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.

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

Expanding the Frontiers of Immunotherapy: Advances in Adoptive Cell Therapy

Jonathan Nair *

School of Medicine, Johns Hopkins University School of Medicine, USA

*Corresponding Author:
Jonathan Nair
Department of Medicine, Johns Hopkins University School of Medicine, USA
E-mail: jnair@jhmi.edu

Received: 03-Feb -2025, Manuscript No. AAJCIT-25-161380; Editor assigned: 04-Feb-2025, PreQC No. AAJCIT-25-161380 (PQ); Reviewed:18-Feb-2025, QC No. AAJCIT-25-161380; Revised:23-Feb-2025, Manuscript No. AAJCIT-25-161380 (R); Published:28-Feb-2025, DOI:10.35841/aajcit-8.1.254

Citation: Nair J. Expanding the frontiers of immunotherapy: Advances in adoptive cell therapy. J Cancer Immunol Ther. 2025;8(1):254

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

Abstract

  

Introduction

Immunotherapy has revolutionized the treatment of cancer and other diseases by harnessing the body's immune system to target and eliminate malignancies. Among the various immunotherapeutic approaches, adoptive cell therapy (ACT) has emerged as a promising strategy, demonstrating significant clinical success. ACT involves the isolation, genetic modification, and expansion of immune cells before reinfusing them into patients to enhance anti-tumor responses. This article explores recent advancements in ACT, including chimeric antigen receptor (CAR) T-cell therapy, T-cell receptor (TCR) therapy, tumor-infiltrating lymphocytes (TILs), and natural killer (NK) cell therapy, and discusses challenges and future directions in this rapidly evolving field [1].

CAR T-cell therapy has been at the forefront of ACT advancements. It involves genetically engineering T-cells to express synthetic receptors (CARs) that recognize specific tumor antigens. CAR T-cells have shown remarkable efficacy in hematological malignancies, leading to FDA approval of therapies such as axicabtagene ciloleucel and tisagenlecleucel for B-cell lymphomas and leukemias. Recent developments aim to improve CAR T-cell therapy by reducing toxicities such as cytokine release syndrome (CRS) and neurotoxicity, enhancing persistence, and expanding applicability to solid tumors [2].

Unlike CAR T-cells, TCR therapy relies on the natural T-cell receptor to recognize tumor antigens presented by major histocompatibility complex (MHC) molecules. TCR-engineered T-cells can target intracellular proteins, broadening their application beyond surface antigens. TCR therapies are being developed for multiple cancers, including melanoma, sarcoma, and lung cancer. The challenge remains in improving antigen specificity and reducing off-target effects [3].

TIL therapy utilizes naturally occurring lymphocytes isolated from a patient’s tumor, expanded ex vivo, and reinfused to enhance immune response. Clinical trials have demonstrated durable responses in melanoma patients, leading to FDA breakthroughs. Advances in TIL therapy include combination approaches with checkpoint inhibitors and cytokine-based enhancements to improve efficacy [4].

NK cells provide an alternative ACT approach due to their innate ability to recognize and eliminate tumor cells without prior sensitization. Advances in genetic modification, such as CAR-NK cells, and the use of induced pluripotent stem cell (iPSC)-derived NK cells, have increased their therapeutic potential. Early-phase clinical trials have shown promising results, especially in hematologic cancers [5].

Despite significant progress, several challenges hinder the widespread application of ACT. These include: While CAR T-cells have been highly effective in blood cancers, their efficacy in solid tumors remains limited due to the immunosuppressive tumor microenvironment and poor tumor infiltration [6].

The complexity and high cost of manufacturing patient-specific ACT products pose barriers to accessibility. Efforts are underway to develop off-the-shelf allogeneic cell therapies [7].

To overcome these challenges, research is focusing on several key areas: Genetic modifications, CRISPR-based gene editing, and metabolic reprogramming are being investigated to improve the longevity and efficacy of ACT products [8].

ACT is being combined with immune checkpoint inhibitors, cytokine therapy, and oncolytic viruses to enhance anti-tumor responses. Allogeneic T and NK cell therapies derived from healthy donors or iPSCs are being developed to provide cost-effective and readily available treatments [9].

CRS and neurotoxicity are major side effects of CAR T-cell therapy. Strategies such as IL-6 inhibitors and suicide gene constructs are being explored to mitigate these effects [10].

Conclusion

Adoptive cell therapy represents a groundbreaking advancement in immunotherapy, with the potential to transform cancer treatment and beyond. Continuous innovations in cell engineering, combination strategies, and manufacturing scalability will be critical in expanding its reach to a broader patient population. As research progresses, ACT is poised to become a cornerstone of precision medicine in oncology and other diseases.

References

  1. Danishefsky SJ, Shue YK, Chang MN, et al., Development of Globo-H cancer vaccine. Acc Chem Res. 2015;48(3):643-52.
  2. Indexed at, Google Scholar, Cross Ref

  3. Bowen WS, Svrivastava AK, Batra L, et al., Current challenges for cancer vaccine adjuvant development. Expert Rev Vaccines. 2018;17(3):207-15.
  4. Indexed at, Google Scholar, Cross Ref

  5. Bowen WS, Svrivastava AK, Batra L, et al., Current challenges for cancer vaccine adjuvant development. Expert Rev Vaccines. 2018;17(3):207-15.
  6. Indexed at, Google Scholar, Cross Ref

  7. Wang RF, Rosenberg SA. Human tumor antigens for cancer vaccine development. Immunol  1999;170(1):85-100.
  8. Indexed at, Google Scholar, Cross Ref

  9. Saxena M, van der Burg SH, Melief CJ, et al., Therapeutic cancer vaccines. Nat Rev Cancer. 2021;21(6):360-78.
  10. Indexed at, Google Scholar, Cross Ref

  11. Goldman B, DeFrancesco L. The cancer vaccine roller coaster. Nat Biotechnol. 2009;27(2):129-39.
  12. Indexed at, Google Scholar, Cross Ref

  13. Buonaguro L, Tagliamonte M. Selecting target antigens for cancer vaccine development. Vaccines. 2020;8(4):615.
  14. Indexed at, Google Scholar, Cross Ref

  15. Jäger E, Jäger D, Knuth A. Clinical cancer vaccine trials. Curr Opin Immunol. 2002;14(2):178-82.
  16. Indexed at, Google Scholar

  17. Gilboa E. DC-based cancer vaccines. J Clin Invest. 2007;117(5):1195-203.
  18. Indexed at, Google Scholar

  19. Finn OJ. Cancer vaccines: between the idea and the reality. Nat Rev Immunol. 2003;3(8):630-41.
  20. Indexed at, Google Scholar, Cross Ref

Get the App