Commentary - Journal of Cancer Immunology & Therapy (2025) Volume 8, Issue 2
Checkpoint Inhibitors and T-Cell Activation: Enhancing Immune Response
Karin Asara *
Department of Experimental Medical Science, Lund University, Sweden
- *Corresponding Author:
- Karin Asara
Department of Experimental Medical Science, Lund University, Sweden
E-mail: karin.a@med.lu.se
Received: 03-Apr -2025, Manuscript No. AAJCIT-25-163914; Editor assigned: 04-Apr-2025, PreQC No. AAJCIT-25-163914 (PQ); Reviewed:18-Apr-2025, QC No. AAJCIT-25-163914; Revised:23-Apr-2025, Manuscript No. AAJCIT-25-163914 (R); Published:28-Apr-2025, DOI:10.35841/aajcit-8.2.259
Citation: Asara K. Checkpoint inhibitors and t-cell activation: Enhancing immune response. J Cancer Immunol Ther. 2025;8(2):259
Introduction
The immune system has evolved complex mechanisms to eliminate pathogens and abnormal cells, including cancer. Central to this defense are T cells, which can target and destroy diseased cells. However, to maintain immune homeostasis and prevent autoimmunity, the body employs regulatory mechanisms known as immune checkpoints. While these checkpoints are essential in normal physiology, they can be co-opted by tumors and chronic infections to evade immune surveillance [1].
Checkpoint inhibitors are a class of immunotherapies that "release the brakes" on T cells, enhancing their activation and boosting immune response. They have revolutionized cancer treatment and show promise in infectious diseases and chronic inflammation. This article explores how checkpoint inhibitors work, their relationship with T-cell activation, and their therapeutic impact [2].
Recognition of antigen via T-cell receptor (TCR) and MHC-peptide complex. Co-stimulatory signals (e.g., CD28 binding to CD80/CD86 on antigen-presenting cells). Cytokine signaling that influences T-cell proliferation and differentiation. Following activation, T cells upregulate inhibitory receptors such as CTLA-4, PD-1, LAG-3, and TIM-3. These immune checkpoints are crucial for dampening immune responses to prevent overactivation and tissue damage [3].
CTLA-4 competes with CD28 for binding to CD80/CD86 and inhibits early T-cell activation in lymph nodes. Its blockade increases T-cell priming and expansion. PD-1 binds to PD-L1/PD-L2 and suppresses T-cell activity in peripheral tissues, particularly within the tumor microenvironment. It reduces cytokine production and promotes T-cell exhaustion [4].
Checkpoint inhibitors targeting CTLA-4 (e.g., ipilimumab) or PD-1/PD-L1 (e.g., nivolumab, pembrolizumab, atezolizumab) block these inhibitory pathways, leading to reinvigoration of exhausted T cells and enhanced immune response. Preventing inhibitory signals that suppress T-cell activity. Reinvigorating exhausted T cells that have lost effector function due to chronic antigen stimulation. Promoting expansion of memory and effector T-cell populations [5].
Modifying the tumor microenvironment to favor immune infiltration. Checkpoint inhibitors not only restore cytotoxic CD8+ T-cell activity but also enhance the function of CD4+ helper T cells, dendritic cells, and natural killer (NK) cells. Checkpoint inhibitors have shown remarkable success in treating several cancers: Ipilimumab and nivolumab have improved survival in metastatic melanoma [6].
PD-1/PD-L1 inhibitors are now first-line treatments. Show high response rates to checkpoint blockade. Combination therapies (e.g., CTLA-4 and PD-1 blockade) often yield better responses, though with increased toxicity. Chronic viral infections like HIV and hepatitis B to reverse T-cell exhaustion. Tuberculosis and Leishmaniasis, where chronic infection leads to T-cell dysfunction [7].
Autoimmune diseases, with caution, to rebalance immune responses without overactivation. Research is ongoing to determine how checkpoint modulation can be safely applied to infectious and inflammatory diseases. Checkpoint blockade may cause immune-related adverse events (irAEs), including: Colitis, Pneumonitis, Hepatitis, Endocrinopathies (e.g., thyroiditis) [8].
These side effects result from increased systemic T-cell activation and loss of peripheral. Moreover, not all patients respond. Resistance can arise due to: Lack of tumor-infiltrating lymphocytes (TILs) Absence of neoantigens, Immunosuppressive tumor environments [9].
Biomarkers like PD-L1 expression, tumor mutational burden (TMB), and T-cell receptor clonality are being explored to predict response. Strategies to enhance checkpoint blockade include: Bi-specific antibodies targeting multiple immune checkpoints. Checkpoint inhibitor combinations with chemotherapy or radiation. Personalized vaccines to increase neoantigen-specific T-cell responses [10].
Conclusion
Checkpoint inhibitors have transformed the therapeutic landscape by harnessing T-cell activation to fight disease. By blocking inhibitory pathways like PD-1 and CTLA-4, these therapies restore immune competence and have shown remarkable clinical benefits, particularly in oncology. Although challenges remain—especially regarding adverse effects and resistance—the integration of checkpoint modulation with other immunotherapies holds promise for broader and more effective treatment paradigms in the future.
References
- Miao L, Zhang Y, Huang L. mRNA vaccine for cancer immunotherapy. Mol Cancer. 2021;20(1):1-23.
- Van Lint S, Renmans D, Broos K, et al., The ReNAissanCe of mRNA-based cancer therapy. Expert Rev Vaccines. 2015;14(2):235-51.
- Heine A, Juranek S, Brossart P. Clinical and immunological effects of mRNA vaccines in malignant diseases. Mol Cancer. 2021;20(1):1-20.
- Barbier AJ, Jiang AY, Zhang P, et al., The clinical progress of mRNA vaccines and immunotherapies. Nat Biotechnol. 2022;40(6):840-54.
- Beck JD, Reidenbach D, Salomon N, et al., mRNA therapeutics in cancer immunotherapy. Mol Cancer. 2021;20(1):1-24.
- Pardi N, Hogan MJ, Porter FW, et al., mRNA vaccines—a new era in vaccinology. Nat Rev Drug Discov. 2018;17(4):261-79.
- Jahanafrooz Z, Baradaran B, Mosafer J, et al., Comparison of DNA and mRNA vaccines against cancer. Drug Discov Today. 2020;25(3):552-60.
- Fotin?Mleczek M, Zanzinger K, Heidenreich R, et al., Highly potent mRNA based cancer vaccines represent an attractive platform for combination therapies supporting an improved therapeutic effect. J Genet Med. 2012;14(6):428-39.
- Diken M, Kranz LM, Kreiter S, et al., mRNA: a versatile molecule for cancer vaccines. Curr Issues Mol Biol. 2017;22(1):113-28.
- Rosa SS, Prazeres DM, Azevedo AM, et al., mRNA vaccines manufacturing: Challenges and bottlenecks. 2021;39(16):2190-200.
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref