0102030405060708
Literature Analysis| Dual CD47 and PD-L1 Blockade Boosts CD8+ T Cell Anti-Tumor Response
2025-04-07
On November 22, 2024, Susan N. Christo et al. published this paper in the Clin Transl Immunology journal, which mainly focused on the dual antibody targets of CD47 and PD-L1. The main content is as follows: Immune checkpoint blockade (ICB) plays an important role in cancer treatment, and therapies targeting PD-1/PD-L1 have been shown to inhibit tumor growth. However, they have problems with low effectiveness and short persistence. Most tumor regression is incomplete, while antibodies targeting CD47 can reduce tumor burden. However, anti-CD47 therapy may cause serious clinical side effects such as red blood cell toxicity and anemia. Therefore, dual-target antibody therapy is more effective in enhancing anti-cancer T cells than current single-target therapy. The authors used CD47 × PD-L1 bispecific antibodies to reduce the affinity of antibodies to CD47, thereby reducing binding to red blood cells while retaining binding to PD-L1-expressing cells in the tumor. The research results show that the aim is to clarify that CD47 × PD-L1 BisAb can elicit stronger CD8+ T cell responses than standard anti-PD-L1 monotherapy. This emphasizes the clinical potential of CD47 × PD-L1 bispecific antibodies.
Background and Knowledge SupplementationLITERATURE
The function of an immune checkpoint is to prevent excessive immune damage to normal cells. When the recognition proteins on the surface of immune cells (such as T cells) bind to partner proteins of tumor cells, the immune system will organize the immune system to kill tumor cells. In the presence of immunosuppressants, the binding of partner proteins to T cells will be blocked, and this process of T cells killing tumor cells is called immune checkpoint blockade therapy. At present, the cancers that can be treated by the blocking method of immune examination include breast cancer, bladder cancer, cervical cancer, and other solid tumors. CTLA-4 and PD-1 are the first checkpoint receptors found. The blocking receptor treatment provides a prospect for expanding cancer therapy in clinical practice. The first blocking drug of the immune checkpoint that enters the oncology clinical trials is the anti-CTLA-4 monoclonal antibody ipilimumab (full human IgG1) and tremelimumab (full human IgG2). With the deepening of research, anti-PD-1 drugs have more extensive anti-tumor activities.
Tab 1: Drugs in clinical development that block PD-1 or PD-L1
Target | Drug Name | Other Names | Isotype and Characteristics |
---|---|---|---|
PD-1 | MEDI0680 | AMP-514 | Information not available |
Nivolumab | Opdivo, BMS-936558, MDX-1106, ONO-4538 | Fully human IgG4a | |
Pembrolizumab | Keytruda, MK-3475, lambrolizumab | Humanized IgG4 | |
Pidilizumab | CT-011 | Humanized IgG1 | |
PD-L1 | BMS-936559 | MDX-1105 | Fully human IgG4a |
MEDI4736 | None | Fc-modified human IgG1b | |
MPDL3280A | RG7446 | Fc-modified human IgG1b | |
MSB0010718C | None | Fully human IgG1a |
Send Inquiry
Contact Us For Best Would you Like to Know more We can Give you the answer, For inquiries about our products and services. please leave your e-mail to us and will reply within 24 hours.
Click for inquiryAs research deepens, some potential immune checkpoints have emerged, such as lymphocyte activation gene 3 (LAG-3) (anti-LAG-3 monoclonal antibody or in combination with anti-PD-1 therapy), killer inhibitory receptor (KIR) (alone or in combination with anti-PD-1 or anti-CTLA-4 therapy), B7-H3, T cell immunoglobulin and mucin-3 (TIM-3), T cell immunoglobulin and immune tyrosine inhibitory motif (ITIM) domain (TIGIT), and other immune checkpoint related drugs are also being studied, broadening the path for cancer treatment.
CD8+ T Cell Introduction
The cytotoxic T cell, also known as CD8+ T cell, is a T cell receptor (TCR) that can kill cancer cells, cells infected by viruses, bacteria, or other forms of damage. It recognizes specific antigens. The antigens inside the cell bind to MHCI molecules and are carried to the cell surface for recognition by T cells. If TCR is specific to the antigen, it will bind to the complex, and T cells will destroy the cell. If the antigen inside the cell binds to MHC class II molecules, it is a CD4+ T cell.

PD-1/PD-L1 Target Introduction
Programmed cell death protein 1 (PD-1), encoded by the PDCD1 gene, is a cell surface receptor on T and B cells. PD-1 is an immune checkpoint that regulates T cell activity by promoting antigen-specific T cell apoptosis in lymph nodes and inhibiting regulatory T cell apoptosis. The expression of PD-L1 on tumor cells inhibits anti-tumor activity by binding to PD-1 on effector T cells, inducing cell apoptosis, and weakening the host's immune response to tumor cells, thereby promoting tumor cell growth and causing immune evasion of tumor cells. Antigen-presenting cells (APCs) present tumor antigens to T cell receptors (TCRs) through the major histocompatibility complex (MHC). When the MHC antigen complex specifically binds to the TCR, it triggers a series of signal transduction pathways, including phosphatidylinositol signaling, mitogen-activated protein kinase signaling pathways, etc., thereby activating the immune response of effector T cells. After PD-L1 binds to PD-1, the cytoplasmic region of PD-1 undergoes phosphorylation, thereby activating SHP2. Subsequently, SHP-2 mediated dephosphorylation of TCR-associated CD3 and ZAP70 signaling bodies while inhibiting CD28 stimulation signals, weakening downstream TCR signal intensity and cytokine secretion (such as IL-2), ultimately inhibiting T cell function.

Fig 2: The immune regulation mechanism of the PD-1/PD-L1. (Reference source: Lin, X., Kang, K., Chen, P. et al. Regulatory mechanisms of PD-1/PD-L1 in cancers. Mol Cancer 23, 108 (2024). https://doi.org/10.1186/s12943-024-02023-w)
CD47 Target Introduction
CD47 (CD47 differentiation cluster 47), also known as integrin-associated protein (IAP), is involved in a series of cellular processes including apoptosis, proliferation, adhesion, and migration. Activation and axonal development of T cells and dendritic cells (DCs), binding of CD47 to its receptor signaling regulatory protein alpha (SIRP alpha), and downregulation of tumor cell phagocytosis, anti-CD47 antibodies can promote tumor cell death through various mechanisms. Firstly, anti-CD47 monoclonal antibodies can inhibit the interaction between CD47 and SIRP alpha, which can lead to macrophage phagocytosis of tumor cells (anti-SIRP alpha antibodies can also inhibit this pathway), reducing immune escape. Secondly, they can eliminate tumor cells through NK cell-mediated antibody-dependent cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). Thirdly, they can directly induce tumor cell apoptosis and finally present antigens to CD4+ and CD8+ T cells, thereby stimulating an anti-tumor adaptive immune response.

Fig 3: Therapeutic targeting of the CD47-SIRPα pathway can cause elimination of cancer cells through multiple mechanisms. (Reference source: Chao MP, Weissman IL, Majeti R. The CD47-SIRPα pathway in cancer immune evasion and potential therapeutic implications. )
The author inoculated tumor cell lines (selected AT3-OVA can express high levels of CD47 and PD-L1) into mice, injected a certain amount of bispecific antibodies (CD47 × PD-L1) at certain intervals, and added corresponding control groups. For CD8+ T cell experiments, anti-CD8a was used to observe. After some time, T cells were isolated from mouse tissues and bodies, and single-cell suspensions were prepared for cell flow cytometry analysis. The data was statistically analyzed, and the tumor growth inhibition rate was estimated by calculating the volume average.
Result and ConclusionLITERATURE
CD47 × PD-L1 BisAb treatment can reduce the burden of AT3 tumors and enhance CD8+ T cells in tumors
Fig. 4a shows the experimental group and control group: CD47 × PD-L1 BisAb and isotype control were injected into mice receiving AT3-OVA tumors. Fig. 4b (process changes) and c (result presentation) show that the tumor volume was significantly reduced compared to receiving isotype control antibodies. Figs. 4d and e represent macrophages, monocytes, and neutrophils in the spleen and tumor, respectively, whose numbers increased after the experiment. Fig. 4f and g represent CD44 hi CD4+ T cells in the spleen and tumor. The number of CD44 hi CD8+ T cells and SIINFEKL+ CD8+ T cells showed significant increases in some cases and no significant changes in others.

Fig 4: CD47 × PD‐L1 BisAb therapy reduces tumor burden and induces CD8+ T cell expansion.
CD8+ T cells play an important role in CD47 × PD-L1 BisAb-mediated tumor control
Fig. 5a Inoculate mice in the experimental group and the control group with anti-CD8 depletion antibodies. Fig. 5b shows the number of CD44 hi CD8+ T cells in the spleen and tumor. It can be seen that the number of CD44 hi CD8+ T cells decreases sharply or even does not exist. The tumor volume in Fig. 5c increases with time. The final tumor volume in Fig. 5d increases compared with other control groups. It can be seen that CD8+ T cells play a direct role in tumor regression of breast cancer, and the tumor control mechanism mediated by CD47 × PD-L1 BisAb is regulated by CD8+ T cells.

Fig 5: CD47 × PD‐L1 BisAb therapy reduces tumor burden and induces CD8+ T cell expansion.
The author also demonstrated that the efficacy of CD47 × PD-L1 BisAb combination therapy is due to the effect of anti-PD-L1 monotherapy.
Therefore, the use of CD47 × PD-L1 BisAb dual checkpoint blockade is the best strategy to promote CD8+ T cell-mediated protection, providing insights for cancer treatment.
about usLITERATURE
Alpha Lifetech can provide biosimilar antibodies targeting CD47 and PD-L1, respectively, as shown in the table below. We can also provide biosimilar antibody development and bispecific antibody development services to accelerate the progress of our client's project research and provide customized services for our clients.
Tab 2: Biosmilar Antibody Products List.
Catalog Number | Product Name | Target | Product Sizes |
---|---|---|---|
ADT1054 | ADT1054-Atezolizumab Biosimilar– Anti-CD274, B7-H1, PDL1 mAb-Research Grade | PD-L1 | 1mg,5mg |
ADT1061 | ADT1061-Avelumab Biosimilar– Anti-PD-L1 mAb-Research Grade | PD-L1 | 1mg,5mg |
ADT1149 | ADT1149-Cosibelimab Biosimilar– Anti-CD274, PD-L1, B7-H1 mAb-Research Grade | PD-L1 | 100ug,1mg,5mg |
ADT1186 | ADT1186-Durvalumab Biosimilar– Anti-PD-L1 mAb-Research Grade | PD-L1 | 1mg,5mg |
ADT1217 | ADT1217-Envafolimab Biosimilar– Anti-CD274, PD-L1, B7-H1 mAb-Research Grade | PD-L1 | 1mg,5mg |
ADT1382 | ADT1382-Manelimab Biosimilar– Anti-CD274, PD-L1, B7-H1 mAb-Research Grade | PD-L1 | 100ug,1mg,5mg |
ADT1485 | ADT1485-Pacmilimab Biosimilar– Anti-CD274, PD-L1, B7-H1 mAb-Research Grade | PD-L1 | 100ug,1mg,5mg |
ADT1347 | ADT1347-lemzoparlimab Biosimilar-Anti CD47 mAb- Research Grade | CD47 | 100ug,1mg,5mg |
ADT1354 | ADT1354-Letaplimab Biosimilar-Anti CD47 mAb- Research Grade | CD47 | 100ug,1mg,5mg |
ADT1361 | ADT1361-Ligufalimab Biosimilar-Anti CD47 mAb- Research Grade | CD47 | 100ug,1mg,5mg |
ADT1681 | ADT1681-Urabrelimab Biosimilar-Anti CD47 mAb- Research Grade | CD47 | 100ug,1mg,5mg |
ADT1381 | ADT1381-Magrolimab Biosimilar-Anti CD47 mAb- Research Grade | CD47 | 100ug,1mg,5mg |
ReferenceLITERATURE
[1] Christo, S.N., McDonald, K.M., Burn, T.N., Kurd, N., Stanfield, J., Kaneda, M.M., Seelige, R., Dillon, C.P., Fisher, T.S., Baaten, B. and Mackay, L.K. (2024), Dual CD47 and PD-L1 blockade elicits anti-tumor immunity by intratumoral CD8+ T cells. Clin Transl Immunol, 13: e70014. https://doi.org/10.1002/cti2.70014
[2] https://www.news-medical.net/news/20250212/New-dual-target-therapy-offers-hope-for-better-breast-cancer-treatment.aspx
[3] Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015 Apr 13;27(4):450-61. doi: 10.1016/j.ccell.2015.03.001. Epub 2015 Apr 6. PMID: 25858804; PMCID: PMC4400238.
[4] https://en.wikipedia.org/wiki/Cytotoxic_T_cell
[5] Lin, X., Kang, K., Chen, P. et al. Regulatory mechanisms of PD-1/PD-L1 in cancers. Mol Cancer 23, 108 (2024). https://doi.org/10.1186/s12943-024-02023-w
[6] Chao MP, Weissman IL, Majeti R. The CD47-SIRPα pathway in cancer immune evasion and potential therapeutic implications. Curr Opin Immunol. 2012 Apr;24(2):225-32. doi: 10.1016/j.coi.2012.01.010. Epub 2012 Feb 4. PMID: 22310103; PMCID: PMC3319521.