International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
In 2020, colorectal cancer (CRC) is the third most prevalent malignancy globally and the second leading cause of cancer-associated deaths (1) Distant metastasis is a key driver of mortality in CRC, with the liver being the most common site of metastasis (2). A total of 15–25% of patients with CRC present with synchronous liver metastases at initial diagnosis, while a further 15–25% develop metachronous liver metastases following primary CRC resection (2). Currently, a limited number of these liver metastases are amenable to surgical resection, and the 5-year survival rate for these patients is 25–44%. Moreover, up to 60% of patients experience rapid recurrence following resection (3).
The exact mechanisms underlying colorectal liver metastasis (CRLM) remain poorly understood. Tumor cell dissemination from the primary site to distant organs and the subsequent formation of metastatic tumors involve a dynamic process regulated by numerous genes and signaling pathways. This process includes tumor cell detachment from the primary site, entry into the circulatory or lymphatic system, extravasation and colonization at secondary sites. Tumor cell invasion, migration, adhesion, extracellular matrix remodeling, neovascularization and immune regulation are key in facilitating this metastatic cascade. The rise of immunotherapy, with its notable clinical success, has highlighted the critical role of the tumor immune microenvironment (TIME) in CRLM. CRLM represents a complex interaction between tumor and microenvironmental cells. Tumor cells, while acquiring invasive phenotypes, maintain intercellular communication with microenvironmental cells, regulating their functions through both direct and indirect mechanisms. This regulation involves the expression of immune checkpoint molecules and the secretion of cytokines, establishing a microenvironment conducive to tumor cell colonization and proliferation (4). Moreover, suppressive immune cells enhance the invasiveness of tumor cells by activating pro-metastatic signaling pathways. This interaction between tumor cells and the microenvironment drives the development of CRLM (Fig. 1).
The development, invasion and metastasis of CRC are complex biological processes involving multiple genetic alterations. Successive mutations and abnormal expression of genes such as APC, KRAS, BRAF and PTEN activate signaling pathways promoting CRC invasion and metastasis (5).
Epithelial-mesenchymal transition (EMT) is a critical precursor to CRLM. EMT refers to the phenotypical transformation of tumor epithelial cells into a mesenchymal phenotype, which enhances their migratory capacity (6). This involves the loss of intercellular junctions, increased secretion of intercellular plasma hydrolases and disruption of apical polarity, resulting in a breakdown of cell-cell adhesion. Concurrently, these changes induce the expression of N-cadherin, vimentin and α-smooth muscle actin, facilitating the transition from polarized epithelial cells to multipolar mesenchymal cells. This transformation increases cell motility, enabling tumor cells to detach from epithelial clusters and migrate individually in a mesenchymal manner, further enhancing the metastatic potential (7). The E-cadherin-β-catenin complex serves a pivotal role in maintaining epithelial integrity. Disruption of this complex leads to the detachment of cells from the primary tumor, enabling invasion and migration through the extracellular matrix and entry into the circulatory or lymphatic system. This is an important initiating step in the development of CRLM (8).
The acquisition of an invasive phenotype in CRC is governed by signaling pathways, including Wnt/β-catenin (9,10), TGF-β (11,12), PI3K/AKT (13–15), MEK/ERK (16,17) and hepatocyte growth factor (HGF)/MET (18). These pathways are regulated by intracellular oncogenes and tumor suppressor genes, as well as tumor microenvironmental signaling factors that influence tumor cell invasion and metastasis (Fig. 2).
Hepatic susceptibility to colonization by circulating tumor cells compared with other metastasis sites such as the lungs and peritoneum is primarily attributed to its highly permeable blood vessels, unique hemodynamic properties and distinct immune microenvironment. Notably, the ability to tolerate immune responses contributes to the creation of an immunosuppressive microenvironment, which protects the organ from excessive immune reactions to antigens. Additionally, liver homeostasis is maintained by specialized resident cells and diverse immune cell populations, which collectively regulate immune responses and oncogenesis (19–21).
During liver metastasis, immune cells undergo functional changes as a result of communication with tumor cells. Typically, under the influence of tumor cells, immune cells shift toward an immunosuppressive phenotype. Simultaneously, these immune cells reverse their roles to promote the invasive behavior of tumor cells by releasing pro-metastatic cytokines. This facilitates the establishment of a pre-metastatic niche, supporting the colonization of metastatic tumor cells and leading to the formation of metastatic foci (Fig. 3) (22).
CD8+ T cells play a critical role in antitumor immunity, and their infiltration is associated with a lower risk of recurrence and extended survival in patients with CRLM (23,24). However, tumors employ various mechanisms to suppress CD8+ T cell activity, facilitating immune evasion immune evasion. The expression of immune checkpoint molecules, such as PD-1/PD-L1, has emerged as a significant immunosuppressive factor and a promising therapeutic target (25). Both tumor and stromal cells exhibit high PD-L1 expression, which binds PD-1 on T cells, thereby inhibiting their activation. This interaction converts cytotoxic into exhausted T cells, promoting immune escape (25). Elevated PD-L1 expression in CRC is associated with advanced tumor stage, lymph node involvement, distant metastasis and poor prognosis (26,27). PD-L1 expression in CRLM is notably higher than in primary tumors, suggesting the activity of infiltrating CD8+ T cells in metastases is more significantly suppressed compared with primary foci (28). Aberrant activation of signaling pathways such as Wnt/β-catenin, PI3K/AKT, MEK/ERK and HGF/MET all contribute to the upregulation of PD-L1 expression in tumor cells (29). Targeted inhibition of these pathways, combined with αPD1 therapy, has shown potential in combating CRC liver metastasis (30).
Increased infiltration of CD4+ T cells in CRC has been associated with better prognosis following resection of liver metastatic tumors (31,32). However, different CD4+ T cell subtypes exert varying effects on tumor behavior. CD4+ T cells within the liver tend to differentiate into immunosuppressive phenotypes, producing high levels of immunosuppressive cytokines and restricting immune responses against metastatic tumors in the liver (33). T helper (Th)1 cells enhance the cytotoxic activity of CD8+ T cells, thereby boosting antitumor immunity. Th1 cell infiltration in CRC is associated with improved disease-free survival, while Th17 cell infiltration, which carries immunosuppressive functions, is associated with poor prognosis (34). Th17 cells are predominantly enriched in the primary lesions of metastatic CRC (mCRC) (35). Th17 cells promote tumor growth and metastasis by secreting cytokines such as IL-17, IL-21 and IL-22 (36). Furthermore, Th17 cells release tumor necrosis factor receptor superfamily member 12A, a cytokine that induces EMT and facilitates liver metastasis in CRC (35). The presence of Th17 cells in resected liver metastases is associated with poor prognosis (37).
Regulatory T cells (Tregs), identified by the expression of CD25 and FoxP3, are classical immune suppressors (38). In CRLM, Tregs are the primary source of IL-10, which increases PD-L1 expression on monocytes. This interaction diminishes CD8+ T cell infiltration and impairs antitumor immunity in CRLM (39). Previous studies have shown a significant increase in the proportion of Tregs in both mouse models (39,40) and resected CRLM specimens from patients (41). Furthermore, Treg-mediated suppression of the antitumor immune response is linked to clinical prognosis of patients with CRLM (42). Studies have revealed a paradox where elevated infiltration of FOXP3+ T cells in CRC is associated with improved relapse-free and disease-specific survival, while low FOXP3+ T cell infiltration is associated with poor prognosis (31,43). This discrepancy may be due to the heterogeneity of Tregs within the tumor microenvironment (TME). Saito et al (44) identified two distinct subpopulations of Tregs in CRC terminally differentiated immunosuppressive FoxP3high and pro-inflammatory FoxP3low subtypes. Inflammatory Treg-infiltrating CRC showed significant upregulation of genes associated with inflammation and immune responses. Functionally distinct subpopulations of Tregs influence CRC prognosis in opposing directions, with high FOXP3 expression in immunosuppressive Treg-infiltrating tumors associated with poorer outcomes. Pedroza-Gonzalez et al (45) demonstrated that, compared with Tregs from primary hepatocellular carcinoma, Tregs in CRLM exhibit higher expression of glucocorticoid-induced tumor necrosis factor receptor and stronger immunosuppressive activity. These findings suggest that a more precise characterization of Treg subpopulations in CRC and its liver metastases may deepen the understanding of Treg function in CRLM and help refine therapeutic strategies.
Macrophages within the TME exhibit notable plasticity and heterogeneity, classified into M1-type macrophages with pro-inflammatory, immune activating and antitumor properties, and M2-type macrophages, which possess immunosuppressive and pro-tumor functions. M2 macrophages are the dominant subtype in liver metastases (46,47) and associated with poor prognosis (48).
M2 macrophages facilitate tumor invasion and metastasis through several mechanisms, such as remodeling the extracellular matrix (49) and inducing EMT (50,51). Additionally, M2 macrophages contribute to tumor angiogenesis through the secretion of VEGF (52) and support immune evasion by releasing immunosuppressive cytokines such as IL-10 and TGF-β (53). Aberrant expression and activation of signaling molecules such as X-box binding protein 1 (XBP1) in M2 macrophages are further amplified by elevated cytokine secretion of cytokines, including IL-6 and VEGFA, which accelerate CRLM progression (54).
Cytokines and exosomes serve pivotal roles in CRC cells by inducing macrophage polarization toward the M2 phenotype. TGF-β, a classical cytokine, regulates macrophage polarization and, in CRLM, is modulated by pro-oncogenic factors such as collagen triple helix repeat containing 1, which further promotes CRLM by remodeling infiltrating macrophages via TGF-β signaling (55). Additionally, CCL2 is a key regulator of M2-type polarization, which fosters CRLM progression. Pro-oncogenic factors such as STAT3, transcription factor 4 (TCF4) and spondin 2 in CRC cells stimulate CCL2 secretion (56–58). Targeting the CCL2/CCR2 chemokine pathway reduces M2-typetumor-associated macrophages (TAMs) at metastatic sites, disrupts the immunosuppressive TME and enhances the susceptibility of mCRC to antitumor T cell responses (59). Tumor-derived factors such as CCL20, IL-10, VEGF and IL-1β also serve key roles in promoting macrophage infiltration and M2 polarization in CRLM (60,61). Exosome-mediated release of pro-oncogenic factors also contributes to the M2 polarization. CRC cells induce M2 polarization and establish an immunosuppressive pre-metastatic niche via the exosomal release of microRNAs (miRs; such as miR-25-3p, miR-130b-3p and miR-425-5p, miR-21-5p, miR-203, miR-934, miR-135a-5p and miR-106a-5p) and signaling molecules such as heat shock protein 90B1, and circ-0034880, which promote CRLM (62–69).
Recent studies have identified new macrophage subtypes (70,71). Liu et al (70) found that complement C1q subcomponent C (C1QC)+ macrophage express genes like CXCL9 and CXCL10, which are associated with favorable responses to immune checkpoint therapy and primarily support cellular phagocytosis and antigen presentation. This suggests that C1QC+ macrophage may have a beneficial role in CRLM treatment. Conversely, secreted phosphoprotein 1(SPP1)+ macrophage exert pro-angiogenic and tumor metastasis-promoting functions, with signature genes associated with poor prognosis. Notably, SPP1+ macrophage are absent in primary hepatocellular carcinoma but significantly enriched in CRLM, suggesting that they may facilitate CRC cell metastasis to the liver (70). Inhibiting their function may benefit CRLM treatment. Wu et al (71) demonstrated notable spatial reprogramming of the metastatic microenvironment, particularly involving immune-suppressive cells such as mannose receptor C-type 1+ CCL18+ M2-like macrophages. These macrophages were notably enriched in metastatic tumors and exhibited heightened metabolic activity.
Neutrophils in the TME exhibit dual roles: In early-stage tumors, they enhance T cell responses, while in advanced tumors, they adopt an immunosuppressive function (72). Similarly to macrophages, neutrophils in the TME can differentiate into distinct subsets, categorized as antitumor N1- and pro-tumor N2-type (73). N1-type neutrophils enhance tumor cell killing by expressing immune-activating cytokines and chemokines while inhibiting arginase expression. TGF-β in the TME induces the polarization of neutrophils from N1 to N2-type. N2-type neutrophils suppress tumor-killing T cell activity (74) and promote tumor invasion and metastasis by stimulating angiogenesis (75).
Neutrophils play a critical role in the formation and progression of CRLM (76). These metastases are characterized by neutrophil infiltration, which is notably more pronounced compared with uninvolved liver tissue. Neutrophils in the metastatic site promote angiogenesis and tumor metastasis by producing high levels of FGF2. Additionally, activated neutrophils can release neutrophil extracellular traps (NETs), and serum NET levels are elevated in patients with CRLM compared with those without liver metastases (77). Furthermore, liver metastasis specimens contain significantly more NETs than primary CRC lesions. NETs trap tumor cells in the liver, enhancing tumor proliferation and invasiveness, thus facilitating liver metastasis formation (78).
Tumor cells induce neutrophil infiltration via multiple pathways. CRC cells secrete granulocyte colony stimulating factor, which recruits neutrophils and upregulates Bv8/Prokineticin 2 expression, promoting immunosuppression and angiogenesis, thereby contributing to CRC metastasis (75,79). Seubert et al (80) demonstrated that tumor-derived tissue inhibitor of metalloproteinases 1(TIMP-1) upregulates stromal derived factor-1, recruiting neutrophils to the liver, and promoting liver metastasis. Aberrantly expressed molecules such as cell migration inducing hyaluronidase 1 (81) and DNA rrimase subunit 1 (82) in CRLM ead to the production of CXCL1 and CXCL3, causing accumulation of immunosuppressive neutrophils, ultimately enhancing CRLM progression. These findings suggest potential therapeutic targets for future CRLM research and treatment.
MDSCs, a heterogeneous group of immature myeloid cells, serve a pivotal role in facilitating CRLM by inducing immunosuppression, remodeling the extracellular matrix and promoting angiogenesis (83). Additionally, MDSCs are involved in the formation of NETs within the pre-metastatic niche (84). Abnormal expression and secretion of cytokines trigger MDSC infiltration, further advancing CRLM. CCL2-CCR2 signaling has been shown to induce MDSC infiltration in a STAT3-dependent manner, enhancing their immunosuppressive functions and contributing to CRC progression (85–87). Furthermore, CRC-derived CCL15 (88) and TME-derived CXCL1 (89,90) recruit MDSCs to establish a pre-metastatic niche and promote CRLM. Cytokines such as CCL7 (91), IL-6 (92), IL-33 (93) and exosomes containing long non-coding RNA MIR181A1HG (94) also serve critical roles in inducing MDSC infiltration, enhancing tumor invasiveness, supporting neovascularization and facilitating the creation of a pre-metastatic ecological niche in the liver. Targeted inhibition of these cytokines may offer an effective therapeutic approach for CRLM.
DCs, as antigen-presenting cells, initiate immune responses by capturing exogenous antigens and presenting them to T cells. To evade immune surveillance, tumor cells suppress antigen presentation by releasing inhibitory cytokines such as TGF-β (95). Compared with DCs from healthy individuals, those from patients with CRC exhibit impaired antigen presentation, decreased expression of costimulatory molecules, increased secretion of immunosuppressive IL-10 and reduced levels of immunostimulatory IL-12 and TNF-α (96). Nagorsen et al (97) found that tumor-infiltrating S100+ DCs in CRC are negatively associated with systemic antigen-specific T cell responses and positively associated with Tregs. Hsu et al (98) discovered elevated CXCL1 expression in CRC patient-derived DCs, which enhanced cell migration, increased matrix metalloproteinase 7 expression and promoted EMT, reflecting the altered functionality of DCs within the CRC TME. Huang et al (99) revealed that tumor-associated fibroblasts in CRC secrete WNT2, which suppresses DC function through the JAK2/STAT3 pathway. Targeting WNT2 may restore DC-mediated antitumor immunity. Further exploration of the mechanisms regulating DC function in CRC may identify new therapeutic targets for CRC treatment.
As a key component of innate immunity, NK cells exert antitumor effects by releasing cytotoxic molecules such as TRAIL and FasL. In a mouse model of CRLM, NK cells were shown to inhibit liver metastasis of CRC (100). Increased NK cell infiltration is associated with improved overall survival (OS) in patients with CRLM (101).
Restoring NK cell function inhibits CRLM progression in CRC. Dupaul-Chicoine et al (102) demonstrated that the NLRP3 inflammasome suppresses metastatic growth of CRC in the liver by enhancing the tumor-killing activity of NK cells. In NLRP3 inflammasome-deficient mice, IL-18 signaling is impaired, affecting NK cell maturation in the liver and their ability to effectively kill tumor cells (102). TRAIL is hypothesized to play a central role in NK cell-mediated tumor killing. TRAIL is constitutively expressed on hepatic NK cells and, together with perforin and FasL, triggers a toxic response to tumor cells in vitro (103). Neutralizing TRAIL with a monoclonal antibody significantly increases the formation of experimental hepatic metastases (103). CXCR3 has been shown to enhance protection against CRLM by promoting NK cell infiltration and plasticity (104). CXCR3 facilitates the accumulation and persistence of CD49a+ NK cells, which exhibit the highest cytotoxic capacity among metastasis-infiltrating NK cells (104).
However, NK cell function is impaired in both CRC and liver metastases compared with NK cells in healthy livers. CRC cells regulate the TME pH by producing lactic acid, which lowers the pH within NK cells, leading to mitochondrial dysfunction and apoptosis. This enables tumor cells to evade the cytotoxic effects of NK cells (105). Metabolic dysfunction in the metastatic niche notably impacts NK cell functionality. Increased glutamine uptake by cancer cells depletes glutamine availability for NK cells, decreasing their activity and cytotoxicity, thereby promoting CRLM progression (106).
Upon entering the portal circulation, tumor cells reach the hepatic sinusoidal capillaries through the portal vein. These tumor cells trigger non-specific liver defense mechanisms, leading to their phagocytosis by resident immune cells such as Kupffer cells (KCs) and NK cells (107).
KCs, the resident macrophages of the liver, serve a pivotal role in maintaining liver homeostasis and are key contributors to the pathogenesis of liver disease. KCs are essential in defending against liver metastasis due to their phagocytic capability, cytokine production and promotion of tertiary lymphoid structures (108). Dysfunction of phagocytosis in KCs is a key driving force in CRLM. Some tumor cells can evade phagocytosis by KCs, highlighting the need for further research into this evasion mechanism to identify novel therapeutic targets for liver metastasis (109). The balance between pro-phagocytic ‘eat me’ signals, such as tumor-associated antigens, calreticulin, SLAM Family Member 7 and Erythroblast Membrane Associated Protein (ERMAP), and anti-phagocytic ‘don't eat me’ signals, including CD47, PD-L1, CD24 and β2-microglobulin, is a key determinant in the phagocytosis process (110). Additionally, the functional reprogramming of KCs warrants attention. Following metastatic colonization of the liver, KCs undergo transcriptional reprogramming typical of TAMs, which facilitates tumor progression (111). The phagocytosis of exosomes released by the primary tumor into the circulation and into the liver by KCs can initiate the formation of a pre-metastatic niche in the liver (66).
Liver-resident specialized NK cells also serve a significant role in the early stages of metastasis by contributing to the establishment of pre-metastatic niches. Invariant NK T cells in the liver promote metastasis by producing fibrogenic cytokines such as IL-4 and IL-13, independent of T cell receptor activation, thereby inducing a fibrotic niche in the liver. Targeted disruption of IL-4 and IL-13 signaling pathways in hepatic stellate cells inhibits their trans differentiation into extracellular matrix-producing myofibroblasts, thereby impeding the metastatic proliferation of disseminated cancer cells (112).
Tumor cells that evade innate immune surveillance extravasate from blood vessels and form metastatic lesions. Tumor cell adhesion to the vascular system is not only driven by mechanical blockage of the vasculature, but also by specific cellular adhesion processes that facilitate tumor cell attachment and extravasation (113,114). Liver sinusoidal endothelial cells express the cell adhesion molecule E-selectin, which promotes tumor cell attachment to hepatic sinusoidal endothelial cells. Inhibition of E-selectin effectively decreases the formation of liver metastases (115). Moreover, tumor cells can trigger the release of pro-inflammatory cytokines, such as TNF-α, from KCs, which upregulates the expression of adhesion molecules, such as E-selectin, vascular cell adhesion molecule 1 and intercellular adhesion molecule 1(ICAM1) on hepatic sinusoidal endothelial cells. This increase in adhesion molecule expression enhances tumor cell colonization in the liver (116).
Once tumor cells extravasate into the Disse interstitium, hepatic stellate cells are activated by cytokines such as TGF-β, which is secreted by KCs. This activation prompts hepatic stellate cells to produce extracellular matrix proteins such as collagen, laminin and fibronectin, creating a supportive environment for tumor cell colonization. Additionally, KCs and neutrophils secrete matrix metalloproteinases and elastases, which degrade and remodel the extracellular matrix, facilitating tumor cell invasion. Concurrently, hepatic stellate cells promote a suppressive microenvironment by inducing the apoptosis of cytotoxic T cells and expanding immune-regulatory T cells, creating a favorable environment for tumor cell colonization in the liver (117).
In the context of chronic liver disease, Zeng et al (118) observed that abnormal activation of hepatocellular cell cycle-related kinase (CCRK) and NF-κB signaling pathways increases CXCL1 expression, which induces MDSC infiltration and facilitates CRC metastasis to the liver. Non-alcoholic fatty liver disease stimulates KCs to secrete the chemokine CXCL5, which recruits CXCR2+ MDSCs, further promoting CRLM progression (119). Additionally, extracellular vesicles in fatty liver are implicated in promoting CRC liver metastasis by fostering cancer cell proliferation and an immunosuppressive microenvironment through M2 macrophage infiltration, thus enhancing the metastatic TME (120).
Research into liver metastasis in CRC has revealed numerous potential therapeutic targets, with targeted therapy and immunotherapy emerging as the leading strategies for managing CRLM (121). In addition to traditional neoadjuvant radiotherapy and chemotherapy and adjuvant therapy following surgical resection, integrating targeted and immunotherapies with radiotherapy/chemotherapy and surgery has established a comprehensive treatment system for CRLM (122). This combination therapy offers dual benefits: By initiating neoadjuvant treatment, previously unresectable CRLM can become surgically resectable, increasing the number of patients eligible for hepatic resection while decreasing perioperative morbidity and mortality (123). Second, these combination therapies show promise in enhancing long-term survival rates for patients (124).
EGFR, a member of the receptor tyrosine kinase (TK) family, serves a key role in CRC development and invasive metastasis through downstream signaling pathways, including the RAS/RAF/MEK/ERK and PI3K/AKT pathways (125). Cetuximab, the first monoclonal antibody targeting EGFR, significantly improves OS and progression-free survival (PFS) in patients with CRC who are resistant to other treatment (126). When combined with chemotherapy, cetuximab has demonstrated favorable therapeutic outcomes. The combination of cetuximab with FOLFIRI (irinotecan, fluorouracil and leucovorin) significantly decreases the risk of disease progression in patients with mCRC compared with FOLFIRI alone (127). The combination of cetuximab with FOLFOX4(oxaliplatin, leucovorin, and fluorouracil) as first-line treatment for mCRC has also shown superior remission rates compared with the FOLFOX4 regimen alone (128). However, mutations in the RAS gene in CRC confer resistance to cetuximab, meaning cetuximab is effective in patients with RAS wild-type mCRC (129) Another EGFR targeting drug, panitumumab, is a fully humanized antibody that does not induce antibody-dependent cell-mediated cytotoxicity (125). The PRIME trial, which examined the efficacy of FOLFOX alone and in combination with panitumumab in patients with mCRC, found that the combination therapy resulted in higher OS and PFS compared with FOLFOX treatment alone (130,131).
Bevacizumab, a monoclonal antibody targeting the angiogenesis inhibitor VEGF, has received US Food and Drug Administration approval for the treatment of mCRC and demonstrated favorable efficacy (132). A meta-analysis by Cao et al (133), which included 1,838 patients with mCRC, showed that chemotherapy combined with bevacizumab following primary tumor resection significantly prolonged OS compared with chemotherapy alone. The study also revealed improved OS in patients who were initially unable to undergo resection of their primary tumor when treated with bevacizumab. Additionally, the combination of bevacizumab with chemotherapy may enhance the resectability of CRLM. In a study by Tang et al (134), the combination of bevacizumab with mFOLFOX6 as first-line treatment for patients with unresectable CRLM harboring RAS mutations demonstrated significantly superior efficacy compared with mFOLFOX6 monotherapy. This combination not only markedly increased the R0 resection rate of liver metastases but also improved the overall resectability of liver metastases, leading to improved PFS and OS in patients. Ramucirumab, a VEGFR antagonist that specifically binds VEGFR2 and blocks ligand-receptor binding, has also shown promising results in mCRC treatment (135). In a study by Tabernero et al (136), the combination of ramucirumab and FOLFIRI was evaluated against a placebo in patients with mCRC. Ramucirumab and FOLFIRI combination significantly improved OS in patients. Ramucirumab is currently approved for use in the second-line treatment of mCRC.
Receptor TKs, located on the cell surface and intracellularly, play a key role in intercellular signaling, which influences cell function. TKIs block the activity of kinase proteins that contribute to tumor cell proliferation and the development of tumor vasculature. Regorafenib is an oral, multi-targeted TKI that inhibits VEGFR1-3, PDGFR, FGFR, KIT, RET1 and BRAF and has been shown to improve survival in patients with refractory mCRC (137). Regorafenib is used to treat mCRC that progresses despite previous chemotherapy, anti-VEGF or anti-EGFR therapy (138). In addition to regorafenib, fruquintinib is an oral TKI that selectively inhibits different subtypes of VEGFR, thereby inhibiting tumor angiogenesis and growth (139). The FRESCO study evaluated the effectiveness and safety of fruquintinib as a third-line or subsequent treatment for patients with mCRC. The results demonstrated that fruquintinib monotherapy significantly prolonged survival in patients with mCRC who had failed second-line or higher chemotherapy (140). FRESCO-2, an international, multicentre, randomised, double-blind, phase 3 study, also demonstrated that fruquintinib significantly improved overall survival in patients with refractory metastatic colorectal cancer compared to placebo (141). Fruquintinib is currently approved by FDA for use in patients with mCRC who have previously undergone fluoropyrimidine-, oxaliplatin-, and irinotecan-based chemotherapy, an anti-VEGF therapy, and if RAS wild-type and medically appropriate, an anti-EGFR therapy (142).
In recent years, immune checkpoint inhibitors have garnered attention due to their success in achieving long-lasting responses in a range of previously difficult-to-treat solid tumors (143). Overman et al (144) demonstrated the significant efficacy of the PD-1 inhibitor nivolumab alone in individuals with deficient mismatch repair/microsatellite instability(dMMR/MSI) CRC in a multicenter phase II clinical trial (CheckMate142). Similarly, the KEYNOTE-177 (145) study showed a significant improvement in PFS in patients with dMMR/MSI mCRC treated with the PD-1 inhibitor pembrolizumab as first-line therapy, compared with standard treatment. PD-1 inhibitors are currently approved by FDA for patients with dMMR/MSI mCRC who experience disease progression following standard chemotherapy (146). This approval highlights their importance as a pivotal immunotherapy approach, particularly for individuals with liver metastases from CRC. However, the efficacy of PD-1 inhibitors varies among patients, and some do not benefit from them. Patients with proficient mismatch repair/microsatellite stability(pMMR/MSS) CRC, which constitutes the majority of the patient population, derive limited benefits from PD-1 inhibitor therapy. For individuals with dMMR/MSI CRC, who are currently considered candidates for PD-1 inhibitor treatment, the observed efficacy rate is suboptimal. In the KEYNOTE-016 trial, pembrolizumab was administered to a cohort of 41 patients with CRC, including both pMMR/MSS and dMMR/MSI subgroups, who experienced disease progression following chemotherapy. In patients with dMMR/MSI CRC, the immune-related objective response rate was 40% and the 20-week PFS rate was 78%. By contrast, patients with pMMR/MSS CRC exhibited an immune-related objective response rate of 0% and a 20-week PFS rate of 11% (147). Furthermore, liver metastases from CRC induce systemic immune tolerance through a unique immunosuppressive mechanism, which may further affect the efficacy of PD-1 inhibitor therapy (148–150). Exploring effective combination therapies may enhance the efficacy of PD-1 inhibitors. The CheckMate-142 study demonstrated that combining PD-1 inhibitors with CTLA4 inhibitors improves antitumor efficacy (151). In addition to immune checkpoint inhibitors, drugs such as regorafenib have gained widespread attention as potential combination therapies with PD-1 inhibitors due to their promising role in modulating immunity and improving the TME (152,153).
Cancer vaccines, adoptive cell transfer (ACT) therapy and oncolytic viruses have emerged as prominent areas of research in CRLM (154–156). As a form of active immunotherapy, cancer vaccines present the immune system with tumor-specific or -associated antigens, inducing antitumor cytotoxic responses that help the immune system recognize and destroy cancer cells (157). ACT therapy enhances the natural anti-cancer response by activating or genetically modifying autologous or allogeneic immune cells in vitro to boost tumor-fighting capabilities, followed by reinfusion into patients. ACT includes therapies such as cytokine-induced killer cells and chimeric antigen receptor T cell therapies (158). Oncolytic virus therapy uses naturally occurring or genetically engineered viruses to selectively target and lyse tumor cells. This strategy not only modulates the TIME but also activates specific anti-tumor immune responses (159). Additionally, it is crucial to explore the role of cytokines, chemokines and adjuvants in enhancing the precision and efficacy of immunotherapies in CRLM.
Liver metastasis is a major factor influencing the prognosis of patients with CRC. The process of liver metastasis in CRC is complex and involves interconnected stages. Key pathways that affect the invasive and metastatic potential of CRC cells have been identified, along with prognostic and therapeutic molecules. Additionally, factors within the TME influencing liver metastasis in CRC have been preliminarily analyzed. Notably, the discovery of immune-associated targets holds promise for treating liver metastasis in CRC and improving prognosis. However, the clinical application of these targets and associated drugs in individuals with CRLM remains limited, and many patients do not benefit from current targeted therapies and immunotherapies. Therapeutic strategies aimed at modulating the immunosuppressive microenvironment, such as depleting immunosuppressive cells, inhibiting immune checkpoint pathways and stimulating cytotoxic cells are critical approaches for enhancing the effectiveness of immunotherapy.
Currently, novel immunotherapies for CRLM remain in preclinical or clinical trials, and their successful integration into clinical practice faces challenges. Firstly, CRLM creates a highly immunosuppressive microenvironment within the liver. This hostile TME actively inhibits the function of effector immune cells (such as cytotoxic T cells and NK cells), rendering many immunotherapies ineffective. Secondly, tumor heterogeneity and evolution make it difficult to identify universal therapeutic targets. Thirdly, lack of predictive biomarkers makes it difficult to select patients most likely to benefit from expensive and potentially toxic immunotherapies, leading to low response rates and inefficient resource use. Lastly, overcoming the aforementioned challenges above often requires combining immunotherapies (e.g., dual checkpoint blockade) with other modalities like chemotherapy, targeted therapy (e.g., anti-VEGF, anti-EGFR), radiotherapy, liver-directed therapies (ablation, embolization), or other immunomodulators. Combinations significantly increase the risk of severe immune-related adverse events (irAEs), including hepatitis, colitis, pneumonitis, and endocrine toxicities. Managing overlapping toxicities, especially in patients with liver involvement, is challenging and can limit dosing or lead to treatment discontinuation.
Overcoming these multifaceted challenges requires further intensive studies to understand the CRLM biology and liver immunology, elucidate the underlying mechanisms, discovering robust biomarkers and identify novel therapeutic targets. Deeper understanding of the key molecules and signaling pathways influencing the invasive and metastatic potential of CRC is key, achievable through the integration of high-throughput genomic technology. Considering the complex and heterogeneous influence of the TME on liver metastasis, it is necessary to investigate the functional characteristics and dynamic changes of distinct cell populations throughout the liver metastasis process, at a single-cell level to identify key cells and molecules driving TME remodeling. Moreover, as the understanding of the mechanisms underlying CRLM advances, the diagnosis and treatment of this condition may shift toward a multidisciplinary approach and enhance patient care by facilitating a comprehensive and integrated treatment strategy.
In conclusion, further research is required into the mechanisms in CRLM, the exploration of novel targets for personalized treatment and the development of innovative intervention strategies to improve CRLM therapy efficacy.
Not applicable.
The present study was supported by the National Natural Science Foundation of China (grant no. 82272841).
Not applicable.
CJY conceptualized the study and wrote the manuscript. LZ performed the literature review. CHW and YJY contributed to conception and design. ZLS revised and edited the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021.PubMed/NCBI | |
|
Horn SR, Stoltzfus KC, Lehrer EJ, Dawson LA, Tchelebi L, Gusani NJ, Sharma NK, Chen H, Trifiletti DM and Zaorsky NG: Epidemiology of liver metastases. Cancer Epidemiol. 67:1017602020. View Article : Google Scholar : PubMed/NCBI | |
|
Rees M, Tekkis PP, Welsh FK, O'Rourke T and John TG: Evaluation of long-term survival after hepatic resection for metastatic colorectal cancer: A multifactorial model of 929 patients. Ann Surg. 247:125–135. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Joyce JA and Pollard JW: Microenvironmental regulation of metastasis. Nat Rev Cancer. 9:239–252. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Medici B, Benatti S, Dominici M and Gelsomino F: New frontiers of biomarkers in metastatic colorectal cancer: Potential and critical issues. Int J Mol Sci. 26:52682025. View Article : Google Scholar : PubMed/NCBI | |
|
Tsubakihara Y and Moustakas A: Epithelial-mesenchymal transition and metastasis under the control of transforming growth factor β. Int J Mol Sci. 19:36722018. View Article : Google Scholar : PubMed/NCBI | |
|
van Zijl F, Krupitza G and Mikulits W: Initial steps of metastasis: Cell invasion and endothelial transmigration. Mutat Res. 728:23–34. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Qi J and Zhu YQ: Targeting the most upstream site of Wnt signaling pathway provides a strategic advantage for therapy in colorectal cancer. Curr Drug Targets. 9:548–557. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Rubinfeld B, Albert I, Porfiri E, Fiol C, Munemitsu S and Polakis P: Binding of GSK3beta to the APC-beta-catenin complex and regulation of complex assembly. Science. 272:1023–1026. 1996. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Z, Gao Y, Qian Y, Wei B, Jiang K, Sun Z, Zhang F, Yang M, Baldi S, Yu X, et al: The Lyn/RUVBL1 complex promotes colorectal cancer liver metastasis by regulating arachidonic acid metabolism through chromatin remodeling. Adv Sci (Weinh). 12:e24065622025. View Article : Google Scholar : PubMed/NCBI | |
|
Zubeldia IG, Bleau AM, Redrado M, Serrano D, Agliano A, Gil-Puig C, Vidal-Vanaclocha F, Lecanda J and Calvo A: Epithelial to mesenchymal transition and cancer stem cell phenotypes leading to liver metastasis are abrogated by the novel TGFβ1-targeting peptides P17 and P144. Exp Cell Res. 319:12–22. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Yang Y, Qi X, Cui P, Kang Y, Liu H, Wei Z and Wang H: SLC14A1 and TGF-β signaling: A feedback loop driving EMT and colorectal cancer metachronous liver metastasis. J Exp Clin Cancer Res. 43:2082024. View Article : Google Scholar : PubMed/NCBI | |
|
Shin AE, Sugiura K, Kariuki SW, Cohen DA, Flashner SP, Klein-Szanto AJ, Nishiwaki N, De D, Vasan N, Gabre JT, et al: LIN28B-mediated PI3K/AKT pathway activation promotes metastasis in colorectal cancer models. J Clin Invest. 135:e1860352025. View Article : Google Scholar : PubMed/NCBI | |
|
Sun X, Zhang J, Dong B, Xiong Q, Wang X, Gu Y, Wang Z, Liu H, Zhang J, He X, et al: Targeting SLITRK4 restrains proliferation and liver metastasis in colorectal cancer via regulating PI3K/AKT/NFκB pathway and tumor-associated macrophage. Adv Sci (Weinh). 12:e24003672025. View Article : Google Scholar : PubMed/NCBI | |
|
Dong Z, She X, Ma J, Chen Q, Gao Y, Chen R, Qin H, Shen B and Gao H: The E3 Ligase NEDD4L prevents colorectal cancer liver metastasis via degradation of PRMT5 to inhibit the AKT/mTOR signaling pathway. Adv Sci (Weinh). 2025:e25047042025. View Article : Google Scholar : PubMed/NCBI | |
|
Urosevic J, Blasco MT, Llorente A, Bellmunt A, Berenguer-Llergo A, Guiu M, Cañellas A, Fernandez E, Burkov I, Clapés M, et al: ERK1/2 signaling induces upregulation of ANGPT2 and CXCR4 to mediate liver metastasis in colon cancer. Cancer Res. 80:4668–4680. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Chu PC, Lin PC, Wu HY, Lin KT, Wu C, Bekaii-Saab T, Lin YJ, Lee CT, Lee JC and Chen CS: 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. 37:3440–3455. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Yao JF, Li XJ, Yan LK, He S, Zheng JB, Wang XR, Zhou PH, Zhang L, Wei GB and Sun XJ: Role of HGF/c-Met in the treatment of colorectal cancer with liver metastasis. J Biochem Mol Toxicol. 33:e223162019. View Article : Google Scholar : PubMed/NCBI | |
|
Xu W, Xu J, Liu J, Wang N, Zhou L and Guo J: Liver metastasis in cancer: Molecular mechanisms and management. MedComm (2020). 6:e701192025. View Article : Google Scholar : PubMed/NCBI | |
|
Dunbar KJ, Efe G, Cunningham K, Esquea E, Navaridas R and Rustgi AK: Regulation of metastatic organotropism. Trends Cancer. 11:216–231. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu C, Liao JY, Liu YY, Chen ZY, Chang RZ, Chen XP, Zhang BX and Liang JN: Immune dynamics shaping pre-metastatic and metastatic niches in liver metastases: From molecular mechanisms to therapeutic strategies. Mol Cancer. 23:2542024. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y, Liu F, Cai Q, Deng L, Ouyang Q, Zhang XH and Zheng J: Invasion and metastasis in cancer: Molecular insights and therapeutic targets. Signal Transduct Target Ther. 10:572025. View Article : Google Scholar : PubMed/NCBI | |
|
Glaire MA, Domingo E, Sveen A, Bruun J, Nesbakken A, Nicholson G, Novelli M, Lawson K, Oukrif D, Kildal W, et al: Tumour-infiltrating CD8+ lymphocytes and colorectal cancer recurrence by tumour and nodal stage. Br J Cancer. 121:474–482. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Trailin A, Ali E, Ye W, Pavlov S, Červenková L, Vyčítal O, Ambrozkiewicz F, Hošek P, Daum O, Liška V and Hemminki K: Prognostic assessment of T-cells in primary colorectal cancer and paired synchronous or metachronous liver metastasis. Int J Cancer. 156:1282–1292. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Yang A, Zhou M, Gao Y and Zhang Y: Mechanisms of CD8+ T cell exhaustion and its clinical significance in prognosis of anti-tumor therapies: A review. Int Immunopharmacol. 159:1148432025. View Article : Google Scholar : PubMed/NCBI | |
|
Shan T, Chen S, Wu T, Yang Y, Li S and Chen X: PD-L1 expression in colon cancer and its relationship with clinical prognosis. Int J Clin Exp Pathol. 12:1764–1769. 2019.PubMed/NCBI | |
|
Zhao T, Li Y, Zhang J and Zhang B: PD-L1 expression increased by IFN-γ via JAK2-STAT1 signaling and predicts a poor survival in colorectal cancer. Oncol Lett. 20:1127–1134. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Wei XL, Luo X, Sheng H, Wang Y, Chen DL, Li JN, Wang FH and Xu RH: PD-L1 expression in liver metastasis: Its clinical significance and discordance with primary tumor in colorectal cancer. J Transl Med. 18:4752020. View Article : Google Scholar : PubMed/NCBI | |
|
Rong D, Sun G, Zheng Z, Liu L, Chen X, Wu F, Gu Y, Dai Y, Zhong W, Hao X, et al: MGP promotes CD8+ T cell exhaustion by activating the NF-κB pathway leading to liver metastasis of colorectal cancer. Int J Biol Sci. 18:2345–2361. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Sun G, Zhao S, Fan Z, Wang Y, Liu H, Cao H, Sun G, Huang T, Cai H, Pan H, et al: CHSY1 promotes CD8+ T cell exhaustion through activation of succinate metabolism pathway leading to colorectal cancer liver metastasis based on CRISPR/Cas9 screening. J Exp Clin Cancer Res. 42:2482023. View Article : Google Scholar : PubMed/NCBI | |
|
Kuwahara T, Hazama S, Suzuki N, Yoshida S, Tomochika S, Nakagami Y, Matsui H, Shindo Y, Kanekiyo S, Tokumitsu Y, et al: Intratumoural-infiltrating CD4+ and FOXP3 + T cells as strong positive predictive markers for the prognosis of resectable colorectal cancer. Br J Cancer. 121:659–665. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Katz SC, Pillarisetty V, Bamboat ZM, Shia J, Hedvat C, Gonen M, Jarnagin W, Fong Y, Blumgart L, D'Angelica M and DeMatteo RP: T cell infiltrate predicts long-term survival following resection of colorectal cancer liver metastases. Ann Surg Oncol. 16:2524–2530. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Katz SC, Pillarisetty VG, Bleier JI, Kingham TP, Chaudhry UI, Shah AB and DeMatteo RP: Conventional liver CD4 T cells are functionally distinct and suppressed by environmental factors. Hepatology. 42:293–300. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Tosolini M, Kirilovsky A, Mlecnik B, Fredriksen T, Mauger S, Bindea G, Berger A, Bruneval P, Fridman WH, Pagès F and Galon J: Clinical impact of different classes of infiltrating T cytotoxic and helper cells (Th1, th2, treg, th17) in patients with colorectal cancer. Cancer Res. 71:1263–1271. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Liu X, Wang X, Yang Q, Luo L, Liu Z, Ren X, Lei K, Li S, Xie Z, Zheng G, et al: Th17 cells Secrete TWEAK to trigger epithelial-mesenchymal transition and promote colorectal cancer liver metastasis. Cancer Res. 84:1352–1371. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
De Simone V, Pallone F, Monteleone G and Stolfi C: Role of T(H)17 cytokines in the control of colorectal cancer. Oncoimmunology. 2:e266172013. View Article : Google Scholar : PubMed/NCBI | |
|
Kroemer M, Turco C, Spehner L, Viot J, Idirène I, Bouard A, Renaude E, Deschamps M, Godet Y, Adotévi O, et al: Investigation of the prognostic value of CD4 T cell subsets expanded from tumor-infiltrating lymphocytes of colorectal cancer liver metastases. J Immunother Cancer. 8:e0014782020. View Article : Google Scholar : PubMed/NCBI | |
|
Olguín JE, Medina-Andrade I, Rodríguez T, Rodríguez-Sosa M and Terrazas LI: Relevance of regulatory T cells during colorectal cancer development. Cancers (Basel). 12:18882020. View Article : Google Scholar : PubMed/NCBI | |
|
Shiri AM, Zhang T, Bedke T, Zazara DE, Zhao L, Lücke J, Sabihi M, Fazio A, Zhang S, Tauriello DVF, et al: IL-10 dampens antitumor immunity and promotes liver metastasis via PD-L1 induction. J Hepatol. 80:634–644. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Huang X, Chen Z, Zhang N, Zhu C, Lin X, Yu J, Chen Z, Lan P and Wan Y: Increase in CD4+FOXP3+ regulatory T cell number and upregulation of the HGF/c-Met signaling pathway during the liver metastasis of colorectal cancer. Oncol Lett. 20:2113–2118. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Katz SC, Bamboat ZM, Maker AV, Shia J, Pillarisetty VG, Yopp AC, Hedvat CV, Gonen M, Jarnagin WR, Fong Y, et al: Regulatory T cell infiltration predicts outcome following resection of colorectal cancer liver metastases. Ann Surg Oncol. 20:946–955. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Brudvik KW, Henjum K, Aandahl EM, Bjørnbeth BA and Taskén K: Regulatory T-cell-mediated inhibition of antitumor immune responses is associated with clinical outcome in patients with liver metastasis from colorectal cancer. Cancer Immunol Immunother. 61:1045–1053. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Salama P, Phillips M, Grieu F, Morris M, Zeps N, Joseph D, Platell C and Iacopetta B: Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer. J Clin Oncol. 27:186–192. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Saito T, Nishikawa H, Wada H, Nagano Y, Sugiyama D, Atarashi K, Maeda Y, Hamaguchi M, Ohkura N, Sato E, et al: Two FOXP3(+)CD4(+) T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat Med. 22:679–684. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Pedroza-Gonzalez A, Verhoef C, Ijzermans JN, Peppelenbosch MP, Kwekkeboom J, Verheij J, Janssen HL and Sprengers D: Activated tumor-infiltrating CD4+ regulatory T cells restrain antitumor immunity in patients with primary or metastatic liver cancer. Hepatology. 57:183–194. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
He Y, Han Y, Fan AH, Li D, Wang B, Ji K, Wang X, Zhao X and Lu Y: Multi-perspective comparison of the immune microenvironment of primary colorectal cancer and liver metastases. J Transl Med. 20:4542022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang D, Wang X, Si M, Yang J, Sun S, Wu H, Cui S, Qu X and Yu X: Exosome-encapsulated miRNAs contribute to CXCL12/CXCR4-induced liver metastasis of colorectal cancer by enhancing M2 polarization of macrophages. Cancer Lett. 474:36–52. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Lee YS, Song SJ, Hong HK, Oh BY, Lee WY and Cho YB: The FBW7-MCL-1 axis is key in M1 and M2 macrophage-related colon cancer cell progression: Validating the immunotherapeutic value of targeting PI3Kγ. Exp Mol Med. 52:815–831. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Afik R, Zigmond E, Vugman M, Klepfish M, Shimshoni E, Pasmanik-Chor M, Shenoy A, Bassat E, Halpern Z, Geiger T, et al: Tumor macrophages are pivotal constructors of tumor collagenous matrix. J Exp Med. 213:2315–2331. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Cai J, Xia L, Li J, Ni S, Song H and 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. 51:252–266. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wei C, Yang C, Wang S, Shi D, Zhang C, Lin X, Liu Q, Dou R and Xiong B: Crosstalk between cancer cells and tumor associated macrophages is required for mesenchymal circulating tumor cell-mediated colorectal cancer metastasis. Mol Cancer. 18:642019. View Article : Google Scholar : PubMed/NCBI | |
|
Suarez-Lopez L, Sriram G, Kong YW, Morandell S, Merrick KA, Hernandez Y, Haigis KM and Yaffe MB: MK2 contributes to tumor progression by promoting M2 macrophage polarization and tumor angiogenesis. Proc Natl Acad Sci USA. 115:E4236–E4244. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhong X, Chen B and Yang Z: The role of Tumor-associated macrophages in colorectal carcinoma progression. Cell Physiol Biochem. 45:356–365. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao Y, Zhang W, Huo M, Wang P, Liu X, Wang Y, Li Y, Zhou Z, Xu N and Zhu H: XBP1 regulates the protumoral function of tumor-associated macrophages in human colorectal cancer. Signal Transduct Target Ther. 6:3572021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang XL, Hu LP, Yang Q, Qin WT, Wang X, Xu CJ, Tian GA, Yang XM, Yao LL, Zhu L, et al: CTHRC1 promotes liver metastasis by reshaping infiltrated macrophages through physical interactions with TGF-β receptors in colorectal cancer. Oncogene. 40:3959–3973. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Huang C, Ou R, Chen X, Zhang Y, Li J, Liang Y, Zhu X, Liu L, Li M, Lin D, et al: Tumor cell-derived SPON2 promotes M2-polarized tumor-associated macrophage infiltration and cancer progression by activating PYK2 in CRC. J Exp Clin Cancer Res. 40:3042021. View Article : Google Scholar : PubMed/NCBI | |
|
Wang X, Wang J, Zhao J, Wang H, Chen J and Wu J: HMGA2 facilitates colorectal cancer progression via STAT3-mediated tumor-associated macrophage recruitment. Theranostics. 12:963–975. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Tu W, Gong J, Zhou Z, Tian D and Wang Z: TCF4 enhances hepatic metastasis of colorectal cancer by regulating tumor-associated macrophage via CCL2/CCR2 signaling. Cell Death Dis. 12:8822021. View Article : Google Scholar : PubMed/NCBI | |
|
Grossman JG, Nywening TM, Belt BA, Panni RZ, Krasnick BA, DeNardo DG, Hawkins WG, Goedegebuure SP, Linehan DC and Fields RC: Recruitment of CCR2+ tumor associated macrophage to sites of liver metastasis confers a poor prognosis in human colorectal cancer. Oncoimmunology. 7:e14707292018. View Article : Google Scholar : PubMed/NCBI | |
|
Xu C, Fan L, Lin Y, Shen W, Qi Y, Zhang Y, Chen Z, Wang L, Long Y, Hou T, et al: Fusobacterium nucleatum promotes colorectal cancer metastasis through miR-1322/CCL20 axis and M2 polarization. Gut Microbes. 13:19803472021. View Article : Google Scholar : PubMed/NCBI | |
|
Ohashi K, Wang Z, Yang YM, Billet S, Tu W, Pimienta M, Cassel SL, Pandol SJ, Lu SC, Sutterwala FS, et al: NOD-like receptor C4 inflammasome regulates the growth of colon cancer liver metastasis in NAFLD. Hepatology. 70:1582–1599. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou J, Song Q, Li H, Han Y, Pu Y, Li L, Rong W, Liu X, Wang Z, Sun J, et al: Targeting circ-0034880-enriched tumor extracellular vesicles to impede SPP1highCD206+ pro-tumor macrophages mediated pre-metastatic niche formation in colorectal cancer liver metastasis. Mol Cancer. 23:1682024. View Article : Google Scholar : PubMed/NCBI | |
|
Shao Y, Chen T, Zheng X, Yang S, Xu K, Chen X, Xu F, Wang L, Shen Y, Wang T, et al: Colorectal cancer-derived small extracellular vesicles establish an inflammatory premetastatic niche in liver metastasis. Carcinogenesis. 39:1368–1379. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Takano Y, Masuda T, Iinuma H, Yamaguchi R, Sato K, Tobo T, Hirata H, Kuroda Y, Nambara S, Hayashi N, et al: Circulating exosomal microRNA-203 is associated with metastasis possibly via inducing tumor-associated macrophages in colorectal cancer. Oncotarget. 8:78598–78613. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao S, Mi Y, Guan B, Zheng B, Wei P, Gu Y, Zhang Z, Cai S, Xu Y, Li X, et al: Tumor-derived exosomal miR-934 induces macrophage M2 polarization to promote liver metastasis of colorectal cancer. J Hematol Oncol. 13:1562020. View Article : Google Scholar : PubMed/NCBI | |
|
Sun H, Meng Q, Shi C, Yang H, Li X, Wu S, Familiari G, Relucenti M, Aschner M, Wang X and Chen R: Hypoxia-inducible exosomes facilitate liver-tropic premetastatic niche in colorectal cancer. Hepatology. 74:2633–2651. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Li S, Fu X, Ning D, Liu Q, Zhao J, Cheng Q, Chen X and Jiang L: Colon cancer exosome-associated HSP90B1 initiates pre-metastatic niche formation in the liver by polarizing M1 macrophage into M2 phenotype. Biol Direct. 20:522025. View Article : Google Scholar : PubMed/NCBI | |
|
Liang Y, Li J, Yuan Y, Ju H, Liao H, Li M, Liu Y, Yao Y, Yang L, Li T and Lei X: Exosomal miR-106a-5p from highly metastatic colorectal cancer cells drives liver metastasis by inducing macrophage M2 polarization in the tumor microenvironment. J Exp Clin Cancer Res. 43:2812024. View Article : Google Scholar : PubMed/NCBI | |
|
Wei X, Ye J, Pei Y, Wang C, Yang H, Tian J, Si G, Ma Y, Wang K and Liu G: Extracellular vesicles from colorectal cancer cells promote metastasis via the NOD1 signalling pathway. J Extracell Vesicles. 11:e122642022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Zhang Q, Xing B, Luo N, Gao R, Yu K, Hu X, Bu Z, Peng J, Ren X and Zhang Z: Immune phenotypic linkage between colorectal cancer and liver metastasis. Cancer Cell. 40:424–437.e5. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Wu Y, Yang S, Ma J, Chen Z, Song G, Rao D, Cheng Y, Huang S, Liu Y, Jiang S, et al: Spatiotemporal immune landscape of colorectal cancer liver metastasis at Single-cell level. Cancer Discov. 12:134–153. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Eruslanov EB, Bhojnagarwala PS, Quatromoni JG, Stephen TL, Ranganathan A, Deshpande C, Akimova T, Vachani A, Litzky L, Hancock WW, et al: Tumor-associated neutrophils stimulate T cell responses in early-stage human lung cancer. J Clin Invest. 124:5466–5480. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Fridlender ZG, Sun J, Kim S, Kapoor V, Cheng G, Ling L, Worthen GS and Albelda SM: Polarization of tumor-associated neutrophil phenotype by TGF-beta: ‘N1’ versus ‘N2’ TAN. Cancer Cell. 16:183–194. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Germann M, Zangger N, Sauvain MO, Sempoux C, Bowler AD, Wirapati P, Kandalaft LE, Delorenzi M, Tejpar S, Coukos G and Radtke F: Neutrophils suppress tumor-infiltrating T cells in colon cancer via matrix metalloproteinase-mediated activation of TGFβ. EMBO Mol Med. 12:e106812020. View Article : Google Scholar : PubMed/NCBI | |
|
Itatani Y, Yamamoto T, Zhong C, Molinolo AA, Ruppel J, Hegde P, Taketo MM and Ferrara N: Suppressing neutrophil-dependent angiogenesis abrogates resistance to anti-VEGF antibody in a genetic model of colorectal cancer. Proc Natl Acad Sci USA. 117:21598–21608. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Gordon-Weeks AN, Lim SY, Yuzhalin AE, Jones K, Markelc B, Kim KJ, Buzzelli JN, Fokas E, Cao Y, Smart S and Muschel R: Neutrophils promote hepatic metastasis growth through fibroblast growth factor 2-dependent angiogenesis in mice. Hepatology. 65:1920–1935. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Yang L, Liu L, Zhang R, Hong J, Wang Y, Wang J, Zuo J, Zhang J, Chen J and Hao H: IL-8 mediates a positive loop connecting increased neutrophil extracellular traps (NETs) and colorectal cancer liver metastasis. J Cancer. 11:4384–4396. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Tan H, Jiang Y, Shen L, Nuerhashi G, Wen C, Gu L, Wang Y, Qi H, Cao F, Huang T, et al: Cryoablation-induced neutrophil Ca2+ elevation and NET formation exacerbate immune escape in colorectal cancer liver metastasis. J Exp Clin Cancer Res. 43:3192024. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang Y, Long G, Huang X, Wang W, Cheng B and Pan W: Single-cell transcriptomic analysis reveals dynamic changes in the liver microenvironment during colorectal cancer metastatic progression. J Transl Med. 23:3362025. View Article : Google Scholar : PubMed/NCBI | |
|
Seubert B, Grünwald B, Kobuch J, Cui H, Schelter F, Schaten S, Siveke JT, Lim NH, Nagase H, Simonavicius N, et al: Tissue inhibitor of metalloproteinases (TIMP)-1 creates a premetastatic niche in the liver through SDF-1/CXCR4-dependent neutrophil recruitment in mice. Hepatology. 61:238–248. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Wang H, Zhang B, Li R, Chen J, Xu G, Zhu Y, Li J, Liang Q, Hua Q, Wang L, et al: KIAA1199 drives immune suppression to promote colorectal cancer liver metastasis by modulating neutrophil infiltration. Hepatology. 76:967–981. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Wu J, Song J, Ge Y, Hou S, Chang Y, Chen X, Nie Z, Guo L and Yin J: PRIM1 enhances colorectal cancer liver metastasis via promoting neutrophil recruitment and formation of neutrophil extracellular trap. Cell Signal. 132:1118222025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang QQ, Hu XW, Liu YL, Ye ZJ, Gui YH, Zhou DL, Qi CL, He XD, Wang H and Wang LJ: CD11b deficiency suppresses intestinal tumor growth by reducing myeloid cell recruitment. Sci Rep. 5:159482015. View Article : Google Scholar : PubMed/NCBI | |
|
Cao X, Lan Q, Xu H, Liu W, Cheng H, Hu X, He J, Yang Q, Lai W and Chu Z: Granulocyte-like myeloid-derived suppressor cells: The culprits of neutrophil extracellular traps formation in the pre-metastatic niche. Int Immunopharmacol. 143:1135002024. View Article : Google Scholar : PubMed/NCBI | |
|
Lim SY, Gordon-Weeks AN, Zhao L, Tapmeier TT, Im JH, Cao Y, Beech J, Allen D, Smart S and Muschel RJ: Recruitment of myeloid cells to the tumor microenvironment supports liver metastasis. Oncoimmunology. 2:e231872013. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao L, Lim SY, Gordon-Weeks AN, Tapmeier TT, Im JH, Cao Y, Beech J, Allen D, Smart S and Muschel RJ: Recruitment of a myeloid cell subset (CD11b/Gr1 mid) via CCL2/CCR2 promotes the development of colorectal cancer liver metastasis. Hepatology. 57:829–839. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Chun E, Lavoie S, Michaud M, Gallini CA, Kim J, Soucy G, Odze R, Glickman JN and Garrett WS: CCL2 promotes colorectal carcinogenesis by enhancing polymorphonuclear myeloid-derived suppressor cell population and function. Cell Rep. 12:244–257. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Inamoto S, Itatani Y, Yamamoto T, Minamiguchi S, Hirai H, Iwamoto M, Hasegawa S, Taketo MM, Sakai Y and Kawada K: Loss of SMAD4 promotes colorectal cancer progression by accumulation of myeloid-derived suppressor cells through the CCL15-CCR1 chemokine axis. Clin Cancer Res. 22:492–501. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Wang D, Sun H, Wei J, Cen B and DuBois RN: CXCL1 is critical for premetastatic niche formation and metastasis in colorectal cancer. Cancer Res. 77:3655–3665. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Dang Y, Yu J, Zhao S, Cao X and Wang Q: HOXA7 promotes the metastasis of KRAS mutant colorectal cancer by regulating myeloid-derived suppressor cells. Cancer Cell Int. 22:882022. View Article : Google Scholar : PubMed/NCBI | |
|
Ren X, Xiao J, Zhang W, Wang F, Yan Y, Wu X, Zeng Z, He Y, Yang W, Liao W, et al: Inhibition of CCL7 derived from Mo-MDSCs prevents metastatic progression from latency in colorectal cancer. Cell Death Dis. 12:4842021. View Article : Google Scholar : PubMed/NCBI | |
|
Lin Q, Ren L, Jian M, Xu P, Li J, Zheng P, Feng Q, Yang L, Ji M, Wei Y and Xu J: The mechanism of the premetastatic niche facilitating colorectal cancer liver metastasis generated from myeloid-derived suppressor cells induced by the S1PR1-STAT3 signaling pathway. Cell Death Dis. 10:6932019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Davis C, Shah S, Hughes D, Ryan JC, Altomare D and Peña MM: IL-33 promotes growth and liver metastasis of colorectal cancer in mice by remodeling the tumor microenvironment and inducing angiogenesis. Mol Carcinog. 56:272–287. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Gu Y, Mi Y, Cao Y, Yu K, Zhang Z, Lian P, Li D, Qin J and Zhao S: The lncRNA MIR181A1HG in extracellular vesicles derived from highly metastatic colorectal cancer cells promotes liver metastasis by remodeling the extracellular matrix and recruiting myeloid-derived suppressor cells. Cell Biosci. 15:232025. View Article : Google Scholar : PubMed/NCBI | |
|
Kobie JJ, Wu RS, Kurt RA, Lou S, Adelman MK, Whitesell LJ, Ramanathapuram LV, Arteaga CL and Akporiaye ET: Transforming growth factor beta inhibits the antigen-presenting functions and antitumor activity of dendritic cell vaccines. Cancer Res. 63:1860–1864. 2003.PubMed/NCBI | |
|
Orsini G, Legitimo A, Failli A, Ferrari P, Nicolini A, Spisni R, Miccoli P and Consolini R: Defective generation and maturation of dendritic cells from monocytes in colorectal cancer patients during the course of disease. Int J Mol Sci. 14:22022–22041. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Nagorsen D, Voigt S, Berg E, Stein H, Thiel E and 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. 5:622007. View Article : Google Scholar : PubMed/NCBI | |
|
Hsu YL, Chen YJ, Chang WA, Jian SF, Fan HL, Wang JY and Kuo PL: Interaction between tumor-associated dendritic cells and colon cancer cells contributes to tumor progression via CXCL1. Int J Mol Sci. 19:24272018. View Article : Google Scholar : PubMed/NCBI | |
|
Huang TX, Tan XY, Huang HS, Li YT, Liu BL, Liu KS, Chen X, Chen Z, Guan XY, Zou C and Fu L: Targeting cancer-associated fibroblast-secreted WNT2 restores dendritic cell-mediated antitumour immunity. Gut. 71:333–344. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Sun Y, Hu H, Liu Z, Xu J, Gao Y, Zhan X, Zhou S, Zhong W, Wu D, Wang P, et al: Macrophage STING signaling promotes NK cell to suppress colorectal cancer liver metastasis via 4-1BBL/4-1BB co-stimulation. J Immunother Cancer. 11:e0064812023. View Article : Google Scholar : PubMed/NCBI | |
|
Donadon M, Hudspeth K, Cimino M, Di Tommaso L, Preti M, Tentorio P, Roncalli M, Mavilio D and Torzilli G: Increased infiltration of natural killer and T cells in colorectal liver metastases improves patient overall survival. J Gastrointest Surg. 21:1226–1236. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Dupaul-Chicoine J, Arabzadeh A, Dagenais M, Douglas T, Champagne C, Morizot A, Rodrigue-Gervais IG, Breton V, Colpitts SL, Beauchemin N and Saleh M: The Nlrp3 inflammasome suppresses colorectal cancer metastatic growth in the liver by promoting natural killer cell tumoricidal activity. Immunity. 43:751–763. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Takeda K, Hayakawa Y, Smyth MJ, Kayagaki N, Yamaguchi N, Kakuta S, Iwakura Y, Yagita H and Okumura K: Involvement of tumor necrosis factor-related apoptosis-inducing ligand in surveillance of tumor metastasis by liver natural killer cells. Nat Med. 7:94–100. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Russo E, D'Aquino C, Di Censo C, Laffranchi M, Tomaipitinca L, Licursi V, Garofalo S, Promeuschel J, Peruzzi G, Sozio F, et al: Cxcr3 promotes protection from colorectal cancer liver metastasis by driving NK cell infiltration and plasticity. J Clin Invest. 135:e1840362025. View Article : Google Scholar : PubMed/NCBI | |
|
Harmon C, Robinson MW, Hand F, Almuaili D, Mentor K, Houlihan DD, Hoti E, Lynch L, Geoghegan J and O'Farrelly C: Lactate-mediated acidification of tumor microenvironment induces apoptosis of liver-resident NK cells in colorectal liver metastasis. Cancer Immunol Res. 7:335–346. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Fang H, Dai W, Gu R, Zhang Y, Li J, Luo W, Tong S, Han L, Wang Y, Jiang C, et al: myCAF-derived exosomal PWAR6 accelerates CRC liver metastasis via altering glutamine availability and NK cell function in the tumor microenvironment. J Hematol Oncol. 17:1262024. View Article : Google Scholar : PubMed/NCBI | |
|
Matsumura H, Kondo T, Ogawa K, Tamura T, Fukunaga K, Murata S and Ohkohchi N: Kupffer cells decrease metastasis of colon cancer cells to the liver in the early stage. Int J Oncol. 45:2303–2310. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Wortzel I, Seo Y, Akano I, Shaashua L, Tobias GC, Hebert J, Kim KA, Kim D, Dror S, Liu Y, et al: Unique structural configuration of EV-DNA primes Kupffer cell-mediated antitumor immunity to prevent metastatic progression. Nat Cancer. 5:1815–1833. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Lu WP, Liu YD, Zhang ZF, Liu J, Ye JW, Wang SY, Lin XY, Lai YR, Li J, Liu SY, et al: m6A-modified MIR670HG suppresses tumor liver metastasis through enhancing Kupffer cell phagocytosis. Cell Mol Life Sci. 82:1852025. View Article : Google Scholar : PubMed/NCBI | |
|
Li J, Liu XG, Ge RL, Yin YP, Liu YD, Lu WP, Huang M, He XY, Wang J, Cai G, et al: The ligation between ERMAP, galectin-9 and dectin-2 promotes Kupffer cell phagocytosis and antitumor immunity. Nat Immunol. 24:1813–1824. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Bresesti C, Carito E, Notaro M, Giacca G, Breggion S, Kerzel T, Mercado CM, Beretta S, Monti M, Merelli I, et al: Reprogramming liver metastasis-associated macrophages toward an anti-tumoral phenotype through enforced miR-342 expression. Cell Rep. 44:1155922025. View Article : Google Scholar : PubMed/NCBI | |
|
Nater M, Brügger M, Cecconi V, Pereira P, Forni G, Köksal H, Dimakou D, Herbst M, Calvanese AL, Lucchiari G, et al: Hepatic iNKT cells facilitate colorectal cancer metastasis by inducing a fibrotic niche in the liver. iScience. 28:1123642025. View Article : Google Scholar : PubMed/NCBI | |
|
Gassmann P, Hemping-Bovenkerk A, Mees ST and Haier J: Metastatic tumor cell arrest in the liver-lumen occlusion and specific adhesion are not exclusive. Int J Colorectal Dis. 24:851–858. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Haier J, Korb T, Hotz B, Spiegel HU and Senninger N: An intravital model to monitor steps of metastatic tumor cell adhesion within the hepatic microcirculation. J Gastrointest Surg. 7:507–515. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Khatib AM, Fallavollita L, Wancewicz EV, Monia BP and Brodt P: Inhibition of hepatic endothelial E-selectin expression by C-raf antisense oligonucleotides blocks colorectal carcinoma liver metastasis. Cancer Res. 62:5393–5398. 2002.PubMed/NCBI | |
|
Khatib AM, Auguste P, Fallavollita L, Wang N, Samani A, Kontogiannea M, Meterissian S and Brodt P: Characterization of the host proinflammatory response to tumor cells during the initial stages of liver metastasis. Am J Pathol. 167:749–759. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Huang WH, Zhou MW, Zhu YF, Xiang JB, Li ZY, Wang ZH, Zhou YM, Yang Y, Chen ZY and Gu XD: The role of hepatic stellate cells in promoting liver metastasis of colorectal carcinoma. Onco Targets Ther. 12:7573–7580. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zeng X, Zhou J, Xiong Z, Sun H, Yang W, Mok MTS, Wang J, Li J, Liu M, Tang W, et al: Cell cycle-related kinase reprograms the liver immune microenvironment to promote cancer metastasis. Cell Mol Immunol. 18:1005–1015. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Yang Y, Chen Y, Liu Z, Chang Z, Sun Z and Zhao L: Concomitant NAFLD facilitates liver metastases and PD-1-refractory by recruiting MDSCs via CXCL5/CXCR2 in Colorectal Cancer. Cell Mol Gastroenterol Hepatol. 18:1013512024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Z, Kim SY, Tu W, Kim J, Xu A, Yang YM, Matsuda M, Reolizo L, Tsuchiya T, Billet S, et al: Extracellular vesicles in fatty liver promote a metastatic tumor microenvironment. Cell Metab. 35:1209–1226.e13. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Ruff SM, Brown ZJ and Pawlik TM: A review of targeted therapy and immune checkpoint inhibitors for metastatic colorectal cancer. Surg Oncol. 51:1019932023. View Article : Google Scholar : PubMed/NCBI | |
|
Hernandez Dominguez O, Yilmaz S and Steele SR: Stage IV colorectal cancer management and treatment. J Clin Med. 12:20722023. View Article : Google Scholar : PubMed/NCBI | |
|
Cheng XF, Zhao F, Chen D and Liu FL: Current landscape of preoperative neoadjuvant therapies for initial resectable colorectal cancer liver metastasis. World J Gastroenterol. 30:663–672. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Tatsuta K, Sakata M, Kojima T, Booka E, Kurachi K and Takeuchi H: Updated insights into the impact of adjuvant chemotherapy on recurrence and survival after curative resection of liver or lung metastases in colorectal cancer: A rapid review and meta-analysis. World J Surg Oncol. 23:562025. View Article : Google Scholar : PubMed/NCBI | |
|
Yarom N and Jonker DJ: The role of the epidermal growth factor receptor in the mechanism and treatment of colorectal cancer. Discov Med. 11:95–105. 2011.PubMed/NCBI | |
|
Jonker DJ, O'Callaghan CJ, Karapetis CS, Zalcberg JR, Tu D, Au HJ, Au HJ, Berry SR, Krahn M, Price T, et al: Cetuximab for the treatment of colorectal cancer. N Engl J Med. 357:2040–2048. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Van Cutsem E, Köhne CH, Hitre E, Zaluski J, Chang Chien CR, Makhson A, D'Haens G, Pintér T, Lim R, Bodoky G, et al: Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med. 360:1408–1417. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Bokemeyer C, Bondarenko I, Makhson A, Hartmann JT, Aparicio J, de Braud F, Donea S, Ludwig H, Schuch G, Stroh C, et al: Fluorouracil, leucovorin, and oxaliplatin with and without cetuximab in the first-line treatment of metastatic colorectal cancer. J Clin Oncol. 27:663–671. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, Valtorta E, Schiavo R, Buscarino M, Siravegna G, et al: Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature. 486:532–536. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Douillard JY, Siena S, Cassidy J, Tabernero J, Burkes R, Barugel M, Humblet Y, Bodoky G, Cunningham D, Jassem J, et al: Randomized, phase III trial of panitumumab with infusional fluorouracil, leucovorin, and oxaliplatin (FOLFOX4) versus FOLFOX4 alone as first-line treatment in patients with previously untreated metastatic colorectal cancer: The PRIME study. J Clin Oncol. 28:4697–4705. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Douillard JY, Siena S, Cassidy J, Tabernero J, Burkes R, Barugel M, Humblet Y, Bodoky G, Cunningham D, Jassem J, et al: Final results from PRIME: Randomized phase III study of panitumumab with FOLFOX4 for first-line treatment of metastatic colorectal cancer. Ann Oncol. 25:1346–1355. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Choi HY and Chang JE: Targeted therapy for cancers: From ongoing clinical trials to FDA-Approved drugs. Int J Mol Sci. 24:136182023. View Article : Google Scholar : PubMed/NCBI | |
|
Cao D, Zheng Y, Xu H, Ge W and Xu X: Bevacizumab improves survival in metastatic colorectal cancer patients with primary tumor resection: A meta-analysis. Sci Rep. 9:203262019. View Article : Google Scholar : PubMed/NCBI | |
|
Tang W, Ren L, Liu T, Ye Q, Wei Y, He G, Lin Q, Wang X, Wang M, Liang F, et al: 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. 38:3175–3184. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Debeuckelaere C, Murgioni S, Lonardi S, Girardi N, Alberti G, Fano C, Gallimberti S, Magro C, Ahcene-Djaballah S, Daniel F, et al: Ramucirumab: The long and winding road toward being an option for mCRC treatment. Expert Opin Biol Ther. 19:399–409. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Tabernero J, Yoshino T, Cohn AL, Obermannova R, Bodoky G, Garcia-Carbonero R, Ciuleanu TE, Portnoy DC, 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. 16:499–508. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Grothey A, Van Cutsem E, Sobrero A, Siena S, Falcone A, Ychou M, Humblet Y, Bouché O, Mineur L, Barone C, et al: Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): An international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet. 381:303–312. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Goel G: Evolution of regorafenib from bench to bedside in colorectal cancer: Is it an attractive option or merely a ‘me too’ drug? Cancer Manag Res. 10:425–437. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Zou JY, Wang Z and Wang Y: Fruquintinib: A novel antivascular endothelial growth factor receptor tyrosine kinase inhibitor for the treatment of metastatic colorectal cancer. Cancer Manag Res. 11:7787–7803. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Li J, Qin S, Xu RH, Shen L, Xu J, Bai Y, Yang L, Deng Y, Chen ZD, Zhong H, et al: Effect of fruquintinib vs placebo on overall survival in patients with previously treated metastatic colorectal cancer: The FRESCO randomized clinical trial. JAMA. 319:2486–2496. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Dasari A, Lonardi S, Garcia-Carbonero R, Elez E, Yoshino T, Sobrero A, Yao J, García-Alfonso P, Kocsis J, Cubillo Gracian A, et al: Fruquintinib versus placebo in patients with refractory metastatic colorectal cancer (FRESCO-2): An international, multicentre, randomised, double-blind, phase 3 study. Lancet. 402:41–53. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Fusco MJ, Casak SJ, Mushti SL, Cheng J, Christmas BJ, Thompson MD, Fu W, Wang H, Yoon M, Yang Y, et al: FDA approval summary: Fruquintinib for the treatment of refractory metastatic colorectal cancer. Clin Cancer Res. 30:3100–3104. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Mc Neil V and Lee SW: Advancing cancer treatment: A review of immune checkpoint inhibitors and combination STrategies. Cancers (Basel). 17:14082025. View Article : Google Scholar : PubMed/NCBI | |
|
Overman MJ, McDermott R, Leach JL, Lonardi S, Lenz HJ, Morse MA, Desai J, Hill A, Axelson M, Moss RA, et al: Nivolumab in patients with metastatic DNA mismatch Repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): An open-label, multicentre, phase 2 study. Lancet Oncol. 18:1182–1191. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
André T, Shiu KK, Kim TW, Jensen BV, Jensen LH, 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. 383:2207–2218. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Weng J, Li S, Zhu Z, Liu Q, Zhang R, Yang Y and Li X: Exploring immunotherapy in colorectal cancer. J Hematol Oncol. 15:952022. View Article : Google Scholar : PubMed/NCBI | |
|
Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, et al: PD-1 Blockade in tumors with Mismatch-repair deficiency. N Engl J Med. 372:2509–2520. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Yu J, Green MD, Li S, Sun Y, Journey SN, Choi JE, Rizvi SM, Qin A, Waninger JJ, Lang X, et al: Liver metastasis restrains immunotherapy efficacy via macrophage-mediated T cell elimination. Nat Med. 27:152–164. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Saberzadeh-Ardestani B, Jones JC, McWilliams RR, Tougeron D, Halfdanarson TR, Guimbaud R, Hubbard JM, Flecchia C, Shi Q, Alouani E, et al: Metastatic site and clinical outcome of patients with deficient mismatch repair metastatic colorectal cancer treated with an immune checkpoint inhibitor in the first-line setting. Eur J Cancer. 196:1134332024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang C, Sandhu J, Ouyang C, Ye J, Lee PP and Fakih M: Clinical response to immunotherapy targeting programmed cell death Receptor 1/Programmed cell death Ligand 1 in patients with treatment-resistant microsatellite stable colorectal cancer with and without liver metastases. JAMA Netw Open. 4:e21184162021. View Article : Google Scholar : PubMed/NCBI | |
|
Overman MJ, Lonardi S, Wong KYM, Lenz HJ, Gelsomino F, Aglietta M, Morse MA, Van Cutsem E, McDermott R, Hill A, et al: Durable clinical benefit with nivolumab plus ipilimumab in DNA mismatch Repair-deficient/microsatellite instability-high metastatic colorectal cancer. J Clin Oncol. 36:773–779. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Fukuoka S, Hara H, Takahashi N, Kojima T, Kawazoe A, Asayama M, Yoshii T, Kotani D, Tamura H, Mikamoto Y, et al: Regorafenib plus nivolumab in patients with advanced gastric or colorectal cancer: An open-label, dose-escalation, and dose-expansion phase Ib trial (REGONIVO, EPOC1603). J Clin Oncol. 38:2053–2061. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Yang C, Zhao L, Lin Y, Wang S, Ye Y and Shen Z: Improving the efficiency of immune checkpoint inhibitors for metastatic pMMR/MSS colorectal cancer: Options and strategies. Crit Rev Oncol Hematol. 200:1042042024. View Article : Google Scholar : PubMed/NCBI | |
|
Gutiu AG, Zhao L, Marrah AJ, Maher AJ, Voss BB, Mayberry TG, Cowan BC, Wakefield MR and Fang Y: Promising immunotherapeutic treatments for colon cancer. Med Oncol. 42:1752025. View Article : Google Scholar : PubMed/NCBI | |
|
Kaviyarasan V, Das A, Deka D, Saha B, Banerjee A, Sharma NR, Duttaroy AK and Pathak S: Advancements in immunotherapy for colorectal cancer treatment: a comprehensive review of strategies, challenges, and future prospective. Int J Colorectal Dis. 40:12024. View Article : Google Scholar : PubMed/NCBI | |
|
Fatemi N, Mirbahari SN, Tierling S, Sanjabi F, Shahrivari S, AmeliMojarad M, Amelimojarad M, Mirzaei Rezaei M, Nobaveh P, Totonchi M and Nazemalhosseini Mojarad E: Emerging frontiers in colorectal cancer therapy: From targeted molecules to immunomodulatory breakthroughs and cell-based approaches. Dig Dis Sci. 70:919–942. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Liu N, Xiao X, Zhang Z, Mao C, Wan M and Shen J: Advances in cancer vaccine research. ACS Biomater Sci Eng. 9:5999–6023. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu C, Liu N, Zhang T and Tu Y: Adoptive immune cell therapy for colorectal cancer. Front Immunol. 16:15579062025. View Article : Google Scholar : PubMed/NCBI | |
|
Pérez-Domínguez F, Quezada-Monrás C, Cárcamo L, Muñoz JP and Carrillo-Beltrán D: Oncolytic viruses as a novel therapeutic approach for colorectal cancer: Mechanisms, current advances, and future directions. Cancers (Basel). 17:18542025. View Article : Google Scholar : PubMed/NCBI |