Journal of Cancer Immunology & Therapy

All submissions of the EM system will be redirected to Online Manuscript Submission System. Authors are requested to submit articles directly to Online Manuscript Submission System of respective journal.

Rapid Communication - Journal of Cancer Immunology & Therapy (2025) Volume 8, Issue 1

Challenges and Future Directions in Adoptive Cell Therapy

Shan Peng *

Department of Pharmacology, Central South University, China

*Corresponding Author:
Shan Peng
Department of Pharmacology, Central South University, China
E-mail: Sh.peng@csu.edu.cn

Received: 03-Feb -2025, Manuscript No. AAJCIT-25-161378; Editor assigned: 04-Feb-2025, PreQC No. AAJCIT-25-161378 (PQ); Reviewed:18-Feb-2025, QC No. AAJCIT-25-161378; Revised:23-Feb-2025, Manuscript No. AAJCIT-25-161378 (R); Published:28-Feb-2025, DOI:10.35841/aajcit-8.1.252

Citation: Peng S. Challenges and future directions in adoptive cell therapy. J Cancer Immunol Ther. 2025;8(1):252

Visit for more related articles at Journal of Cancer Immunology & Therapy

Abstract

  

Introduction

Adoptive cell therapy (ACT) has emerged as a revolutionary approach in cancer immunotherapy, harnessing the power of the immune system to target malignancies. The most prominent forms of ACT include chimeric antigen receptor (CAR) T-cell therapy, tumor-infiltrating lymphocyte (TIL) therapy, and T-cell receptor (TCR) therapy. While ACT has demonstrated remarkable success in treating hematologic malignancies, significant challenges hinder its broader application. This article explores the key challenges associated with ACT and discusses future directions to enhance its efficacy, safety, and accessibility [1].

Despite the success of CAR T-cell therapy in hematologic malignancies, its application in solid tumors remains challenging. Solid tumors create an immunosuppressive microenvironment that limits the infiltration and persistence of adoptively transferred cells. Additionally, the heterogeneity of tumor antigens complicates the identification of suitable targets [2].

CAR T-cells can recognize antigens present on both malignant and normal tissues, leading to off-tumor effects. For example, HER2-targeted CAR T-cells have caused fatal toxicity due to expression of HER2 in normal lung tissue. Developing strategies to enhance tumor specificity while minimizing off-tumor recognition is crucial for safety improvements [3].

One of the most serious complications of ACT, particularly CAR T-cell therapy, is cytokine release syndrome (CRS). This systemic inflammatory response can lead to multi-organ failure if not properly managed. Additionally, neurotoxicity, characterized by confusion, seizures, and cerebral edema, poses a significant risk to patients. Strategies to mitigate these toxicities, such as the use of IL-6 inhibitors (e.g., tocilizumab), have been explored, but further improvements are needed [4].

ACT is a personalized therapy that requires ex vivo expansion and genetic modification of patient-derived T-cells, making it a labor-intensive and costly process. The manufacturing time for CAR T-cells can take weeks, delaying treatment for patients with aggressive disease. Developing allogeneic (off-the-shelf) cell therapies and optimizing automation in cell processing could address these limitations [5].

The long-term persistence of adoptively transferred cells is essential for sustained antitumor effects. However, T-cell exhaustion, driven by chronic antigen exposure and inhibitory signaling pathways, limits their efficacy. Enhancing T-cell persistence through genetic modifications, checkpoint blockade, or metabolic reprogramming is an area of active research [6].

Tumors employ multiple mechanisms to evade immune detection, including loss of antigen expression and upregulation of inhibitory ligands such as PD-L1. Overcoming immune evasion strategies by combining ACT with immune checkpoint inhibitors or engineering multi-targeted T-cells is a promising approach [7].

The advent of CRISPR-Cas9 and other gene-editing tools has revolutionized ACT by enabling precise modifications to enhance T-cell function. Knockout of inhibitory receptors (e.g., PD-1) or insertion of synthetic receptors can improve T-cell persistence and efficacy [8].

Combining ACT with immune checkpoint inhibitors, vaccines, or cytokine therapies may enhance efficacy. For instance, PD-1 blockade has been shown to improve CAR T-cell persistence and function. Such combinatorial approaches are likely to play a crucial role in overcoming resistance mechanisms [9].

Engineering T-cells to resist immunosuppressive signals in the tumor microenvironment is a major focus of research. Strategies such as incorporating dominant-negative TGF-β receptors or secreting pro-inflammatory cytokines like IL-12 could improve T-cell infiltration and activity in solid tumors [10].

Conclusion

Adoptive cell therapy has transformed the landscape of cancer treatment, offering hope to patients with refractory malignancies. However, challenges such as immune evasion, manufacturing complexities, and toxicity must be addressed to maximize its potential. Future advancements in gene editing, combinatorial therapies, and automation are likely to enhance the safety, efficacy, and affordability of ACT. As research progresses, the next generation of ACT may achieve broader applicability, ultimately improving patient outcomes across various cancer types.

References

  1. A Baudino T. Targeted cancer therapy: the next generation of cancer treatment. Curr Drug Discov Technol. 2015;12(1):3-20.
  2. Indexed at, Google Scholar

  3. Efferth T, Li PC, Konkimalla VS, et al., From traditional Chinese medicine to rational cancer therapy. Trends Mol Med. 2007;13(8):353-61.
  4. Indexed at, Google Scholar, Cross Ref

  5. Moslehi JJ. Cardiovascular toxic effects of targeted cancer therapies. New England Journal of Medicine. 2016;375(15):1457-67.
  6. Indexed at, Google Scholar, Cross Ref

  7. Sullivan R, Smith JE, Rowan NJ. Medicinal mushrooms and cancer therapy: translating a traditional practice into Western medicine. Perspect Biol Med. 2006;49(2):159-70.
  8. Indexed at, Google Scholar,

  9. Zhang H, Berezov A, Wang Q, et al., ErbB receptors: from oncogenes to targeted cancer therapies. J Clin Investig. 2007;117(8):2051-8.
  10. Indexed at, Google Scholar, Cross Ref

  11. Perera PK. Traditional medicine-based therapies for cancer management. Sys Rev Pharm. 2019;10(1):90-2.
  12. Indexed at, Google Scholar, Cross Ref

  13. Ouyang L, Luo Y, Tian M, et al., Plant natural products: from traditional compounds to new emerging drugs in cancer therapy. Cell Prolif. 2014;47(6):506-15.
  14. Indexed at, Google Scholar, Cross Ref

  15. Ky B, Vejpongsa P, Yeh ET, et al., Emerging paradigms in cardiomyopathies associated with cancer therapies. Circ Res.2013;113(6):754-64.
  16. Indexed at, Google Scholar, Cross Ref

  17. Herrmann J. Adverse cardiac effects of cancer therapies: cardiotoxicity and arrhythmia. Nat Rev Cardiol. 2020;17(8):474-502.
  18. Indexed at, Google Scholar, Cross Ref

  19. Chen H, Zhou X, Gao Y, et al., Recent progress in development of new sonosensitizers for sonodynamic cancer therapy. Drug Discov Today. 2014;19(4):502-9.
  20. Indexed at, Google Scholar, Cross Ref

Get the App