PDF
Abstract
Background: Melanoma is recognized as a highly malignant cancer with a generally poor prognosis, underscoring the critical need for effective therapeutic strategies. Adoptive cell therapy has emerged as a promising modality to improve treatment outcomes in melanoma. For endogenous cell therapy (ECT), peripheral blood (PB) has traditionally served as the primary cell source. However, the potential of umbilical cord blood (UCB) as an alternative source for ECT remains unclear. Furthermore, the repertoire of TCRs remains limited. These deficiencies impede the optimization and broader application of ECT for melanoma, highlighting the necessity for focused investigations to resolve these issues.
Methods: To evaluate the effects of HLA-A2 restricted antigen-specific CD8+ T cells on melanoma cells, the cytotoxic activity of CD8+ T cells derived from UCB and PB were conducted in vivo and in vitro assays. Single-cell RNA sequencing combined with TCR V(D)J sequencing was employed to characterize cellular composition and quantify the frequencies of specific TCR clonotypes. The generation probability and peripheral occurrence probability of antigen-specific CD8+ TCR sequences from UCB and PB were computed using the Simple Olga Sonia algorithm. Finally, molecular docking simulations were conducted to predict the binding affinity between isolated TCRs and pMHC.
Results: No significant differences were observed in the cytotoxic effects mediated by antigen-specific CD8+ T cells derived from UCB versus PB. Phenotypic analysis revealed that PB-derived antigen-specific CD8+ T cells were predominantly effector and proliferating cells, whereas those from UCB consisted largely of memory cells. TCR sequencing identified a greater diversity of antigen-specific TCR clonotypes in PB, meanwhile UCB-derived TCRs exhibited strong pMHC binding. Molecular docking simulations confirmed high binding affinity between pMHC and TCR clones isolated from both sources.
Conclusions: Antigen-specific CD8+ T cells from UCB and PB display comparable cytotoxic efficacy against melanoma, albeit with distinct compositional profiles of antigen-specific CD8+ T cell subsets. Candidate TCRs can be effectively activated by the tumor-associated antigens MART1 and gp100. This activation promotes the expansion of the available TCR repertoires, thereby mitigating the previous constraint of a limited TCR library.
Keywords
antigen-specific TCR
/
cancer immunotherapy
/
gp100
/
MART1
/
melanoma
/
umbilical cord blood
Cite this article
Download citation ▾
Jiaji Liang, Xiao Jiang, Xi He, Zhiqin Dong, Jinqiang Lu, Shuxian Jiang, Hengyu Du, Haoran Mao, Songtao Luo, Xifeng An, Hongwei Liu.
Umbilical cord blood-derived cytotoxic T lymphocytes target melanoma via HLA-A2-restricted tumour antigens.
Clinical and Translational Medicine, 2025, 15(10): e70444 DOI:10.1002/ctm2.70444
| [1] |
Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019; 69(1): 7-34.
|
| [2] |
Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: gLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2019; 69(1): 7-34.
|
| [3] |
Jr GPG, Thomas CC, Thompson T, et al. Vital signs: melanoma incidence and mortality trends and projections-United States, 1982–2030. MMWR Morb Mortal Wkly Rep. 2015; 64(21): 591-596.
|
| [4] |
Neil Crowson A, Magro CM, Mihm MC. Prognosticators of melanoma, the melanoma report, and the sentinel lymph node. Mod Pathol. 2006; 19(2): S71-S87.
|
| [5] |
Barnhill RL, Katzen J, Spatz A, et al. The importance of mitotic rate as a prognostic factor for localized cutaneous melanoma. J Cutan Pathol. 2005; 32(4): 268-273.
|
| [6] |
Huang YS, Chen XX, Yang SX, et al. Preliminary exploration of the clinical features of Chinese patients with skin malignancies and premalignancies: a retrospective study of 1420 cases from Peking University First Hospital. J Eur Acad Dermatol Venereol. 2013; 27(9): 1114-1119.
|
| [7] |
Sheen Y-S, Tan K-T, Tse K-P, et al. Genetic alterations in primary melanoma in Taiwan. Br J Dermatol. 2020; 182(5): 1205-1213.
|
| [8] |
Yamazaki N, Takenouchi T, Fujimoto M, et al. Phase 1b study of pembrolizumab (MK-3475; anti-PD-1 monoclonal antibody) in Japanese patients with advanced melanoma (KEYNOTE-041). Cancer Chemother Pharmacol. 2017; 79(4): 651-660.
|
| [9] |
Shoushtari AN, Bao R, Luke JJ. PD-1 blockade in Chinese versus Western patients with melanoma. Clin Cancer Res. 2020; 26(16): 4171-4173.
|
| [10] |
Jhunjhunwala S, Hammer C, Delamarre L. Antigen presentation in cancer: insights into tumour immunogenicity and immune evasion. Nat Rev Cancer. 2021; 21(5): 298-312.
|
| [11] |
Salmaninejad A, Zamani MR, Pourvahedi M, et al. Cancer/Testis antigens: expression, regulation, tumor invasion, and use in immunotherapy of cancers. Immunol Invest. 2016; 45(7): 619-640.
|
| [12] |
Aris M, Zubieta MR, Colombo M, et al. MART-1- and gp100-expressing and -non-expressing melanoma cells are equally proliferative in tumors and clonogenic in vitro. J Invest Dermatol. 2012; 132(2): 365-374.
|
| [13] |
Yee C. The use of endogenous T cells for adoptive transfer. Immunol Rev Immunol Rev. 2014; 257(1): 250-263.
|
| [14] |
Pollack SM, Jones RL, Farrar EA, et al. Tetramer guided, cell sorter assisted production of clinical grade autologous NY-ESO-1 specific CD8(+) T cells. J Immunother Cancer. 2014; 2(1): 36.
|
| [15] |
Wölfl M, Greenberg PD. Antigen-specific activation and cytokine-facilitated expansion of naive, human CD8+ T cells. Nat Protoc. 2014; 9(4): 950-966.
|
| [16] |
Sutherland DR, Keating A. The CD34 antigen: structure, biology, and potential clinical applications. J Hematother. 1992; 1(2): 115-129.
|
| [17] |
Sutherland DR, Keating A, Nayar R, et al. Sensitive detection and enumeration of CD34+ cells in peripheral and cord blood by flow cytometry. Exp Hematol. 1994; 22(10): 1003-1010.
|
| [18] |
Rocha V, Wagner Jr JE, Sobocinski KA, et al. Graft-versus-host disease in children who have received a cord-blood or bone marrow transplant from an HLA-identical sibling. Eurocord and International Bone Marrow Transplant Registry Working Committee on Alternative Donor and Stem Cell Sources. N Engl J Med. 2000; 342(25): 1846-1854.
|
| [19] |
Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998; 392(6673): 245-252.
|
| [20] |
Yee C, Thompson JA, Byrd D, et al. Adoptive T cell therapy using antigen-specific CD8 T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells. Proc Natl Acad Sci U S A. 2002; 99(25): 16168-16173.
|
| [21] |
Li Y, Bleakley M, Yee C. IL-21 influences the frequency, phenotype, and affinity of the antigen-specific CD8 T cell response. J Immunol. 2005; 175(4): 2261-2269.
|
| [22] |
Kim Ji-H, Lee K-J, Lee S-W. Cancer immunotherapy with T-cell targeting cytokines: iL-2 and IL-7. BMB Rep. 2021; 54(1): 21-30.
|
| [23] |
Brucker C, Reimann J, Wagner H, et al. Clonal activation of cytotoxic lymphocyte precursors by monoclonal anti-CD3 antibody: analysis of feeder cell requirements. Immunol Lett. 1987; 14(2): 121-125.
|
| [24] |
Zhong M, Gao R, Zhao R, et al. BET bromodomain inhibition rescues PD-1-mediated T-cell exhaustion in acute myeloid leukemia. Cell Death Dis. 2022; 13(8): 671.
|
| [25] |
Hunder NN, Wallen H, Cao J, et al. Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1. N Engl J Med. 2008; 358(25): 2698-2703.
|
| [26] |
Altman JD, Moss PA, Goulder PJ, et al. Phenotypic analysis of antigen-specific T lymphocytes. Science. 1996; 274(5284): 94-96.
|
| [27] |
García-Tuñón I, Ricote M, Antonio Ruiz A, et al. Influence of IFN-gamma and its receptors in human breast cancer. BMC Cancer. 2007; 7(1): 158.
|
| [28] |
Chen Y, Wang D, Li Y, et al. Spatiotemporal single-cell analysis decodes cellular dynamics underlying different responses to immunotherapy in colorectal cancer. Cancer Cell. 2024; 42(7): 1268-1285.e7.
|
| [29] |
Zhang B, Ren Z, Zhao J, et al. Global analysis of HLA-A2 restricted MAGE-A3 tumor antigen epitopes and corresponding TCRs in non-small cell lung cancer. Theranostics. 2023; 13(13): 4449-4468.
|
| [30] |
Isacchini G, Olivares C, Nourmohammad A, Walczak AM, Mora T. SOS: online probability estimation and generation of T- and B-cell receptors. Bioinformatics. 2020; 36(16): 4510-4512.
|
| [31] |
Zhang Ze, Xiong D, Wang X, et al. Mapping the functional landscape of T cell receptor repertoires by single-T cell transcriptomics. Nat Methods. 2021; 18(1): 92-99.
|
| [32] |
Takanashi M, Atsuta Y, Fujiwara K, et al. The impact of anti-HLA antibodies on unrelated cord blood transplantations. Blood. 2010; 116(15): 2839-2846.
|
| [33] |
Ruggeri A, Rocha V, Masson E, et al. Impact of donor-specific anti-HLA antibodies on graft failure and survival after reduced intensity conditioning-unrelated cord blood transplantation: a Eurocord, Societe Francophone d'Histocompatibilite et d'Immunogenetique (SFHI) and Societe Francaise de Greffe de Moelle et de Therapie Cellulaire (SFGM-TC) analysis. Haematologica. 2013; 98(7): 1154-1160.
|
| [34] |
Rocha V, Spellman S, Zhang M-J, et al. Effect of HLA-matching recipients to donor noninherited maternal antigens on outcomes after mismatched umbilical cord blood transplantation for hematologic malignancy. Biol Blood Marrow Transplant. 2012; 18(12): 1890-1896.
|
| [35] |
van Rood JJ, Stevens CE, Smits J, et al. Reexposure of cord blood to noninherited maternal HLA antigens improves transplant outcome in hematological malignancies. Proc Natl Acad Sci U S A. 2009; 106(47): 19952-19957.
|
| [36] |
Kurtzberg J. Update on umbilical cord blood transplantation. Curr Opin Pediatr. 2009; 21(1): 22-29.
|
| [37] |
Paloczi K. Immunophenotypic and functional characterization of human umbilical cord blood mononuclear cells. Leukemia. 1999; 13(1): S87-S89.
|
| [38] |
Okas M, Gertow J, Uzunel M, et al. Clinical expansion of cord blood-derived T cells for use as donor lymphocyte infusion after cord blood transplantation. J Immunother. 2010; 33(1): 96-105.
|
| [39] |
Fabricius WA, Ramanathan M. Review on haploidentical hematopoietic cell transplantation in patients with hematologic malignancies. Adv Hematol. 2016; 2016: 5726132.
|
| [40] |
Gluckman EG, Roch VV, Chastang C. Use of cord blood cells for banking and transplant. Oncologist. 1997; 2(5): 340-343.
|
| [41] |
Tanay A, Regev A. Scaling single-cell genomics from phenomenology to mechanism. Nature. 2017; 541(7637): 331-338.
|
| [42] |
Gutcher I, Becher B. APC-derived cytokines and T cell polarization in autoimmune inflammation. J Clin Investig. 2007; 117(5): 1119-1127.
|
| [43] |
Smith-Garvin JE, Koretzky GA, Jordan MS. T cell activation. Annu Rev Immunol. 2009; 27: 591-619.
|
| [44] |
Neurauter AA, Bonyhadi M, Lien E, et al. Cell isolation and expansion using Dynabeads. Adv Biochem Eng Biotechnol. 2007; 106: 41-73.
|
| [45] |
Maus MV, Thomas AK, Leonard DG, et al. Ex vivo expansion of polyclonal and antigen-specific cytotoxic T lymphocytes by artificial APCs expressing ligands for the T-cell receptor, CD28 and 4-1BB. Nat Biotechnol. 2002; 20(2): 143-148.
|
| [46] |
Xu H, Wang N, Cao W, Huang L, Zhou J, Sheng L. Influence of various medium environment to in vitro human T cell culture. In Vitro Cell Dev Biol Anim. 2018; 54(8): 559-566.
|
| [47] |
Sato K, Kondo M, Sakuta K, et al. Impact of culture medium on the expansion of T cells for immunotherapy. Cytotherapy. 2009; 11(7): 936-946.
|
| [48] |
Gebhardt T, Park SL, Parish IA. Stem-like exhausted and memory CD8+ T cells in cancer. Nat Rev Cancer. 2023; 23(11): 780-798.
|
| [49] |
Okła K, Farber DL, Zou W. Tissue-resident memory T cells in tumor immunity and immunotherapy. J Exp Med. 2021; 218(4): e20201605.
|
| [50] |
Dolton G, Rius C, Wall A, et al. Targeting of multiple tumor-associated antigens by individual T cell receptors during successful cancer immunotherapy. Cell. 2023; 186(16): 3333-3349.e27.
|
| [51] |
Eltahla AA, Rizzetto S, Pirozyan MR, et al. Linking the T cell receptor to the single cell transcriptome in antigen-specific human T cells. Immunol Cell Biol. 2016; 94(6): 604-611.
|
| [52] |
Tirosh I, Izar B, Prakadan SM, et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science. 2016; 352(6282): 189-196.
|
| [53] |
Li H, van der Leun AM, Yofe I, et al. Dysfunctional CD8 T cells form a proliferative, dynamically regulated compartment within human melanoma. Cell. 2019; 176(4): 775-789.e18.
|
| [54] |
Sade-Feldman M, Yizhak K, Bjorgaard SL, et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell. 2018; 175(4): 998-1013.e20.
|
| [55] |
Sibener LV, Fernandes RA, Kolawole EM, et al. Isolation of a structural mechanism for uncoupling T cell receptor signaling from peptide-MHC binding. Cell. 2018; 174(3): 672-687.e27.
|
| [56] |
Yang X, Garner LI, Zvyagin IV, et al. Autoimmunity-associated T cell receptors recognize HLA-B*27-bound peptides. Nature. 2022; 612(7941): 771-777.
|
| [57] |
Sewell AK. Why must T cells be cross-reactive? Nat Rev Immunol. 2012; 12(9): 669-677.
|
| [58] |
Song I, Gil A, Mishra R, Ghersi D, Selin LK, Stern LJ. Broad TCR repertoire and diverse structural solutions for recognition of an immunodominant CD8+ T cell epitope. Nat Struct Mol Biol. 2017; 24: 395-406.
|
| [59] |
Montemurro A, Schuster V, Povlsen HR, et al. NetTCR-2.0 enables accurate prediction of TCR-peptide binding by using paired TCRα and β sequence data. Commun Biol. 2021; 4(1): 1060.
|
| [60] |
Gielis S, Meysman P, et al. Identification of epitope-specific T cells in T-cell receptor repertoires. Methods Mol Biol. 2020; 2120: 183-195.
|
| [61] |
Homma F, Huang J, van der Hoorn RAL. AlphaFold-Multimer predicts cross-kingdom interactions at the plant-pathogen interface. Nat Commun. 2023; 14(1): 6040.
|
| [62] |
Chuprin J, Buettner H, Seedhom MO, et al. Humanized mouse models for immuno-oncology research. Nat Rev Clin Oncol. 2023; 20(3): 192-206.
|
RIGHTS & PERMISSIONS
2025 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.