology. 2018;36(4):359–366. doi: 10.1200/jco.2017.74.9564. [DOI] [PubMed] [Google Scholar]
- 234.Eton O., Legha S. S., Bedikian A. Y., et al. Sequential biochemotherapy versus chemotherapy for metastatic melanoma: results from a phase III randomized trial. Journal of Clinical Oncology. 2002;20(8):2045–2052. doi: 10.1200/JCO.2002.07.044. [DOI] [PubMed] [Google Scholar]
- 235.Hurwitz H., Van Cutsem E., Bendell J., et al. Ruxolitinib + capecitabine in advanced/metastatic pancreatic cancer after disease progression/intolerance to first-line therapy: JANUS 1 and 2 randomized phase III studies. Investigational New Drugs. 2018;36(4):683–695. doi: 10.1007/s10637-018-0580-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Ohtsu A., Shah M. A., Van Cutsem E., et al. Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a randomized, double-blind, placebo-controlled phase III study. Journal of Clinical Oncology. 2011;29(30):3968–3976. doi: 10.1200/jco.2011.36.2236. [DOI] [PubMed] [Google Scholar]
- 237.Bailey J. M., Swanson B. J., Hamada T., et al. Sonic hedgehog promotes desmoplasia in pancreatic cancer. Clinical Cancer Research. 2008;14(19):5995–6004. doi: 10.1158/1078-0432.ccr-08-0291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Ramanathan R. K., McDonough S. L., Philip P. A., et al. Phase Ib/II randomized study of FOLFIRINOX plus pegylated recombinant human hyaluronidase versus FOLFIRINOX alone in patients with metastatic pancreatic adenocarcinoma: SWOG S1313. Journal of Clinical Oncology. 2019;37(13):1062–1069. doi: 10.1200/jco.18.01295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Santos A. M., Jung J., Aziz N., Kissil J. L., Puré E. Targeting fibroblast activation protein inhibits tumor stromagenesis and growth in mice. Journal of Clinical Investigation. 2009;119(12):3613–3625. doi: 10.1172/jci38988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Ostermann E., Garin-Chesa P., Heider K. H., et al. Effective immunoconjugate therapy in cancer models targeting a serine protease of tumor fibroblasts. Clinical Cancer Research. 2008;14(14):4584–4592. doi: 10.1158/1078-0432.ccr-07-5211. [DOI] [PubMed] [Google Scholar]
- 241.Lo A., Wang L.-C. S., Scholler J., et al. Tumor-promoting desmoplasia is disrupted by depleting FAP-expressing stromal cells. Cancer Research. 2015;75(14):2800–2810. doi: 10.1158/0008-5472.can-14-3041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Duperret E. K., Trautz A., Ammons D., et al. Alteration of the tumor stroma using a consensus DNA vaccine targeting fibroblast activation protein (FAP) synergizes with antitumor vaccine therapy in mice. Clinical Cancer Research. 2018;24(5):1190–1201. doi: 10.1158/1078-0432.ccr-17-2033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Wang Y., Wang H., Yao H., Li C., Fang J.-Y., Xu J. Regulation of PD-L1: emerging routes for targeting tumor immune evasion. Frontiers in Pharmacology. 2018;9:p. 536. doi: 10.3389/fphar.2018.00536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Kang Y.-K., Boku N., Satoh T., et al. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomised, double-blind, placebo-controlled, phase 3 trial. The Lancet. 2017;390(10111):2461–2471. doi: 10.1016/s0140-6736(17)31827-5. [DOI] [PubMed] [Google Scholar]
- 245.Fuchs C. S., Doi T., Jang R. W., et al. Safety and efficacy of pembrolizumab monotherapy in patients with previously treated advanced gastric and gastroesophageal junction cancer. JAMA Oncology. 2018;4(5) doi: 10.1001/jamaoncol.2018.0013.e180013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Yun S., Vincelette N. D., Green M. R., Wahner Hendrickson A. E., Abraham I. Targeting immune checkpoints in unresectable metastatic cutaneous melanoma: a systematic review and meta-analysis of anti-CTLA-4 and anti-PD-1 agents trials. Cancer Medicine. 2016;5(7):1481–1491. doi: 10.1002/cam4.732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Royal R. E., Levy C., Turner K., et al. Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. Journal of Immunotherapy. 2010;33(8):828–833. doi: 10.1097/cji.0b013e3181eec14c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Brahmer J. R., Tykodi S. S., Chow L. Q. M., et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. New England Journal of Medicine. 2012;366(26):2455–2465. doi: 10.1056/NEJMoa1200694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Le D. T., Durham J. N., Smith K. N., et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357(6349):409–413. doi: 10.1126/science.aan6733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.El-Khoueiry A. B., Sangro B., Yau T., et al. Nivolumab in patients with advanced hepatocellular carcinoma (checkmate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial. The Lancet. 2017;389(10088):2492–2502. doi: 10.1016/s0140-6736(17)31046-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Zhu A. X. Pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib (KEYNOTE-224): a non-randomised, open-label phase 2 trial. The Lancet Oncology. 2018;19(7):940–952. doi: 10.1016/s1470-2045(18)30351-6. [DOI] [PubMed] [Google Scholar]
- 252.Kenilworth N. J. Merck Provides Update on KEYNOTE-240, a Phase 3 Study of KEYTRUDA® (Pembrolizumab) in Previously Treated Patients with Advanced Hepatocellular Carcinoma. Kenilworth, NJ, USA: Merck & Co., Inc.; 2019. [Google Scholar]
- 253.Chen Y., Ramjiawan R. R., Reiberger T., et al. CXCR4 inhibition in tumor microenvironment facilitates anti-programmed death receptor-1 immunotherapy in sorafenib-treated hepatocellular carcinoma in mice. Hepatology. 2015;61(5):1591–1602. doi: 10.1002/hep.27665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Seo Y. D., Jiang X., Sullivan K. M., et al. Mobilization of CD8+ T cells via CXCR4 blockade facilitates PD-1 checkpoint therapy in human pancreatic cancer. Clinical Cancer Research. 2019;25(1):3934–3945. doi: 10.1158/1078-0432.ccr-19-0081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Naito Y., Saito K., Shiiba K., et al. CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Research. 1998;58(16):3491–3494. [PubMed] [Google Scholar]
- 256.Bossard C., Bézieau S., Matysiak-Budnik T., et al. HLA-E/β2 microglobulin overexpression in colorectal cancer is associated with recruitment of inhibitory immune cells and tumor progression. International Journal of Cancer. 2012;131(4):855–863. doi: 10.1002/ijc.26453. [DOI] [PubMed] [Google Scholar]
- 257.Lunavat T. R., Cheng L., Einarsdottir B. O., et al. BRAFV600 inhibition alters the microRNA cargo in the vesicular secretome of malignant melanoma cells. Proceedings of the National Academy of Sciences. 2017;114(29):E5930–E5939. doi: 10.1073/pnas.1705206114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Havel J. J., Chowell D., Chan T. A. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nature Reviews Cancer. 2019;19(3):133–150. doi: 10.1038/s41568-019-0116-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Duan X., Chan C., Han W., Guo N., Weichselbaum R. R., Lin W. Immunostimulatory nanomedicines synergize with checkpoint blockade immunotherapy to eradicate colorectal tumors. Nature Communications. 2019;10(1):p. 1899. doi: 10.1038/s41467-019-09221-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Michaloglou C., Vredeveld L. C. W., Soengas M. S., et al. BRAFE600-associated senescence-like cell cycle arrest of human naevi. Nature. 2005;436(7051):720–724. doi: 10.1038/nature03890. [DOI] [PubMed] [Google Scholar]
- 261.Nywening T. M., Belt B. A., Cullinan D. R., et al. Targeting both tumour-associated CXCR2+ neutrophils and CCR2+ macrophages disrupts myeloid recruitment and improves chemotherapeutic responses in pancreatic ductal adenocarcinoma. Gut. 2018;67(6):1112–1123. doi: 10.1136/gutjnl-2017-313738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Lascorz J., Bevier M., Schönfels W. V., et al. Association study identifying polymorphisms in CD47 and other extracellular matrix pathway genes as putative prognostic markers for colorectal cancer. International Journal of Colorectal Disease. 2013;28(2):173–181. doi: 10.1007/s00384-012-1541-4. [DOI] [PubMed] [Google Scholar]
- 263.Weiskopf K. Cancer immunotherapy targeting the CD47/SIRPα axis. European Journal of Cancer. 2017;76:100–109. doi: 10.1016/j.ejca.2017.02.013. [DOI] [PubMed] [Google Scholar]
- 264.Sikic B. I., Lakhani N. J., Patnaik A., et al. A first-in-class, first-in-human phase 1 pharmacokinetic (PK) and pharmacodynamic (PD) study of Hu5F9-G4, an anti-CD47 monoclonal antibody (mAb), in patients with advanced solid tumors. Journal of Clinical Oncology. 2018;36(15_suppl):p. 3002. doi: 10.1200/JCO.2018.36.15_suppl.3002. [DOI] [Google Scholar]
- 265.Murata Y., Saito Y., Kotani T., Matozaki T. CD47-signal regulatory protein α signaling system and its application to cancer immunotherapy. Cancer Science. 2018;109(8):2349–2357. doi: 10.1111/cas.13663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Pyonteck S. M., Gadea B. B., Wang H.-W., et al. Deficiency of the macrophage growth factor CSF-1 disrupts pancreatic neuroendocrine tumor development. Oncogene. 2012;31(11):1459–1467. doi: 10.1038/onc.2011.337. [DOI] [PubMed] [Google Scholar]
- 267.Zhu Y., Knolhoff B. L., Meyer M. A., et al. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Research. 2014;74(18):5057–5069. doi: 10.1158/0008-5472.can-13-3723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Fuchs C. S., Shitara K., Bartolomeo M. D., et al. Ramucirumab with cisplatin and fluoropyrimidine as first-line therapy in patients with metastatic gastric or junctional adenocarcinoma (RAINFALL): a double-blind, randomised, placebo-controlled, phase 3 trial. The Lancet Oncology. 2019;20(3):420–435. doi: 10.1016/s1470-2045(18)30791-5. [DOI] [PubMed] [Google Scholar]
- 269.Fuchs C. S., Tomasek T., Yong C. J., et al. Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): an international, randomised, multicentre, placebo-controlled, phase 3 trial. The Lancet. 2014;383(9911):31–39. doi: 10.1016/s0140-6736(13)61719-5. [DOI] [PubMed] [Google Scholar]
- 270.Llovet J. M., Ricci S., Mazzaferro V., et al. Sorafenib in advanced hepatocellular carcinoma. New England Journal of Medicine. 2008;359(4):378–390. doi: 10.1056/NEJMoa0708857. [DOI] [PubMed] [Google Scholar]
- 271.Bruix J., Qin S., Merle P., et al. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): a randomised, double-blind, placebo-controlled, phase 3 trial. The Lancet. 2017;389(10064):56–66. doi: 10.1016/s0140-6736(16)32453-9. [DOI] [PubMed] [Google Scholar]
- 272.Abou-Alfa G. K., Meyer T., Cheng A.-L., et al. Cabozantinib in patients with advanced and progressing hepatocellular carcinoma. New England Journal of Medicine. 2018;379(1):54–63. doi: 10.1056/NEJMoa1717002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273.Rosen L. S., Jacobs I. A., Burkes R. L. Bevacizumab in colorectal cancer: current role in treatment and the potential of biosimilars. Targeted Oncology. 2017;12(5):599–610. doi: 10.1007/s11523-017-0518-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274.Kerr R. S., Love S., Segelov E., et al. Adjuvant capecitabine plus bevacizumab versus capecitabine alone in patients with colorectal cancer (QUASAR 2): an open-label, randomised phase 3 trial. The Lancet Oncology. 2016;17(11):1543–1557. doi: 10.1016/s1470-2045(16)30172-3. [DOI] [PubMed] [Google Scholar]
- 275.Van Cutsem E., Joulain F., Hoff P. M., et al. Aflibercept plus FOLFIRI vs. placebo plus FOLFIRI in second-line metastatic colorectal cancer: a post hoc analysis of survival from the phase III VELOUR study subsequent to exclusion of patients who had recurrence during or within 6 months of completing adjuvant oxaliplatin-based therapy. Targeted Oncology. 2016;11(3):383–400. doi: 10.1007/s11523-015-0402-9. [DOI] [PubMed] [Google Scholar]
- 276.Tabernero J., Yoshino T., Cohn A. L., et al. Ramucirumab versus placebo in combination with second-line FOLFIRI in patients with metastatic colorectal carcinoma that progressed during or after first-line therapy with bevacizumab, oxaliplatin, and a fluoropyrimidine (RAISE): a randomised, double-blind, multicentre, phase 3 study. The Lancet Oncology. 2015;16(5):499–508. doi: 10.1016/s1470-2045(15)70127-0. [DOI] [PubMed] [Google Scholar]