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.
Key immune cells in the tumor immune microenvironment of colorectal cancer: Roles and research advances (Review)
Colorectal cancer (CRC) is the third most common cancer globally and the second leading cause of cancer‑related mortalities. Surgery‑centered multimodal therapy remains the cornerstone of care, yet outcomes are poor in advanced or drug‑resistant disease. The tumor immune microenvironment (TIME), a network of immune cells, cytokines and stromal elements, shapes antitumor immunity and can either restrain or encourage tumor growth. Specific immune cells within the TIME influence CRC biology, while immune‑checkpoint blockade has delivered notable benefits, especially in microsatellite instability‑high tumors. The present review discusses the principal immune cell populations in the CRC TIME, outlines their mechanisms of action and discusses emerging cell‑based immunotherapies that may guide future precision treatment.
Colorectal cancer (CRC) is one of the most prevalent malignancies of the digestive system. According to global cancer statistics, >1.9 million new CRC cases and ~900,000 mortalities due to CRC were recorded in 2022, making it the third most commonly diagnosed cancer and the second leading cause of cancer-related mortality worldwide (1). The incidence and progression of CRC are influenced by various factors, including age, genetic predisposition, lifestyle choices and environmental exposures. Because early symptoms are subtle, several patients present with advanced disease or distant spread, leading to a poor outcome (2). Currently, the mainstay of CRC treatment is comprehensive therapy centered around surgical resection, complemented by radiotherapy, chemotherapy, targeted therapy and immunotherapy. Preoperative neoadjuvant therapies and postoperative adjuvant chemotherapy or radiotherapy have improved the chances of curative outcomes. Standard adjuvant chemotherapy regimens include oxaliplatin based xeloda and oxaliplatin and folinic acid and fluorouracil and oxaliplatin regimens (3,4). Additionally, targeted therapies focusing on EGFR and vascular endothelial growth factor (VEGF) pathways have demonstrated clinical benefit in select patient populations (5,6). Nevertheless, response rates are modest and adverse effects substantial (7–9).
Immunotherapy has transformed outcomes in several malignancies and is now a key focus in CRC. Immune checkpoint inhibitors (ICIs), in particular, have shown favorable efficacy in patients with microsatellite instability-high (MSI-H) CRC. Nonetheless, ~85% of CRC cases exhibit microsatellite stability (MSS) or proficient mismatch repair (pMMR) status, and these patients typically exhibit limited responses to ICIs (10). Thus, elucidating the immune microenvironment of CRC and developing novel immunotherapeutic strategies have emerged as key areas of current research.
The tumor microenvironment (TME) comprises various components associated with tumor tissues, carrying out key roles in tumor initiation and progression. Multiple immune cells, including but not limited to macrophages, T cells, B cells, natural killer (NK) cells, dendritic cells (DCs) and neutrophils, are recruited into the vicinity of tumor cells, interacting with extracellular matrix (ECM) components and cytokines to collectively form the tumor immune microenvironment (TIME) (11) (Fig. 1). These cells can mount potent antitumor responses yet also enable immune escape. Their relative abundance and functional state help determine tumor immunogenicity, therapy response and patient survival. Intensive research has explored the roles and regulatory mechanisms of various immune cells within the CRC TIME.
Macrophages are abundant and versatile residents of the CRC TIME (12). TAMs interact directly with tumor cells, secreting various growth factors, cytokines and ECM-modifying enzymes, thus promoting tumor growth, migration and invasion (13). TAMs also regulate immune responses within the TME, creating an immunosuppressive milieu conducive to tumor progression by inhibiting cytotoxic T lymphocytes, expanding regulatory T cells (Tregs) and NK cell functions. Moreover, macrophages release MMPs and other ECM remodeling factors, enhancing tumor cell migration (14).
Based on functional characteristics, TAMs are typically classified into pro-inflammatory M1 and anti-inflammatory M2 phenotypes. M1 macrophages, activated by IFN-γ, lipopolysaccharide or Toll-like receptor ligands, express markers such as major histocompatibility complex (MHC)-II, CD68, CD80 and CD86. They enhance anti-tumor immune responses through secretion of cytokines including IL-12, TNF-α and IFN-γ, and directly exert cytotoxic effects on tumor cells via reactive oxygen species and nitric oxide (NO) production (15–19). Conversely, M2 macrophages exhibit immunosuppressive roles, expressing inhibitory receptors such as programmed death-ligand 1 (PD-L1) and B7-H3 (20–22). Activated by Th2 cytokines IL-4 and IL-13, M2 macrophages express CD163, CD206 and ARG1, and secrete cytokines such as IL-10, TGF-β and VEGF, suppressing T cell activity, promoting angiogenesis and facilitating immune escape (Fig. 2) (23–25).
High infiltration of M1 macrophages associates with improved prognosis in CRC. Studies have shown an inverse relationship between the proportion of M1 macrophages and lymph node and distant metastases (26–28). Wang et al (29) reported that ANKRD22 expression in M1 macrophages positively associates with survival and macrophage infiltration levels in patients with colon cancer. Nussbaum et al (30)Q analyzing single-cell RNA sequencing (scRNA-seq) data from 23 patients with CRC, identified high SPP1-CD44 interactions within inflammatory macrophages. Targeting SPP1+ macrophages exhibiting an anti-inflammatory phenotype could disrupt these interactions, potentially reducing tumor progression and immunosuppression.
Conversely, M2 macrophage infiltration in CRC is associated with tumor progression and immune evasion. Their presence associates notably with increased tumor aggressiveness, metastatic potential and immunosuppressive capabilities (31,32). Tumor-derived exosomes serve as key mediators of intercellular communication, facilitating tumor-macrophage interactions. After internalization by macrophages, these exosomes activate polarization pathways, driving M2 macrophage polarization, creating pre-metastatic niches and subsequently promoting CRC metastasis (33–36). M2 macrophages further enhance tumor invasion and metastasis by secreting pro-tumor factors such as VEGF, MMPs, IL-4 and IL-13, contributing to angiogenesis and ECM degradation (37–39). Additionally, M2 macrophages facilitate immune escape through expression of PD-L1, engaging with PD-1 and cytotoxic T-lymphocyte associated protein 4 (CTLA-4), impairing T-cell receptor (TCR) signaling and inhibiting cytotoxic T cell function (40). They also expand Tregs via TGF-β secretion, further suppressing CD8+ T cell activities and promoting immune evasion (41).
Studies have demonstrated the dual influence of TAMs on the effectiveness of radiotherapy and chemotherapy, either antagonizing therapeutic efficacy by facilitating tissue repair and immunosuppression or enhancing overall antitumor effects (42,43). De Palma et al (44) found that TAMs also drive repair mechanisms following vascular-targeted therapy. Furthermore, low-dose γ irradiation of macrophages during neoadjuvant therapy can induce vascular normalization and counteract immunosuppression, diminishing pro-tumor effects of TAMs (45). Studies indicate that ICIs, such as PD-1/PD-L1 inhibitors and agents such as selicrelumab, can enhance macrophage phagocytosis, reducing tumor burden (46,47). Current clinical trials targeting TAMs primarily follow three major strategies, with differential therapeutic efficacy revealing distinct underlying mechanisms of action. First, strategy targeting the C-C motif chemokine receptor 2/C-C motif chemokine ligand 2 or colony-stimulating factor 1 receptor (CSF1R) axis represent a common therapeutic approach. This strategy inhibits TAM recruitment, effectively reducing their overall density within the TIME. Published results from clinical studies largely support the feasibility of this approach (48–51).
However, likely due to limited anti-tumor clinical activity, the majority of ongoing trials in solid tumors are Phase I/II studies focusing on safety and preliminary efficacy (50,52,53). This constrained efficacy may be partly attributed to the concurrent suppression of potentially anti-tumor M1-type TAM recruitment, thereby weakening anti-tumor immunity. Furthermore, the high plasticity of TAMs may trigger compensatory activation of other pro-tumor signaling pathways upon CSF1R inhibition, potentially restoring the pro-tumor functions of TAMs and diminishing therapeutic effectiveness (54). Second, blocking the ‘don't eat me’ signal using anti-CD47/SIRPα antibodies to activate macrophage phagocytosis constitutes another common strategy. This approach bypasses the dependency on TAM quantity and directly enhances their intrinsic tumor-killing capacity (55). Results from the NCT02953782 trial demonstrated that combined anti-CD47 and anti-EGFR therapy was tolerable and showed potential anti-tumor activity in heavily pre-treated patients with CRC, despite no notable changes in immune cell infiltration levels being observed (56). Additionally, since CD47 is involved in maintaining erythrocyte homeostasis, off-target toxicity of CD47 antibodies often limits the efficacy of monotherapy (57). Third, Chimeric Antigen Receptor Macrophage (CAR-M) therapy, which utilizes genetically engineered macrophages, represents a precision empowerment strategy. This approach not only enables specific tumor antigen recognition but may also promote macrophage polarization towards the M1 phenotype upon activation. The subsequent secretion of pro-inflammatory cytokines such as IL-12 could potentially convert ‘cold’ tumors into ‘hot’ tumors. Clinical trial results (NCT04660929) indicated that CAR-M therapy is safe and feasible in patients with HER2-overexpressing solid tumors and can induce TME remodeling and anti-tumor immune responses (58), offering a novel strategy to address prevalent immunosuppression in CRC (Table I).
Substantial advancements have been realized in the field of cancer immunotherapy over recent years (59–61). Although numerous early-phase clinical trials have explored targeting TAMs, these studies (62–64) have revealed several key and unresolved challenges: i) The lack of predictive biomarkers to prospectively identify patients who are likely to benefit; ii) difficulties in managing toxicities associated with combination therapies, as well as uncertainties regarding the optimal sequencing of treatments and their underlying mechanistic basis; iii) obstacles in genetically engineering macrophages, including their poor expansion in vitro and limited self-renewal capacity following adoptive transfer. Therefore, future efforts should focus on integrating specific biomarkers to identify patients most likely to benefit, and exploring combination regimens with chemotherapy, radiotherapy, immunotherapy or therapies targeting other immune cells. This strategy is essential to overcome treatment resistance and maximize therapeutic efficacy.
T cells are prominent immune cell populations within the CRC TIME, playing essential roles in tumor defense. They are categorized primarily as CD8+ cytotoxic T cells and CD4+ helper T cells based on functional differentiation. CD8+ T cells serve as key effectors in adaptive immunity, recognizing tumor antigens presented via MHC class I molecules and directly killing tumor cells through secretion of perforin and granzyme B (65). They also release IFN-γ and TNF-α, strengthening local immune responses and initiating apoptosis via the Fas/FasL signaling pathway (66). Increased infiltration of activated CD8+ T cells is associated with improved patient prognosis (67).
Multiple factors suppress CD8+ T cell functionality in CRC. High expression of checkpoint molecules such as PD-1 and CTLA-4 within the tumor environment induces CD8+ T cell exhaustion (68). In addition, the peripheral blood count of CD8+ T lymphocytes in patients with CRC is markedly reduced compared with that observed in healthy individuals (69). Exhausted T cells (Tex) arise from chronic antigen stimulation, exhibiting impaired responsiveness (70,71). Early-stage Tex cells retain proliferative and differentiation capacities and can partially recover with ICIs, while terminal Tex cells remain largely irreversible (72,73). Tex cells express high levels of inhibitory receptors, losing proliferative and cytotoxic abilities (74–76). Single-cell studies indicate that baseline Tex proportions and dynamic changes following PD-1 blockade can predict immunotherapy outcomes (77–79). Additional immunosuppressive factors, such as Tregs, MDSCs, TAM-derived cytokines such as IL-10, TGF-β and metabolic by-products lactate, further impair CD8+ T cell functions (25,80–82). PD-1/PD-L1 inhibitors demonstrate notable efficacy in MSI-H CRC by restoring CD8+ T cell activity, however, their efficacy remains limited in MSS CRC due to lower T-cell infiltration (83,84). Thus, combination therapies involving cancer vaccines or engineered T-cell therapies (CAR-T/TCR-T) might enhance antitumor responses.
CD4+ T helper cells modulate immune responses by regulating CD8+ T and B cell activities, differentiating into Th1, Th2, Th17 and Treg subsets. Th1 cells promote antitumor immunity through IFN-γ and IL-2 production (85–87), whereas Th2 cells facilitate tumor progression via pro-inflammatory cytokines IL-4, IL-5 and IL-13 (88,89). The role of Th17 cells remains controversial, potentially promoting or inhibiting CRC progression through cytokines such as IL-17 and IL-22 (90–94). Tregs, characterized by Foxp3 expression, suppress immune responses via cytokines such as TGF-β, IL-10 and IL-35, associating with poor prognosis and increased metastasis in CRC (95–100). Thus, targeting Treg metabolism or TGF-β pathways may enhance anti-tumor immunity in CRC.
B cells represent a key component of adaptive immunity, mediating anti-tumor responses primarily through antibody production, antigen presentation and the promotion of T cell activation (101). The antibodies secreted by B cells can bind to tumor cells, facilitating cytotoxic activities mediated by NK cells, macrophages and the complement system. Additionally, B cells function as antigen-presenting cells, stimulating CD4+ T cells via MHC class II molecules, thereby enhancing antitumor immune responses (102). However, the role of B cells within the TME is not exclusively protective.
Certain subsets of B cells, particularly regulatory B cells (Bregs), may promote tumor progression (103). Bregs secrete immunosuppressive cytokines such as IL-10 and TGF-β, inhibiting CD8+ T cell and NK cell functions, thus facilitating tumor immune escape (104). Moreover, B cells can exacerbate tumor-associated inflammation by producing pro-inflammatory cytokines, including IL-6, thereby enhancing cancer cell proliferation and metastasis (105). Clinical studies demonstrate that increased infiltration of Bregs in CRC tissues associates positively with tumor progression and predicts poor patient prognosis (106,107).
Targeting B cells for immunomodulation has emerged as a novel direction for CRC treatment. On one hand, enhancing antigen-specific B cell responses, such as developing B-cell-based vaccines, might potentiate antitumor immunity (108). On the other hand, inhibiting Breg functions to reduce their immunosuppressive effects could improve responses to immunotherapy. Monoclonal antibodies targeting Breg-specific markers, such as CD19 or CD38, have been proposed as promising therapeutic approaches (109,110). Additionally, interactions between B cells and T cells are key within the CRC immune microenvironment (111). Thus, future therapeutic strategies should comprehensively consider dynamic interactions among various immune cell subsets to optimize immunotherapy outcomes.
NK cells constitute essential components of innate immunity, capable of directly recognizing and killing tumor cells independently of antigen presentation. NK cells induce apoptosis in target cells through the secretion of perforin and granzyme B, and enhance antitumor immune responses via IFN-γ production (112). Additionally, NK cells contribute markedly to antibody-dependent cellular cytotoxicity, enhancing the efficacy of antibody-based therapies (113). NK cells are principally stratified into two distinct functional subsets contingent upon the surface density of CD56 expression: The CD56dim and CD56bright subpopulations. The former constitutes the numerically predominant fraction, characterized by potent cytotoxicity and serving as the primary effector killer cells; in contradistinction, the latter subpopulation is specialized chiefly in cytokine secretion and the orchestration of immune responses (114).
NK cell activity is notably impaired. CRC tissues often exhibit reduced NK cell infiltration and functionality due to immunosuppressive factors present within the TME (115). For example, cytokines such as TGF-β and IL-10, abundant in the CRC microenvironment, directly inhibit NK cell cytotoxicity, inducing a state of functional exhaustion (116,117). Furthermore, increased expression of inhibitory receptors, such as PD-1 on NK cells within CRC tumors, reduces their capacity to recognize and eliminate tumor cells, thereby allowing tumor cells to evade immune surveillance (118). The modalities underpinning this immune escape are multifaceted. Initially, CRC neoplastic cells elude immunosurveillance mediated by the NK activating receptor NKG2D via the specific downregulation of MHC-I related molecular expression (119). Concomitantly, NK cells exhibit elevated surface expression patterns of novel immune checkpoint receptors beyond PD-1, namely NKG2A/CD94, TIGIT and TIM-3. The cognate ligands for these inhibitory receptors are abundantly expressed by tumor cells or residing within the TME, collaboratively engendering a state of profound NK exhaustion phenotype (120). Moreover, the deleterious metabolic milieu within the TME constitutes another key determinant: Overexpressed CD73 enzymatically catabolizes extracellular ATP into adenosine, which subsequently engages the A2AR receptor on the NK cell surface, thereby abrogating their proliferative capacity and cytotoxic effector functions (121).
Strategies to enhance NK cell antitumor activities include CAR-NK cell therapy, NK cell stimulants, such as IL-15 and combination treatments involving PD-1/PD-L1 blockade (122). Preclinical studies indicate CAR-NK cells exhibit potent antitumor activity and possess superior safety profiles compared with CAR-T cell therapies (123–125). However, due to challenges associated with NK cell proliferation and survival in vitro, further optimization of NK cell-based immunotherapies remains essential to improve their clinical utility in CRC treatment.
DCs, originating from hematopoietic stem cells in the bone marrow, are the most potent professional antigen-presenting cells. DCs capture exogenous antigens, process them into antigenic peptides and present them in complex with MHC molecules on their surface, initiating naïve T cell activation. DCs are a highly heterogeneous group, primarily categorized into conventional DCs (cDCs) and plasmacytoid DCs (pDCs). Functionally, cDCs specialize in antigen processing and presentation, while pDCs are less proficient in antigen presentation but produce substantial amounts of type I interferons (126).
Within the TME, DCs serve as pivotal antigen-presenting cells, capturing, processing and presenting tumor antigens to activate T cell-mediated antitumor immune responses. DCs stimulate CD4+ T cells via MHC-II molecules and also facilitate CD8+ T cell activation through cross-presentation (127). cDCs constitute the predominant DC population within the TME and are further delineated into distinct cDC1 and cDC2 subsets. Notably, the cDC1 subpopulation, whose development is dependent upon the transcription factor BATF3, assumes an indispensable role in orchestrating anti-tumor T cell immune responses (128), whereas cDC2s are principally tasked with initiating Th2-type immunity. However, DC functionality in the CRC microenvironment is frequently compromised, impairing effective antitumor immunity. Immunosuppressive factors, such as prostaglandin E2 and IL-6, inhibit DC maturation, thereby limiting their capacity to activate T cells effectively. This specific functional deficit is particularly salient within the context of CRC. Investigations have elucidated that in MSS CRC phenotypes, aberrant intrinsic activation of the Wnt/β-catenin signaling axis constitutes a pivotal mechanism underpinning immunosuppression (129). Mechanistically, this pathway acts to repress the expression of chemokines CCL4 and CCL5, thereby specifically impeding the efficacious intratumoral infiltration of the key cDC1 subset; this cascade consequently renders the TME an immunologically ‘cold’ landscape, ultimately conferring primary resistance to ICIs (130). Additionally, some DC populations may acquire tolerogenic properties, failing to effectively present antigens and instead promoting Treg expansion, further suppressing antitumor responses (131).
Current strategies aiming to restore DC-mediated antitumor immunity include DC vaccines, autologous DC infusion and combination therapies with ICIs (132,133). DC vaccines hold promise for CRC treatment, with several clinical trials currently evaluating their efficacy in combination with ICIs. Furthermore, prospective therapeutic avenues could involve targeted intervention against the Wnt/β-catenin signaling axis to ameliorate the established impediments to cDC1 recruitment within the TME, thereby orchestrating the conversion of immunologically ‘cold’ tumors into a ‘hot’ phenotype (134–136). This strategy could subsequently be leveraged in synergistic combinations with ICIs to augment overall antitumor therapeutic efficacy.
Neutrophils represent a primary barrier of innate immunity, carrying out key roles in inflammation and immune responses. Within the TME, neutrophils can be further categorized into N1 phenotypes, which exert anti-tumor effects and N2 phenotypes, which facilitate tumor progression. Growth factors, cytokines and chemokines present in the TME recruit neutrophils into the tumor tissue, where they participate in the regulation of diverse and complex biological processes (137). Prevailingly, elevated concentrations of TGF-β function as a cardinal impetus driving the phenotypic skewing of neutrophils from an N1 state towards the pro-tumorigenic N2 phenotype (138). N2-polarized neutrophils predominantly manifest immunosuppressive functionalities, exerting these effects via the regulated secretion of a complex array of chemokines, pro-angiogenic factors and immunosuppressive moieties. Consequently, this bioactive secretome actively recruits and activates collateral immunosuppressive cell populations, facilitates neovascularization and concurrently subverts host immune system recognition and cytotoxic clearance of tumors (139).
Neutrophils carry out a key role in the metastatic process of CRC. Under specific signaling stimuli, neutrophils undergo a unique form of cell death known as neutrophil extracellular trap (NET) formation. The released NETs not only physically capture circulating tumor cells (140), but studies have also revealed that NETs interact via histones on DNA with specific receptors on tumor cell surfaces, activating downstream signaling pathways that promote the colonization and survival of tumor cells in distant organs (141,142). Furthermore, NETs facilitate immune evasion by degrading anti-tumor molecules within the TME (143). Clinical evidence further confirms that an elevated peripheral blood neutrophil-to-lymphocyte ratio serves as an independent predictor of poor prognosis and resistance to immunotherapy in patients with CRC (144).
Therapeutic approaches targeting neutrophils include inhibition of NET formation and blockade of the CXCR2 signaling pathway (145,146), with the objective of diminishing neutrophil recruitment within the TME and augmenting the efficacy of immunotherapeutic interventions. Consequently, the strategic modulation of neutrophil polarization states, the suppression of NET generation and the targeted disruption of their recruitment signals represent viable avenues for enhancing therapeutic outcomes in CRC management.
Multi-omics approaches integrate multiple molecular analyses to elucidate interactions among various biological components. Recently, notable advancements in scRNA-seq and spatial transcriptomics have profoundly enhanced the understanding of the CRC TIME (77,147,148), illuminating immune cell heterogeneity, functional states and interactions with tumor cells, thereby offering novel insights into CRC pathophysiology.
ScRNA-seq provides comprehensive information at the transcriptomic, genomic and epigenomic levels for individual cells, enabling researchers to identify distinct immune cell subsets within the TME, characterize their functional states and reveal potential intercellular communication pathways. Zhang et al (149) conducted a detailed scRNA-seq analysis of tumor-infiltrating myeloid cells in patients with CRC, elucidating the characteristics and lineage trajectories of TAM and DC subpopulations, as well as their interactions with T cells and other cell types. Their study revealed a specific enrichment of secreted phosphoprotein 1+ (SPP1+) TAMs in CRC tissues and demonstrated the similarity between CSF1R inhibitor-resistant TAM subsets and SPP1+ TAM populations, providing a mechanistic explanation for the limited efficacy of anti-CSF1R monotherapy and highlighting the role of SPP1+ macrophages in promoting tumor metastasis (149,150). Importantly, the identification of this SPP1+ TAM subset provides the molecular evidence for the therapeutic resistance mentioned in the TAM section, validating that the compensatory activation limiting CSF1R inhibitor efficacy is driven by distinct, intrinsically resistant macrophage subpopulations. The investigation by Zhang et al (149) had elucidated, at single-cell resolution, a landscape of TAM heterogeneity that was markedly more intricate when compared with the canonical M1/M2 stratification paradigm. This had substantially augmented the comprehension of intratumoral TAM diversity by precisely pinpointing an SPP1+ TAM subset exhibiting inherent resistance to CSF1R inhibitors. This key discovery not only offers a mechanistic rationale for the aforementioned therapeutic predicaments but further delineated that prospective targeting avenues must focus on the precise therapeutic interrogation of such distinct pro-tumorigenic subpopulations. Additionally, Wu et al (151) applied combined scRNA-seq and spatial transcriptomics to CRC liver metastasis samples, systematically illustrating spatial expression patterns of SPP1+ macrophages and mannose receptor C-type 1+, CCL18+ M2-like macrophages, suggesting a key immunosuppressive role of the TAM in CRC liver metastasis (151).
Moreover, Zhang et al (152) performed comprehensive analyses of CRC-associated T cells via single-cell sequencing, thoroughly characterizing their tissue distribution, clonality, migration and dynamic transitions. Through integrated single-cell TCR sequencing, they discovered that both the TME and TCR repertoire influenced the differentiation of tumor-infiltrating CD8+ effector-memory T cells toward Tex and Treg. This high-resolution trajectory analysis refines the concept of the aforementioned T-cell exhaustion, demonstrating that the shift from effector to terminal exhausted states is a dynamic continuum governed by specific TCR repertoires rather than a static classification. This finding had profoundly augmented the comprehension regarding the dynamics of T cell dysfunction, thereby furnishing a theoretical underpinning for their inherent plasticity and potential for reversibility. Further studies identified that Tex or tumor-reactive-like CD8+ T (Ttr-like) cells were associated with the efficacy of anti-PD-1 immunotherapy (78,79). Specifically, a higher abundance of blood-associated Ttr-like cells exhibiting exhaustion phenotypes associated with improved therapeutic outcomes (77). Another notable study integrating spatially enhanced resolution omics sequencing and scRNA-seq identified physical interactions and abundance of lysosomal associated membrane protein 3+ DCs and CXCL13+ T cells, potentially shaping an immune checkpoint blockade (ICB) responsive tumor-stroma boundary. This spatial arrangement and immune status were distinctly characterized dMMR and pMMR CRC and were considerably associated with responses to ICB therapy (153). Collectively, these observations underscore that the therapeutic efficacy of ICB was not predicated merely upon the quantitative abundance of immune infiltrates, but rather hinges upon the spatially orchestrated crosstalk between distinct functional T cell subsets and specific antigen-presenting cells. This spatial organization constituted the structural architectural basis governing ICB outcomes. Such findings proffered a profound mechanistic rationale elucidating why treatment efficacy from immunotherapy remain restricted to a select fraction of the patient population.
The advancement and integrated application of multi-omics technologies have shown considerable potential in dissecting immune cell composition, functional dynamics and tumor-immune cell interactions within the CRC microenvironment. Such comprehensive insights provide key foundations for developing novel therapeutic strategies and advancing personalized medicine.
Notwithstanding the remarkable preclinical strides in TIME-targeted immunotherapies, translating these findings into tangible clinical benefits for patients with CRC is thwarted by multifaceted impediments, principally stemming from the profound heterogeneity of the CRC microenvironment (77). While ICIs demonstrate efficacy in MSI-H subsets, the vast majority of patients with MSS exhibit an ‘immune desert’ or ‘immune exclusion’ phenotype. This intrinsic immunological dormancy renders single-agent TIME-directed strategies insufficient to revert their immune-silent status. Moreover, the dynamic adaptability of the TIME is another key determinant of therapeutic failure. The TME possesses a high degree of compensatory capacity. This mechanism of compensatory immune evasion severely curtails the durable efficacy achievable with monotherapies.
The safety profile and the predictive value of preclinical models also urgently warrant resolution. Several TIME targets, such as CD47 and TGF-β, are ubiquitously expressed in healthy tissues, meaning that off-target systemic toxicity severely constrains the therapeutic window for these agents (154,155). While combinatorial regimens are deemed the principal strategy to circumvent resistance, this approach invariably precipitates compounding toxicities and an escalated risk of immune-related adverse events. Concomitantly, clear clinical guidance remains lacking for the precise determination of the optimal sequencing for distinct therapeutic agents. Furthermore, the widely employed subcutaneous xenograft models exhibit rapid growth dynamics and lack the complex human CRC stromal architecture and the long-term co-evolutionary interplay between the immune system and the tumor (156). The disparity between these models and clinical pathological features often results in a failure to recapitulate efficacy in human trials, despite promising results observed in mice. Therefore, future research should focus on developing more clinically relevant models and exploring combination therapy strategies to overcome the limitations of targeting single pathways.
Accumulating evidence indicates that tumor progression involves not only the intrinsic characteristics of tumor cells but also key interactions within the TIME (157,158). Immune cells within the TIME carry out pivotal roles in CRC initiation, progression and immune evasion processes. Specifically, these immune cells shape the host immune responses, considerably influencing tumor development, thereby positioning the TIME as a promising therapeutic target for cancer treatment. Immunotherapy has evolved from simply enhancing immune effector functions to specifically targeting immunosuppressive factors within the TIME. Consequently, strategies aimed at effectively modulating the TIME to amplify the therapeutic effects of immunotherapy are emerging as a key area of ongoing and future research.
To advance the transition from targeting single cells toward systematically reshaping the TIME through combination therapies, future research should focus on multidisciplinary integration and overcoming challenges such as drug resistance and the limitations of immunotherapy, thereby enabling the development of personalized treatment regimens. Although patients with dMMR/MSI-H CRC benefit from ICIs, the majority of CRC cases are pMMR/MSS and remain unresponsive to ICI-based immunotherapy (10). Therefore, a primary objective is to overcome the immunosuppressive state in pMMR/MSS CRC by enhancing immunogenicity to render these tumors susceptible to immunotherapy. For instance, immunosuppressive M2-type TAMs express inhibitory ligands such as PD-L1 and B7H3, which contribute to immune suppression and diminish the efficacy of T cell-directed ICIs. Combining therapies that target these immunosuppressive TAMs with PD-1/PD-L1 blockade is thus a rational strategy. Clinical evidence supports this approach: The combination of the CSF1R inhibitor emactuzumab with the anti-PD-L1 agent atezolizumab has demonstrated a notable objective response rate in ICI-naïve patients (159). This provides a promising avenue for improving outcomes in pMMR/MSS patients with CRC. Furthermore, breakthroughs in drug development technologies and insights from spatial transcriptomics into TIME-mediated immunosuppression and resistance mechanisms will be instrumental in achieving precision medicine and addressing treatment insensitivity in a subset of patients with CRC. For example, a previous study showed that deep learning can predict MSI status directly from routine histology slides with high concordance with standard molecular testing (160), underscoring the potential of multidisciplinary integration to inform individualized precision therapy.
Comprehensive understanding of the composition and functional dynamics of the TIME, along with its interplay in CRC development and immune escape mechanisms, can provide key insights into the underlying pathogenesis of CRC. Such knowledge offers a robust theoretical framework for developing novel immunotherapeutic approaches. Future research dedicated to unraveling the regulatory mechanisms and interactive networks within the TIME will facilitate individualized, precision-based therapeutic strategies, ultimately improving both survival outcomes and quality of life for patients with CRC.
Not applicable.
This work was supported by grants from the Guangxi Natural Science Foundation (grant no. 2025GXNSFAA069131) and First-class discipline innovation-driven talent program of Guangxi Medical University.
Not applicable.
MQ, HM and ML wrote the manuscript. CL contributed by generating Table I and contributed to manuscript writing. HM and ML revised the manuscript. All authors actively contributed to different aspects of the present study and all authors read and approved the final manuscript. Data authentication not applicable.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: Globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024.PubMed/NCBI | |
|
Sargent D, Sobrero A, Grothey A, O'Connell MJ, Buyse M, Andre T, Zheng Y, Green E, Labianca R, O'Callaghan C, et al: Evidence for cure by adjuvant therapy in colon cancer: Observations based on individual patient data from 20,898 patients on 18 randomized trials. J Clin Oncol. 27:872–877. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Maughan TS, Adams RA, Smith CG, Meade AM, Seymour MT, Wilson RH, Idziaszczyk S, Harris R, Fisher D, Kenny SL, et al: Addition of cetuximab to oxaliplatin-based first-line combination chemotherapy for treatment of advanced colorectal cancer: Results of the randomised phase 3 mrc coin trial. Lancet. 377:2103–2114. 2011. 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 | |
|
Larsen AK, Poindessous V, Ouaret D, El Ouadrani K, Megalophonos VF, Batistella A, Petitprez A, Escargueil AE, Tournigand C and De Gramont A: Feasibility of combining EGFR- and VEGF(R)-targeted agents in colorectal cancer. J Clin Oncol. 29 (4_suppl):S4432011. View Article : Google Scholar | |
|
Chibaudel B, Henriques J, Rakez M, Brenner B, Kim TW, Martinez-Villacampa M, Gallego-Plazas J, Cervantes A, Shim K, Jonker D, et al: Association of bevacizumab plus oxaliplatin-based chemotherapy with disease-free survival and overall survival in patients with stage II colon cancer: A secondary analysis of the avant trial. JAMA Netw Open. 3:e20204252020. View Article : Google Scholar : PubMed/NCBI | |
|
Cutroneo PM, Giardina C, Ientile V, Potenza S, Sottosanti L, Ferrajolo C, Trombetta CJ and Trifirò G: Overview of the safety of anti-VEGF drugs: Analysis of the Italian spontaneous reporting system. Drug Saf. 40:1131–1140. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Passaro A, Jänne PA, Mok T and Peters S: Overcoming therapy resistance in EGFR-mutant lung cancer. Nat Cancer. 2:377–391. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Oxnard GR, Yang JC, Yu H, Kim SW, Saka H, Horn L, Goto K, Ohe Y, Mann H, Thress KS, et al: TATTON: A multi-arm, phase Ib trial of osimertinib combined with selumetinib, savolitinib, or durvalumab in egfr-mutant lung cancer. Ann Oncol. 31:507–516. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Ganesh K, Stadler ZK, Cercek A, Mendelsohn RB, Shia J, Segal NH and Diaz LA Jr: Immunotherapy in colorectal cancer: Rationale, challenges and potential. Nat Rev Gastroenterol Hepatol. 16:361–375. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Tang T, Huang X, Zhang G, Hong Z, Bai X and Liang T: Advantages of targeting the tumor immune microenvironment over blocking immune checkpoint in cancer immunotherapy. Signal Transduct Target Ther. 6:722021. View Article : Google Scholar : PubMed/NCBI | |
|
Coussens LM and Werb Z: Inflammation and cancer. Nature. 420:860–867. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Sedighzadeh SS, Khoshbin AP, Razi S, Keshavarz-Fathi M and Rezaei N: A narrative review of tumor-associated macrophages in lung cancer: Regulation of macrophage polarization and therapeutic implications. Transl Lung Cancer Res. 10:1889–1916. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Mantovani A, Sozzani S, Locati M, Allavena P and Sica A: Macrophage polarization: Tumor-associated macrophages as a paradigm for polarized m2 mononuclear phagocytes. Trends Immunol. 23:549–555. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Müller E, Christopoulos PF, Halder S, Lunde A, Beraki K, Speth M, Øynebråten I and Corthay A: Toll-like receptor ligands and interferon-γ synergize for induction of antitumor m1 macrophages. Front Immunol. 8:13832017. View Article : Google Scholar : PubMed/NCBI | |
|
Jing M, Ma M, Zhang M, Mei Y, Wang L, Jiang Y, Li J, Song R, Yang Z, Pu Y, et al: The photothermal effect induces m1 macrophage-derived tnf-α-type exosomes to inhibit bladder tumor growth. Chem Eng J. 498:1550232024. View Article : Google Scholar | |
|
Aslam M, Ashfaq-Khan M, Qureshi A, Celik S, Werle J-T, Senkowski M, Kim YO, KAPS L and Schuppan D: Fri-453-rapamycin promotes tumoricidal immunity in murine HCC by inducing M1-type macrophages and dentritic cells polarization and enhancement of cytotoxic T-cells. J Hepatol. 70:e5952019. View Article : Google Scholar | |
|
Han Y, Sun J, Yang Y, Liu Y, Lou J, Pan H, Yao J and Han W: TMP195 exerts antitumor effects on colorectal cancer by promoting M1 macrophages polarization. Int J Biol Sci. 18:5653–5666. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Griess B, Mir S, Datta K and Teoh-Fitzgerald M: Scavenging reactive oxygen species selectively inhibits M2 macrophage polarization and their pro-tumorigenic function in part, via Stat3 suppression. Free Radic Biol Med. 147:48–60. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Cassetta L and Pollard JW: Tumor-associated macrophages. Curr Biol. 30:R246–R248. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng Y, Ren S, Zhang Y, Liu S, Meng L, Liu F, Gu L, Ai N and Sang M: Circular RNA circWWC3 augments breast cancer progression through promoting M2 macrophage polarization and tumor immune escape via regulating the expression and secretion of IL-4. Cancer Cell Int. 22:2642022. View Article : Google Scholar : PubMed/NCBI | |
|
Lee YH, Martin-Orozco N, Zheng P, Li J, Zhang P, Tan H, Park HJ, Jeong M, Chang SH, Kim BS, et al: Inhibition of the B7-H3 immune checkpoint limits tumor growth by enhancing cytotoxic lymphocyte function. Cell Res. 27:1034–1045. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Kee JY, Ito A, Hojo S, Hashimoto I, Igarashi Y, Tsuneyama K, Tsukada K, Irimura T, Shibahara N, Takasaki I, et al: CXCL16 suppresses liver metastasis of colorectal cancer by promoting TNF-α-induced apoptosis by tumor-associated macrophages. BMC Cancer. 14:9492014. View Article : Google Scholar : PubMed/NCBI | |
|
Brigati C, Noonan DM, Albini A and Benelli R: Tumors and inflammatory infiltrates: Friends or foes? Clin Exp Metastasis. 19:247–258. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Locati M, Curtale G and Mantovani A: Diversity, mechanisms, and significance of macrophage plasticity. Annu Rev Pathol. 15:123–147. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Inagaki K, Kunisho S, Takigawa H, Yuge R, Oka S, Tanaka S, Shimamoto F, Chayama K and Kitadai Y: Role of tumor-associated macrophages at the invasive front in human colorectal cancer progression. Cancer Sci. 112:2692–2704. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Ma J, Liu L, Che G, Yu N, Dai F and You Z: The M1 form of tumor-associated macrophages in non-small cell lung cancer is positively associated with survival time. BMC Cancer. 10:1122010. View Article : Google Scholar : PubMed/NCBI | |
|
Edin S, Wikberg ML, Dahlin AM, Rutegård J, Öberg Å, Oldenborg PA and Palmqvist R: The distribution of macrophages with a M1 or M2 phenotype in relation to prognosis and the molecular characteristics of colorectal cancer. PLoS One. 7:e470452012. View Article : Google Scholar : PubMed/NCBI | |
|
Wang X, Yang K, Yang B, Wang R, Zhu Y and Pan T: ANKRD22 participates in the proinflammatory activities of macrophages in the colon cancer tumor microenvironment. Cancer Immunol Immunother. 74:862025. View Article : Google Scholar : PubMed/NCBI | |
|
Nussbaum YI, Manjunath Y, Kaifi JT, Warren W and Mitchem J: Analysis of tumor-associated macrophages' heterogeneity in colorectal cancer patients using single-cell RNA-seq data. J Clin Oncol. 40 (4_suppl):S1462025. View Article : Google Scholar | |
|
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 | |
|
Liu Q, Yang C, Wang S, Shi D, Wei C, Song J, Lin X, Dou R, Bai J, Xiang Z, et al: Wnt5a-induced M2 polarization of tumor-associated macrophages via IL-10 promotes colorectal cancer progression. Cell Commun Signal. 18:512020. 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 Z, Xu Y, Shao B, Dang P, Hu S, Sun H, Chen C, Wang C, Liu J, Liu Y and Hu J: Exosomal circPOLQ promotes macrophage M2 polarization via activating Il-10/STAT3 axis in a colorectal cancer model. J Immunother Cancer. 12:e0084912024. View Article : Google Scholar : PubMed/NCBI | |
|
Yang C, Dou R, Wei C, Liu K, Shi D, Zhang C, Liu Q, Wang S and Xiong B: Tumor-derived exosomal microRNA-106b-5p activates EMT-cancer cell and M2-subtype tam interaction to facilitate CRC metastasis. Mol Ther. 29:2088–2107. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Wu Y, Xiao Y, Ding Y, Ran R, Wei K, Tao S, Mao H, Wang J, Pang S, Shi J, et al: Colorectal cancer cell-derived exosomal miRNA-372-5p induces immune escape from colorectal cancer via PTEN/AKT/NF-κB/PD-L1 pathway. Int Immunopharmacol. 143((Pt 1)): 1132612024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu X, Liang R, Lan T, Ding D, Huang S, Shao J, Zheng Z, Chen T, Huang Y, Liu J, et al: Tumor-associated macrophage-specific CD155 contributes to M2-phenotype transition, immunosuppression, and tumor progression in colorectal cancer. J Immunother Cancer. 10:e0042192022. 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 | |
|
Hardbower DM, Coburn LA, Asim M, Singh K, Sierra JC, Barry DP, Gobert AP, Piazuelo MB, Washington MK and Wilson KT: EGFR-mediated macrophage activation promotes colitis-associated tumorigenesis. Oncogene. 36:3807–3819. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Kuang DM, Zhao Q, Peng C, Xu J, Zhang JP, Wu C and Zheng L: Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J Exp Med. 206:1327–1337. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Yu X, Qian J, Ding L, Pan C, Liu X, Wu Q, Wang S, Liu J, Shang M, Su R, et al: Galectin-1-induced tumor associated macrophages repress antitumor immunity in hepatocellular carcinoma through recruitment of tregs. Adv Sci (Weinh). 12:e24087882025. View Article : Google Scholar : PubMed/NCBI | |
|
Mantovani A and Allavena P: The interaction of anticancer therapies with tumor-associated macrophages. J Exp Med. 212:435–445. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Mantovani A, Marchesi F, Malesci A, Laghi L and Allavena P: Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol. 14:399–416. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
De Palma M and Lewis CE: Macrophage regulation of tumor responses to anticancer therapies. Cancer Cell. 23:277–286. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Klug F, Prakash H, Huber PE, Seibel T, Bender N, Halama N, Pfirschke C, Voss RH, Timke C, Umansky L, et al: Low-dose irradiation programs macrophage differentiation to an iNOS+/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell. 24:589–602. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Gordon SR, Maute RL, Dulken BW, Hutter G, George BM, McCracken MN, Gupta R, Tsai JM, Sinha R, Corey D, et al: PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature. 545:495–499. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Byrne KT, Betts CB, Mick R, Sivagnanam S, Bajor DL, Laheru DA, Chiorean EG, O'Hara MH, Liudahl SM, Newcomb C, et al: Neoadjuvant selicrelumab, an agonist CD40 antibody, induces changes in the tumor microenvironment in patients with resectable pancreatic cancer. Clin Cancer Res. 27:4574–4586. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Dowlati A, Harvey RD, Carvajal RD, Hamid O, Klempner SJ, Kauh JSW, Peterson DA, Yu D, Chapman SC, Szpurka AM, et al: LY3022855, an anti-colony stimulating factor-1 receptor (CSF-1R) monoclonal antibody, in patients with advanced solid tumors refractory to standard therapy: Phase 1 dose-escalation trial. Invest New Drugs. 39:1057–1071. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Voissière A, Gomez-Roca C, Chabaud S, Rodriguez C, Nkodia A, Berthet J, Montane L, Bidaux AS, Treilleux I, Eberst L, et al: The CSF-1R inhibitor pexidartinib affects FLT3-dependent DC differentiation and may antagonize durvalumab effect in patients with advanced cancers. Sci Transl Med. 16:eadd18342024. View Article : Google Scholar : PubMed/NCBI | |
|
Brana I, Calles A, LoRusso PM, Yee LK, Puchalski TA, Seetharam S, Zhong B, de Boer CJ, Tabernero J and Calvo E: Carlumab, an anti-C-C chemokine ligand 2 monoclonal antibody, in combination with four chemotherapy regimens for the treatment of patients with solid tumors: An open-label, multicenter phase 1b study. Target Oncol. 10:111–123. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Gomez-Roca CA, Italiano A, Le Tourneau C, Cassier PA, Toulmonde M, D'Angelo SP, Campone M, Weber KL, Loirat D, Cannarile MA, et al: Phase I study of emactuzumab single agent or in combination with paclitaxel in patients with advanced/metastatic solid tumors reveals depletion of immunosuppressive M2-like macrophages. Ann Oncol. 30:1381–1392. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Falchook GS, Peeters M, Rottey S, Dirix LY, Obermannova R, Cohen JE, Perets R, Frommer RS, Bauer TM, Wang JS, et al: A phase 1a/1b trial of CSF-1R inhibitor ly3022855 in combination with durvalumab or tremelimumab in patients with advanced solid tumors. Invest New Drugs. 39:1284–1297. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Nywening TM, Wang-Gillam A, Sanford DE, Belt BA, Panni RZ, Cusworth BM, Toriola AT, Nieman RK, Worley LA, Yano M, et al: Targeting tumour-associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: A single-centre, open-label, dose-finding, non-randomised, phase 1b trial. Lancet Oncol. 17:651–662. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Cannarile MA, Weisser M, Jacob W, Jegg AM, Ries CH and Rüttinger D: Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy. J Immunother Cancer. 5:532017. View Article : Google Scholar : PubMed/NCBI | |
|
Qu T, Li B and Wang Y: Targeting CD47/SIRPα as a therapeutic strategy, where we are and where we are headed. Biomark Res. 10:202022. View Article : Google Scholar : PubMed/NCBI | |
|
Eng C, Lakhani NJ, Philip PA, Schneider C, Johnson B, Kardosh A, Chao MP, Patnaik A, Shihadeh F, Lee Y, et al: A phase 1b/2 study of the anti-CD47 antibody magrolimab with cetuximab in patients with colorectal cancer and other solid tumors. Target Oncol. 20:519–530. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Yamada-Hunter SA, Theruvath J, McIntosh BJ, Freitas KA, Lin F, Radosevich MT, Leruste A, Dhingra S, Martinez-Velez N, Xu P, et al: Engineered CD47 protects T cells for enhanced antitumour immunity. Nature. 630:457–465. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Reiss KA, Angelos MG, Dees EC, Yuan Y, Ueno NT, Pohlmann PR, Johnson ML, Chao J, Shestova O, Serody JS, et al: Car-macrophage therapy for HER2-overexpressing advanced solid tumors: A phase 1 trial. Nat Med. 31:1171–1182. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Tan PB, Verschoor YL, van den Berg JG, Balduzzi S, Kok NFM, Ijsselsteijn ME, Moore K, Jurdi A, Tin A, Kaptein P, et al: Neoadjuvant immunotherapy in mismatch-repair-proficient colon cancers. Nature. 648:726–735. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang F, Chen G, Qiu M, Ma J, Mo X, Liu H, Li Y, Ding P, Wan X, Hu Y, et al: Neoadjuvant treatment of IBI310 plus sintilimab in locally advanced MSI-H/DMMR colon cancer: A randomized phase 1b study. Cancer Cell. 43:1958–1967.e2. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang X, Wang J, Wang G, Zhang Y, Fan Q, Lu C, Hu C, Sun M, Wan Y, Sun S, et al: First-line sugemalimab plus chemotherapy for advanced gastric cancer: The gemstone-303 randomized clinical trial. JAMA. 333:1305–1314. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Ohtani Y, Ross K, Dandekar A, Gabbasov R and Klichinsky M: 128 development of an M1-polarized, non-viral chimeric antigen receptor macrophage (car-m) platform for cancer immunotherapy. Journal for ImmunoTherapy of Cancer. 8 (Suppl 3):doi.org/10.1136/jitc-2020-SITC2020.0128. 2020. | |
|
Razak AR, Cleary JM, Moreno V, Boyer M, Aller EC, Edenfield W, Tie J, Harvey RD, Rutten A, Shah MA, et al: Safety and efficacy of AMG 820, an anti-colony-stimulating factor 1 receptor antibody, in combination with pembrolizumab in adults with advanced solid tumors. J Immunother Cancer. 8:e0010062020. View Article : Google Scholar : PubMed/NCBI | |
|
Spierenburg G, van der Heijden L, van Langevelde K, Szuhai K, Bovée JVGM, van de Sande MAJ and Gelderblom H: Tenosynovial giant cell tumors (TGCT): Molecular biology, drug targets and non-surgical pharmacological approaches. Expert Opin Ther Targets. 26:333–345. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Golstein P and Griffiths GM: An early history of T cell-mediated cytotoxicity. Nat Rev Immunol. 18:527–535. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Pei S, Pollyea DA, Gustafson A, Minhajuddin M, Stevens BM, Ye HY, Inguva A, Amaya ML, Krug A, Jones CL, et al: Developmental plasticity of acute myeloid leukemia mediates resistance to venetoclax-based therapy. Blood. 134 (Suppl_1):S1852019. View Article : Google Scholar | |
|
Li X, Gruosso T, Zuo D, Omeroglu A, Meterissian S, Guiot MC, Salazar A, Park M and Levine H: Infiltration of CD8(+) T cells into tumor cell clusters in triple-negative breast cancer. Proc Natl Acad Sci USA. 116:3678–3687. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Gajewski TF, Schreiber H and Fu YX: Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol. 14:1014–1022. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang L, Shi YC, Yang YX, Wang ZG, Wang SS and Zhang H: Association of T lymphocyte subset counts with the clinical features of colorectal cancer. J Nutr Oncol. 8:178–185. 2023. View Article : Google Scholar | |
|
Doering TA, Crawford A, Angelosanto JM, Paley MA, Ziegler CG and Wherry EJ: Network analysis reveals centrally connected genes and pathways involved in CD8+ T cell exhaustion versus memory. Immunity. 37:1130–1144. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Wherry EJ and Kurachi M: Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 15:486–499. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Angelosanto JM, Blackburn SD, Crawford A and Wherry EJ: Progressive loss of memory T cell potential and commitment to exhaustion during chronic viral infection. J Virol. 86:8161–8170. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Im SJ, Hashimoto M, Gerner MY, Lee J, Kissick HT, Burger MC, Shan Q, Hale JS, Lee J, Nasti TH, et al: Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature. 537:417–421. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Alfei F and Zehn D: T cell exhaustion: An epigenetically imprinted phenotypic and functional makeover. Trends Mol Med. 23:769–771. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Baitsch L, Baumgaertner P, Devêvre E, Raghav SK, Legat A, Barba L, Wieckowski S, Bouzourene H, Deplancke B, Romero P, et al: Exhaustion of tumor-specific CD8+ T cells in metastases from melanoma patients. J Clin Invest. 121:2350–2360. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Odorizzi PM, Pauken KE, Paley MA, Sharpe A and Wherry EJ: Genetic absence of PD-1 promotes accumulation of terminally differentiated exhausted Cd8+ T cells. J Exp Med. 212:1125–1137. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Y, Wang D, Li Y, Qi L, Si W, Bo Y, Chen X, Ye Z, Fan H, Liu B, et al: Spatiotemporal single-cell analysis decodes cellular dynamics underlying different responses to immunotherapy in colorectal cancer. Cancer Cell. 42:1268–1285.e7. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Z, Zhang Y, Ma N, Yang Y, Ma Y, Wang F, Wang Y, Wei J, Chen H, Tartarone A, et al: Progenitor-like exhausted SPRY1(+)CD8(+) t cells potentiate responsiveness to neoadjuvant PD-1 blockade in esophageal squamous cell carcinoma. Cancer Cell. 41:1852–1870.e9. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Z, Yang Z, Wu J, Zhang W, Sun Y, Zhang C, Bai G, Yang L, Fan H, Chen Y, et al: A single-cell atlas reveals immune heterogeneity in anti-PD-1-treated non-small cell lung cancer. Cell. 188:3081–3096.e19. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Kim YJ, Park SJ and Broxmeyer HE: Phagocytosis, a potential mechanism for myeloid-derived suppressor cell regulation of CD8+ T cell function mediated through programmed cell death-1 and programmed cell death-1 ligand interaction. J Immunol. 187:2291–2301. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Tanaka A and Sakaguchi S: Regulatory T cells in cancer immunotherapy. Cell Res. 27:109–118. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Ma S, Dahabieh MS, Mann TH, Zhao S, McDonald B, Song WS, Chung HK, Farsakoglu Y, Garcia-Rivera L, Hoffmann FA, et al: Nutrient-driven histone code determines exhausted CD8(+) T cell fates. Science. 387:eadj30202025. View Article : Google Scholar : PubMed/NCBI | |
|
Li J, Wu C, Hu H, Qin G, Wu X, Bai F, Zhang J, Cai Y, Huang Y, Wang C, et al: Remodeling of the immune and stromal cell compartment by pd-1 blockade in mismatch repair-deficient colorectal cancer. Cancer Cell. 41:1152–1169.e7. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Borràs DM, Verbandt S, Ausserhofer M, Sturm G, Lim J, Verge GA, Vanmeerbeek I, Laureano RS, Govaerts J, Sprooten J, et al: Single cell dynamics of tumor specificity vs bystander activity in CD8(+) T cells define the diverse immune landscapes in colorectal cancer. Cell Discov. 9:1142023. View Article : Google Scholar : PubMed/NCBI | |
|
Donia M, Andersen R, Kjeldsen JW, Fagone P, Munir S, Nicoletti F, Andersen MH, Straten PT and Svane IM: Aberrant expression of MHC class II in melanoma attracts inflammatory tumor-specific CD4+ T- cells, which dampen CD8+ T-cell antitumor reactivity. Cancer Res. 75:3747–3759. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Borst J, Ahrends T, Bąbała N, Melief CJM and Kastenmüller W: CD4(+) T cell help in cancer immunology and immunotherapy. Nat Rev Immunol. 18:635–647. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Ahrends T, Spanjaard A, Pilzecker B, Bąbała N, Bovens A, Xiao Y, Jacobs H and Borst J: CD4(+) T cell help confers a cytotoxic T cell effector program including coinhibitory receptor downregulation and increased tissue invasiveness. Immunity. 47:848–861.e5. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Shamoun L, Skarstedt M, Andersson RE, Wågsäter D and Dimberg J: Association study on IL-4, IL-4Rα and Il-13 genetic polymorphisms in swedish patients with colorectal cancer. Clin Chim Acta. 487:101–106. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Li W, Chen F, Gao H, Xu Z, Zhou Y, Wang S, Lv Z, Zhang Y, Xu Z, Huo J, et al: Cytokine concentration in peripheral blood of patients with colorectal cancer. Front Immunol. 14:11755132023. View Article : Google Scholar : PubMed/NCBI | |
|
Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, Cua DJ and Littman DR: The orphan nuclear receptor rorgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell. 126:1121–1133. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Huber S, Gagliani N, Zenewicz LA, Huber FJ, Bosurgi L, Hu B, Hedl M, Zhang W, O'Connor W Jr, Murphy AJ, et al: IL-22BP is regulated by the inflammasome and modulates tumorigenesis in the intestine. Nature. 491:259–263. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Kryczek I, Wei S, Szeliga W, Vatan L and Zou W: Endogenous IL-17 contributes to reduced tumor growth and metastasis. Blood. 114:357–359. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Bai Y, Li T, Wang Q, You W, Yang H, Xu X, Li Z, Zhang Y, Yan C, Yang L, et al: Shaping immune landscape of colorectal cancer by cholesterol metabolites. EMBO Mol Med. 16:334–360. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
De Simone V, Pallone F, Monteleone G and Stolfi C: Role of TH17 cytokines in the control of colorectal cancer. Oncoimmunology. 2:e266172013. View Article : Google Scholar : PubMed/NCBI | |
|
Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, Evdemon-Hogan M, Conejo-Garcia JR, Zhang L, Burow M, et al: Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 10:942–949. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Sayour EJ, McLendon P, McLendon R, De Leon G, Reynolds R, Kresak J, Sampson JH and Mitchell DA: Increased proportion of FoxP3+ regulatory T cells in tumor infiltrating lymphocytes is associated with tumor recurrence and reduced survival in patients with glioblastoma. Cancer Immunol Immunother. 64:419–427. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Merlo A, Casalini P, Carcangiu ML, Malventano C, Triulzi T, Mènard S, Tagliabue E and Balsari A: Foxp3 expression and overall survival in breast cancer. J Clin Oncol. 27:1746–1752. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Zi R, Zhao X, Liu L, Wang Y, Zhang R, Bian Z, Jiang H, Liu T, Sun Y, Peng H, et al: Metabolic-immune suppression mediated by the SIRT1-CX3CL1 axis induces functional enhancement of regulatory T cells in colorectal carcinoma. Adv Sci (Weinh). 12:e24047342025. View Article : Google Scholar : PubMed/NCBI | |
|
Gu J, Zhou J, Chen Q, Xu X, Gao J, Li X, Shao Q, Zhou B, Zhou H, Wei S, et al: Tumor metabolite lactate promotes tumorigenesis by modulating MOESIN lactylation and enhancing TGF-beta signaling in regulatory T cells. Cell Rep. 39:1109862022. View Article : Google Scholar : PubMed/NCBI | |
|
Adamczyk A, Pastille E, Kehrmann J, Vu VP, Geffers R, Wasmer MH, Kasper S, Schuler M, Lange CM, Muggli B, et al: GPR15 facilitates recruitment of regulatory T cells to promote colorectal cancer. Cancer Res. 81:2970–2982. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Akkaya M, Kwak K and Pierce SK: B cell memory: Building two walls of protection against pathogens. Nat Rev Immunol. 20:229–238. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Choi IK, Wang Z, Ke Q, Hong M, Paul DW Jr, Fernandes SM, Hu Z, Stevens J, Guleria I, Kim HJ, et al: Mechanism of EBV inducing anti-tumour immunity and its therapeutic use. Nature. 590:157–162. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Manfroi B and Fillatreau S: Regulatory B cells gain muscles with a leucine-rich diet. Immunity. 55:970–972. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Murakami Y, Saito H, Shimizu S, Kono Y, Shishido Y, Miyatani K, Matsunaga T, Fukumoto Y, Ashida K, Sakabe T, et al: Increased regulatory B cells are involved in immune evasion in patients with gastric cancer. Sci Rep. 9:130832019. View Article : Google Scholar : PubMed/NCBI | |
|
Griss J, Bauer W, Wagner C, Simon M, Chen M, Grabmeier-Pfistershammer K, Maurer-Granofszky M, Roka F, Penz T, Bock C, et al: B cells sustain inflammation and predict response to immune checkpoint blockade in human melanoma. Nat Commun. 10:41862019. View Article : Google Scholar : PubMed/NCBI | |
|
Mao H, Pan F, Wu Z, Wang Z, Zhou Y, Zhang P, Gou M and Dai G: Colorectal tumors are enriched with regulatory plasmablasts with capacity in suppressing T cell inflammation. Int Immunopharmacol. 49:95–101. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Liao Y, Zhuo X, Huang Y, Xu H, Hao Z, Huang L, Zheng H and Zhou J: A SENP7-SIRT1-IL-10 axis driven by DeSUMOylation promotes breg differentiation and immune evasion in colorectal cancer. Int J Biol Sci. 22:111–125. 2026. View Article : Google Scholar : PubMed/NCBI | |
|
Hou D, Castro B, Dapash M, Rashidi A, Zhang P, Han Y, Lopez-Rosas A, Lesniak M, Miska J and Chang C: Immu-36. B cell-vaccine elicits long term immunity against glioblastoma via activation and differentiation of tumor-specific CD8+ memory T cells. Neuro Oncol. 23 (Suppl_6):vi100. 2021. View Article : Google Scholar | |
|
Irimia RM, Gerke MB, Thakar M, Ren Z, Helmenstine E, Imus PH, Ghiaur G, Leone R and Gocke GB: CD38 is a key regulator of tumor growth by modulating the metabolic signature of malignant plasma cells. Blood. 138 (Suppl 1):S26522021. View Article : Google Scholar | |
|
Malik H, Buelow B, Rangaswamy U, Balasubramani A, Boudreau A, Dang K, Davison L, Aldred SF, Harris KM, Iyer S, Jorgensen B, Pham D, Prabhakar K, Schellenberger U, Ugamraj H, Trinklein N and Van Schooten W: Tnb-486, a novel fully human bispecific CD19 × CD3 antibody that kills CD19-positive tumor cells with minimal cytokine secretion. Blood. 134 (Suppl_1):S4070. 2019. View Article : Google Scholar | |
|
Wang W, Zhong Y, Zhuang Z, Xie J, Lu Y, Huang C, Sun Y, Wu L, Yin J, Yu H, et al: Multiregion single-cell sequencing reveals the transcriptional landscape of the immune microenvironment of colorectal cancer. Clin Transl Med. 11:e2532021. View Article : Google Scholar : PubMed/NCBI | |
|
Nicolai CJ and Raulet DH: Killer cells add fire to fuel immunotherapy. Science. 368:943–944. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Dixon K, Hullsiek R, Snyder K, Davis Z, Khaw M, Lee T, Chu HY, Abujarour R, Dinella J, Rogers P, et al: Engineered iPSC-derived NK cells expressing recombinant CD64 for enhanced ADCC. Blood. 136 (Suppl 1):S10–S11. 2020. View Article : Google Scholar | |
|
Fauriat C, Long EO, Ljunggren HG and Bryceson YT: Regulation of human NK-cell cytokine and chemokine production by target cell recognition. Blood. 115:2167–2176. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Liu YJ, Han M, Li JP, Zeng SH, Ye QW, Yin ZH, Liu SL and Zou X: An analysis regarding the association between connexins and colorectal cancer (CRC) tumor microenvironment. J Inflamm Res. 15:2461–2476. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Yang L, Yi J, He W, Kong P, Xie Q, Jin Y, Xiong Z and Xia L: Death receptors 4/5 mediate tumour sensitivity to natural killer cell-mediated cytotoxicity in mismatch repair deficient colorectal cancer. Br J Cancer. 131:334–346. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Otegbeye F, Ojo E, Moreton S, Mackowski N, Lee DA, de Lima M and Wald DN: Inhibiting TGF-beta signaling preserves the function of highly activated, in vitro expanded natural killer cells in AML and colon cancer models. PLoS One. 13:e01913582018. View Article : Google Scholar : PubMed/NCBI | |
|
Vuletić A, Martinović KM, Miletić NT, Zoidakis J, Castellvi-Bel S and Čavić M: Cross-talk between tumor cells undergoing epithelial to mesenchymal transition and natural killer cells in tumor microenvironment in colorectal cancer. Front Cell Dev Biol. 9:7500222021. View Article : Google Scholar : PubMed/NCBI | |
|
Tang S, Fu H, Xu Q and Zhou Y: Mir-20a regulates sensitivity of colorectal cancer cells to NK cells by targeting mica. Biosci Rep. 39:BSR201806952019. View Article : Google Scholar : PubMed/NCBI | |
|
Lin A, Ye P, Li Z, Jiang A, Liu Z, Cheng Q, Zhang J and Luo P: Natural killer cell immune checkpoints and their therapeutic targeting in cancer treatment. Research (Wash DC). 8:07232025.PubMed/NCBI | |
|
Lupo K and Matosevic S: 123 natural killer cells engineered with an inducible, responsive genetic construct targeting tigit and CD73 to relieve immunosuppression within the gbm microenvironment. Journal for ImmunoTherapy of Cancer. 8 (Suppl 3):doi.org/10.1136/jitc-2020-SITC2020.0123. 2020.PubMed/NCBI | |
|
Strassheimer F, Strecker MI, Alekseeva T, Macas J, Demes MC, Mildenberger IC, Tonn T, Wild PJ, Sevenich L, Reiss Y, et al: Os12.6.A combination therapy of CAR-NK-cells and anti-pd-1 results in high efficacy against advanced-stage glioblastoma in a syngeneic mouse model and induces protective anti-tumor immunity in vivo. Neuro Oncol. 23 (Suppl_2):ii152021. View Article : Google Scholar | |
|
Kong R, Liu B, Wang H, Lu T and Zhou X: CAR-NK cell therapy: Latest updates from the 2024 ASH annual meeting. J Hematol Oncol. 18:222025. View Article : Google Scholar : PubMed/NCBI | |
|
Duan J, Zhao S, Duan Y, Sun D, Zhang G, Yu D, Lou Y, Liu H, Yang S, Liang X, et al: Mnox nanoenzyme armed CAR-NK cells enhance solid tumor immunotherapy by alleviating the immunosuppressive microenvironment. Adv Healthc Mater. 13:e23039632024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang W, Liu Y, He Z, Li L, Liu S, Jiang M, Zhao B, Deng M, Wang W, Mi X, et al: Breakthrough of solid tumor treatment: CAR-NK immunotherapy. Cell Death Discov. 10:402024. View Article : Google Scholar : PubMed/NCBI | |
|
Reizis B: Plasmacytoid dendritic cells: Development, regulation, and function. Immunity. 50:37–50. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wculek SK, Cueto FJ, Mujal AM, Melero I, Krummel MF and Sancho D: Dendritic cells in cancer immunology and immunotherapy. Nat Rev Immunol. 20:7–24. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Jhunjhunwala S, Hammer C and Delamarre L: Antigen presentation in cancer: Insights into tumour immunogenicity and immune evasion. Nat Rev Cancer. 21:298–312. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Klempner SJ, Cecchini M, Khushman M, Kummar S, Pelster M, Rodon J, Sharma S, Yaeger R, Rojo J, Wells AL, et al: Abstract B031: A phase 1/2 trial of fog-001, a first-in-class direct β-catenin:Tcf4 inhibitor, preliminary safety and efficacy in patients with solid tumors bearing wnt pathway-activating mutations (WPAM+). Mol Cancer Ther. 24 (10_Suppl):B0312025. View Article : Google Scholar | |
|
Suryawanshi A, Hussein MS, Prasad PD and Manicassamy S: Wnt signaling cascade in dendritic cells and regulation of anti-tumor immunity. Front Immunol. 11:1222020. View Article : Google Scholar : PubMed/NCBI | |
|
Dhodapkar MV, Dhodapkar KM and Palucka AK: Interactions of tumor cells with dendritic cells: Balancing immunity and tolerance. Cell Death Differ. 15:39–50. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Itai YS, Barboy O, Salomon R, Bercovich A, Xie K, Winter E, Shami T, Porat Z, Erez N, Tanay A, et al: Bispecific dendritic-T cell engager potentiates anti-tumor immunity. Cell. 187:375–389.e18. 2024. View Article : Google Scholar | |
|
Aznar MA, Planelles L, Perez-Olivares M, Molina C, Garasa S, Etxeberría I, Perez G, Rodriguez I, Bolaños E, Lopez-Casas P, et al: Immunotherapeutic effects of intratumoral nanoplexed poly I:C. J Immunother Cancer. 7:1162019. View Article : Google Scholar : PubMed/NCBI | |
|
He F, Wu Z, Liu C, Zhu Y, Zhou Y, Tian E, Rosin-Arbesfeld R, Yang D, Wang MW and Zhu D: Targeting BCL9/BCL9L enhances antigen presentation by promoting conventional type 1 dendritic cell (cDC1) activation and tumor infiltration. Signal Transduct Target Ther. 9:1392024. View Article : Google Scholar : PubMed/NCBI | |
|
Singer M, Valerin J, Zhang Z, Zhang Z, Dayyani F, Yaghmai V, Choi A, Imagawa D and Abi-Jaoudeh N: Promising cellular immunotherapy for colorectal cancer using classical dendritic cells and natural killer T cells. Cells. 14:1662025. View Article : Google Scholar : PubMed/NCBI | |
|
Passardi A, Sullo FG, Bittoni A, Matteucci L, De Rosa F, Bulgarelli J, Tazzari M, Petrini M, Scarpi E, Testoni S, et al: CombiCoR-Vax trial: Study protocol for a phase II, single-arm, multicenter trial of sequential pembrolizumab plus dendritic cell vaccine followed by trifluridine/tipiracil and bevacizumab in refractory microsatellite-stable metastatic colorectal cancer. BMC Cancer. 25:19212025. View Article : Google Scholar : PubMed/NCBI | |
|
Guo FF and Cui JW: The role of neutrophils in cancer development. Journal of Nutritional Oncology. 4:85–90. 2019.https://journals.lww.com/jno/Fulltext/2019/05150/The_Role_of_Neutrophils_in_Cancer_Development.5.aspx View Article : Google Scholar | |
|
Peng H, Shen J, Long X, Zhou X, Zhang J, Xu X, Huang T, Xu H, Sun S, Li C, et al: Local release of TGF-β inhibitor modulates tumor-associated neutrophils and enhances pancreatic cancer response to combined irreversible electroporation and immunotherapy. Adv Sci (Weinh). 9:e21052402022. View Article : Google Scholar : PubMed/NCBI | |
|
Giese MA, Hind LE and Huttenlocher A: Neutrophil plasticity in the tumor microenvironment. Blood. 133:2159–2167. 2019. 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 | |
|
Yang L, Liu Q, Zhang X, Liu X, Zhou B, Chen J, Huang D, Li J, Li H, Chen F, et al: DNA of neutrophil extracellular traps promotes cancer metastasis via CCDC25. Nature. 583:133–138. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Tohme S, Yazdani HO, Al-Khafaji AB, Chidi AP, Loughran P, Mowen K, Wang Y, Simmons RL, Huang H and Tsung A: Neutrophil extracellular traps promote the development and progression of liver metastases after surgical stress. Cancer Res. 76:1367–1380. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Wang X, He S, Gong X, Lei S, Zhang Q, Xiong J and Liu Y: Neutrophils in colorectal cancer: Mechanisms, prognostic value, and therapeutic implications. Front Immunol. 16:15386352025. View Article : Google Scholar : PubMed/NCBI | |
|
Yuan Y, Chen YF, Liu XM, Hu Y, Hao S and Dai XY: Analysis of risk factors and prognostic prediction in advanced colorectal cancer undergoing immunotherapy combined with targeted therapy. Front Med (Lausanne). 12:16404692025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu T, Zou X, Yang C, Li L, Wang B, Li R, Li H, Xu Z, Huang D and Wu Q: Neutrophil extracellular traps promote gastric cancer metastasis by inducing epithelial-mesenchymal transition. Int J Med. 48:1272021. | |
|
Cheng Y, Mo F, Li Q, Han X, Shi H, Chen S, Wei Y and Wei X: Targeting CXCR2 inhibits the progression of lung cancer and promotes therapeutic effect of cisplatin. Mol Cancer. 20:622021. View Article : Google Scholar : PubMed/NCBI | |
|
Chu X, Li X, Zhang Y, Dang G, Miao Y, Xu W, Wang J, Zhang Z and Cheng S: Integrative single-cell analysis of human colorectal cancer reveals patient stratification with distinct immune evasion mechanisms. Nat Cancer. 5:1409–1426. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Oliveira MF, Romero JP, Chung M, Williams SR, Gottscho AD, Gupta A, Pilipauskas SE, Mohabbat S, Raman N, Sukovich DJ, et al: High-definition spatial transcriptomic profiling of immune cell populations in colorectal cancer. Nat Genet. 57:1512–1523. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang L, Li Z, Skrzypczynska KM, Fang Q, Zhang W, O'Brien SA, He Y, Wang L, Zhang Q, Kim A, et al: Single-cell analyses inform mechanisms of myeloid-targeted therapies in colon cancer. Cell. 181:442–459.e29. 2020. 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 | |
|
Zhang L, Yu X, Zheng L, Zhang Y, Li Y, Fang Q, Gao R, Kang B, Zhang Q, Huang JY, et al: Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature. 564:268–272. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Feng Y, Ma W, Zang Y, Guo Y, Li Y, Zhang Y, Dong X, Liu Y, Zhan X, Pan Z, et al: Spatially organized tumor-stroma boundary determines the efficacy of immunotherapy in colorectal cancer patients. Nat Commun. 15:102592024. View Article : Google Scholar : PubMed/NCBI | |
|
Massagué J and Sheppard D: TGF-β signaling in health and disease. Cell. 186:4007–4037. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Sikic BI, Lakhani N, Patnaik A, Shah SA, Chandana SR, Rasco D, Colevas AD, O'Rourke T, Narayanan S, Papadopoulos K, et al: First-in-human, first-in-class phase I trial of the anti-CD47 antibody Hu5f9-G4 in patients with advanced cancers. J Clin Oncol. 37:946–953. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Pyo DH, Hong HK, Lee WY and Cho YB: Patient-derived cancer modeling for precision medicine in colorectal cancer: Beyond the cancer cell line. Cancer Biol Ther. 21:495–502. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Balaban E and Cohen M: Decoding multicellular interaction networks-a new horizon in tumor microenvironment research. Mol Oncol. 19:957–960. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Quail DF and Joyce JA: Microenvironmental regulation of tumor progression and metastasis. Nat Med. 19:1423–1437. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Gomez-Roca C, Cassier P, Zamarin D, Machiels JP, Gracia JL, Hodi FS, Taus A, Garcia M, Boni V, Eder JP, et al: Anti-CSF-1R emactuzumab in combination with anti-PD-L1 atezolizumab in advanced solid tumor patients naïve or experienced for immune checkpoint blockade. J Immunother Cancer. 10:e0040762022. View Article : Google Scholar : PubMed/NCBI | |
|
Wagner SJ, Reisenbüchler D, West NP, Niehues JM, Zhu J, Foersch S, Veldhuizen GP, Quirke P, Grabsch HI, van den Brandt PA, et al: Transformer-based biomarker prediction from colorectal cancer histology: A large-scale multicentric study. Cancer Cell. 41:1650–1661.e4. 2023. View Article : Google Scholar : PubMed/NCBI |