Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2595 word(s) 2595 2021-12-15 04:11:55 |
2 Format change -14 word(s) 2581 2022-01-13 02:26:04 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Buchbinder, E. Cancer Therapy Targeting CD47. Encyclopedia. Available online: https://encyclopedia.pub/entry/18169 (accessed on 10 August 2024).
Buchbinder E. Cancer Therapy Targeting CD47. Encyclopedia. Available at: https://encyclopedia.pub/entry/18169. Accessed August 10, 2024.
Buchbinder, Elizabeth. "Cancer Therapy Targeting CD47" Encyclopedia, https://encyclopedia.pub/entry/18169 (accessed August 10, 2024).
Buchbinder, E. (2022, January 12). Cancer Therapy Targeting CD47. In Encyclopedia. https://encyclopedia.pub/entry/18169
Buchbinder, Elizabeth. "Cancer Therapy Targeting CD47." Encyclopedia. Web. 12 January, 2022.
Cancer Therapy Targeting CD47
Edit

The interaction between cluster of differentiation 47 (CD47) on cancer cells and signal regulatory protein alpha (SIRPα) on immune cells, such as macrophages and dendritic cells, generates a “don’t eat me” signal. This is a common mechanism that provides cancer cells an escape from the innate immune system. Several therapeutics directed to CD47 or SIRPα have entered early clinical trials in recent years.

CD47 cancer

1. Background

In the past decade, the field of cancer immunotherapy has rapidly advanced, establishing a crucial role for immune checkpoint blockers in the treatment of a variety of cancer types. In parallel with these remarkable clinical developments, further efforts have focused on ways of unleashing adaptive immune responses against cancer. CD47, a cell surface molecule overexpressed by several cancer types that facilitates immune escape from macrophages, dendritic cells and natural killer cells, and its ligand SIRPα, have emerged as potential therapeutic targets. A number of agents directed to CD47/SIRPα have been developed and demonstrated preclinical activity. Early phase clinical trials are investigating CD47/SIRPα directed agents with available data, suggesting safety and preliminary activity. 

2. Role of CD47/SIRPα in Cancer

In the early 1990s, the first oncological studies of CD47 identified it as a potential tumor marker for ovarian cancer [1]. This was followed by investigations on a wide variety of solid and hematological cancer types, including head and neck small-cell carcinoma (HNSCC), breast cancer, acute myeloid leukemia (AML), non-Hodgkin’s lymphoma (NHL), myeloma demonstrating differential overexpression of CD47 between cancer cells and matched normal cells [2][3][4][5][6][7][8][9].
The role of the CD47/SIRPα interaction in providing an escape mechanism for cancer cells from macrophage targeting has been well described. Human-derived xenograft models for several types of malignancies demonstrated sensitivity to CD47-blocking antibodies. In culture, these antibodies induced the macrophage-mediated phagocytosis of tumor cells [10][11][12][13][14][15][16][17]. The impact of the CD47 blockade on macrophage populations within the tumor microenvironment was also studied. In brief, TAMs display different polarization states between M1 macrophages with anti-tumor phenotypes and M2 macrophages with pro-tumor and immunosuppressive phenotypes [18][19][20]. In a human glioblastoma model, anti-CD47 therapy increased M1 macrophages within the tumor. This finding suggests that anti-CD47 therapy may play a role in shifting the phenotype of macrophages toward the anti-tumorigenic M1 subtype [21]. CD47 signaling also participates in macrophage recruitment into tumors. Weiskopf and colleagues showed that phagocytosis, following anti-CD47 treatment, causes systemic and local secretion of chemokines and cytokines that recruit macrophages into tumors in mice engrafted with small-cell lung cancer (SCLC) cell lines [10].
Beyond the activation of macrophage-mediated tumor killing, CD47-SIRPα interruption exerts other multidimensional positive effects on the immune response against cancer cells. For example, CD47-SIRPα blockade augments antibody dependent cellular cytotoxicity (ADCC) via the inhibition of SIRPα, expressed on the surface of NK cells [22][23]. Kim and colleagues demonstrated that impaired NK cell activity present in HNSCC cell lines overexpressing CD47 could be reversed with anti-CD47 antibodies [24]. CD47-SIRPα antagonist agents with an intact or even partially inactive Fc portion embedded in their structure may foster anti-tumor activity via antibody opsonization and destruction of target cells through ADCC or antibody-dependent cellular phagocytosis (ADCP) [25]. In addition, CD47/SIRPα interaction also has roles in tumor cell apoptosis, proliferation and migration [26][27][28]. CD47 inhibition can also negatively impact the function of other CD47 ligands, such as TSP-1 and integrins. These indirect effects may contribute to the anti-tumor and pro-inflammatory activity of CD47 inhibition. Despite contrasting evidence, a growing body of research highlights the role of TSP-1 in cell proliferation, invasion, metastatic potential, and worse survival rates, either through its interaction with CD47 or independently [29][30][31]. Notably, Kamijo and colleagues reported an association between high TSP-1 expression and worse disease-free survival in cutaneous T cell lymphoma patients. TSP-1 was found to be overexpressed in cutaneous T cell lymphoma, and anti-CD47 antibodies led to the inhibition of TSP-1-mediated cell proliferation in vivo [32].
Preclinical work has suggested a synergy between the cytotoxic agents and the CD47 inhibitors, especially when cytotoxic therapies were introduced prior to CD47-directed therapies. Neoantigens and nucleic acid remnants, produced from dying cancer cells and released into the tumor microenvironment after chemotherapy, may potentiate anti-CD47 activity [33]. In the context of hematologic malignancies, in vitro studies showed that azacytidine (a standard of care DNA hypomethylating agent used in the treatment of AML) and myelodysplastic syndrome and venetoclax (a B-cell lymphoma-2 inhibitor used in AML), induces the expression of other pro-phagocytic pathway components such as calreticulin and CD47 [34].
Perhaps more intriguingly, the macrophages involved in phagocytosis function as antigen-presenting cells, linking innate and adaptive immunity [9][33][35]. Thus, targeting the CD47-SIRPα axis, either through the CD47 or SIRPα blockade, may also promote antigen-presenting cell function, and stimulate T cell-mediated anti-cancer immunity (Figure 1) [36][37]. Studies in preclinical models with cancer types including chronic lymphocytic leukemia, colon cancer, melanoma, HNSCC, and glioblastoma, showed the induction of antitumor cytotoxic T cell populations, and reduced regulatory T cell populations in response to anti-CD47 treatment [6][35][38][39][40]. These observations were replicated in ex vivo studies. For example, Tao and colleagues assessed tumor samples from esophageal squamous cell cancer patients, showing an inverse relationship between CD8 T cell density and CD47 expression. In mice models with esophageal squamous cell cancer, treatment with anti-CD47 antibodies led to an increase in PD-1 and CTLA-4 expression. Treatment with the combination of CD47, PD-1 and CTLA-4 inhibitors yielded significantly improved survival in mice, compared with anti-CD47 monotherapy or PD-1 and CTLA-4 inhibitor combination, suggesting a rationale for combinatory therapeutic approaches to obtain synergistic effects [41].
Figure 1. CD47/SIRPα interaction leading to repression of phagocytosis and therapeutic approaches blocking CD47/SIRPα axis.
Clinical implications of CD47 overexpression were also studied in various cancer types with the majority showing an inverse relationship between CD47 overexpression and clinical outcomes [42]. Chao et al. used flow cytometry and found that NHL cells had two-fold greater CD47 expression than normal germinal center and peripheral blood B cells. Grouping patient samples based on CD47 mRNA expression levels, investigators showed improved overall survival in patients with CD47 low tumors, especially diffuse large B cell lymphoma (DLBCL), B cell chronic lymphocytic leukemia, and mantle cell lymphoma subsets [43]. Majeti and colleagues, showed high CD47 expression by gene expression arrays and flow cytometry in leukemia stem cells, compared with normal counterparts in a group of 137 AML patients. Compared with those with low CD47 expression, patients with high CD47 expression had significantly worse overall survival rates (22.1 vs 9.1 months, hazard ratio (HR): 2.02) and event free survival (17.1 vs 6.8 months, HR 1.94) [12]. Analyzing immunohistochemistry staining of CD47 in bone marrow biopsy samples from 248 AML patients, Galli et al. detected high CD47 staining in one-fourth of the patient samples. Samples with high CD47 staining had higher median blast count, median bone marrow infiltration, and disease burden. Although there was a trend towards unfavorable progression free survival in patients with high CD47, no statistical difference was observed in median progression-free survival, or overall survival [44]. Melanoma patients with tumors bearing CD47 overexpression were found to have worse overall survival rates and higher rates of distant metastasis [45]. Similarly, head and neck cancer patients with tumors bearing robust CD47 immunohistochemistry staining had diminished overall survival, compared with those with low CD47 staining [6]. A study of ovarian cancer demonstrated that increased CD47 expression is associated with worse prognosis, increased migration and invasion, and the induction of epithelial-mesenchymal transition [46].

3. Therapies Targeting CD47/SIRPα in Cancer

As a result of the promising preclinical data regarding the anti-tumor activity of CD47/SIPRα blockade obtained from in vivo and in vitro studies, several molecules have been developed and are undergoing clinical testing. Functionally, therapeutics under investigation may be classified as (1) CD47 targeting agents, (2) SIRPα targeting agents and (3) bispecific targeting agents. Table 1 provides a comprehensive list of the ongoing clinical trials of the CD47/SIRPα targeting therapeutics at the time of this publication. Although most of those approaches are currently being tested in early-phase clinical trials to assess safety and tolerability, available data from a number of published studies has revealed promising activity and favorable tolerability. In addition to being tested on their own, trials of combinations with other anti-tumor agents are underway. Inspired by the fact that CD47/SIRPα signaling limits the efficacy of tumor-opsonizing antibodies, a number of clinical trials are evaluating agents targeting this axis in combination with agents such as rituximab, cetuximab and trastuzumab [10][43][47]. Histone deacetylase (HDAC) inhibitors have been shown to enhance checkpoint inhibitor therapy by decreasing immune suppressive cells and increasing tumor antigen presentation [48][49]. Given the possible enhancement of tumor immunity, combinations of HDAC inhibitors and CD47 targeting therapies are underway. Other strategies employ a combination of CD47 targeted therapies with immune checkpoint inhibitors and chemotherapies.
Table 1. Clinical trials testing agents targeting CD47/SIRPα axis.
Agent Therapeutic
Target
Design Phase Disease Site Accrual Goal Identifier
Monoclonal Antibodies
IBI188 (Letaplimab) CD47 IBI188 +/− rituximab I Metastatic solid tumors or lymphoma 92 NCT03717103
IBI188 +/− azacitidine I Myelodysplastic syndrome 12 NCT04485065
Hu5F9-G4 (Magrolimab) CD47 Hu5F9-G4 (Magrolimab) + Pembrolizumab II Hodgkin’s lymphoma 24 NCT04788043
Hu5F9-G4 (Magrolimab) I Hematologic malignancies 20 NCT02678338
Hu5F9-G4 (Magrolimab) + acalabrutinib + rituximab or other combinations without Hu5F9-G4 (Magrolimab) I Non-Hodgkin’s Lymphoma 30 NCT03527147
Hu5F9-G4 (Magrolimab) + Obinutuzumab + venetoclax I Non-Hodgkin’s Lymphoma 76 NCT04599634
ZL-1201 CD47 ZL-1201 I Metastatic solid tumors or refractory lymphomas 66 NCT04257617
STI-6643 CD47 STI-6643 I Metastatic solid tumors 24 NCT04900519
CC-9002 CD47 CC-90002 +/−rituximab   Part A: Metastatic solid tumors, multiple Myeloma or non-Hodgkin’s lymphoma
Part B, relapsed and/or refractory CD20-positive NHL
60 NCT02367196
AK117 CD47 AK117 I Metastatic solid tumors or lymphoma 162 NCT04728334
AK117 + azacitidine I/II Myelodysplastic syndrome 190 NCT04900350
AO-176 CD47 AO-176 +/− paclitaxel I/II Metastatic solid tumors 132 NCT03834948
AO-176 +/− dexamethasone or dexhamethasone + bortezomide I Multiple myeloma 102 NCT04445701
IMC-002 CD47 IMC-002 I Metastatic solid tumors or lymphoma 24 NCT04306224
TQB2928 CD47 TQB2928 I Metastatic solid tumors or hematologic malignancies 20 NCT04854681
FSI-189 SIRPα FSI-189 +/− rituximab I Non-Hodgkin’s lymphoma (B-cell) 63 NCT04502706
BI 765063 SIRPα BI 765063 +/− PD-1 inhibitor I Metastatic solid tumors with SIRPα polymorphism 116 NCT03990233
Bispecific antibodies
HX009 CD47 and PD-1 HX009 II Metastatic solid tumors 210 NCT04886271
PF-07257876 CD47 and PD-L1 PF-07257876 I Non small-cell lung cancer, head and neck squamous cell carcinoma, ovarian cancer 90 NCT04881045
CPO107 (JMP601) CD47 and CD20 CPO107 (JMP601) I Non-Hodgkin’s lymphoma (CD-20 positive) 75 NCT04853329
IBI322 CD47 and PD-L1 IBI322 I Hematologic malignancies 182 NCT04795128
IBI322 Ia Metastatic solid tumors 45 NCT04338659
IBI322 Ia/Ib Metastatic solid tumors 218 NCT04328831
SL-172154 SIRPα and CD40L SL-172154 (intravenous) I Ovarian cancer 40 NCT04406623
SL-172154 (intratumoral) I Head and neck or cutaneous squamous cell carcinoma 18 NCT04502888
TG-1801 CD47 and CD19 TG-1801 +/− ubitixumab Ib Hematologic malignancies 60 NCT04806035
IMM0306 CD47 and CD20 IMM0306 I Refractory or Relapsed CD20-positive B cell Non-Hodgkin’s Lymphoma 131 NCT04746131
Fusion proteins
TTI-622 CD47 via SIRPαFc (IgG4) structure TTI-622 + rituximab, PD-1 inhibitor, Proteasome inhibitor regimen or rituximab Ia/Ib Lymphoma or myeloma 156 NCT03530683
ALX148 CD47 via SIRPαFc (IgG1) structure ALX148 + azacitidine I/II Myelodysplastic syndrome 173 NCT04417517
ALX148 + venetoclax or azacitidine I/II Acute myleoid leukemia 97 NCT04755244
ALX148 II Head and neck squamous cell carcinoma 112 NCT04675333
ALX148 + pembrolizumab II Head and neck squamous cell carcinoma 111 NCT04675294
 
 
Major concerns regarding the use of CD47-targeted agents are driven by the ubiquitous expression of CD47, which leads to rapid drug elimination, “antigen sink” and hematologic toxicity, such as anemia and thrombocytopenia [50]. The impact on hematopoietic cells, particularly red blood cells, presents a substantial issue with CD47/SIRPα targeted drugs. Given that older red blood cells are more sensitive to phagocytosis, red blood cell destruction remains a limiting toxicity with these drugs, and may influence the age of patients that can be treated with these agents [51]. Notably, as opposed to red blood cells, other normal functioning cells are less vulnerable to macrophage-mediated immune destruction with anti-CD47 therapies, as further activation of prophagocytic signals and involvement of calreticulin are suggested to be necessary steps to generate immune-related adverse events [52][53]. Furthermore, the use of CD47 or SIPRα-directed antibodies structured with an intact Fc portion raises similar concerns, due to the widespread expression of Fc receptors on normal cells, and the risks for potentially causing immune destruction of “self” cells [25]. From the drug development standpoint, the efficacy of the CD47/SIRPα blockade can vary based upon drug delivery method and compartmentalization of the drug. In addition, tumor type and stage, tumor immune microenvironment and acquired drug resistance, constitute other potential determinants of the efficacy of CD47/SIRPα inhibitors [54].

4. Anti-CD47 Antibodies and CD47-Targeting Recombinant Proteins

CD47-directed monoclonal antibodies and fusion proteins with SIRPα immunoglobulin structure competitively bind CD47 and block the interaction between CD47 and SIRPα. This class of therapeutics constitute the majority of the available in-human data testing CD47/SIRPα inhibition in solid tumors and hematologic malignancies, although data remains limited.
Hu5F9-G4 (5F9, magrolimab) is a humanized antibody with an IgG4 Fc fragment [55]. In a preclinical setting, magrolimab demonstrated anti-tumor activity against AML in-vitro and in vivo. Furthermore, complete disease elimination was observed in human B lymphoblastoid cell-engrafted mice, after treatment with magrolimab in combination with rituximab [55]. Preclinical models testing magrolimab in solid tumors such as colon, liver, ovarian and breast cancers demonstrated promising anti-tumor activity [56]. Another study in which patient-derived NHL xenografted mice were treated with magrolimab/rituximab combination showed an 89% cure rate, defined as over 4 months of disease free survival following the discontinuation of therapy [43]. In-depth analyses suggested that rituximab plays a complementary role in further stimulation of innate immunity, via its active Fc effector function-inducing natural killer cell and macrophage-mediated cellular cytotoxicity. Accordingly, the data from a phase Ib study of 22 patients with relapsed or refractory NHL, 95% of whom were previously treated with rituximab, demonstrated encouraging outcomes with an objective response rate of 50%, and a complete response rate of 36%, with magrolimab and rituximab in combination [57]. Adverse events experienced by patients on trial included chills, anemia and headaches (41% each), all of which occurred only in the first weeks of the trial. There were no significant safety signals in the latter stages of the trial. A simultaneously conducted phase I study of single agent magrolimab in metastatic solid tumors demonstrated a similar safety profile, with transient treatment-related adverse events [58]. Of note, trends in anemia development and transfusion requirements with magrolimab were further examined using the data from the patient population in the phase I dose escalation part of these studies [59]. Patients on escalating doses of magrolimab experienced a median 1.0 g/dL decrease in hemoglobin levels, and subsequent doses were associated with a lesser degree of hemoglobin decline. Red blood cell transfusion yielded appropriate responses in hemoglobin concentration, supporting the evidence regarding the transient nature of anemia after magrolimab administration [59]. A number of clinical trials evaluating magrolimab, either as a single agent or in combination with cytotoxic therapies, targeted therapies or immune checkpoint inhibitors to treat hematologic neoplasms, are ongoing.
Other CD-47-targeting monoclonal antibodies that have entered clinical development include IBI188 (letaplimab), AK117 and SRF231. A phase I study of letaplimab in patients with advanced solid tumors and lymphomas was recently completed. Letaplimab demonstrated a favorable toxicity profile, with no dose-limiting toxicities. The majority of the treatment-related adverse events were grade 1–2. The rate of anemia was 15%, and only one patient developed grade 3 anemia. Notably, infusion related reactions were seen in 65% of the patient population, but all were grade 1–2 and manageable with a standard infusion-related reaction treatment algorithm [60]. AK117 monotherapy in patients with metastatic solid tumors demonstrated safety with no dose-limiting toxicities, no infusion-related reactions, or grade ≥ 3 treatment-related adverse events observed with up to 20 mg/kg dosing. Further dose escalation is underway with 30 mg/kg dosing [61]. For SRF231, further exploration in clinical trials was held by the pharmaceutical company.

References

  1. Mawby, W.J.; Holmes, C.H.; Anstee, D.J.; Spring, F.A.; Tanner, M.J.A. Isolation and Characterization of CD47 Glycoprotein: A Multispanning Membrane Protein Which Is the Same as Integrin-Associated Protein (IAP) and the Ovarian Tumour Marker OA3. Biochem. J. 1994, 304, 525–530.
  2. Jaiswal, S.; Jamieson, C.H.M.; Pang, W.W.; Park, C.Y.; Chao, M.P.; Majeti, R.; Traver, D.; van Rooijen, N.; Weissman, I.L. CD47 Is Upregulated on Circulating Hematopoietic Stem Cells and Leukemia Cells to Avoid Phagocytosis. Cell 2009, 138, 271–285.
  3. Oldenborg, P.A.; Zheleznyak, A.; Fang, Y.F.; Lagenaur, C.F.; Gresham, H.D.; Lindberg, F.P. Role of CD47 as a Marker of Self on Red Blood Cells. Science 2000, 288, 2051–2054.
  4. Burger, P.; Hilarius-Stokman, P.; de Korte, D.; van den Berg, T.K.; van Bruggen, R. CD47 Functions as a Molecular Switch for Erythrocyte Phagocytosis. Blood 2012, 119, 5512–5521.
  5. Matlung, H.L.; Szilagyi, K.; Barclay, N.A.; van den Berg, T.K. The CD47-SIRPα Signaling Axis as an Innate Immune Checkpoint in Cancer. Immunol. Rev. 2017, 276, 145–164.
  6. Wu, L.; Yu, G.-T.; Deng, W.-W.; Mao, L.; Yang, L.-L.; Ma, S.-R.; Bu, L.-L.; Kulkarni, A.B.; Zhang, W.-F.; Zhang, L.; et al. Anti-CD47 Treatment Enhances Anti-Tumor T-Cell Immunity and Improves Immunosuppressive Environment in Head and Neck Squamous Cell Carcinoma. OncoImmunology 2018, 7, e1397248.
  7. Chao, M.P.; Tang, C.; Pachynski, R.K.; Chin, R.; Majeti, R.; Weissman, I.L. Extranodal Dissemination of Non-Hodgkin Lymphoma Requires CD47 and Is Inhibited by Anti-CD47 Antibody Therapy. Blood 2011, 118, 4890–4901.
  8. Willingham, S.B.; Volkmer, J.-P.; Gentles, A.J.; Sahoo, D.; Dalerba, P.; Mitra, S.S.; Wang, J.; Contreras-Trujillo, H.; Martin, R.; Cohen, J.D.; et al. The CD47-Signal Regulatory Protein Alpha (SIRPa) Interaction Is a Therapeutic Target for Human Solid Tumors. Proc. Natl. Acad. Sci. USA 2012, 109, 6662–6667.
  9. Molecular Pathways: Activating T Cells after Cancer Cell Phagocytosis from Blockade of CD47 “Don’t Eat Me” Signals|Clinical Cancer Research. Available online: https://clincancerres.aacrjournals.org/content/21/16/3597 (accessed on 24 August 2021).
  10. Weiskopf, K.; Jahchan, N.S.; Schnorr, P.J.; Cristea, S.; Ring, A.M.; Maute, R.L.; Volkmer, A.K.; Volkmer, J.-P.; Liu, J.; Lim, J.S.; et al. CD47-Blocking Immunotherapies Stimulate Macrophage-Mediated Destruction of Small-Cell Lung Cancer. J. Clin. Investig. 2016, 126, 2610–2620.
  11. Edris, B.; Weiskopf, K.; Volkmer, A.K.; Volkmer, J.-P.; Willingham, S.B.; Contreras-Trujillo, H.; Liu, J.; Majeti, R.; West, R.B.; Fletcher, J.A.; et al. Antibody Therapy Targeting the CD47 Protein Is Effective in a Model of Aggressive Metastatic Leiomyosarcoma. Proc. Natl. Acad. Sci. USA 2012, 109, 6656–6661.
  12. Majeti, R.; Chao, M.P.; Alizadeh, A.A.; Pang, W.W.; Jaiswal, S.; Gibbs, K.D.; van Rooijen, N.; Weissman, I.L. CD47 Is an Adverse Prognostic Factor and Therapeutic Antibody Target on Human Acute Myeloid Leukemia Stem Cells. Cell 2009, 138, 286–299.
  13. Xu, L.; Wang, S.; Li, J.; Li, B. CD47/SIRPα Blocking Enhances CD19/CD3-Bispecific T Cell Engager Antibody-Mediated Lysis of B Cell Malignancies. Biochem. Biophys. Res. Commun. 2019, 509, 739–745.
  14. Yoshida, K.; Tsujimoto, H.; Matsumura, K.; Kinoshita, M.; Takahata, R.; Matsumoto, Y.; Hiraki, S.; Ono, S.; Seki, S.; Yamamoto, J.; et al. CD47 Is an Adverse Prognostic Factor and a Therapeutic Target in Gastric Cancer. Cancer Med. 2015, 4, 1322–1333.
  15. Xiao, Z.; Chung, H.; Banan, B.; Manning, P.T.; Ott, K.C.; Lin, S.; Capoccia, B.J.; Subramanian, V.; Hiebsch, R.R.; Upadhya, G.A.; et al. Antibody Mediated Therapy Targeting CD47 Inhibits Tumor Progression of Hepatocellular Carcinoma. Cancer Lett. 2015, 360, 302–309.
  16. Ring, N.G.; Herndler-Brandstetter, D.; Weiskopf, K.; Shan, L.; Volkmer, J.-P.; George, B.M.; Lietzenmayer, M.; McKenna, K.M.; Naik, T.J.; McCarty, A.; et al. Anti-SIRPα Antibody Immunotherapy Enhances Neutrophil and Macrophage Antitumor Activity. Proc. Natl. Acad. Sci. USA 2017, 114, E10578–E10585.
  17. Vaeteewoottacharn, K.; Kariya, R.; Pothipan, P.; Fujikawa, S.; Pairojkul, C.; Waraasawapati, S.; Kuwahara, K.; Wongkham, C.; Wongkham, S.; Okada, S. Attenuation of CD47-SIRPα Signal in Cholangiocarcinoma Potentiates Tumor-Associated Macrophage-Mediated Phagocytosis and Suppresses Intrahepatic Metastasis. Transl. Oncol. 2018, 12, 217–225.
  18. Solinas, G.; Germano, G.; Mantovani, A.; Allavena, P. Tumor-Associated Macrophages (TAM) as Major Players of the Cancer-Related Inflammation. J. Leukoc. Biol. 2009, 86, 1065–1073.
  19. Gordon, S. Alternative Activation of Macrophages. Nat. Rev. Immunol. 2003, 3, 23–35.
  20. Mantovani, A.; Marchesi, F.; Malesci, A.; Laghi, L.; Allavena, P. Tumour-Associated Macrophages as Treatment Targets in Oncology. Nat. Rev. Clin. Oncol. 2017, 14, 399–416.
  21. Zhang, M.; Hutter, G.; Kahn, S.A.; Azad, T.D.; Gholamin, S.; Xu, C.Y.; Liu, J.; Achrol, A.S.; Richard, C.; Sommerkamp, P.; 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.
  22. Nath, P.R.; Pal-Nath, D.; Mandal, A.; Cam, M.C.; Schwartz, A.L.; Roberts, D.D. Natural Killer Cell Recruitment and Activation Are Regulated by CD47 Expression in the Tumor Microenvironment. Cancer Immunol. Res. 2019, 7, 1547–1561.
  23. Deuse, T.; Hu, X.; Agbor-Enoh, S.; Jang, M.K.; Alawi, M.; Saygi, C.; Gravina, A.; Tediashvili, G.; Nguyen, V.Q.; Liu, Y.; et al. The SIRPα–CD47 Immune Checkpoint in NK Cells. J. Exp. Med. 2021, 218, e20200839.
  24. Kim, M.J.; Lee, J.-C.; Lee, J.-J.; Kim, S.; Lee, S.G.; Park, S.-W.; Sung, M.W.; Heo, D.S. Association of CD47 with Natural Killer Cell-Mediated Cytotoxicity of Head-and-Neck Squamous Cell Carcinoma Lines. Tumor Biol. 2008, 29, 28–34.
  25. Veillette, A.; Chen, J. SIRPα–CD47 Immune Checkpoint Blockade in Anticancer Therapy. Trends Immunol. 2018, 39, 173–184.
  26. Kikuchi, Y.; Uno, S.; Kinoshita, Y.; Yoshimura, Y.; Iida, S.-I.; Wakahara, Y.; Tsuchiya, M.; Yamada-Okabe, H.; Fukushima, N. Apoptosis Inducing Bivalent Single-Chain Antibody Fragments against CD47 Showed Antitumor Potency for Multiple Myeloma. Leuk Res. 2005, 29, 445–450.
  27. Boukhari, A.; Alhosin, M.; Bronner, C.; Sagini, K.; Truchot, C.; Sick, E.; Schini-Kerth, V.B.; André, P.; Mély, Y.; Mousli, M.; et al. CD47 Activation-Induced UHRF1 over-Expression Is Associated with Silencing of Tumor Suppressor Gene P16INK4A in Glioblastoma Cells. Anticancer Res. 2015, 35, 149–157.
  28. Uluçkan, O.; Becker, S.N.; Deng, H.; Zou, W.; Prior, J.L.; Piwnica-Worms, D.; Frazier, W.A.; Weilbaecher, K.N. CD47 Regulates Bone Mass and Tumor Metastasis to Bone. Cancer Res. 2009, 69, 3196–3204.
  29. Huang, T.; Sun, L.; Yuan, X.; Qiu, H. Thrombospondin-1 Is a Multifaceted Player in Tumor Progression. Oncotarget 2017, 8, 84546–84558.
  30. Byrne, G.J.; Hayden, K.E.; McDowell, G.; Lang, H.; Kirwan, C.C.; Tetlow, L.; Kumar, S.; Bundred, N.J. Angiogenic Characteristics of Circulating and Tumoural Thrombospondin-1 in Breast Cancer. Int. J. Oncol. 2007, 31, 1127–1132.
  31. Borsotti, P.; Ghilardi, C.; Ostano, P.; Silini, A.; Dossi, R.; Pinessi, D.; Foglieni, C.; Scatolini, M.; Lacal, P.M.; Ferrari, R.; et al. Thrombospondin-1 Is Part of a Slug-Independent Motility and Metastatic Program in Cutaneous Melanoma, in Association with VEGFR-1 and FGF-2. Pigment. Cell Melanoma Res. 2015, 28, 73–81.
  32. Kamijo, H.; Miyagaki, T.; Takahashi-Shishido, N.; Nakajima, R.; Oka, T.; Suga, H.; Sugaya, M.; Sato, S. Thrombospondin-1 Promotes Tumor Progression in Cutaneous T-Cell Lymphoma via CD47. Leukemia 2020, 34, 845–856.
  33. Liu, X.; Pu, Y.; Cron, K.; Deng, L.; Kline, J.; Frazier, W.A.; Xu, H.; Peng, H.; Fu, Y.-X.; Xu, M.M. CD47 Blockade Triggers T Cell-Mediated Destruction of Immunogenic Tumors. Nat. Med. 2015, 21, 1209–1215.
  34. Chen, A.; Harrabi, O.; Fong, A.P.; Ruffner, K.L.; Forgie, A.J.; Sim, J.; Randolph, S.S.; Wan, H.; Pons, J.; Kuo, T.C. ALX148 Enhances the Depth and Durability of Response to Multiple AML Therapies. Blood 2020, 136, 15–16.
  35. Tseng, D.; Volkmer, J.-P.; Willingham, S.B.; Contreras-Trujillo, H.; Fathman, J.W.; Fernhoff, N.B.; Seita, J.; Inlay, M.A.; Weiskopf, K.; Miyanishi, M.; et al. Anti-CD47 Antibody-Mediated Phagocytosis of Cancer by Macrophages Primes an Effective Antitumor T-Cell Response. Proc. Natl. Acad. Sci. USA 2013, 110, 11103–11108.
  36. Yang, H.; Shao, R.; Huang, H.; Wang, X.; Rong, Z.; Lin, Y. Engineering Macrophages to Phagocytose Cancer Cells by Blocking the CD47/SIRPɑ Axis. Cancer Med. 2019, 8, 4245–4253.
  37. Gauttier, V.; Pengam, S.; Durand, J.; Biteau, K.; Mary, C.; Morello, A.; Néel, M.; Porto, G.; Teppaz, G.; Thepenier, V.; et al. Selective SIRPα Blockade Reverses Tumor T Cell Exclusion and Overcomes Cancer Immunotherapy Resistance. J. Clin. Investig. 2020, 130, 6109–6123.
  38. Soto-Pantoja, D.R.; Terabe, M.; Ghosh, A.; Ridnour, L.A.; DeGraff, W.G.; Wink, D.A.; Berzofsky, J.A.; Roberts, D.D. CD47 in the Tumor Microenvironment Limits Cooperation between Antitumor T-Cell Immunity and Radiotherapy. Cancer Res. 2014, 74, 6771–6783.
  39. Von Roemeling, C.A.; Wang, Y.; Qie, Y.; Yuan, H.; Zhao, H.; Liu, X.; Yang, Z.; Yang, M.; Deng, W.; Bruno, K.A.; et al. Therapeutic Modulation of Phagocytosis in Glioblastoma Can Activate Both Innate and Adaptive Antitumour Immunity. Nat. Commun. 2020, 11, 1508.
  40. Martinez-Torres, A.-C.; Quiney, C.; Attout, T.; Boullet, H.; Herbi, L.; Vela, L.; Barbier, S.; Chateau, D.; Chapiro, E.; Nguyen-Khac, F.; et al. CD47 Agonist Peptides Induce Programmed Cell Death in Refractory Chronic Lymphocytic Leukemia B Cells via PLCγ1 Activation: Evidence from Mice and Humans. PLoS Med. 2015, 12, e1001796.
  41. Tao, H.; Qian, P.; Wang, F.; Yu, H.; Guo, Y. Targeting CD47 Enhances the Efficacy of Anti-PD-1 and CTLA-4 in an Esophageal Squamous Cell Cancer Preclinical Model. Oncol. Res. 2017, 25, 1579–1587.
  42. Nagahara, M.; Mimori, K.; Kataoka, A.; Ishii, H.; Tanaka, F.; Nakagawa, T.; Sato, T.; Ono, S.; Sugihara, K.; Mori, M. Correlated Expression of CD47 and SIRPA in Bone Marrow and in Peripheral Blood Predicts Recurrence in Breast Cancer Patients. Clin. Cancer Res. 2010, 16, 4625–4635.
  43. Chao, M.P.; Alizadeh, A.A.; Tang, C.; Myklebust, J.H.; Varghese, B.; Gill, S.; Jan, M.; Cha, A.C.; Chan, C.K.; Tan, B.T.; et al. Anti-CD47 Antibody Synergizes with Rituximab to Promote Phagocytosis and Eradicate Non-Hodgkin Lymphoma. Cell 2010, 142, 699–713.
  44. Galli, S.; Zlobec, I.; Schürch, C.; Perren, A.; Ochsenbein, A.F.; Banz, Y. CD47 Protein Expression in Acute Myeloid Leukemia: A Tissue Microarray-Based Analysis. Leuk Res. 2015, 39, 749–756.
  45. Fu, W.; Li, J.; Zhang, W.; Li, P. High Expression of CD47 Predicts Adverse Prognosis in Chinese Patients and Suppresses Immune Response in Melanoma. Biomed. Pharmacother. 2017, 93, 1190–1196.
  46. Overexpression of CD47 Predicts Poor Prognosis and Promotes Cancer Cell Invasion in High-Grade Serous Ovarian Carcinoma. Available online: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5489890/ (accessed on 24 August 2021).
  47. Upton, R.; Banuelos, A.; Feng, D.; Biswas, T.; Kao, K.; McKenna, K.; Willingham, S.; Ho, P.Y.; Rosental, B.; Tal, M.C.; et al. Combining CD47 Blockade with Trastuzumab Eliminates HER2-Positive Breast Cancer Cells and Overcomes Trastuzumab Tolerance. Proc. Natl. Acad. Sci. USA 2021, 118, e2026849118.
  48. Briere, D.; Sudhakar, N.; Woods, D.M.; Hallin, J.; Engstrom, L.D.; Aranda, R.; Chiang, H.; Sodré, A.L.; Olson, P.; Weber, J.S.; et al. The Class I/IV HDAC Inhibitor Mocetinostat Increases Tumor Antigen Presentation, Decreases Immune Suppressive Cell Types and Augments Checkpoint Inhibitor Therapy. Cancer Immunol. Immunother. 2018, 67, 381–392.
  49. Orillion, A.; Hashimoto, A.; Damayanti, N.; Shen, L.; Adelaiye-Ogala, R.; Arisa, S.; Chintala, S.; Ordentlich, P.; Kao, C.; Elzey, B.; et al. Entinostat Neutralizes Myeloid-Derived Suppressor Cells and Enhances the Antitumor Effect of PD-1 Inhibition in Murine Models of Lung and Renal Cell Carcinoma. Clin. Cancer Res. 2017, 23, 5187–5201.
  50. Catani, L.; Sollazzo, D.; Ricci, F.; Polverelli, N.; Palandri, F.; Baccarani, M.; Vianelli, N.; Lemoli, R.M. The CD47 Pathway Is Deregulated in Human Immune Thrombocytopenia. Exp. Hematol. 2011, 39, 486–494.
  51. Khandelwal, S.; van Rooijen, N.; Saxena, R.K. Reduced Expression of CD47 during Murine Red Blood Cell (RBC) Senescence and Its Role in RBC Clearance from the Circulation. Transfusion 2007, 47, 1725–1732.
  52. Takimoto, C.H.; Chao, M.P.; Gibbs, C.; McCamish, M.A.; Liu, J.; Chen, J.Y.; Majeti, R.; Weissman, I.L. The Macrophage ‘Do Not Eat Me’ Signal, CD47, Is a Clinically Validated Cancer Immunotherapy Target. Ann. Oncol. 2019, 30, 486–489.
  53. Chao, M.P.; Majeti, R.; Weissman, I.L. Programmed Cell Removal: A New Obstacle in the Road to Developing Cancer. Nat. Rev. Cancer 2012, 12, 58–67.
  54. Zhang, W.; Huang, Q.; Xiao, W.; Zhao, Y.; Pi, J.; Xu, H.; Zhao, H.; Xu, J.; Evans, C.E.; Jin, H. Advances in Anti-Tumor Treatments Targeting the CD47/SIRPα Axis. Front. Immunol. 2020, 11, 18.
  55. Liu, J.; Wang, L.; Zhao, F.; Tseng, S.; Narayanan, C.; Shura, L.; Willingham, S.; Howard, M.; Prohaska, S.; Volkmer, J.; et al. Pre-Clinical Development of a Humanized Anti-CD47 Antibody with Anti-Cancer Therapeutic Potential. PLoS ONE 2015, 10, e0137345.
  56. Gholamin, S.; Mitra, S.S.; Feroze, A.H.; Liu, J.; Kahn, S.A.; Zhang, M.; Esparza, R.; Richard, C.; Ramaswamy, V.; Remke, M.; et al. Disrupting the CD47-SIRPα Anti-Phagocytic Axis by a Humanized Anti-CD47 Antibody Is an Efficacious Treatment for Malignant Pediatric Brain Tumors. Sci. Transl. Med. 2017, 9, eaaf2968.
  57. Advani, R.; Flinn, I.; Popplewell, L.; Forero, A.; Bartlett, N.L.; Ghosh, N.; Kline, J.; Roschewski, M.; LaCasce, A.; Collins, G.P.; et al. CD47 Blockade by Hu5F9-G4 and Rituximab in Non-Hodgkin’s Lymphoma. N. Engl. J. Med. 2018, 379, 1711–1721.
  58. Sikic, B.I.; Lakhani, N.; Patnaik, A.; Shah, S.A.; Chandana, S.R.; Rasco, D.; Colevas, A.D.; O’Rourke, T.; Narayanan, S.; Papadopoulos, K.; et al. First-in-Human, First-in-Class Phase I Trial of the Anti-CD47 Antibody Hu5F9-G4 in Patients with Advanced Cancers. J. Clin. Oncol. 2019, 37, 946–953.
  59. Brierley, C.K.; Staves, J.; Roberts, C.; Johnson, H.; Vyas, P.; Goodnough, L.T.; Murphy, M.F. The Effects of Monoclonal Anti-CD47 on RBCs, Compatibility Testing, and Transfusion Requirements in Refractory Acute Myeloid Leukemia. Transfusion 2019, 59, 2248–2254.
  60. Lakhani, N.; Orloff, M.; Fu, S.; Liu, Y.; Wang, Y.; Zhou, H.; Lin, K.; Liu, F.; Yan, S.; Patnaik, A. 295 First-in-Human Phase I Trial of IBI188, an Anti-CD47 Targeting Monoclonal Antibody, in Patients with Advanced Solid Tumors and Lymphomas. J. Immunother. Cancer 2020, 8.
  61. Gan, H.K.; Coward, J.; Mislang, A.R.A.; Cosman, R.; Nagrial, A.; Jin, X.; Li, B.; Wang, Z.M.; Kwek, K.Y.; Xia, D.; et al. Safety of AK117, an Anti-CD47 Monoclonal Antibody, in Patients with Advanced or Metastatic Solid Tumors in a Phase I Study. J. Clin. Oncol. 2021, 39, 2630.
More
Information
Subjects: Oncology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 783
Revisions: 2 times (View History)
Update Date: 29 Mar 2022
1000/1000
Video Production Service