Open Access

Progress in cancer research on the regulator of phagocytosis CD47, which determines the fate of tumor cells (Review)

  • Authors:
    • Fan Wu
    • Hongyuan Pang
    • Fan Li
    • Mengqing Hua
    • Chuanwang Song
    • Jie Tang
  • View Affiliations

  • Published online on: April 9, 2024     https://doi.org/10.3892/ol.2024.14389
  • Article Number: 256
  • Copyright: © Wu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cluster of differentiation 47 (CD47) is a transmembrane protein that is widely and moderately expressed on the surface of various cells and can have an essential role in mediating cell proliferation, migration, phagocytosis, apoptosis, immune homeostasis and other related responses by binding to its ligands, integrins, thrombospondin‑1 and signal regulatory protein α. The poor prognosis of cancer patients is closely associated with high expression of CD47 in glioblastoma, ovarian cancer, breast cancer, bladder cancer, colon cancer and hepatocellular carcinoma. Upregulation of CD47 expression facilitates the growth of numerous types of tumor cells, while downregulation of its expression promotes phagocytosis of tumor cells by macrophages, thereby limiting tumor growth. In addition, blocking CD47 activates the cyclic GMP‑AMP (cGAMP) synthase/cGAMP/interferon gene stimulating factor signaling pathway and initiates an adaptive immune response that kills tumor cells. The present review describes the structure, function and interactions of CD47 with its ligands, as well as its regulation of phagocytosis and tumor cell fate. It summarizes the therapeutics, mechanisms of action, research advances and challenges of targeting CD47. In addition, this paper provides an overview of the latest therapeutic options for targeting CD47, such as chimeric antigen receptor (CAR) T‑cells, CAR macrophages and nanotechnology‑based delivery systems, which are essential for future clinical research on targeting CD47.

Introduction

Cluster of differentiation 47 (CD47) expression levels are influenced by an organism's physiological state and cell type (1). Under normal physiological conditions, the expression level of CD47 has a vital role in maintaining homeostasis. For instance, young erythrocytes have high CD47 expression on their surface. By contrast, senescent erythrocytes have low CD47 expression on their surface, which allows macrophages to eliminate CD47 for erythrocyte renewal (2). As previously reported, binding different ligands to CD47 also results in different biological effects. For example, CD47 binds to signal regulatory protein α (SIRPα) to activate a signaling pathway that inhibits phagocytosis and the killing of tumor cells by macrophages in the tumor microenvironment (TME) by modulating the immune response (3). Under pathological conditions, CD47 is highly expressed in hematological tumors, and by binding to its ligand SIRPα, CD47 transmits a series of inhibitory signals to macrophages; consequently, the phagocytosis of tumor cells by macrophages is prevented (4). The expression level of CD47 and blockade of the signaling pathway activated by CD47 also significantly impact the fate of tumor cells, and the upregulation or downregulation of CD47 expression and blockade of CD47 signaling can determine the fate of tumor cells. Blocking CD47 signaling can also determine the survival of tumor cells. In recent years, blocking CD47 has emerged as a potential therapeutic strategy for tumor immunotherapy (5), and immunotherapies targeting CD47 have achieved significant success in certain cancer patients. However, remission rates vary; not all individuals benefit from current treatments (6). Various drugs targeting CD47, such as monoclonal antibodies (mAbs), SIRPα fusion proteins (SIRPα-Fc), bispecific antibodies (BsAbs), small-molecule inhibitors and nanotechnology-based delivery systems, are being developed (7,8). Preclinical studies and early clinical trials have demonstrated that CD47-targeted therapies have a promising future for application. In addition, CD47-targeted therapies have potential limitations and challenges, including adverse reactions such as anemia and thrombocytopenia, as well as resistance to drugs (9).

CD47

In 1990, Brown et al (10) first identified CD47 as a cell surface protein associated with αvβ3 integrins in placenta and platelets. This protein was subsequently shown to regulate integrins and leukocyte responsiveness to extracellular matrix proteins; hence, it was named integrin-associated protein (IAP) (11,12).

CD47 has a molecular weight of 45–55 kDa and is a member of the immunoglobulin (Ig) superfamily (IgSF) (4,13,14). Its molecular structure includes one N-terminal extracellular IgG-like domain, five highly hydrophobic transmembrane segments and one hydrophilic cytoplasmic tail at the C-terminus. Hatherley et al (15,16) investigated the crystal structure of the IgG structural domain. They showed that the structure of CD47 has a typical IgV-like fold and is similar to that of myelin oligodendrocyte glycoprotein. CD47 mediates vascular smooth muscle cell (VSMC) proliferation and migration (17), as well as platelet activation and spreading (18), and recruits granulocytes and T cells to the site of infection (11).

Although an existing review (19) has summarised the structure and function of CD47, a description of the structure and function of CD47 isoforms is lacking. This review refers to previous studies reporting that CD47 has four traditional isoforms in human cell lines and tissues, all of which have different amino acid lengths in the cytoplasmic tail (19). In a recent study, sequence analysis of cDNA cloned from human skeletal muscle revealed that, in addition to the four traditional isoforms, human CD47 has a new isoform 5, which features an entirely different cytoplasmic tail amino acid length and amino acid sequence compared with the four traditional isoforms (20,21).

The distribution and functions of different isoforms within tissues vary. bladder, ovarian and breast cancer cells are examples of keratinocytes and tumor cells expressing type 1 CD47 (22,23). The most extensively expressed form, type 2 CD47, is mainly involved in signaling between astrocytes and is primarily expressed in hematopoietic cells, epithelial cells and vascular endothelial cells. Neuronal, testicular and intestinal mucosal cells explicitly express type 3 and 4 CD47 (24). Types 3 and 4 are thought to be closely related to memory mechanisms because their expression is markedly elevated in the brains of rats with good memory (25,26). Protein linking IAP with cytoskeleton 1 (PLIC-1) cytoplasmic protein regulates cyclic adenosine monophosphate (cAMP) signaling by CD47 by binding to the cytoplasmic tails of types 2 and 4, recruiting heterotrimeric G proteins to CD47 (27), inhibiting chemotactic signaling induced by the Gi-coupled receptor C-X-C motif chemokine receptor 4 (28) and activating the PI3K/Akt pathway in astrocytomas (29). More research is required to determine the functional distinctions between the cytoplasmic tails of the various CD47 isoforms, as the studies on this aspect of CD47 isoforms have been minimal in recent years, resulting in a limited understanding of the regulatory mechanisms and roles of the cytoplasmic tails of different isoforms of CD47.

In addition, certain cells can adapt to various physiological and pathological changes by switching their subtype, e.g., Reinhold et al (23) used PCR to detect mRNA expression and found that primary mouse endothelial cells cultured in vitro predominantly expressed CD47 type 2 mRNA, and endothelial cells transformed with intermediate T antigen expressed all four types of mRNA. However, certain researchers dispute this; for instance, Mateo et al (30) observed no change in the expression of CD47 isoforms. These studies indicate that the role of the CD47 types in tumorigenesis and development and the mechanism of interconversion require further investigation.

CD47 receptors

CD47 receptors include integrin, thrombospondin-1 (TSP-1) and SIRPα. Based on published reviews, it may be summarized that CD47 affects multiple biological functions of target cells by binding to these ligands (31,32). In addition, the gene expression of the three ligands of CD47 under physiological conditions and in tumors may be summarized through the GEPIA database (http://gepia.cancer-pku.cn/index.html) and the Human Protein Atlas (https://www.proteinatlas.org/), as elaborated below.

Interaction with integrins

Integrins are transmembrane ligands that bridge the gap between cells and the extracellular matrix and regulate signaling processes such as the cell cycle, morphology and motility (26,32,33). CD47 was initially found to interact with αvβ3 intergrin, hence the designation IAP. Under normal physiological conditions, αvβ3 integrins are mainly expressed in cardiomyocytes, oligodendrocytes and astrocytes, while under pathological conditions, they may be widely expressed mainly in cancers, such as glioblastoma, esophageal, thyroid and pancreatic cancers. The CD47-integrin complex may activate multiple heterotrimeric G proteins by linking IAP to PLIC-1, thereby inducing CD47 to activate cAMP signaling (34). Lindberg et al (35), through a study using a CD47-deficient human cell line, showed that CD47 is required for αvβ3 integrin-mediated binding of hyaluronan to encapsulated microbeads. In addition to αvβ3 integrin, CD47 binds to αIIbβ3 integrin and induces platelet aggregation and increased adhesion spot kinase tyrosine phosphorylation (18). In addition, CD47 binds to α4β1 integrin and mediates reticulocyte adhesion (36); CD47 binds to α5β1 integrin and is involved in chondrocyte mechanotransduction (37); and CD47 binds to α6β1 integrin and has a role in fibrillar β-amyloid-mediated activation and phagocytosis of microglia (38).

Interaction with TSP

TSP is an extracellular matrix calcium-binding glycoprotein that is highly expressed on monocytes, mucus cells and macrophages under normal physiological conditions and is widely expressed in cancers, such as breast adenocarcinoma carcinoma, lung adenocarcinoma, pancreatic adenocarcinoma and gastric adenocarcinoma, mainly under pathological conditions. There are currently five known isoforms of TSP, i.e., TSP-1-5 (39). TSP-1 is the first identified endogenous ligand of CD47 and it has a variety of biological functions, including the inhibition of angiogenesis, activation of transforming growth factor-β and participation in tissue repair (40). Protein-related studies have shown that TSP-1 binds to the CD47 extracellular IgV structural domain through its C-terminal structural domain peptide 4N1K and has a role in several biological processes, including inflammation, immune response, cell proliferation, apoptosis, adhesion and migration (41). The mechanism of CD47-TSP-1 interaction has not been studied in detail because the crystal structure of the CD47-TSP-1 complex still needs to be clarified. Early experiments have shown that CD47 affects signaling through heterotrimeric Gi proteins in a pertussis toxin-sensitive manner (28), thereby modulating TSP-1-induced cell spreading and platelet activation. Isenberg et al (42) measured cGMP levels by immunoassay, indicating that binding of CD47 to TSP-1 inhibits nitric oxide signaling in endothelial and VSMCs, thereby promoting platelet aggregation. To date, we have found that CD47-TSP-1 expression serves as a marker for predicting patient response to immune checkpoint blockade therapy, but there is no targeted therapy for the CD47-TSP-1 axis. It is hypothesized that this may be because CD47 has little effect on the adaptive immune response through its interaction with TSP-1, and therefore, blocking the CD47-TSP-1 axis has little clinical therapeutic significance. However, a novel immunotherapeutic drug, TAX2 peptide, which acts as an orthosteric antagonist of the interaction between TSP-1 and CD47, has shown a good safety profile in mouse models of ovarian cancer and is effective in killing tumor cells (43).

Interaction with SIRPα

SIRPα, the ligand with the highest affinity for CD47, is a member of the SIRP family and was first identified by Kharitonenkov et al (44) in the 1990s. Belonging to the IgSF, under normal physiological conditions, SIRPα is extensively expressed on the surface of cells such as monocytes, macrophages, neutrophils, dendritic cells (DCs) and microglia. Under pathological conditions, it is widely expressed in cancers such as glioblastoma, melanoma, renal cancer and head and neck cancer (45,46).

The intracellular region of SIRPα contains four tyrosine phosphorylation sites and two immunoreceptor tyrosine inhibition motifs (ITIMs), and the extracellular region has three IgSF structural domains, namely, one N-terminal IgV-like domain and two C-like domains (47,48). The crystal structure of the N-terminal IgV-like domain of SIRPα suggested an IgV-like fold and four-loop structure (BC, CD, DE and FG loops) with an overall structure similar to that of the T-cell receptor (16,49).

SIRPα binds to CD47 through its N-terminal FG and BC loop, thus forming a highly entangled, well-fitted complex structure (15). The long disulfide bond between Cys33 of the IgV structural domain and Cys263 of the transmembrane structural domain in CD47 is essential for enhancing binding to SIRPα (5052). According to X-ray computational crystallography calculations and analysis, when CD47 interacts with SIRPα, the total distance between the two cell types approximates the entire distance of the immune synapse (~14 nm) (53). Therefore, the binding of SIRPα to CD47 may occur via an antigen receptor rather than through the usual cell-cell structural domain binding interaction (16).

The binding of SIRPα to CD47 promotes the phosphorylation of the intracellular region of the ITIM (15,47). Phosphorylated ITIM recruits and activates Src homology region 2 (SH2)-containing tyrosine phosphatase-1 (SHP-1) and SHP-2 (54), which affects cytoskeletal function by inactivating motor myosin IIA (55), thereby blocking tyrosine phosphorylation-dependent signaling pathways and limiting phagocytosis by macrophages and others (47). Although SHP-1 and SHP-2 are typically inactive, phosphorylated ITIM recruits the SH2 structural domain to the cell membrane, and a change in its conformation activates SHP-1 and SHP-2. SHP-1 is present mainly in hematopoietic and epithelial cells and is selectively expressed in myeloid cells, which function as a negative regulator of phagocytosis. By contrast, SHP-2 is widely expressed and promotes cell proliferation, growth and migration mainly by regulating the GTP-binding proteins RAS and Rho (26).

CD47-SIRPα interactions not only regulate the maintenance of lymphocyte homeostasis (56), DC maturation and activation (57), the correct localization of DC subpopulations in sub-lymphoid organs and cell migration (58) but also have an essential role during remodeling of the nervous system and bone tissues (59). The cellular responses regulated by CD47-SIRPα interactions depend upon bidirectional signaling between CD47 and SIRPα: CD47 on host cells acts as a ‘self-tag’ (60) and regulates phagocytosis by binding to SIRPα. How this regulates phagocytosis will be further discussed in a later section.

CD47 and SIRPα: Bidirectional regulation of the immune system

Complex cellular communication systems in multicellular organisms have evolved to ensure adequate intercellular communication, which is crucial for cell differentiation, tissue and organ formation, individual development in multicellular organisms and immune function regulation (61).

The interaction between CD47 and SIRPα constitutes an intercellular communication system whose role in regulating immune system function is bidirectional (62). The CD47-SIRPα signaling pathway negatively regulates DC activation. The fusion protein of CD47, when bound to SIRPα, inhibits the phenotype and function of immature DCs and the production of cytokines by mature DCs (63). However, considering its role in antigen presentation, SIRPα has a positive regulatory effect. SIRPα is abundantly expressed on the surface of mature DCs. When the immune system responds to pathogens, SIRPα helps DCs present relevant antigens to T cells and costimulatory molecules associated with initiating T cells, thus promoting T-cell activation and proliferation (1,19).

CD47-SIRPα regulates phagocytosis

Phagocytosis is the process by which tissue cell debris and apoptotic cells are engulfed and digested, and this process helps maintain a stable balance in the body's internal environment. CD47 has a vital role in regulating phagocytosis. This regulatory function is mediated by binding to the inhibitory receptor SIRPα on phagocytes to activate the CD47-SIRPα signaling pathway. CD47 binds to SIRPα and sends an inhibitory ‘do not eat me’ signal to phagocytes, thus limiting phagocytosis (3,64) (Fig. 1).

The most characterized function of the CD47-SIRPα signaling pathway in vivo is the clearance of senescent and apoptotic erythrocytes. Okazawa et al (65) reported that the primary site of erythrocyte macrophage clearance is the red pulp of the spleen, suggesting that erythrocyte clearance is mediated by splenic red pulp macrophages. SIRPα is abundant in these macrophages and Ishikawa-Sekigami et al (66) demonstrated that erythrocyte clearance was significantly increased in SIRPα mutant mice injected with normal erythrocytes. This increase is because the mutated form of SIRPα expressed by SIRPα mutant mice cannot bind to SHP-1 or SHP-2 due to the lack of cytoplasmic domains, and SIRPα fails to exert an inhibitory effect on the CD47-SIRPα signaling pathway. As a result, the phagocytosis of red blood cells by splenic red pulp macrophages is enhanced (65). This phenomenon is observed not only in erythrocytes but also in platelets. Previous studies have demonstrated that SIRPα mutant mice lacking the cytoplasmic structural domains exhibit thrombocytopenia in the SIRPα mutant mouse model and clear platelets from the blood of the mutant mice at a more rapid rate when compared to wild-type mice (13,67).

CD47-SIRPα signaling also has an essential regulatory role in hematopoietic stem cell (HSC) transplantation. HSCs upregulate CD47 expression to protect themselves from phagocytosis by macrophages, thus achieving successful implantation (68,69). In general, the CD47 of one species has little interaction with the SIRPα of another species. However, higher-polymorphism SIRPα on macrophages was observed in a nonobese diabetic (NOD)-severe combined immunodeficiency xenograft mouse model when compared with other mouse lines. These cells have an exceptionally high affinity for human CD47, even higher than the mouse-mouse or human-human affinity of CD47 and SIRPα (70,71). Theocharides et al (72) demonstrated that implantation of normal human HSCs in NOD mice was also dependent on the interaction of human CD47 with SIRPa in NOD mice by implanting HSCs into a NOD mouse model. These studies demonstrated that the interaction between CD47 on human HSCs and SIRPα on macrophages is critical for the successful implantation of HSCs. In addition, human SIRPα is polymorphic and each polymorphic variant has a different affinity for human CD47 in vitro. This finding suggested that the human SIRPα polymorphism is critical for successfully implanting HSCs (73,74).

The ‘do not eat me’ signal from the CD47-SIRPα signaling pathway is also used to maintain homeostasis in body. The body must remove various cells, including those that are overproduced, damaged or aged. One removal mechanism is apoptosis, through which macrophages clear apoptotic cells precisely and efficiently. This mechanism is a key ‘do not eat me’ signal from CD47 that occurs on the surface of healthy cells, and binds to SIRPα inhibitory receptors on macrophages to prevent them from being eaten by macrophages. CD47 expression on the surface of apoptotic cells is downregulated, thereby attenuating the inhibitory signal generated by CD47 binding to SIRPα. By contrast, low IgG or C3b opsonization levels can cause the phagocytosis of apoptotic cells by macrophages (75).

CD47 signaling regulates tumor cell fate

Malignant tumors like glioblastoma, acute lymphoblastic leukemia, as well as ovarian, breast, gastric and lung cancers express high levels of CD47 (7678). Liu et al (79) used flow cytometry to detect the expression of CD47 in isolated primary lung cancer cells and adjacent normal cells and the results showed that the expression level of CD47 in tumor cells was higher than that in normal cells. There were apparent differences between subtypes of lung cancer, with the highest expression of CD47 in small-cell lung cancer, followed by lung adenocarcinoma, and the lowest in lung squamous carcinoma (79). Furthermore, compared to normal myeloid cells from healthy individuals, acute myeloid leukemia (AML) and chronic myeloid leukemia cells expressed higher levels of CD47. Furthermore, a positive association was found between high levels of CD47 expression and poor treatment response and patient prognosis (80). A study confirmed that CD47 mRNA and protein levels were higher in leukemic stem cells of patients with AML than in normal healthy stem cells (81). In a study on Epstein-Barr virus (EBV)-associated gastric cancer (EBVaGC), the expression of CD47 in EBVaGC was higher than that in EBV-negative gastric cancer tissue samples, which also indicated that high expression of CD47 was associated with poor prognosis in EBVaGC (82). Yu et al (83) detected the expression of CD47 in ovarian cancer tissues by immunohistochemistry, which showed that the prognosis of patients with low expression of CD47 was better than that of patients with high CD47 expression. The above studies indicate that CD47 expression levels are closely related to the prognosis of patients with cancer.

Recent research has demonstrated that controlling the expression of CD47 in tumor cells and inhibiting the signaling pathway that CD47 activates have crucial regulatory roles in determining the fate of tumor cells. The mechanisms of action include the following: i) Upregulation of CD47 expression, which binds to the macrophage surface receptor SIRPα and transmits the ‘do not eat me’ signal to promote phagocytosis of tumors by macrophages; ii) blockade of CD47 enhances the phagocytosis of tumor cells by DCs and promotes antigen delivery from DCs to T lymphocytes, initiating an antitumor adaptive immune response; iii) blockade of CD47 is capable of clearing tumor cells through natural killer cell-mediated antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) to clear tumor cells; iv) blockade of CD47 also activates the apoptotic pathway and directly induces apoptosis in tumor cells.

The expression level of CD47 is regulated by transcription factors such as nuclear factor κB (NF-κB), the MYC oncogene and hypoxia-inducible factor-1 (HIF-1), which regulate the phagocytosis of tumor cells by upregulating or downregulating the expression of CD47 (4). In a T-cell acute lymphoblastic leukemia xenograft model, MYC directly binds to the CD47 promoter and upregulates its expression, thus promoting the growth of tumor cells. By contrast, inactivation of MYC downregulates the expression of CD47 and enhances macrophage infiltration and phagocytosis, thereby inhibiting tumor cell growth (84). In addition, activated NF-κB directly binds to specific enhancer components of CD47 and upregulates CD47 expression in breast cancer cells, thereby promoting tumor growth (85). In a clinical analysis of thousands of patients with breast cancer, Zhang et al (78) reported a strong correlation between CD47 and HIF-1. Under hypoxic conditions, HIF-1 binds to the CD47 promoter and upregulates its expression, thereby inhibiting the phagocytosis of breast cancer cells (84). In addition, ERK signaling inhibits tumor-cell phagocytosis by activating nuclear respiratory factor-1 and upregulating CD47 expression in melanoma cells. Conversely, microRNA (miRNA)-mediated downregulation of CD47 expression promotes tumor-cell phagocytosis. MiR-708 is inversely associated with CD47 expression and its binding to the 3′-untranslated region of CD47 induces tumor-cell phagocytosis by suppressing CD47 expression (86). In multiple myeloma, CD47 expression on the surface of myeloma cells can be inhibited by upregulating the expression of the tumor suppressor gene miRNA-155, thereby inducing phagocytosis of tumor cells by macrophages (87) (Fig. 2).

Reputable reviews (64,88) have shown that transcription factors, oncogenes and miRNAs may control CD47 expression in tumor cells; however, regulation of CD47 expression occurs in the tumor immune microenvironment and the immune response of other cell types. A recent study found that blocking CD47-SIRPα signaling enhances antitumor immune responses (89). Tumor DNA in DCs activates the cell membrane DNA sensor cGMP-AMP (cGAMP) synthase (cGAS), which subsequently exerts potent antitumor effects by binding to the second messenger cGAMP and activating interferon gene stimulating factor (STING) (89). By contrast, CD47 inhibits this signaling pathway and aids in the immune escape of tumor cells. For instance, in treating glioblastoma, blocking CD47 not only enhances DC phagocytosis but also promotes the initiation of the adaptive immune response by T cells by activating the cGAS-cGAMP-STING signaling pathway (90). In addition, a study by Xu et al (91) found that in mouse models of colon cancer, lymphoma and melanoma, blocking CD47-SIRPα signaling activates NADPH oxidase in DCs to inhibit the degradation of tumor-derived mitochondrial DNA (mtDNA), which leads to an increase in the level of mtDNA and its recognition by cGAS in the cytoplasm of DCs. As a result, the cGAS-cGAMP-STING signaling pathway is activated, which releases interferon-γ to initiate the CD8+ T-cell mediated adaptive immune response, thereby killing tumor cells (47); i.e., the tumor-killing effect of T cells is dependent on the blockade of the CD47-activated cGAS-cGAMP-STING signaling pathway (4) (Fig. 3).

In addition, blocking CD47 induces tumor cell death only when endogenous activation signals are present (64). A study by Chen et al (92) revealed the presence of an endogenous activation signal on the surface of tumor cells called SLAM family member 7 (SLAMF7), a prophagocytic signal of SLAMF7, which is a prophagocytic ‘eat-me’ signal and usually interacts with the macrophage-1 antigen, promoting the phagocytosis of tumor cells by macrophages. Furthermore, they contended that CD47-mediated phagocytosis requires SLAMF7. However, He et al (93) refuted this view by finding that phagocytosis was also effectively induced in SLAMF7-negative diffuse large B-cell lymphomas cells after they blocked CD47 by the CD47 antibody Inhibrix. Further studies are needed to determine whether SLAMF7 is required to mediate CD47.

Therapeutic strategies targeting CD47

Clinical research has shown that CD47 is an intrinsic immune checkpoint with high clinical development value and promising application prospects. Numerous domestic and foreign companies are actively developing drugs targeting CD47, particularly mAbs, BsAbs, fusion proteins and small-molecule antibodies, and many of them have already entered the clinical research stage. A search of the PubMed database (https://pubmed.ncbi.nlm.nih.gov/) and online open resources from the US National Clinical Trials Registry system (www.clinical trials.gov) was performed as part of the present review. Compared with previously published reviews (48,94,95), not only the names, structures and clinical trials of various CD47-targeted representative drugs were summarized, but the current state of clinical research and the results of clinical trials were also outlined (Table I).

Table I.

Summary of representative CD47-targeted drugs in clinical trials.

Table I.

Summary of representative CD47-targeted drugs in clinical trials.

Drug nameFormatTargetForms of treatmentIndicationsPhaseStart dateStatusNCT no.
Magrolimab (Hu5F9-G4)CD47 mAbCD47MonotherapySolid tumorINovember 2, 2016CompletedNCT02953782
MonotherapyAML, MDSINovember 2015CompletedNCT02678338
ObinutuzumabFollicularIDecember 16, 2021Active, not recruitingNCT04599634
lymphoma
AvelumabOvarian cancerIMay 23, 2018CompletedNCT03558139
RituximabR/R B-NHLI/IINovember 21, 2016Active, not recruitingNCT02953509
Lemzoparlimab (TIJC4)CD47 mAbCD47Pembrolizumab,Solid tumor/IApril 16, 2019CompletedNCT03934814
Rituximablymphoma
MonotherapyAML, MDSI/IIMarch 25, 2020CompletedNCT04202003
ToripalimabR/R advancedI/IIDecember 30, 2021TerminatedNCT05148533
solid tumor
Dexamethasone,Multiple myelomaIJanuary 17, 2022TerminatedNCT04895410
Pomalidomide,
Daratumumab
Ligufalimab (AK117)CD47 mAbCD47MonotherapyMalignantIApril 23, 2020CompletedNCT04349969
neoplasms
AzacitidineAMLI/IIAugust 13, 2021RecruitingNCT04980885
AzacitidineMDSI/IIJune 18, 2021RecruitingNCT04900350
AO-176CD47 mAbCD47Monotherapy, Paclitaxel,Solid tumorI/IIFebruary 4, 2019CompletedNCT03834948
Pembrolizumab
Dexamethasone,R/R multipleI/IINovember 30, 2020CompletedNCT04445701
Bortezomibmyeloma
CC-90002CD47 mAbCD47MonotherapyAML, MDSIMarch 1, 2016TerminatedNCT02641002
RituximabHematologicalIMarch 12, 2015CompletedNCT02367196
cancer
TII-621SIRPa-FcCD47Monotherapy, RituximabHematologicIJanuary 28, 2016TerminatedNCT02663518
mAb and Nivolumabmalignancies
Monotherapy,R/R solid tumorsISeptember 2016TerminatedNCT02890368
PD-1/PD-L1 inhibitor
DaratumumabMultiple myelomaIOctober 28, 2021Active, not recruitingNCT05139225
Hyaluronidase-fihj
TII-622SIRPa-Fc mAbCD47Monotherapy,HematologicalIJune 7, 2018Active, not recruitingNCT03530683
Rituximab, PD-1malignancies
inhibitor, proteasome-
inhibitor regimen
Evorpacept (ALX-148)SIRPa-D1SIRPaMonotherapy,Solid tumorIFebruary 3, 2017Active, not recruitingNCT03013218
mAb Pembrolizumab,
Trastuzumab, Rituximab
Lenalidomide,Indolent andI/IIOctober 13, 2021RecruitingNCT05025800
Rituximabaggressive
B-NHL
AzacitidineHigher-risk MDSI/IIFebruary 2020Active, not recruitingNCT04417517
Venetoclax, AzacitidineAMLI/IIMay 5, 2021Active, not recruitingNCT04755244
IMM-306BsAbCD47,RituximabB-cell NHLIJanuary 15, 2021SuspendedNCT04746131
CD20
IBI-322BsAbCD47,AzacitidineAdvanced solidIJuly 21, 2021CompletedNCT04912466
PD-L1 tumor
HematologicIMay 7, 2021RecruitingNCT04795128
malignancy
Myeloid tumorIDecember 28, 2021TerminatedNCT05148442
HX009BsAbCD47, Advanced solidIJune 12, 2019CompletedNCT04097769
PD-L1 tumor
R/R lymphomaI/IIDecember 31, 2021UnknownNCT05189093
NI1701BsAbCD47,RituximabB-cell lymphomaIMarch 5, 2019Active, not recruitingNCT03804996
CD19
SL-172154BsAbSIRPa- Ovarian cancerIJune 29, 2020CompletedNCT04406623
Fc,
CD40L

[i] NCT, US National Clinical Trials Registry (www.clinical trials.gov); mAb, monoclonal antibody; CD47, cluster of differentiation 47; SIRPa-Fc, signal regulatory protein α fusion proteins; BsAb, bispecific antibodies; AML, acute myeloid leukemia; MDS, myelodysplastic syndromes; PD-1, programmed cell death 1; PD-L1, PD-1 ligand 1; R/R, relapsed/refractory; B-NHL, B-cell non-Hodgkin's lymphoma.

mAbs targeting CD47 or SIRPα

Immunotherapies targeting CD47 can be divided into two categories: First, blocking or inhibiting the ‘do not eat me’ signal with SIRPα via antibodies to promote the phagocytosis of tumor cells by macrophages (96); second, the activation of innate and adaptive immune responses. Tumor cells are recognized, taken up by antigen-presenting cells (APCs) and delivered to the initial T cells, activating T cells. T cells activate when APCs identify, pick up and transfer tumor cells to initial T cells. Antibodies targeting CD47 can kill tumor cells by inhibiting protein kinase A (97,98).

Closure of CD47 on tumor cells using mAbs targeting CD47 or soluble SIRPα-Fc structures triggers macrophage antibody-dependent cellular phagocytosis in vitro. It significantly promotes the killing of tumor cells (99). In addition, the CD47-targeted fusion protein SIRPαD1-Fc was found to inhibit the Akt/mTOR signaling pathway, upregulate reactive oxygen species production and promote autophagy in non-small cell lung cancer cells, thereby enhancing the antitumor effect (100).

More than 10 antibodies targeting CD47 have entered clinical trials (Table I), among which Magrolimab (Hu5F9-G4) was the first CD47 antibody to enter clinical trials and is already in clinical trials for various types of cancers, including AML, myelodysplastic syndromes (MDS) and solid tumors (45,101,102).

Furthermore, CD47-targeting antibodies can synergize with various mAbs, and combining the two can provide a better antitumor effect. Commonly used combinations include combination therapy with other therapeutic antibodies, chemotherapy or radiation therapy. A phase I clinical study revealed that the combination of a CD47 mAb and Rituximab resulted in an objective response rate (ORR) of 40% and a complete response rate (CRR) of 33% in patients with diffuse large B-cell lymphoma, with an ORR of 71% and a CRR of 43% in patients with follicular lymphoma (45).

In addition, several clinical trials have evaluated the safety and efficacy of CD47-targeted drugs in different stages and types of tumors. For instance, Lemzoparlimab (TJC4), which targets CD47, was screened using a phage display system. A phase I clinical trial is evaluating the efficacy effects of TJC4 alone or in combination with Pembrolizumab or Rituximab in the treatment of relapsed or refractory (R/R) advanced solid tumors and lymphomas (103). The results of the preclinical study demonstrated a favorable safety profile and clinical efficacy in five patients with AML and high-risk MDS who had received at least two treatments. Of particular note, one patient with R/R AML achieved a morphologic leukemia-free status after treatment with TJC4 (104). Humanized CD47 antibody Ligufalimab (AK117) is an anti-CD47 mAb with a unique structure, which not only has anti-tumor effects but also eliminates erythrocyte agglutination and significantly reduces phagocytosis of erythrocytes by macrophages. Phase I trials have been completed in Australia and phase II trials are underway in China and Australia. Results from a clinical trial enrolling 15 patients with advanced solid tumors showed that AK117 was safe and well tolerated, with no infusion- or treatment-related adverse effects observed (105). AO-176, a mAb targeting CD47, is being evaluated in a phase I clinical trial for treating R/R multiple myeloma (106,107). AO-176 binds preferentially to tumor cells (rather than normal cells), can bind tumor cells more efficiently in an acidic microenvironment and can kill tumor cells directly in a cell-autonomous manner (108). Current clinical data show that of the 27 patients treated with AO-176, one patient with endometrial cancer did not respond to its treatment regimen and seven patients had the best response of stable disease (SD) (109). CC-90002 is the first generation of humanized CD47 antibody to enter clinical studies that block CD47-SIRPα binding to achieve the killing of hematological tumor cells (96). Clinical trials for AML and MDS revealed that CC-90002 had poor efficacy and safety, which led to its forced discontinuation. Researchers restarted clinical trials after improving the CC-90002 treatment regimen and safety (110,111). In a mouse transplantation tumor model of multiple myeloma, CC-90002 showed significant dose-dependent antitumor activity. In addition, in non-primate animals, CC-90002 exhibited favorable pharmacokinetic properties and toxicity (96). TTI-621 and TTI-622 are SIRPs-Fc fusion proteins that have been used in the treatment of hematologic malignancies, solid tumors and mycosis fungoides (112), and such agents are currently being evaluated in a phase I clinical trial for R/R B-cell lymphomas (113). In 164 patients with B-cell non-Hodgkin's lymphoma (B-NHL), TT-621 plus rituximab was used to treat the disease in a phase I trial. The study showed that TT1-621 was well tolerated and that monotherapy is a promising therapeutic option. The ORR for all patients treated with TTI-621 monotherapy was 13%, while it was 29% for diffuse large B-cell lymphoma and 25% for T-cell NHL (113). Clinical studies of TTI-622 in patients with advanced R/R lymphomas showed that one patient with non-growth center B cells who had received five prior therapies achieved partial remission (PR) at week 8 and overall response at week 36 (114). The SIRPs-Fc fusion protein Evorpacept (ALX148) is presently undergoing evaluation in several programs (95), such as a phase I/II trial for patients with advanced solid tumors and a phase I trial for patients with aggressive and indolent NHL (115). PR rates were 22% with trastuzumab combination therapy in patients with Her2-positive gastric cancer and 16% with pembrolizumab combination therapy in patients with head and neck squamous cell carcinoma (116).

BsAbs

BsAbs are genetically engineered artificial antibodies that contain two specific antigen-binding sites. The BsAb backbone has two binding arms, one blocking the CD47-SIRPα pathway and the other binding tumor-specific antigens, thus ensuring the killing of tumor cells by BsAbs (99). Compared with combination therapy, using BsAbs also reduces the cost of drug development and clinical trials.

Several CD47-related BsAbs are in early clinical trials. For instance, IMM0306, a BsAbs targeting CD20 and CD47, avoids binding to CD47 in normal cells due to its high affinity for CD20, thus reducing the toxicity associated with the CD47 target. IMM0306 has demonstrated vigorous antitumor activity in a mouse model of human NHL transplantation tumor (117). It is currently being evaluated in a phase I clinical trial in B-NHL (118). IBI322 is a drug that inhibits both the programmed cell death 1 (PD-1)/PD-1 ligand 1 and CD47-SIRPα signaling pathways for treating intermediate to advanced malignancies. Repeated weekly injections of IBI322 showed good tolerability in non-human primates (119). IBI322 is currently being evaluated in a phase I clinical trial for advanced malignancies. HX009 is a BsAb targeting PD-1 and CD47 for treating advanced tumors such as gastric, colorectal and hepatocellular carcinomas and is currently being evaluated in a phase I trial for advanced solid tumors (120). Clinical studies demonstrated that of the 18 patients with at least one post-baseline tumor assessment, three patients achieved a PR and six achieved SD (121). CC-96673, a humanized BsAbs co-targeting CD47 and CD20, was able to efficiently promote phagocytosis by macrophages by blocking CD47-SIRPα interactions and mediated the selective removal of CD20-expressing tumor cells by ADCC and CDC to selectively clear CD20-expressing tumor cells. A phase I clinical trial is presently assessing it for R/R NHL. NI-1701 is a novel BsAb constructed using spinopore technology to target CD47 and CD19 (122). Previous studies have found that NI-1701 selectively binds to CD47 and CD19 co-expressing cells and has poor binding ability with normal cells by interacting poorly with normal cells, avoiding binding to normal cells and thus improving biosafety (121,123). SL-172154, a fusion protein targeting SIRPs-Fc and CD40L, is being evaluated in a phase I clinical trial for solid tumors (124).

Other treatment strategies

Chimeric antigen receptor T cell (CAR T cell) immunotherapy has made significant progress in oncology, and combining CD47 blockade therapy with CAR T-cell therapy has become a hot research topic. A previous review (125) described CAR T cells and their future prospects and directions in detail; however, there is a lack of description of the role of CD47-CAR T cells in various types of tumor. CAR T-cell therapy is a cell-over-cell immunotherapy that does not depend on major histocompatibility complex (126). Beckett et al (127) examined the role of CD47 in CAR T-cell function by knocking down CD47 in T cells for downstream functional analysis. They showed that CD47 expression is critical for CAR T-cell survival in vivo and is required for successful overt T-cell therapy. Golubovskaya et al (128) reported that CD47 CAR T cells had antitumor activity and significantly inhibited the growth of transplanted pancreatic cancer tumors. Shu et al (129) constructed a CAR T cell targeting both CD47 and tumor-associated glycoprotein 72 (TAG-72), which showed vigorous antitumor activity in both in vitro and in vivo models of ovarian cancer. The specific targeting of TAG-72 could reduce its killing of normal cells. Chen et al (130) developed a SIRPα-Fc fusion protein CAR T cell, which promoted the phagocytosis of macrophages, recruited more DCs into tumor tissues, inhibited the apoptosis of CAR T cells themselves and reduced the expression of PD-1 on the surface of CAR T cells, thus enhancing the antitumor effect.

The understanding of chimeric antigen receptor macrophages (CAR-Ms) is minimal. CAR-Ms is the engineering of macrophages to modify CARs in order to enhance macrophage antigen-specific phagocytosis and tumor clearance (131,132). Klichinsky et al (133) first proposed the CAR-M concept, constructed CAR-Ms and reported that CAR-Ms have strong antitumor effects and can promote the secretion of proinflammatory factors, promote M2-type to M1-type polarization and increase T-lymphocyte antigen presentation (134).

In addition, a new therapeutic strategy for targeting CD47 has emerged in recent years, namely reprogramming the immunogenicity of cancer cells, whereby specific chemotherapeutic agents or radiation therapy stimulate tumor cells to undergo tumor immunogenic cell death (ICD), which is a form of apoptosis that activates the immune system (9). Abdel-Bar et al (135) developed nucleic acid lipid particles for the delivery of ICD-inducing Adriamycin and CD47 proteins, which could enhance phagocytosis by macrophages by increasing the amount of cell surface calreticulin.

Challenges of antitumor therapy targeting CD47

Due to its high expression on the surface of tumor cells, CD47 has become an ideal target for tumor immunotherapy, and antitumor drugs targeting CD47 were shown to have promising applications. However, chemotherapeutic drugs targeting CD47 have numerous adverse effects, a limitation that makes targeted CD47 therapy a significant challenge. First, CD47 is widely expressed on the surface of tumor cells and normal cells, leading to inevitable injury to normal red blood cells in the process of killing tumor cells. Many red blood cells will become the best ‘cover’ for tumor cells, and red blood cells will be exhausted by targeted drugs before tumor cells, resulting in adverse effects such as red blood-cell aggregation, anemia and thrombocytopenia (100,136). The degree of toxicity is dose-, time- and patient-specific and can be reduced by optimizing the dosage and combining drugs with erythropoietin. Second, there may be differences in the level of CD47 expression on the surface of different tumor cells, resulting in different sensitivities to targeted CD47 therapy (137,138). Finally, due to the presence of multiple immunosuppressive cells in the human body, such as myeloid-derived suppressor cells, tumor-associated macrophages and tumor-associated DCs, tumor cells may evade the surveillance of immune cells by upregulating the expression other immune checkpoint molecules (139141), thus altering the therapeutic efficacy of targeting CD47 (6).

Challenges in immunotherapy targeting CD47 have led to the proposal of new therapeutic regimens to improve the effectiveness of treatment. One such approach is to combine CD47-targeting drugs with other immune checkpoint inhibitors to reduce immune escape by tumor cells (142,143). Furthermore, the development of BsAbs has provided new ideas for achieving improved specificity of targeted therapy (144,145). In addition, solutions to modulate the TME to enhance the efficacy of CD47-targeted therapies are also being explored (141,146,147). These solutions are expected to improve the efficacy of CD47-targeted therapies and reduce resistance.

Conclusion and prospects

In recent years, an increasing number of studies on CD47 have been conducted, and this topic has become a significant hotspot in various research fields. CD47 binds to SIRPα to activate a signaling pathway that regulates DC activation and antigen presentation in both directions and regulates macrophage phagocytosis during erythrocyte and HSC transplantation. Upregulating or downregulating the expression of CD47 has an essential regulatory role in tumor-cell growth or death, and blocking CD47 expression also initiates an adaptive immune response that kills tumor cells (148). Although the combination of targeted CD47-SIRPα axis blockade therapy with other antibody drugs or therapies has shown good antitumor efficacy, CD47 is widely expressed in erythrocytes, myeloid cells and other hematopoietic cells, and anemia remains the most significant challenge associated with CD47-targeted drug therapy (149); furthermore, relevant antibody drugs have shown good efficacy. These drugs effectively attenuate the adverse effects of CD47-SIRPα blockade and significantly improve safety (117,143). However, much progress is needed before immunotherapy targeting the CD47-SIRPα axis can be applied in the clinic. To date, numerous clinical studies have shown that metabolic reprogramming has an essential role in the regulation of macrophage activation and study of the regulation of phagocytosis by the CD47-SIRPα axis from the point of view of metabolic reprogramming will be a promising direction; however, the underlying mechanisms of metabolism during phagocytosis, which are associated with the CD47-SIRPα axis, remain elusive (9). Further scientific research will clarify the mechanisms of action.

Acknowledgements

Not applicable.

Funding

This work was supported by the Natural Science Research Project of the Anhui Educational Committee (grant no. KJ2020ZD49) and the 512 Talent Cultivation Program of Bengbu Medical College (grant no. by51201103).

Availability of data and materials

Not applicable.

Authors' contributions

FW was involved in writing of the original draft and searching the literature. HP, FL and MH performed the literature search and reviewed the draft. JT and CS were involved in supervision, writing and editing. All of the authors discussed the article, and have read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Chao MP, Weissman IL and Majeti R: The CD47-SIRPα pathway in cancer immune evasion and potential therapeutic implications. Curr Opin Immunol. 24:225–232. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Khandelwal S, van Rooijen N and Saxena RK: Reduced expression of CD47 during murine red blood cell (RBC) senescence and its role in RBC clearance from the circulation. Transfusion. 47:1725–1732. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Matlung HL, Szilagyi K, Barclay NA and van den Berg TK: The CD47-SIRPα signaling axis as an innate immune checkpoint in cancer. Immunol Rev. 276:145–164. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Jia X, Yan B, Tian X, Liu Q, Jin J, Shi J and Hou Y: CD47/SIRPα pathway mediates cancer immune escape and immunotherapy. Int J Biol Sci. 17:3281–3287. 2021. View Article : Google Scholar : PubMed/NCBI

5 

Lin F, Xiong M, Hao W, Song Y, Liu R, Yang Y, Yuan X, Fan D, Zhang Y, Hao M, et al: A novel blockade CD47 antibody with therapeutic potential for cancer. Front Oncol. 10:6155342020. View Article : Google Scholar : PubMed/NCBI

6 

Chen Q, Guo X and Ma W: Opportunities and challenges of CD47-targeted therapy in cancer immunotherapy. Oncol Res. 32:49–60. 2023. View Article : Google Scholar : PubMed/NCBI

7 

Liu J, Meng Z, Xu T, Kuerban K, Wang S, Zhang X, Fan J, Ju D, Tian W, Huang X, et al: A SIRPαFc fusion protein conjugated with the Collagen-Binding domain for targeted immunotherapy of non-small cell lung cancer. Front Immunol. 13:8452172022. View Article : Google Scholar : PubMed/NCBI

8 

Ozaniak A, Smetanova J, Bartolini R, Rataj M, Capkova L, Hacek J, Fialova M, Krupickova L, Striz I, Lischke R, et al: A novel anti-CD47-targeted blockade promotes immune activation in human soft tissue sarcoma but does not potentiate anti-PD-1 blockade. J Cancer Res Clin Oncol. 149:3789–3801. 2023. View Article : Google Scholar : PubMed/NCBI

9 

Hao Y, Zhou X, Li Y, Li B and Cheng L: The CD47-SIRPα axis is a promising target for cancer immunotherapies. Int Immunopharmacol. 120:1102552023. View Article : Google Scholar : PubMed/NCBI

10 

Brown E, Hooper L, Ho T and Gresham H: Integrin-associated protein: A 50-kD plasma membrane antigen physically and functionally associated with integrins. J Cell Biol. 111:2785–2794. 1990. View Article : Google Scholar : PubMed/NCBI

11 

Lindberg FP, Bullard DC, Caver TE, Gresham HD, Beaudet AL and Brown EJ: Decreased resistance to bacterial infection and granulocyte defects in IAP-deficient mice. Science. 274:795–798. 1996. View Article : Google Scholar : PubMed/NCBI

12 

van Helden MJ, Zwarthoff SA, Arends RJ, Reinieren-Beeren IMJ, Paradé MCBC, Driessen-Engels L, de Laat-Arts K, Damming D, Santegoeds-Lenssen EWH, van Kuppeveld DWJ, et al: BYON4228 is a pan-allelic antagonistic SIRPα antibody that potentiates destruction of antibody-opsonized tumor cells and lacks binding to SIRPγ on T cells. J Immunother Cancer. 11:e0065672023. View Article : Google Scholar : PubMed/NCBI

13 

Navarro-Alvarez N and Yang YG: CD47: A new player in phagocytosis and xenograft rejection. Cell Mol Immunol. 8:285–288. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Deng H, Wang G, Zhao S, Tao Y, Zhang Z, Yang J and Lei Y: New hope for tumor immunotherapy: The macrophage-related ‘do not eat me’ signaling pathway. Front Pharmacol. 14:12289622023. View Article : Google Scholar : PubMed/NCBI

15 

Hatherley D, Graham SC, Turner J, Harlos K, Stuart DI and Barclay AN: Paired receptor specificity explained by structures of signal regulatory proteins alone and complexed with CD47. Mol Cell. 31:266–277. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Hatherley D, Harlos K, Dunlop DC, Stuart DI and Barclay AN: The structure of the macrophage signal regulatory protein alpha (SIRPalpha) inhibitory receptor reveals a binding face reminiscent of that used by T cell receptors. J Biol Chem. 282:14567–14575. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Lymn JS, Patel MK, Clunn GF, Rao SJ, Gallagher KL and Hughes AD: Thrombospondin-1 differentially induces chemotaxis and DNA synthesis of human venous smooth muscle cells at the receptor-binding level. J Cell Sci. 115:4353–4360. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Chung J, Gao AG and Frazier WA: Thrombspondin acts via integrin-associated protein to activate the platelet integrin alphaIIbbeta3. J Biol Chem. 272:14740–14746. 1997. View Article : Google Scholar : PubMed/NCBI

19 

Hayat SMG, Bianconi V, Pirro M, Jaafari MR, Hatamipour M and Sahebkar A: CD47: Role in the immune system and application to cancer therapy. Cell Oncol (Dordr). 43:19–30. 2020. View Article : Google Scholar : PubMed/NCBI

20 

Zhang T, Wang F, Xu L and Yang YG: Structural-functional diversity of CD47 proteoforms. Front Immunol. 15:13295622024. View Article : Google Scholar : PubMed/NCBI

21 

Sadallah S, Eken C, Martin PJ and Schifferli JA: Microparticles (ectosomes) shed by stored human platelets downregulate macrophages and modify the development of dendritic cells. J Immunol. 186:6543–6552. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Aversa R, Sorrentino A, Esposito R, Ambrosio MR, Amato A, Zambelli A, Ciccodicola A, D'Apice L and Costa V: Alternative splicing in adhesion- and motility-related genes in breast cancer. Int J Mol Sci. 17:1212016. View Article : Google Scholar : PubMed/NCBI

23 

Reinhold MI, Lindberg FP, Plas D, Reynolds S, Peters MG and Brown EJ: In vivo expression of alternatively spliced forms of integrin-associated protein (CD47). J Cell Sci. 108:3419–3425. 1995. View Article : Google Scholar : PubMed/NCBI

24 

Barclay AN and Van den Berg TK: The interaction between signal regulatory protein alpha (SIRPα) and CD47: Structure, function, and therapeutic target. Annu Rev Immunol. 32:25–50. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Lee EH, Hsieh YP, Yang CL, Tsai KJ and Liu CH: Induction of integrin-associated protein (IAP) mRNA expression during memory consolidation in rat hippocampus. Eur J Neurosci. 12:1105–1112. 2000. View Article : Google Scholar : PubMed/NCBI

26 

Ratnikova NM, Lezhnin YN, Frolova EI, Kravchenko JE and Chumakov SP: CD47 receptor as a primary target for cancer therapy. Mol Biol (Mosk). 51:251–261. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Frazier WA, Gao AG, Dimitry J, Chung J, Brown EJ, Lindberg FP and Linder ME: The thrombospondin receptor integrin-associated protein (CD47) functionally couples to heterotrimeric Gi. J Biol Chem. 274:8554–8560. 1999. View Article : Google Scholar : PubMed/NCBI

28 

N'Diaye EN and Brown EJ: The ubiquitin-related protein PLIC-1 regulates heterotrimeric G protein function through association with Gbetagamma. J Cell Biol. 163:1157–1165. 2003. View Article : Google Scholar : PubMed/NCBI

29 

Sick E, Boukhari A, Deramaudt T, Rondé P, Bucher B, André P, Gies JP and Takeda K: Activation of CD47 receptors causes proliferation of human astrocytoma but not normal astrocytes via an Akt-dependent pathway. Glia. 59:308–319. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Mateo V, Brown EJ, Biron G, Rubio M, Fischer A, Deist FL and Sarfati M: Mechanisms of CD47-induced caspase-independent cell death in normal and leukemic cells: Link between phosphatidylserine exposure and cytoskeleton organization. Blood. 100:2882–2890. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Soto-Pantoja DR, Kaur S and Roberts DD: CD47 signaling pathways controlling cellular differentiation and responses to stress. Crit Rev Biochem Mol Biol. 50:212–230. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Brown EJ and Frazier WA: Integrin-associated protein (CD47) and its ligands. Trends Cell Biol. 11:130–135. 2001. View Article : Google Scholar : PubMed/NCBI

33 

Murata Y, Saito Y, Kotani T and Matozaki T: Blockade of CD47 or SIRPα: A new cancer immunotherapy. Expert Opin Ther Targets. 24:945–951. 2020. View Article : Google Scholar : PubMed/NCBI

34 

Manna PP and Frazier WA: The mechanism of CD47-dependent killing of T cells: Heterotrimeric Gi-dependent inhibition of protein kinase A. J Immunol. 170:3544–3553. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Lindberg FP, Gresham HD, Reinhold MI and Brown EJ: Integrin-associated protein immunoglobulin domain is necessary for efficient vitronectin bead binding. J Cell Biol. 134:1313–1322. 1996. View Article : Google Scholar : PubMed/NCBI

36 

Brittain JE, Han J, Ataga KI, Orringer EP and Parise LV: Mechanism of CD47-induced alpha4beta1 integrin activation and adhesion in sickle reticulocytes. J Biol Chem. 279:42393–42402. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Orazizadeh M, Lee HS, Groenendijk B, Sadler SJ, Wright MO, Lindberg FP and Salter DM: CD47 associates with alpha 5 integrin and regulates responses of human articular chondrocytes to mechanical stimulation in an in vitro model. Arthritis Res Ther. 10:R42008. View Article : Google Scholar : PubMed/NCBI

38 

Koenigsknecht J and Landreth G: Microglial phagocytosis of fibrillar beta-amyloid through a beta1 integrin-dependent mechanism. J Neurosci. 24:9838–9846. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Zhang K, Li M, Yin L, Fu G and Liu Z: Role of thrombospondin-1 and thrombospondin-2 in cardiovascular diseases (Review). Int J Mol Med. 45:1275–1293. 2020.PubMed/NCBI

40 

Adams JC and Lawler J: The thrombospondins. Cold Spring Harb Perspect Biol. 3:a0097122011. View Article : Google Scholar : PubMed/NCBI

41 

Leclair P and Lim CJ: CD47-independent effects mediated by the TSP-derived 4N1K peptide. PLoS One. 9:e983582014. View Article : Google Scholar : PubMed/NCBI

42 

Isenberg JS, Romeo MJ, Yu C, Yu CK, Nghiem K, Monsale J, Rick ME, Wink DA, Frazier WA and Roberts DD: Thrombospondin-1 stimulates platelet aggregation by blocking the antithrombotic activity of nitric oxide/cGMP signaling. Blood. 111:613–623. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Jeanne A, Sarazin T, Charlé M, Moali C, Fichel C, Boulagnon-Rombi C, Callewaert M, Andry MC, Diesis E, Delolme F, et al: Targeting ovarian carcinoma with TSP-1: CD47 antagonist TAX2 activates Anti-Tumor immunity. Cancers (Basel). 13:50192021. View Article : Google Scholar : PubMed/NCBI

44 

Kharitonenkov A, Chen Z, Sures I, Wang H, Schilling J and Ullrich A: A family of proteins that inhibit signalling through tyrosine kinase receptors. Nature. 386:181–186. 1997. View Article : Google Scholar : PubMed/NCBI

45 

Advani R, Flinn I, Popplewell L, Forero A, Bartlett NL, Ghosh N, Kline J, Roschewski M, LaCasce A, Collins GP, et al: CD47 Blockade by Hu5F9-G4 and rituximab in Non-Hodgkin's lymphoma. N Engl J Med. 379:1711–1721. 2018. View Article : Google Scholar : PubMed/NCBI

46 

Barclay AN and Brown MH: The SIRP family of receptors and immune regulation. Nat Rev Immunol. 6:457–464. 2006. View Article : Google Scholar : PubMed/NCBI

47 

Feng M, Jiang W, Kim BYS, Zhang CC, Fu YX and Weissman IL: Phagocytosis checkpoints as new targets for cancer immunotherapy. Nat Rev Cancer. 19:568–586. 2019. View Article : Google Scholar : PubMed/NCBI

48 

Zhao H, Song S, Ma J, Yan Z, Xie H, Feng Y and Che S: CD47 as a promising therapeutic target in oncology. Front Immunol. 13:7574802022. View Article : Google Scholar : PubMed/NCBI

49 

Nakaishi A, Hirose M, Yoshimura M, Oneyama C, Saito K, Kuki N, Matsuda M, Honma N, Ohnishi H, Matozaki T, et al: Structural insight into the specific interaction between murine SHPS-1/SIRP alpha and its ligand CD47. J Mol Biol. 375:650–660. 2008. View Article : Google Scholar : PubMed/NCBI

50 

Vernon-Wilson EF, Kee WJ, Willis AC, Barclay AN, Simmons DL and Brown MH: CD47 is a ligand for rat macrophage membrane signal regulatory protein SIRP (OX41) and human SIRPalpha 1. Eur J Immunol. 30:2130–2137. 2000. View Article : Google Scholar : PubMed/NCBI

51 

Han X, Sterling H, Chen Y, Saginario C, Brown EJ, Frazier WA, Lindberg FP and Vignery A: CD47, a ligand for the macrophage fusion receptor, participates in macrophage multinucleation. J Biol Chem. 275:37984–37992. 2000. View Article : Google Scholar : PubMed/NCBI

52 

Rebres RA, Vaz LE, Green JM and Brown EJ: Normal ligand binding and signaling by CD47 (integrin-associated protein) requires a long range disulfide bond between the extracellular and membrane-spanning domains. J Biol Chem. 276:34607–34616. 2001. View Article : Google Scholar : PubMed/NCBI

53 

Hatherley D, Graham SC, Harlos K, Stuart DI and Barclay AN: Structure of signal-regulatory protein alpha: A link to antigen receptor evolution. J Biol Chem. 284:26613–26619. 2009. View Article : Google Scholar : PubMed/NCBI

54 

Takada T, Matozaki T, Takeda H, Fukunaga K, Noguchi T, Fujioka Y, Okazaki I, Tsuda M, Yamao T, Ochi F and Kasuga M: Roles of the complex formation of SHPS-1 with SHP-2 in insulin-stimulated mitogen-activated protein kinase activation. J Biol Chem. 273:9234–9242. 1998. View Article : Google Scholar : PubMed/NCBI

55 

Tsai RK and Discher DE: Inhibition of ‘self’ engulfment through deactivation of myosin-II at the phagocytic synapse between human cells. J Cell Biol. 180:989–1003. 2008. View Article : Google Scholar : PubMed/NCBI

56 

Sato-Hashimoto M, Saito Y, Ohnishi H, Iwamura H, Kanazawa Y, Kaneko T, Kusakari S, Kotani T, Mori M, Murata Y, et al: Signal regulatory protein α regulates the homeostasis of T lymphocytes in the spleen. J Immunol. 187:291–297. 2011. View Article : Google Scholar : PubMed/NCBI

57 

Latour S, Tanaka H, Demeure C, Mateo V, Rubio M, Brown EJ, Maliszewski C, Lindberg FP, Oldenborg A, Ullrich A, et al: Bidirectional negative regulation of human T and dendritic cells by CD47 and its cognate receptor signal-regulator protein-alpha: Down-regulation of IL-12 responsiveness and inhibition of dendritic cell activation. J Immunol. 67:2547–2554. 2001. View Article : Google Scholar

58 

Saito Y, Iwamura H, Kaneko T, Ohnishi H, Murata Y, Okazawa H, Kanazawa Y, Sato-Hashimoto M, Kobayashi H, Oldenborg PA, et al: Regulation by SIRPα of dendritic cell homeostasis in lymphoid tissues. Blood. 116:3517–3525. 2010. View Article : Google Scholar : PubMed/NCBI

59 

Maile LA, DeMambro VE, Wai C, Lotinun S, Aday AW, Capps BE, Beamer WG, Rosen CJ and Clemmons DR: An essential role for the association of CD47 to SHPS-1 in skeletal remodeling. J Bone Miner Res. 26:2068–2081. 2011. View Article : Google Scholar : PubMed/NCBI

60 

Oldenborg PA, Zheleznyak A, Fang YF, Lagenaur CF, Gresham HD and Lindberg FP: Role of CD47 as a marker of self on red blood cells. Science. 288:2051–2054. 2000. View Article : Google Scholar : PubMed/NCBI

61 

Clevers H, Loh KM and Nusse R: Stem cell signaling. An integral program for tissue renewal and regeneration: Wnt signaling and stem cell control. Science. 346:12480122014. View Article : Google Scholar : PubMed/NCBI

62 

Murata Y, Kotani T, Ohnishi H and Matozaki T: The CD47-SIRPα signalling system: Its physiological roles and therapeutic application. J Biochem. 155:335–344. 2014. View Article : Google Scholar : PubMed/NCBI

63 

Ferrari D, Gorini S, Callegari G and la Sala A: Shaping immune responses through the activation of dendritic cells' P2 receptors. Purinergic Signal. 3:99–107. 2007. View Article : Google Scholar : PubMed/NCBI

64 

Logtenberg MEW, Scheeren FA and Schumacher TN: The CD47-SIRPα Immune Checkpoint. Immunity. 52:742–752. 2020. View Article : Google Scholar : PubMed/NCBI

65 

Okazawa H, Motegi S, Ohyama N, Ohnishi H, Tomizawa T, Kaneko Y, Oldenborg PA, Ishikawa O and Matozaki T: Negative regulation of phagocytosis in macrophages by the CD47-SHPS-1 system. J Immunol. 174:2004–2011. 2005. View Article : Google Scholar : PubMed/NCBI

66 

Ishikawa-Sekigami T, Kaneko Y, Okazawa H, Tomizawa T, Okajo J, Saito Y, Okuzawa C, Sugawara-Yokoo M, Nishiyama U, Ohnishi H, et al: SHPS-1 promotes the survival of circulating erythrocytes through inhibition of phagocytosis by splenic macrophages. Blood. 107:341–348. 2006. View Article : Google Scholar : PubMed/NCBI

67 

Yamao T, Noguchi T, Takeuchi O, Nishiyama U, Morita H, Hagiwara T, Akahori H, Kato T, Inagaki K, Okazawa H, et al: Negative regulation of platelet clearance and of the macrophage phagocytic response by the transmembrane glycoprotein SHPS-1. J Biol Chem. 277:39833–39839. 2002. View Article : Google Scholar : PubMed/NCBI

68 

Wang C, Wang H, Ide K, Wang Y, Van Rooijen N, Ohdan H and Yang YG: Human CD47 expression permits survival of porcine cells in immunodeficient mice that express SIRPα capable of binding to human CD47. Cell Transplant. 20:1915–1920. 2011. View Article : Google Scholar : PubMed/NCBI

69 

Griesemer A, Yamada K and Sykes M: Xenotransplantation: Immunological hurdles and progress toward tolerance. Immunol Rev. 258:241–258. 2014. View Article : Google Scholar : PubMed/NCBI

70 

Takenaka K, Prasolava TK, Wang JC, Mortin-Toth SM, Khalouei S, Gan OI, Dick JE and Danska JS: Polymorphism in Sirpa modulates engraftment of human hematopoietic stem cells. Nat Immunol. 8:1313–1323. 2007. View Article : Google Scholar : PubMed/NCBI

71 

Kwong LS, Brown MH, Barclay AN and Hatherley D: Signal-regulatory protein α from the NOD mouse binds human CD47 with an exceptionally high affinity-implications for engraftment of human cells. Immunology. 143:61–67. 2014. View Article : Google Scholar : PubMed/NCBI

72 

Theocharides AP, Jin L, Cheng PY, Prasolava TK, Malko AV, Ho JM, Poeppl AG, van Rooijen N, Minden MD, Danska JS, et al: Disruption of SIRPα signaling in macrophages eliminates human acute myeloid leukemia stem cells in xenografts. J Exp Med. 209:1883–1899. 2012. View Article : Google Scholar : PubMed/NCBI

73 

Rodriguez PL, Harada T, Christian DA, Pantano DA, Tsai RK and Discher DE: Minimal ‘Self’ peptides that inhibit phagocytic clearance and enhance delivery of nanoparticles. Science. 339:971–975. 2013. View Article : Google Scholar : PubMed/NCBI

74 

Iwamoto C, Takenaka K, Urata S, Yamauchi T, Shima T, Kuriyama T, Daitoku S, Saito Y, Miyamoto T, Iwasaki H, et al: The BALB/c-specific polymorphic SIRPA enhances its affinity for human CD47, inhibiting phagocytosis against human cells to promote xenogeneic engraftment. Exp Hematol. 42:163–171.e1. 2014. View Article : Google Scholar : PubMed/NCBI

75 

Ishikawa-Sekigami T, Kaneko Y, Saito Y, Murata Y, Okazawa H, Ohnishi H, Oldenborg PA, Nojima Y and Matozaki T: Enhanced phagocytosis of CD47-deficient red blood cells by splenic macrophages requires SHPS-1. Biochem Biophys Res Commun. 343:1197–1200. 2006. View Article : Google Scholar : PubMed/NCBI

76 

Chao MP, Alizadeh AA, Tang C, Myklebust JH, Varghese B, Gill S, Jan M, Cha AC, Chan CK, Tan BT, et al: Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma. Cell. 142:699–713. 2010. View Article : Google Scholar : PubMed/NCBI

77 

Xiao Z, Chung H, Banan B, Manning PT, Ott KC, Lin S, Capoccia BJ, Subramanian V, Hiebsch RR, Upadhya GA, et al: Antibody mediated therapy targeting CD47 inhibits tumor progression of hepatocellular carcinoma. Cancer Lett. 360:302–309. 2015. View Article : Google Scholar : PubMed/NCBI

78 

Zhang H, Lu H, Xiang L, Bullen JW, Zhang C, Samanta D, Gilkes DM, He J and Semenza GL: 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. 112:E6215–6223. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Liu L, Zhang L, Yang L, Li H, Li R, Yu J, Yang L, Wei F, Yan C, Sun Q, et al: Anti-CD47 antibody as a targeted therapeutic agent for human lung cancer and cancer stem cells. Front Immunol. 8:4042017. View Article : Google Scholar : PubMed/NCBI

80 

Russ A, Hua AB, Montfort WR, Rahman B, Riaz IB, Khalid MU, Carew JS, Nawrocki ST, Persky D and Anwer F: Blocking ‘don't eat me’ signal of CD47-SIRPα in hematological malignancies, an in-depth review. Blood Rev. 32:480–489. 2018. View Article : Google Scholar : PubMed/NCBI

81 

Yang K, Xu J, Liu Q, Li J and Xi Y: Expression and significance of CD47, PD1 and PDL1 in T-cell acute lymphoblastic lymphoma/leukemia. Pathol Res Pract. 215:265–271. 2019. View Article : Google Scholar : PubMed/NCBI

82 

Abe H, Saito R, Ichimura T, Iwasaki A, Yamazawa S, Shinozaki-Ushiku A, Morikawa T, Ushiku T, Yamashita H, Seto Y and Fukayama M: CD47 expression in Epstein-Barr virus-associated gastric carcinoma: Coexistence with tumor immunity lowering the ratio of CD8+/Foxp3+ T cells. Virchows Arch. 472:643–651. 2018. View Article : Google Scholar : PubMed/NCBI

83 

Yu L, Ding Y, Wan T, Deng T, Huang H and Liu J: Significance of CD47 and its association with tumor immune microenvironment heterogeneity in ovarian cancer. Front Immunol. 12:7681152021. View Article : Google Scholar : PubMed/NCBI

84 

Casey SC, Tong L, Li Y, Do R, Walz S, Fitzgerald KN, Gouw AM, Baylot V, Gütgemann I, Eilers M and Felsher DW: MYC regulates the antitumor immune response through CD47 and PD-L1. Science. 352:227–231. 2016. View Article : Google Scholar : PubMed/NCBI

85 

Betancur PA, Abraham BJ, Yiu YY, Willingham SB, Khameneh F, Zarnegar M, Kuo AH, McKenna K, Kojima Y, Leeper NJ, et al: A CD47-associated super-enhancer links pro-inflammatory signalling to CD47 upregulation in breast cancer. Nat Commun. 8:148022017. View Article : Google Scholar : PubMed/NCBI

86 

Suzuki S, Yokobori T, Tanaka N, Sakai M, Sano A, Inose T, Sohda M, Nakajima M, Miyazaki T, Kato H and Kuwano H: CD47 expression regulated by the miR-133a tumor suppressor is a novel prognostic marker in esophageal squamous cell carcinoma. Oncol Rep. 28:465–472. 2012. View Article : Google Scholar : PubMed/NCBI

87 

Rastgoo N, Wu J, Liu A, Pourabdollah M, Atenafu EG, Reece D, Chen W and Chang H: Targeting CD47/TNFAIP8 by miR-155 overcomes drug resistance and inhibits tumor growth through induction of phagocytosis and apoptosis in multiple myeloma. Haematologica. 105:2813–2823. 2020. View Article : Google Scholar : PubMed/NCBI

88 

Huang CY, Ye ZH, Huang MY and Lu JJ: Regulation of CD47 expression in cancer cells. Transl Oncol. 13:1008622020. View Article : Google Scholar : PubMed/NCBI

89 

Ma R, Ortiz Serrano TP, Davis J, Prigge AD and Ridge KM: The cGAS-STING pathway: The role of self-DNA sensing in inflammatory lung disease. FASEB J. 34:13156–13170. 2020. View Article : Google Scholar : PubMed/NCBI

90 

von Roemeling CA, Wang Y, Qie Y, Yuan H, Zhao H, Liu X, Yang Z, Yang M, Deng W, Bruno KA, et al: Therapeutic modulation of phagocytosis in glioblastoma can activate both innate and adaptive antitumour immunity. Nat Commun. 11:15082020. View Article : Google Scholar : PubMed/NCBI

91 

Xu MM, Pu Y, Han D, Shi Y, Cao X, Liang H, Chen X, Li XD, Deng L, Chen ZJ, et al: Dendritic cells but not macrophages sense tumor mitochondrial DNA for cross-priming through signal regulatory protein α signaling. Immunity. 47:363–373.e5. 2017. View Article : Google Scholar : PubMed/NCBI

92 

Chen J, Zhong MC, Guo H, Davidson D, Mishel S, Lu Y, Rhee I, Pérez-Quintero LA, Zhang S, Cruz-Munoz ME, et al: SLAMF7 is critical for phagocytosis of haematopoietic tumour cells via Mac-1 integrin. Nature. 544:493–497. 2017. View Article : Google Scholar : PubMed/NCBI

93 

He Y, Bouwstra R, Wiersma VR, de Jong M, Jan Lourens H, Fehrmann R, de Bruyn M, Ammatuna E, Huls G, van Meerten T and Bremer E: Cancer cell-expressed SLAMF7 is not required for CD47-mediated phagocytosis. Nat Commun. 10:5332019. View Article : Google Scholar : PubMed/NCBI

94 

Yang Y, Yang Z and Yang Y: Potential role of CD47-directed bispecific antibodies in cancer immunotherapy. Front Immunol. 12:6860312021. View Article : Google Scholar : PubMed/NCBI

95 

Zhang W, Huang Q, Xiao W, Zhao Y, Pi J, Xu H, Zhao H, Xu J, Evans CE and Jin H: Advances in anti-tumor treatments targeting the CD47/SIRPα axis. Front Immunol. 11:182020. View Article : Google Scholar : PubMed/NCBI

96 

Narla RK, Modi H, Bauer D, Abbasian M, Leisten J, Piccotti JR, Kopytek S, Eckelman BP, Deveraux Q, Timmer J, et al: Modulation of CD47-SIRPα innate immune checkpoint axis with Fc-function detuned anti-CD47 therapeutic antibody. Cancer Immunol Immunother. 71:473–489. 202 View Article : Google Scholar : PubMed/NCBI

97 

Kuo TC, Chen A, Harrabi O, Sockolosky JT, Zhang A, Sangalang E, Doyle LV, Kauder SE, Fontaine D, Bollini S, et al: Targeting the myeloid checkpoint receptor SIRPα potentiates innate and adaptive immune responses to promote anti-tumor activity. J Hematol Oncol. 13:1602020. View Article : Google Scholar : PubMed/NCBI

98 

Bian HT, Shen YW, Zhou YD, Nagle DG, Guan YY, Zhang WD and Luan X: CD47: Beyond an immune checkpoint in cancer treatment. Biochim Biophys Acta Rev Cancer. 1877:1887712022. View Article : Google Scholar : PubMed/NCBI

99 

Luo X, Shen Y, Huang W, Bao Y, Mo J, Yao L and Yuan L: Blocking CD47-SIRPα signal axis as promising immunotherapy in ovarian cancer. Cancer Control. 30:107327482311597062023. View Article : Google Scholar : PubMed/NCBI

100 

Liu Y, Wang Y, Yang Y, Weng L, Wu Q, Zhang J, Zhao P, Fang L, Shi Y and Wang P: Emerging phagocytosis checkpoints in cancer immunotherapy. Signal Transduct Target Ther. 8:1042023. View Article : Google Scholar : PubMed/NCBI

101 

Maute R, Xu J and Weissman IL: CD47-SIRPα-targeted therapeutics: Status and prospects. Immunooncol Technol. 13:1000702022. View Article : Google Scholar : PubMed/NCBI

102 

Kayser S and Levis MJ: The clinical impact of the molecular landscape of acute myeloid leukemia. Haematologica. 108:308–320. 2023. View Article : Google Scholar : PubMed/NCBI

103 

Berlin J, Harb W, Adjei A, Xing Y, Swiecicki P, Seetharam M, Nandagopal L, Gopal A, Xu C, Meng Y, et al: 385 A first-in-human study of lemzoparlimab, a differentiated anti-CD47 antibody, in subjects with relapsed/refractory malignancy: Initial monotherapy results. J Immuno Ther Res Cancer. 8 (Suppl 3):A233–A234. 2020.

104 

Qi J, Li J, Jiang B, Jiang B, Liu H, Cao X, Zhang M, Meng Y, MA X, Jia Y, et al: A Phase I/IIa study of lemzoparlimab, a monoclonal antibody targeting CD47, in patients with relapsed and/or refractory acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS): Initial phase I results. Blood. 136:30–31. 2020. View Article : Google Scholar

105 

Gan HK, Coward J, Mislang A, Cosman R, Nagrial A, Jin X, Li B, Wang ZM, Kwek KY, Xia D and Xia Y: Safety of AK117, an anti-CD47 monoclonal antibody, in patients with advanced or metastatic solid tumors in a phase I study. J Clini Oncol. 39 (Suppl 15):S26302021. View Article : Google Scholar

106 

Jiang Z, Sun H, Yu J, Tian W and Song Y: Targeting CD47 for cancer immunotherapy. J Hematol Oncol. 14:1802021. View Article : Google Scholar : PubMed/NCBI

107 

Qu T, Li B and Wang Y: Targeting CD47/SIRPα as a therapeutic strategy, where we are and where we are headed. Biomark Res. 10:202022. View Article : Google Scholar : PubMed/NCBI

108 

Puro RJ, Bouchlaka MN, Hiebsch RR, Capoccia BJ, Donio MJ, Manning PT, Frazier WA, Karr RW and Pereira DS: Development of AO-176, a Next-Generation Humanized Anti-CD47 antibody with novel anticancer properties and negligible red blood cell binding. Mol Cancer Ther. 19:835–846. 2020. View Article : Google Scholar : PubMed/NCBI

109 

III HAB, Spira AI, Taylor MH, Yeku OO, Liu JF, Munster P, Hamilton EP, Thomas JS, Gatlin F, Penson RT, et al: A first-in-human study of AO-176, a highly differentiated anti-CD47 antibody, in patients with advanced solid tumors. J Clin Oncol. 39 (15_Suppl):S25162021. View Article : Google Scholar

110 

Zeidan AM, DeAngelo DJ, Palmer J, Seet CS, Tallman MS, Wei X, Raymon H, Sriraman P, Kopytek S, Bewersdorf JP, et al: Phase 1 study of anti-CD47 monoclonal antibody CC-90002 in patients with relapsed/refractory acute myeloid leukemia and high-risk myelodysplastic syndromes. Ann Hematol. 101:557–569. 2022. View Article : Google Scholar : PubMed/NCBI

111 

Zeidan AM, DeAngelo DJ, Palmer JM, DeAngelo DJ, Palmer JM, Seet CS, Tallman MS, Wei X, Li YF, Hock R, et al: A Phase I study of CC-90002, a monoclonal antibody targeting CD47, in patients with relapsed and/or refractory (R/R) acute myeloid leukemia (AML) and High-risk myelodysplastic syndromes (MDS): Final results. Blood. 134:13202019. View Article : Google Scholar

112 

Velliquette RW, Aeschlimann J, Kirkegaard J, Shakarian G, Lomas-Francis C and Westhoff CM: Monoclonal anti-CD47 interference in red cell and platelet testing. Transfusion. 59:730–737. 2019. View Article : Google Scholar : PubMed/NCBI

113 

Ansell SM, Maris MB, Lesokhin AM, Chen RW, Flinn IW, Sawas A, Minden MD, Villa D, Percival MM, Advani AS, et al: Phase I study of the CD47 Blocker TTI-621 in patients with relapsed or refractory hematologic malignancies. Clin Cancer Res. 27:2190–2199. 2021. View Article : Google Scholar : PubMed/NCBI

114 

Patel K, Maris MB, Cheson BD, Zonder JA, Lesokhin AM, Keudell GV, Seymour EK, Lin GHY, Catalano T, Shou Y, et al: Ongoing, first-in-human, phase I dose escalation study of the investigational CD47-blocker TTI-622 in patients with advanced relapsed or refractory lymphoma. J Clin Oncol. 38 (15_Suppl):S30302020. View Article : Google Scholar

115 

Yang H, Xun Y and You H: The landscape overview of CD47-based immunotherapy for hematological malignancies. Biomark Res. 11:152023. View Article : Google Scholar : PubMed/NCBI

116 

Chow LQ, Gainor J, Lakhani N, Chunget HC, Lee KW, Lee J, Lorusso P, Bang YJ, Hodi FS, Fanning P, et al: A phase 1 study of ALX148, a CD47 blocker, in combination with established anticancer antibodies in patients with advanced malignancy. Safety. 1:362019.

117 

Piccione EC, Juarez S, Liu J, Tseng S, Ryan CE, Narayanan C, Wang L, Weiskopf K and Majeti R: A bispecific antibody targeting CD47 and CD20 selectively binds and eliminates dual antigen expressing lymphoma cells. MAbs. 7:946–956. 2015. View Article : Google Scholar : PubMed/NCBI

118 

Yu J, Li S, Chen D, Guo H, Yang C, Zhang W, Zhang L, Zhao G, Tu X, Peng L, et al: IMM0306, a fusion protein of CD20 mAb with the CD47 binding domain of SIRPα, exerts excellent cancer killing efficacy by activating both macrophages and NK cells via blockade of CD47-SIRPα interaction and FcɣR engagement by simultaneously binding to CD47 and CD20 of B cells. Leukemia. 37:695–698. 2023. View Article : Google Scholar : PubMed/NCBI

119 

Wang Y, Ni H, Zhou S, He K, Gao Y, Wu W, Wu M, Wu Z, Qiu X, Zhou Y, et al: Tumor-selective blockade of CD47 signaling with a CD47/PD-L1 bispecific antibody for enhanced anti-tumor activity and limited toxicity. Cancer Immunol Immunother. 70:365–376. 2021. View Article : Google Scholar : PubMed/NCBI

120 

Ke H, Zhang F, Wang J, Xiong L, An X, Tu X, Chen C, Wang Y, Mao M, Guo S, et al: HX009, a novel BsAb dual targeting PD1 × CD47, demonstrates potent anti-lymphoma activity in preclinical models. Sci Rep. 13:54192023. View Article : Google Scholar : PubMed/NCBI

121 

Roohullah A, Ganju V, Zhang F, Zhang L, Yu T, Wilkinson K, Cooper A and de Souza P: First-in-human phase 1 dose escalation study of HX009, a novel recombinant humanized anti-PD-1 and CD47 bispecific antibody, in patients with advanced malignancies. J Clin Oncol. 39:2517. 2021. View Article : Google Scholar

122 

Dheilly E, Moine V, Broyer L, Salgado-Pires S, Johnson Z, Papaioannou A, Cons L, Calloud S, Majocchi S, Rousseau F, et al: Selective blockade of the ubiquitous checkpoint receptor CD47 is enabled by dual-targeting bispecific antibodies. Mol Ther. 25:523–533. 2017. View Article : Google Scholar : PubMed/NCBI

123 

Buatois V, Johnson Z, Salgado-Pires S, Papaioannou A, Hatterer E, Chauchet X, Richard F, Barba L, Daubeuf B, Cons L, et al: Preclinical development of a bispecific antibody that safely and effectively targets CD19 and CD47 for the treatment of B-Cell lymphoma and leukemia. Mol Cancer Ther. 17:1739–1751. 2018. View Article : Google Scholar : PubMed/NCBI

124 

de Silva S, Fromm G, Shuptrine CW, Johannes K, Patel A, Yoo KJ, Huang K and Schreiber TH: CD40 enhances type I interferon responses downstream of CD47 blockade, bridging innate and adaptive immunity. Cancer Immunol Res. 8:230–245. 2020. View Article : Google Scholar : PubMed/NCBI

125 

Golubovskaya V: CAR-T cells targeting immune checkpoint pathway players. Front Biosci (Landmark Ed). 27:1212022. View Article : Google Scholar : PubMed/NCBI

126 

Feins S, Kong W, Williams EF, Milone MC and Fraietta JA: An introduction to chimeric antigen receptor (CAR) T-cell immunotherapy for human cancer. Am J Hematol. 94 (Suppl):S3–S9. 2019. View Article : Google Scholar : PubMed/NCBI

127 

Beckett AN, Chockley P, Pruett-Miller SM, Nguyen P, Vogel P, Sheppard H, Krenciute G, Gottschalk S and DeRenzo C: CD47 expression is critical for CAR T-cell survival in vivo. J Immunother Cancer. 11:e0058572023. View Article : Google Scholar : PubMed/NCBI

128 

Golubovskaya V, Berahovich R, Zhou H, Xu S, Harto H, Li L, Chao CC, Mao MM and Wu L: CD47-CAR-T cells effectively kill target cancer cells and block pancreatic tumor growth. Cancers (Basel). 9:1392017. View Article : Google Scholar : PubMed/NCBI

129 

Shu R, Evtimov VJ, Hammett MV, Nguyen NN, Zhuang J, Hudson PJ, Howard MC, Pupovac A, Trounson AO and Boyd RL: Engineered CAR-T cells targeting TAG-72 and CD47 in ovarian cancer. Mol Ther Oncolytics. 20:325–341. 2021. View Article : Google Scholar : PubMed/NCBI

130 

Chen H, Yang Y, Deng Y, Wei F, Zhao Q, Liu Y, Liu Z, Yu B and Huang Z: Delivery of CD47 blocker SIRPα-Fc by CAR-T cells enhances antitumor efficacy. J Immunother Cancer. 10:e0037372022. View Article : Google Scholar : PubMed/NCBI

131 

Sloas C, Gill S and Klichinsky M: Engineered CAR-macrophages as adoptive immunotherapies for solid tumors. Front Immunol. 12:7833052021. View Article : Google Scholar : PubMed/NCBI

132 

Chen Y, Yu Z, Tan X, Jiang H, Xu Z, Fang Y, Han D, Hong W, Wei W and Tu J: CAR-macrophage: A new immunotherapy candidate against solid tumors. Biomed Pharmacother. 139:1116052021. View Article : Google Scholar : PubMed/NCBI

133 

Klichinsky M, Ruella M, Shestova O, Lu XM, Best A, Zeeman M, Schmierer M, Gabrusiewicz K, Anderson NR, Petty NE, et al: Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat Biotechnol. 38:947–953. 2020. View Article : Google Scholar : PubMed/NCBI

134 

Zhang L, Tian L, Dai X, Yu H, Wang J, Lei A, Zhu M, Xu J, Zhao W, Zhu Y, et al: Pluripotent stem cell-derived CAR-macrophage cells with antigen-dependent anti-cancer cell functions. J Hematol Oncol. 13:1532020. View Article : Google Scholar : PubMed/NCBI

135 

Abdel-Bar HM, Walters AA, Lim Y, Rouatbi N, Qin Y, Gheidari F, Han S, Osman R, Wang JT and Al-Jamal KT: An ‘eat me’ combinatory nano-formulation for systemic immunotherapy of solid tumors. Theranostics. 11:8738–8754. 2021. View Article : Google Scholar : PubMed/NCBI

136 

Chen YC, Shi W, Shi JJ and Lu JJ: Progress of CD47 immune checkpoint blockade agents in anticancer therapy: A hematotoxic perspective. J Cancer Res Clin Oncol. 148:1–14. 2022. View Article : Google Scholar : PubMed/NCBI

137 

Yan X, Lai B, Zhou X, Yang S, Ge Q, Zhou M, Shi C, Xu Z and Ouyang G: The differential expression of CD47 may be related to the pathogenesis from myelodysplastic syndromes to acute myeloid leukemia. Front Oncol. 12:8729992022. View Article : Google Scholar : PubMed/NCBI

138 

Shi M, Gu Y, Jin K, Fang H, Chen Y, Cao Y, Liu X, Lv K, He X, Lin C, et al: CD47 expression in gastric cancer clinical correlates and association with macrophage infiltration. Cancer Immunol Immunother. 70:1831–1840. 2021. View Article : Google Scholar : PubMed/NCBI

139 

Li K, Shi H, Zhang B, Ou X, Ma Q, Chen Y, Shu P, Li D and Wang Y: Myeloid-derived suppressor cells as immunosuppressive regulators and therapeutic targets in cancer. Signal Transduct Target Ther. 6:3622021. View Article : Google Scholar : PubMed/NCBI

140 

Xu S, Wang C, Yang L, Wu J, Li M, Xiao P, Xu Z, Xu Y and Wang K: Targeting immune checkpoints on tumor-associated macrophages in tumor immunotherapy. Front Immunol. 14:11996312023. View Article : Google Scholar : PubMed/NCBI

141 

Zhang H, Liu L, Liu J, Dang P, Hu S, Yuan W, Sun Z, Liu Y and Wang C: Roles of tumor-associated macrophages in anti-PD-1/PD-L1 immunotherapy for solid cancers. Mol Cancer. 22:582023. View Article : Google Scholar : PubMed/NCBI

142 

Torres ETR and Emens LA: Emerging combination immunotherapy strategies for breast cancer: Dual immune checkpoint modulation, antibody-drug conjugates and bispecific antibodies. Breast Cancer Res Treat. 191:291–302. 2022. View Article : Google Scholar : PubMed/NCBI

143 

Chen SH, Dominik PK, 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. 9:e0034642021. View Article : Google Scholar : PubMed/NCBI

144 

van de Donk N and Zweegman S: T-cell-engaging bispecific antibodies in cancer. Lancet. 402:142–158. 2023. View Article : Google Scholar : PubMed/NCBI

145 

Zhang T, Lin Y and Gao Q: Bispecific antibodies targeting immunomodulatory checkpoints for cancer therapy. Cancer Biol Med. 20:181–195. 2023. View Article : Google Scholar : PubMed/NCBI

146 

Olaoba OT, Ayinde KS, Lateef OM, Akintubosun MO, Lawal KA and Adelusi TI: Is the new angel better than the old devil? Challenges and opportunities in CD47-SIRPα-based cancer therapy. Crit Rev Oncol Hematol. 184:1039392023. View Article : Google Scholar : PubMed/NCBI

147 

Cao A, Yi J, Tang X, Szeto CW, Wu R, Wan B, Fang X, Li S, Wang L, Wang L, et al: CD47-blocking antibody ZL-1201 promotes Tumor-associated macrophage phagocytic activity and enhances the efficacy of the therapeutic antibodies and chemotherapy. Cancer Res Commun. 2:1404–1417. 2022. View Article : Google Scholar : PubMed/NCBI

148 

Chen Q, Sun L and Chen ZJ: Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nat Immunol. 17:1142–1149. 2016. View Article : Google Scholar : PubMed/NCBI

149 

Brierley CK, Staves J, Roberts C, Johnson H, Vyas P, Goodnough LT and Murphy MF: The effects of monoclonal anti-CD47 on RBCs, compatibility testing, and transfusion requirements in refractory acute myeloid leukemia. Transfusion. 59:2248–2254. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2024
Volume 27 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wu F, Pang H, Li F, Hua M, Song C and Tang J: Progress in cancer research on the regulator of phagocytosis CD47, which determines the fate of tumor cells (Review). Oncol Lett 27: 256, 2024
APA
Wu, F., Pang, H., Li, F., Hua, M., Song, C., & Tang, J. (2024). Progress in cancer research on the regulator of phagocytosis CD47, which determines the fate of tumor cells (Review). Oncology Letters, 27, 256. https://doi.org/10.3892/ol.2024.14389
MLA
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)". Oncology Letters 27.6 (2024): 256.
Chicago
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)". Oncology Letters 27, no. 6 (2024): 256. https://doi.org/10.3892/ol.2024.14389