Bispecific antibodies (BsAbs) are artificial molecules that can simultaneously bind to two different antigens or epitopes, thereby enabling novel modes of action and therapeutic applications. BsAbs have been explored for various purposes, such as targeting multiple pathways, recruiting immune cells, enhancing effector functions, and delivering drugs or toxins. Among these, one of the most promising and attractive applications of BsAbs is cancer immunotherapy, which aims to harness the power of the immune system to fight against tumors. Cancer immunotherapy has been revolutionized by the discovery and development of checkpoint inhibitors, which are monoclonal antibodies (mAbs) that block the inhibitory signals that suppress the anti-tumor activity of T cells. However, checkpoint inhibitors have several limitations, such as low response rates, resistance mechanisms, toxicity issues, and high costs. To overcome these challenges, BsAbs have been developed to achieve dual checkpoint blockade, which is a strategy that simultaneously targets two inhibitory receptors or ligands on T cells or tumor cells, such as PD-1, PD-L1, CTLA-4, LAG-3, TIM-3, etc. Dual checkpoint blockade can potentially enhance the efficacy and safety of cancer immunotherapy by overcoming the immune suppression and resistance mediated by multiple checkpoints.
The immune system is composed of various cells and molecules that work together to protect the body from foreign invaders and eliminate abnormal or damaged cells. Among these, T cells are the key players of adaptive immunity, which can recognize and kill specific antigens, such as tumor-associated antigens (TAAs). However, T cells are also regulated by various signals that modulate their activation, proliferation, differentiation, and function. These signals can be classified into two categories: co-stimulatory signals and co-inhibitory signals. Co-stimulatory signals enhance the T cell response, while co-inhibitory signals suppress the T cell response. Co-inhibitory signals are also known as checkpoints, which are essential for maintaining immune tolerance and preventing autoimmunity. However, tumors can exploit these checkpoints to evade immune surveillance and resistance by expressing or inducing the expression of inhibitory receptors or ligands on T cells or tumor cells, such as PD-1, PD-L1, CTLA-4, LAG-3, TIM-3, etc. These checkpoints can inhibit the T cell activation, proliferation, survival, effector function, and memory formation, resulting in T cell exhaustion and dysfunction.
To overcome immune suppression and resistance mediated by multiple checkpoints, a strategy called dual checkpoint blockade has been developed. Dual checkpoint blockade uses bispecific antibodies (BsAbs) to simultaneously target two inhibitory receptors or ligands on T cells or tumor cells. This blocks their interaction and restores the T cell response. Dual checkpoint blockade has the potential to enhance the efficacy and safety of cancer immunotherapy by achieving synergistic effects, overcoming heterogeneity, reducing toxicity, and improving pharmacokinetics.
For example, dual checkpoint blockade can target PD-1 and CTLA-4, which are two major checkpoints that regulate T cell activation at different stages. PD-1 inhibits the T cell response in peripheral tissues, while CTLA-4 inhibits the T cell response in lymph nodes. By blocking both PD-1 and CTLA-4, dual checkpoint blockade can enhance the priming and effector phases of the T cell response.
Similarly, dual checkpoint blockade can target PD-1 and LAG-3, which are two co-expressed checkpoints that induce T cell exhaustion in chronic infections and cancers. By blocking both PD-1 and LAG-3, dual checkpoint blockade can reverse T cell exhaustion and restore T cell function.
Moreover, dual checkpoint blockade can target PD-L1 and 4-1BB, which are two ligands expressed on tumor cells and activated T cells, respectively. By blocking PD-L1 and engaging 4-1BB, dual checkpoint blockade can not only inhibit tumor-induced immune suppression but also stimulate T cell activation and survival. These are some examples of how dual checkpoint blockade can achieve superior outcomes compared to monospecific checkpoint blockade or combination therapy in cancer immunotherapy.
Application scenarios of dual checkpoint blockade using bispecific antibodies have been seen in various types of cancers, such as melanoma, non-small cell lung cancer (NSCLC), renal cell carcinoma (RCC), head and neck squamous cell carcinoma (HNSCC), and colorectal cancer (CRC). These cancers are characterized by high immunogenicity, high expression of inhibitory checkpoints, and low response to monospecific checkpoint inhibitors. By simultaneously blocking two inhibitory checkpoints, bispecific antibodies can enhance anti-tumor immunity and overcome resistance mechanisms in these cancers.
Table 1. Examples of bispecific antibodies for dual checkpoint blockade in different types of cancers
Bispecific antibody | Checkpoints | Cancer type | Clinical outcome |
---|---|---|---|
REGN5678 (PD-1 x LAG-3) | PD-1 on T cells and LAG-3 on tumor cells | Melanoma | Significant clinical activity in patients who have progressed on prior PD-1 or PD-L1 inhibitors |
MGD013 (PD-1 x LAG-3) | PD-1 and LAG-3 on T cells | NSCLC | Durable responses and disease control in patients who have failed prior PD-1 or PD-L1 inhibitors |
RO7121661 (PD-L1 x 4-1BB) | PD-L1 on tumor cells and 4-1BB on T cells | RCC | Tumor regression and immune activation in patients who have received prior PD-1 or PD-L1 inhibitors |
XmAb20717 (PD-1 x CTLA-4) | PD-1 and CTLA-4 on T cells | HNSCC | Objective responses and stable disease in patients who have not received prior PD-1 or PD-L1 inhibitors |
MGD019 (PD-1 x CTLA-4) | PD-1 and CTLA-4 on T cells | CRC | Preliminary anti-tumor activity and manageable safety in patients who have high microsatellite instability or high tumor mutational burden |
References
1. Kontermann RE, et al. Bispecific antibodies. Drug Discov Today. 2015 Jun;20(7):838-47.
2. Spiess C, et al. Alternative molecular formats and therapeutic applications for bispecific antibodies. Mol Immunol. 2015 Oct;67(2 Pt A):95-106.
3. Labrijn AF, et al. Bispecific antibodies: a mechanistic review of the pipeline. Nat Rev Drug Discov. 2019 Aug;18(8):585-608.
4. Bacac M, et al. CEA TCB: a novel head-to-tail 2:1 T cell bispecific antibody for treatment of CEA-positive solid tumors. Oncoimmunology. 2016 Sep 16;5(10):e1203498.
5. Bacac M, et al. A novel carcinoembryonic antigen T-cell bispecific antibody (CEA TCB) for the treatment of solid tumors. Clin Cancer Res. 2016 Jul 1;22(13):3286-97.
6. Fan G, et al. Bispecific antibodies and their applications. J Hematol Oncol. 2015 Dec 29;8:130.
7. Fridman WH, et al. The immune contexture in cancer prognosis and treatment. Nat Rev Clin Oncol. 2017 Dec;14(12):717-34.
8. Gantner F, et al. The potential of dual-specific antibodies as therapeutics for infectious diseases and cancer. Expert Opin Biol Ther. 2020 Mar;20(3):249-62.
9. He XJ, et al. Bispecific antibodies as a development platform for new concepts and treatment strategies. Int J Mol Sci. 2017 Jan 4;18(1):48.
10. Kontermann RE. Dual targeting strategies with bispecific antibodies. MAbs. 2012 Mar-Apr;4(2):182-97.
Welcome! For price inquiries, we will get back to you as soon as possible.
INQUIRYSERVICES
PRODUCTS
PLATFORMS