BPG is committed to discovery and dissemination of knowledge
Minireviews Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Sep 15, 2025; 17(9): 109824
Published online Sep 15, 2025. doi: 10.4251/wjgo.v17.i9.109824
Immune checkpoint molecules signal regulatory protein alpha in the development of hepatocellular carcinoma
Xue Zhang, Dong-Bo Chen, Shao-Ping She, Yao Yang, Li-Ying Ren, Hong-Song Chen, Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Peking University People’s Hospital, Beijing 100044, China
Rui Zhang, Department of Anesthesiology, Peking University Third Hospital, Beijing 100191, China
Pu Chen, Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu Province, China
Hong-Song Chen, Peking University Third Hospital, Beijing 100191, China
ORCID number: Hong-Song Chen (0000-0001-6858-8398).
Co-first authors: Xue Zhang and Dong-Bo Chen.
Author contributions: Zhang X and Chen DB completed the literature review and article writing; Zhang R and Chen P completed the chart drawing; Chen HS designed the framework and content of the article; She SP, Yang Y and Ren LY participated in the revision of the article.
Supported by the National Key Sci-Tech Special Project of China, No. 2018ZX10302207; the Beijing Natural Science Foundation, No. 7222191; the Beijing Natural Science Foundation, No. 7244426; the Fundamental Research Funds for the Central Universities, Peking University, No. PKU2024XGK005; the Peking University Medicine Seed Fund for Interdisciplinary Research, No. BMU2021MX007 and No. BMU2022MX001; and Fundamental Research Funds for the Central Universities, Peking University People’s Hospital Scientific Research Development Funds, No. RDY2020-06 and No. RDJ2022-14.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Hong-Song Chen, Professor, Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Peking University People’s Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, China. chenhongsong2999@163.com
Received: May 26, 2025
Revised: June 29, 2025
Accepted: July 30, 2025
Published online: September 15, 2025
Processing time: 115 Days and 14.6 Hours

Abstract

Hepatocellular carcinoma (HCC) is a primary malignant tumor of the liver and one of the most common malignant tumors, as well as the third leading cause of cancer-related death. In recent years, immune checkpoint inhibitors have emerged as a key strategy in cancer treatment. However, anti-programmed cell death 1/programmed death ligand 1 therapies, one of the main immunotherapeutic approaches, only elicit a response in only approximately 20% of advanced HCC. This suggests that there may be other immune checkpoints playing important roles in HCC immunotherapy. Recent studies have highlighted Signal regulatory protein alpha (SIRPα) is a phagocytic checkpoint in macrophages and other immune cells, as a promising novel therapeutic target in tumor immunotherapy. This review summarizes current progress on SIRPα in HCC and identifies key challenges for future related research.

Key Words: Signal regulatory protein alpha; Hepatocellular carcinoma; Immunotherapy; Immune checkpoint molecules; Immune checkpoint inhibitors

Core Tip: Hepatocellular carcinoma (HCC) is a primary malignant tumor of the liver and one of the most common malignant tumors, as well as the third leading cause of cancer-related death. In recent years, immune checkpoint inhibitors have emerged as a key strategy in cancer treatment. However, anti-programmed cell death 1/programmed death ligand 1 therapies, one of the main immunotherapeutic approaches, only elicit a response in only approximately 20% of advanced HCC. This suggests that there may be other immune checkpoints playing important roles in HCC immunotherapy. Recent studies have highlighted Signal regulatory protein alpha (SIRPα) is a phagocytic checkpoint in macrophages and other immune cells, as a promising novel therapeutic target in tumor immunotherapy. This review summarizes current progress on SIRPα in HCC and identifies key challenges for future related research.



INTRODUCTION

Primary liver cancer is one of the most prevalent malignancies worldwide and ranks third in cancer-related mortality. According to global cancer statistics in 2022, liver cancer ranked sixth-highest incidence[1], with hepatocellular carcinoma (HCC) accounting for approximately 90% of cases[2-5]. In early-stage HCC, radical surgical resection remains the primary treatment option. However, HCC often develops asymptomatically, and most patients (approximately 80%) are diagnosed at intermediate or advanced stages, rendering them ineligible for curative surgery[6]. Moreover, HCC exhibits a high postoperative recurrence rate of 70%, presenting substantial challenges to achieving long-term patient survival[7].

With the completion of the Human Genome Project and advances in genomic technologies, it has become evident that the key driver genes play a crucial role in tumorigenesis. The study of these genes is of strategic importance for understanding tumor origins, predicting prognosis, and developing targeted therapies and immunotherapy strategies. Among these, cancer immunotherapies, particularly immune checkpoint inhibitors, have become an essential treatment option for various cancers[8]. Clinical trials have confirmed that blocking immunosuppressive receptor pathways is a key strategy for HCC immunotherapy, yielding significant therapeutic benefits[9]. The 2020 IMbrave150 clinical trial demonstrated that in previously untreated, unresectable patients, the combination of atezolizumab [anti-programmed death ligand 1 (PD-L1)] and bevacizumab (anti-VEGF) significantly extended overall survival and progression-free survival by 6.8 months, and improved the response rate and duration of response vs standard therapy[10]. Moreover, PD-L1-high advanced HCC patients exhibit a higher objective response rate to programmed cell death 1 (PD-1)/PD-L1 inhibitors compared to controls[11]. However, only approximately 20% of patients with advanced HCC respond to anti-PD-1/PD-L1 therapy, suggesting the involvement of alternative immune checkpoints[12,13].

Signal regulatory protein alpha (SIRPα) is one of the most extensively studied immune checkpoints and a key mediator of tumor-immune crosstalk. This review synthesizes recent advances on SIRPα in HCC, delineating its mechanistic roles in tumor initiation and progression, and discusses its potential clinical applications.

THE PROTEIN STRUCTURE OF SIRPΑ

Signal regulatory protein SIRPα (also known as CD172a, PTPNS1, SHPS1, CD172A and P84), as a member of the SIRP family (SIRPα, SIRPβ1, SIRPβ2, SIRPγ and SIRPδ), is a typical inhibitory immune receptor on myeloid cell membranes (including monocytes, macrophages, neutrophils, a subset of dendritic cells and microglia). Structurally, SIRPα is a transmembrane protein encoded on chromosome 20p13 and composed of 504 amino acids (Figure 1A). Its extracellular domain contains three immunoglobulin (Ig)-like domains, classifying it within the Ig superfamily (Figure 1B). Its N-terminal domain interacts with CD47, thereby mediating signal transduction (Figure 1C). The intracellular region contains four tyrosine residues forming two immunoreceptor tyrosine-based inhibitory motifs (ITIMs), which associate with Src homology 2 domain-containing phosphatases SHP-1 and SHP-2, underscoring its immunosuppressive function[14-16]. Notably, among SIRP family members, SIRPα has the longest intracellular domain.

Figure 1
Figure 1 Structural characteristics of signal regulatory protein alpha. A: The three-dimensional predicted structure of signal regulatory protein alpha (SIRPα). It contains an intracellular domain, an extracellular domain, and four tyrosine residues in the cytoplasmic region; B: The structure of SIRPα. It contains three immunoglobulin like domains in the extracellular domain. It contains four tyrosine residues in the cytoplasmic region, forming two typical tyrosine immunosuppressive motifs; C: CD47-SIRPα signaling pathway. ITIM: Immunoreceptor tyrosine-based inhibitory motif; SIRPα: Signal regulatory protein alpha. Parts of Figure 1 were created in pymol.

SIRPα is selectively expressed on the membrane surface of myeloid cells and nerve cells such as macrophages, neutrophils, dendritic cells[17,18], CD8+ T cell subsets during chronic infection[19], and brain tissue[20]. In contrast, it is absent on normal mature red blood cells, B cells, and T cells (https://www.proteinatlas.org). As an immunosuppressive receptor, SIRPα plays a critical role in immune regulation, modulating immune responses to maintain homeostasis[15] (Figure 1).

THE CLASSICAL SIGNALING PATHWAY OF SIRPΑ

The CD47-SIRPα signaling pathway is currently the most extensively characterized classical signaling pathway mediating tumor immune escape in the occurrence and development of tumors. As a ligand for SIRPα, CD47 has five transmembrane domains and a single extracellular IgSF domain[21,22]. Unlike SIRPα, which selectively expresses on myelloid cells such as macrophages, CD47 exhibits ubiqutions expression across various cell types[23], with frequent overexpression in cancer cells[24], including HCC cells[25]. After binding to CD47, SIRPα on macrophages triggers an inhibitory signaling cascade that suppresses phagocytosis[23]. This interaction enables CD47 to transmit a "don't eat me" signal to macrophages and mediates the phosphorylation of ITIMs in SIRPα’s cytoplasmic tail, promoting immune escape and protecting tumor cells[26] (Figure 1C). Beyond SIRPα, CD47 also binds to its family member SIRPγ. Multiple studies have confirmed that CD47 is highly expressed on normal red blood cells, preventing macrophage-mediated phagocytosis, whereas the loss of CD47 expression in aging red blood cells can triggers their phagocytic clearance[27].

In addition, abnormal expression of SIRPα on macrophages heightens their susceptibility to red blood cells, leading to increased clearance. Similarly, platelets and lymphocytes also inhibit macrophage phagocytosis through the CD47-SIRPα signaling pathway. These findings demonstrate that SIRPα serves as an innate immune sensor that recognizes self-antigen signals on the surface of host cells, especially CD47. Although CD47 is the main ligand for SIRPα, surfactant protein (SP)-A and SP-D have been identified as ligands for SIRPα expressed on alveolar macrophages, potentially serving as potential inhibitors of apoptotic cell phagocytosis[28].

THE ROLE AND CLINICAL SIGNIFICANCE OF SIRPΑ IN THE DEVELOPMENT OF HEPATOCELLULAR CARCINOMA
Molecular mechanisms of SIRPα in HCC progression

SIRPα primarily functions as an inhibitory immune receptor on macrophages, interacting with highly expressed CD47 on tumor cells to facilitate immune evasion[29-31]. Inhibiting SIRPα function can block the CD47-SIRPα pathway to promote tumor phagocytosis and myeloid cell clearance[32,33]. Analysis of The Cancer Genome Atlas (TCGA) data showed that both SIRPα and CD47 were highly expressed in several tumor types, including cholangiocarcinoma, esophageal cancer, head and neck squamous cell carcinoma, and gastric adenocarcinoma. This overexpression pattern suggests these tumors may utilize CD47-SIRPα signaling pathway to mediate immune suppression and promote immune escape. While SIRPα expression is at an intermediate level in HCC, TCGA data analysis shows that high expression of SIRPα is positively correlated with poor prognosis in HCC patients (Figure 2A). Further analysis of the HCCDB database indicates that SIRPα expression is higher in tumors compared to normal tissues (Figure 2B). Additionally, SIRPα expression levels showed an increasing trend in normal tissues, stromal tissues, and tumor tissues of HCC (Figure 2C) (http://Lifeome.net/database/hccdb/home.html). Although current studies support the role of SIRPα in the occurrence and development of HCC, the precise molecular mechanism underlying its function remain to be fully elucidated.

Figure 2
Figure 2 The expression of signal regulatory protein alpha in adjacent and tumor tissues of hepatocellular carcinoma. A: The overall survival of signal regulatory protein alpha (SIRPα) (The Cancer Genome Atlas, http://gepia.cancer-pku.cn/). High expression of SIRPα is positively correlated with poor prognosis in hepatocellular carcinoma (HCC) patients; B and C: Expression patterns of SIRPα in tumor, stromal and normal tissues of HCC. The expression of SIRPα showed an increasing trend in normal tissues, stromal tissues, and tumor tissues of HCC (http://Lifeome.net/database/hccdb/home.html). SIRPα: Signal regulatory protein alpha.

Qin et al[34] found that SIRPα was closely related to the occurrence, progression, and liver regeneration processes of HCC. This study showed that SIRPα overexpression in Sk-Hep-1 cells significantly upregulated the protein expression of nuclear factor kappa-B (NF-κB), and downregulated the protein expression of P65, P50, and cyclin D1. They proposed that SIRPα may negatively regulate the abnormal proliferation of HCC cells by modulating the protein content and localization of NF-κB, thereby affecting the expression of cyclins (particularly cyclin D1) in the signaling pathway. This is the first study to explore the mechanism of SIRPα in HCC development, providing a theoretical foundation and direction for future research. Meanwhile, Pan et al[35] reported that SIRPα expression was downregulated in monocytes/macrophages derived from the paracancerous tissues of HCC patients, with a partial recovery observed in intratumoral macrophages. Co-culture with HCC cells further suppressed SIRPα expression in macrophages. Mechanistically, SIRPα knockdown sustained the activation of NF-κB and PI3K-Akt signaling upon macrophage-tumor cell interaction, promoting macrophage migration, survival, and proinflammatory cytokine secretion. These finding suggest that SIRPα plays a key role in macrophage functional polarization within the tumor microenvironment and may contribute to tumor progression. Thus, SIRPα represents a potential therapeutic target for HCC treatment (Figure 2).

Therapeutic value of the CD47-SIRPα signaling pathway

Willingham et al[24] focused on the traditional mechanism of SIRPα in tumors: The CD47-SIRPα signaling pathway. They found that CD47 expression was 3.3 times higher in tumor cells compared to their corresponding normal counterparts. Based on the author's previous research: Blocking CD47-mediated SIRPα signaling with targeted monoclonal antibodies (mAbs) has been shown to promote phagocytosis of leukemia, lymphoma, and bladder cancer cells by macrophages[36-39]. Recent studies have expanded this approach to solid tumors, demonstrating that solid tumor cells treated with anti-CD47 blocking antibodies are efficiently phagocytosed by macrophages. Additionally, anti-CD47 treatment significantly inhibits the growth of various solid tumors, including HCC. This shows that the expression of CD47 is common mechanism used by human solid tumor cells (including HCC) to escape phagocytosis. Disrupting the CD47-SIRPα signaling pathway enables phagocytosis of solid tumor cells in vitro and inhibit tumor growth in the orthotopic xenotransplantation model through blocking CD47 mAbs. These findings are expected to apply all approaches of interfering with CD47-SIRPα interactions.

Acidic microenvironment[40], like hypoxia[41,42], inflammation or immune response, is a hallmark of the tumor microenvironment[43-45]. Jiang et al[46] compared risk scores with immune checkpoint expression and found that the high-risk group exhibited significantly higher levels of TNFSF4, SIRPα, CD276, and TNFSF15., indicating that high-risk individuals may potentially response to immunotherapy targeting these molecules. This suggests that high-risk patients may be more responsive to certain immunotherapies (such as anti-TNFSF4, anti-SIRPα, anti-CD276, and anti-TNFSF15), providing valuable clinical treatment insights for HCC patients.

Novel mechanism of SIRPα in regulating T-cell immune responses

Tomiyama et al[47] analyzed the expression of SIRPα by studying RNA sequencing data from 372 HCC tissues from the TCGA dataset and immunohistochemical staining of a cohort of 189 HCC patients. In a cohort of 189 patients, high expression of SIRPα was associated with lower recurrence free survival (RFS). High expression of SIRPα is associated with higher microvascular infiltration rates and lower serum albumin levels, as well as increased intratumoral infiltration of CD68 positive macrophages and myeloid derived suppressor cells (MDSCs). In spatial transcriptome sequencing, SIRPα expression is significantly correlated with CD163 expression. The high expression of SIRPα in HCC suggests poor prognosis and may be achieved by inhibiting macrophage phagocytosis of tumor cells, promoting MDSC infiltration, and inducing anti-tumor immunity. In HCC patients, SIRPα blockade can be considered to inhibit the development of HCC. Meanwhile, multiple factor analysis showed that patients with high expression of SIRPα, low infiltration of CD8+ T cells and high infiltration of MDSCs had significantly reduced RFS and OS rates[47]. This observation raises the question whether SIRPα exerts additional mechanisms of action involving CD8+ T cells beyond the conventional CD47-SIRPα signaling pathway. Huang et al[48] precisely confirmed this point. They found that compared to WT mice, KO-Sirpα (-/-) mice had significantly slower tumor growth and significantly prolonged overall survival after subcutaneous tumor loading with hepa 1-6 cells. At the same time, by injecting three times the number of tumor cells into SIRPα (-/-) mice whose subcutaneous tumors disappeared, they found that the tumor was completely suppressed, indicating that SIRPα (-/-) mice established strong immune memory after tumor stimulation, which usually relies on adaptive immune cells, particularly T cells. They found that SIRPα deficiency not only greatly inhibits the development of multiple solid tumors, but also is not related to CD47 expression on tumor cells. Instead, SIRPα (-/-) macrophages promote T cell recruitment into tumors through Syk/Btk dependent Ccl8 secretion[48]. This suggests that targeting SIRPα on bone marrow-derived cells may be a new approach to optimize solid tumor immunotherapy.

FUTURE CHALLENGES FOR SIRPΑ IN HEPATOCELLULAR CARCINOMA
Open up new avenues for research and treatment of tumor immunity

Although many mechanisms and functions of SIRPα in tumor immune processes remain unclear, some studies have shown that SIRPα is expressed on macrophages[49] and reduces macrophage phagocytic capacity through the CD47-SIRPα signaling pathway, leading tumor immune escape. This has been confirmed in various tumors[36-39]. This suggests that SIRPα may have important biological functions between tumors and the immune system. Consequently, SIRPα can serve not only as a biomarker, but also as a target for specific antibodies to relieve immune suppression, and enhance the anti-tumor activity of immune cells[24]. Additionally, preclinical studies demonstrate that combining SIRPα-CD47 inhibitors with antibodies that promote antibody-dependent cellular phagocytosis (ADCP), such as rituximab, enhance ADCP against cancer cells and improve anti-tumor efficacy[50-53]. According to ClinicalTrials.gov (https://clinicaltrials.gov/ct2/home), since 2016, it has been a total of 16 clinical trials related to SIRPα (Table 1), mostly focused on blocking the CD47-SIRPα signaling pathway (such as anti-SIRPα Ab or anti-CD47 Ab) to relieve tumor immune suppression, as well as combination therapy with other immune checkpoint antibodies or chemotherapy for various tumors. Currently, numerous clinical trials targeting SIRPα are underway, but there is only one clinical trial on HCC.

Table 1 Clinical trials related to signal regulatory protein alpha.
Tumor
Interventions
Phase
Number enrolled
NCT number
Study start
Sponsor/collaborators
LymphomaOntorpaceptII (recruiting)41NCT05507541April 19, 2023Mayo Clinic
Advanced solid tumor; breast cancerDrug: DS-1103a; Drug: T-DXdI (recruiting)78NCT05765851May 30, 2023Daiichi Sankyo
Colorectal cancerDrug: Evorpacept (ALX148); Drug: Cetuximab; Drug: PembrolizumabII (active, not recruiting)80NCT05167409July 28, 2022University of Colorado, Denver, CO, United States
Leukemia, myeloid, acute; myelodysplastic syndromesDrug: CC-95251; Drug: Azacitidine; Drug: VenetoclaxI (completed)56NCT05168202January 19, 2022Bristol-Myers Squibb
Solid tumors; mycosis fungoides; melanomaTTI-621 + PD-1/PD-L1 InhibitorI (terminated)56NCT02890368September 2016Pfizer
LymphomaDrug: CD47 Antagonist ALX148; Drug: Lenalidomide; Biological: RituximabI/II (recruiting)60NCT05025800October 13, 2021M.D. Anderson Cancer Center
Hepatocellular carcinomaSIRPα AbComplete30NCT02868255January 13, 2016Nantes University Hospital
Acute myeloid leukemiaDrug: Evorpacept; Drug: Venetoclax; Drug: AzacitidineI (terminated)14NCT04755244May 5, 2021ALX Oncology Inc.
Higher risk myelodysplastic syndromesDrug: Evorpacept; Drug: AzacitidineI/II (active, not recruiting)65NCT04417517October 2, 2020ALX Oncology Inc.
Multiple myelomaDrug: Elranatamab; Drug: Carfilzomib; Drug: MaplirpaceptI (recruiting)90NCT05675449December 14, 2022Pfizer
Brain cancerDrug: MagrolimabI (completed)13NCT05169944April 22, 2022University of California, San Francisco, CA, United States
Multiple myelomaDrug: TTI-622; Drug: Daratumumab Hyaluronidase-fihjI (active, not recruiting)7NCT05139225October 28, 2021Memorial Sloan Kettering Cancer Center
Bladder cancer; urothelial carcinomaDrug: Evorpacept; Drug: Enfortumab VedotinI (recruiting)30NCT05524545November 2, 2022ALX Oncology Inc.
Advanced solid cancers; hematologic cancersDrug: SRF231I (completed)148NCT03512340March 13, 2018Surface Oncology
Head and neck squamous cell carcinomaDrug: BI 765063; Drug: EzabenlimabI (active, not recruiting)48NCT05249426April 12, 2022Boehringer Ingelheim
Breast cancerDrug: ALX148; Drug: Fam-Trastuzumab Deruxtecan-Nxki; Drug: ZanidatamabI (recruiting)54NCT05868226December 22, 2022QuantumLeap Healthcare Collaborative
QuantumLeap Healthcare Collaborative

Since 2016, it has been a total of 16 clinical trials related to SIRPα, mostly focused on blocking the CD47-SIRPα signaling pathway, but there is relatively little research related to HCC.

The main problems about the research of SIRPα

Currently, the roles, functions, and mechanisms of SIRPα in HCC remain largely unclear, presenting several unresolved questions. Beyond its established role in reducing macrophage phagocytic capacity and promoting tumor immune escape through the CD47-SIRPα signaling pathways, it is uncertain whether SIRPα engages other signaling pathways in HCC, as observed in other cancers. For instance, studies have identified alternative pathways such as SIRPα/interleukin-6 axis, which may promote lung cancer development[54] (Figure 3A), the upregulation of SIRPα driving osteosarcoma metastasis through the "SP1 stable loop" and SLC 7A3 mediated arginine uptake[55] (Figure 3B), and SIRPα (-/-) macrophages enhancing T cell recruitment into tumors through Syk/Btk dependent Ccl8 secretion[48] (Figure 3C). These pathways have not been thoroughly investigated in HCC, especially the upstream and downstream genes directly affected by SIRPα. Studies have shown that sustained anti-tumor immune responses require stimulation from adaptive immune cells[56], suggesting that SIRPα plays important roles in other immune cells besides macrophages. The study by Yamada-Hunter et al[57] showed that enhancing macrophage phagocytosis through CD47 blockade can improve the efficacy of CAR T cell therapy. Although T cell depletion was greatly enhanced by CD47 blockade, macrophage phagocytosis limits the persistence of adoptively transferred T cells even in the absence of CD47-SIRPα blockade[57]. It is well known that CD47 blockade selectively depletes aging red blood cells. Prolonged CD47 blockade leads to increased expression of the "eat me" signal on T cells while reducing CD47 expression. This raises the possibility that the CD47-SIRPα signaling pathway may similarly regulate the clearance of aged T cells[53]. Additionally, future research should further explore the impact of SIRPα on the functional state of T cells. For example, can SIRPA directly regulate the function of T cells (such as exhaustion or apoptosis, etc.) to promote tumor progression (Figure 3)?

Figure 3
Figure 3 The mechanism of signal regulatory protein alpha. A: The signal regulatory protein alpha (SIRPα)/interleukin-6 axis may promote lung cancer development; B: Upregulation of SIRPα promotes osteosarcoma metastasis through the "SP1 stable loop" and SLC 7A3 mediated arginine uptake; C: SIRPα (-/-) macrophages promote T cell recruitment into tumors through Syk/Btk dependent Ccl8 secretion.
CONCLUSION

HCC remains a major global health challenge. Although immunotherapies such as PD-1/PD-L1 inhibitors have shown promise, their efficacy is limited to some patients, and there is an urgent need to explore new immune escape mechanisms. Studies have shown that the inhibitory receptor SIRPα of myeloid cells conveys the "Don't eat me" signal by binding to tumor cell CD47. SIRPα is highly expressed in HCC and is associated with poor prognosis, macrophage dysfunction and increased infiltration of MDSC. In addition to inhibiting phagocytosis, SIRPα can also regulate NF-κB signaling, T cell recruitment and cytokine production, suggesting that it has a broader immunomodulatory function in the progression of HCC. In the future, exploring the extracellular functions of SIRPα in macrophages may become an important research direction. Future research on SIRPα in HCC could focus on: (1) Elucidating its role in ICI resistance through PD-1/CTLA-4 pathway interactions and treatment response dynamics; (2) Developing targeted therapies via HCC-specific antagonists and TACE combinations; and (3) Investigating non-canonical functions using spatial transcriptomics and metabolic profiling. These studies will employ advanced techniques (CyTOF, scRNA-seq) and models (humanized mice, organoids) to expand therapeutic strategies beyond PD-1/PD-L1 inhibition.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade B, Grade C, Grade C

Novelty: Grade B, Grade C, Grade C

Creativity or Innovation: Grade B, Grade C, Grade C

Scientific Significance: Grade B, Grade C, Grade D

P-Reviewer: Chiang ZC, PhD, Professor, Taiwan; Ke Y, MD, PhD, Associate Professor, China S-Editor: Li L L-Editor: A P-Editor: Wang WB

References
1.  Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, Jemal A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74:229-263.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5690]  [Cited by in RCA: 8303]  [Article Influence: 8303.0]  [Reference Citation Analysis (2)]
2.  Villanueva A. Hepatocellular Carcinoma. N Engl J Med. 2019;380:1450-1462.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2066]  [Cited by in RCA: 3181]  [Article Influence: 530.2]  [Reference Citation Analysis (37)]
3.  Jemal A, Ward EM, Johnson CJ, Cronin KA, Ma J, Ryerson B, Mariotto A, Lake AJ, Wilson R, Sherman RL, Anderson RN, Henley SJ, Kohler BA, Penberthy L, Feuer EJ, Weir HK. Annual Report to the Nation on the Status of Cancer, 1975-2014, Featuring Survival. J Natl Cancer Inst. 2017;109:djx030.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 748]  [Cited by in RCA: 1112]  [Article Influence: 139.0]  [Reference Citation Analysis (0)]
4.  Vogel A, Meyer T, Sapisochin G, Salem R, Saborowski A. Hepatocellular carcinoma. Lancet. 2022;400:1345-1362.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1284]  [Cited by in RCA: 1244]  [Article Influence: 414.7]  [Reference Citation Analysis (41)]
5.  Li J, Liang YB, Wang QB, Li YK, Chen XM, Luo WL, Lakang Y, Yang ZS, Wang Y, Li ZW, Ke Y. Tumor-associated lymphatic vessel density is a postoperative prognostic biomarker of hepatobiliary cancers: a systematic review and meta-analysis. Front Immunol. 2024;15:1519999.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 38]  [Article Influence: 38.0]  [Reference Citation Analysis (0)]
6.  Wang QB, Li J, Zhang ZJ, Li YK, Liang YB, Chen XM, Luo WL, Lakang Y, Yang ZS, Liu GY, Liu Y, Li SX, Ke Y. The effectiveness and safety of therapies for hepatocellular carcinoma with tumor thrombus in the hepatic vein, inferior vena cave and/or right atrium: a systematic review and single-arm meta-analysis. Expert Rev Anticancer Ther. 2025;25:561-570.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 20]  [Cited by in RCA: 17]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
7.  Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, Lencioni R, Koike K, Zucman-Rossi J, Finn RS. Hepatocellular carcinoma. Nat Rev Dis Primers. 2021;7:6.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4432]  [Cited by in RCA: 3903]  [Article Influence: 975.8]  [Reference Citation Analysis (3)]
8.  Koyama S, Akbay EA, Li YY, Herter-Sprie GS, Buczkowski KA, Richards WG, Gandhi L, Redig AJ, Rodig SJ, Asahina H, Jones RE, Kulkarni MM, Kuraguchi M, Palakurthi S, Fecci PE, Johnson BE, Janne PA, Engelman JA, Gangadharan SP, Costa DB, Freeman GJ, Bueno R, Hodi FS, Dranoff G, Wong KK, Hammerman PS. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun. 2016;7:10501.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 870]  [Cited by in RCA: 1196]  [Article Influence: 132.9]  [Reference Citation Analysis (0)]
9.  Wilky BA. Immune checkpoint inhibitors: The linchpins of modern immunotherapy. Immunol Rev. 2019;290:6-23.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 107]  [Cited by in RCA: 143]  [Article Influence: 28.6]  [Reference Citation Analysis (0)]
10.  Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, Kudo M, Breder V, Merle P, Kaseb AO, Li D, Verret W, Xu DZ, Hernandez S, Liu J, Huang C, Mulla S, Wang Y, Lim HY, Zhu AX, Cheng AL; IMbrave150 Investigators. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N Engl J Med. 2020;382:1894-1905.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2542]  [Cited by in RCA: 4720]  [Article Influence: 944.0]  [Reference Citation Analysis (2)]
11.  Yang Y, Chen D, Zhao B, Ren L, Huang R, Feng B, Chen H. The predictive value of PD-L1 expression in patients with advanced hepatocellular carcinoma treated with PD-1/PD-L1 inhibitors: A systematic review and meta-analysis. Cancer Med. 2023;12:9282-9292.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 22]  [Cited by in RCA: 24]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
12.  Sangro B, Sarobe P, Hervás-Stubbs S, Melero I. Advances in immunotherapy for hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2021;18:525-543.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 264]  [Cited by in RCA: 859]  [Article Influence: 214.8]  [Reference Citation Analysis (0)]
13.  Suzuki H, Iwamoto H, Tanaka T, Sakaue T, Imamura Y, Masuda A, Nakamura T, Koga H, Hoshida Y, Kawaguchi T. Fibroblast growth factor inhibition by molecular-targeted agents mitigates immunosuppressive tissue microenvironment in hepatocellular carcinoma. Hepatol Int. 2024;18:610-622.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 5]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
14.  Fujioka Y, Matozaki T, Noguchi T, Iwamatsu A, Yamao T, Takahashi N, Tsuda M, Takada T, Kasuga M. A novel membrane glycoprotein, SHPS-1, that binds the SH2-domain-containing protein tyrosine phosphatase SHP-2 in response to mitogens and cell adhesion. Mol Cell Biol. 1996;16:6887-6899.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 350]  [Cited by in RCA: 369]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
15.  Logtenberg MEW, Scheeren FA, Schumacher TN. The CD47-SIRPα Immune Checkpoint. Immunity. 2020;52:742-752.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 100]  [Cited by in RCA: 439]  [Article Influence: 87.8]  [Reference Citation Analysis (0)]
16.  Kharitonenkov A, Chen Z, Sures I, Wang H, Schilling J, Ullrich A. A family of proteins that inhibit signalling through tyrosine kinase receptors. Nature. 1997;386:181-186.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 490]  [Cited by in RCA: 527]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
17.  Adams S, van der Laan LJ, Vernon-Wilson E, Renardel de Lavalette C, Döpp EA, Dijkstra CD, Simmons DL, van den Berg TK. Signal-regulatory protein is selectively expressed by myeloid and neuronal cells. J Immunol. 1998;161:1853-1859.  [PubMed]  [DOI]
18.  Seiffert M, Cant C, Chen Z, Rappold I, Brugger W, Kanz L, Brown EJ, Ullrich A, Bühring HJ. Human signal-regulatory protein is expressed on normal, but not on subsets of leukemic myeloid cells and mediates cellular adhesion involving its counterreceptor CD47. Blood. 1999;94:3633-3643.  [PubMed]  [DOI]
19.  Myers LM, Tal MC, Torrez Dulgeroff LB, Carmody AB, Messer RJ, Gulati G, Yiu YY, Staron MM, Angel CL, Sinha R, Markovic M, Pham EA, Fram B, Ahmed A, Newman AM, Glenn JS, Davis MM, Kaech SM, Weissman IL, Hasenkrug KJ. A functional subset of CD8(+) T cells during chronic exhaustion is defined by SIRPα expression. Nat Commun. 2019;10:794.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 43]  [Cited by in RCA: 45]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
20.  Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson Å, Kampf C, Sjöstedt E, Asplund A, Olsson I, Edlund K, Lundberg E, Navani S, Szigyarto CA, Odeberg J, Djureinovic D, Takanen JO, Hober S, Alm T, Edqvist PH, Berling H, Tegel H, Mulder J, Rockberg J, Nilsson P, Schwenk JM, Hamsten M, von Feilitzen K, Forsberg M, Persson L, Johansson F, Zwahlen M, von Heijne G, Nielsen J, Pontén F. Proteomics. Tissue-based map of the human proteome. Science. 2015;347:1260419.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7696]  [Cited by in RCA: 10565]  [Article Influence: 1056.5]  [Reference Citation Analysis (0)]
21.  Campbell IG, Freemont PS, Foulkes W, Trowsdale J. An ovarian tumor marker with homology to vaccinia virus contains an IgV-like region and multiple transmembrane domains. Cancer Res. 1992;52:5416-5420.  [PubMed]  [DOI]
22.  Mushegian A. Refining structural and functional predictions for secretasome components by comparative sequence analysis. Proteins. 2002;47:69-74.  [PubMed]  [DOI]
23.  Brown EJ, Frazier WA. Integrin-associated protein (CD47) and its ligands. Trends Cell Biol. 2001;11:130-135.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 604]  [Cited by in RCA: 685]  [Article Influence: 28.5]  [Reference Citation Analysis (0)]
24.  Willingham SB, Volkmer JP, Gentles AJ, Sahoo D, Dalerba P, Mitra SS, Wang J, Contreras-Trujillo H, Martin R, Cohen JD, Lovelace P, Scheeren FA, Chao MP, Weiskopf K, Tang C, Volkmer AK, Naik TJ, Storm TA, Mosley AR, Edris B, Schmid SM, Sun CK, Chua MS, Murillo O, Rajendran P, Cha AC, Chin RK, Kim D, Adorno M, Raveh T, Tseng D, Jaiswal S, Enger PØ, Steinberg GK, Li G, So SK, Majeti R, Harsh GR, van de Rijn M, Teng NN, Sunwoo JB, Alizadeh AA, Clarke MF, Weissman IL. The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc Natl Acad Sci U S A. 2012;109:6662-6667.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 905]  [Cited by in RCA: 1292]  [Article Influence: 99.4]  [Reference Citation Analysis (0)]
25.  Kim H, Bang S, Jee S, Paik SS, Jang K. Clinicopathological significance of CD47 expression in hepatocellular carcinoma. J Clin Pathol. 2021;74:111-115.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 19]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
26.  Oldenborg PA, Zheleznyak A, Fang YF, Lagenaur CF, Gresham HD, Lindberg FP. Role of CD47 as a marker of self on red blood cells. Science. 2000;288:2051-2054.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1241]  [Cited by in RCA: 1450]  [Article Influence: 58.0]  [Reference Citation Analysis (0)]
27.  Burger P, Hilarius-Stokman P, de Korte D, van den Berg TK, van Bruggen R. CD47 functions as a molecular switch for erythrocyte phagocytosis. Blood. 2012;119:5512-5521.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 179]  [Cited by in RCA: 247]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
28.  Janssen WJ, McPhillips KA, Dickinson MG, Linderman DJ, Morimoto K, Xiao YQ, Oldham KM, Vandivier RW, Henson PM, Gardai SJ. Surfactant proteins A and D suppress alveolar macrophage phagocytosis via interaction with SIRP alpha. Am J Respir Crit Care Med. 2008;178:158-167.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 144]  [Cited by in RCA: 168]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
29.  Nagahara M, Mimori K, Kataoka A, Ishii H, Tanaka F, Nakagawa T, Sato T, Ono S, Sugihara K, Mori M. Correlated expression of CD47 and SIRPA in bone marrow and in peripheral blood predicts recurrence in breast cancer patients. Clin Cancer Res. 2010;16:4625-4635.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 39]  [Cited by in RCA: 52]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
30.  Gholamin S, Mitra SS, Feroze AH, Liu J, Kahn SA, Zhang M, Esparza R, Richard C, Ramaswamy V, Remke M, Volkmer AK, Willingham S, Ponnuswami A, McCarty A, Lovelace P, Storm TA, Schubert S, Hutter G, Narayanan C, Chu P, Raabe EH, Harsh G 4th, Taylor MD, Monje M, Cho YJ, Majeti R, Volkmer JP, Fisher PG, Grant G, Steinberg GK, Vogel H, Edwards M, Weissman IL, Cheshier SH. Disrupting the CD47-SIRPα anti-phagocytic axis by a humanized anti-CD47 antibody is an efficacious treatment for malignant pediatric brain tumors. Sci Transl Med. 2017;9:eaaf2968.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 206]  [Cited by in RCA: 339]  [Article Influence: 42.4]  [Reference Citation Analysis (0)]
31.  Li B, Hao Y, He H, Fan Y, Ren B, Peng X, Zhou X, Cheng L. CD47-SIRPα Blockade Sensitizes Head and Neck Squamous Cell Carcinoma to Cetuximab by Enhancing Macrophage Adhesion to Cancer Cells. Cancer Res. 2024;84:3189-3206.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
32.  Murata Y, Saito Y, Kotani T, Matozaki T. Blockade of CD47 or SIRPα: a new cancer immunotherapy. Expert Opin Ther Targets. 2020;24:945-951.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 12]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
33.  Alvey CM, Spinler KR, Irianto J, Pfeifer CR, Hayes B, Xia Y, Cho S, Dingal PCPD, Hsu J, Smith L, Tewari M, Discher DE. SIRPA-Inhibited, Marrow-Derived Macrophages Engorge, Accumulate, and Differentiate in Antibody-Targeted Regression of Solid Tumors. Curr Biol. 2017;27:2065-2077.e6.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 73]  [Cited by in RCA: 88]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
34.  Qin JM, Wan XW, Zeng JZ, Wu MC. Effect of Sirpalpha1 on the expression of nuclear factor-kappa B in hepatocellular carcinoma. Hepatobiliary Pancreat Dis Int. 2007;6:276-283.  [PubMed]  [DOI]
35.  Pan YF, Tan YX, Wang M, Zhang J, Zhang B, Yang C, Ding ZW, Dong LW, Wang HY. Signal regulatory protein α is associated with tumor-polarized macrophages phenotype switch and plays a pivotal role in tumor progression. Hepatology. 2013;58:680-691.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 32]  [Cited by in RCA: 34]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
36.  Jaiswal S, Jamieson CH, Pang WW, Park CY, Chao MP, Majeti R, Traver D, van Rooijen N, Weissman IL. CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell. 2009;138:271-285.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1313]  [Cited by in RCA: 1240]  [Article Influence: 77.5]  [Reference Citation Analysis (0)]
37.  Chao MP, Alizadeh AA, Tang C, Myklebust JH, Varghese B, Gill S, Jan M, Cha AC, Chan CK, Tan BT, Park CY, Zhao F, Kohrt HE, Malumbres R, Briones J, Gascoyne RD, Lossos IS, Levy R, Weissman IL, Majeti R. Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma. Cell. 2010;142:699-713.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 906]  [Cited by in RCA: 895]  [Article Influence: 59.7]  [Reference Citation Analysis (0)]
38.  Majeti R, Chao MP, Alizadeh AA, Pang WW, Jaiswal S, Gibbs KD Jr, van Rooijen N, Weissman IL. CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell. 2009;138:286-299.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1418]  [Cited by in RCA: 1368]  [Article Influence: 85.5]  [Reference Citation Analysis (0)]
39.  Chan KS, Espinosa I, Chao M, Wong D, Ailles L, Diehn M, Gill H, Presti J Jr, Chang HY, van de Rijn M, Shortliffe L, Weissman IL. Identification, molecular characterization, clinical prognosis, and therapeutic targeting of human bladder tumor-initiating cells. Proc Natl Acad Sci U S A. 2009;106:14016-14021.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 516]  [Cited by in RCA: 497]  [Article Influence: 31.1]  [Reference Citation Analysis (0)]
40.  Helmlinger G, Yuan F, Dellian M, Jain RK. Interstitial pH and pO2 gradients in solid tumors in vivo: high-resolution measurements reveal a lack of correlation. Nat Med. 1997;3:177-182.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1206]  [Cited by in RCA: 1153]  [Article Influence: 41.2]  [Reference Citation Analysis (0)]
41.  Zhang MS, Cui JD, Lee D, Yuen VW, Chiu DK, Goh CC, Cheu JW, Tse AP, Bao MH, Wong BPY, Chen CY, Wong CM, Ng IO, Wong CC. Hypoxia-induced macropinocytosis represents a metabolic route for liver cancer. Nat Commun. 2022;13:954.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 66]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
42.  Yao B, Li Y, Chen T, Niu Y, Wang Y, Yang Y, Wei X, Liu Q, Tu K. Hypoxia-induced cofilin 1 promotes hepatocellular carcinoma progression by regulating the PLD1/AKT pathway. Clin Transl Med. 2021;11:e366.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 28]  [Cited by in RCA: 38]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
43.  Terashima Y, Toda E, Itakura M, Otsuji M, Yoshinaga S, Okumura K, Shand FHW, Komohara Y, Takeda M, Kokubo K, Chen MC, Yokoi S, Rokutan H, Kofuku Y, Ohnishi K, Ohira M, Iizasa T, Nakano H, Okabe T, Kojima H, Shimizu A, Kanegasaki S, Zhang MR, Shimada I, Nagase H, Terasawa H, Matsushima K. Targeting FROUNT with disulfiram suppresses macrophage accumulation and its tumor-promoting properties. Nat Commun. 2020;11:609.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 28]  [Cited by in RCA: 74]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
44.  Massara M, Bonavita O, Savino B, Caronni N, Mollica Poeta V, Sironi M, Setten E, Recordati C, Crisafulli L, Ficara F, Mantovani A, Locati M, Bonecchi R. ACKR2 in hematopoietic precursors as a checkpoint of neutrophil release and anti-metastatic activity. Nat Commun. 2018;9:676.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 45]  [Cited by in RCA: 77]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
45.  Corbet C, Bastien E, Santiago de Jesus JP, Dierge E, Martherus R, Vander Linden C, Doix B, Degavre C, Guilbaud C, Petit L, Michiels C, Dessy C, Larondelle Y, Feron O. TGFβ2-induced formation of lipid droplets supports acidosis-driven EMT and the metastatic spreading of cancer cells. Nat Commun. 2020;11:454.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 95]  [Cited by in RCA: 218]  [Article Influence: 43.6]  [Reference Citation Analysis (0)]
46.  Jiang P, Xue W, Xi C, Zhuang L, Yuan Z, Liu Z, Sun T, Xu X, Tan Y, Ding W. A new acidic microenvironment related lncRNA signature predicts the prognosis of liver cancer patients. Front Oncol. 2022;12:1016721.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 22]  [Cited by in RCA: 14]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
47.  Tomiyama T, Itoh S, Iseda N, Toshida K, Kosai-Fujimoto Y, Tomino T, Kurihara T, Nagao Y, Morita K, Harada N, Liu YC, Ozaki D, Kohashi K, Oda Y, Mori M, Yoshizumi T. Clinical Significance of Signal Regulatory Protein Alpha (SIRPα) Expression in Hepatocellular Carcinoma. Ann Surg Oncol. 2023;30:3378-3389.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]
48.  Huang C, Wang X, Wang Y, Feng Y, Wang X, Chen S, Yan P, Liao J, Zhang Q, Mao C, Li Y, Wang L, Wang X, Yi W, Cai W, Chen S, Hong N, He W, Chen J, Jin W. Sirpα on tumor-associated myeloid cells restrains antitumor immunity in colorectal cancer independent of its interaction with CD47. Nat Cancer. 2024;5:500-516.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 41]  [Article Influence: 41.0]  [Reference Citation Analysis (0)]
49.  Barclay AN, Van den Berg TK. The interaction between signal regulatory protein alpha (SIRPα) and CD47: structure, function, and therapeutic target. Annu Rev Immunol. 2014;32:25-50.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 431]  [Cited by in RCA: 448]  [Article Influence: 40.7]  [Reference Citation Analysis (0)]
50.  Liu J, Wang L, Zhao F, Tseng S, Narayanan C, Shura L, Willingham S, Howard M, Prohaska S, Volkmer J, Chao M, Weissman IL, Majeti R. Pre-Clinical Development of a Humanized Anti-CD47 Antibody with Anti-Cancer Therapeutic Potential. PLoS One. 2015;10:e0137345.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 255]  [Cited by in RCA: 383]  [Article Influence: 38.3]  [Reference Citation Analysis (0)]
51.  Kauder SE, Kuo TC, Harrabi O, Chen A, Sangalang E, Doyle L, Rocha SS, Bollini S, Han B, Sim J, Pons J, Wan HI. ALX148 blocks CD47 and enhances innate and adaptive antitumor immunity with a favorable safety profile. PLoS One. 2018;13:e0201832.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 89]  [Cited by in RCA: 122]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
52.  Voets E, Paradé M, Lutje Hulsik D, Spijkers S, Janssen W, Rens J, Reinieren-Beeren I, van den Tillaart G, van Duijnhoven S, Driessen L, Habraken M, van Zandvoort P, Kreijtz J, Vink P, van Elsas A, van Eenennaam H. Functional characterization of the selective pan-allele anti-SIRPα antibody ADU-1805 that blocks the SIRPα-CD47 innate immune checkpoint. J Immunother Cancer. 2019;7:340.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 31]  [Cited by in RCA: 57]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
53.  Liu J, Xavy S, Mihardja S, Chen S, Sompalli K, Feng D, Choi T, Agoram B, Majeti R, Weissman IL, Volkmer JP. Targeting macrophage checkpoint inhibitor SIRPα for anticancer therapy. JCI Insight. 2020;5:e134728.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 15]  [Cited by in RCA: 53]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
54.  Wang B, Pan L, Chen M, Ma Y, Gao J, Tang D, Jiang Z. SIRP-alpha-IL-6 axis induces immunosuppressive macrophages in non-small-cell lung cancer. Biochem Biophys Res Commun. 2023;682:386-396.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
55.  Wang P, Song Y, Li H, Zhuang J, Shen X, Yang W, Mi R, Lu Y, Yang B, Ma M, Shen H. SIRPA enhances osteosarcoma metastasis by stabilizing SP1 and promoting SLC7A3-mediated arginine uptake. Cancer Lett. 2023;576:216412.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 18]  [Reference Citation Analysis (0)]
56.  Sockolosky JT, Dougan M, Ingram JR, Ho CC, Kauke MJ, Almo SC, Ploegh HL, Garcia KC. Durable antitumor responses to CD47 blockade require adaptive immune stimulation. Proc Natl Acad Sci U S A. 2016;113:E2646-E2654.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 200]  [Cited by in RCA: 295]  [Article Influence: 32.8]  [Reference Citation Analysis (0)]
57.  Yamada-Hunter SA, Theruvath J, McIntosh BJ, Freitas KA, Lin F, Radosevich MT, Leruste A, Dhingra S, Martinez-Velez N, Xu P, Huang J, Delaidelli A, Desai MH, Good Z, Polak R, May A, Labanieh L, Bjelajac J, Murty T, Ehlinger Z, Mount CW, Chen Y, Heitzeneder S, Marjon KD, Banuelos A, Khan O, Wasserman SL, Spiegel JY, Fernandez-Pol S, Kuo CJ, Sorensen PH, Monje M, Majzner RG, Weissman IL, Sahaf B, Sotillo E, Cochran JR, Mackall CL. Engineered CD47 protects T cells for enhanced antitumour immunity. Nature. 2024;630:457-465.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 48]  [Cited by in RCA: 37]  [Article Influence: 37.0]  [Reference Citation Analysis (0)]