Next Article in Journal
Clear Cell Renal Cell Carcinoma Metastasis to the Thyroid: A Narrative Review of the Literature
Previous Article in Journal
Detection of Human Papillomavirus (HPV) in HPV-Associated Oropharyngeal Squamous Cell Carcinoma: A Review of Diagnostic Approach and Its Importance for the Head and Neck Oncologist
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Expression and Clinical Significance of CD47 in Colorectal Cancer: A Review

1
Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, 56126 Pisa, Italy
2
Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
3
Unit of Pathological Anatomy, University Hospital of Pisa, 56126 Pisa, Italy
*
Author to whom correspondence should be addressed.
Cancers 2026, 18(1), 54; https://doi.org/10.3390/cancers18010054
Submission received: 18 November 2025 / Revised: 19 December 2025 / Accepted: 22 December 2025 / Published: 24 December 2025
(This article belongs to the Section Cancer Immunology and Immunotherapy)

Simple Summary

Colorectal cancer is one of the most common and deadly cancers worldwide. Despite advances in treatment, many patients still experience relapse or poor outcomes, highlighting the need for new immune-based strategies. One promising target is CD47, a molecule on tumor cells that sends a “don’t eat me” signal to immune cells and helps cancer escape immune destruction. However, previous studies have reported inconsistent CD47 expression levels, making its clinical interpretation challenging. This review explores what is currently known about CD47 expression in colorectal cancer, how it interacts with the immune environment, and how it relates to patient prognosis and molecular subtypes. By bringing together findings from different studies, we aim to clarify the potential of CD47 as a biomarker and therapeutic target, providing insights that may support the development of more effective and personalized immunotherapy approaches for colorectal cancer.

Abstract

Cluster of Differentiation 47 (CD47), an innate immune checkpoint, facilitates immune escape by binding signal regulatory protein alpha (SIRPα) to inhibit macrophage phagocytosis. Its significance in colorectal cancer (CRC) has garnered heightened interest. This review summarizes five immunohistochemistry (IHC) studies and complementary transcriptomic analyses assessing CD47 in CRC. IHC results consistently indicated membrane overexpression, though positivity rates varied widely (16–91%) due to methodological heterogeneity. Transcriptomic results confirmed CD47 upregulation, especially in Consensus Molecular Subtype 1 (CMS1) and CMS4 subtypes and revealed co-expression with immune checkpoints and oncogenic pathways. Clinically, high CD47 levels were associated with advanced TNM stage, metastasis, poor differentiation, and altered immune infiltration; however, the prognostic significance varied among cohorts. Overall, CD47 appears to be a promising biomarker and therapeutic target, but clinical translation requires standardized evaluation, including harmonized antibody selection and scoring cut-offs, and prospective validation.

1. Introduction

CRC ranks as the third most prevalent cancer globally and the second foremost cause of cancer-related mortality, with around 1.8 million new cases and 880,000 deaths annually [1,2]. In addition, a colorectal cancer-like subtype of cancer of unknown primary has recently been described and is managed according to CRC treatment paradigms, which may partly contribute to observed CRC incidence trends [3]. CRC is a disease with significant molecular heterogeneity, among which approximately 15% of the cases show microsatellite instability (MSI) caused by deficient DNA mismatch repair (dMMR) [4,5]. However, both biological and methodological heterogeneity in dMMR assessment using immunohistochemistry (IHC) and MSI testing may lead to discordant results, thereby complicating the interpretation and comparison of immune-related findings across studies [6].
The tumor microenvironment (TME) has received increasing attention for its role in CRC progression and treatment response [7,8]. Immune infiltration patterns affect both tumor heterogeneity and response to immunotherapies such as checkpoint inhibitors [9,10,11,12]. Tumors avoid immune surveillance through multiple mechanisms [13,14,15,16]. Among these, tumor-associated macrophages (TAMs) promote immune escape by inducing T cell (Tregs) exhaustion [17,18]. However, reported CD47 expression levels in CRC vary considerably across studies, highlighting the need for a review of the available evidence. CD47, an innate immune checkpoint frequently overexpressed in CRC, serves as a major “don’t eat me” signal, by binding to SIRPα on the surface of macrophages, thereby inhibiting phagocytosis and facilitating tumor immune evasion [19,20,21,22,23].
Although therapeutic strategies targeting CD47 are being gradually explored, especially in metastatic CRC, its potential as a predictive or prognostic marker remains unclear [24,25]. Moreover, published studies report inconsistent CD47 expression patterns, methodologies, and clinical associations, leaving the overall significance of CD47 in CRC insufficiently defined. Therefore, we conducted this review to systematically summarize the expression characteristics of CD47 in CRC and its relationship with clinicopathological characteristics.

2. Mechanism of CD47-Mediated Immune Escape

Tumors achieve immune escape partly through adaptive checkpoints such as Programmed cell death protein 1 (PD-1)/Programmed death-ligand 1 (PD-L1) and Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), which normally restrain T-cell activity but are co-opted by cancers to induce T-cell exhaustion [26,27,28,29,30,31]. In the TME, immunosuppressive populations (Tregs, MDSCs, TAMs) further enhance PD-1/PD-L1 signaling and promote tumor progression [32,33,34,35].
Studies have found that TAMs not only highly express PD-L1 itself, but also enhance the expression of PD-1 in T cells via cell–cell contact or cytokine release, thereby inducing functional exhaustion of CD8+ T cells [36,37,38]. Antigen presentation by TAMs may trigger excessive T-cell activation, which leads to T-cell dysfunction [39,40]. During this process, the Interferon regulatory factor 8 acts as a key regulator [41,42]. Therefore, the activation of immune checkpoint pathways plays a bridging role in immune escape, reflecting not only the immune suppression state of the TME but also revealing the core functions of immune cells such as TAMs in regulating this process [43,44,45].
Beyond adaptive checkpoints, innate immune checkpoints also contribute to tumor immune evasion [46,47]. Among them, the CD47–SIRPα axis has emerged as a central mechanism by which tumor cells suppress macrophage-mediated phagocytosis [48,49]. CD47 is a multi-pass transmembrane protein, with molecular weight between 45 and 55 kDa, composed of an extracellular immunoglobulin superfamily domain, five transmembrane regions, and a short alternatively spliced cytoplasmic tail [50,51,52,53].
As a ligand of inhibitory receptor SIRPα, CD47 is differentially expressed and functions differently between normal and tumor cells, regulates the outcome of myeloid cell-target cells in many homeostasis processes, and plays important roles in cellular functions such as migration, adhesion, angiogenesis, proliferation, and apoptosis [50,54,55,56]. Under physiological conditions, CD47 is broadly expressed and helps maintain self-tolerance, particularly in red blood cells and hematopoietic stem cells [57,58]. In tumor cells, CD47 is frequently overexpressed and binds to the inhibitory receptor SIRPα, primarily expressed on myeloid cells, thus facilitating innate immune escape [59,60]. Moreover, CD47 expression is implicated in epithelial–mesenchymal transition (EMT), tumor stemness, and the shaping of the immunosuppressive microenvironment [61,62,63,64]. Upon CD47 binding, the intracellular immunoreceptor tyrosine-based inhibitory motifs (ITIMs) of SIRPα undergo phosphorylation by Src family kinases [62,64,65], resulting in the recruitment of Src homology region 2 domain-containing phosphatase-1/2 (SHP-1/2), which are normally autoinhibited through Src homology 2 domains [66,67,68]. Binding to phosphorylated ITIMs elicits conformational alterations that activate SHP-1/2 [62,69,70].These phosphatases dephosphorylate downstream targets such as myosin IIA, thereby suppressing cytoskeletal remodeling [53,71]. As a result, macrophage phagocytosis is blocked and tumor cells escape immune clearance [59]. In summary, CD47–SIRPα signaling enables tumor cells to avoid macrophage-mediated clearance, making it a key innate immune checkpoint and an attractive therapeutic target.
Strategies to block the CD47–SIRPα axis include monoclonal antibodies targeting CD47 or SIRPα to disrupt their interaction [72,73,74]; engineered high-affinity SIRPα variants that enhance antibody-dependent phagocytosis [75,76], and gene- or nanotechnology-based approaches to downregulate CD47 expression on tumor cells [25] (Figure 1).
While the majority of research on CD47 blockade utilizes T cell-deficient xenograft models that emphasize macrophage effects, immunocompetent models reveal that CD47 inhibition also promotes CD8+ T cell-mediated antitumor responses [77,78,79]. This results from enhanced antigen processing and presentation by dendritic cells (DCs), facilitating superior T cell priming [65,80,81]. Moreover, CD47 inhibits the function of natural killer (NK) cells [81,82]. Collectively, CD47 suppresses both innate and adaptive immunity to promote tumor immune escape [83,84]. In CRC, this may be particularly relevant in MSI-high tumors, where CD47-mediated suppression of antigen presentation could impair the initiation of adaptive anti-tumor responses.
Mechanistically, upregulation of CD47 in tumors is induced by pro-inflammatory cytokines like TNF-α through the activation of NF-κB signaling, signifying an adaptive response to the TME [66,85,86,87]. These findings identify the CD47-SIRPα axis as a crucial target in tumor immunotherapy [72].

3. Expression of CD47 in CRC

3.1. CD47 Expression in Immunohistochemistry Studies

Three IHC studies have consistently reported that CD47 is markedly elevated in CRC tissues [22,23,88]. Oh et al. evaluated CD47 expression in 468 CRC patient samples, including tumor tissues, normal colonic mucosa, and metastatic/non-metastatic lymph nodes [23]. They found that CD47 was mainly localized on tumor cell membranes (detected in 250 of 468 cases, 53.4%), with low expression in the stroma and normal mucosa. CD47 expression intensity and total IHC scores were considerably elevated in metastatic lymph nodes compared to non-metastatic ones (p = 0.029). Fujiwara-Tani et al. assessed 95 stage II–IV CRC samples and reported a high CD47 positive rate of 86.3% (82/95), markedly higher than in normal tissues [22]. Despite inter-patient heterogeneity, no distinct spatial distribution pattern was identified. Tian et al. analyzed 90 colon adenocarcinoma (COAD) cases and found a CD47 expression rate of 91.11%, mainly localized to the cell membrane and cytoplasm [88].
Nonetheless, two studies have shown divergent results [89,90]. Kim et al. analyzed 328 colorectal adenocarcinoma (CRA) samples and identified a low CD47 positive rate of 16.2% (H-score ≥ 50, calculated as 1 × % of weakly, 2 × % of moderately, and 3 × % of strongly stained tumor cells; total range 0–300), with no positive expression in normal mucosa. The average expression score in CRA was considerably elevated compared to normal mucosa (p = 0.013) [89]. Sugimura-Nagata et al. reported a CD47 positive rate of 35% on the tumor cell membrane in CRC, which was lower than the rates observed in several other studies [90]. In addition to positivity rate-based analyses, two IHC studies categorized CD47 expression using score-based cut-off values rather than binary positivity. Hu et al. classified CRC cases into CD47-high and CD47-low groups based on an IHC score threshold (<4 vs. ≥4) [91], whereas Aktepe et al. defined CD47 positivity using a median staining index [92]. These methodological differences further highlight the heterogeneity in CD47 assessment across IHC studies. Characteristics of included studies are summarized in Table 1.

3.2. CD47 Expression in Transcriptomic Analyses

Recent studies utilizing public databases, including The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), and Gene Expression Profiling Interactive Analysis (GEPIA), have examined CD47 expression profiles and their clinical implications in CRC. These studies consistently demonstrate increased CD47 expression in CRC tissues and its correlation with numerous clinicopathological characteristics, TME, and prognosis.
Tian et al. found that high CD47 expression was substantially correlated with altered M1/M2 macrophage ratios subtypes in CRC, using TCGA-COAD data [88]. Furthermore, co-expression of CD47 with adaptive immune checkpoint molecules such as PD-1 and PD-L1 was significantly elevated, suggesting a pivotal function for CD47 in tumor immune evasion. Oh et al. revealed that CD47 mRNA levels were considerably elevated in CRC tissues compared to matched normal mucosa (p = 0.048) via Reverse Transcription Polymerase Chain Reaction (RT-PCR) [23]. These results suggest CD47 is transcriptionally upregulated.
Consistent elevation of CD47 at both mRNA and protein levels has been documented in various studies, indicating a robust molecular correlation. Importantly, this transcriptional upregulation is consistently observed across independent datasets employing different platforms, reinforcing its reliability beyond technical artifacts. Prior research has associated elevated CD47 expression with EMT, stem-like transcriptional characteristics, and PD-1/PD-L1 resistance [94]. These associations indicate that CD47 may aid in diagnosis, molecular subtyping, and personalized CRC therapy.
Arai et al. performed a large-scale molecular profiling study of 14,287 CRC cases and discovered that CD47-high tumors were markedly enriched for CMS1 and CMS4 subtypes, both linked to immune activation and heightened immune cell infiltration [93]. Elevated CD47 expression correlated with the activation of oncogenic pathways (e.g., Mitogen-activated protein kinase, Phosphoinositide 3-kinase, TGF-β, angiogenesis) and upregulation of multiple immune checkpoints, including PD-1, PD-L1, CTLA-4, and Lymphocyte-activation gene 3 (LAG3). Consistently, Tumor Immune Estimation Resource (TIMER)-based analysis of TCGA-COAD data confirmed increased infiltration of CD8+ T cells, macrophages, and dendritic cells in CD47-high tumors, supporting a potential role for CD47 in modulating an immunosuppressive TME [95].

4. Clinical and Prognostic Correlations of CD47 Expression in CRC

4.1. Correlation Between CD47 and TNM Staging

Tian et al. examined 90 COAD cases and found significantly higher CD47 expression in patients with T3–T4, N1–N2, and Tumor–Node–Metastasis (TNM) stage III–IV tumors (all p < 0.05), supporting a positive correlation with disease advancement [88]. Similarly, Oh et al. reported in 468 CRC samples that elevated CD47 expression was significantly associated with deeper tumor invasion (p = 0.009), lymph node metastasis (p < 0.001), and distant metastasis (p = 0.004) [23]. These findings suggest that CD47 is elevated in advanced TNM stages. In another cohort of 328 CRC patients, Kim et al. used an H-score system and showed an association between CD47 expression and AJCC staging, although specific p values were not provided [89].

4.2. Correlation Between CD47 and Tumor Differentiation

Oh et al. found a significant correlation between CD47 expression and tumor differentiation, with higher expression noted in poorly differentiated tumors (p = 0.020) [23]. Sugimura-Nagata et al. similarly found a higher prevalence of poorly differentiated adenocarcinoma and mucinous adenocarcinoma subtypes in CD47-positive patients (p = 0.032 and p = 0.040, respectively) [90]. Nonetheless, Fujiwara-Tani et al. reported no statistically significant correlation between CD47 expression and tumor differentiation (p = 0.289), indicating that differences in cohort composition and scoring methodologies may affect the observed associations [22]. Other IHC studies primarily focused on prognostic stratification rather than histological grading and did not consistently report independent associations between CD47 expression and tumor differentiation [91,92].

4.3. Correlation Between CD47 and Lymphatic/Distant Metastasis

Oh et al. pointed out that high CD47 expression was notably correlated with both lymph node metastasis (p < 0.001) and distant metastasis (p = 0.004) [23]. Tian et al. also noted increased CD47 expression in N1/N2 stage patients [88]. Although Fujiwara-Tani et al. found a significant correlation between CD47 and metastasis, they did not find a comparable relationship with local T staging (p = 0.472) [22]. Additionally, Kim et al. discovered that CD47 expression was notably associated with multiple metastasis-related markers, such as lymphovascular invasion, perineural invasion, and tumor budding [89]. Similarly, Aktepe et al. observed that CD47-positive tumors exhibited more aggressive clinicopathological features and poorer outcomes, indirectly supporting its association with metastatic potential [92]. Moreover, Sugimura-Nagata et al. reported heightened infiltration of SIRPα and CD163 TAIs in CD47-positive samples, indicating that CD47 positivity is linked to macrophage infiltration and immune evasion [90].

4.4. Prognostic Impact of CD47 Expression on Survival and Recurrence

Oh et al. identified a correlation between elevated CD47 expression and reduced OS in univariate analysis; however, this association lacked significance after controlling for age, sex, tumor size, and TNM staging in multivariate Cox regression [23]. Tian et al. reported a paradoxical finding: while high CD47 expression was associated with advanced TNM stage, it also correlated with longer progression-free survival (PFI) (HR = 0.57, p = 0.025) in their cohort [88]. The authors themselves noted this discrepancy and investigated it, finding a positive correlation between CD47 expression and markers of M1-polarized macrophages (e.g., CD86, CXCL9). They therefore speculated that in this specific context, the prognostic signal of CD47 might be modulated or overridden by a concomitant anti-tumor (M1) immune response, highlighting the microenvironment-dependent nature of its role. Fujiwara-Tani et al. found no significant correlation between CD47 expression and overall survival (p = 0.289) in colorectal cancer patients [22]. In another study, Kim et al. reported that while high CD47 expression was associated with shorter recurrence-free survival (RFS) in univariate analysis (HR = 1.792, 95% CI 1.014–3.084; p = 0.035), it did not retain independent prognostic significance in multivariate analysis when adjusted for AJCC stage and treatment (HR = 1.406, 95% CI 0.815–2.428; p = 0.221). No significant association was found with cancer-specific survival (CSS) [89]. In contrast, Sugimura-Nagata et al. revealed that the 5-year survival was significantly lower in CD47-positive patients (64.0% vs. 79.0%, p = 0.0268) [90]. Multivariate Cox regression identified high CD47 expression as an independent adverse prognostic factor (HR = 1.75, p = 0.038), comparable to lymph node (HR = 2.26) and peritoneal metastasis (HR = 5.80). Consistent with these findings, Hu et al. analyzed a tissue microarray-based CRC cohort and demonstrated that CD47-high expression was significantly associated with both shorter overall survival and disease-free survival, remaining an independent adverse prognostic factor in multivariate Cox regression analyses [91]. Similarly, Aktepe et al. reported that CD47-positive tumors were independently associated with poorer overall survival after adjustment for clinicopathological variables, further supporting the prognostic relevance of CD47 in CRC [92]. In a large cohort of 14,287 CRC cases from the Caris Life Sciences database, Arai et al. reported that high CD47 tumors were enriched in CMS1 and CMS4, both associated with immune activation and unfavorable outcomes [93]. The prognostic findings from published studies are summarized in Table 1.
Collectively, CD47 expression shows consistent prognostic associations in CRC, although its effect may be modulated by the immune microenvironment, molecular subtype (e.g., CMS1/4), and co-expression of other immune checkpoints. Future studies ought to focus on integrative multi-omics methodologies to clarify the independent predictive value of CD47 and assess its potential utility as a personalized prognostic biomarker.

4.5. Correlation Between CD47 Expression and Consensus Molecular Subtypes

The CMS system stratifies CRC into four subtypes with distinct transcriptional and immune features. Among the four established subtypes, CMS1 and CMS4 are closely linked to immune microenvironmental characteristics and exhibit distinct expression levels of CD47 [93].
Arai et al. performed an analysis utilizing 14,287 CRC samples. It was found that the high expression of CD47 mRNA was significantly enriched in the CMS1 (characterized by hypermutation, high microsatellite instability, pronounced immunogenicity, mutations of the BRAF gene) and CMS4 (activation of the TGF-β signaling pathway, epithelial–mesenchymal transition, severe stromal infiltration, active neoangiogenesis, and poor prognosis) [93,96]. These two subtypes belong to the “immune hot” type [97], but they exhibit a paradoxical relationship with immune escape, indicating that CD47 may function as a compensatory “Don’t eat me” signal within the inflammatory microenvironment.
Valenzuela et al. revealed that patients with CMS4 exhibited heightened expression of genes associated with matrix and angiogenesis, diminished response to chemotherapy, and reduced OS [95]. The high expression of CD47 in the CMS4 subtype may enhance its immunosuppressive and treatment resistance. As an innate immune checkpoint, high CD47 expression may contribute to immune exclusion in CMS4 tumors [98]. This subtype is also marked by strong matrix remodeling and angiogenesis-related gene expression [99,100]. Although the early prognosis of CMS1 patients is relatively good, their survival significantly decreases after recurrence, which may be related to the persistent expression of immune checkpoints, including CD47. By delivering a potent “Don’t eat me” signal, CD47 suppresses macrophage phagocytosis and dendritic cell antigen presentation. This innate immune checkpoint activity likely synergizes with adaptive checkpoints (e.g., PD-L1) to create a compounded immunosuppressive barrier, facilitating immune evasion and disease progression upon recurrence. Data from Arai et al. show that CD47-high tumors are enriched in CMS1 and CMS4 subtypes, and key CMS subtype features are summarized according to Valenzuela et al., which are summarized in Table 2.
Overall, the prognosis and therapeutic significance of CD47 may depend on the CMS context. Future research should incorporate CMS classification when analyzing CD47 expression to better predict prognosis and guide immunotherapy, especially for high MSI and interstitial CRC.

4.6. The Relationship Between CD47 and the Tumor Immune Microenvironment

Multiple studies have shown that the high expression of CD47 in CRC is closely related to specific immune cell infiltration patterns and co-expression of immune checkpoints, suggesting that it plays an important role in shaping the TME.
Tian et al. found through TIMER analysis based on TCGA-COAD data that the infiltration of CD8+ T cells, macrophages, and dendritic cells were significantly increased in tumors with high CD47 expression [88]. CD47 expression correlated positively with M1 macrophage markers (CD86, CXCL9, CD80; p < 0.001) but negatively with M2 markers (p < 0.001), suggesting context-dependent effects. Furthermore, Sugimura-Nagata et al. reported that in CD47-positive CRC samples, the numbers of SIRPα and CD163 cells in TAIs significantly increased (p = 0.044; p < 0.0001), which was consistent with the enrichment of immunosuppressive macrophages [90]. The increase in CD163+ cells, a hallmark of M2-like, pro-tumorigenic TAMs, suggests that CD47 may actively shape an immunosuppressive TME by promoting the polarization or recruitment of such cells. These findings indicate that CD47 not only facilitates innate immune evasion via the CD47-SIRPα axis but may also enhance the recruitment of immunosuppressive myeloid cells, thus establishing an immune-tolerant TME. This paradox might reflect dysfunctional or “exhausted” macrophages that express M1 markers but have lost anti-tumor function, possibly due to chronic CD47-SIRPα signaling.

5. Discussion

Although an increasing amount of evidence indicates that CD47 is a promising biomarker and immunotherapy target, there are still some issues that need to be addressed in its clinical translation. More broadly, the clinical implementation of emerging biomarkers in colorectal cancer is often limited by a lack of standardization in molecular testing, tumor heterogeneity, and real-world constraints such as cost and access, which may hinder consistent interpretation and clinical applicability [101].
First, there are significant differences in the positive rate of CD47 recorded in different IHC studies, ranging from 16.2% to 91.1%. This heterogeneity may stem from multiple methodological factors, including: (a) Antibody differences: These five studies used different antibody clones or antibodies from different companies (such as Abcam, CST, different or unspecified), which differed in epitope affinity and signal strength; (b) Scoring system: The definition of high expression varies in different studies. Some studies use an H-score of ≥50, while others use a combination of percentage thresholds, intensity and area scores or do not specify them. For example, a sample with 60% weak (1+) staining would score 60 by H-score (1 × 60), qualifying as positive per Kim et al.’s cutoff (H-score ≥ 50). However, under a scoring system that emphasizes staining intensity, the same weak staining might only score 1 point, potentially classifying it as low or negative (e.g., as implied in intensity-based methods like Tian et al.’s [88]). This directly illustrates how scoring criteria alone can dramatically alter positivity rates. (c) Cut-off value: Arbitrary or research-specific positive thresholds hinder comparisons between studies. (d) Cohort size: The included studies varied considerably in cohort size (ranging from n = 90 to n = 468). Smaller cohorts are inherently more susceptible to random sampling error and have lower statistical power, which may partially explain divergent results in studies of CD47’s correlation with specific clinicopathological features or its prognostic value. These inconsistencies reflect the urgent need to develop a standardized assessment plan for CD47 IHC. Future meta-analyses or cross-platform TCGA comparisons may be helpful in determining reliable evaluation parameters and standardizing cross-cohort expressions. In addition to methodological factors, differences in inclusion criteria and tumor subtypes can also lead to heterogeneity. Some studies only evaluate COAD, while others assess all CRC (colon and rectum), or specifically evaluate CRA. These differences cannot be ignored: Right-sided colon cancers are more frequently characterized by MSI-high and CMS1 subtypes, whereas rectal cancers are predominantly CMS2/3, which can reshape the TME and may indirectly affect the expression of CD47 [102,103]. This is biologically grounded in their distinct embryological origins (midgut vs. hindgut), which establish divergent immune milieus that likely modulate checkpoint expression. In addition, pathological variant types (CRA, mucinous, serrated) exhibit different immune infiltration characteristics, further altering the association between CD47 and prognosis [104,105,106]. In conclusion, these biological and clinical differences have led to variations in the results among studies.
Second, most of the existing studies are retrospective, which essentially limits the reliability and universality of their research results. Retrospective analysis is prone to be influenced by factors such as patient selection, heterogeneity of detection methods, and data deficiency. Therefore, the association between the reported CD47 expression and adverse pathological features (including advanced TNM stage, lymphovascular invasion and tumor budding) should be interpreted with caution [107], as the current evidence only indicates a correlation rather than a causal relationship. It is still uncertain whether the high expression of CD47 really promotes tumor invasiveness or is merely a coincidence. Therefore, the lack of prospective clinical validation and mechanism studies further limits the ability to establish a causal relationship between high CD47 in CRC and adverse clinicopathological outcomes. Future prospective studies using standardized CD47 assessment and predefined clinical endpoints will be essential to clarify causal relationships between CD47 expression and clinical outcomes.
Third, conventional methodologies such as bulk RNA-sequencing and IHC have limitations because they only measure average expression levels without distinguishing between cell types or spatial distribution in the TME [108,109]. This simplification method cannot reflect the complex cellular and spatial heterogeneity specific to CRC [110]. Recent studies have shown that the biological effects of CD47 may vary depending on the immune environment [89,111]. Especially, the balance between M1 and M2 type TAMs may affect tumor progression in the opposite way [88]. Therefore, transcriptome analysis indicated that CD47 was abundantly expressed in the CMS1 (MSI-immune) and CMS4 (mesenchymal) subtypes, both of which are characterized by intense immune infiltration [93]. But paradoxically, they are associated with poor prognosis. These findings suggest that CD47-mediated immune escape may be environmentally dependent and influenced by macrophage polarization and molecular subtypes [112]. Specifically, in tumors dominated by immunosuppressive M2-like TAMs, high CD47 expression would reinforce an existing “Don’t eat me” barrier, aggravating immune evasion and correlating with poor outcomes. Conversely, in tumors with a high infiltration of inflammatory M1-like macrophages, elevated CD47 might represent a compensatory, but ultimately insufficient, response by tumor cells to resist phagocytic pressure. In this context, the prognostic signal of CD47 could be confounded or even reversed by the dominant anti-tumor immune activity, as suggested by Tian et al. [88]. Thus, the net prognostic value of CD47 is not intrinsic but is interpreted through the lens of the local myeloid cell composition. Advanced methods such as single-cell transcriptomics and spatial proteomics are crucial for clarifying these interactions and improving the prognostic and therapeutic value of CD47.
Recent spatial transcriptomic studies have improved the understanding of the colorectal cancer tumor microenvironment by revealing spatially distinct immune and stromal compartments, which cannot be captured by bulk transcriptomic analyses [113].
In addition to its immunoregulatory role, CD47 has been reported to exert tumor-intrinsic, immune-independent functions in colorectal cancer [91]. Hu et al. demonstrated that CD47 overexpression promotes CRC cell proliferation, invasion, and metastasis in vitro and in vivo, even in macrophage-depleted nude mouse models, indicating that these effects are independent of the CD47–SIRPα immune axis [91]. Mechanistically, CD47 was shown to interact with the glycolytic enzyme ENO1, protecting it from FBXW7-mediated ubiquitin degradation [114]. Stabilization of ENO1 was associated with enhanced aerobic glycolysis and ERK signaling, thereby facilitating tumor growth and metastatic potential [91,115]. Collectively, these findings expand the functional landscape of CD47 beyond immune evasion and suggest that CD47 may also contribute to CRC progression through metabolic reprogramming and tumor-intrinsic signaling pathways.
Preclinical studies using experimental models have provided important evidence supporting the therapeutic potential of CD47 targeting in colorectal cancer. In vitro studies in CRC cell lines and in vivo xenograft models have shown that genetic or pharmacological modulation of CD47 can suppress tumor growth and metastatic potential, even under immune cell-depleted conditions, highlighting tumor-intrinsic vulnerabilities [91]. In addition, translational analyses have suggested that CD47-directed strategies may enhance therapeutic efficacy and support further development of CD47-targeting approaches in CRC [92].
Fourth, the rationale for simultaneously blocking the CD47–SIRPα and PD-1/PD-L1 pathways is strongly supported by emerging translational evidence. This dual-targeting strategy has been validated by a CD47/PD-L1 bispecific antibody (6MW3211), which demonstrated potent synergistic anti-tumor efficacy in preclinical models and a favorable safety profile in GLP non-human primate studies, underscoring its clinical translation potential [116]. Furthermore, histological analysis confirmed the co-expression of PD-L1 and CD47 on several human tumor tissues, providing a biomarker rationale for patient selection in clinical trials [117]. In this context, our discovery of SMC18 (a novel small-molecule dual inhibitor of both CD47/SIRPα and PD-1/PD-L1) provides a complementary therapeutic approach with a distinct pharmacokinetic profile to overcome potential cross-resistance associated with single-pathway blockade. The translational potential of co-targeting these pathways is underscored by ongoing clinical efforts, such as a phase I/II trial combining a CD47 antibody with a small-molecule TLR7 agonist and chemotherapy in advanced solid tumors (NCT04588324). This validates the clinical rationale for our dual-targeting strategy. In this context, SMC18—a single small-molecule inhibitor of both CD47/SIRPα and PD-1/PD-L1—represents a direct and integrated approach to overcome potential cross-resistance. Future research should explore the interaction mechanism between the two pathways to investigate potential synergistic or redundant effects, thereby potentially discovering new targets for combined immunotherapy.
Emerging clinical data indicate that CD47-targeted strategies are being actively evaluated in CRC, with early results suggesting heterogeneous activity and notable safety considerations. In an open-label phase 1b/2 study, the anti-CD47 antibody magrolimab combined with cetuximab showed modest efficacy in heavily pretreated CRC, with ORR 6.3% in KRAS wild-type tumors versus 0% in KRAS-mutant tumors (DCR 50.0% vs. 38.1%), and manageable toxicities; grade ≥ 3 anemia occurred in 11.5% of patients and no treatment-related deaths were reported [118]. In contrast, a phase II trial testing evorpacept (ALX148) plus cetuximab and pembrolizumab in refractory MSS mCRC reported an ORR of 6.3% and a DCR of 12.5%, but enrollment was terminated early due to serious immune-related toxicities, including treatment-related grade 5 hemophagocytic lymphohistiocytosis and cytokine release syndrome [119]. Collectively, these findings support the feasibility of CD47-based combinations in CRC while underscoring the need for careful patient selection and safety optimization.
Ultimately, although CD47 is associated with poor prognosis in some cohorts [120], its predictive value for treatment response, especially to immune checkpoint inhibitors, still awaits verification through clinical trials. Combining CD47 expression with multi-dimensional immune features (such as CMS, TAM polarization, and interferon features) may help develop more accurate risk stratification models.

6. Conclusions

CD47 is widely overexpressed in CRC and is associated with adverse pathological features, immune infiltration and prognosis. However, the inconsistencies in detection methods, scoring criteria and cohort characteristics have limited its clinical application. We should further focus on standardizing the unified IHC assessment protocol, including the selection of antibody clones, scoring criteria and positive cut-off values.
Integrating CD47 expression data with single-cell and spatial transcriptomics methods may help us better understand the role of CD47 in immune escape. Furthermore, co-targeting CD47 with adaptive checkpoints such as PD-1 or LAG3 holds promising therapeutic prospects, especially CMS4 CRC. Overcoming issues such as extratumoral toxicity and weak phagocytic activation remains a key challenge in the development of anti-CD47 antibodies.

Author Contributions

Conceptualization, C.U.; methodology, Q.L. and C.U.; data curation, P.V.; visualization, D.T.; formal analysis, G.M. and B.F.; writing—original draft preparation, Q.L.; writing—review and editing, A.M.P. and R.S.; investigation, R.B. and E.M.; supervision, C.U. All authors have read and agreed to the published version of the manuscript.

Funding

This research is funded by PNRR-Missione 4 “Istruzione e ricerca”—Componente 2 “Dalla ricerca all’impresa”—Progetto Ecosistema dell’Innovazione THE—Tuscany Health Ecosystem (ECS—Ecosistema dell’innovazione sulle scienze e le tecnologie della vita in Toscana)—CUP I53C22000780001—Spoke n. 6 and the China Scholarship Council (CSC), grant numbers 202408450049 and 202408340009.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

The authors thank Cristina Niccoli and the technical staff of the Unit of Pathological Anatomy, University Hospital of Pisa, Pisa, Italy, for their valuable assistance in immunohistochemistry and related experimental procedures.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AJCCAmerican Joint Committee on Cancer
BRAFv-Raf murine sarcoma viral oncogene homolog B1
CD47Cluster of Differentiation 47
CIMPCpG island methylator phenotype
CMSConsensus Molecular Subtypes (CMS1–4)
COADColon adenocarcinoma
CRAColorectal adenocarcinoma
CRCColorectal cancer
CSSCancer-specific survival
CTLA-4Cytotoxic T-lymphocyte-associated protein 4
DCsDendritic cells
DFSDisease-free survival
dMMRDeficient DNA mismatch repair
DSSDisease-specific survival
EGFREpidermal growth factor receptor
EMTEpithelial–mesenchymal transition
GEOGene Expression Omnibus
GEPIAGene Expression Profiling Interactive Analysis
IHCImmunohistochemistry
ITIMsImmunoreceptor tyrosine-based inhibitory motifs
KRASKirsten rat sarcoma viral oncogene homolog
LAG3Lymphocyte-activation gene 3
MDSCsMyeloid-derived suppressor cells
MSIMicrosatellite instability
MYCMyelocytomatosis oncogene
NKNatural killer cells
OSOverall survival
PD-1Programmed cell death protein 1
PD-L1Programmed death-ligand 1
PFIProgression-free interval
RFSRecurrence-free survival
RT-PCRReverse Transcription Polymerase Chain Reaction
SHP-1/2Src homology region 2 domain-containing phosphatase-1/2
SIRPαSignal regulatory protein alpha
TAMsTumor-associated macrophages
TCGAThe Cancer Genome Atlas
TGF-βTransforming growth factor-β
TIMERTumor Immune Estimation Resource
TMETumor immune microenvironment
TNMTumor–Node–Metastasis staging system
TregsRegulatory T cells
WNTWingless-related integration site

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef]
  3. Mathew, B.G.; Aliyuda, F.; Taiwo, D.; Adekeye, K.; Agada, G.; Sanchez, E.; Ghose, A.; Rassy, E.; Boussios, S. From Biology to Diagnosis and Treatment: The Ariadne’s Thread in Cancer of Unknown Primary. Int. J. Mol. Sci. 2023, 24, 5588. [Google Scholar] [CrossRef] [PubMed]
  4. Taieb, J.; Svrcek, M.; Cohen, R.; Basile, D.; Tougeron, D.; Phelip, J.M. Deficient mismatch repair/microsatellite unstable colorectal cancer: Diagnosis, prognosis and treatment. Eur. J. Cancer 2022, 175, 136–157. [Google Scholar] [CrossRef]
  5. Vilar, E.; Tabernero, J. Molecular dissection of microsatellite instable colorectal cancer. Cancer Discov. 2013, 3, 502–511. [Google Scholar] [CrossRef]
  6. Adeleke, S.; Haslam, A.; Choy, A.; Diaz-Cano, S.; Galante, J.R.; Mikropoulos, C.; Boussios, S. Microsatellite instability testing in colorectal patients with Lynch syndrome: Lessons learned from a case report and how to avoid such pitfalls. Pers. Med. 2022, 19, 277–286. [Google Scholar] [CrossRef] [PubMed]
  7. Oshi, M.; Asaoka, M.; Tokumaru, Y.; Yan, L.; Matsuyama, R.; Ishikawa, T.; Endo, I.; Takabe, K. CD8 T Cell Score as a Prognostic Biomarker for Triple Negative Breast Cancer. Int. J. Mol. Sci. 2020, 21, 6968. [Google Scholar] [CrossRef]
  8. Binnewies, M.; Roberts, E.W.; Kersten, K.; Chan, V.; Fearon, D.F.; Merad, M.; Coussens, L.M.; Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Hedrick, C.C.; et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 2018, 24, 541–550. [Google Scholar] [CrossRef] [PubMed]
  9. Inaguma, S.; Lasota, J.; Wang, Z.; Felisiak-Golabek, A.; Ikeda, H.; Miettinen, M. Clinicopathologic profile, immunophenotype, and genotype of CD274 (PD-L1)-positive colorectal carcinomas. Mod. Pathol. 2017, 30, 278–285. [Google Scholar] [CrossRef]
  10. Kim, J.H.; Park, H.E.; Cho, N.Y.; Lee, H.S.; Kang, G.H. Characterisation of PD-L1-positive subsets of microsatellite-unstable colorectal cancers. Br. J. Cancer 2016, 115, 490–496. [Google Scholar] [CrossRef]
  11. Lee, L.H.; Cavalcanti, M.S.; Segal, N.H.; Hechtman, J.F.; Weiser, M.R.; Smith, J.J.; Garcia-Aguilar, J.; Sadot, E.; Ntiamoah, P.; Markowitz, A.J.; et al. Patterns and prognostic relevance of PD-1 and PD-L1 expression in colorectal carcinoma. Mod. Pathol. 2016, 29, 1433–1442. [Google Scholar] [CrossRef]
  12. Rosenbaum, M.W.; Bledsoe, J.R.; Morales-Oyarvide, V.; Huynh, T.G.; Mino-Kenudson, M. PD-L1 expression in colorectal cancer is associated with microsatellite instability, BRAF mutation, medullary morphology and cytotoxic tumor-infiltrating lymphocytes. Mod. Pathol. 2016, 29, 1104–1112. [Google Scholar] [CrossRef] [PubMed]
  13. Lei, X.; Lei, Y.; Li, J.K.; Du, W.X.; Li, R.G.; Yang, J.; Li, J.; Li, F.; Tan, H.B. Immune cells within the tumor microenvironment: Biological functions and roles in cancer immunotherapy. Cancer Lett. 2020, 470, 126–133. [Google Scholar] [CrossRef]
  14. Cavallo, F.; De Giovanni, C.; Nanni, P.; Forni, G.; Lollini, P.L. 2011: The immune hallmarks of cancer. Cancer Immunol. Immunother. 2011, 60, 319–326. [Google Scholar] [CrossRef]
  15. McGranahan, N.; Favero, F.; de Bruin, E.C.; Birkbak, N.J.; Szallasi, Z.; Swanton, C. Clonal status of actionable driver events and the timing of mutational processes in cancer evolution. Sci. Transl. Med. 2015, 7, 283ra254. [Google Scholar] [CrossRef] [PubMed]
  16. Wu, Q.; Tsai, H.I.; Zhu, H.; Wang, D. The Entanglement between Mitochondrial DNA and Tumor Metastasis. Cancers 2022, 14, 1862. [Google Scholar] [CrossRef]
  17. Kerr, S.C.; Morgan, M.M.; Gillette, A.A.; Livingston, M.K.; Lugo-Cintron, K.M.; Favreau, P.F.; Florek, L.; Johnson, B.P.; Lang, J.M.; Skala, M.C.; et al. A bioengineered organotypic prostate model for the study of tumor microenvironment-induced immune cell activation. Integr. Biol. 2020, 12, 250–262. [Google Scholar] [CrossRef]
  18. Cortellino, S.; Longo, V.D. Metabolites and Immune Response in Tumor Microenvironments. Cancers 2023, 15, 3898. [Google Scholar] [CrossRef] [PubMed]
  19. Veillette, A.; Chen, J. SIRPα-CD47 Immune Checkpoint Blockade in Anticancer Therapy. Trends Immunol. 2018, 39, 173–184. [Google Scholar] [CrossRef]
  20. Chao, M.P.; Weissman, I.L.; Majeti, R. The CD47-SIRPα pathway in cancer immune evasion and potential therapeutic implications. Curr. Opin. Immunol. 2012, 24, 225–232. [Google Scholar] [CrossRef]
  21. Feng, M.; Jiang, W.; Kim, B.Y.S.; Zhang, C.C.; Fu, Y.X.; Weissman, I.L. Phagocytosis checkpoints as new targets for cancer immunotherapy. Nat. Rev. Cancer 2019, 19, 568–586. [Google Scholar] [CrossRef]
  22. Fujiwara-Tani, R.; Sasaki, T.; Ohmori, H.; Luo, Y.; Goto, K.; Nishiguchi, Y.; Mori, S.; Nakashima, C.; Mori, T.; Miyagawa, Y.; et al. Concurrent Expression of CD47 and CD44 in Colorectal Cancer Promotes Malignancy. Pathobiology 2019, 86, 182–189. [Google Scholar] [CrossRef] [PubMed]
  23. Oh, H.H.; Park, Y.L.; Park, S.Y.; Myung, E.; Im, C.M.; Yu, H.J.; Han, B.; Seo, Y.J.; Kim, K.H.; Myung, D.S.; et al. CD47 mediates the progression of colorectal cancer by inducing tumor cell apoptosis and angiogenesis. Pathol. Res. Pract. 2022, 240, 154220. [Google Scholar] [CrossRef]
  24. Feng, R.; Zhao, H.; Xu, J.; Shen, C. CD47: The next checkpoint target for cancer immunotherapy. Crit. Rev. Oncol. Hematol. 2020, 152, 103014. [Google Scholar] [CrossRef] [PubMed]
  25. Li, Z.; Li, Y.; Gao, J.; Fu, Y.; Hua, P.; Jing, Y.; Cai, M.; Wang, H.; Tong, T. The role of CD47-SIRPα immune checkpoint in tumor immune evasion and innate immunotherapy. Life Sci. 2021, 273, 119150. [Google Scholar] [CrossRef] [PubMed]
  26. Bai, Z.; Zhou, Y.; Ye, Z.; Xiong, J.; Lan, H.; Wang, F. Tumor-Infiltrating Lymphocytes in Colorectal Cancer: The Fundamental Indication and Application on Immunotherapy. Front. Immunol. 2021, 12, 808964. [Google Scholar] [CrossRef]
  27. Zhang, L.; Pan, K.; Huang, S.; Zhang, X.; Zhu, X.; He, Y.; Chen, X.; Tang, Y.; Yuan, L.; Yu, D. Graphdiyne Oxide-Mediated Photodynamic Therapy Boosts Enhancive T-Cell Immune Responses by Increasing Cellular Stiffness. Int. J. Nanomed. 2023, 18, 797–812. [Google Scholar] [CrossRef]
  28. Waibl Polania, J.; Lerner, E.C.; Wilkinson, D.S.; Hoyt-Miggelbrink, A.; Fecci, P.E. Pushing Past the Blockade: Advancements in T Cell-Based Cancer Immunotherapies. Front. Immunol. 2021, 12, 777073. [Google Scholar] [CrossRef]
  29. Huang, K.K.; Han, S.S.; He, L.Y.; Yang, L.L.; Liang, B.Y.; Zhen, Q.Y.; Zhu, Z.B.; Zhang, C.Y.; Li, H.Y.; Lin, Y. Combination therapy (toripalimab and lenvatinib)-associated toxic epidermal necrolysis in a patient with metastatic liver cancer: A case report. World J. Clin. Cases 2022, 10, 3478–3484. [Google Scholar] [CrossRef]
  30. Tseng, J.C.; Chang, Y.C.; Huang, C.M.; Hsu, L.C.; Chuang, T.H. Therapeutic Development Based on the Immunopathogenic Mechanisms of Psoriasis. Pharmaceutics 2021, 13, 1064. [Google Scholar] [CrossRef]
  31. Sas, Z.; Cendrowicz, E.; Weinhäuser, I.; Rygiel, T.P. Tumor Microenvironment of Hepatocellular Carcinoma: Challenges and Opportunities for New Treatment Options. Int. J. Mol. Sci. 2022, 23, 3778. [Google Scholar] [CrossRef]
  32. He, Q.; Gao, H.; Tan, D.; Zhang, H.; Wang, J.Z. mRNA cancer vaccines: Advances, trends and challenges. Acta Pharm. Sin. B 2022, 12, 2969–2989. [Google Scholar] [CrossRef]
  33. Lin, K.X.; Istl, A.C.; Quan, D.; Skaro, A.; Tang, E.; Zheng, X. PD-1 and PD-L1 inhibitors in cold colorectal cancer: Challenges and strategies. Cancer Immunol. Immunother. 2023, 72, 3875–3893. [Google Scholar] [CrossRef]
  34. Imani, S.; Farghadani, R.; Roozitalab, G.; Maghsoudloo, M.; Emadi, M.; Moradi, A.; Abedi, B.; Jabbarzadeh Kaboli, P. Reprogramming the breast tumor immune microenvironment: Cold-to-hot transition for enhanced immunotherapy. J. Exp. Clin. Cancer Res. 2025, 44, 131. [Google Scholar] [CrossRef]
  35. Natsuki, S.; Yoshii, M.; Tanaka, H.; Mori, T.; Deguchi, S.; Miki, Y.; Tamura, T.; Toyokawa, T.; Lee, S.; Maeda, K. Involvement of CX3CR1+ cells appearing in the abdominal cavity in the immunosuppressive environment immediately after gastric cancer surgery. World J. Surg. Oncol. 2024, 22, 74. [Google Scholar] [CrossRef]
  36. Zhu, Z.; Jin, Y.; Zhou, J.; Chen, F.; Chen, M.; Gao, Z.; Hu, L.; Xuan, J.; Li, X.; Song, Z.; et al. PD1/PD-L1 blockade in clear cell renal cell carcinoma: Mechanistic insights, clinical efficacy, and future perspectives. Mol. Cancer 2024, 23, 146. [Google Scholar] [CrossRef] [PubMed]
  37. Lubitz, G.S.; Brody, J.D. Not just neighbours: Positive feedback between tumour-associated macrophages and exhausted T cells. Nat. Rev. Immunol. 2022, 22, 3. [Google Scholar] [CrossRef] [PubMed]
  38. Zha, H.; Xu, Z.; Xu, X.; Lu, X.; Shi, P.; Xiao, Y.; Tsai, H.I.; Su, D.; Cheng, F.; Cheng, X.; et al. PD-1 Cellular Nanovesicles Carrying Gemcitabine to Inhibit the Proliferation of Triple Negative Breast Cancer Cell. Pharmaceutics 2022, 14, 1263. [Google Scholar] [CrossRef] [PubMed]
  39. Tang, S.; Che, X.; Wang, J.; Li, C.; He, X.; Hou, K.; Zhang, X.; Guo, J.; Yang, B.; Li, D.; et al. T-bet+CD8+ T cells govern anti-PD-1 responses in microsatellite-stable gastric cancers. Nat. Commun. 2025, 16, 3905. [Google Scholar] [CrossRef]
  40. Cheng, M.; Zain, J.; Rosen, S.T.; Querfeld, C. Emerging drugs for the treatment of cutaneous T-cell lymphoma. Expert Opin. Emerg. Drugs 2022, 27, 45–54. [Google Scholar] [CrossRef]
  41. Nixon, B.G.; Kuo, F.; Ji, L.; Liu, M.; Capistrano, K.; Do, M.; Franklin, R.A.; Wu, X.; Kansler, E.R.; Srivastava, R.M.; et al. Tumor-associated macrophages expressing the transcription factor IRF8 promote T cell exhaustion in cancer. Immunity 2022, 55, 2044–2058.e2045. [Google Scholar] [CrossRef]
  42. Dalmau Gasull, A.; Glavan, M.; Samawar, S.K.R.; Kapupara, K.; Kelk, J.; Rubio, M.; Fumagalli, S.; Sorokin, L.; Vivien, D.; Prinz, M. The niche matters: Origin, function and fate of CNS-associated macrophages during health and disease. Acta Neuropathol. 2024, 147, 37. [Google Scholar] [CrossRef]
  43. Gao, J.; Tan, W.; Yuan, L.; Wang, H.; Wen, J.; Sun, K.; Chen, X.; Wang, S.; Deng, W. Antitumour mechanisms of traditional Chinese medicine elicited by regulating tumour-associated macrophages in solid tumour microenvironments. Heliyon 2024, 10, e27220. [Google Scholar] [CrossRef] [PubMed]
  44. Gao, H.; Ma, L.; Zou, Q.; Hu, B.; Cai, K.; Sun, Y.; Lu, L.; Ren, D. Unraveling dynamic interactions between tumor-associated macrophages and consensus molecular subtypes in colorectal cancer: An integrative analysis of single-cell and bulk RNA transcriptome. Heliyon 2023, 9, e19224. [Google Scholar] [CrossRef]
  45. Li, J.; Ding, Z.; Liu, J.; Li, G.; Li, Y.; Wang, W.; Nundlall, K.; Deng, Y.; Miao, J.; Hu, M.; et al. Reshaping tumor immune microenvironment and modulating T cell function based on hierarchical nanotherapeutics for synergistically inhibiting osteosarcoma. Mater. Today Bio 2025, 34, 102095. [Google Scholar] [CrossRef]
  46. Li, W.; Xu, M.; Cheng, M.; Wei, J.; Zhu, L.; Deng, Y.; Guo, F.; Bi, F.; Liu, M. Current Advances and Future Directions for Sensitizing Gastric Cancer to Immune Checkpoint Inhibitors. Cancer Med. 2025, 14, e71065. [Google Scholar] [CrossRef] [PubMed]
  47. Zhang, C.; Zhou, Y.; Gao, Y.; Zhu, Z.; Zeng, X.; Liang, W.; Sun, S.; Chen, X.; Wang, H. Radiated glioblastoma cell-derived exosomal circ_0012381 induce M2 polarization of microglia to promote the growth of glioblastoma by CCL2/CCR2 axis. J. Transl. Med. 2022, 20, 388. [Google Scholar] [CrossRef] [PubMed]
  48. Shin, S.; Baek, D.S.; Mellors, J.W.; Dimitrov, D.S.; Li, W. Development of Fully Human Antibodies Targeting SIRPα and PLA2G7 for Cancer Therapy. Antibodies 2025, 14, 21. [Google Scholar] [CrossRef]
  49. Chan, C.; Stip, M.; Nederend, M.; Jansen, M.; Passchier, E.; van den Ham, F.; Wienke, J.; van Tetering, G.; Leusen, J. Enhancing IgA-mediated neutrophil cytotoxicity against neuroblastoma by CD47 blockade. J. Immunother. Cancer 2024, 12, e008478. [Google Scholar] [CrossRef]
  50. Brown, E.J.; Frazier, W.A. Integrin-associated protein (CD47) and its ligands. Trends Cell Biol. 2001, 11, 130–135. [Google Scholar] [CrossRef]
  51. Folkes, A.S.; Feng, M.; Zain, J.M.; Abdulla, F.; Rosen, S.T.; Querfeld, C. Targeting CD47 as a cancer therapeutic strategy: The cutaneous T-cell lymphoma experience. Curr. Opin. Oncol. 2018, 30, 332–337. [Google Scholar] [CrossRef]
  52. Wu, F.; Pang, H.; Li, F.; Hua, M.; Song, C.; Tang, J. Progress in cancer research on the regulator of phagocytosis CD47, which determines the fate of tumor cells (Review). Oncol. Lett. 2024, 27, 256. [Google Scholar] [CrossRef]
  53. Jalil, A.R.; Andrechak, J.C.; Discher, D.E. Macrophage checkpoint blockade: Results from initial clinical trials, binding analyses, and CD47-SIRPα structure-function. Antib. Ther. 2020, 3, 80–94. [Google Scholar] [CrossRef]
  54. Oldenborg, P.A. CD47: A Cell Surface Glycoprotein Which Regulates Multiple Functions of Hematopoietic Cells in Health and Disease. ISRN Hematol. 2013, 2013, 614619. [Google Scholar] [CrossRef] [PubMed]
  55. Xie, Z.; Liao, J.; Chen, J. Targeting macrophages to reprogram the tumor immune microenvironment. Blood Sci. 2024, 6, e00203. [Google Scholar] [CrossRef] [PubMed]
  56. Qin, L.; Li, Y.; Zeng, R.; He, Y.; Chen, X.; Xiao, L.; Zhou, H. A novel anti-CD47 antibody with therapeutic potential for NK/T-cell lymphoma. Hum. Vaccin. Immunother. 2024, 20, 2408088. [Google Scholar] [CrossRef]
  57. Eladl, E.; Tremblay-LeMay, R.; Rastgoo, N.; Musani, R.; Chen, W.; Liu, A.; Chang, H. Role of CD47 in Hematological Malignancies. J. Hematol. Oncol. 2020, 13, 96. [Google Scholar] [CrossRef]
  58. Tal, M.C.; Hansen, P.S.; Ogasawara, H.A.; Feng, Q.; Volk, R.F.; Lee, B.; Casebeer, S.E.; Blacker, G.S.; Shoham, M.; Galloway, S.D.; et al. P66 is a bacterial mimic of CD47 that binds the anti-phagocytic receptor SIRPα and facilitates macrophage evasion by Borrelia burgdorferi. bioRxiv 2024. [Google Scholar] [CrossRef]
  59. Saini, P.; Mirji, G.; Islam, S.M.S.; Simons, L.M.; Bhat, S.A.; Bonfanti, A.P.; Muthumani, K.; Agrawal, P.; Cassel, J.; Tang, H.Y.; et al. Targeting Siglec-10/α3β1 Integrin Interactions Enhances Macrophage-Mediated Phagocytosis of Pancreatic Cancer. bioRxiv 2025. [Google Scholar] [CrossRef] [PubMed]
  60. Matlung, H.L.; Szilagyi, K.; Barclay, N.A.; van den Berg, T.K. The CD47-SIRPα signaling axis as an innate immune checkpoint in cancer. Immunol. Rev. 2017, 276, 145–164. [Google Scholar] [CrossRef]
  61. Chao, M.P.; Jaiswal, S.; Weissman-Tsukamoto, R.; Alizadeh, A.A.; Gentles, A.J.; Volkmer, J.; Weiskopf, K.; Willingham, S.B.; Raveh, T.; Park, C.Y.; et al. Calreticulin is the dominant pro-phagocytic signal on multiple human cancers and is counterbalanced by CD47. Sci. Transl. Med. 2010, 2, 63ra94. [Google Scholar] [CrossRef]
  62. Pai, S.; Bamodu, O.A.; Lin, Y.K.; Lin, C.S.; Chu, P.Y.; Chien, M.H.; Wang, L.S.; Hsiao, M.; Yeh, C.T.; Tsai, J.T. CD47-SIRPα Signaling Induces Epithelial-Mesenchymal Transition and Cancer Stemness and Links to a Poor Prognosis in Patients with Oral Squamous Cell Carcinoma. Cells 2019, 8, 1658. [Google Scholar] [CrossRef]
  63. Zhang, H.; Lu, H.; Xiang, L.; Bullen, J.W.; Zhang, C.; Samanta, D.; Gilkes, D.M.; He, J.; Semenza, G.L. HIF-1 regulates CD47 expression in breast cancer cells to promote evasion of phagocytosis and maintenance of cancer stem cells. Proc. Natl. Acad. Sci. USA 2015, 112, E6215–E6223. [Google Scholar] [CrossRef]
  64. Marcucci, F.; Rumio, C.; Corti, A. Tumor cell-associated immune checkpoint molecules—Drivers of malignancy and stemness. Biochim. Biophys. Acta Rev. Cancer 2017, 1868, 571–583. [Google Scholar] [CrossRef] [PubMed]
  65. Schwartz, A.L.; Nath, P.R.; Allgauer, M.; Lessey-Morillon, E.C.; Sipes, J.M.; Ridnour, L.A.; Morillon Ii, Y.M.; Yu, Z.; Restifo, N.P.; Roberts, D.D. Antisense targeting of CD47 enhances human cytotoxic T-cell activity and increases survival of mice bearing B16 melanoma when combined with anti-CTLA4 and tumor irradiation. Cancer Immunol. Immunother. 2019, 68, 1805–1817. [Google Scholar] [CrossRef]
  66. Betancur, P.A.; Abraham, B.J.; Yiu, Y.Y.; Willingham, S.B.; Khameneh, F.; Zarnegar, M.; Kuo, A.H.; McKenna, K.; Kojima, Y.; Leeper, N.J.; et al. A CD47-associated super-enhancer links pro-inflammatory signalling to CD47 upregulation in breast cancer. Nat. Commun. 2017, 8, 14802. [Google Scholar] [CrossRef] [PubMed]
  67. Satoh, J.; Kawana, N.; Yamamoto, Y. Pathway Analysis of ChIP-Seq-Based NRF1 Target Genes Suggests a Logical Hypothesis of their Involvement in the Pathogenesis of Neurodegenerative Diseases. Gene Regul. Syst. Bio 2013, 7, 139–152. [Google Scholar] [CrossRef] [PubMed]
  68. Liu, F.; Jiang, C.C.; Yan, X.G.; Tseng, H.Y.; Wang, C.Y.; Zhang, Y.Y.; Yari, H.; La, T.; Farrelly, M.; Guo, S.T.; et al. BRAF/MEK inhibitors promote CD47 expression that is reversible by ERK inhibition in melanoma. Oncotarget 2017, 8, 69477–69492. [Google Scholar] [CrossRef]
  69. Gowda, P.; Patrick, S.; Singh, A.; Sheikh, T.; Sen, E. Mutant Isocitrate Dehydrogenase 1 Disrupts PKM2-β-Catenin-BRG1 Transcriptional Network-Driven CD47 Expression. Mol. Cell. Biol. 2018, 38, e00001-18. [Google Scholar] [CrossRef]
  70. Deng, H.; Wang, G.; Zhao, S.; Tao, Y.; Zhang, Z.; Yang, J.; Lei, Y. New hope for tumor immunotherapy: The macrophage-related “do not eat me” signaling pathway. Front. Pharmacol. 2023, 14, 1228962. [Google Scholar] [CrossRef]
  71. Dizman, N.; Buchbinder, E.I. Cancer Therapy Targeting CD47/SIRPα. Cancers 2021, 13, 6229. [Google Scholar] [CrossRef]
  72. Willingham, S.B.; Volkmer, J.P.; Gentles, A.J.; Sahoo, D.; Dalerba, P.; Mitra, S.S.; Wang, J.; Contreras-Trujillo, H.; Martin, R.; Cohen, J.D.; et al. The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc. Natl. Acad. Sci. USA 2012, 109, 6662–6667. [Google Scholar] [CrossRef]
  73. Majeti, R.; Chao, M.P.; Alizadeh, A.A.; Pang, W.W.; Jaiswal, S.; Gibbs, K.D., Jr.; van Rooijen, N.; Weissman, I.L. CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell 2009, 138, 286–299. [Google Scholar] [CrossRef] [PubMed]
  74. Chao, M.P.; Alizadeh, A.A.; Tang, C.; Jan, M.; Weissman-Tsukamoto, R.; Zhao, F.; Park, C.Y.; Weissman, I.L.; Majeti, R. Therapeutic antibody targeting of CD47 eliminates human acute lymphoblastic leukemia. Cancer Res. 2011, 71, 1374–1384. [Google Scholar] [CrossRef]
  75. Ho, C.C.; Guo, N.; Sockolosky, J.T.; Ring, A.M.; Weiskopf, K.; Özkan, E.; Mori, Y.; Weissman, I.L.; Garcia, K.C. “Velcro” engineering of high affinity CD47 ectodomain as signal regulatory protein α (SIRPα) antagonists that enhance antibody-dependent cellular phagocytosis. J. Biol. Chem. 2015, 290, 12650–12663. [Google Scholar] [CrossRef]
  76. Weiskopf, K.; Ring, A.M.; Ho, C.C.; Volkmer, J.P.; Levin, A.M.; Volkmer, A.K.; Ozkan, E.; Fernhoff, N.B.; van de Rijn, M.; Weissman, I.L.; et al. Engineered SIRPα variants as immunotherapeutic adjuvants to anticancer antibodies. Science 2013, 341, 88–91. [Google Scholar] [CrossRef]
  77. Ingram, J.R.; Blomberg, O.S.; Sockolosky, J.T.; Ali, L.; Schmidt, F.I.; Pishesha, N.; Espinosa, C.; Dougan, S.K.; Garcia, K.C.; Ploegh, H.L.; et al. Localized CD47 blockade enhances immunotherapy for murine melanoma. Proc. Natl. Acad. Sci. USA 2017, 114, 10184–10189. [Google Scholar] [CrossRef]
  78. Liu, X.; Pu, Y.; Cron, K.; Deng, L.; Kline, J.; Frazier, W.A.; Xu, H.; Peng, H.; Fu, Y.X.; Xu, M.M. CD47 blockade triggers T cell-mediated destruction of immunogenic tumors. Nat. Med. 2015, 21, 1209–1215. [Google Scholar] [CrossRef] [PubMed]
  79. Zhao, H.; Song, S.; Ma, J.; Yan, Z.; Xie, H.; Feng, Y.; Che, S. CD47 as a promising therapeutic target in oncology. Front. Immunol. 2022, 13, 757480. [Google Scholar] [CrossRef]
  80. Soto-Pantoja, D.R.; Terabe, M.; Ghosh, A.; Ridnour, L.A.; DeGraff, W.G.; Wink, D.A.; Berzofsky, J.A.; Roberts, D.D. CD47 in the tumor microenvironment limits cooperation between antitumor T-cell immunity and radiotherapy. Cancer Res. 2014, 74, 6771–6783. [Google Scholar] [CrossRef] [PubMed]
  81. Bond, N.G.; Fahlberg, M.D.; Yu, S.; Rout, N.; Tran, D.; Fitzpatrick-Schmidt, T.; Sprehe, L.M.; Scheef, E.A.; Mudd, J.C.; Schaub, R.; et al. Immunomodulatory potential of in vivo natural killer T (NKT) activation by NKTT320 in Mauritian-origin cynomolgus macaques. iScience 2022, 25, 103889. [Google Scholar] [CrossRef]
  82. Nath, P.R.; Gangaplara, A.; Pal-Nath, D.; Mandal, A.; Maric, D.; Sipes, J.M.; Cam, M.; Shevach, E.M.; Roberts, D.D. CD47 Expression in Natural Killer Cells Regulates Homeostasis and Modulates Immune Response to Lymphocytic Choriomeningitis Virus. Front. Immunol. 2018, 9, 2985. [Google Scholar] [CrossRef]
  83. Matthews, A.H.; Pratz, K.W.; Carroll, M.P. Targeting Menin and CD47 to Address Unmet Needs in Acute Myeloid Leukemia. Cancers 2022, 14, 5906. [Google Scholar] [CrossRef]
  84. Chen, S.H.; Dominik, P.K.; Stanfield, J.; Ding, S.; Yang, W.; Kurd, N.; Llewellyn, R.; Heyen, J.; Wang, C.; Melton, Z.; et al. Dual checkpoint blockade of CD47 and PD-L1 using an affinity-tuned bispecific antibody maximizes antitumor immunity. J. Immunother. Cancer 2021, 9, e003464. [Google Scholar] [CrossRef] [PubMed]
  85. Reissfelder, C.; Stamova, S.; Gossmann, C.; Braun, M.; Bonertz, A.; Walliczek, U.; Grimm, M.; Rahbari, N.N.; Koch, M.; Saadati, M.; et al. Tumor-specific cytotoxic T lymphocyte activity determines colorectal cancer patient prognosis. J. Clin. Investig. 2015, 125, 739–751. [Google Scholar] [CrossRef] [PubMed]
  86. Broekgaarden, M.; Kos, M.; Jurg, F.A.; van Beek, A.A.; van Gulik, T.M.; Heger, M. Inhibition of NF-κB in Tumor Cells Exacerbates Immune Cell Activation Following Photodynamic Therapy. Int. J. Mol. Sci. 2015, 16, 19960–19977. [Google Scholar] [CrossRef] [PubMed]
  87. Liu, Z.; Lei, M.; Bai, Y. Chronic Stress Mediates Inflammatory Cytokines Alterations and Its Role in Tumorigenesis. J. Inflamm. Res. 2025, 18, 1067–1090. [Google Scholar] [CrossRef]
  88. Tian, Q.S.; Zhang, C.; Bao, Z.J.; Pei, Z. The role of CD47 in immune escape of colon cancer and its correlation with heterogeneity of tumor immune microenvironment. PeerJ 2024, 12, e18579. [Google Scholar] [CrossRef]
  89. Kim, H.; Jee, S.; Kim, Y.; Sim, J.; Bang, S.; Son, H.K.; Park, H.; Myung, J.; Ko, Y.H.; Paik, S.S. Correlation of CD47 Expression with Adverse Clinicopathologic Features and an Unfavorable Prognosis in Colorectal Adenocarcinoma. Diagnostics 2021, 11, 668. [Google Scholar] [CrossRef]
  90. Sugimura-Nagata, A.; Koshino, A.; Inoue, S.; Matsuo-Nagano, A.; Komura, M.; Riku, M.; Ito, H.; Inoko, A.; Murakami, H.; Ebi, M.; et al. Expression and Prognostic Significance of CD47-SIRPA Macrophage Checkpoint Molecules in Colorectal Cancer. Int. J. Mol. Sci. 2021, 22, 2690. [Google Scholar] [CrossRef]
  91. Hu, T.; Liu, H.; Liang, Z.; Wang, F.; Zhou, C.; Zheng, X.; Zhang, Y.; Song, Y.; Hu, J.; He, X.; et al. Tumor-intrinsic CD47 signal regulates glycolysis and promotes colorectal cancer cell growth and metastasis. Theranostics 2020, 10, 4056–4072. [Google Scholar] [CrossRef]
  92. Aktepe, O.H.; Kurtulan, O.; Dama, P.E.; Yildirim, E.C.; Terzi, A.; Kalyoncu, S.A.; Atag, E.; Yavuzsen, T.; Uner, M.; Karaoglu, A.; et al. Prognostic importance of CD47 expression on survival of colorectal cancer. BMC Gastroenterol. 2025, 25, 757. [Google Scholar] [CrossRef]
  93. Arai, H.; Gandhi, N.; Battaglin, F.; Wang, J.; Algaze, S.; Jayachandran, P.; Soni, S.; Zhang, W.; Yang, Y.; Millstein, J.; et al. Role of CD47 gene expression in colorectal cancer: A comprehensive molecular profiling study. J. Immunother. Cancer 2024, 12, e010326. [Google Scholar] [CrossRef]
  94. Hu, Z.; Li, W.; Chen, S.; Chen, D.; Xu, R.; Zheng, D.; Yang, X.; Li, S.; Zhou, X.; Niu, X.; et al. Design of a novel chimeric peptide via dual blockade of CD47/SIRPα and PD-1/PD-L1 for cancer immunotherapy. Sci. China Life Sci. 2023, 66, 2310–2328. [Google Scholar] [CrossRef] [PubMed]
  95. Valenzuela, G.; Canepa, J.; Simonetti, C.; Solo de Zaldívar, L.; Marcelain, K.; González-Montero, J. Consensus molecular subtypes of colorectal cancer in clinical practice: A translational approach. World J. Clin. Oncol. 2021, 12, 1000–1008. [Google Scholar] [CrossRef] [PubMed]
  96. Larionova, I.; Tuguzbaeva, G.; Ponomaryova, A.; Stakheyeva, M.; Cherdyntseva, N.; Pavlov, V.; Choinzonov, E.; Kzhyshkowska, J. Tumor-Associated Macrophages in Human Breast, Colorectal, Lung, Ovarian and Prostate Cancers. Front. Oncol. 2020, 10, 566511. [Google Scholar] [CrossRef]
  97. Zhang, J.; Zhu, H.; Liu, W.; Miao, J.; Mao, Y.; Li, Q. Prognostic and predictive molecular biomarkers in colorectal cancer. Front. Oncol. 2025, 15, 1532924. [Google Scholar] [CrossRef]
  98. Leonard, N.A.; Corry, S.M.; Reidy, E.; Egan, H.; O’Malley, G.; Thompson, K.; McDermott, E.; O’Neill, A.; Zakaria, N.; Egan, L.J.; et al. Tumor-associated mesenchymal stromal cells modulate macrophage phagocytosis in stromal-rich colorectal cancer via PD-1 signaling. iScience 2024, 27, 110701. [Google Scholar] [CrossRef]
  99. Plundrich, D.; Chikhladze, S.; Fichtner-Feigl, S.; Feuerstein, R.; Briquez, P.S. Molecular Mechanisms of Tumor Immunomodulation in the Microenvironment of Colorectal Cancer. Int. J. Mol. Sci. 2022, 23, 2782. [Google Scholar] [CrossRef] [PubMed]
  100. Loomans-Kropp, H.A. The utility of liquid biopsy-based methylation biomarkers for colorectal cancer detection. Front. Oncol. 2024, 14, 1351514. [Google Scholar] [CrossRef]
  101. Kyrochristou, I.; Lianos, G.D.; Kyrochristou, G.D.; Anagnostopoulos, G.; Bali, C.; Boussios, S.; Mitsis, M.; Schizas, D.; Vlachos, K. Agnostic Biomarkers and Molecular Signatures in Colorectal Cancer-Guiding Chemotherapy and Predicting Response. Biomedicines 2025, 13, 2038. [Google Scholar] [CrossRef]
  102. Hasbullah, H.H.; Sulong, S.; Che Jalil, N.A.; Abdul Aziz, A.A.; Musa, N.; Musa, M. KRAS Mutational Profiles among Colorectal Cancer Patients in the East Coast of Peninsular Malaysia. Diagnostics 2023, 13, 822. [Google Scholar] [CrossRef]
  103. Burnett-Hartman, A.N.; Lee, J.K.; Demb, J.; Gupta, S. An Update on the Epidemiology, Molecular Characterization, Diagnosis, and Screening Strategies for Early-Onset Colorectal Cancer. Gastroenterology 2021, 160, 1041–1049. [Google Scholar] [CrossRef]
  104. Marzouk, O.; Schofield, J. Review of histopathological and molecular prognostic features in colorectal cancer. Cancers 2011, 3, 2767–2810. [Google Scholar] [CrossRef]
  105. Li, E.; Qiao, H.; Sun, J.; Ma, Q.; Lin, L.; He, Y.; Li, S.; Mao, X.; Zhang, X.; Liao, B. Cuproptosis-related gene expression is associated with immune infiltration and CD47/CD24 expression in glioblastoma, and a risk score based on these genes can predict the survival and prognosis of patients. Front. Oncol. 2023, 13, 1011476. [Google Scholar] [CrossRef]
  106. Elomaa, H.; Tarkiainen, V.; Äijälä, V.K.; Sirniö, P.; Ahtiainen, M.; Sirkiä, O.; Karjalainen, H.; Kastinen, M.; Tapiainen, V.V.; Rintala, J.; et al. Associations of mucinous differentiation and mucin expression with immune cell infiltration and prognosis in colorectal adenocarcinoma. Br. J. Cancer 2025, 132, 660–669. [Google Scholar] [CrossRef] [PubMed]
  107. Chen, C.; Lu, F.; Huang, H.; Pan, Y. Translating CD47-targeted therapy in gastrointestinal cancers: Insights from preclinical to clinical studies. iScience 2024, 27, 111478. [Google Scholar] [CrossRef]
  108. Zhao, Z.; Mak, T.K.; Shi, Y.; Li, K.; Huo, M.; Zhang, C. Integrative analysis of cancer-associated fibroblast signature in gastric cancer. Heliyon 2023, 9, e19217. [Google Scholar] [CrossRef] [PubMed]
  109. Kang, Y.; Zhang, H.; Guan, J. scINRB: Single-cell gene expression imputation with network regularization and bulk RNA-seq data. Brief. Bioinform. 2024, 25, bbae148. [Google Scholar] [CrossRef] [PubMed]
  110. Pirrotta, S.; Masatti, L.; Bortolato, A.; Corrà, A.; Pedrini, F.; Aere, M.; Esposito, G.; Martini, P.; Risso, D.; Romualdi, C.; et al. Exploring public cancer gene expression signatures across bulk, single-cell and spatial transcriptomics data with signifinder Bioconductor package. NAR Genom. Bioinform. 2024, 6, lqae138. [Google Scholar] [CrossRef]
  111. Parcesepe, P.; Giordano, G.; Laudanna, C.; Febbraro, A.; Pancione, M. Cancer-Associated Immune Resistance and Evasion of Immune Surveillance in Colorectal Cancer. Gastroenterol. Res. Pract. 2016, 2016, 6261721. [Google Scholar] [CrossRef]
  112. Celepli, P.; Karabulut, S.; Bigat, İ.; Celepli, S.; Hücümenoğlu, S. CD47 expression and tumor-associated immune cells in breast cancer and their correlation with molecular subtypes and prognostic factors. Pathol. Res. Pract. 2022, 238, 154107. [Google Scholar] [CrossRef]
  113. Roelands, J.; van der Ploeg, M.; Ijsselsteijn, M.E.; Dang, H.; Boonstra, J.J.; Hardwick, J.C.H.; Hawinkels, L.; Morreau, H.; de Miranda, N. Transcriptomic and immunophenotypic profiling reveals molecular and immunological hallmarks of colorectal cancer tumourigenesis. Gut 2023, 72, 1326–1339. [Google Scholar] [CrossRef]
  114. Zhan, P.; Wang, Y.; Zhao, S.; Liu, C.; Wang, Y.; Wen, M.; Mao, J.H.; Wei, G.; Zhang, P. FBXW7 negatively regulates ENO1 expression and function in colorectal cancer. Lab. Investig. 2015, 95, 995–1004. [Google Scholar] [CrossRef] [PubMed]
  115. Polara, R.; Ganesan, R.; Pitson, S.M.; Robinson, N. Cell autonomous functions of CD47 in regulating cellular plasticity and metabolic plasticity. Cell Death Differ. 2024, 31, 1255–1266. [Google Scholar] [CrossRef] [PubMed]
  116. Wang, R.; Zhang, C.; Cao, Y.; Wang, J.; Jiao, S.; Zhang, J.; Wang, M.; Tang, P.; Ouyang, Z.; Liang, W.; et al. Blockade of dual immune checkpoint inhibitory signals with a CD47/PD-L1 bispecific antibody for cancer treatment. Theranostics 2023, 13, 148–160. [Google Scholar] [CrossRef]
  117. Jin, S.; Wang, H.; Li, Y.; Yang, J.; Li, B.; Shi, P.; Zhang, X.; Zhou, X.; Zhou, X.; Niu, X.; et al. Discovery of a novel small molecule as CD47/SIRPα and PD-1/PD-L1 dual inhibitor for cancer immunotherapy. Cell Commun. Signal. 2024, 22, 173. [Google Scholar] [CrossRef]
  118. Eng, C.; Lakhani, N.J.; Philip, P.A.; Schneider, C.; Johnson, B.; Kardosh, A.; Chao, M.P.; 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. 2025, 20, 519–530. [Google Scholar] [CrossRef]
  119. Lentz, R.W.; Lang, J.; Pitts, T.M.; Blatchford, P.; Hu, J.; Jordan, K.R.; Van Bokhoven, A.; Bagby, S.M.; Dominguez, A.T.A.; Binns, C.A.; et al. Phase II Clinical Trial and Preclinical Evaluation of a Novel CD47 Blockade Combination in Refractory Microsatellite-Stable Metastatic Colorectal Cancer. Cancer Res. Commun. 2025, 5, 2039–2052. [Google Scholar] [CrossRef] [PubMed]
  120. Fan, Y.N.; Zhao, G.; Zhang, Y.; Ye, Q.N.; Sun, Y.Q.; Shen, S.; Liu, Y.; Xu, C.F.; Wang, J. Progress in nanoparticle-based regulation of immune cells. Med. Rev. 2023, 3, 152–179. [Google Scholar] [CrossRef]
Figure 1. CD47 expressed on tumor cells binds SIRPα on macrophages, activating ITIM–SHP-1/2 signaling that blocks cytoskeletal remodeling and phagocytosis, thereby transmitting a “don’t eat me” signal. Created with BioRender.com. https://BioRender.com/joi5t5i.
Figure 1. CD47 expressed on tumor cells binds SIRPα on macrophages, activating ITIM–SHP-1/2 signaling that blocks cytoskeletal remodeling and phagocytosis, thereby transmitting a “don’t eat me” signal. Created with BioRender.com. https://BioRender.com/joi5t5i.
Cancers 18 00054 g001
Table 1. Summary of studies evaluating the prognostic significance of CD47 in colorectal cancer.
Table 1. Summary of studies evaluating the prognostic significance of CD47 in colorectal cancer.
StudyCohortMethodSample SizeCD47 AssessmentStaining LocalizationScoring MethodAntibody UsedMain Finding (Standardized)
Oh et al. [23]CRC (South Korea)IHC46853.4% (250/468)Cell membraneIntensity (0–3) × Density (1–4); Mean IHC score ≥ 5 = positiveClone not specified, Abcam, Cambridge, UK;OS ↓ (poorer OS in univariate; not significant in multivariate)
Fujiwara-Tani et al. [22]CRC (Japan)IHC9586.3% (82/95)Cell membraneStaining index = intensity (0–3) × area (%); intensity 1 = normal tissue baselineClone not specified, Abcam, Cambridge, UK;OS –
Tian et al. [88]COAD (China)IHC9091.11% (80/90)Cell membrane and cytoplasmA scale of 0 to 3: 0 points for no staining, 1 point for 1–25% positive staining, 2 points for 26–50%, and 3 points for 51–100%. High CD47 expression was defined by an IHC score of 3 or higher.1:200, CST, 63000SOS –; DSS –; PFI ↑
Kim et al. [89]CRA (South Korea)IHC32816.2% (53/328)MembraneH-score = 1 × (% of 1 + cells) + 2 × (% of 2 + cells) + 3 × (% of 3 + cells). Positive expression groups (H-score ≥ 50)Abcam, Cambridge, UK, EPR21794 (1:200)CSS ↓; RFS ↓ (high CD47 expression associated with poorer CSS and shorter RFS)
Sugimura-Nagata et al. [90]CRC (Japan)IHC26935% (95/269)CytomembraneNot specifiedSP279, Abcam (Cambridge, UK)5-year survival ↓ in CD47-positive tumors; high CD47 was an independent risk factor
Arai et al. [93]CRC (USA)Whole-transcriptome RNA sequencing analysis14,287NANANANAOS—(CD47 mRNA not correlated with OS; enriched in CMS1/4)
Hu et al. [91]CRC (China)IHC (TMA)293High vs. Low (IHC score cut-off: <4 vs. ≥4)NAIHC score (cut-off: <4 vs. ≥4)NAOS ↓; DFS ↓ (CD47-high associated with poorer OS and DFS; independent prognostic factor in multivariate analysis)
Aktepe et al. [92]CRC (Turkey)IHC (TMA)9856.1% (55/98)Membranous stainingArea score (1–4 by % cells: 1% = 1–25%, 2 = 26–50%, 3 = 51–75%, 4 > 75%) × Intensity (0–3+); staining index = intensity × area; median IHC score = 4; negative < median, positive ≥ medianAnti-CD47, Abcam (Cambridge, UK)OS ↓ (CD47-positive had shorter OS; independent prognostic factor in multivariate Cox: HR 2.142, p = 0.006)
Abbreviations: CD47, cluster of differentiation 47; CRC, colorectal cancer; COAD, colon adenocarcinoma; CRA, colorectal adenocarcinoma; IHC, immunohistochemistry; TMA, tissue microarray; OS, overall survival; DSS, disease-specific survival; PFI, progression-free interval; CSS, cancer-specific survival; RFS, recurrence-free survival; CMS, consensus molecular subtype; NA, not applicable. Legend: ↓ indicates worse outcome for CD47-high or CD47-positive tumors; ↑ indicates better outcome; – indicates no significant association.
Table 2. Correlation between CD47 expression and CMS in CRC.
Table 2. Correlation between CD47 expression and CMS in CRC.
CMS SubtypePredominanceCD47 Association Key Features
CMS1 (Immune)Proximal colonEnriched in CD47-high tumors (17.9% vs. 14.5%)MSI-high, high CIMP/BRAF mutations, low prevalence of SNCA, immune infiltration
CMS2 (Canonical)Distal colon and rectumReduced in CD47-high tumors (31.1% vs. 35.4%)Epithelial phenotype, WNT/MYC activation, chromosomal instability
CMS3 (Metabolic)Without predominanceReduced in CD47-high tumors (10.9% vs. 23.3%)KRAS mutations, metabolic reprogramming, intermediate MSI/CIMP
CMS4 (Mesenchymal)Distal colon and rectumEnriched in CD47-high tumors (40.1% vs. 26.8%)Stromal infiltration, EMT/TGF-β activation, angiogenesis, poor prognosis
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Li, Q.; Vignali, P.; Tang, D.; Martinelli, G.; Fuochi, B.; Sparavelli, R.; Poma, A.M.; Bruno, R.; Macerola, E.; Ugolini, C. Expression and Clinical Significance of CD47 in Colorectal Cancer: A Review. Cancers 2026, 18, 54. https://doi.org/10.3390/cancers18010054

AMA Style

Li Q, Vignali P, Tang D, Martinelli G, Fuochi B, Sparavelli R, Poma AM, Bruno R, Macerola E, Ugolini C. Expression and Clinical Significance of CD47 in Colorectal Cancer: A Review. Cancers. 2026; 18(1):54. https://doi.org/10.3390/cancers18010054

Chicago/Turabian Style

Li, Qijie, Paola Vignali, Donghao Tang, Giulia Martinelli, Beatrice Fuochi, Rebecca Sparavelli, Anello Marcello Poma, Rossella Bruno, Elisabetta Macerola, and Clara Ugolini. 2026. "Expression and Clinical Significance of CD47 in Colorectal Cancer: A Review" Cancers 18, no. 1: 54. https://doi.org/10.3390/cancers18010054

APA Style

Li, Q., Vignali, P., Tang, D., Martinelli, G., Fuochi, B., Sparavelli, R., Poma, A. M., Bruno, R., Macerola, E., & Ugolini, C. (2026). Expression and Clinical Significance of CD47 in Colorectal Cancer: A Review. Cancers, 18(1), 54. https://doi.org/10.3390/cancers18010054

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

Article Metrics

Back to TopTop