Next Article in Journal
The Impact of Post-Mastectomy Radiotherapy on Survival Outcomes in Breast Cancer Patients Who Underwent Neoadjuvant Chemotherapy
Next Article in Special Issue
The Complex Network between Inflammation and Colorectal Cancer: A Systematic Review of the Literature
Previous Article in Journal
Heavy Ion Minibeam Therapy: Side Effects in Normal Brain
Previous Article in Special Issue
Oxidative Distress Induces Wnt/β-Catenin Pathway Modulation in Colorectal Cancer Cells: Perspectives on APC Retained Functions
 
 
Comment published on 31 August 2022, see Cancers 2022, 14(17), 4235.
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Colorectal Cancer Tumor Microenvironment and Its Impact on Liver and Lung Metastasis

1
Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
2
Hamon Center for Therapeutic Oncology Research, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
3
School of Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
4
Department of Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
5
Division of Colon & Rectal Surgery, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
*
Author to whom correspondence should be addressed.
Submission received: 3 November 2021 / Revised: 2 December 2021 / Accepted: 2 December 2021 / Published: 9 December 2021
(This article belongs to the Special Issue Molecular Aspects of Tumor Microenvironment of Colorectal Cancers)

Abstract

:

Simple Summary

Colorectal cancer (CRC) is the third most common cancer worldwide. Metastasis to secondary organs, such as the liver and lungs, is a key driver of CRC-related mortality. The tumor microenvironment, which consists of the primary cancer cells, as well as associated support and immune cells, significantly affects the behavior of CRC cells at the primary tumor site, as well as in metastatic lesions. In this paper, we review the role of the individual components of the tumor microenvironment on tumor progression, immune evasion, and metastasis, and we discuss the implications of these components on antitumor therapies.

Abstract

Colorectal cancer (CRC) is the third most common malignancy and the second most common cause of cancer-related mortality worldwide. A total of 20% of CRC patients present with distant metastases, most frequently to the liver and lung. In the primary tumor, as well as at each metastatic site, the cellular components of the tumor microenvironment (TME) contribute to tumor engraftment and metastasis. These include immune cells (macrophages, neutrophils, T lymphocytes, and dendritic cells) and stromal cells (cancer-associated fibroblasts and endothelial cells). In this review, we highlight how the TME influences tumor progression and invasion at the primary site and its function in fostering metastatic niches in the liver and lungs. We also discuss emerging clinical strategies to target the CRC TME.

1. Introduction

Colorectal cancer (CRC) is the third most common malignancy and the second leading cause of cancer-related mortality worldwide [1]. Metastasis is a primary driver of CRC-related mortality, with the liver and lungs representing the most frequently involved organs. While surgical resection of colorectal liver metastases (CRLMs) and colorectal pulmonary metastases (CRPMs) provides the only potentially curative treatment, select patients with unresectable metastatic CRC (mCRC) may benefit from other locoregional therapies, including radiofrequency ablation and stereotactic radiotherapy [2,3]. However, for most patients, systemic chemotherapy is the cornerstone of treatment. Recently, immune checkpoint inhibitors (ICIs) have been shown to have a significant clinical benefit in CRC that has high microsatellite instability or mismatch repair deficiencies [4]. This advance reflects the potential of tumor microenvironment (TME)-directed therapies for the treatment of mCRC. Here, we review the functions of the individual components of the TME in CRC, highlight the importance of the TME during the engraftment and growth of hepatic and pulmonary metastases, and discuss potential therapeutic implications of the TME in mCRC.

2. Limitations of Conventional Approaches for mCRC

Pulmonary metastasectomy and hepatic metastasectomy are the widely accepted first-line courses of management and the only cure for CRPM and CRLM, respectively [5,6,7,8]. The 2021 National Comprehensive Cancer Network guidelines recommend the resection of CRPMs in patients who have undergone curative (R0) resection of the primary tumor, have adequate cardiopulmonary reserve, have technically resectable disease, and have no extrathoracic metastases (with the exception of resectable CRLM) [5]. Similarly, for CRLM, those patients for whom an R0 margin is achievable with an adequate future functional liver remnant (20% in chemotherapy-naïve, noncirrhotic livers, and 40% for cirrhotic or post-treatment livers) are candidates for resection with curative intent [5,9]. A meta-analysis of 21 studies with 8361 patients demonstrated 5-year overall survival ranging between 24 and 82% after pulmonary metastasectomy [10]. Large-scale randomized controlled trials assessing the efficacy of pulmonary metastasectomy are lacking, and the only phase III randomized controlled trial (PulMiCC) comparing pulmonary metastasectomy versus active clinical monitoring for CRPM was stopped early due to poor recruitment [11]. A recent population-based Surveillance, Epidemiology and End Results Program study of 10,325 patients demonstrated no improvement in cancer-specific survival with pulmonary metastasectomy alone or in combination with hepatic metastasectomy on multivariable analysis [12].
Survival in CRLM is driven by the ability to completely extirpate hepatic disease, as positive margins incur an overall recurrence rate of 52% compared to 39% with R0 margins. Choti et al., Figueras et al., and Abdalla et al. [13,14,15] each demonstrated survival rates for CRLM patients approaching 60%, higher than earlier studies, despite more liberal indications for surgery. Unfortunately, up to 90% of mCRC patients present with unresectable disease, including up to 50% of patients undergoing metastasectomy [15,16,17,18]. Surgical trauma itself may confer a risk of recurrence. In lung resections, trauma to the nearby lung parenchyma has been associated with increased infiltration of myeloid cells and regulatory T cells into the TME, which may suppress CD8+ T-cell recruitment to the resection bed and increase the risk of recurrence [19]. Furthermore, the extent of resection and the magnitude of subsequent local tissue injury may be correlated to the immunosuppressive M2 polarization of local macrophages; a study of resection versus biopsy of oral squamous cell carcinomas in animal models demonstrated that resected lesions had higher levels of alternatively activated macrophages compared to biopsied lesions [20]. Thus, greater local trauma may be correlated with a favorable TME for tumor progression.
Chemotherapy alone has not been shown to be effective for CRLM or CRPM. A retrospective series of 2541 patients with mCRC demonstrated a 5-year overall survival of 10.8% for this approach. Of long-term survivors, only 2.2% (0.24% of the overall patient population) achieved a complete response to chemotherapy alone [21]. This underlies the importance of neoadjuvant and adjuvant systemic therapies, as well as continued research into novel therapeutics [17,22]. These mixed findings reflect the critical need to further investigate the role of the CRC TME on CRPM and CRLM pathogenesis and therapy.

3. Colorectal Liver Metastasis (CRLM)

The liver is the most common site of CRC metastases [23,24]. Up to 25% CRC patients may have synchronous CRLM, with an additional 50% developing CLRM during their disease course [25]. Oncogenotype may predict CRLM; Chu et al. [26] demonstrated that mutant KRAS+ CRC (found in 30–40% of cases) is associated with CRLM through the upregulation of YB-1 and insulin-like growth factor-1 receptor via the MEK-miR137 signaling pathway. Consistently, Brudvik et al. [27] identified that mutant KRAS status was inversely associated with overall and recurrence-free survival after CRLM resection in a meta-analysis of 1809 patients. The liver microenvironment represents a unique metastatic niche, and there may be differences in the immune landscape between liver metastases, the primary tumor, and other metastatic sites. Indeed, Tian et al. [28] demonstrated that CRC cells cultured on decellularized liver and lung scaffolds formed spheroid “metastases” in vitro and exhibited organ-specific tropism when injected into murine-models. Wei et al. showed in a 74-patient cohort that programmed death-ligand 1 (PD-L1) expression, a potential biomarker for ICI efficacy, and CD4 T-cell density were higher in CRLMs than the primary tumor [29]. An improved understanding of the CRLM TME, therefore, has implications on treatment strategy and may justify biopsy excisional biopsy of a CRLM to analyze both the metastasis and its microenvironment.

3.1. Pathophysiology of Colorectal Liver Metastasis

CRCs are thought to seed the liver primarily through the portal venous circulation. In contrast, cancer cells originating from outside of the gastrointestinal tract likely seed through the systemic circulation via the hepatic artery. These two sources of hepatic blood flow join at the point of entry into the liver sinusoids, where blood perfuses the liver parenchyma and returns to the systemic circulation via the centrilobular veins. Once circulating CRC tumor cells enter the liver sinusoids, the seeding of metastasis to the liver occurs in four dynamic, overlapping phases (Figure 1) [30,31,32].

3.1.1. Microvascular Phase

Once in the portal vascular system, CRC circulating tumor cells pass through the portal venules into the sinusoidal capillaries. There, the cells encounter liver-specific defense mechanisms, and many are phagocytosed by stellate macrophages (Kupffer cells (KCs)), a process potentiated by natural killer cells (NKs) [33,34]. However, cells that escape innate immune surveillance must stop in the sinusoids to extravasate and form a metastatic lesion. While this mechanical arrest contributes to the adhesion of circulating tumor cells to the sinusoidal vasculature, intravital microscopy has shown that CRC circulating cells are usually smaller than the vessel diameter [35,36]. This suggests that specific cell-adhesion interactions can facilitate tumor cell arrest.
The liver sinusoids are lined by liver sinusoidal endothelial cells (LSECs), which express the critical cell-adhesion protein, E-selectin. E-selectin is a cytokine-inducible protein implicated in CRC cell adhesion to liver sinusoidal endothelial cells [37]. Notably, the inhibition of endothelial E-selectin has been shown to reduce the number of CRLMs up to 97% relative to the placebo in syngeneic murine models [37,38]. Furthermore, Khatib et al. [39,40] showed that CRC circulating cells trigger the release of proinflammatory TNF-α and IL-1β from KCs, thereby upregulating the expression of E-selectin and other adhesion molecules on liver sinusoidal endothelial cells and enhancing liver metastases [39,40].

3.1.2. Extravascular/Pre-Angiogenic Phase

Once tumor cells have extravasated from the sinusoidal microvasculature into the space of Disse, hepatic stellate cells (HSCs) are activated by the pro-inflammatory cascade that was initiated in the microvascular phase. HSCs produce extracellular matrix (ECM) proteins, including collagen, laminin, fibrillin, and fibronectin, and provide a scaffold for tumor cells [41,42]. Additionally, KCs and neutrophils secrete matrix metalloproteinases (MMPs) and elastase, which degrade and remodel the ECM, facilitating tumor cell invasion [31,43]. Furthermore, cells in the TME have been implicated in the induction of an immunosuppressive niche, facilitating tumor cell growth. Huang et al. [44] demonstrated that HSCs co-cultured with T cells and CRC antigen-pulsed dendritic cells (DCs) significantly abrogated T-cell responsiveness and induced the expansion of immunosuppressive regulatory T cells (Tregs). In addition, portal injections of HSCs with tumor cells significantly increased CRLM compared to tumor cells alone [44].

3.1.3. Angiogenic Phase

CRLMs are predominantly supplied by hepatic arteries [45]. The vascularization of liver metastases can occur through vascular endothelial growth factor (VEGF)-mediated angiogenesis or by the “co-opting” of existing vasculature. Importantly, a phase III trial by Hurwitz et al. [46] showed that the addition of bevacizumab, an anti-VEGF agent, to standard chemotherapy improved the median survival of patients with mCRC; however, the majority of patients did not exhibit a response to therapy [46]. This suggests the importance of other mechanisms of tumor vascularization independent of VEGF in some CRCs [42,47]. Notably, Frentzas et al. [48] described the co-opting of vessels in CRLM, a process by which cancer cells invade along the existing liver microvasculature, replacing hepatocytes and occupying the space adjacent to sinusoidal vessels. This commonly occurs in tumors that display a replacement growth pattern (see Section 3.1.4. below below) and is associated with resistance to anti-VEGF therapy. Importantly, the inhibition of actin-related protein 2/3 (Arp2/3), which is associated with the co-opting of vessels, was found to reduce vascular density in combination bevacizumab significantly more than bevacizumab by itself. Combination therapy also potently inhibited the growth of CRLMs with reduction in lesion area by approximately 75% compared to controls [48].

3.1.4. Growth Phase

The growth of vascularized micrometastases leads to tumor cell proliferation and the establishment of clinically detectable metastases. Three histologic growth patterns have been described by Eefsen et al. [25] in a series of 24 patients with resected CRLMs: desmoplastic, pushing, and replacement. In the desmoplastic pattern, a band of fibrotic tissue separates tumor cells from the surrounding liver parenchyma. In the pushing pattern, liver cells are pushed aside by the growing metastatic lesion and assume a flattened morphology with no fibrotic band separating the invasive tumor front from the hepatic parenchyma. The replacement pattern describes tumor cell infiltration into the existing liver parenchyma, such that it supplants the parenchyma instead of simply pushing it away. In 83% of patients, identical growth patterns were identified in all metastases [25]. This classification is notable because the dominant replacement growth pattern is associated with a worse prognosis [49]. These differences indicate that different interactions of tumor cells, TME cells, and cells of the hepatic parenchyma lead to varying CRLM phenotypes.

4. Colorectal Lung Metastasis (CRPM)

The lungs are the second most common site of CRC metastasis. Studies suggest that 10–18% of rectal cancers and 5–6% of colon cancers metastasize to the lungs [50]. According to population-based studies, the incidence of synchronous CRPMs has been estimated to be up to 11%, and has increased over time [50,51,52]. Similar to CRLM, specific oncogenes are implicated in CRPM. Zhang et al. [53] demonstrated through extensive genomic profiling studies of clinical samples from five CRC patients that CRPM shared clonality with primary tumors. Furthermore, 14 of 27 mutated genes identified in CRPM (i.e., KRAS, APC, and TP53) were also found in the primary lesion, thus suggesting that these mutations likely existed in the primary lesion itself. As such, the identification of these mutations in the primary tumor during diagnostic workup or on post-resection pathologic analysis may be prognostically significant [53]. While surgical resection for isolated CRPMs is the only curative option for select patients, there are no high-quality clinical-trial data to guide practice. For most patients, systemic therapy is the cornerstone of treatment. Consistent with this, in their decellularized liver and lung scaffolding models, Tian et al. [28] found that CRPM-model cells were significantly more sensitive to chemotherapy compared to CRLM-model cells, suggesting that unique microenvironment features affect therapeutic response. As such, an improved understanding of the TME of CRPM may identify new therapeutic targets.

Pathophysiology of Colorectal Metastasis to the Lungs

CRCs metastasize to the lungs through different mechanisms. Lymphatic vessels from the colon and proximal rectum drain into the inferior mesenteric nodes, which follows venous drainage into the portal circulation. Outflow from the distal rectum through the middle and inferior rectal veins drains into the internal iliac veins and directly into systemic circulation, thereby bypassing the portal system. Distal rectal lymphatics follow a similar course into the internal iliac nodes. As such, distal rectal tumors are more likely to present with early CRPM due to direct systemic drainage, as opposed to proximal tumors that must first filter through the liver [51]. Additionally, lymphatic vessels drain into the cisterna chyli and ultimately into the left subclavian vein through the thoracic duct, which may also lead to the development of CRPM [54].
The microenvironment of the lung parenchyma is enriched with a diverse array of immune cells that reside along the airway, including alveolar macrophages (dust cells), lymphocytes, and DCs, which are crucial for defense against airborne pathogens, toxins, and inflammatory substances [55,56]. Chronic inflammatory conditions, such as cigarette smoking and chronic obstructive pulmonary disease, alter the microenvironment in a way that lends itself to primary tumor development and the establishment of a pre-metastatic niche [57,58,59]. Indeed, current smoking status was found to be an independent risk factor for CRPM in a multivariable analysis of outcomes from 567 CRC patients [60].
Clearly, CRPMs (as well as other metastatic sites) represent an escape from immune surveillance. Thus, it is of great interest that a key feature of lung metastases is a tendency toward having a more immune responsive TME relative to metastases in other organs, such as the brain, liver, or bone [61]. García-Mulero et al. [61] analyzed resected tumor tissue and found that lung metastases from various primary tumors, including CRC, were enriched for genes associated with antigen presentation and immune effector cells, such as cytotoxic T lymphocytes (CTLs) and B-lineage cells. Moreover, they observed a low density of suppressor cells [61]. Lung metastases had a higher infiltration of CTLs, B-lineage cells, and DCs, but also expressed high levels of PD-L1 and cytotoxic T-lymphocyte–associated protein 4 (CTLA-4), which could partly explain their ability to escape immune surveillance [61]. However, infiltration by neutrophils, NK cells, and myeloid lineage cells was similar to other metastatic sites [61]. The overall high immunogenicity of the lung metastatic TME, as well as the elevated expression of PD-L1 and CTLA-4 in these lesions, may confer increased sensitivity to ICIs.
Altkori et al. [56] described a general mechanism for the formation of lung metastasis from extra-thoracic tumors. First, primary tumors secrete extracellular vesicles and pro-metastatic factors, including TGF-β, VEGF, and others that remodel the ECM, promote epithelial–mesenchymal transition (EMT), and facilitate invasion into the systemic circulation. These factors also promote recruitment of bone-marrow-derived cells into the microenvironment. Circulating tumor cells then extravasate into the pulmonary tissue. Type II alveolar cells recruit neutrophils, which suppress CTL activity and work with fibroblasts to facilitate seeding of tumor cells into the lung parenchyma. In addition, macrophages in the metastatic niche promote tumor cell survival and proliferation. Altogether, the established metastatic niche sustains tumor cell growth, promotes mesenchymal–epithelial transition, and stimulates angiogenesis [56].

5. The Tumor Microenvironment in CRC

To strengthen our understanding of the pathophysiology of CRLM and CRPM, it is crucial to first investigate in detail the function of individual components of the TME in CRC and their contribution to primary tumor invasiveness and subsequent metastasis to the liver and lung. The TME is the environment surrounding tumor cells and includes a complex array of immune cells, fibroblasts, endothelial cells, and stromal proteins. It is implicated in virtually every aspect of tumor progression and the metastatic cascade, including initial engraftment, growth, and immune evasion. Here, we review the function of selected cell types of the CRC TME (Figure 2).

5.1. Immune Cells

5.1.1. Macrophages

Tumor-associated macrophages (TAMs) have historically been dichotomized as “M1-like” macrophages with pro-inflammatory, immunostimulatory, and anti-tumorigenic properties or “M2-like” macrophages with immunosuppressive and pro-tumorigenic properties [62]. In reality, TAMs exist on a spectrum between these two states; however, this traditional classification provides a useful framework for discussion. M1 macrophages produce pro-inflammatory immunostimulatory cytokines, including IL-1β, IL-6, and TNF-α [62,63]. Although the M1 phenotype is generally thought to be anti-tumorigenic, this notion is context-specific. For example, M1 TAMs can promote chronic inflammation in induced colitis models, and this is a risk factor for the development of CRC [64]. In a colitis-associated CRC model, Wang et al. [65] demonstrated that the densities of M1 and M2 macrophages vary throughout the inflammation–carcinoma sequence, but overall macrophage count increased as tumors became metastatic. Exposure of CRC cell lines to conditioned media from M1 TAMs decreased viability and promoted pro-apoptotic morphologic changes, while treatment with M2-derived conditioned media increased cell proliferation and expression of the anti-apoptotic markers survivin and BMI-1 compared to control conditioned media [66]. Activated M2 TAMs also contribute to tumor angiogenesis through secretion of VEGF along with other TME cells [67,68,69].
M2 TAMs promote invasion and metastasis [70,71,72,73,74]. TAM secrete of MMP-9, a key proteolytic enzyme that contributes to ECM remodeling in multiple cancers, including CRC [72]. Concordantly, Afik et al. [74] demonstrated that CRC TAMs were enriched for molecular signatures associated with ECM remodeling in transcriptomic and proteomic analyses [74]. M2 TAMs also contribute to the production of TGF-β, which potentiates ECM remodeling, EMT, and metastasis [71]. EMT pertains to tumor cell loss of epithelial features (i.e., the loss of apical–basal polarity, dissolution of tight junctions, etc.) and assumption of a mesenchymal-like phenotype [75]. The mesenchymal phenotype is associated with increased invasiveness and migratory potential [75]. The loss of E-cadherin expression, a hallmark of the epithelial phenotype, is associated with a poor prognosis in CRC [76]. TAMs also promote EMT through IL-6-mediated activation of the JAK2/STAT3 pathway and subsequent inhibition of the miRNA tumor suppressor miR-506-3p and its target, FoxQ1, as well as through secretion of CCL22 and subsequent activation of the PI3K/AKT pathway [73,77].
M2 TAMs also facilitate immune evasion through secretion of immunosuppressive cytokines IL-10 and TGF-β, which facilitate suppression of T lymphocytes [65,78]. Herbeuval et al. [79] demonstrated that TAMs induced an immunosuppressive environment through the secretion of IL-6, leading to STAT3-mediated production of IL-10 by CRC cells. Thus, M2 TAMs in the CRC TME drive tumor progression, invasiveness, and a more aggressive tumor phenotype.

TAMs in CRLM and CRPM

TAMs are directly implicated in CRLM and may have context-specific pro-metastatic and anti-metastatic effects. In a microarray analysis of 159 resected CRLM specimens, the presence of CD68+ TAMs was associated with longer disease-free survival (DFS) on multivariable analysis [80]. Conversely, Wang et al. [81] demonstrated that CD206+ M2 TAMs were positively correlated with the presence of CRLM in patient samples; activation of the CXCL12/CXCR4 axis in CRC tumor cells promoted the exosome-mediated release of PTEN-suppressing miRNAs (miR-25-3p, miR-130b-3p, and miR-425-5p), which polarized TAMs to an M2 phenotype. Notably, murine tumors derived from HCT116 and MC38 CRC cells plus macrophages transfected with these miRNAs significantly increased tumor volume, and tail vein co-injection resulted in a significantly greater number and size of CRLM nodules compared to an injection of tumor cells alone [81]. KCs may also contribute to CRLM through the promotion of ECM remodeling and facilitation of tumor cell invasion for those CRC cells that escaped initial phagocytosis [43]. In this sense, they may serve a protective role against tumor cell infiltration earlier during metastasis. Indeed, Wen et al. demonstrated that the depletion of KCs at the earlier exponential growth phase increased CRLM burden in orthotopic murine models, while delayed KC depletion restricted tumor burden. This suggests that the pro- or anti-metastatic function of KCs may be time-dependent [82]. KCs and TAMs may also be impacted by CRLM resection. In a murine model of associated liver partition and portal vein ligation for staged hepatectomy for CRLM, mice who underwent this combined procedure had a significantly higher infiltration of KCs, which were polarized to the M1 phenotype, while TAMs were polarized to the M2 phenotype. This suggests that M1 KCs may be involved in a reactive inflammatory response, and that TAMs, perhaps M2-polarized as a consequence of induced hypoxia after the procedure, may contribute to persistent tumor progression and mixed therapeutic response [83].
How TAMs directly contribute to CRPM formation is not as well-elucidated. Cai et al. [71] demonstrated in CT26 CRC cell line–derived xenografts that the injection of TAMs into the tumor site significantly increased the number of CRPM nodules and total lung weight, a process dependent on TAM-derived TGF-β secretion and the induction of EMT through the modulation of the Smad2/3/4–Snail–E-cadherin pathway. Recently, KRAS mutational status was identified as a metastatic tropic factor for CRPM development, significantly more so than CRLM [84]. Patients with mutant KRAS tumors had a significantly shorter time to CRPM development and CRPM-free survival compared to those wild-type KRAS status [84]. Concordantly, Liu et al. [85] demonstrated that mutant KRAS reprograms macrophages to a M2 TAM-like phenotype through colony-stimulating factor 2 and lactate production, which promoted tumor progression and conferred EGFR inhibitor resistance.

5.1.2. Helper T Cells

CD4+ helper T cells are key mediators of the adaptive immune system, secreting cytokines to modulate the activity of other immune cells in response to infection or cancer. The presence of tumor-infiltrating lymphocytes (TILs) is associated with improved survival in CRC. In a study of 342 resected CRC specimens, high CD4+ T-cell density was associated with improved relapse-free survival and disease-specific survival [86]. However, CD4+ T-cell subtypes in the TME have varying impacts on tumor behavior. T-helper 1 cells (TH1) are traditionally thought to enhance CTL effector function and are instrumental for augmentation of the antitumor immune response [87]. Increased expression of TH1-cluster genes in resected CRC specimens is associated with improved disease-free survival, while higher numbers of T-helper 17 cells (TH17), which have an immunosuppressive function, are associated with poor survival [88]. This is consistent with prior preclinical studies, which suggest that TH17-derived cytokines, such as IL-17, IL-21, and IL-22, promote CRC tumor growth [89].

Helper T Cells in CRLM and CRPM

Kroemer et al. [90] demonstrated that the presence of expanded TH17 CD4+ T cells in resected CRLM was associated with poor prognosis. At the tumor site, while CD4+ T cells may be the most frequently encountered T-cell population, their ability to proliferate in response to tumor antigens after DC-mediated pulsing was significantly diminished [91]. These findings are consistent with those from Katz et al. [92], who demonstrated that high CD4+ T-cell density was inversely associated with survival after CRLM resection. In another study, Katz et al. [93] demonstrated through murine models that hepatic CD4+ T cells are generally polarized toward the TH2 phenotype and produced immunosuppressive cytokines. This may, in part, explain why the presence of CD4+ T cells negatively impacts prognosis. Little is known about how CD4+ T cells specifically impact CRPM. In other lung metastasis models including breast cancer, TH2-CD4+ T cells have been shown to augment metastasis through the potentiation of macrophage activity [94]. Further investigation into how CD4+ T cells specifically impact CRPM is necessary.

5.1.3. Dendritic Cells (DCs)

DCs are professional antigen-presenting cells that are crucial for the initiation of the immune response to specific antigens through internalization of foreign antigens and subsequent presentation to T cells. To avoid immune surveillance, cancer cells may suppress DCs through multiple mechanisms, including the secretion of immunosuppressive TGF-β [95]. In CRC, myeloid DCs, the most common DC subtype associated with cell-mediated immunity and stimulation of naïve CD4+ T cells to the TH1 phenotype, are found in increased frequency at the tumor invasive front and are associated with lymph node invasion [96]. In contrast, Gulubova et al. [97] noted that advanced-stage CRC tumors had a lower density of CD83+ DCs in the stroma and invasive margin [97].
The mechanism through which these DCs enhance invasiveness has not been well-elucidated. Orsini et al. [98] demonstrated that DCs from CRC patients had impaired antigen-presenting capacity and reduced co-stimulatory molecule expression compared to DCs from healthy controls. Furthermore, CRC patient-derived DCs secreted increased levels of immunosuppressive IL-10 and decreased levels of immunostimulatory IL-12 and TNF-α [98]. Concordantly, Nagorsen et al. [99] demonstrated that while S100+ DCs were found more frequently in limited disease and were associated with improved survival, their presence in the CRC TME was also positively associated with Treg infiltration, further highlighting the paradoxical pro-tumorigenic function of CRC-hijacked DCs. Indeed, Hsu et al. [100] demonstrated that DCs harvested from CRC patients highly expressed CXCL1, which enhanced tumor cell migration, cancer cell stemness, and EMT. Thus, DC immunostimulatory capacity may be suppressed by CRC cells and modulated into an invasive, immunosuppressive phenotype, perhaps reflecting a reversion back to an immature phenotype [101].

DCs in CRLM and CRPM

Enhancing the DC presence in the CRLM TME may be a therapeutic target. Fused allogenic peripheral blood DCs with SW620 CRC cells promoted significantly higher secretion of IFN-γ by CD8+ T cells and, when injected into highly immunodeficient CRLM mouse models, markedly reduced tumor growth compared to sham-vaccinated CRC cells [102]. These findings are consistent with prognostic data suggesting that the absence of CD83+ DCs in resected CRLM specimens is associated with worse 5-year OS, partially due to diminished antigen presentation and immunostimulation [103]. This vaccination strategy was being investigated in a CRLM clinical trial which, despite being terminated early, demonstrated evidence of improved median DFS [104].
Hsu et al. [100] demonstrated that CXCL1 secretion by DCs augments CRPM. The sequencing of CXCL1-enriched SW620 CRC cells resulted in enrichment of prognostically significant genes including PTHLH (parathyroid hormone-like hormone). When these cells were implanted into murine livers, they were found to have a heightened capacity for CRPM formation compared to native SW620 cells, which was dependent on suppression of the p38/MAPK pathway and subsequent upregulation of PTHLH [105]. The inhibition of PTHLH selectively inhibited CRPM formation from CRLM, but not the growth of directly implanted cells or of liver metastases themselves [105].

5.1.4. Regulatory T Cells (Tregs)

Tregs, characterized by CD25 and FoxP3 expression, are potent mediators of immunosuppression. Their presence in the TME is associated with increased metastasis and poor outcomes in numerous malignancies [106,107,108]. In their physiologic state, Tregs prevent autoimmunity and regulate the immune response by downregulating IL-2, releasing adenosine, and secreting immunosuppressive cytokines including TGF-β, IL-10, and IL-35 [109]. In CRC, the impact of Tregs in the TME is complex, and evidence suggests their function as pro- or anti-tumorigenic is context-specific. Ji et al. [110] demonstrated through mRNA profiling of 81 pretreatment biopsies of locally advanced rectal cancer patients that the Tregs were inversely correlated with prognosis. Conversely, other studies demonstrate that elevated FoxP3+ cell infiltration correlate with improved survival [86,111].
A potential explanation for the apparent contradictory effect of Tregs is likely due to their heterogeneity. Saito et al. [112] demonstrated that naïve Tregs (FoxP3low/CD45RA+) in CRC specimens could be stratified into terminally differentiated immunosuppressive FoxP3high/CD45RA-, and pro-inflammatory FoxP3low/CD45RA- subgroups. Compared to other tumors, such as melanoma, some CRC specimens had significantly higher numbers of FoxP3low/CD45RA- Tregs [112]. The authors therefore characterized CRC tumors as “type A” with a low expression of these inflammatory Tregs or “type B” with a high expression of inflammatory Tregs. Notably, type-B CRC demonstrated significant upregulation of genes involved with inflammation and immune response, including IL-12, TNF-β, and TGF-β [112]. Consistent with these findings, high FOXP3 expression in type-A CRC tumors was associated with a worse prognosis [112].

Regulatory T Cells in CRLM and CRPM

While Treg heterogeneity has not been investigated in detail specifically for CRLM, their overall presence markedly impacts prognosis. Pedroza-Gonzalez et al. [91] demonstrated that CRLMs contain sequestered activated Tregs at the tumor site, which highly expressed glucocorticoid-induced TNF receptor (GITR) and were more potently immunosuppressive compared to Tregs from primary hepatocellular carcinoma. The ablation of GITR prevented Treg-mediated suppression of effector T-cell activity, suggesting a possible immunostimulatory therapeutic target [91]. Katz et al. demonstrated immunohistochemical analysis of 188 resected CRLM specimens that higher ratios of FoxP3+ Treg:CD4+ and FoxP3+ Treg:CD8+ ratios were associated with shorter 5-year overall survival compared to lower ratios (34% vs. 51%, and 35% vs. 46%, respectively) [113].
Tregs are prevalent in CRPM, but little is known about specific functions in this niche [114]. Resident Tregs may contribute to the pulmonary pre-metastatic niche. In physiologic states, pulmonary tissue is enriched with Tregs, which are crucial in regulating immune responses to inhaled environmental antigens. Through many of the previously discussed mechanisms, pulmonary Tregs may promote immunosuppression in the lung microenvironment and foster a niche for pulmonary metastasis [115].

5.1.5. Neutrophils

Neutrophils are the first responders to acute infection or inflammation [116]. They secrete lytic enzymes, phagocytose certain microbes, generate neutrophil extracellular traps, and secrete numerous cytokines that sustain the inflammatory response and recruit other immune cells as part of the innate immune response. Tumor-associated neutrophils (TANs) are associated with prognosis; as such, understanding the function of TANs is an emerging area of investigation [117]. Fridlender et al. [118] demonstrated that TGF-β production in the CRC TME polarizes TANs from an immunostimulatory “N1” phenotype to a pro-tumorigenic “N2” phenotype. TANs promote invasiveness through modulation of angiogenesis and resistance to VEGF inhibition. In a colitis-associated CRC mouse model, Itatani et al. [119] demonstrated that the induction of colitis increased granulocyte colony stimulating factor (G-CSF) levels in anti-VEGF therapy-resistant tumors, which was associated with increased Bv8/PROK2+ neutrophil infiltration. The dual inhibition of G-CSF or its receptor along with VEGF inhibition resulted in significantly suppressed tumor formation and angiogenesis [119].
TANs also contribute to immune evasion. In an inducible CRC murine model, Germann et al. [120] demonstrated that presence of T cells in adenomas was diminished and inversely correlated with neutrophil infiltration. These TANs were enriched for immunosuppressive gene expression and secreted MMP-9, which suppressed T-cell proliferation through the activation of TGF-β [120]. Overall, TANs may also facilitate the dissemination of tumor cells by augmenting degradation of the basement membrane, which enhances tumor mobility and promotes tumor cell extravasation [121].

TANs in CRLM and CRPM

TANs contribute to the growth of CRLM through multiple mechanisms. In murine models, Gordon-Weeks et al. [122] demonstrated that CRLM-associated neutrophils were enriched in tumors, expressed high levels of fibroblast growth factor-2 (FGF2), and enhanced the growth of CRLM in mice. The depletion of neutrophils 7 days after an intrasplenic injection of CRC cells diminished CRLM development and angiogenesis. Monoclonal antibody (GAL-F2) blockade of FGF2 also restricted tumor growth and normalized vasculature, suggesting an FGF2-dependent mechanism through which TANs contribute to CRLM [122]. This is consistent with the previously discussed findings of Itatani et al., who also demonstrated that granulocyte-colony stimulating factor (G-CSF) secretion by CRC cells enhanced TAN recruitment in CRLM as well as in primary CRC tumors, and conferred similar resistance to anti-VEGF therapy [119]. Additionally, Yang et al. [123] demonstrated that neutrophils derived from CRC patient sera prominently secreted neutrophil extracellular traps (web-like consolidations of chromatin and proteases thought to be released by activated neutrophils and believed to enhance tumorigenesis perhaps through trapping cells at metastatic sites), which correlated with the risk of CRLM. Furthermore, CXCL8 secretion from CRC cells mediated the production of neutrophil extracellular traps, which enhanced CRLM formation in vivo [123].
A combination of SMAD4 deficiency in CRC cells plus CCR1+ TANs is implicated in the progression of CRPM [124]. SMAD4 is a tumor suppressor, and its inactivation is associated with multiple cancers, including CRC [125,126,127]. SMAD4-deficient CRC cells secrete higher levels of CCL15, which recruits CCR1+ TANs through the CCL15-CCR1 chemokine axis to promote CRPM [124]. TANs are also critical drivers for the development of the pre-metastatic niche at secondary sites like the lung [128,129,130]. In a model of melanoma pulmonary metastasis, El Rayes et al. [59] demonstrated that LPS-inflamed lungs were enriched for Ly6G+ neutrophils and developed a greater metastatic burden compared to controls. These TANs proteolytically degraded thrombospondin-1 (Tsp-1), an ECM protein that regulates inflammation, inhibits angiogenesis, and restricts tumor growth [59].

5.1.6. Cytotoxic T Cells

CTLs are the primary effector cells of the adaptive immune response and are a crucial component of antitumor immunity. The presence of CTLs in patient-derived CRC tumor cores is associated with a reduced risk of recurrence, particularly in higher-stage tumors [131]. Concordantly, the CRC Immunoscore prognostic tool was developed to supplement the TNM staging paradigm and is based on the prevalence of infiltrating CD3+ cells and CTLs. Higher Immunoscore values positively correlate with improved survival and are corroborated by internal and external validation studies [132,133].
To promote immune evasion, CTL activity is suppressed through multiple mechanisms. O’Malley et al. [134] demonstrated that TNF-α–mediated inflammation in CT26 CRC cells induced PD-L1 expression in stromal cells, resulting in the inhibition of activated granzyme-secreting CD8+ T cells [134]. Tumor and stromal cells express PD-L1, which, upon binding to the PD-1 receptor on T cells, suppresses T-cell activation. In CRC, a higher expression of PD-L1 is associated with higher stage, higher grade, lymph node involvement, distant metastasis, and reduced overall survival [135,136,137]. The PD-1/PD-L1 axis is an attractive target to increase immune targeting. Indeed, PD-1 blockade with pembrolizumab was FDA-approved for MSI-high/mismatch repair-deficient subtype advanced tumors in the pivotal KEYNOTE 177 trial [4,138]. Other TME cells also curtail CTL function through TGF-β secretion, which inhibits CTL expression of the key cytolytic enzymes: perforin, granzyme, and Fas ligand [139]. Furthermore, Tregs recruited via the CCL5/CCR5 signaling pathway have a heightened ability to kill CTLs in CRC murine models [140].

CTLs in CRLM and CRPM

CRLMs are more highly enriched for PD-L1 expression compared to primary tumors, particularly rectal cancer liver metastases [29]. In a study of 74 patients with resected CRLM and primary tumors, Wei et al. [29] found that high PD-L1 expression was significantly associated with an enrichment of CD4+ T cells and CTLs. Concordantly, Katz et al. [92] observed that a high density of CTLs positively correlated with 10-year survival after CRLM resection, and on multivariable analysis, a high CD8+/low CD4+ phenotype was significantly associated with better long-term survival.
Similarly, Remark et al. [141] found that an increased presence of LAMP+ DCs and CTLs was positively associated with improved overall survival in CRPM, but not in renal cell carcinoma lung metastases, suggesting a context-specific function. Furthermore, the density of CTLs, as well as DCs and NKs, in primary CRC tumors correlated with densities in CRPM, suggesting a reproducible immune pattern from the primary tumor possibly secondary to TME “imprinting” from the primary site or the development of educated immune cells [141]. The authors suggest that, since tertiary lymphoid structures are prevalent in CRPM, these T cells may be more educated compared to renal cell carcinoma metastases, which tend to have a scarce presence of tertiary lymphoid structures [141]. This may point toward a sensitivity of CRPM and CRLM to ICIs, particularly in patients with MSI-high/mismatch repair-deficient subtype advanced CRC [4,138].

5.2. Stromal Cells and the Extracellular Matrix

5.2.1. Extracellular Matrix

The ECM comprises structural proteins, including collagen, proteoglycans, hyaluronic acid, and glycosaminoglycans, whose structure and function can be remodeled by cells in the TME [142]. In addition to being the physical scaffolding for tumor cells, the ECM dynamically contributes to cell–cell adhesion, paracrine signaling, tumor proliferation, immune evasion, and metastasis [143]. Remodeling of the ECM is a consistent feature of the TME in many indications including CRC. Yang et al. [144] demonstrated that density of the remodeling enzyme MMP-9 was markedly greater in CRC compared to normal mucosa and higher expression was associated with worse prognosis. MMP-2 has also been implicated in ECM degradation and correlates with lymphatic invasion and advanced stage [145,146]. MMP-2 and MMP-9 encode collagenases that target type IV collagen, which is found in the basement membrane; and the degradation of type IV collagen can enhance tumor cell motility and invasion [145,147,148]. The biomechanics of the ECM also contribute to CRC proliferation. Using atomic force microscopy, Brauchle et al. [149] demonstrated that collagen-rich regions of the CRC ECM were stiffer compared to normal tissue. Stiffness of CRC has been associated with metastasis and EMT [150]. Li et al. [143] observed that the density of type I collagen positively correlated with stage, while type IV collagen density was reduced in higher stage tumors. This latter finding is consistent with invasion through the basement membrane [151]. The inverse relationship between density of type IV collagen and CRC stage likely reflects increased ECM remodeling, partly through MMP-2 and -9 activity [147]. At distant sites, proteomic analyses of control vs. matched CRLM patient tissues suggest that, while the ECM of CRLM more closely resembles the primary tumor compared to liver parenchyma, it is enriched for several unique proteins (i.e., SPP1 and COMP) [152]. Yuzhalin et al. [153] found that the CRLM ECM had higher levels of citrullinating enzyme peptidylarginine deiminase 4 (PAD4) compared to benign liver tissue, primary CRC, or colonic mucosa. CRC cells grown in citrullinated type I collagen exhibited mesenchymal-to-epithelial transition (reversion to a proliferative epithelial phenotype of tumor cells) in vitro and enhancement of CRLM growth in vivo [153].

5.2.2. Cancer-Associated Fibroblasts

Cancer-associated fibroblasts (CAFs) contribute significantly to ECM maintenance, desmoplasia, angiogenesis, immunosuppression, invasion, and chemoresistance [154,155]. While a majority of CAFs arise from resident stromal fibroblasts, mesenchymal stem cells (MSCs) and mesothelial cells also contribute a significant proportion of the CAF progenitor population [154,156]. Bone-marrow-derived MSCs, which are pluripotent stem cells involved in tissue remodeling, may be recruited to tumor sites to aid in tumor growth and progression [157]. Once localized at the tumor site, MSCs are induced by tumor-derived factors to assume a CAF-like phenotype [158]. Meanwhile, mesothelial cells contribute to the CAF population in highly invasive tumors via mesothelial-to-mesenchymal transition, which has been implicated in CRC peritoneal carcinomatosis [159]. Overall, in CRC, two major subpopulations of CAFs have been identified through single-cell RNA sequencing: CAF-A (which express ECM remodeling-associated genes, including MMP2, DCN, and COL1A2) and CAF-B (myofibroblast-like that express ACTA2, TAGLN, and PDGFA) [160].
CAFs promote immune evasion [120,154,161,162]. In microsatellite stable CRC samples, Tauriello et al. demonstrated an inverse correlation between the immunostimulatory TH1 to naïve T-cell ratio and the mean expression of CAF-specific genes [162]. Tumor invasive margins had high levels of stromal TGF-β, of which CAFs were the primary source. The elevated expression of TGF-β in the TME inhibited TH1 T-cell function. Additionally, CAFs secrete CXCL8, which attracts monocytes to the CRC TME and promotes M2 polarization, further contributing to immunosuppression [163].
CAFs promote metastasis through ECM remodeling and EMT in multiple cancers, including CRC [164,165,166,167]. Their presence in the CRC intratumoral stroma is associated with lymphatic invasion [168]. Through the secretion of ECM remodeling enzymes, collagen, and other cytoskeletal proteins, CAFs promote desmoplasia, which can be identified in up to 78% of CRC tumor specimens [168]. TGF-β/Smad2 signaling is prominently activated in CAFs, and CRC cells themselves can stimulate TGF-β production in CAFs with resulting expression of α-SMA and differentiation to a myofibroblastic phenotype [169]. These upregulate the expression of invasion-related proteinases, including multiple MMPs [169]. Notably, TAMs may regulate CAF activity in the CRC ECM with respect to the expression of collagen types I and XIV [74]. The expression of collagen crosslinking enzyme lysyl oxidase–like 2 (LOXL2) by CAFs in CRC tumors is associated with a higher rate of recurrence and worse overall survival and disease-free survival [170]. Through LOXL2, CAFs also stimulate EMT through activation of the FAK pathway, with a resultant reduction in E-cadherin protein expression. CRC cells co-cultured with these activated fibroblasts demonstrated increased migration rates [167]. CAFs also promote angiogenesis through secretion of IL-6; Nagasaki et al. [171] showed that CRC cells potentiated IL-6 secretion from CAFs with resulting upregulation of VEGFA expression. Thus, CAFs contribute to CRC progression through immunosuppression, ECM remodeling, and the promotion of EMT.

CAFs in CRLM and CRPM

In murine models, CRLM tumor cells recruit CAFs to the metastatic site, which contributes to tumor progression [172]. Circulating levels of TGF-β are a predictor of future development of CRLM after resection of the primary tumor [173]. Calon et al. [174] demonstrated that increased stromal TGF-β signaling significantly promoted CRPM and CRLM in murine models through the upregulation of GP130–p-STAT3 signaling in CRC cells. TGF-β–stimulated CAFs in the CRC TME secreted IL-11, a key ligand for GP130, resulting in the upregulation of anti-apoptotic factors MCL-1 and Bcl-2 [174]. Intrasplenic injection of HT29-M6 cells autonomously producing IL-11 more robustly colonized the liver and exhibited reduced apoptosis within the first hours of colonization compared to controls [174]. CAFs also potentiate metastasis through secretion of hepatocyte growth factor (HGF). Zhang et al. [175] demonstrated that secretion of HGF by CAFs upregulated CD44, and enhanced adhesive and migratory capacity of CRC cells in vitro and increased CRLM and CRPM formation in vivo. Resident fibroblasts also contribute to CRLM formation as well as the TME CAF population. As discussed, activated HSCs secrete ECM components and provide scaffolding for newly seeded CRC cells and contribute to immune evasion [44]. Furthermore, HSC-derived myofibroblasts secrete stromal-cell-derived factor 1 (a CXCR4 ligand), which can promote primary tumorigenesis in nude mice. Concordantly, CXCR4 may be a therapeutic target, as its inhibition with AMD3100 significantly diminished CRLM [176]. HSCs may not be the primary progenitors for CRLM CAFs, but rather resident portal fibroblasts. Mueller et al. [177] demonstrated that CRLM CAFs are similar to resident portal fibroblasts in their myofibroblastic phenotypes (α-SMA+, ICAM-1+, Thy-1+) and do not closely resemble HSCs (in contrast to the HSC phenotype, they are negative for glial fibrillary acidic protein, desmin, and neural cell adhesion molecules).
CAFs impact the prognosis for CRPM. In a study of 181 CRC-patient primary-tumor and metastatic-site specimens, Kwak et al. [178] demonstrated that CRPM had lower density of CAFs compared to primary tumors, but similar to the density at other tumor sites. The presence of CAFs at these metastatic sites and the loss of their PTEN expression were significantly associated with a worse prognosis [178]. CAFs in the CRPM TME prominently secrete heat-shock protein-27 (Hsp27), a protein implicated in angiogenesis, EMT, and fibroblast motility [179]. Hsp27 expression was associated with reduced disease-free survival and overall survival after CRPM metastasectomy [179].

5.2.3. Endothelial Cells

Tumor vasculature prominently impacts CRC invasion and is an established therapeutic target [180,181]. Endothelial cells (ECs), along with pericytes, smooth muscle cells, and progenitor cells, comprise the vasculature [182]. While EC generally remain quiescent in healthy tissue, malignancy can induce vascular turnover and cell proliferation [183]. Local injury, hypoxia, and rapid malignant proliferation initiate paracrine signaling networks where factors such as VEGF, PDGF, CXCL8, and TNF-α secreted by multiple TME cells drive neovascularization to perfuse proliferating tumor tissue [184]. ECs are implicated in invasion and metastasis. Zhang et al. [185] demonstrated that expression of vascular cell-adhesion molecular 1 (VCAM1) expression in CRC was associated with more invasive features and poorer prognosis. Forced expression of VCAM1 in CRC cells promoted pseudopodia formation and increased trans-endothelial migration in vitro. Furthermore, after CRC cells have entered the systemic vasculature, circulating tumor-derived ECs have been recently identified that may reflect a unique EC state that contributes directly to tumor progression and metastasis [186].

Endothelial Cells in CRLM and CRPM

As discussed, LSECs are central in early metastatic seeding, which is driven by E-selectin mediated adhesion to the endothelium and VEGF-mediated lymphangiogenesis [37,187]. LSEC lectin (LSECtin) promotes migration in malignant colorectal cell lines in vitro and in vivo and inhibition of LSECtin reduced CRLM formation [188]. Additionally, tumor-activated LSECs secrete ICAM-1, which drives IL-1 production and decreased antitumor immune activity through impaired IFN-γ production and mannose receptor-dependent endocytosis [189]. LSECs have also been shown to induce chemotaxis and outgrowth in CRC by secreting macrophage inhibitory factors, whose levels correlate with CRLM development [190].
In CRPM, CUB domain-containing protein 1 (CDCP1) is a cell-surface marker which identifies colorectal circulating stem cells that exhibit tropism to the lung [191]. CDCP1 expression is associated with increased risk of CRPM formation and inferior metastasis-free survival, and it may be implicated in the seeding and adhesion of CTCs to the pulmonary vasculature [191]. Elevated VEGF expression in CRPMs has been identified as a prognostic marker which predicts lack of benefit from CRPM resection [192].
Figure 2. (A) Contributions of the key individual components of the CRC TME on tumor progression, invasion, ECM remodeling, immunosuppression, and metastasis in the primary tumor. (B) CRLM- and (C) CRPM-specific functions are depicted in the lower two panels. Green headings signify pro-tumorigenic functions, and red headings signify anti-tumorigenic functions. Abbreviations: CRC, colorectal cancer; ECM, extracellular matrix; EMT, epithelial–mesenchymal transition; MMP, matrix metalloproteinase; TME, tumor microenvironment.
Figure 2. (A) Contributions of the key individual components of the CRC TME on tumor progression, invasion, ECM remodeling, immunosuppression, and metastasis in the primary tumor. (B) CRLM- and (C) CRPM-specific functions are depicted in the lower two panels. Green headings signify pro-tumorigenic functions, and red headings signify anti-tumorigenic functions. Abbreviations: CRC, colorectal cancer; ECM, extracellular matrix; EMT, epithelial–mesenchymal transition; MMP, matrix metalloproteinase; TME, tumor microenvironment.
Cancers 13 06206 g002

6. Implications of TME Factors on the Treatment of mCRC

We have thus far reviewed the contribution individual cellular components of the CRC TME have on tumor progression, metastasis, and prognosis. Understanding these functions may be crucial for understanding how they impact the efficacy of established and emerging and novel therapies. Below, we highlight some of the key advancements made in the treatment of CRC and how they are influenced by the TME. Additionally, a summary of meta-analyses examining the efficacy of surgery and local ablative therapies for the treatment of CRPM and CRLM is provided in Table 1.

6.1. Local Ablative Techniques

6.1.1. Stereotactic Body Radiation Therapy

Patients who are not candidates for hepatic metastasectomy or pulmonary metastasectomy may be managed with stereotactic body radiation therapy (SBRT), through which focused radiation is used to ablate tumors while minimizing nearby tissue damage. A meta-analysis of 18 studies showed that SBRT for CRPM resulted in 3-year local control, overall survival, and progression-free survival rates of 60%, 52%, and 13%, respectively [193]. Rectal cancer pulmonary metastases may be more radioresistant, as primary rectal tumors are frequently treated with neoadjuvant chemoradiation; this may select for the survival of radioresistant cells or higher rates of KRAS mutations, which are associated with poorer survival after SBRT [194]. Genetic studies have also identified CRPMs as more intrinsically radioresistant [193]. Despite this, SBRT may enhance the immunogenicity of CRPMs. For example, in primary lung cancer, SBRT has been demonstrated to prime the immune TME through the upregulation of immunogenic cell surface markers (i.e., ICAM-1, MHC-1, or Fas), increase the release of tumor antigens, and enhance immunostimulatory cytokine production. Through the abscopal effect, even distant metastases not primarily targeted by radiation may exhibit enhanced antitumor immune response and cell death, perhaps in response to the “vaccine effect” of SBRT, with the release of CD8+ T cells stimulated by danger signals and cytokines. Conversely, SBRT increases the release of TGF-β and increases PD-L1 expression, which may simultaneously increase immunosuppression [195]. To this end, a phase Ib/II trial investigating fusion blockade of TGF-β and PD-L1 (M7284) and radiation therapy for MSI+ mCRC [196] and a phase Ib trial investigating SBRT + pembrolizumab for CRLM are ongoing [197].

6.1.2. Radiofrequency Ablation

Radiofrequency ablation therapy (RFA) relies on the insertion of electrodes into a tumor and passing high-frequency electrical currents to heat and destroy cells [15,198]. It is an established approach for CRLM with 5-year survivals ranging from 20 to 48.5% [198]. It has also been shown to be safe and effective for inoperable CRPM, though studies are limited by study size and patient heterogeneity [199,200]. According to a large meta-analysis, the 1-year overall survival following RFA ranges between 83.9 and 95%, while the 3-year survival ranges between 46 and 59.6% [199]. Unfortunately, recurrence at the ablative site is a persistent concern. This may be due, in part, to the modulation of the immune microenvironment. Stimulation of the antitumor immune response by RFA may contribute to its effectiveness but may also trigger reactive immunosuppression. In a case-controlled study of 38 patients with synchronous CRLM who underwent neoadjuvant RFA prior to hepatic metastasectomy, Shi et al. [201] demonstrated that RFA significantly increased the number of infiltrating T cells with a higher CD8+/CD4+ ratio. Interestingly, PD-L1 expression on tumor cells and tumor-associated immune cells was also upregulated, suggesting a “self-limiting” phenomenon, which was further corroborated by murine studies that demonstrated a decrease in the initially high CD8+/Treg ratio after RFA as well as a failure to generate a durable suppression of tumor growth [201]. The authors similarly demonstrated that incomplete RFA was also associated with CRLM progression and resistance to anti-PD-1 therapy in murine models [202]. While disappointing, the authors showed that combination therapy with complete RFA and anti-PD-1 therapy resulted in marked, durable reduction in tumor growth as well as increased infiltration of tumor-infiltrating lymphocytes [201].
Table 1. Summary of meta-analyses investigating 3-year PFS and OS after surgical resection or local ablation of CRLM and CRPM.
Table 1. Summary of meta-analyses investigating 3-year PFS and OS after surgical resection or local ablation of CRLM and CRPM.
Modality3-Year PFS3-Year OSReference
CRPM
 SurgeryNot reported68.6%Zabaleta et al., 2018 [203]
 RFANot reported35–65%Lyons et al., 2015 [204]
 SBRT13%52%Cao et al., 2019 [193]
CRLM
 Surgery31.2%63.8%Beppu et al., 2012 [205]
 RFA24%60%Di Martino et al., 2020 [206]
 SBRT11.5 months *31.5 months *Petrelli et al., 2018 [207]
* Only median PFS and OS reported.

6.2. TME-Specific Targeted Therapy

6.2.1. Amelioration of Inflammation

The CRC TME promotes a pro-inflammatory milieu, which has become a promising therapeutic target with repurposed agents. Aspirin, as an example, became the first drug recommended for chemoprevention based on compelling retrospective data [208]. Platelet activation is known to result in an immunosuppressive TME that spares cancer cells from immune surveillance. Aspirin inhibits platelet function and induces apoptosis of tumor cells by upregulation of TNFα and by inhibiting the mTOR pathway [209]. It also inhibits cell proliferation, metastasis, and angiogenesis through the inhibition of COX pathways [210]. Celecoxib, a COX-2 inhibitor, along with chemotherapy, demonstrated clinical benefit in COX-2-positive gastric cancer, but it did not meaningfully improve disease-free survival in combination with adjuvant FOLFOX for stage III CRC [211,212]. It is currently being evaluated in combination with anti-PD-1 immunotherapy in an ongoing phase II study of MSI-high/mismatch repair-deficient and high-tumor-mutational-burden CRC [213].

6.2.2. Anti-Angiogenic Therapy

Anti-angiogenic agents impede the formation of new vasculature, which is essential for tumor growth and metastasis. The most well-studied anti-angiogenic agent is bevacizumab, a humanized IgG monoclonal antibody that targets and inhibits VEGF-A, which is prominently produced by tumor cells, TAMs, and other cells in the TME [69]. It is FDA-approved for mCRC [180]. In a meta-analysis of seven clinical trials comprising 1838 patients, Cao et al. demonstrated that the addition of bevacizumab to chemotherapy after primary tumor resection prolonged overall survival compared to chemotherapy alone [214]. Similarly, the phase III TRIBE2 trial found that bevacizumab + FOLFOXIRI resulted in superior median overall survival (27.4 vs. 22.5 months) and median progression-free survival after first-line treatment (12 vs. 9.8 months) [215]. The inhibition of angiogenesis in the TME may even increase the chance for resectability of CRLM; for example, the BECOME trial demonstrated that the R0 resection rate of initially unresectable RAS-mutant CRLMs was 22.3% in the bevacizumab + FOLFOX6 group, compared with 5.8% in the FOLFOX6-only group [216,217,218]. The efficacy of bevacizumab specifically for pulmonary metastasectomy is less understood. A case series of 11 patients who received neoadjuvant bevacizumab + FOLFOX/capecitabine followed by pulmonary metastasectomy had a 5-year disease-free survival of 10.9% [219]. A case report of rectal pulmonary metastasis treated with pulmonary metastasectomy as well as adjuvant S-1, irinotecan, and bevacizumab yielded a complete response [220]. It is possible that the presence of tumor cavitation predicts response to bevacizumab; in a study of 60 CRPM patients treated with bevacizumab + chemotherapy, the presence of tissue cavitation yielded a longer median overall survival (42.1 vs. 30.8 months) [221]. For those patients who progress despite bevacizumab + 5-FU-oxaliplatin-based chemotherapy, ramucirumab (anti-VEGF-R2 monoclonal antibody) + FOLFIRI has been shown to significantly increase OS and PFS in the phase III RAISE trial of 1072 patients and is now FDA-approved [181,222].

6.2.3. Modulation of CAFs

Inhibition of CAFs is under investigation for mCRC in numerous clinical trials with mixed results thus far [154]. Targeting CAF activity through inhibition of the pro-desmoplastic Sonic Hedgehog pathway yielded no added clinical benefit in combination with FOLFOX or FOLFIRI + bevacizumab in a phase II trial of previously untreated mCRC [223]. Similarly, inhibition of LOXL2 via simtuzumab in combination with FOLFIRI yielded no clinical benefit in a phase II trial, and 90.6% of patients discontinued treatment due to progression [224]. CAFs are stimulated by and are a source of TGF-β. Thus, targeting TGF- β has garnered significant interest in CRC. A recent phase 1b trial of regorafenib with PF-03446962, a monoclonal antibody targeting TGF-β receptor activin-receptor-like kinase 1 (ALK-1) demonstrated significant toxicity and no clinical activity [225]. One of the emerging anti-TGF-β agents currently under clinical investigation is galunisertib, a small molecule selective inhibitor that suppresses the TGF-β pathway by inhibiting TGF-β receptor type 1 [226]. Galunisertib has been shown to reduce TGF-β-mediated suppression of T cells, provoking a strong cytotoxic T-cell response against CRC cells and enhancing the efficacy of ICIs [227]. In syngeneic CRC mouse models, galunisertib reduced tumor size, extent of peritoneal metastases, and CRLM, an effect that was dependent on CD8+ T-cell activity [162]. Clinical trials currently evaluating the efficacy of galunisertib or LY3200882 (another TGF-β receptor type 1 inhibitor) combined with capecitabine or 5-FU for advanced CRC, as well as neoadjuvant galunisertib with chemoradiation for rectal cancer, are underway [228,229,230].

6.2.4. Repolarizing Macrophages

The conversion of macrophage phenotype from a pro-tumorigenic to an anti-tumorigenic phenotype is an attractive strategy. In a key study of therapeutic targeting of TAMs for CRLM, Halama et al. [231] demonstrated that cytokines, including CCL5 (ligand for CCR5) were highly expressed at the invasive margin. CCL5 was exclusively secreted by CD3+ T cells, which polarized TAMs (which contain CCR5) into a pro-tumorigenic phenotype. Treatment with maraviroc (a CCR5 inhibitor) in a CRLM tumor explant model induced macrophage-dependent tumor cell death [231]. These findings led the authors to launch a phase I trial in 14 mCRC patients treated with maraviroc, which resulted in a partial response in three patients, including a partial remission of CRPM. Histologically, all post-treatment CRLM samples showed a reduced proliferative index, tumor cell death, and a reduction in the expression of pro-tumorigenic, pro-angiogenic, and chemoresistance-promoting cytokines [231]. These promising results led to a phase I trial of maraviroc in combination with PD-1 inhibition for patients with microsatellite-stable mCRC, who experienced higher-than-expected overall survival but limited clinical activity, suggesting that further investigation is necessary [232].

6.2.5. Inhibition of Tregs

While the impact of Tregs in CRC is controversial, the depletion of Tregs in CRPM may be clinically beneficial. In a murine CRPM model, Wang et al. [114] demonstrated that the administration of arsenic trioxide (As2O3) significantly reduced Treg infiltration and FoxP3 expression and reduced metastatic tumor size and number, and increased the cytotoxic activity of cytokine-induced killer cells [114]. A pilot study of 17 patients demonstrated that the addition of As2O3 to FOLFIRI for mCRC refractory to first-line FOLFOX/CAPOX yielded stable disease in 65% of patients, a partial response in 18%, and a median progression-free survival of 5.3 months [233].

6.2.6. Augmentation of the Antitumor T-Cell Response

An emerging approach for CRC is chimeric antigen receptor (CAR) T-cell therapy, in which a patient’s own T cells are extracted, genetically modified to recognize tumor antigens, and reinfused. The application of CAR T cells in CRLM faces several key hurdles, including the immunosuppressive nature of the metastatic TME, the challenge of efficiently trafficking the cells to the tumor site, and the need to target the tumor-associated antigen [234,235]. In a preclinical murine model, Burga et al. [236] found that the regional infusion of anti-carcinoembryonic antigen (CEA) CAR T cells via the portal vein delayed the tumor progression of CEA+ CRLMs. However, the growth of CRLMs was associated with granulocyte colony macrophage stimulating factor (GM-CSF)-mediated expansion of the CD11b+ Gr-1+ myeloid-derived tumor suppressor cell (L-MDSC) population, which abrogated CAR T-cell proliferation within the tumor via STAT3-dependent PD-L1 expression [236]. Through the use of potent STAT3 inhibitors (e.g., JSI-124 or celastrol) combined with anti-GM-CSF therapy, the suppression of L-MDSC expansion improved CAR T-cell antitumor activity [236]. Furthermore, infusions of CAR T cells via the portal vein resulted in superior antitumor efficacy compared to systemic infusion [236]. These preclinical findings have been translated into ongoing and completed phase I and II trials investigating regional CAR T-cell therapy for CRLM [234,235].
Although they have established efficacy for other cancers, ICIs have only recently become FDA-approved for MSI-high/mismatch repair-deficient CRC [138]. This subtype is more receptive to PD-1 blockade [237]. The recent phase III KEYNOTE 177 trial of 307 patients with treatment-naïve metastatic MSI-high/mismatch repair-deficient CRC demonstrated that pembrolizumab was significantly superior to 5-FU-based chemotherapy (with or without VEGF (bevacizumab) or EGFR (cetuximab) blockade) with respect to progression-free survival (16.5 vs. 8.2 months) and was better tolerated [4]. Similarly, ipilimumab (CTLA-4 inhibitor) and nivolumab (another PD-1 inhibitor) have been approved for MSI-high/mismatch repair deficient mCRC [238,239]. Given their efficacy and favorable side effect profile, these agents are now being investigated in the neoadjuvant setting for early stage disease with a stage I trial demonstrating good tolerability and high rate of pathologic response (100%) in MMR-deficient tumors and moderate rate (27%) in MMR-proficient tumors [240]. Another combination of anti-CTLA-4/anti-PD-L1 blockade (tremelimumab/durvalumab) has also yielded improved overall survival in microsatellite stable cases in a recent phase II trial [241]. Notably, the administration of first-line FOLFOX (5-FU, oxaliplatin, and leucovorin) increased CD8+ T-cell infiltration and ameliorated CD8+ T-cell exhaustion in a CRC mouse model; combination FOLFOX with anti-PD-1 therapy significantly reduced tumor growth [242]. Thus, combination chemo-immunotherapy with FOLFOX and immune checkpoint blockade may be particularly promising for CRC and warrants further investigation in clinical trials. Additionally, as previously discussed, combinations with other ablative therapies, such as RFA, also warrant continued investigation.
A summary of TME-specific targeted therapies currently under investigation in clinical trials for CRC is provided in Table 2.

7. Conclusions

The TME of CRC is involved in virtually every step of tumor progression, invasion, immune evasion, and metastasis. The liver and lungs are the most frequently involved organs for CRC metastasis. While most research has focused on the primary tumor, the body of knowledge addressing the unique microenvironments in these organs and how their different cell types contribute to metastasis continues to evolve. There is a paucity of the literature specifically investigating the impact of TME components on CRPM. This may reflect the difficulty in obtaining adequate CRPM tissue for translational investigation, as image-guided biopsies may not yield adequate spare tissue, and surgical biopsies, even with minimally invasive thoracoscopic approaches, may place patients with advanced, unresectable disease at prohibitive risk. Further investigation in preclinical models of CRPM to better elucidate the unique features of the TME is, therefore, crucial. A clear observation in the CRC TME is that immune cells are important in cancer progression. The local TME may suppress anti-tumorigenic functions or modulate immune cells into a pro-tumorigenic phenotype. Abrogating pro-tumorigenic and augmenting anti-tumorigenic functions of these TME components has been demonstrated to enhance antitumor immune activity in preclinical models and clinical trials, but further investigation is warranted to maximize the benefit of novel immunotherapies. Indeed, while CAF-targeted trials in combination with chemotherapy have yielded mixed results, the efficacy of combination therapy with immunotherapy remains to be elucidated. A continued focus on how each of the individual components of the TME contributes to CRC progression and metastasis is critical for the determination of potential targets for novel therapies.

Author Contributions

R.C., J.D.K., C.L., G.Z.M. and R.A.B. were involved in conceptualization of the article and writing of the original draft. R.C., J.D.K., C.L., G.Z.M., J.V.P., C.A.H., S.I.R., E.H., J.D.M. and R.A.B. were involved in reviewing and editing the final draft. R.C. and R.A.B. conducted the project administration. All authors have read and agreed to the published version of the manuscript.

Funding

R.C., J.D.K.: and G.Z.M. are supported by a Physician-Scientist Institutional Award from the Burroughs-Wellcome Fund (award No. 1018897). The University of Texas Southwestern Medical Center—Dallas holds a Physician-Scientist Institutional Award from the Burroughs Wellcome Fund. G.Z.M. is additionally supported by R01CA243577-02S1. EHH is supported by U01 CA214300 and R01 CA237304. JDM is supported by the Lung Cancer SPORE P50 CA070907. RAB is supported by R01 CA243577, U54 CA210181, project 2 and the Effie Marie Cain Scholarship in Angiogenesis Research.

Acknowledgments

The authors would like to thank Dave Primm of the University of Texas Southwestern Department of Surgery for help in editing this article. Figures constructed using BioRender.com (accessed on 15 October 2021).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Keum, N.; Giovannucci, E. Global burden of colorectal cancer: Emerging trends, risk factors and prevention strategies. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 713–732. [Google Scholar] [CrossRef]
  2. Kinj, R.; Bondiau, P.Y.; Francois, E.; Gerard, J.P.; Naghavi, A.O.; Leysalle, A.; Chamorey, E.; Evesque, L.; Padovani, B.; Ianessi, A.; et al. Radiosensitivity of Colon and Rectal Lung Oligometastasis Treated with Stereotactic Ablative Radiotherapy. Clin. Color. Cancer 2017, 16, e211–e220. [Google Scholar] [CrossRef] [PubMed]
  3. Widder, J.; Klinkenberg, T.J.; Ubbels, J.F.; Wiegman, E.M.; Groen, H.J.; Langendijk, J.A. Pulmonary oligometastases: Metastasectomy or stereotactic ablative radiotherapy? Radiother. Oncol. 2013, 107, 409–413. [Google Scholar] [CrossRef] [PubMed]
  4. Andre, T.; Shiu, K.K.; Kim, T.W.; Jensen, B.V.; Jensen, L.H.; Punt, C.; Smith, D.; Garcia-Carbonero, R.; Benavides, M.; Gibbs, P.; et al. Pembrolizumab in Microsatellite-Instability-High Advanced Colorectal Cancer. N. Engl. J. Med. 2020, 383, 2207–2218. [Google Scholar] [CrossRef]
  5. Benson, A.B.; Venook, A.P.; Al-Hawary, M.M.; Arain, M.A.; Chen, Y.-J.; Ciombor, K.K.; Cohen, S.; Cooper, H.S.; Deming, D.; Farkas, L. Colon cancer, version 2.2021, NCCN clinical practice guidelines in oncology. J. Natl. Compr. Cancer Netw. 2021, 19, 329–359. [Google Scholar] [CrossRef]
  6. Koodziejski, L.; Goralczyk, J.; Dyczek, S.; Duda, K.; Nabiaek, T. The role of surgery in lung metastases. Eur. J. Surg. Oncol. 1999, 25, 410–417. [Google Scholar] [CrossRef]
  7. Rotolo, N.; De Monte, L.; Imperatori, A.; Dominioni, L. Pulmonary resections of single metastases from colorectal cancer. Surg. Oncol. 2007, 16, 141–144. [Google Scholar] [CrossRef] [PubMed]
  8. Villeneuve, P.J.; Sundaresan, R.S. Surgical management of colorectal lung metastasis. Clin. Colon Rectal Surg. 2009, 22, 233–241. [Google Scholar] [CrossRef] [Green Version]
  9. Guglielmi, A.; Ruzzenente, A.; Conci, S.; Valdegamberi, A.; Iacono, C. How much remnant is enough in liver resection? Dig. Surg. 2012, 29, 6–17. [Google Scholar] [CrossRef]
  10. Zellweger, M.; Abdelnour-Berchtold, E.; Krueger, T.; Ris, H.-B.; Perentes, J.Y.; Gonzalez, M. Surgical treatment of pulmonary metastasis in colorectal cancer patients: Current practice and results. Crit. Rev. Oncol. Hematol. 2018, 127, 105–116. [Google Scholar] [CrossRef] [PubMed]
  11. Treasure, T.; Farewell, V.; Macbeth, F.; Monson, K.; Williams, N.R.; Brew-Graves, C.; Lees, B.; Grigg, O.; Fallowfield, L. Pulmonary metastasectomy versus continued active monitoring in colorectal cancer (PulMiCC): A multicentre randomised clinical trial. Trials 2019, 20, 718. [Google Scholar] [CrossRef] [Green Version]
  12. Siebenhüner, A.R.; Güller, U.; Warschkow, R. Population-based SEER analysis of survival in colorectal cancer patients with or without resection of lung and liver metastases. BMC Cancer 2020, 20, 246. [Google Scholar] [CrossRef] [PubMed]
  13. Choti, M.A.; Sitzmann, J.V.; Tiburi, M.F.; Sumetchotimetha, W.; Rangsin, R.; Schulick, R.D.; Lillemoe, K.D.; Yeo, C.J.; Cameron, J.L. Trends in long-term survival following liver resection for hepatic colorectal metastases. Ann. Surg. 2002, 235, 759–766. [Google Scholar] [CrossRef]
  14. Figueras, J.; Valls, C.; Rafecas, A.; Fabregat, J.; Ramos, E.; Jaurrieta, E. Resection rate and effect of postoperative chemotherapy on survival after surgery for colorectal liver metastases. Br. J. Surg. 2001, 88, 980–985. [Google Scholar] [CrossRef] [PubMed]
  15. Abdalla, E.K.; Vauthey, J.N.; Ellis, L.M.; Ellis, V.; Pollock, R.; Broglio, K.R.; Hess, K.; Curley, S.A. Recurrence and outcomes following hepatic resection, radiofrequency ablation, and combined resection/ablation for colorectal liver metastases. Ann. Surg. 2004, 239, 818–825, discussion 825–827. [Google Scholar] [CrossRef] [PubMed]
  16. Chiorean, E.G.; Nandakumar, G.; Fadelu, T.; Temin, S.; Alarcon-Rozas, A.E.; Bejarano, S.; Croitoru, A.E.; Grover, S.; Lohar, P.V.; Odhiambo, A.; et al. Treatment of Patients With Late-Stage Colorectal Cancer: ASCO Resource-Stratified Guideline. JCO Glob. Oncol. 2020, 6, 414–438. [Google Scholar] [CrossRef]
  17. Kanas, G.P.; Taylor, A.; Primrose, J.N.; Langeberg, W.J.; Kelsh, M.A.; Mowat, F.S.; Alexander, D.D.; Choti, M.A.; Poston, G. Survival after liver resection in metastatic colorectal cancer: Review and meta-analysis of prognostic factors. Clin. Epidemiol. 2012, 4, 283. [Google Scholar]
  18. Pawlik, T.M.; Scoggins, C.R.; Zorzi, D.; Abdalla, E.K.; Andres, A.; Eng, C.; Curley, S.A.; Loyer, E.M.; Muratore, A.; Mentha, G.; et al. Effect of surgical margin status on survival and site of recurrence after hepatic resection for colorectal metastases. Ann. Surg. 2005, 241, 715–722, discussion 722–714. [Google Scholar] [CrossRef]
  19. Alieva, M.; van Rheenen, J.; Broekman, M.L. Potential impact of invasive surgical procedures on primary tumor growth and metastasis. Clin. Exp. Metastasis 2018, 35, 319–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Weber, M.; Moebius, P.; Büttner-Herold, M.; Amann, K.; Preidl, R.; Neukam, F.; Wehrhan, F. Macrophage polarisation changes within the time between diagnostic biopsy and tumour resection in oral squamous cell carcinomas—An immunohistochemical study. Br. J. Cancer 2015, 113, 510–519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Ferrarotto, R.; Pathak, P.; Maru, D.; Agarwal, A.; Overman, M.; Hoff, P.M.; Kopetz, S. Durable complete responses in metastatic colorectal cancer treated with chemotherapy alone. Clin. Color. Cancer 2011, 10, 178–182. [Google Scholar] [CrossRef]
  22. Xie, Y.-H.; Chen, Y.-X.; Fang, J.-Y. Comprehensive review of targeted therapy for colorectal cancer. Signal Transduct. Target. Ther. 2020, 5, 1–30. [Google Scholar] [CrossRef]
  23. Robinson, J.R.; Newcomb, P.A.; Hardikar, S.; Cohen, S.A.; Phipps, A.I. Stage IV colorectal cancer primary site and patterns of distant metastasis. Cancer Epidemiol. 2017, 48, 92–95. [Google Scholar] [CrossRef] [PubMed]
  24. Horn, S.R.; Stoltzfus, K.C.; Lehrer, E.J.; Dawson, L.A.; Tchelebi, L.; Gusani, N.J.; Sharma, N.K.; Chen, H.; Trifiletti, D.M.; Zaorsky, N.G. Epidemiology of liver metastases. Cancer Epidemiol. 2020, 67, 101760. [Google Scholar] [CrossRef] [PubMed]
  25. Eefsen, R.L.; Van Den Eynden, G.G.; Høyer-Hansen, G.; Brodt, P.; Laerum, O.D.; Vermeulen, P.B.; Christensen, I.J.; Wettergren, A.; Federspiel, B.; Willemoe, G.L.; et al. Histopathological Growth Pattern, Proteolysis and Angiogenesis in Chemonaive Patients Resected for Multiple Colorectal Liver Metastases. J. Oncol. 2012, 2012, 907971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Chu, P.C.; Lin, P.C.; Wu, H.Y.; Lin, K.T.; Wu, C.; Bekaii-Saab, T.; Lin, Y.J.; Lee, C.T.; Lee, J.C.; Chen, C.S. Mutant KRAS promotes liver metastasis of colorectal cancer, in part, by upregulating the MEK-Sp1-DNMT1-miR-137-YB-1-IGF-IR signaling pathway. Oncogene 2018, 37, 3440–3455. [Google Scholar] [CrossRef] [PubMed]
  27. Brudvik, K.W.; Kopetz, S.E.; Li, L.; Conrad, C.; Aloia, T.A.; Vauthey, J.N. Meta-analysis of KRAS mutations and survival after resection of colorectal liver metastases. Br. J. Surg. 2015, 102, 1175–1183. [Google Scholar] [CrossRef] [Green Version]
  28. Tian, X.; Werner, M.E.; Roche, K.C.; Hanson, A.D.; Foote, H.P.; Yu, S.K.; Warner, S.B.; Copp, J.A.; Lara, H.; Wauthier, E.L.; et al. Organ-specific metastases obtained by culturing colorectal cancer cells on tissue-specific decellularized scaffolds. Nat. Biomed. Eng. 2018, 2, 443–452. [Google Scholar] [CrossRef]
  29. Wei, X.-L.; Luo, X.; Sheng, H.; Wang, Y.; Chen, D.-L.; Li, J.-N.; Wang, F.-H.; Xu, R.-H. PD-L1 expression in liver metastasis: Its clinical significance and discordance with primary tumor in colorectal cancer. J. Transl. Med. 2020, 18, 475. [Google Scholar] [CrossRef] [PubMed]
  30. Giakoustidis, A.; Mudan, S.; Hagemann, T. Tumour microenvironment: Overview with an emphasis on the colorectal liver metastasis pathway. Cancer Microenviron. 2015, 8, 177–186. [Google Scholar] [CrossRef] [Green Version]
  31. Brodt, P. Role of the Microenvironment in Liver Metastasis: From Pre- to Prometastatic Niches. Clin. Cancer Res. 2016, 22, 5971–5982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Milette, S.; Sicklick, J.K.; Lowy, A.M.; Brodt, P. Molecular Pathways: Targeting the Microenvironment of Liver Metastases. Clin. Cancer Res. 2017, 23, 6390–6399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Timmers, M.; Vekemans, K.; Vermijlen, D.; Asosingh, K.; Kuppen, P.; Bouwens, L.; Wisse, E.; Braet, F. Interactions between rat colon carcinoma cells and Kupffer cells during the onset of hepatic metastasis. Int. J. Cancer 2004, 112, 793–802. [Google Scholar] [CrossRef] [PubMed]
  34. Matsumura, H.; Kondo, T.; Ogawa, K.; Tamura, T.; Fukunaga, K.; Murata, S.; Ohkohchi, N. Kupffer cells decrease metastasis of colon cancer cells to the liver in the early stage. Int. J. Oncol. 2014, 45, 2303–2310. [Google Scholar] [CrossRef] [Green Version]
  35. Haier, J.; Korb, T.; Hotz, B.; Spiegel, H.-U.; Senninger, N. An intravital model to monitor steps of metastatic tumor cell adhesion within the hepatic microcirculation. J. Gastrointest. Surg. 2003, 7, 507–515. [Google Scholar] [CrossRef]
  36. Gassmann, P.; Hemping-Bovenkerk, A.; Mees, S.T.; Haier, J. Metastatic tumor cell arrest in the liver–lumen occlusion and specific adhesion are not exclusive. Int. J. Color. Dis. 2009, 24, 851–858. [Google Scholar] [CrossRef] [PubMed]
  37. Brodt, P.; Fallavollita, L.; Bresalier, R.S.; Meterissian, S.; Norton, C.R.; Wolitzky, B.A. Liver endothelial E-selectin mediates carcinoma cell adhesion and promotes liver metastasis. Int. J. Cancer 1997, 71, 612–619. [Google Scholar] [CrossRef]
  38. Khatib, A.-M.; Fallavollita, L.; Wancewicz, E.V.; Monia, B.P.; Brodt, P. Inhibition of hepatic endothelial E-selectin expression by C-raf antisense oligonucleotides blocks colorectal carcinoma liver metastasis. Cancer Res. 2002, 62, 5393–5398. [Google Scholar]
  39. Khatib, A.-M.; Auguste, P.; Fallavollita, L.; Wang, N.; Samani, A.; Kontogiannea, M.; Meterissian, S.; Brodt, P. Characterization of the host proinflammatory response to tumor cells during the initial stages of liver metastasis. Am. J. Pathol. 2005, 167, 749–759. [Google Scholar] [CrossRef] [Green Version]
  40. Khatib, A.-M.; Kontogiannea, M.; Fallavollita, L.; Jamison, B.; Meterissian, S.; Brodt, P. Rapid induction of cytokine and E-selectin expression in the liver in response to metastatic tumor cells. Cancer Res. 1999, 59, 1356–1361. [Google Scholar]
  41. Lee, Y.; Friedman, S.L. Fibrosis in the Liver; Elsevier: Amsterdam, The Netherlands, 2010; pp. 151–200. [Google Scholar]
  42. Döme, B.; Hendrix, M.J.C.; Paku, S.; Tóvári, J.; Tímár, J. Alternative Vascularization Mechanisms in Cancer. Am. J. Pathol. 2007, 170, 1–15. [Google Scholar] [CrossRef] [Green Version]
  43. Tsilimigras, D.I.; Brodt, P.; Clavien, P.-A.; Muschel, R.J.; D’Angelica, M.I.; Endo, I.; Parks, R.W.; Doyle, M.; De Santibañes, E.; Pawlik, T.M. Liver metastases. Nat. Rev. Dis. Primers 2021, 7, 1–23. [Google Scholar] [CrossRef] [PubMed]
  44. Huang, W.-H.; Zhou, M.-W.; Zhu, Y.-F.; Xiang, J.-B.; Li, Z.-Y.; Wang, Z.-H.; Zhou, Y.-M.; Yang, Y.; Chen, Z.-Y.; Gu, X.-D. The role of hepatic stellate cells in promoting liver metastasis of colorectal carcinoma. OncoTargets Ther. 2019, 12, 7573. [Google Scholar] [CrossRef] [Green Version]
  45. Breedis, C.; Young, G. The blood supply of neoplasms in the liver. Am. J. Pathol. 1954, 30, 969. [Google Scholar]
  46. Hurwitz, H.; Fehrenbacher, L.; Novotny, W.; Cartwright, T.; Hainsworth, J.; Heim, W.; Berlin, J.; Baron, A.; Griffing, S.; Holmgren, E.; et al. Bevacizumab plus Irinotecan, Fluorouracil, and Leucovorin for Metastatic Colorectal Cancer. N. Engl. J. Med. 2004, 350, 2335–2342. [Google Scholar] [CrossRef] [Green Version]
  47. Donnem, T.; Hu, J.; Ferguson, M.; Adighibe, O.; Snell, C.; Harris, A.L.; Gatter, K.C.; Pezzella, F. Vessel co-option in primary human tumors and metastases: An obstacle to effective anti-angiogenic treatment? Cancer Med. 2013, 2, 427–436. [Google Scholar] [CrossRef] [Green Version]
  48. Frentzas, S.; Simoneau, E.; Bridgeman, V.L.; Vermeulen, P.B.; Foo, S.; Kostaras, E.; Nathan, M.R.; Wotherspoon, A.; Gao, Z.-H.; Shi, Y.; et al. Vessel co-option mediates resistance to anti-angiogenic therapy in liver metastases. Nat. Med. 2016, 22, 1294–1302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Fernández Moro, C.; Bozóky, B.; Gerling, M. Growth patterns of colorectal cancer liver metastases and their impact on prognosis: A systematic review. BMJ Open Gastroenterol. 2018, 5, e000217. [Google Scholar] [CrossRef] [Green Version]
  50. Parnaby, C.N.; Bailey, W.; Balasingam, A.; Beckert, L.; Eglinton, T.; Fife, J.; Frizelle, F.A.; Jeffery, M.; Watson, A.J. Pulmonary staging in colorectal cancer: A review. Color. Dis. 2012, 14, 660–670. [Google Scholar] [CrossRef] [PubMed]
  51. Mitry, E.; Guiu, B.; Cosconea, S.; Jooste, V.; Faivre, J.; Bouvier, A.M. Epidemiology, management and prognosis of colorectal cancer with lung metastases: A 30-year population-based study. Gut 2010, 59, 1383–1388. [Google Scholar] [CrossRef]
  52. Van der Geest, L.G.; Lam-Boer, J.; Koopman, M.; Verhoef, C.; Elferink, M.A.; de Wilt, J.H. Nationwide trends in incidence, treatment and survival of colorectal cancer patients with synchronous metastases. Clin. Exp. Metastasis 2015, 32, 457–465. [Google Scholar] [CrossRef] [PubMed]
  53. Zhang, N.; Di, J.; Wang, Z.; Gao, P.; Jiang, B.; Su, X. Genomic profiling of colorectal cancer with isolated lung metastasis. Cancer Cell Int. 2020, 20, 281. [Google Scholar] [CrossRef]
  54. Riihimäki, M.; Hemminki, A.; Sundquist, J.; Hemminki, K. Patterns of metastasis in colon and rectal cancer. Sci. Rep. 2016, 6, 29765. [Google Scholar] [CrossRef] [Green Version]
  55. Agostini, C.; Chilosi, M.; Zambello, R.; Trentin, L.; Semenzato, G. Pulmonary immune cells in health and disease: Lymphocytes. Eur. Respir J. 1993, 6, 1378–1401. [Google Scholar]
  56. Altorki, N.K.; Markowitz, G.J.; Gao, D.; Port, J.L.; Saxena, A.; Stiles, B.; McGraw, T.; Mittal, V. The lung microenvironment: An important regulator of tumour growth and metastasis. Nat. Rev. Cancer 2019, 19, 9–31. [Google Scholar] [CrossRef] [Green Version]
  57. Houghton, A.M. Mechanistic links between COPD and lung cancer. Nat. Rev. Cancer 2013, 13, 233–245. [Google Scholar] [CrossRef] [PubMed]
  58. Wood, S.L.; Pernemalm, M.; Crosbie, P.A.; Whetton, A.D. The role of the tumor-microenvironment in lung cancer-metastasis and its relationship to potential therapeutic targets. Cancer Treat. Rev. 2014, 40, 558–566. [Google Scholar] [CrossRef] [PubMed]
  59. El Rayes, T.; Catena, R.; Lee, S.; Stawowczyk, M.; Joshi, N.; Fischbach, C.; Powell, C.A.; Dannenberg, A.J.; Altorki, N.K.; Gao, D. Lung inflammation promotes metastasis through neutrophil protease-mediated degradation of Tsp-1. Proc. Natl. Acad. Sci. USA 2015, 112, 16000–16005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Yahagi, M.; Tsuruta, M.; Hasegawa, H.; Okabayashi, K.; Toyoda, N.; Iwama, N.; Morita, S.; Kitagawa, Y. Smoking is a risk factor for pulmonary metastasis in colorectal cancer. Color. Dis. 2017, 19, O322–O328. [Google Scholar] [CrossRef] [PubMed]
  61. García-Mulero, S.; Alonso, M.H.; Pardo, J.; Santos, C.; Sanjuan, X.; Salazar, R.; Moreno, V.; Piulats, J.M.; Sanz-Pamplona, R. Lung metastases share common immune features regardless of primary tumor origin. J. Immunother. Cancer 2020, 8, 1–12. [Google Scholar] [CrossRef] [PubMed]
  62. Orecchioni, M.; Ghosheh, Y.; Pramod, A.B.; Ley, K. Macrophage polarization: Different gene signatures in M1 (LPS+) vs. classically and M2 (LPS–) vs. alternatively activated macrophages. Front. Immunol. 2019, 10, 1084. [Google Scholar] [CrossRef]
  63. Grivennikov, S.I. Inflammation and colorectal cancer: Colitis-associated neoplasia. In Seminars in Immunopathology; Springer: Berlin/Heidelberg, Germany, 2013; pp. 229–244. [Google Scholar]
  64. Zhu, W.; Yu, J.; Nie, Y.; Shi, X.; Liu, Y.; Li, F.; Zhang, X.-l. Disequilibrium of M1 and M2 macrophages correlates with the development of experimental inflammatory bowel diseases. Immunol. Investig. 2014, 43, 638–652. [Google Scholar] [CrossRef]
  65. Wang, W.; Li, X.; Zheng, D.; Zhang, D.; Peng, X.; Zhang, X.; Ai, F.; Wang, X.; Ma, J.; Xiong, W.; et al. Dynamic changes and functions of macrophages and M1/M2 subpopulations during ulcerative colitis-associated carcinogenesis in an AOM/DSS mouse model. Mol. Med. Rep. 2015, 11, 2397–2406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Lee, Y.S.; Song, S.J.; Hong, H.K.; Oh, B.Y.; Lee, W.Y.; Cho, Y.B. The FBW7-MCL-1 axis is key in M1 and M2 macrophage-related colon cancer cell progression: Validating the immunotherapeutic value of targeting PI3Kgamma. Exp. Mol. Med. 2020, 52, 815–831. [Google Scholar] [CrossRef]
  67. Barbera-Guillem, E.; Nyhus, J.K.; Wolford, C.C.; Friece, C.R.; Sampsel, J.W. Vascular endothelial growth factor secretion by tumor-infiltrating macrophages essentially supports tumor angiogenesis, and IgG immune complexes potentiate the process. Cancer Res. 2002, 62, 7042–7049. [Google Scholar] [PubMed]
  68. Suarez-Lopez, L.; Sriram, G.; Kong, Y.W.; Morandell, S.; Merrick, K.A.; Hernandez, Y.; Haigis, K.M.; Yaffe, M.B. MK2 contributes to tumor progression by promoting M2 macrophage polarization and tumor angiogenesis. Proc. Natl. Acad. Sci. USA 2018, 115, E4236–E4244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Burmeister, K.; Quagliata, L.; Andreozzi, M.; Eppenberger-Castori, S.; Matter, M.S.; Perrina, V.; Grobholz, R.; Jochum, W.; Horber, D.; Moosmann, P.; et al. Vascular endothelial growth factor A amplification in colorectal cancer is associated with reduced M1 and M2 macrophages and diminished PD-1-expressing lymphocytes. PLoS ONE 2017, 12, e0175563. [Google Scholar] [CrossRef]
  70. Kaler, P.; Augenlicht, L.; Klampfer, L. Macrophage-derived IL-1β stimulates Wnt signaling and growth of colon cancer cells: A crosstalk interrupted by vitamin D 3. Oncogene 2009, 28, 3892–3902. [Google Scholar] [CrossRef] [Green Version]
  71. Cai, J.; Xia, L.; Li, J.; Ni, S.; Song, H.; Wu, X. Tumor-associated macrophages derived TGF-β-induced epithelial to mesenchymal transition in colorectal cancer cells through smad2, 3-4/snail signaling pathway. Cancer Res. Treat. Off. J. Korean Cancer Assoc. 2019, 51, 252. [Google Scholar]
  72. Illemann, M.; Bird, N.; Majeed, A.; Sehested, M.; Laerum, O.D.; Lund, L.R.; Danø, K.; Nielsen, B.S. MMP-9 Is Differentially Expressed in Primary Human Colorectal Adenocarcinomas and Their Metastases. Mol. Cancer Res. 2006, 4, 293–302. [Google Scholar] [CrossRef] [Green Version]
  73. Wei, C.; Yang, C.; Wang, S.; Shi, D.; Zhang, C.; Lin, X.; Liu, Q.; Dou, R.; Xiong, B. Crosstalk between cancer cells and tumor associated macrophages is required for mesenchymal circulating tumor cell-mediated colorectal cancer metastasis. Mol. Cancer 2019, 18, 64. [Google Scholar] [CrossRef]
  74. Afik, R.; Zigmond, E.; Vugman, M.; Klepfish, M.; Shimshoni, E.; Pasmanik-Chor, M.; Shenoy, A.; Bassat, E.; Halpern, Z.; Geiger, T. Tumor macrophages are pivotal constructors of tumor collagenous matrix. J. Exp. Med. 2016, 213, 2315–2331. [Google Scholar] [CrossRef] [PubMed]
  75. Vu, T.; Datta, P.K. Regulation of EMT in Colorectal Cancer: A Culprit in Metastasis. Cancers 2017, 9, 171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Yun, J.A.; Kim, S.H.; Hong, H.K.; Yun, S.H.; Kim, H.C.; Chun, H.K.; Cho, Y.B.; Lee, W.Y. Loss of E-Cadherin expression is associated with a poor prognosis in stage III colorectal cancer. Oncology 2014, 86, 318–328. [Google Scholar] [CrossRef] [PubMed]
  77. Wei, C.; Yang, C.; Wang, S.; Shi, D.; Zhang, C.; Lin, X.; Xiong, B. M2 macrophages confer resistance to 5-fluorouracil in colorectal cancer through the activation of CCL22/PI3K/AKT signaling. OncoTargets Ther. 2019, 12, 3051. [Google Scholar] [CrossRef] [Green Version]
  78. Zhong, X.; Chen, B.; Yang, Z. The Role of Tumor-Associated Macrophages in Colorectal Carcinoma Progression. Cell. Physiol. Biochem. 2018, 45, 356–365. [Google Scholar] [CrossRef]
  79. Herbeuval, J.-P.; Lelievre, E.; Lambert, C.; Dy, M.; Genin, C. Recruitment of STAT3 for production of IL-10 by colon carcinoma cells induced by macrophage-derived IL-6. J. Immunol. 2004, 172, 4630–4636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Cavnar, M.J.; Turcotte, S.; Katz, S.C.; Kuk, D.; Gönen, M.; Shia, J.; Allen, P.J.; Balachandran, V.P.; D’Angelica, M.I.; Kingham, T.P. Tumor-associated macrophage infiltration in colorectal cancer liver metastases is associated with better outcome. Ann. Surg. Oncol. 2017, 24, 1835–1842. [Google Scholar] [CrossRef] [PubMed]
  81. Wang, D.; Wang, X.; Si, M.; Yang, J.; Sun, S.; Wu, H.; Cui, S.; Qu, X.; Yu, X. Exosome-encapsulated miRNAs contribute to CXCL12/CXCR4-induced liver metastasis of colorectal cancer by enhancing M2 polarization of macrophages. Cancer Lett. 2020, 474, 36–52. [Google Scholar] [CrossRef]
  82. Wen, S.W.; Ager, E.I.; Christophi, C. Bimodal role of Kupffer cells during colorectal cancer liver metastasis. Cancer Biol. Ther. 2013, 14, 606–613. [Google Scholar] [CrossRef] [Green Version]
  83. García-Pérez, R.; Fábrega, J.F.; Varona-Bosque, A.; Martínez, C.M.; Revilla-Nuin, B.; Cabellos, L.; Pena, R.; Vilana, R.; Gonzalez-Abós, C.; García-Valdecasas, J.C. Role of Kupffer cells in the progression of CRC liver metastases after the first stage of ALPPS. Sci. Rep. 2018, 8, 8089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Pereira, A.A.L.; Rego, J.; Morris, V.; Overman, M.J.; Eng, C.; Garrett, C.R.; Boutin, A.; Ferrarotto, R.; Lee, M.; Jiang, Z. Association between KRAS mutation and lung metastasis in advanced colorectal cancer. Br. J. Cancer 2015, 112, 424–428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Liu, H.; Liang, Z.; Zhou, C.; Zeng, Z.; Wang, F.; Hu, T.; He, X.; Wu, X.; Wu, X.; Lan, P. Mutant KRAS triggers functional reprogramming of tumor-associated macrophages in colorectal cancer. Signal Transduct. Target. Ther. 2021, 6, 144. [Google Scholar] [CrossRef] [PubMed]
  86. Kuwahara, T.; Hazama, S.; Suzuki, N.; Yoshida, S.; Tomochika, S.; Nakagami, Y.; Matsui, H.; Shindo, Y.; Kanekiyo, S.; Tokumitsu, Y. Intratumoural-infiltrating CD4+ and FOXP3+ T cells as strong positive predictive markers for the prognosis of resectable colorectal cancer. Br. J. Cancer 2019, 121, 659–665. [Google Scholar] [CrossRef] [Green Version]
  87. Ahrends, T.; Spanjaard, A.; Pilzecker, B.; Babala, N.; Bovens, A.; Xiao, Y.; Jacobs, H.; Borst, J. CD4(+) T Cell Help Confers a Cytotoxic T Cell Effector Program Including Coinhibitory Receptor Downregulation and Increased Tissue Invasiveness. Immunity 2017, 47, 848–861. [Google Scholar] [CrossRef] [Green Version]
  88. Tosolini, M.; Kirilovsky, A.; Mlecnik, B.; Fredriksen, T.; Mauger, S.; Bindea, G.; Berger, A.; Bruneval, P.; Fridman, W.H.; Pages, F.; et al. Clinical impact of different classes of infiltrating T cytotoxic and helper cells (Th1, th2, treg, th17) in patients with colorectal cancer. Cancer Res. 2011, 71, 1263–1271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. De Simone, V.; Pallone, F.; Monteleone, G.; Stolfi, C. Role of TH17 cytokines in the control of colorectal cancer. Oncoimmunology 2013, 2, e26617. [Google Scholar] [CrossRef] [Green Version]
  90. Kroemer, M.; Turco, C.; Spehner, L.; Viot, J.; Idirène, I.; Bouard, A.; Renaude, E.; Deschamps, M.; Godet, Y.; Adotévi, O. Investigation of the prognostic value of CD4 T cell subsets expanded from tumor-infiltrating lymphocytes of colorectal cancer liver metastases. J. Immunother. Cancer 2020, 8, e001478. [Google Scholar] [CrossRef]
  91. Pedroza-Gonzalez, A.; Verhoef, C.; Ijzermans, J.N.; Peppelenbosch, M.P.; Kwekkeboom, J.; Verheij, J.; Janssen, H.L.; Sprengers, D. Activated tumor-infiltrating CD4+ regulatory T cells restrain antitumor immunity in patients with primary or metastatic liver cancer. Hepatology 2013, 57, 183–194. [Google Scholar] [CrossRef]
  92. Katz, S.C.; Pillarisetty, V.; Bamboat, Z.M.; Shia, J.; Hedvat, C.; Gonen, M.; Jarnagin, W.; Fong, Y.; Blumgart, L.; D’Angelica, M. T cell infiltrate predicts long-term survival following resection of colorectal cancer liver metastases. Ann. Surg. Oncol. 2009, 16, 2524–2530. [Google Scholar] [CrossRef]
  93. Katz, S.C.; Pillarisetty, V.G.; Bleier, J.I.; Kingham, T.P.; Chaudhry, U.I.; Shah, A.B.; DeMatteo, R.P. Conventional liver CD4 T cells are functionally distinct and suppressed by environmental factors. Hepatology 2005, 42, 293–300. [Google Scholar] [CrossRef]
  94. DeNardo, D.G.; Barreto, J.B.; Andreu, P.; Vasquez, L.; Tawfik, D.; Kolhatkar, N.; Coussens, L.M. CD4+ T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell 2009, 16, 91–102. [Google Scholar] [CrossRef] [Green Version]
  95. Kobie, J.J.; Wu, R.S.; Kurt, R.A.; Lou, S.; Adelman, M.K.; Whitesell, L.J.; Ramanathapuram, L.V.; Arteaga, C.L.; Akporiaye, E.T. Transforming growth factor β inhibits the antigen-presenting functions and antitumor activity of dendritic cell vaccines. Cancer Res. 2003, 63, 1860–1864. [Google Scholar] [PubMed]
  96. Pryczynicz, A.; Cepowicz, D.; Zaręba, K.; Gryko, M.; Hołody-Zaręba, J.; Kędra, B.; Kemona, A.; Guzińska-Ustymowicz, K. Dysfunctions in the mature dendritic cells are associated with the presence of metastases of colorectal cancer in the surrounding lymph nodes. Gastroenterol. Res. Pract. 2016, 2016, 1–5. [Google Scholar] [CrossRef] [PubMed]
  97. Gulubova, M.V.; Ananiev, J.R.; Vlaykova, T.I.; Yovchev, Y.; Tsoneva, V.; Manolova, I.M. Role of dendritic cells in progression and clinical outcome of colon cancer. Int. J. Color. Dis. 2012, 27, 159–169. [Google Scholar] [CrossRef] [PubMed]
  98. Orsini, G.; Legitimo, A.; Failli, A.; Ferrari, P.; Nicolini, A.; Spisni, R.; Miccoli, P.; Consolini, R. Defective generation and maturation of dendritic cells from monocytes in colorectal cancer patients during the course of disease. Int. J. Mol. Sci. 2013, 14, 22022–22041. [Google Scholar] [CrossRef] [Green Version]
  99. Nagorsen, D.; Voigt, S.; Berg, E.; Stein, H.; Thiel, E.; Loddenkemper, C. Tumor-infiltrating macrophages and dendritic cells in human colorectal cancer: Relation to local regulatory T cells, systemic T-cell response against tumor-associated antigens and survival. J. Transl. Med. 2007, 5, 62. [Google Scholar] [CrossRef] [Green Version]
  100. Hsu, Y.-L.; Chen, Y.-J.; Chang, W.-A.; Jian, S.-F.; Fan, H.-L.; Wang, J.-Y.; Kuo, P.-L. Interaction between tumor-associated dendritic cells and colon cancer cells contributes to tumor progression via CXCL1. Int. J. Mol. Sci. 2018, 19, 2427. [Google Scholar] [CrossRef] [Green Version]
  101. Legitimo, A.; Consolini, R.; Failli, A.; Orsini, G.; Spisni, R. Dendritic cell defects in the colorectal cancer. Hum. Vaccines Immunother. 2014, 10, 3224–3235. [Google Scholar] [CrossRef] [Green Version]
  102. Xu, F.; Ye, Y.-J.; Liu, W.; Kong, M.; He, Y.; Wang, S. Dendritic cell/tumor hybrids enhances therapeutic efficacy against colorectal cancer liver metastasis in SCID mice. Scand. J. Gastroenterol. 2010, 45, 707–713. [Google Scholar] [CrossRef] [PubMed]
  103. Miyagawa, S.; Soeda, J.; Takagi, S.; Miwa, S.; Ichikawa, E.; Noike, T. Prognostic significance of mature dendritic cells and factors associated with their accumulation in metastatic liver tumors from colorectal cancer. Hum. Pathol. 2004, 35, 1392–1396. [Google Scholar] [CrossRef] [PubMed]
  104. Rodriguez, J.; Castañón, E.; Perez-Gracia, J.L.; Rodriguez, I.; Viudez, A.; Alfaro, C.; Oñate, C.; Perez, G.; Rotellar, F.; Inogés, S. A randomized phase II clinical trial of dendritic cell vaccination following complete resection of colon cancer liver metastasis. J. Immunother. Cancer 2018, 6, 96. [Google Scholar] [CrossRef] [Green Version]
  105. Urosevic, J.; Garcia-Albéniz, X.; Planet, E.; Real, S.; Céspedes, M.V.; Guiu, M.; Fernandez, E.; Bellmunt, A.; Gawrzak, S.; Pavlovic, M. Colon cancer cells colonize the lung from established liver metastases through p38 MAPK signalling and PTHLH. Nat. Cell Biol. 2014, 16, 685–694. [Google Scholar] [CrossRef]
  106. Zhao, S.; Jiang, T.; Zhang, L.; Yang, H.; Liu, X.; Jia, Y.; Zhou, C. Clinicopathological and prognostic significance of regulatory T cells in patients with non-small cell lung cancer: A systematic review with meta-analysis. Oncotarget 2016, 7, 36065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Tang, Y.; Xu, X.; Guo, S.; Zhang, C.; Tang, Y.; Tian, Y.; Ni, B.; Lu, B.; Wang, H. An increased abundance of tumor-infiltrating regulatory T cells is correlated with the progression and prognosis of pancreatic ductal adenocarcinoma. PLoS ONE 2014, 9, e91551. [Google Scholar] [CrossRef] [Green Version]
  108. Merlo, A.; Casalini, P.; Carcangiu, M.L.; Malventano, C.; Triulzi, T.; Mènard, S.; Tagliabue, E.; Balsari, A. FOXP3 expression and overall survival in breast cancer. J. Clin. Oncol. 2009, 27, 1746–1752. [Google Scholar] [CrossRef] [PubMed]
  109. Olguín, J.E.; Medina-Andrade, I.; Rodríguez, T.; Rodríguez-Sosa, M.; Terrazas, L.I. Relevance of regulatory T Cells during colorectal cancer development. Cancers 2020, 12, 1888. [Google Scholar] [CrossRef]
  110. Ji, D.; Song, C.; Li, Y.; Xia, J.; Wu, Y.; Jia, J.; Cui, X.; Yu, S.; Gu, J. Combination of radiotherapy and suppression of Tregs enhances abscopal antitumor effect and inhibits metastasis in rectal cancer. J. Immunother. Cancer 2020, 8, e000826. [Google Scholar] [CrossRef]
  111. Salama, P.; Phillips, M.; Grieu, F.; Morris, M.; Zeps, N.; Joseph, D.; Platell, C.; Iacopetta, B. Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer. J. Clin. Oncol. 2009, 27, 186–192. [Google Scholar] [CrossRef] [PubMed]
  112. Saito, T.; Nishikawa, H.; Wada, H.; Nagano, Y.; Sugiyama, D.; Atarashi, K.; Maeda, Y.; Hamaguchi, M.; Ohkura, N.; Sato, E. Two FOXP3+ CD4+ T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat. Med. 2016, 22, 679–684. [Google Scholar] [CrossRef] [PubMed]
  113. Katz, S.C.; Bamboat, Z.M.; Maker, A.V.; Shia, J.; Pillarisetty, V.G.; Yopp, A.C.; Hedvat, C.V.; Gonen, M.; Jarnagin, W.R.; Fong, Y. Regulatory T cell infiltration predicts outcome following resection of colorectal cancer liver metastases. Ann. Surg. Oncol. 2013, 20, 946–955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Wang, L.; Hu, X.; Xu, Y.; Liu, Z. Arsenic trioxide inhibits lung metastasis of mouse colon cancer via reducing the infiltration of regulatory T cells. Tumour Biol. 2016, 37, 15165–15173. [Google Scholar] [CrossRef] [Green Version]
  115. Huppert, L.A.; Green, M.D.; Kim, L.; Chow, C.; Leyfman, Y.; Daud, A.I.; Lee, J.C. Tissue-specific tregs in cancer metastasis: Opportunities for precision immunotherapy. Cell. Mol. Immunol. 2021, 1–13. [Google Scholar] [CrossRef]
  116. Ellis, S.; Lin, E.J.; Tartar, D. Immunology of wound healing. Curr. Dermatol. Rep. 2018, 7, 350–358. [Google Scholar] [CrossRef] [Green Version]
  117. Mazaki, J.; Katsumata, K.; Kasahara, K.; Tago, T.; Wada, T.; Kuwabara, H.; Enomoto, M.; Ishizaki, T.; Nagakawa, Y.; Tsuchida, A. Neutrophil-to-lymphocyte ratio is a prognostic factor for colon cancer: A propensity score analysis. BMC Cancer 2020, 20, 922. [Google Scholar] [CrossRef]
  118. Fridlender, Z.G.; Sun, J.; Kim, S.; Kapoor, V.; Cheng, G.; Ling, L.; Worthen, G.S.; Albelda, S.M. Polarization of tumor-associated neutrophil phenotype by TGF-β:“N1” versus “N2” TAN. Cancer Cell 2009, 16, 183–194. [Google Scholar] [CrossRef] [Green Version]
  119. Itatani, Y.; Yamamoto, T.; Zhong, C.; Molinolo, A.A.; Ruppel, J.; Hegde, P.; Taketo, M.M.; Ferrara, N. Suppressing neutrophil-dependent angiogenesis abrogates resistance to anti-VEGF antibody in a genetic model of colorectal cancer. Proc. Natl. Acad. Sci. USA 2020, 117, 21598–21608. [Google Scholar] [CrossRef]
  120. Germann, M.; Zangger, N.; Sauvain, M.O.; Sempoux, C.; Bowler, A.D.; Wirapati, P.; Kandalaft, L.E.; Delorenzi, M.; Tejpar, S.; Coukos, G. Neutrophils suppress tumor-infiltrating T cells in colon cancer via matrix metalloproteinase-mediated activation of TGF β. EMBO Mol. Med. 2020, 12, e10681. [Google Scholar] [CrossRef]
  121. Uribe-Querol, E.; Rosales, C. Neutrophils in Cancer: Two Sides of the Same Coin. J. Immunol. Res. 2015, 2015, 983698. [Google Scholar] [CrossRef] [Green Version]
  122. Gordon-Weeks, A.N.; Lim, S.Y.; Yuzhalin, A.E.; Jones, K.; Markelc, B.; Kim, K.J.; Buzzelli, J.N.; Fokas, E.; Cao, Y.; Smart, S. Neutrophils promote hepatic metastasis growth through fibroblast growth factor 2–dependent angiogenesis in mice. Hepatology 2017, 65, 1920–1935. [Google Scholar] [CrossRef]
  123. Yang, L.; Liu, L.; Zhang, R.; Hong, J.; Wang, Y.; Wang, J.; Zuo, J.; Zhang, J.; Chen, J.; Hao, H. IL-8 mediates a positive loop connecting increased neutrophil extracellular traps (NETs) and colorectal cancer liver metastasis. J. Cancer 2020, 11, 4384–4396. [Google Scholar] [CrossRef]
  124. Yamamoto, T.; Kawada, K.; Itatani, Y.; Inamoto, S.; Okamura, R.; Iwamoto, M.; Miyamoto, E.; Chen-Yoshikawa, T.F.; Hirai, H.; Hasegawa, S.; et al. Loss of SMAD4 Promotes Lung Metastasis of Colorectal Cancer by Accumulation of CCR1+ Tumor-Associated Neutrophils through CCL15-CCR1 Axis. Clin. Cancer Res. 2017, 23, 833–844. [Google Scholar] [CrossRef] [Green Version]
  125. Zhao, M.; Mishra, L.; Deng, C.X. The role of TGF-beta/SMAD4 signaling in cancer. Int. J. Biol. Sci. 2018, 14, 111–123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Lin, L.H.; Chang, K.W.; Cheng, H.W.; Liu, C.J. SMAD4 Somatic Mutations in Head and Neck Carcinoma Are Associated With Tumor Progression. Front. Oncol. 2019, 9, 1379. [Google Scholar] [CrossRef] [Green Version]
  127. Liu, J.; Cho, S.N.; Akkanti, B.; Jin, N.; Mao, J.; Long, W.; Chen, T.; Zhang, Y.; Tang, X.; Wistub, I.; et al. ErbB2 Pathway Activation upon Smad4 Loss Promotes Lung Tumor Growth and Metastasis. Cell Rep. 2015, 10, 1599–1613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Jablonska, J.; Lang, S.; Sionov, R.V.; Granot, Z. The regulation of pre-metastatic niche formation by neutrophils. Oncotarget 2017, 8, 112132–112144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Wu, C.F.; Andzinski, L.; Kasnitz, N.; Kroger, A.; Klawonn, F.; Lienenklaus, S.; Weiss, S.; Jablonska, J. The lack of type I interferon induces neutrophil-mediated pre-metastatic niche formation in the mouse lung. Int. J. Cancer 2015, 137, 837–847. [Google Scholar] [CrossRef] [PubMed]
  130. Kowanetz, M.; Wu, X.; Lee, J.; Tan, M.; Hagenbeek, T.; Qu, X.; Yu, L.; Ross, J.; Korsisaari, N.; Cao, T.; et al. Granulocyte-colony stimulating factor promotes lung metastasis through mobilization of Ly6G+Ly6C+ granulocytes. Proc. Natl. Acad. Sci. USA 2010, 107, 21248–21255. [Google Scholar] [CrossRef] [Green Version]
  131. Glaire, M.A.; Domingo, E.; Sveen, A.; Bruun, J.; Nesbakken, A.; Nicholson, G.; Novelli, M.; Lawson, K.; Oukrif, D.; Kildal, W. Tumour-infiltrating CD8+ lymphocytes and colorectal cancer recurrence by tumour and nodal stage. Br. J. Cancer 2019, 121, 474–482. [Google Scholar] [CrossRef]
  132. Angell, H.K.; Bruni, D.; Barrett, J.C.; Herbst, R.; Galon, J. The immunoscore: Colon cancer and beyond. Clin. Cancer Res. 2020, 26, 332–339. [Google Scholar] [CrossRef] [Green Version]
  133. Pagès, F.; Mlecnik, B.; Marliot, F.; Bindea, G.; Ou, F.-S.; Bifulco, C.; Lugli, A.; Zlobec, I.; Rau, T.T.; Berger, M.D. International validation of the consensus Immunoscore for the classification of colon cancer: A prognostic and accuracy study. Lancet 2018, 391, 2128–2139. [Google Scholar] [CrossRef]
  134. O’Malley, G.; Treacy, O.; Lynch, K.; Naicker, S.D.; Leonard, N.A.; Lohan, P.; Dunne, P.D.; Ritter, T.; Egan, L.J.; Ryan, A.E. Stromal cell PD-L1 inhibits CD8+ T-cell antitumor immune responses and promotes colon cancer. Cancer Immunol. Res. 2018, 6, 1426–1441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Shan, T.; Chen, S.; Wu, T.; Yang, Y.; Li, S.; Chen, X. PD-L1 expression in colon cancer and its relationship with clinical prognosis. Int. J. Clin. Exp. Pathol. 2019, 12, 1764. [Google Scholar] [PubMed]
  136. Zhao, T.; Li, Y.; Zhang, J.; Zhang, B. PD-L1 expression increased by IFN-γ via JAK2-STAT1 signaling and predicts a poor survival in colorectal cancer. Oncol. Lett. 2020, 20, 1127–1134. [Google Scholar] [CrossRef] [PubMed]
  137. Elfishawy, M.; Abd, E.S.A.; Hegazy, A.; El-Yasergy, D.F. Immunohistochemical Expression of Programmed Death Ligand-1 (PDL-1) in Colorectal carcinoma and Its Correlation with Stromal Tumor Infiltrating Lymphocytes. Asian Pac. J. Cancer Prev. 2020, 21, 225–232. [Google Scholar] [CrossRef] [Green Version]
  138. FDA Approves First-Line Immunotherapy for Patients with MSI-H/dMMR Metastatic Colorectal Cancer. Available online: https://www.fda.gov/news-events/press-announcements/fda-approves-first-line-immunotherapy-patients-msi-hdmmr-metastatic-colorectal-cancer (accessed on 1 October 2021).
  139. Thomas, D.A.; Massagué, J. TGF-β directly targets cytotoxic T cell functions during tumor evasion of immune surveillance. Cancer Cell 2005, 8, 369–380. [Google Scholar] [CrossRef] [Green Version]
  140. Chang, L.-Y.; Lin, Y.-C.; Mahalingam, J.; Huang, C.-T.; Chen, T.-W.; Kang, C.-W.; Peng, H.-M.; Chu, Y.-Y.; Chiang, J.-M.; Dutta, A. Tumor-derived chemokine CCL5 enhances TGF-β–mediated killing of CD8+ T cells in colon cancer by T-regulatory cells. Cancer Res. 2012, 72, 1092–1102. [Google Scholar] [CrossRef] [Green Version]
  141. Remark, R.; Alifano, M.; Cremer, I.; Lupo, A.; Dieu-Nosjean, M.-C.; Riquet, M.; Crozet, L.; Ouakrim, H.; Goc, J.; Cazes, A. Characteristics and clinical impacts of the immune environments in colorectal and renal cell carcinoma lung metastases: Influence of tumor origin. Clin. Cancer Res. 2013, 19, 4079–4091. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Frantz, C.; Stewart, K.M.; Weaver, V.M. The extracellular matrix at a glance. J. Cell Sci. 2010, 123, 4195–4200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Li, Z.L.; Wang, Z.J.; Wei, G.H.; Yang, Y.; Wang, X.W. Changes in extracellular matrix in different stages of colorectal cancer and their effects on proliferation of cancer cells. World J. Gastrointest. Oncol. 2020, 12, 267–275. [Google Scholar] [CrossRef]
  144. Yang, B.; Tang, F.; Zhang, B.; Zhao, Y.; Feng, J.; Rao, Z. Matrix metalloproteinase-9 overexpression is closely related to poor prognosis in patients with colon cancer. World J. Surg. Oncol. 2014, 12, 24. [Google Scholar] [CrossRef] [Green Version]
  145. Dong, W.; Li, H.; Zhang, Y.; Yang, H.; Guo, M.; Li, L.; Liu, T. Matrix metalloproteinase 2 promotes cell growth and invasion in colorectal cancer. Acta Biochim. Biophys. Sin. 2011, 43, 840–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Sis, B.; Sagol, O.; Kupelioglu, A.; Sokmen, S.; Terzi, C.; Fuzun, M.; Ozer, E.; Bishop, P. Prognostic significance of matrix metalloproteinase-2, cathepsin D, and tenascin-C expression in colorectal carcinoma. Pathol. Res. Prat. 2004, 200, 379–387. [Google Scholar] [CrossRef] [PubMed]
  147. Zeng, Z.S.; Cohen, A.M.; Guillem, J.G. Loss of basement membrane type IV collagen is associated with increased expression of metalloproteinases 2 and 9 (MMP-2 and MMP-9) during human colorectal tumorigenesis. Carcinogenesis 1999, 20, 749–755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Sand, J.M.; Larsen, L.; Hogaboam, C.; Martinez, F.; Han, M.; Rossel Larsen, M.; Nawrocki, A.; Zheng, Q.; Karsdal, M.A.; Leeming, D.J. MMP mediated degradation of type IV collagen alpha 1 and alpha 3 chains reflects basement membrane remodeling in experimental and clinical fibrosis--validation of two novel biomarker assays. PLoS ONE 2013, 8, e84934. [Google Scholar] [CrossRef]
  149. Brauchle, E.; Kasper, J.; Daum, R.; Schierbaum, N.; Falch, C.; Kirschniak, A.; Schaffer, T.E.; Schenke-Layland, K. Biomechanical and biomolecular characterization of extracellular matrix structures in human colon carcinomas. Matrix Biol. 2018, 68–69, 180–193. [Google Scholar] [CrossRef] [PubMed]
  150. Bauer, J.; Emon, M.A.B.; Staudacher, J.J.; Thomas, A.L.; Zessner-Spitzenberg, J.; Mancinelli, G.; Krett, N.; Saif, M.T.; Jung, B. Increased stiffness of the tumor microenvironment in colon cancer stimulates cancer associated fibroblast-mediated prometastatic activin A signaling. Sci. Rep. 2020, 10, 50. [Google Scholar] [CrossRef] [PubMed]
  151. Tanjore, H.; Kalluri, R. The role of type IV collagen and basement membranes in cancer progression and metastasis. Am. J. Pathol. 2006, 168, 715–717. [Google Scholar] [CrossRef] [Green Version]
  152. Naba, A.; Clauser, K.R.; Whittaker, C.A.; Carr, S.A.; Tanabe, K.K.; Hynes, R.O. Extracellular matrix signatures of human primary metastatic colon cancers and their metastases to liver. BMC Cancer 2014, 14, 518. [Google Scholar] [CrossRef] [Green Version]
  153. Yuzhalin, A.E.; Gordon-Weeks, A.N.; Tognoli, M.L.; Jones, K.; Markelc, B.; Konietzny, R.; Fischer, R.; Muth, A.; O’Neill, E.; Thompson, P.R.; et al. Colorectal cancer liver metastatic growth depends on PAD4-driven citrullination of the extracellular matrix. Nat. Commun. 2018, 9, 4783. [Google Scholar] [CrossRef]
  154. Ganguly, D.; Chandra, R.; Karalis, J.; Teke, M.; Aguilera, T.; Maddipati, R.; Wachsmann, M.B.; Ghersi, D.; Siravegna, G.; Zeh, H.J. Cancer-Associated Fibroblasts: Versatile Players in the Tumor Microenvironment. Cancers 2020, 12, 2652. [Google Scholar] [CrossRef]
  155. Garvey, C.M.; Lau, R.; Sanchez, A.; Sun, R.X.; Fong, E.J.; Doche, M.E.; Chen, O.; Jusuf, A.; Lenz, H.-J.; Larson, B. Anti-EGFR Therapy Induces EGF Secretion by Cancer-Associated Fibroblasts to Confer Colorectal Cancer Chemoresistance. Cancers 2020, 12, 1393. [Google Scholar] [CrossRef]
  156. Chan, T.S.; Shaked, Y.; Tsai, K.K. Targeting the Interplay between Cancer Fibroblasts, Mesenchymal Stem Cells, and Cancer Stem Cells in Desmoplastic Cancers. Front. Oncol. 2019, 9, 688. [Google Scholar] [CrossRef] [Green Version]
  157. Jung, Y.; Kim, J.K.; Shiozawa, Y.; Wang, J.; Mishra, A.; Joseph, J.; Berry, J.E.; McGee, S.; Lee, E.; Sun, H.; et al. Recruitment of mesenchymal stem cells into prostate tumours promotes metastasis. Nat. Commun. 2013, 4, 1795. [Google Scholar] [CrossRef] [Green Version]
  158. Mishra, P.J.; Mishra, P.J.; Humeniuk, R.; Medina, D.J.; Alexe, G.; Mesirov, J.P.; Ganesan, S.; Glod, J.W.; Banerjee, D. Carcinoma-associated fibroblast-like differentiation of human mesenchymal stem cells. Cancer Res. 2008, 68, 4331–4339. [Google Scholar] [CrossRef] [Green Version]
  159. Rynne-Vidal, A.; Jimenez-Heffernan, J.A.; Fernandez-Chacon, C.; Lopez-Cabrera, M.; Sandoval, P. The Mesothelial Origin of Carcinoma Associated-Fibroblasts in Peritoneal Metastasis. Cancers 2015, 7, 1994–2011. [Google Scholar] [CrossRef] [Green Version]
  160. Li, H.; Courtois, E.T.; Sengupta, D.; Tan, Y.; Chen, K.H.; Goh, J.J.L.; Kong, S.L.; Chua, C.; Hon, L.K.; Tan, W.S. Reference component analysis of single-cell transcriptomes elucidates cellular heterogeneity in human colorectal tumors. Nat. Genet. 2017, 49, 708–718. [Google Scholar] [CrossRef]
  161. Wang, L.; Yang, J.; Huang, J.; Wen, Z.-Q.; Xu, N.; Liu, X.; Zhang, J.-H.; Li, W.-L. MicroRNA expression profile in the N2 phenotype neutrophils of colorectal cancer and screen of putative key mircRNAs. Cancer Manag Res. 2020, 12, 5491–5503. [Google Scholar] [CrossRef]
  162. Tauriello, D.V.; Palomo-Ponce, S.; Stork, D.; Berenguer-Llergo, A.; Badia-Ramentol, J.; Iglesias, M.; Sevillano, M.; Ibiza, S.; Cañellas, A.; Hernando-Momblona, X. TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 2018, 554, 538–543. [Google Scholar] [CrossRef] [Green Version]
  163. Zhang, R.; Qi, F.; Zhao, F.; Li, G.; Shao, S.; Zhang, X.; Yuan, L.; Feng, Y. Cancer-associated fibroblasts enhance tumor-associated macrophages enrichment and suppress NK cells function in colorectal cancer. Cell Death Dis. 2019, 10, 273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Zhuang, J.; Lu, Q.; Shen, B.; Huang, X.; Shen, L.; Zheng, X.; Huang, R.; Yan, J.; Guo, H. TGFbeta1 secreted by cancer-associated fibroblasts induces epithelial-mesenchymal transition of bladder cancer cells through lncRNA-ZEB2NAT. Sci. Rep. 2015, 5, 11924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. 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-beta signalling. Br. J. Cancer 2014, 110, 724–732. [Google Scholar] [CrossRef] [Green Version]
  166. You, J.; Li, M.; Cao, L.M.; Gu, Q.H.; Deng, P.B.; Tan, Y.; Hu, C.P. Snail1-dependent cancer-associated fibroblasts induce epithelial-mesenchymal transition in lung cancer cells via exosomes. QJM Int. J. Med. 2019, 112, 581–590. [Google Scholar] [CrossRef]
  167. Xuefeng, X.; Hou, M.X.; Yang, Z.W.; Agudamu, A.; Wang, F.; Su, X.L.; Li, X.; Shi, L.; Terigele, T.; Bao, L.L.; et al. Epithelial-mesenchymal transition and metastasis of colon cancer cells induced by the FAK pathway in cancer-associated fibroblasts. J. Int. Med. Res. 2020, 48, 300060520931242. [Google Scholar] [CrossRef] [PubMed]
  168. Shin, N.; Son, G.M.; Shin, D.H.; Kwon, M.S.; Park, B.S.; Kim, H.S.; Ryu, D.; Kang, C.D. Cancer-Associated Fibroblasts and Desmoplastic Reactions Related to Cancer Invasiveness in Patients With Colorectal Cancer. Ann. Coloproctol. 2019, 35, 36–46. [Google Scholar] [CrossRef] [Green Version]
  169. Hawinkels, L.; Paauwe, M.; Verspaget, H.; Wiercinska, E.; Van Der Zon, J.; Van Der Ploeg, K.; Koelink, P.; Lindeman, J.; Mesker, W.; Ten Dijke, P. Interaction with colon cancer cells hyperactivates TGF-β signaling in cancer-associated fibroblasts. Oncogene 2014, 33, 97–107. [Google Scholar] [CrossRef] [Green Version]
  170. Torres, S.; Garcia-Palmero, I.; Herrera, M.; Bartolome, R.A.; Pena, C.; Fernandez-Acenero, M.J.; Padilla, G.; Pelaez-Garcia, A.; Lopez-Lucendo, M.; Rodriguez-Merlo, R.; et al. LOXL2 Is Highly Expressed in Cancer-Associated Fibroblasts and Associates to Poor Colon Cancer Survival. Clin. Cancer Res. 2015, 21, 4892–4902. [Google Scholar] [CrossRef] [Green Version]
  171. 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. Br. J. Cancer 2014, 110, 469–478. [Google Scholar] [CrossRef] [Green Version]
  172. Suetsugu, A.; Osawa, Y.; Nagaki, M.; Saji, S.; Moriwaki, H.; Bouvet, M.; Hoffman, R.M. Imaging the recruitment of cancer-associated fibroblasts by liver-metastatic colon cancer. J. Cell. Biochem. 2011, 112, 949–953. [Google Scholar] [CrossRef]
  173. Tsushima, H.; Ito, N.; Tamura, S.; Matsuda, Y.; Inada, M.; Yabuuchi, I.; Imai, Y.; Nagashima, R.; Misawa, H.; Takeda, H. Circulating transforming growth factor β1 as a predictor of liver metastasis after resection in colorectal cancer. Clin. Cancer Res. 2001, 7, 1258–1262. [Google Scholar]
  174. Calon, A.; Espinet, E.; Palomo-Ponce, S.; Tauriello, D.V.; Iglesias, M.; Cespedes, M.V.; Sevillano, M.; Nadal, C.; Jung, P.; Zhang, X.H.; et al. Dependency of colorectal cancer on a TGF-beta-driven program in stromal cells for metastasis initiation. Cancer Cell 2012, 22, 571–584. [Google Scholar] [CrossRef] [Green Version]
  175. Zhang, R.; Qi, F.; Shao, S.; Li, G.; Feng, Y. Human colorectal cancer-derived carcinoma associated fibroblasts promote CD44-mediated adhesion of colorectal cancer cells to endothelial cells by secretion of HGF. Cancer Cell Int. 2019, 19, 192. [Google Scholar] [CrossRef]
  176. Matsusue, R.; Kubo, H.; Hisamori, S.; Okoshi, K.; Takagi, H.; Hida, K.; Nakano, K.; Itami, A.; Kawada, K.; Nagayama, S. Hepatic stellate cells promote liver metastasis of colon cancer cells by the action of SDF-1/CXCR4 axis. Ann. Surg. Oncol. 2009, 16, 2645–2653. [Google Scholar] [CrossRef] [Green Version]
  177. Mueller, L.; Goumas, F.A.; Affeldt, M.; Sandtner, S.; Gehling, U.M.; Brilloff, S.; Walter, J.; Karnatz, N.; Lamszus, K.; Rogiers, X. Stromal fibroblasts in colorectal liver metastases originate from resident fibroblasts and generate an inflammatory microenvironment. Am. J. Pathol. 2007, 171, 1608–1618. [Google Scholar] [CrossRef] [Green Version]
  178. Kwak, Y.; Lee, H.E.; Kim, W.H.; Kim, D.-W.; Kang, S.-B.; Lee, H.S. The clinical implication of cancer-associated microvasculature and fibroblast in advanced colorectal cancer patients with synchronous or metachronous metastases. PLoS ONE 2014, 9, e91811. [Google Scholar] [CrossRef]
  179. Schweiger, T.; Nikolowsky, C.; Starlinger, P.; Traxler, D.; Zimmermann, M.; Birner, P.; Hegedüs, B.; Dome, B.; Bergmann, M.; Mildner, M. Stromal expression of heat-shock protein 27 is associated with worse clinical outcome in patients with colorectal cancer lung metastases. PLoS ONE 2015, 10, e0120724. [Google Scholar] [CrossRef] [Green Version]
  180. Bevacizumab. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/125085s301lbl.pdf (accessed on 10 September 2021).
  181. Ramucirumab. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/125477s034lbl.pdf (accessed on 10 September 2021).
  182. Chen, W.Z.; Jiang, J.X.; Yu, X.Y.; Xia, W.J.; Yu, P.X.; Wang, K.; Zhao, Z.Y.; Chen, Z.G. Endothelial cells in colorectal cancer. World J. Gastrointest. Oncol. 2019, 11, 946–956. [Google Scholar] [CrossRef] [PubMed]
  183. Quiroz-Reyes, A.G.; Islas, J.F.; Delgado-Gonzalez, P.; Franco-Villarreal, H.; Garza-Trevino, E.N. Therapeutic Approaches for Metastases from Colorectal Cancer and Pancreatic Ductal Carcinoma. Pharmaceutics 2021, 13, 103. [Google Scholar] [CrossRef]
  184. Hida, K.; Maishi, N.; Annan, D.A.; Hida, Y. Contribution of Tumor Endothelial Cells in Cancer Progression. Int. J. Mol. Sci. 2018, 19, 1272. [Google Scholar] [CrossRef] [Green Version]
  185. Zhang, D.; Bi, J.; Liang, Q.; Wang, S.; Zhang, L.; Han, F.; Li, S.; Qiu, B.; Fan, X.; Chen, W.; et al. VCAM1 Promotes Tumor Cell Invasion and Metastasis by Inducing EMT and Transendothelial Migration in Colorectal Cancer. Front. Oncol. 2020, 10, 1066. [Google Scholar] [CrossRef]
  186. Cima, I.; Kong, S.L.; Sengupta, D.; Tan, I.B.; Phyo, W.M.; Lee, D.; Hu, M.; Iliescu, C.; Alexander, I.; Goh, W.L.; et al. Tumor-derived circulating endothelial cell clusters in colorectal cancer. Sci. Transl. Med. 2016, 8, 345ra389. [Google Scholar] [CrossRef] [Green Version]
  187. Braet, F.; Nagatsuma, K.; Saito, M.; Soon, L.; Wisse, E.; Matsuura, T. The hepatic sinusoidal endothelial lining and colorectal liver metastases. World J. Gastroenterol. 2007, 13, 821–825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Zuo, Y.; Ren, S.; Wang, M.; Liu, B.; Yang, J.; Kuai, X.; Lin, C.; Zhao, D.; Tang, L.; He, F. Novel roles of liver sinusoidal endothelial cell lectin in colon carcinoma cell adhesion, migration and in-vivo metastasis to the liver. Gut 2013, 62, 1169–1178. [Google Scholar] [CrossRef] [PubMed]
  189. Arteta, B.; Lasuen, N.; Lopategi, A.; Sveinbjornsson, B.; Smedsrod, B.; Vidal-Vanaclocha, F. Colon carcinoma cell interaction with liver sinusoidal endothelium inhibits organ-specific antitumor immunity through interleukin-1-induced mannose receptor in mice. Hepatology 2010, 51, 2172–2182. [Google Scholar] [CrossRef]
  190. Zeng, X.; Ward, S.; Zhou, J.; Cheng, A. Liver Immune Microenvironment and Metastasis from Colorectal Cancer-Pathogenesis and Therapeutic Perspectives. Cancers 2021, 13, 2418. [Google Scholar] [CrossRef]
  191. Gao, W.; Chen, L.; Ma, Z.; Du, Z.; Zhao, Z.; Hu, Z.; Li, Q. Isolation and phenotypic characterization of colorectal cancer stem cells with organ-specific metastatic potential. Gastroenterology 2013, 145, 636–646 e635. [Google Scholar] [CrossRef]
  192. Tamura, M.; Oda, M.; Tsunezuka, Y.; Matsumoto, I.; Kawakami, K.; Watanabe, G. Vascular endothelial growth factor expression in metastatic pulmonary tumor from colorectal carcinoma: Utility as a prognostic factor. J. Thorac. Cardiovasc. Surg. 2004, 128, 517–522. [Google Scholar] [CrossRef] [Green Version]
  193. Cao, C.; Wang, D.; Tian, D.H.; Wilson-Smith, A.; Huang, J.; Rimner, A. A systematic review and meta-analysis of stereotactic body radiation therapy for colorectal pulmonary metastases. J. Thorac. Dis. 2019, 11, 5187. [Google Scholar] [CrossRef]
  194. Ahmed, K.A.; Scott, J.G.; Arrington, J.A.; Naghavi, A.O.; Grass, G.D.; Perez, B.A.; Caudell, J.J.; Berglund, A.E.; Welsh, E.A.; Eschrich, S.A. Radiosensitivity of lung metastases by primary histology and implications for stereotactic body radiation therapy using the genomically adjusted radiation dose. J. Thorac. Oncol. 2018, 13, 1121–1127. [Google Scholar] [CrossRef] [Green Version]
  195. Chen, Y.; Gao, M.; Huang, Z.; Yu, J.; Meng, X. SBRT combined with PD-1/PD-L1 inhibitors in NSCLC treatment: A focus on the mechanisms, advances, and future challenges. J. Hematol. Oncol. 2020, 13, 105. [Google Scholar] [CrossRef]
  196. ClinicalTrials.gov. M7824 in Patients with Metastatic Colorectal Cancer or with Advanced Solid Tumors with Microsatellite Instability. Available online: https://clinicaltrials.gov/ct2/show/NCT03436563 (accessed on 30 September 2021).
  197. ClinicalTrials.gov. PI Pembro in Combination with Stereotactic Body Radiotherapy for Liver Metastatic Colorectal Cancer. Available online: https://clinicaltrials.gov/ct2/show/NCT02837263 (accessed on 15 October 2021).
  198. Minami, Y.; Kudo, M. Radiofrequency ablation of liver metastases from colorectal cancer: A literature review. Gut Liver 2013, 7, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Ridge, C.A.; Solomon, S.B. Percutaneous ablation of colorectal lung metastases. J. Gastrointest. Oncol. 2015, 6, 685–692. [Google Scholar] [PubMed]
  200. Steinke, K.; Sewell, P.E.; Dupuy, D.; Lencioni, R.; Helmberger, T.; Kee, S.T.; Jacob, A.L.; Glenn, D.W.; King, J.; Morris, D.L. Pulmonary radiofrequency ablation-an international study survey. Anticancer Res. 2004, 24, 339–344. [Google Scholar]
  201. Shi, L.; Chen, L.; Wu, C.; Zhu, Y.; Xu, B.; Zheng, X.; Sun, M.; Wen, W.; Dai, X.; Yang, M. PD-1 blockade boosts radiofrequency ablation–elicited adaptive immune responses against tumor. Clin. Cancer Res. 2016, 22, 1173–1184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Shi, L.; Wang, J.; Ding, N.; Zhang, Y.; Zhu, Y.; Dong, S.; Wang, X.; Peng, C.; Zhou, C.; Zhou, L. Inflammation induced by incomplete radiofrequency ablation accelerates tumor progression and hinders PD-1 immunotherapy. Nat. Commun. 2019, 10, 5421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Zabaleta, J.; Iida, T.; Falcoz, P.E.; Salah, S.; Jarabo, J.R.; Correa, A.M.; Zampino, M.G.; Matsui, T.; Cho, S.; Ardissone, F.; et al. Individual data meta-analysis for the study of survival after pulmonary metastasectomy in colorectal cancer patients: A history of resected liver metastases worsens the prognosis. Eur. J. Surg. Oncol. 2018, 44, 1006–1012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Lyons, N.J.; Pathak, S.; Daniels, I.R.; Spiers, A.; Smart, N.J. Percutaneous management of pulmonary metastases arising from colorectal cancer; a systematic review. Eur. J. Surg. Oncol. 2015, 41, 1447–1455. [Google Scholar] [CrossRef]
  205. Beppu, T.; Sakamoto, Y.; Hasegawa, K.; Honda, G.; Tanaka, K.; Kotera, Y.; Nitta, H.; Yoshidome, H.; Hatano, E.; Ueno, M.; et al. A nomogram predicting disease-free survival in patients with colorectal liver metastases treated with hepatic resection: Multicenter data collection as a Project Study for Hepatic Surgery of the Japanese Society of Hepato-Biliary-Pancreatic Surgery. J. Hepatobiliary Pancreat Sci. 2012, 19, 72–84. [Google Scholar] [CrossRef] [PubMed]
  206. Di Martino, M.; Rompianesi, G.; Mora-Guzman, I.; Martin-Perez, E.; Montalti, R.; Troisi, R.I. Systematic review and meta-analysis of local ablative therapies for resectable colorectal liver metastases. Eur. J. Surg. Oncol. 2020, 46, 772–781. [Google Scholar] [CrossRef]
  207. Petrelli, F.; Comito, T.; Barni, S.; Pancera, G.; Scorsetti, M.; Ghidini, A.; Metastases, S.F.C.L. Stereotactic body radiotherapy for colorectal cancer liver metastases: A systematic review. Radiother. Oncol. 2018, 129, 427–434. [Google Scholar] [CrossRef]
  208. Bibbins-Domingo, K.; Force, U.S.P.S.T. Aspirin Use for the Primary Prevention of Cardiovascular Disease and Colorectal Cancer: U.S. Preventive Services Task Force Recommendation Statement. Ann. Intern. Med. 2016, 164, 836–845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Zhang, X.; Feng, Y.; Liu, X.; Ma, J.; Li, Y.; Wang, T.; Li, X. Beyond a chemopreventive reagent, aspirin is a master regulator of the hallmarks of cancer. J. Cancer Res. Clin. Oncol. 2019, 145, 1387–1403. [Google Scholar] [CrossRef]
  210. Ma, J.; Cai, Z.; Wei, H.; Liu, X.; Zhao, Q.; Zhang, T. The anti-tumor effect of aspirin: What we know and what we expect. Biomed. Pharmacother. 2017, 95, 656–661. [Google Scholar] [CrossRef] [PubMed]
  211. Guo, Q.; Liu, X.; Lu, L.; Yuan, H.; Wang, Y.; Chen, Z.; Ji, R.; Zhou, Y. Comprehensive evaluation of clinical efficacy and safety of celecoxib combined with chemotherapy in management of gastric cancer. Medicine 2017, 96, e8857. [Google Scholar] [CrossRef]
  212. Meyerhardt, J.A.; Shi, Q.; Fuchs, C.S.; Meyer, J.; Niedzwiecki, D.; Zemla, T.; Kumthekar, P.; Guthrie, K.A.; Couture, F.; Kuebler, P.; et al. Effect of Celecoxib vs Placebo Added to Standard Adjuvant Therapy on Disease-Free Survival Among Patients With Stage III Colon Cancer: The CALGB/SWOG 80702 (Alliance) Randomized Clinical Trial. JAMA 2021, 325, 1277–1286. [Google Scholar] [CrossRef] [PubMed]
  213. ClinicalTrials.gov. PD-1 Antibody Combined with COX Inhibitor in MSI-H/dMMR or High TMB Colorectal Cancer (PCOX). Available online: https://clinicaltrials.gov/ct2/show/NCT03638297 (accessed on 15 October 2021).
  214. Cao, D.; Zheng, Y.; Xu, H.; Ge, W.; Xu, X. Bevacizumab improves survival in metastatic colorectal cancer patients with primary tumor resection: A meta-analysis. Sci. Rep. 2019, 9, 20326. [Google Scholar] [CrossRef] [Green Version]
  215. Cremolini, C.; Antoniotti, C.; Rossini, D.; Lonardi, S.; Loupakis, F.; Pietrantonio, F.; Bordonaro, R.; Latiano, T.P.; Tamburini, E.; Santini, D. Upfront FOLFOXIRI plus bevacizumab and reintroduction after progression versus mFOLFOX6 plus bevacizumab followed by FOLFIRI plus bevacizumab in the treatment of patients with metastatic colorectal cancer (TRIBE2): A multicentre, open-label, phase 3, randomised, controlled trial. Lancet Oncol. 2020, 21, 497–507. [Google Scholar]
  216. Tang, W.; Ren, L.; Liu, T.; Ye, Q.; Wei, Y.; He, G.; Lin, Q.; Wang, X.; Wang, M.; Liang, F. Bevacizumab plus mFOLFOX6 versus mFOLFOX6 alone as first-line treatment for RAS mutant unresectable colorectal liver-limited metastases: The BECOME randomized controlled trial. J. Clin. Oncol. 2020, 38, 3175–3184. [Google Scholar] [CrossRef] [PubMed]
  217. Gruenberger, T.; Bridgewater, J.; Chau, I.; Alfonso, P.G.; Rivoire, M.; Mudan, S.; Lasserre, S.; Hermann, F.; Waterkamp, D.; Adam, R. Bevacizumab plus mFOLFOX-6 or FOLFOXIRI in patients with initially unresectable liver metastases from colorectal cancer: The OLIVIA multinational randomised phase II trial. Ann. Oncol. 2015, 26, 702–708. [Google Scholar] [CrossRef]
  218. Macedo, L.T.; da Costa Lima, A.B.; Sasse, A.D. Addition of bevacizumab to first-line chemotherapy in advanced colorectal cancer: A systematic review and meta-analysis, with emphasis on chemotherapy subgroups. BMC Cancer 2012, 12, 89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Nakagiri, T.; Tokunaga, T.; Kunoh, H.; Noura, S.; Ishida, H.; Miyoshi, N.; Ohue, M.; Okami, J.; Higashiyama, M. Surgical treatment following chemo-targeted therapy with bevacizumab for lung metastasis from colorectal carcinoma: Analysis of safety and histological therapeutic effects in patients treated at a single institution. Case Rep. Oncol. 2018, 11, 98–108. [Google Scholar] [CrossRef]
  220. Shimazaki, J.; Motohashi, G.; Nishida, K.; Tabuchi, T.; Ubukata, H.; Tabuchi, T. Complete response of lung metastases from rectal cancer to combination first-line therapy of S-1 and irinotecan plus bevacizumab: A case report and review of the literature. Oncol. Lett. 2014, 7, 1455–1458. [Google Scholar] [CrossRef] [PubMed]
  221. Peng, Y.; Chen, Y.; Zhang, X.; Yang, Y.; Cao, D.; Bi, F.; Li, Z.; Gou, H. Tumoral cavitation in colorectal cancer patients with unresectable lung metastasis treated with bevacizumab and chemotherapy. J. Cancer Res. Clin. Oncol. 2018, 144, 1339–1346. [Google Scholar] [CrossRef] [Green Version]
  222. Tabernero, J.; Yoshino, T.; Cohn, A.L.; Obermannova, R.; Bodoky, G.; Garcia-Carbonero, R.; Ciuleanu, T.E.; Portnoy, D.C.; Van Cutsem, E.; Grothey, A.; 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. Lancet Oncol. 2015, 16, 499–508. [Google Scholar] [CrossRef]
  223. Berlin, J.; Bendell, J.C.; Hart, L.L.; Firdaus, I.; Gore, I.; Hermann, R.C.; Mulcahy, M.F.; Zalupski, M.M.; Mackey, H.M.; Yauch, R.L.; et al. A randomized phase II trial of vismodegib versus placebo with FOLFOX or FOLFIRI and bevacizumab in patients with previously untreated metastatic colorectal cancer. Clin. Cancer Res. 2013, 19, 258–267. [Google Scholar] [CrossRef] [Green Version]
  224. Hecht, J.R.; Benson, A.B., 3rd; 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. Oncologist 2017, 22, 243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Clarke, J.M.; Blobe, G.C.; Strickler, J.H.; Uronis, H.E.; Zafar, S.Y.; Morse, M.; Dropkin, E.; Howard, L.; O’Neill, M.; Rushing, C.N.; et al. A phase Ib study of the combination regorafenib with PF-03446962 in patients with refractory metastatic colorectal cancer (REGAL-1 trial). Cancer Chemother. Pharm. 2019, 84, 909–917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Kelley, R.; Gane, E.; Assenat, E.; Siebler, J.; Galle, P.; Merle, P.; Hourmand, I.; Cleverly, A.; Zhao, Y.; Gueorguieva, I. A phase 2 study of galunisertib (TGF-β1 receptor type I inhibitor) and sorafenib in patients with advanced hepatocellular carcinoma. Clin. Transl. Gastroenterol. 2019, 10, e00056. [Google Scholar] [CrossRef] [PubMed]
  227. Holmgaard, R.B.; Schaer, D.A.; Li, Y.; Castaneda, S.P.; Murphy, M.Y.; Xu, X.; Inigo, I.; Dobkin, J.; Manro, J.R.; Iversen, P.W. Targeting the TGFβ pathway with galunisertib, a TGFβRI small molecule inhibitor, promotes anti-tumor immunity leading to durable, complete responses, as monotherapy and in combination with CheckpoInt. blockade. J. Immunother. Cancer 2018, 6, 47. [Google Scholar] [CrossRef]
  228. ClinicalTrials.gov. Galunisertib and Capecitabine in Advanced Resistant TGF-Beta Activated Colorectal Cancer (EORTC1615). Available online: https://clinicaltrials.gov/ct2/show/NCT03470350 (accessed on 15 October 2021).
  229. ClinicalTrials.gov. LY3200882 and Capecitabine in Advanced Resistant TGF-Beta Activated Colorectal Cancer (EORTC1615). Available online: https://www.clinicaltrials.gov/ct2/show/NCT04031872 (accessed on 15 October 2021).
  230. Available online: https://clinicaltrials.gov/ct2/show/NCT02688712 (accessed on 15 October 2021).
  231. Halama, N.; Zoernig, I.; Berthel, A.; Kahlert, C.; Klupp, F.; Suarez-Carmona, M.; Suetterlin, T.; Brand, K.; Krauss, J.; Lasitschka, F. Tumoral immune cell exploitation in colorectal cancer metastases can be targeted effectively by anti-CCR5 therapy in cancer patients. Cancer Cell 2016, 29, 587–601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Haag, G.M.; Halama, N.; Springfeld, C.; Grün, B.; Apostolidis, L.; Zschaebitz, S.; Dietrich, M.; Berger, A.-K.; Weber, T.F.; Zoernig, I. Combined PD-1 inhibition (Pembrolizumab) and CCR5 inhibition (Maraviroc) for the treatment of refractory microsatellite stable (MSS) metastatic colorectal cancer (mCRC): First results of the PICCASSO phase I trial. J. Clin. Oncol. 2020, 38, 3010. [Google Scholar] [CrossRef]
  233. Lakshmaiah, K.; Chaudhuri, T.; Babu, G.K.; Lokanatha, D.; Jacob, L.A.; Babu, M.S.; Rudresha, A.; Lokesh, K.; Rajeev, L. Safety and antitumor activity of arsenic trioxide plus infusional 5-fluorouracil, leucovorin, and irinotecan as second-line chemotherapy for refractory metastatic colorectal cancer: A pilot study from South India. Indian J. Cancer 2017, 54, 631. [Google Scholar] [PubMed]
  234. Sur, D.; Havasi, A.; Cainap, C.; Samasca, G.; Burz, C.; Balacescu, O.; Lupan, I.; Deleanu, D.; Irimie, A. Chimeric antigen receptor T-cell therapy for colorectal cancer. J. Clin. Med. 2020, 9, 182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Hege, K.M.; Bergsland, E.K.; Fisher, G.A.; Nemunaitis, J.J.; Warren, R.S.; McArthur, J.G.; Lin, A.A.; Schlom, J.; June, C.H.; Sherwin, S.A. Safety, tumor trafficking and immunogenicity of chimeric antigen receptor (CAR)-T cells specific for TAG-72 in colorectal cancer. J. Immunother. Cancer 2017, 5, 22. [Google Scholar] [CrossRef] [Green Version]
  236. Burga, R.A.; Thorn, M.; Point, G.R.; Guha, P.; Nguyen, C.T.; Licata, L.A.; DeMatteo, R.P.; Ayala, A.; Espat, N.J.; Junghans, R.P. Liver myeloid-derived suppressor cells expand in response to liver metastases in mice and inhibit the anti-tumor efficacy of anti-CEA CAR-T. Cancer Immunol. Immunother. 2015, 64, 817–829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  237. Le, D.T.; Durham, J.N.; Smith, K.N.; Wang, H.; Bartlett, B.R.; Aulakh, L.K.; Lu, S.; Kemberling, H.; Wilt, C.; Luber, B.S. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017, 357, 409–413. [Google Scholar] [CrossRef] [Green Version]
  238. Ipilimumab. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/125377s096lbl.pdf (accessed on 15 October 2021).
  239. Nivolumab. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/125554s097lbl.pdf (accessed on 15 October 2021).
  240. Chalabi, M.; Fanchi, L.F.; Dijkstra, K.K.; Van den Berg, J.G.; Aalbers, A.G.; Sikorska, K.; Lopez-Yurda, M.; Grootscholten, C.; Beets, G.L.; Snaebjornsson, P.; et al. Neoadjuvant immunotherapy leads to pathological responses in MMR-proficient and MMR-deficient early-stage colon cancers. Nat. Med. 2020, 26, 566–576. [Google Scholar] [CrossRef]
  241. Chen, E.X.; Jonker, D.J.; Loree, J.M.; Kennecke, H.F.; Berry, S.R.; Couture, F.; Ahmad, C.E.; Goffin, J.R.; Kavan, P.; Harb, M.; et al. Effect of Combined Immune CheckpoInt. Inhibition vs. Best Supportive Care Alone in Patients with Advanced Colorectal Cancer: The Canadian Cancer Trials Group CO.26 Study. JAMA Oncol. 2020, 6, 831–838. [Google Scholar] [CrossRef] [PubMed]
  242. Guan, Y.; Kraus, S.G.; Quaney, M.J.; Daniels, M.A.; Mitchem, J.B.; Teixeiro, E. FOLFOX chemotherapy ameliorates CD8 T lymphocyte exhaustion and enhances checkpoint blockade efficacy in colorectal Cancer. Front. Oncol. 2020, 10, 586. [Google Scholar] [CrossRef]
Figure 1. Formation of colorectal liver metastases through four overlapping phases: microvascular, extravascular, angiogenic, and growth.
Figure 1. Formation of colorectal liver metastases through four overlapping phases: microvascular, extravascular, angiogenic, and growth.
Cancers 13 06206 g001
Table 2. TME-specific targeted therapies currently under recent investigation in clinical trials. Red highlights reflect trials with negative findings. Green highlights reflect agents which are FDA-approved or recommended by the United States Preventive Services Task Force (Aspirin).
Table 2. TME-specific targeted therapies currently under recent investigation in clinical trials. Red highlights reflect trials with negative findings. Green highlights reflect agents which are FDA-approved or recommended by the United States Preventive Services Task Force (Aspirin).
TME Target Agent MechanismReference
InflammationAspirinCOX inhibitorBibbins-Domingo et al., 2016 [208]
CelecoxibCOX-2 inhibitorMeyerhardt et al., 2021 [212]; NCT03638297 [213]
AngiogenesisBevacizumab Anti-VEGF-A monoclonal antibodyFDA-Approved [180]
RamucirumabAnti-VEGF-R2 monoclonal antibodyFDA-Approved [181]
CAFsGalunisertib TGFβR1 inhibitorNCT03470350 [228]
NCT02688712 [230]
M7284TGF-β inhibitorNCT03436563 [196]
LY3200882TGFβR1 inhibitorNCT04031872 [229]
SimtuzumabLOXL2 inhibitorHecht et al., 2017 [224]
VismodegibSHH inhibitorBerlin et al., 2013 [223]
PF-03446962ALK-1 inhibitorClarke et al., 2019 [225]
TAMSMaravirocCCR5 inhibitorHalama et al., 2016 [231]; Haag et al., 2020 [232]
TregsArsenic TrioxideDepletion of TregsLakshmaiah et al., 2017 [233]
CD8+ T cellsPembrolizumabAnti-PD-1 monoclonal antibodyFDA-Approved [138]
NivolumabAnti-PD-1 monoclonal antibodyFDA-Approved [239]
DurvalumabAnti-PD-L1 monoclonal antibodyChen et al., 2020 [241]
IpilimumabAnti-CTLA-4 monoclonal antibodyFDA-Approved [238]
TremelimumabAnti-CTLA-4 monoclonal antibodyChen et al., 2020 [241]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Chandra, R.; Karalis, J.D.; Liu, C.; Murimwa, G.Z.; Voth Park, J.; Heid, C.A.; Reznik, S.I.; Huang, E.; Minna, J.D.; Brekken, R.A. The Colorectal Cancer Tumor Microenvironment and Its Impact on Liver and Lung Metastasis. Cancers 2021, 13, 6206. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13246206

AMA Style

Chandra R, Karalis JD, Liu C, Murimwa GZ, Voth Park J, Heid CA, Reznik SI, Huang E, Minna JD, Brekken RA. The Colorectal Cancer Tumor Microenvironment and Its Impact on Liver and Lung Metastasis. Cancers. 2021; 13(24):6206. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13246206

Chicago/Turabian Style

Chandra, Raghav, John D. Karalis, Charles Liu, Gilbert Z. Murimwa, Josiah Voth Park, Christopher A. Heid, Scott I. Reznik, Emina Huang, John D. Minna, and Rolf A. Brekken. 2021. "The Colorectal Cancer Tumor Microenvironment and Its Impact on Liver and Lung Metastasis" Cancers 13, no. 24: 6206. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13246206

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop