Mechanism And Application Status Of CAR-T Cell Immunotherapy
DOI:
https://doi.org/10.62051/p6afxj50Keywords:
CAR-T cells; immunotherapy; tumor microenvironment.Abstract
With the continuous increase in the incidence and mortality of cancer, finding more accurate and effective treatment methods has become an important direction of medical research. As an important technological breakthrough in the field of tumor treatment in recent years, chimeric antigen receptor T cell (CAR-T) immunotherapy transforms T cells through genetic engineering to enable them to specifically recognize and kill tumor cells, overcoming many limitations of traditional treatment methods such as chemotherapy, radiotherapy and surgery. This article systematically introduces the basic principles and mechanisms of CAR-T cell immunotherapy, and reviews its clinical application and initial results in hematological malignancies. At the same time, this article also deeply analyzes the challenges currently faced by CAR-T therapy in clinical promotion, including the impact of immunosuppressive tumor microenvironment on efficacy, the difficulty in controlling side effects such as cytokine release syndrome (CRS) and neurotoxicity, and the high cost of treatment. To address these challenges, researchers are actively exploring improvement strategies such as multi-target CAR-T cells and "armored" CAR-T cells. Looking to the future, CAR-T cell therapy is expected to achieve a wider range of disease applications, and make greater breakthroughs in ensuring safety and improving treatment effects, becoming one of the core forces in anti-tumor treatment.
Downloads
References
[1] World Health Organization. Global Cancer Burden growing, Amidst Mounting Need for Services [EB/OL]. (2024-02-01). https://www.who.int/news/item/01-02-2024-global-cancer-burden-growing--amidst-mounting-need-for-services.
[2] Zheng Nairong, Xu Jianqing. Research Progress of CAR-T Cell Immunotherapy[J]. Fudan Journal of Medicine, 2022, (02).
[3] Sterner R C, Sterner R M. CAR-T cell therapy: current limitations and potential strategies[J]. Blood Cancer Journal, 2021, 11(4): 69.
[4] Laetsch T W, Maude S L, Rives S, et al. Three-Year Update of Tisagenlecleucel in Pediatric and Young Adult Patients with Relapsed/Refractory Acute Lymphoblastic Leukemia in the ELIANA Trial[J]. Journal of Clinical Oncology, 2022, 41(9): JCO2200642.
[5] Jacobson C A, Chavez J C, Sehgal A R, et al. Axicabtagene ciloleucel in relapsed or refractory indolent non-Hodgkin lymphoma (ZUMA-5): a single-arm, multicentre, phase 2 trial[J]. The Lancet Oncology, 2022, 23(1): 91–103.
[6] Fala L. Abecma (Idecabtagene Vicleucel) First-in-Class BCMA-Directed CAR T-Cell Therapy Approved for Relapsed or Refractory Multiple Myeloma[J]. 2023.
[7] Longer-term Data from CARTITUDE-1 Study Demonstrate Continued Deep and Durable Responses to CARVYKTI™ (ciltacabtagene autoleucel) in Heavily Pretreated Patients with Relapsed or Refractory Multiple Myeloma [EB/OL].
[8] Ulloa-Navas M J, Luo Y, Sanchez-Garavito J E, et al. Single CAR-Dual target: Intracranial Delivery of Anti-PD-L1 CAR T Cells Effectively Eradicates Glioma and Immunosuppressive Cells in the Tumor Microenvironment[J/OL]. bioRxiv, 2024.
[9] Ferris R L, Jaffee E M, Ferrone S. Tumor antigen–targeted, monoclonal antibody–based immunotherapy: clinical response, cellular immunity, and immunoescape[J]. Journal of Clinical Oncology, 2010, 28(28): 4390–4399.
[10] Mansurov A, Ishihara J, Hosseinchi P, et al. Collagen-binding IL-12 enhances tumour inflammation and drives the complete remission of established immunologically cold mouse tumours[J]. Nature Biomedical Engineering, 2020, 4(5): 531–543.
Downloads
Published
Issue
Section
License
Copyright (c) 2025 Transactions on Materials, Biotechnology and Life Sciences

This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.






