Short abstract
The function of the immune system in cancer initiation and progression has been widely examined. Notably, immunotherapy has become a promising approach for cancer treatment. CD47, a member of the immunoglobulin superfamily, plays an important role in the immune regulation of cancer by binding to SIRPα. Multiple studies have detected high CD47 expression on the surface of tumor cells, which indicates poor prognosis. Treatments that block the interaction of CD47 and SIRPα significantly suppress tumor growth and metastasis through diverse mechanisms, such as phagocytosis, antibody-dependent cellular cytotoxicity, and apoptosis. Recently, several studies have reported increased CD47 expression on different types of lymphoma cells, indicating that the CD47-SIRPα pathway can be used as a therapeutic target in lymphoma. This review focuses on the role of CD47-SIRPα in B-cell lymphoma and discusses promising therapeutic strategies targeting the CD47-SIRPα axis, which yield insights into the immunotherapy of B-cell lymphoma.
Keywords: CD47, SIRPα, immunotherapy, combination strategy, immunoglobulin, B-cell lymphoma
Introduction
Non-Hodgkin lymphoma (NHL) is a common lymphoid malignancy. According to the American Cancer Society, the morbidity of NHL currently ranks seventh among all cancers.1 Although chemo-immunotherapy improves the survival of CD20-positive B-cell lymphoma, many patients experience disease relapse, and some show drug resistance to both conventional chemotherapy and rituximab, suggesting that additional approaches are needed to more effectively treat lymphoma patients.2 Immunotherapy is a novel approach for treatment of B-cell lymphoma that is relapsed or refractory after chemotherapy. Chimeric antigen receptor T cell (CAR-T) therapy uses genetic engineering to alter T cells to produce transmembrane proteins on the cell surface with an extracellular antibody fragment domain that recognizes a tumor antigen; such therapy shows high response rates in refractory B-cell lymphoma.3 As a fusion protein containing both an antigen recognition domain and T cell signaling domain, the CAR specifically activates T cell-mediated anti-tumor immune responses.4 In contrast, immune checkpoint inhibitors, such as programmed cell death-1 or programmed cell death ligand-1 (PD-1/PD-L1) monoclonal antibody (mAb), can activate T cells to attack cancer cells; thus, they offer new options for patients with lymphoma.5 PD-1 is an immune checkpoint receptor expressed on the surface of T cells; by binding to PD-L1, predominantly expressed on tumor cells, PD-1 attenuates the T cell-mediated anti-tumor immune response.6
The innate immune system also plays an important role in anti-tumor responses. Macrophages are important components of innate immunity, which can inhibit tumor growth through phagocytosis. CD47 is a 50-kDa ubiquitous cell membrane protein in the immunoglobulin superfamily. By interacting with integrin αvβ3 and thrombospondin, CD47 participates in regulation of cell motility, adhesion, migration, and platelet activation.7–10 Signal regulatory protein-α (SIRP-α) is another immunoglobulin superfamily transmembrane receptor primarily expressed on the surface of myeloid cells, including macrophages, granulocytes, monocytes, and dendritic cells.11 The interaction of SIRP-α with CD47 phosphorylates its immunoreceptor tyrosine-based inhibition motif, then activates the inhibitory tyrosine phosphatases SHP-1 and SHP-2 to suppress phagocytosis.12,13 CD47 was first identified on human ovarian tumors. A growing number of studies have reported high CD47 expression on diverse types of cancers, including breast cancer, hepatocellular carcinoma, and colon glioblastoma; high CD47 expression is associated with poor prognosis.14–17 Recently, CD47 was reported to be expressed on NHL.18 Therefore, targeting CD47 may be a novel strategy for lymphoma treatment. This review focuses on the role of the CD47-SIRPα pathway and effects of therapeutic strategies targeting this pathway in B-cell lymphoma.
Mechanisms and effects of blocking CD47 in cancer
CD47 expressed on tumor cells and tumor stem cells has been identified as a “don’t eat me” signal, and binding of CD47 to SIRPα contributes to inhibition of macrophage phagocytosis, thus facilitating cancer immune evasion.19–21 Indeed, Liu et al.22 reported that high CD47 expression led to the progression of ovarian cancer by inhibiting macrophage phagocytosis; CD47 downregulation by shRNA, or its inhibition by mAb, promoted phagocytosis and macrophage infiltration in tumor cells.22 Anti-CD47 antibodies also induced macrophage-mediated phagocytosis and suppressed tumor growth in myeloma, hepatocellular carcinoma, and glioblastoma23–26; additionally, blockage of CD47 increased M1 macrophages, affected overall macrophage distribution, and promoted the migration of macrophages into the tumor.25–26 These studies demonstrated that blocking the CD47 inhibitory signal promoted an innate immune response through macrophage-dependent phagocytosis.
The anti-tumor mechanisms of blocking CD47 include: 1) Activation of the antibody-dependent cellular cytotoxicity (ADCC)-mediated innate immune response. For example, Kim et al.27 showed that high CD47 expression was associated with a reduction in natural killer (NK) cell-mediated cytotoxicity in head-and-neck squamous cell carcinoma (HNSCC); treatment with anti-CD47 antibody remarkably increased NK cell-mediated cytotoxicity against HNSCC.27 Chao et al.28 observed that the use of anti-CD47 antibody caused NK cell-mediated ADCC in lymphoma, in an Fc receptor-dependent manner. 2) Promotion of adaptive immunity. Tseng et al.29 reported that antigen-specific CD8+ T cells proliferated after macrophage phagocytosis induced by the use of an anti-CD47 antibody in colon cancer, whereas the number of regulatory T cells was reduced; an increase in CD8+ T cells showed good anti-tumor effects in vivo, indicating that the use of an anti-CD47 antibody enabled adaptive T cell immune responses and overwhelmed the regulatory T cell-mediated immune evasion in cancer.29 Similarly, another study showed that the anti-CD47 antibody inhibited tumor progression by enhancing the antigen-specific CD8+ T cell response, which was activated by dendritic cell-mediated tumor antigen presentation to T cells.30 Recently, Soto-Pantoja et al.31 reported that blocking of CD47 induced a cytotoxic T cell-dependent anti-tumor immune response and enhanced the effects of irradiation in fibrosarcoma; CD47 knockdown in CD8+ T cells also significantly enhanced their tumoricidal activity. Moreover, elevated CD47 expression was associated with reduced CD8+ T cell infiltration in melanoma.31 3) Direct induction of apoptosis. Several studies showed that the blockage of CD47 by the antibody MABL (specific for an extracellular domain of CD47) promoted apoptosis of leukemia and myeloma cells.32–33 Additionally, MABL-induced apoptotic activity in myeloma cells was not affected by chemotherapy, indicating an independent anti-myeloma role for MABL-induced apoptosis.33 4) HIF-1-mediated inhibition of cancer stem cells (CSCs). Zhang et al.34 reported that CD47 gene transcription was dependent on HIF-1 in breast cancer. CD47 expression also contributed to maintenance of breast CSCs. Therefore, downregulation of CD47 by blockage of HIF-1 may inhibit breast CSCs.
Therapeutic strategies targeting CD47-SIRPα in B-cell lymphoma
Increasing evidence has demonstrated that increased CD47 expression by different types of B-cell lymphoid malignancies was associated with tumor progression and dissemination. For example, Chao et al.28,35 reported that CD47 mRNA expression was significantly increased in various B-cell lymphoma cells, and that high CD47 expression indicated poor survival and disease progression in diffuse large B-cell lymphoma (DLBCL), B-cell chronic lymphocytic leukemia (B-CLL), and mantle cell lymphoma. Subsequently, increased CD47 expression was detected in disseminated lymphoma samples; further, CD47 knockdown reduced disease involvement at secondary sites in a lymphoma xenograft model, indicating that lymphoma dissemination was CD47-dependent.28,35 Starr et al.36 also reported that CD47 was highly expressed in both nodal and intravascular DLBCL cells, indicating that CD47 plays an important role in the intravascular dissemination of DLBCL. Therefore, blocking the CD47-SIRPα pathway may be an effective approach to treat B-cell lymphoma.
Strategies targeting the CD47-SIRPα pathway in B-cell lymphoma are diverse. First, CD47 can be directly blocked by monoclonal antibodies. Second, CD47 can bind to recombinant polypeptides derived from SIRPα, such as the SIRPα-Fc fusion protein. Additionally, the interaction of CD47 and SIRPα can be inhibited by an anti-SIRPα antibody.37 Goto et al.38 reported increased expression levels of CD47 on the surface of primary effusion lymphoma (PEL) cell lines, and showed that CD47 knockdown by siRNA or anti-CD47 antibody increased the phagocytosis of PEL cells by macrophages; they also demonstrated that the use of an anti-CD47 antibody significantly inhibited the growth and metastasis of PEL cells in a xenograft mouse model.38 Similarly, Chao et al.28,35 demonstrated that both anti-CD47 and anti-SIRPα antibodies promoted macrophage phagocytosis of NHL cells, in a manner dependent on the level of CD47 expression; additionally, the use of an anti-CD47 antibody inhibited tumor growth and extended the survival of both localized and disseminated lymphoma in vivo. Interestingly, although the anti-CD47 antibody did not fully eliminate the lymphoma, it reduced the growth rate of the tumor and prevented both extranodal and hematogenous spread in a macrophage-dependent manner, demonstrating the inhibitory effect of anti-CD47 antibody on lymphoma dissemination.28,35 Uno et al.39 reported that the anti-CD47 monoclonal antibody (mAb), MABL, enabled apoptosis in JOK-1 cells, but did not induce cell death in CD34+ progenitor cells; additionally, treatment with MABL prolonged the survival of xenograft mice; the median survival of mice injected with MABL was longer than that of mice treated with fludarabine, demonstrating the efficacy of MABL in eradicating B-CLL.39 To reduce hemagglutination from MABL, Sagawa et al.40 developed an S-S diabody, a disulfide-stabilized dimer of a single-chain antibody fragment of MABL; the S-S diabody induced apoptosis in B-CLL cells, but not in normal leukocytes. The experiment in B-CLL-transplanted mice also revealed the anti-tumor effects of the S-S diabody, which blocked tumor growth and improved survival.40 Additionally, CD47 ligation by an anti-CD47 mAb induced apoptosis and cell death in B-CLL through the caspase-independent pathway; apoptotic cells were then eliminated by dendritic cell-mediated phagocytosis.41–42
However, CD47 is also widely expressed on normal cells, including red blood cells, platelets, and mesenchymal stem cells, weakening the specificity of the antibody towards tumor cells.43 Therefore, several studies have developed bispecific antibodies (BsAbs) to specifically limit CD47 neutralization to tumor cells. Piccione et al.44 reported that BsAbs co-targeting CD47 and CD20 increased the phagocytosis of NHL cells; BsAb also reduced the tumor burden in both localized and disseminated NHL mouse models and significantly prolonged survival, compared with either anti-CD47 antibody or rituximab monotherapy.44 Métayer et al.45 showed that both anti-CD47 and anti-CD19 antibodies induced phagocytosis in Burkitt’s lymphoma cells. Subsequently, Dheilly et al.46 developed a CD47/CD19 dual-targeting BsAb and demonstrated its binding selectivity and anti-lymphoma effects, which were mediated by antibody-dependent cellular phagocytosis. Although these BsAbs showed satisfactory binding selectivity, the interaction of the functional Fc fragment of these BsAbs with the Fc receptor of macrophages led to phagocytic systemic toxicity and premature off-target effects that reduced the accumulation of BsAbs on the tumor cell surface. Recently, van Bommel et al.47 built a novel BsAb containing single-chain fragments of variable regions (scFv) of anti-CD47 antibody and scFv (derived from the anti-CD20 antibody rituximab) to resolve this limitation. They demonstrated that this novel BsAb specifically induced antibody-dependent cellular phagocytosis in CD20+/CD47+ malignant B-cell lymphoma in an Fc-independent manner, and that it enhanced the anti-tumor effect of the mAbs daratumumab, alemtuzumab, and obinutuzumab.47
In addition, combination strategies based on blockage of the CD47-SIRPα pathway exerted synergistic anti-lymphoma effects. Liu et al.48 reported that the combination of Hu5F9-G4 and rituximab resulted in phagocytic elimination of lymphoma and significantly prolonged the survival time of NHL model mice. Similarly, Chao et al.28 reported that an anti-CD47 antibody enhanced phagocytosis induced by rituximab in different lymphoma cell lines, but not in normal peripheral blood cells; notably, combination therapy with an anti-CD47 antibody and rituximab eliminated lymphoma in some xenograft mice and induced long-term survival without disease relapse. The improved anti-tumor effect originated from anti-CD47 antibody-mediated Fc-independent and rituximab-mediated Fc-dependent macrophage phagocytosis, but not through NK cells or complement.28 The anti-SIRPα mAb MY-1 also facilitated the phagocytosis of Burkitt’s lymphoma cells induced by rituximab; MY-1 significantly enhanced the inhibitory effect of rituximab on lymphoma growth in vivo.49 Furthermore, the safety and anti-lymphoma effects of several agents blocking CD47 alone or in combination with rituximab have been investigated in phase 1 and 2 clinical trials (Table 1).
Table 1.
NCT number | Agent | Strategy | Type of lymphoma | Phase |
---|---|---|---|---|
NCT02663518 | TTI-621 | Single agent; combination with rituximab | NHL | Phase I |
NCT02367196 | CC-90002 | Single agent; combination with rituximab | NHL | Phase I |
NCT02953509 | Hu5F9-G4 | Single agent; combination with rituximab | NHL; DLCBL; indolent lymphoma | Phase I+II |
NCT03013218 | ALX148 | Combination with rituximab | NHL | Phase I |
Abbreviations: DLBCL: diffuse large B-cell lymphoma; NHL: non-Hodgkin lymphoma
In addition to their synergistic effects with tumor antigen-specific antibodies, antibodies targeting CD47-SIRPα may combine with other agents to augment treatment efficacy. For example, an anti-CD47 mAb triggered type III programmed cell death in B-CLL cells, demonstrating involvement of the caspase-independent pathway in CD47-mediated tumor destruction; anti-CD47 mAb-induced type III programmed cell death was associated with F-actin dynamics. Thus, additional F-actin regulators may enhance the anti-tumor effect of the anti-CD47 mAb. Additionally, because caspase-dependent apoptosis enables cell death in B-CLL, therapeutic strategies that combine anti-CD47 mAb with caspase modulators may constitute promising approaches.50 TTI-621, a soluble SIRPα Fc fusion protein that blocks CD47, significantly increased the phagocytosis of lymphoma cells by macrophages in vitro and tumor-associated macrophages derived from xenograft DLCBL tumors. TTI-621-stimulated macrophages exhibited a highly phagocytic phenotype upon exposure to cytokines (interferon (IFN)-γ, IFN-α, and interleukin-10) or Toll-like receptor agonists (lipopolysaccharides, Poly (I:C), and R848), suggesting that blockage of CD47 with macrophage regulators may serve as a potential combination therapy.51 Furthermore, Gautam et al.52 observed that the Hsp70-peptide complex transformed M2 macrophages into tumor-inhibiting M1 macrophages in Dalton’s lymphoma; additionally, SIRPα expression on macrophages was elevated after treatment with Hsp70-peptide complex. Therefore, the combination of Hsp70 with an anti-SIRPα antibody may have synergistic anti-lymphoma effects52 (Table 2).
Table 2.
Drug | Type of tumor | Mechanism | Combined agents | Refs |
---|---|---|---|---|
Anti-CD47 antibody | PEL | Phagocytosis | / | 38 |
MABL | B-CLL | Apoptosis | / | 39 |
S-S diabody | NHL | Apoptosis | / | 40 |
Anti-CD47 antibody | B-CLL | Apoptosis | / | 41 |
CD47/CD20 BsAb | NHL | Phagocytosis | / | 44 |
CD47/CD19 BsAb | Burkitt’s lymphoma | ADCP | / | 46 |
CD47/CD20 scFv | B-cell lymphoma | ADCP | / | 47 |
Anti-CD47 antibody; Anti-SIRPα antibody | NHL | Phagocytosis | Rituximab | 28 |
Anti-CD47 antibody | Burkitt’s lymphoma | ADCP | Anti-CD10 antibody; anti-CD19 antibody | 45 |
Hu5F9-G4 | NHL | Not mentioned | Rituximab | 48 |
MY-1 | Burkitt’s lymphoma | Phagocytosis | Rituximab | 49 |
Anti-CD47 antibody | B-CLL | Type III PCD | F-actin regulators; caspase modulators | 50 |
TTI-621 | DLBCL | Phagocytosis | Macrophage agonists | 51 |
Abbreviations: ADCP: antibody-dependent cellular phagocytosis; BsAb: bispecific antibody; B-CLL: B-chronic lymphocytic leukemia; DLBCL: diffuse large B-cell lymphoma; NHL: non-Hodgkin lymphoma; PCD: programmed cell death; PEL: primary effusion lymphoma; scFv: single-chain fragment of variable regions
Conclusion
Tumor immune escape is a primary mechanism of lymphoma progression and dissemination. Therefore, immunotherapy has become a hotspot of lymphoma treatment in recent years. The CD47-SIRPα axis plays an important role in the immune regulation of lymphoma. Studies targeting the CD47-SIRPα pathway have shown significant anti-lymphoma effects, mainly through the activation of innate immunity, mediated by macrophage phagocytosis, or direct promotion of apoptosis. However, anti-CD47 antibodies have some limitations: 1) CD47 is not solely expressed on lymphoma cells; it is also expressed on normal cells, resulting in toxic effects and antibody exhaustion. Bispecific antibodies co-targeting CD47 and other tumor-specific antigens may improve the binding specificity of antibodies and tumor cells, enhancing safety and efficacy. 2) Most studies have reported that anti-CD47 antibody monotherapy does not fully eliminate lymphoma; combination strategies that activate adoptive immunity or involve the use of the anti-CD20 antibody, macrophage agonists such as IFN-γ, IFN-α, interleukin-10, and other agents (e.g., caspase modulators and F-actin regulators), may have lasting and effective anti-lymphoma activities. 3) The efficacies of different methods of blocking CD47, such as anti-CD47 antibody or scFv derived from an antibody, remain unknown. Therefore, strategies based on blockage of the CD47-SIRPα axis require further evaluation in pre-clinical studies and clinical trials, and may provide new directions for lymphoma treatment.
Declaration of conflicting interest
The authors declare no potential conflicts of interest with respect to the research, authorship and/or publication of this article.
Funding
The author(s) disclosed receipt of the following financial support for the research, authorship and/or publication of this article: This research was supported by the National Natural Science Foundation of China (No. 81670178), The National Key Research and Development Program of China (No. 2016YFC090150X), the Research Project for Practice Development of National TCM Clinical Research Bases (No. JDZX2015113), and the Funds of Science Technology Department of Zhejiang Province (No. 2018C03016-1).
References
- 1.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin 2018; 68: 7–30. doi: 10.3322/caac.21442. [DOI] [PubMed] [Google Scholar]
- 2.Cheson BD, Leonard JP. Monoclonal antibody therapy for B-cell non-Hodgkin's lymphoma. N Engl J Med 2008; 359: 613–626. doi: 10.1056/NEJMra0708875. [DOI] [PubMed] [Google Scholar]
- 3.Brudno JN, Kochenderfer JN. Chimeric antigen receptor T-cell therapies for lymphoma. Nat Rev Clin Oncol 2018; 15: 31–46. doi: 10.1038/nrclinonc. [DOI] [PubMed] [Google Scholar]
- 4.Neelapu SS, Locke FL, Bartlett NL, et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. N Engl J Med 2017; 377: 2531–2544. doi: 10.1056/NEJMoa1707447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Hude I, Sasse S, Engert A, et al. The emerging role of immune checkpoint inhibition in malignant lymphoma. Haematologica 2017; 102: 30–42. doi: 10.3324/haematol.2016.150656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Lesokhin AM, Ansell SM, Armand P, et al. Nivolumab in patients with relapsed or refractory hematologic malignancy: preliminary results of a phase Ib study. J Clin Oncol 2016; 34: 2698–2704. doi: 10.1200/JCO.2015.65.9789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Zhang L, Huang H. Targeting the cancer biomarker CD47: a review on the diverse mechanisms of CD47 pathway in cancer treatment. Anticancer Agents Med Chem 2016; 16: 658–667. [DOI] [PubMed] [Google Scholar]
- 8.Brown E, Hooper L, Ho T, et al. Integrin associated protein: a 50-kD plasma membrane antigen physically and functionally associated with integrins. J Cell Biol 1990; 111: 2785–2794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Lindberg FP, Gresham HD, Reinhold MI, et al. Integrin-associated protein immunoglobulin domain is necessary for efficient vitronectin bead binding. J Cell Biol 1996; 134: 1313–1322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Isenberg JS, Ridnour LA, Dimitry J, et al. CD47 is necessary for inhibition of nitric oxide-stimulated vascular cell responses by thrombospondin-1. J Biol Chem 2006; 281: 26069–26080. [DOI] [PubMed] [Google Scholar]
- 11.Kong F, Gao F, Li H, et al. CD47: a potential immunotherapy target for eliminating cancer cells. Clin Transl Oncol 2016; 18: 1051–1055. [DOI] [PubMed] [Google Scholar]
- 12.van Beek EM, Cochrane F, Barclay AN, et al. Signal regulatory proteins in the immune system. J Immunol 2005; 175: 7781–7787. [DOI] [PubMed] [Google Scholar]
- 13.Timms JF, Carlberg K, Gu H, et al. Identification of major binding proteins and substrates for the SH2-containing protein tyrosine phosphatase SHP-1 in macrophages. Mol Cell Biol 1998; 18: 3838–3850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Zhao XW, van Beek EM, Schornagel K, et al. CD47-signal regulatory protein-alpha (SIRP alpha) interactions form a barrier for antibody-mediated tumor cell destruction. Proc Natl Acad Sci U S A 2011; 108: 18342–18347. doi: 10.1073/pnas.1106550108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Baccelli I, Stenzinger A, Vogel V, et al. Co-expression of MET and CD47 is a novel prognosticator for survival of luminal breast cancer patients. Oncotarget 2014; 5: 8147–8160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lee TK, Cheung VC, Lu P, et al. Blockade of CD47-mediated cathepsin S/protease-activated receptor 2 signaling provides a therapeutic target for hepatocellular carcinoma. Hepatology 2014; 60: 179–191. doi: 10.1002/hep.27070. [DOI] [PubMed] [Google Scholar]
- 17.Willingham SB, Volkmer JP, Gentles AJ, et al. : 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. doi: 10.1073/pnas.1121623109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Chao MP, Weissman IL, Majeti R. The CD47-SIRPα pathway in cancer immune evasion and potential therapeutic implications. Curr Opin Immunol 2012; 24: 225–232. doi: 10.1016/j.coi.2012.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Jaiswal S, Jamieson CH, Pang WW, et al. CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell 2009; 138: 271–285. doi: 10.1016/j.cell.2009.05.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Murata Y, Kotani T, Ohnishi H, et al. The CD47-SIRPα signalling system: its physiological roles and therapeutic application. J Biochem 2014; 155: 335–344. doi: 10.1093/jb/mvu017. [DOI] [PubMed] [Google Scholar]
- 21.Majeti R, Chao MP, Alizadeh AA, et al. CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell 2009; 138: 286–299. doi: 10.1016/j.cell.2009.05.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Liu R, Wei H, Gao P, et al. CD47 promotes ovarian cancer progression by inhibiting macrophage phagocytosis. Oncotarget 2017; 8: 39021–39032. doi: 10.18632/oncotarget.16547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kim D, Wang J, Willingham SB, et al. Anti-CD47 antibodies promote phagocytosis and inhibit the growth of human myeloma cells. Leukemia 2012; 26: 2538–2545. doi: 10.1038/leu.2012.141. [DOI] [PubMed] [Google Scholar]
- 24.Lo J, Lau EY, So FT, et al. : Anti-CD47 antibody suppresses tumour growth and augments the effect of chemotherapy treatment in hepatocellular carcinoma. Liver Int 2016; 36: 737–745. doi: 10.1111/liv.12963. [DOI] [PubMed] [Google Scholar]
- 25.Xiao Z, Chung H, Banan B, et al. Antibody mediated therapy targeting CD47 inhibits tumor progression of hepatocellular carcinoma. Cancer Lett 2015; 360: 302–309. doi: 10.1016/j.canlet.2015.02.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Zhang M, Hutter G, Kahn SA, et al. Anti-CD47 treatment stimulates phagocytosis of glioblastoma by M1 and M2 polarized macrophages and promotes M1 polarized macrophages In Vivo. PLoS One 2016; 11: e0153550. doi: 10.1371/journal.pone.0153550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Kim MJ, Lee JC, Lee JJ, et al. Association of CD47 with natural killer cell-mediated cytotoxicity of head-and-neck squamous cell carcinoma lines. Tumour Biol 2008; 29: 28–34. doi: 10.1159/000132568. [DOI] [PubMed] [Google Scholar]
- 28.Chao MP, Alizadeh AA, Tang C, et al. Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma. Cell 2010; 142: 699–713. doi: 10.1016/j.cell.2010.07.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Tseng D, Volkmer JP, Willingham SB, et al. Anti-CD47 antibody–mediated phagocytosis of cancer by macrophages primes an effective antitumor T-cell response. Proc Natl Acad Sci U S A 2013; 110: 11103–11108. doi: 10.1073/pnas.1305569110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Liu X, Pu Y, Cron K, et al. CD47 blockade triggers t cell-mediated destruction of immunogenic tumors. Nat Med 2015; 21: 1209–1215. doi: 10.1038/nm.3931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Soto-Pantoja DR, Terabe M, Ghosh A, et al. CD47 in the tumor microenvironment limits cooperation between anti-tumor T cell immunity and radiation therapy. Cancer Res 2014; 74: 6771–6783. doi: 10.1158/0008-5472.CAN-14-0037-T. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Kikuchi Y, Uno S, Yoshimura Y, et al. A bivalent single-chain Fv fragment against CD47 induces apoptosis for leukemic cells. Biochem Biophys Res Commun 2004; 315: 912–918. [DOI] [PubMed] [Google Scholar]
- 33.Kikuchi Y, Uno S, Kinoshita Y, et al. Apoptosis inducing bivalent single-chain antibody fragments against CD47 showed antitumor potency for multiple myeloma. Leuk Res 2005; 29: 445–450. [DOI] [PubMed] [Google Scholar]
- 34.Zhang H, Lu H, Xiang L, et al. HIF-1 regulates CD47 expression in breast cancer cells to promote evasion of phagocytosis and maintenance of cancer stem cells. Proc Natl Acad Sci U S A 2015; 112: E6215–E6223. doi: 10.1073/pnas.1520032112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Chao MP, Tang C, Pachynski RK, et al. Extranodal dissemination of non-Hodgkin lymphoma requires CD47 and is inhibited by anti-CD47 antibody therapy. Blood 2011; 118: 4890–4901. doi: 10.1182/blood-2011-02-338020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Starr JS, Jiang L, Li Z, et al. CD47 and osteopontin expression in diffuse large B-cell lymphoma with nodal and intravascular involvement. Clin Lymphoma Myeloma Leuk 2013; 13: 597–601. doi: 10.1016/j.clml.2013.05.001. [DOI] [PubMed] [Google Scholar]
- 37.Weiskopf K. Cancer immunotherapy targeting the CD47/SIRPα axis. Eur J Cancer 2017; 76: 100–109. doi: 10.1016/j.ejca.2017.02.013. [DOI] [PubMed] [Google Scholar]
- 38.Goto H, Kojima Y, Matsuda K, et al. Efficacy of anti-CD47 antibody-mediated phagocytosis with macrophages against primary effusion lymphoma. Eur J Cancer 2014; 50: 1836–1846. doi: 10.1016/j.ejca.2014.03.004. [DOI] [PubMed] [Google Scholar]
- 39.Uno S, Kinoshita Y, Azuma Y, et al. Antitumor activity of a monoclonal antibody against CD47 in xenograft models of human leukemia. Oncol Rep 2007; 17: 1189–1194. [PubMed] [Google Scholar]
- 40.Sagawa M, Shimizu T, Fukushima N, et al. A new disulfide-linked dimer of a single-chain antibody fragment against human CD47 induces apoptosis in lymphoid malignant cells via the hypoxia inducible factor-1α pathway. Cancer Sci 2011; 102: 1208–1215. doi: 10.1111/j.1349-7006.2011.01925.x. [DOI] [PubMed] [Google Scholar]
- 41.Mateo V, Lagneaux L, Bron D, et al. CD47 ligation induces caspase-independent cell death in chronic lymphocytic leukemia. Nat Med 1999; 5: 1277–1284. [DOI] [PubMed] [Google Scholar]
- 42.Mateo V, Brown EJ, Biron G, et al. Mechanisms of CD47-induced caspase-independent cell death in normal and leukemic cells: link between phosphatidylserine exposure and cytoskeleton organization. Blood 2002; 100: 2882–2890. [DOI] [PubMed] [Google Scholar]
- 43.Subramanian S, Parthasarathy R, Sen S, et al. Species- and cell type-specific interactions between CD47 and human SIRPalpha. Blood 2006; 107: 2548–2556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Piccione EC, Juarez S, Liu J, et al. A bispecific antibody targeting CD47 and CD20 selectively binds and eliminates dual antigen expressing lymphoma cells. MAbs 2015; 7: 946–956. doi: 10.1080/19420862.2015.1062192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Métayer LE, Vilalta A, Burke GAA, et al. Anti-CD47 antibodies induce phagocytosis of live, malignant B cells by macrophages via the Fc domain, resulting in cell death by phagoptosis. Oncotarget 2017; 8: 60892–60903. doi: 10.18632/oncotarget.18492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Dheilly E, Moine V, Broyer L, et al. Selective blockade of the ubiquitous checkpoint receptor CD47 is enabled by dual-targeting bispecific antibodies. Mol Ther 2017; 25: 523–533. doi: 10.1016/j.ymthe.2016.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.van Bommel PE, He Y, Schepel I, et al. CD20-selective inhibition of CD47-SIRPα “don't eat me” signaling with a bispecific antibody-derivative enhances the anticancer activity of daratumumab, alemtuzumab and obinutuzumab. Oncoimmunology 2017; 7: e1386361. doi: 10.1080/2162402X.2017.1386361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Liu J, Wang L, Zhao F, et al. Pre-clinical development of a humanized anti-CD47 antibody with anti-cancer therapeutic potential. PLoS One 2015; 10: e0137345. doi: 10.1371/journal.pone.0137345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Yanagita T, Murata Y, Tanaka D, et al. Anti-SIRPα antibodies as a potential new tool for cancer immunotherapy. JCI Insight 2017; 2: e89140. doi: 10.1172/jci.insight.89140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Barbier S, Chatre L, Bras M, et al. Caspase-independent type III programmed cell death in chronic lymphocytic leukemia: the key role of the F-actin cytoskeleton. Haematologica 2009; 94: 507–517. doi: 10.3324/haematol.13690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Lin GHY, Chai V, Lee V, et al. TTI-621 (SIRPαFc), a CD47-blocking cancer immunotherapeutic, triggers phagocytosis of lymphoma cells by multiple polarized macrophage subsets. PLoS One 2017; 12: e0187262. doi: 10.1371/journal.pone.0187262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Gautam PK, Acharya A. Suppressed expression of homotypic multinucleation, extracellular domains of CD172α (SIRP-α) and CD47 (IAP) receptors in TAMs upregulated by Hsp70-peptide complex in Dalton's lymphoma. Scand J Immunol 2014; 80: 22–35. doi: 10.1111/sji.12180. [DOI] [PubMed] [Google Scholar]