Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1221 2023-04-24 15:19:58 |
2 format corrected. Meta information modification 1221 2023-04-25 04:57:40 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Want, M.Y.; Bashir, Z.; Najar, R.A.; Want, M.Y. T Cell Based Immunotherapy for Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/43400 (accessed on 11 September 2024).
Want MY, Bashir Z, Najar RA, Want MY. T Cell Based Immunotherapy for Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/43400. Accessed September 11, 2024.
Want, Muzamil Y., Zeenat Bashir, Rauf A. Najar, Muzamil Yaqub Want. "T Cell Based Immunotherapy for Cancer" Encyclopedia, https://encyclopedia.pub/entry/43400 (accessed September 11, 2024).
Want, M.Y., Bashir, Z., Najar, R.A., & Want, M.Y. (2023, April 24). T Cell Based Immunotherapy for Cancer. In Encyclopedia. https://encyclopedia.pub/entry/43400
Want, Muzamil Y., et al. "T Cell Based Immunotherapy for Cancer." Encyclopedia. Web. 24 April, 2023.
T Cell Based Immunotherapy for Cancer
Edit

T cells are critical in destroying cancer cells by recognizing antigens presented by MHC molecules on cancer cells or antigen-presenting cells. Identifying and targeting cancer-specific or overexpressed self-antigens is essential for redirecting T cells against tumors, leading to tumor regression. This is achieved through the identification of mutated or overexpressed self-proteins in cancer cells, which guide the recognition of cancer cells by T-cell receptors. There are two main approaches to T cell-based immunotherapy: HLA-restricted and HLA-non-restricted Immunotherapy. 

T cells immunotherapy neoantigens cancer antigens TCR engineering

1. Introduction

Cancer is a major public health challenge, affecting millions of people worldwide. Despite significant advances in cancer therapy, cancer remains a leading cause of death, accounting for approximately 10 million deaths annually. The immune system plays a vital role in protecting the host from malignant cells, and harnessing its potential has been a focus of cancer therapy research for decades. T cells are a critical component of the adaptive immune system, and their ability to recognize and eliminate cancer cells has led to significant advances in cancer immunotherapy. In the mid-twentieth century, the concept of lymphocytes as mediators of anti-tumor surveillance was first proposed by Thomas and Burnett, but it was not until later that T cells were identified as the major mediators of adaptive immunity [1][2][3][4][5][6][7]. Today, the role of T cells in cancer immunotherapy is a rapidly expanding area of research.
The development of T cells begins in the bone marrow, where they originate from progenitors that migrate to the thymus for maturation. Upon maturation in the thymus, T cells differentiate into two main lineages—conventional αβ T cells and unconventional γδ T cells—that can be distinguished by the expression of αβ or γδ T cell receptors, respectively. Conventional αβ T cells comprise various subsets, including cytotoxic T cells, helper T cells, regulatory T cells, natural killer T cells, and memory T cells, each with distinct functions in immune response. While the thymus output of naive T cells decreases with age, human naive T cells have a long intrinsic lifespan and turnover rate, making them a promising candidate for immunotherapy [8][9][10][11][12].
T cells have revolutionized cancer treatment, and their use as adoptive T cell therapy has shown great success for some cancers. However, the success of adoptive T cell therapy depends on various factors, such as the choice of T cell subset, target antigen, and immune evasion mechanisms employed by cancer cells. Additionally, the tumor microenvironment can influence T cell function and promote immune escape, highlighting the need for combination therapies targeting both the cancer cells and the immune system. Nevertheless, the potential of T cells to recognize and eliminate cancer cells has opened new avenues for cancer immunotherapy, providing hope for a future where cancer can be effectively treated and cured. 

2. Antigenic Targets for T Cell Immunotherapy

Antigenic targets for T cell immunotherapy are crucial for the development of effective cancer treatments. The first human tumor antigen was discovered by Van der Bruggen et al. in 1991; it was named MZ2-E and was expressed in melanoma cells [7]. Consequently, cytotoxic T lymphocytes from melanoma patients recognized the melanoma cell line of HLA-A1 patients expressing the MZ2-E. Therefore, their group proposed that precise immunotherapy could be provided to HLA-A1+ melanoma patients expressing the MZ2-E antigen. Since then, various biochemical and genetic approaches have been developed to identify tumor antigens recognized by T cells. The biochemical approach involves eluting the HLA-peptide complex from tumor cells, followed by mass spectrometric sequencing to identify the antigenic peptides presented by the HLA molecules. In contrast, the genetic approach comprises two strategies: forward immunology and reverse immunology. The forward immunology approach involves cloning tumor-reactive T cells from patients and screening a cDNA library from patient-derived tumor cells to identify the antigens recognized by these T cells. On the other hand, the reverse immunology approach uses genome sequencing to identify mutations and predict potential HLA-binding antigens in the patient’s tumor. The peptide candidates are then screened for their ability to activate T cells in vitro [13]. T cells can target both HLA-restricted and HLA-non-restricted antigens found on the target cells. HLA-independent T cell immunotherapy targets antigens expressed on the surface of tumor cells. In contrast, HLA-dependent T cell immunotherapy targets antigens bound to HLA molecules on the surface of cells, known as the immunopeptidome.

2.1. Identification of Tumor Antigenic Epitopes for T Cell Immunotherapy

T cell immunotherapy is a promising approach for cancer treatment, but identifying tumor-reactive T cells and their target antigens is crucial for its success. There are two approaches to identifying these antigens: the indirect method, which involves screening candidate antigen targets of T cells from tumors in the laboratory, and the direct method, which involves isolating and identifying tumor-reactive T cells from tumor-infiltrating lymphocytes (TILs) or peripheral blood lymphocytes (PBLs) in patients. It is crucial to validate the reactivity of T cells towards tumors by studying the interactions between cancer cells and T cells in more depth. However, identifying T cells and their corresponding receptors with the ideal affinity for cancer antigens is challenging, as these antigens are often unmutated self-proteins expressed at higher levels in cancer cells than in normal tissues.
To generate antitumor T cells with optimal affinity of T cell receptor (TCR) to cancer-associated antigens, Obenaus et al. (2015) reported the potential of using antigen-negative humanized mice to generate T cells with a diverse human TCR repertoire and isolated the TCR specific to cancer antigens MAGE-A1 and NY-ESO-1, which have better affinity compared to human-derived TCRs [14]. Several other studies have attempted to enrich naturally occurring T cells from cancer patients by co-culturing TILs or PBLs with autologous tumors and expanding tumor-specific T cells before infusing them back into the patient [15][16][17][18]. However, the low frequency of T cells specific for these antigens makes it difficult to identify TCR T cells specific for cancer antigens. More recently, Arnaud et al. (2022) described a prediction method called NeoScreen that potentially identifies rare tumor antigens and their cognate TCR from TILs, enabling the selective expansion of antigen-specific T cells [19]. They reported that T cells transduced with target specific TCR identification via NeoScreen mediate tumor regression in preclinical models of cancer. Several T cell-based strategies have been developed to target tumor antigens that are well tolerated and show clinical promise in cancer patients. Overall, identifying tumor antigenic epitopes for T cell immunotherapy is crucial for the success of this promising approach to cancer treatment.

2.2. HLA Restricted T Cell-Based Immunotherapy

HLA dependent T cell-based immunotherapy employs the use of lymphocytes with minimally modified TCR or naturally occurring TCR directed against the tumor-specific antigen processed and presented by antigen-presenting cells via HLA molecules. This approach utilizes the ability of T cells to recognize the tumor antigens including cancer-associated or tumor-specific antigens presented by the antigen-presenting molecules on the cell surface. Many approaches have been employed for cancer immunotherapy in patients, such as endogenous T cells targeting undefined or defined antigens including cancer-associated or cancer-specific antigens.

2.3. Non-HLA Restricted T Cell-Based Immunotherapy

HLA-independent TCR-based T cell immunotherapy employs use of lymphocytes modified with TCR that are directed against the tumor antigens directly presented on the tumor cells. This approach utilizes a chimeric receptor introduced into the immune effector cells, such as T cells, natural killer cells or gamma delta (γδ) T cells to recognize tumor cell surface proteins and are commonly called as chimeric antigen receptor T or natural killer cells.

References

  1. Dunn, G.P.; Old, L.J.; Schreiber, R.D. The three Es of cancer immunoediting. Annu. Rev. Immunol. 2004, 22, 329–360.
  2. Burnet, M. Cancer: A biological approach. III. Viruses associated with neoplastic conditions. IV. Practical applications. Br. Med. J. 1957, 1, 841–847.
  3. Kim, R.; Emi, M.; Tanabe, K. Cancer immunoediting from immune surveillance to immune escape. Immunology 2007, 121, 1–14.
  4. Miller, J.F.; Mitchell, G.F.; Weiss, N.S. Cellular basis of the immunological defects in thymectomized mice. Nature 1967, 214, 992–997.
  5. Shankaran, V.; Ikeda, H.; Bruce, A.T.; White, J.M.; Swanson, P.E.; Old, L.J.; Schreiber, R.D. IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 2001, 410, 1107–1111.
  6. Dunn, G.P.; Bruce, A.T.; Ikeda, H.; Old, L.J.; Schreiber, R.D. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998.
  7. van der Bruggen, P.; Traversari, C.; Chomez, P.; Lurquin, C.; De Plaen, E.; van den Eynde, B.; Knuth, A.; Boon, T. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science 1991, 254, 1643–1647.
  8. Douek, D.C.; McFarland, R.D.; Keiser, P.H.; Gage, E.A.; Massey, J.M.; Haynes, B.F.; Polis, M.A.; Haase, A.T.; Feinberg, M.B.; Sullivan, J.L.; et al. Changes in thymic function with age and during the treatment of HIV infection. Nature 1998, 396, 690–695.
  9. Junge, S.; Kloeckener-Gruissem, B.; Zufferey, R.; Keisker, A.; Salgo, B.; Fauchere, J.C.; Scherer, F.; Shalaby, T.; Grotzer, M.; Siler, U.; et al. Correlation between recent thymic emigrants and CD31+ (PECAM-1) CD4+ T cells in normal individuals during aging and in lymphopenic children. Eur. J. Immunol. 2007, 37, 3270–3280.
  10. Hazenberg, M.D.; Verschuren, M.C.; Hamann, D.; Miedema, F.; van Dongen, J.J. T cell receptor excision circles as markers for recent thymic emigrants: Basic aspects, technical approach, and guidelines for interpretation. J. Mol. Med. 2001, 79, 631–640.
  11. Vrisekoop, N.; den Braber, I.; de Boer, A.B.; Ruiter, A.F.; Ackermans, M.T.; van der Crabben, S.N.; Schrijver, E.H.; Spierenburg, G.; Sauerwein, H.P.; Hazenberg, M.D.; et al. Sparse production but preferential incorporation of recently produced naive T cells in the human peripheral pool. Proc. Natl. Acad. Sci. USA 2008, 105, 6115–6120.
  12. Nikolich-Zugich, J. Ageing and life-long maintenance of T-cell subsets in the face of latent persistent infections. Nat. Rev. Immunol. 2008, 8, 512–522.
  13. Want, M.Y.; Konstorum, A.; Huang, R.Y.; Jain, V.; Matsueda, S.; Tsuji, T.; Lugade, A.; Odunsi, K.; Koya, R.; Battaglia, S. Neoantigens retention in patient derived xenograft models mediates autologous T cells activation in ovarian cancer. Oncoimmunology 2019, 8, e1586042.
  14. Obenaus, M.; Leitao, C.; Leisegang, M.; Chen, X.; Gavvovidis, I.; van der Bruggen, P.; Uckert, W.; Schendel, D.J.; Blankenstein, T. Identification of human T-cell receptors with optimal affinity to cancer antigens using antigen-negative humanized mice. Nat. Biotechnol. 2015, 33, 402–407.
  15. Dijkstra, K.K.; Cattaneo, C.M.; Weeber, F.; Chalabi, M.; van de Haar, J.; Fanchi, L.F.; Slagter, M.; van der Velden, D.L.; Kaing, S.; Kelderman, S.; et al. Generation of Tumor-Reactive T Cells by Co-culture of Peripheral Blood Lymphocytes and Tumor Organoids. Cell 2018, 174, 1586–1598.
  16. Chen, F.; Zou, Z.; Du, J.; Su, S.; Shao, J.; Meng, F.; Yang, J.; Xu, Q.; Ding, N.; Yang, Y.; et al. Neoantigen identification strategies enable personalized immunotherapy in refractory solid tumors. J. Clin. Invest. 2019, 129, 2056–2070.
  17. Prickett, T.D.; Crystal, J.S.; Cohen, C.J.; Pasetto, A.; Parkhurst, M.R.; Gartner, J.J.; Yao, X.; Wang, R.; Gros, A.; Li, Y.F.; et al. Durable Complete Response from Metastatic Melanoma after Transfer of Autologous T Cells Recognizing 10 Mutated Tumor Antigens. Cancer Immunol. Res. 2016, 4, 669–678.
  18. Tran, E.; Turcotte, S.; Gros, A.; Robbins, P.F.; Lu, Y.C.; Dudley, M.E.; Wunderlich, J.R.; Somerville, R.P.; Hogan, K.; Hinrichs, C.S.; et al. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science 2014, 344, 641–645.
  19. Arnaud, M.; Chiffelle, J.; Genolet, R.; Navarro Rodrigo, B.; Perez, M.A.S.; Huber, F.; Magnin, M.; Nguyen-Ngoc, T.; Guillaume, P.; Baumgaertner, P.; et al. Sensitive identification of neoantigens and cognate TCRs in human solid tumors. Nat. Biotechnol. 2022, 40, 656–660.
More
Information
Subjects: Immunology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 257
Revisions: 2 times (View History)
Update Date: 25 Apr 2023
1000/1000
ScholarVision Creations