Skip to main content
Top
Published in: Journal of Hematology & Oncology 1/2019

Open Access 01-12-2019 | Melanoma | Review

Targeting cancers through TCR-peptide/MHC interactions

Authors: Qinghua He, Xianhan Jiang, Xinke Zhou, Jinsheng Weng

Published in: Journal of Hematology & Oncology | Issue 1/2019

Login to get access

Abstract

Adoptive T cell therapy has achieved dramatic success in a clinic, and the Food and Drug Administration approved two chimeric antigen receptor-engineered T cell (CAR-T) therapies that target hematological cancers in 2018. A significant issue faced by CAR-T therapies is the lack of tumor-specific biomarkers on the surfaces of solid tumor cells, which hampers the application of CAR-T therapies to solid tumors. Intracellular tumor-related antigens can be presented as peptides in the major histocompatibility complex (MHC) on the cell surface, which interact with the T cell receptors (TCR) on antigen-specific T cells to stimulate an anti-tumor response. Multiple immunotherapy strategies have been developed to eradicate tumor cells through targeting the TCR-peptide/MHC interactions. Here, we summarize the current status of TCR-based immunotherapy strategies, with particular focus on the TCR structure, activated signaling pathways, the effects and toxicity associated with TCR-based therapies in clinical trials, preclinical studies examining immune-mobilizing monoclonal TCRs against cancer (ImmTACs), and TCR-fusion molecules. We propose several TCR-based therapeutic strategies to achieve optimal clinical responses without the induction of autoimmune diseases.
Literature
3.
go back to reference Zheng P-P, Kros JM, Li J. Approved CAR T cell therapies: ice bucket challenges on glaring safety risks and long-term impacts. Drug Discov Today. 2018;23(6):1175–82.PubMedCrossRef Zheng P-P, Kros JM, Li J. Approved CAR T cell therapies: ice bucket challenges on glaring safety risks and long-term impacts. Drug Discov Today. 2018;23(6):1175–82.PubMedCrossRef
4.
go back to reference Zhao J, Song Y, Liu D. Clinical trials of dual-target CAR T cells, donor-derived CAR T cells, and universal CAR T cells for acute lymphoid leukemia. J Hematol Oncol. 2019;12(1):17.PubMedPubMedCentralCrossRef Zhao J, Song Y, Liu D. Clinical trials of dual-target CAR T cells, donor-derived CAR T cells, and universal CAR T cells for acute lymphoid leukemia. J Hematol Oncol. 2019;12(1):17.PubMedPubMedCentralCrossRef
5.
go back to reference Lamers CH, et al. Gene-modified T cells for adoptive immunotherapy of renal cell cancer maintain transgene-specific immune functions in vivo. Cancer Immunol Immunother. 2007;56(12):1875–83.PubMedCrossRef Lamers CH, et al. Gene-modified T cells for adoptive immunotherapy of renal cell cancer maintain transgene-specific immune functions in vivo. Cancer Immunol Immunother. 2007;56(12):1875–83.PubMedCrossRef
6.
go back to reference Kershaw MH, et al. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res. 2006;12(20 Pt 1):6106–15.PubMedPubMedCentralCrossRef Kershaw MH, et al. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res. 2006;12(20 Pt 1):6106–15.PubMedPubMedCentralCrossRef
8.
go back to reference Lamers CHJ, et al. Treatment of metastatic renal cell carcinoma with CAIX CAR-engineered T cells: clinical evaluation and management of on-target toxicity. Mol Ther. 2013;21(4):904–12.PubMedPubMedCentralCrossRef Lamers CHJ, et al. Treatment of metastatic renal cell carcinoma with CAIX CAR-engineered T cells: clinical evaluation and management of on-target toxicity. Mol Ther. 2013;21(4):904–12.PubMedPubMedCentralCrossRef
9.
go back to reference Park JR, et al. Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. Mol Ther. 2007;15(4):825–33.PubMedCrossRef Park JR, et al. Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. Mol Ther. 2007;15(4):825–33.PubMedCrossRef
11.
go back to reference Weekes MP, et al. Comparative analysis of techniques to purify plasma membrane proteins. J Biomol Tech. 2010;21(3):108–15.PubMedPubMedCentral Weekes MP, et al. Comparative analysis of techniques to purify plasma membrane proteins. J Biomol Tech. 2010;21(3):108–15.PubMedPubMedCentral
12.
go back to reference Zinkernagel RM, Doherty PC. Restriction of in vitro T cell-mediated cytotoxicity in lymphocytic choriomeningitis within a syngeneic or semiallogeneic system. Nature. 1974;248(5450):701–2.PubMedCrossRef Zinkernagel RM, Doherty PC. Restriction of in vitro T cell-mediated cytotoxicity in lymphocytic choriomeningitis within a syngeneic or semiallogeneic system. Nature. 1974;248(5450):701–2.PubMedCrossRef
13.
go back to reference Hedrick SM, et al. Isolation of cDNA clones encoding T cell-specific membrane-associated proteins. Nature. 1984;308(5955):149–53.PubMedCrossRef Hedrick SM, et al. Isolation of cDNA clones encoding T cell-specific membrane-associated proteins. Nature. 1984;308(5955):149–53.PubMedCrossRef
14.
go back to reference Yanagi Y, et al. A human T cell-specific cDNA clone encodes a protein having extensive homology to immunoglobulin chains. Nature. 1984;308(5955):145–9.PubMedCrossRef Yanagi Y, et al. A human T cell-specific cDNA clone encodes a protein having extensive homology to immunoglobulin chains. Nature. 1984;308(5955):145–9.PubMedCrossRef
15.
go back to reference Neefjes J, et al. Towards a systems understanding of MHC class I and MHC class II antigen presentation. Nat Rev Immunol. 2011;11:823.PubMedCrossRef Neefjes J, et al. Towards a systems understanding of MHC class I and MHC class II antigen presentation. Nat Rev Immunol. 2011;11:823.PubMedCrossRef
17.
go back to reference Novellino L, Castelli C, Parmiani G. A listing of human tumor antigens recognized by T cells: March 2004 update. Cancer Immunol Immunother. 2005;54(3):187–207.PubMedCrossRef Novellino L, Castelli C, Parmiani G. A listing of human tumor antigens recognized by T cells: March 2004 update. Cancer Immunol Immunother. 2005;54(3):187–207.PubMedCrossRef
18.
go back to reference Andersen RS, et al. Dissection of T-cell antigen specificity in human melanoma. Cancer Res. 2012;72(7):1642–50.PubMedCrossRef Andersen RS, et al. Dissection of T-cell antigen specificity in human melanoma. Cancer Res. 2012;72(7):1642–50.PubMedCrossRef
19.
go back to reference Corse E, Gottschalk RA, Allison JP. Strength of TCR–peptide/MHC interactions and in vivo T cell responses. J Immunol. 2011;186(9):5039–45.PubMedCrossRef Corse E, Gottschalk RA, Allison JP. Strength of TCR–peptide/MHC interactions and in vivo T cell responses. J Immunol. 2011;186(9):5039–45.PubMedCrossRef
20.
go back to reference Coulie PG, et al. Tumour antigens recognized by T lymphocytes: at the core of cancer immunotherapy. Nat Rev Cancer. 2014;14:135.PubMedCrossRef Coulie PG, et al. Tumour antigens recognized by T lymphocytes: at the core of cancer immunotherapy. Nat Rev Cancer. 2014;14:135.PubMedCrossRef
21.
go back to reference Kunert A, et al. TCR-engineered T cells meet new challenges to treat solid tumors: choice of antigen, T cell fitness, and sensitization of tumor milieu. Front Immunol. 2013;4:363.PubMedPubMedCentralCrossRef Kunert A, et al. TCR-engineered T cells meet new challenges to treat solid tumors: choice of antigen, T cell fitness, and sensitization of tumor milieu. Front Immunol. 2013;4:363.PubMedPubMedCentralCrossRef
22.
go back to reference Ping Y, Liu C, Zhang Y. T-cell receptor-engineered T cells for cancer treatment: current status and future directions. Protein Cell. 2018;9(3):254–66.CrossRefPubMed Ping Y, Liu C, Zhang Y. T-cell receptor-engineered T cells for cancer treatment: current status and future directions. Protein Cell. 2018;9(3):254–66.CrossRefPubMed
23.
go back to reference Zhang J, Wang L. The emerging world of TCR-T cell trials against cancer: a systematic review. Technol Cancer Res Treat. 2019;18:1533033819831068.PubMedPubMedCentral Zhang J, Wang L. The emerging world of TCR-T cell trials against cancer: a systematic review. Technol Cancer Res Treat. 2019;18:1533033819831068.PubMedPubMedCentral
24.
go back to reference van der Merwe PA, Dushek O. Mechanisms for T cell receptor triggering. Nat Rev Immunol. 2010;11:47.PubMedCrossRef van der Merwe PA, Dushek O. Mechanisms for T cell receptor triggering. Nat Rev Immunol. 2010;11:47.PubMedCrossRef
25.
go back to reference Davis MM, Bjorkman PJ. T-cell antigen receptor genes and T-cell recognition. Nature. 1988;334(6181):395–402.PubMedCrossRef Davis MM, Bjorkman PJ. T-cell antigen receptor genes and T-cell recognition. Nature. 1988;334(6181):395–402.PubMedCrossRef
26.
go back to reference Pannetier C, Even J, Kourilsky P. T-cell repertoire diversity and clonal expansions in normal and clinical samples. Immunol Today. 1995;16(4):176–81.PubMedCrossRef Pannetier C, Even J, Kourilsky P. T-cell repertoire diversity and clonal expansions in normal and clinical samples. Immunol Today. 1995;16(4):176–81.PubMedCrossRef
27.
go back to reference Wucherpfennig KW, et al. Structural biology of the T-cell receptor: insights into receptor assembly, ligand recognition, and initiation of signaling. Cold Spring Harb Perspect Biol. 2010;2(4):a005140.PubMedPubMedCentralCrossRef Wucherpfennig KW, et al. Structural biology of the T-cell receptor: insights into receptor assembly, ligand recognition, and initiation of signaling. Cold Spring Harb Perspect Biol. 2010;2(4):a005140.PubMedPubMedCentralCrossRef
28.
go back to reference Alarcon B, et al. Initiation of TCR signaling: regulation within CD3 dimers. Immunol Rev. 2003;191:38–46.PubMedCrossRef Alarcon B, et al. Initiation of TCR signaling: regulation within CD3 dimers. Immunol Rev. 2003;191:38–46.PubMedCrossRef
29.
go back to reference Koretzky GA, Abtahian F, Silverman MA. SLP76 and SLP65: complex regulation of signalling in lymphocytes and beyond. Nat Rev Immunol. 2006;6(1):67–78.PubMedCrossRef Koretzky GA, Abtahian F, Silverman MA. SLP76 and SLP65: complex regulation of signalling in lymphocytes and beyond. Nat Rev Immunol. 2006;6(1):67–78.PubMedCrossRef
31.
go back to reference Dembic Z, et al. Transfer of specificity by murine alpha and beta T-cell receptor genes. Nature. 1986;320(6059):232–8.PubMedCrossRef Dembic Z, et al. Transfer of specificity by murine alpha and beta T-cell receptor genes. Nature. 1986;320(6059):232–8.PubMedCrossRef
32.
33.
go back to reference Calogero A, et al. Retargeting of a T cell line by anti MAGE-3/HLA-A2 alpha beta TCR gene transfer. Anticancer Res. 2000;20(3a):1793–9.PubMed Calogero A, et al. Retargeting of a T cell line by anti MAGE-3/HLA-A2 alpha beta TCR gene transfer. Anticancer Res. 2000;20(3a):1793–9.PubMed
34.
go back to reference Clay TM, et al. Efficient transfer of a tumor antigen-reactive TCR to human peripheral blood lymphocytes confers anti-tumor reactivity. J Immunol. 1999;163(1):507–13.PubMed Clay TM, et al. Efficient transfer of a tumor antigen-reactive TCR to human peripheral blood lymphocytes confers anti-tumor reactivity. J Immunol. 1999;163(1):507–13.PubMed
35.
go back to reference Hughes MS, et al. Transfer of a TCR gene derived from a patient with a marked antitumor response conveys highly active T-cell effector functions. Hum Gene Ther. 2005;16(4):457–72.PubMedCrossRef Hughes MS, et al. Transfer of a TCR gene derived from a patient with a marked antitumor response conveys highly active T-cell effector functions. Hum Gene Ther. 2005;16(4):457–72.PubMedCrossRef
36.
go back to reference Cole DJ, et al. Characterization of the functional specificity of a cloned T-cell receptor heterodimer recognizing the MART-1 melanoma antigen. Cancer Res. 1995;55(4):748–52.PubMed Cole DJ, et al. Characterization of the functional specificity of a cloned T-cell receptor heterodimer recognizing the MART-1 melanoma antigen. Cancer Res. 1995;55(4):748–52.PubMed
37.
go back to reference Cooper LJ, et al. Transfer of specificity for human immunodeficiency virus type 1 into primary human T lymphocytes by introduction of T-cell receptor genes. J Virol. 2000;74(17):8207–12.PubMedPubMedCentralCrossRef Cooper LJ, et al. Transfer of specificity for human immunodeficiency virus type 1 into primary human T lymphocytes by introduction of T-cell receptor genes. J Virol. 2000;74(17):8207–12.PubMedPubMedCentralCrossRef
38.
go back to reference Ueno T, et al. Reconstitution of anti-HIV effector functions of primary human CD8 T lymphocytes by transfer of HIV-specific αβ TCR genes. Eur J Immunol. 2004;34(12):3379–88.PubMedCrossRef Ueno T, et al. Reconstitution of anti-HIV effector functions of primary human CD8 T lymphocytes by transfer of HIV-specific αβ TCR genes. Eur J Immunol. 2004;34(12):3379–88.PubMedCrossRef
39.
go back to reference Callender GG, et al. Identification of a hepatitis C virus–reactive T cell receptor that does not require CD8 for target cell recognition. Hepatology. 2006;43(5):973–81.PubMedCrossRef Callender GG, et al. Identification of a hepatitis C virus–reactive T cell receptor that does not require CD8 for target cell recognition. Hepatology. 2006;43(5):973–81.PubMedCrossRef
40.
go back to reference Heemskerk MH, et al. Dual HLA class I and class II restricted recognition of alloreactive T lymphocytes mediated by a single T cell receptor complex. Proc Natl Acad Sci U S A. 2001;98(12):6806–11.PubMedPubMedCentralCrossRef Heemskerk MH, et al. Dual HLA class I and class II restricted recognition of alloreactive T lymphocytes mediated by a single T cell receptor complex. Proc Natl Acad Sci U S A. 2001;98(12):6806–11.PubMedPubMedCentralCrossRef
41.
go back to reference Orentas RJ, et al. Retroviral transduction of a T cell receptor specific for an Epstein–Barr virus-encoded peptide. Clin Immunol. 2001;98(2):220–8.PubMedCrossRef Orentas RJ, et al. Retroviral transduction of a T cell receptor specific for an Epstein–Barr virus-encoded peptide. Clin Immunol. 2001;98(2):220–8.PubMedCrossRef
42.
go back to reference Stanislawski T, et al. Circumventing tolerance to a human MDM2-derived tumor antigen by TCR gene transfer. Nat Immunol. 2001;2(10):962–70.PubMedCrossRef Stanislawski T, et al. Circumventing tolerance to a human MDM2-derived tumor antigen by TCR gene transfer. Nat Immunol. 2001;2(10):962–70.PubMedCrossRef
43.
go back to reference Zhao Y, et al. Primary human lymphocytes transduced with NY-ESO-1 antigen-specific TCR genes recognize and kill diverse human tumor cell lines. J Immunol. 2005;174(7):4415–23.PubMedCrossRef Zhao Y, et al. Primary human lymphocytes transduced with NY-ESO-1 antigen-specific TCR genes recognize and kill diverse human tumor cell lines. J Immunol. 2005;174(7):4415–23.PubMedCrossRef
44.
go back to reference Willemsen RA, et al. Grafting primary human T lymphocytes with cancer-specific chimeric single chain and two chain TCR. Gene Ther. 2000;7(16):1369–77.PubMedCrossRef Willemsen RA, et al. Grafting primary human T lymphocytes with cancer-specific chimeric single chain and two chain TCR. Gene Ther. 2000;7(16):1369–77.PubMedCrossRef
45.
go back to reference Morgan RA, et al. High efficiency TCR gene transfer into primary human lymphocytes affords avid recognition of melanoma tumor antigen glycoprotein 100 and does not alter the recognition of autologous melanoma antigens. J Immunol. 2003;171(6):3287–95.PubMedCrossRef Morgan RA, et al. High efficiency TCR gene transfer into primary human lymphocytes affords avid recognition of melanoma tumor antigen glycoprotein 100 and does not alter the recognition of autologous melanoma antigens. J Immunol. 2003;171(6):3287–95.PubMedCrossRef
46.
go back to reference Schaft N, et al. Peptide fine specificity of anti-glycoprotein 100 CTL is preserved following transfer of engineered TCRαβ genes into primary human T lymphocytes. J Immunol. 2003;170(4):2186–94.PubMedCrossRef Schaft N, et al. Peptide fine specificity of anti-glycoprotein 100 CTL is preserved following transfer of engineered TCRαβ genes into primary human T lymphocytes. J Immunol. 2003;170(4):2186–94.PubMedCrossRef
47.
go back to reference Cohen CJ, et al. Recognition of fresh human tumor by human peripheral blood lymphocytes transduced with a bicistronic retroviral vector encoding a murine anti-p53 TCR. J Immunol. 2005;175(9):5799–808.PubMedCrossRef Cohen CJ, et al. Recognition of fresh human tumor by human peripheral blood lymphocytes transduced with a bicistronic retroviral vector encoding a murine anti-p53 TCR. J Immunol. 2005;175(9):5799–808.PubMedCrossRef
48.
go back to reference Scholten KBJ, et al. Preservation and redirection of HPV16E7-specific T cell receptors for immunotherapy of cervical cancer. Clin Immunol. 2005;114(2):119–29.PubMedCrossRef Scholten KBJ, et al. Preservation and redirection of HPV16E7-specific T cell receptors for immunotherapy of cervical cancer. Clin Immunol. 2005;114(2):119–29.PubMedCrossRef
49.
go back to reference Heemskerk MHM, et al. Redirection of antileukemic reactivity of peripheral T lymphocytes using gene transfer of minor histocompatibility antigen HA-2-specific T-cell receptor complexes expressing a conserved alpha joining region. Blood. 2003;102(10):3530–40.PubMedCrossRef Heemskerk MHM, et al. Redirection of antileukemic reactivity of peripheral T lymphocytes using gene transfer of minor histocompatibility antigen HA-2-specific T-cell receptor complexes expressing a conserved alpha joining region. Blood. 2003;102(10):3530–40.PubMedCrossRef
50.
go back to reference Mommaas B, et al. Adult and cord blood T cells can acquire HA-1 specificity through HA-1 T-cell receptor gene transfer. Haematologica. 2005;90(10):1415–21.PubMed Mommaas B, et al. Adult and cord blood T cells can acquire HA-1 specificity through HA-1 T-cell receptor gene transfer. Haematologica. 2005;90(10):1415–21.PubMed
51.
go back to reference Ivanov R, et al. UTY-specific TCR-transfer generates potential graft-versus-leukaemia effector T cells. Br J Haematol. 2005;129(3):392–402.PubMedCrossRef Ivanov R, et al. UTY-specific TCR-transfer generates potential graft-versus-leukaemia effector T cells. Br J Haematol. 2005;129(3):392–402.PubMedCrossRef
52.
go back to reference Ivanov R, et al. T cell receptor-transgenic primary T cells as a tool for discovery of leukaemia-associated antigens. Clin Exp Immunol. 2006;143(1):78–84.PubMedPubMedCentralCrossRef Ivanov R, et al. T cell receptor-transgenic primary T cells as a tool for discovery of leukaemia-associated antigens. Clin Exp Immunol. 2006;143(1):78–84.PubMedPubMedCentralCrossRef
53.
go back to reference Roszkowski JJ, et al. Simultaneous generation of CD8<sup>+</sup> and CD4<sup>+</sup> melanoma-reactive T cells by retroviral-mediated transfer of a single T-cell receptor. Cancer Res. 2005;65(4):1570–6.PubMedCrossRef Roszkowski JJ, et al. Simultaneous generation of CD8<sup>+</sup> and CD4<sup>+</sup> melanoma-reactive T cells by retroviral-mediated transfer of a single T-cell receptor. Cancer Res. 2005;65(4):1570–6.PubMedCrossRef
54.
go back to reference van der Veken LT, et al. HLA class II restricted T-cell receptor gene transfer generates CD4+ T cells with helper activity as well as cytotoxic capacity. Gene Ther. 2005;12(23):1686–95.PubMedCrossRef van der Veken LT, et al. HLA class II restricted T-cell receptor gene transfer generates CD4+ T cells with helper activity as well as cytotoxic capacity. Gene Ther. 2005;12(23):1686–95.PubMedCrossRef
55.
go back to reference Aarnoudse CA, et al. TCR reconstitution in Jurkat reporter cells facilitates the identification of novel tumor antigens by cDNA expression cloning. Int J Cancer. 2002;99(1):7–13.PubMedCrossRef Aarnoudse CA, et al. TCR reconstitution in Jurkat reporter cells facilitates the identification of novel tumor antigens by cDNA expression cloning. Int J Cancer. 2002;99(1):7–13.PubMedCrossRef
56.
go back to reference Tsuji T, et al. Generation of tumor-specific, HLA class I–restricted human Th1 and Tc1 cells by cell engineering with tumor peptide–specific T-cell receptor genes. Blood. 2005;106(2):470–6.PubMedCrossRef Tsuji T, et al. Generation of tumor-specific, HLA class I–restricted human Th1 and Tc1 cells by cell engineering with tumor peptide–specific T-cell receptor genes. Blood. 2005;106(2):470–6.PubMedCrossRef
57.
go back to reference Xue S-A, et al. Elimination of human leukemia cells in <em>NOD/SCID</em> mice by <em>WT1-TCR</em> gene–transduced human T cells. Blood. 2005;106(9):3062–7.PubMedCrossRef Xue S-A, et al. Elimination of human leukemia cells in <em>NOD/SCID</em> mice by <em>WT1-TCR</em> gene–transduced human T cells. Blood. 2005;106(9):3062–7.PubMedCrossRef
58.
go back to reference Engels B, et al. Redirecting human T lymphocytes toward renal cell carcinoma specificity by retroviral transfer of T cell receptor genes. Hum Gene Ther. 2005;16(7):799–810.PubMedCrossRef Engels B, et al. Redirecting human T lymphocytes toward renal cell carcinoma specificity by retroviral transfer of T cell receptor genes. Hum Gene Ther. 2005;16(7):799–810.PubMedCrossRef
59.
go back to reference Tahara H, et al. Reconstitution of CD8<sup>+</sup> T cells by retroviral transfer of the TCR αβ-chain genes isolated from a clonally expanded P815-infiltrating lymphocyte. J Immunol. 2003;171(4):2154–60.PubMedCrossRef Tahara H, et al. Reconstitution of CD8<sup>+</sup> T cells by retroviral transfer of the TCR αβ-chain genes isolated from a clonally expanded P815-infiltrating lymphocyte. J Immunol. 2003;171(4):2154–60.PubMedCrossRef
60.
go back to reference Parkhurst MR, et al. Characterization of genetically modified T-cell receptors that recognize the CEA:691-699 peptide in the context of HLA-A2.1 on human colorectal cancer cells. Clin Cancer Res. 2009;15(1):169–80.PubMedPubMedCentralCrossRef Parkhurst MR, et al. Characterization of genetically modified T-cell receptors that recognize the CEA:691-699 peptide in the context of HLA-A2.1 on human colorectal cancer cells. Clin Cancer Res. 2009;15(1):169–80.PubMedPubMedCentralCrossRef
61.
go back to reference Voss RH, Kuball J, Theobald M. Designing TCR for cancer immunotherapy. Methods Mol Med. 2005;109:229–56.PubMed Voss RH, Kuball J, Theobald M. Designing TCR for cancer immunotherapy. Methods Mol Med. 2005;109:229–56.PubMed
62.
go back to reference Derbinski J, et al. Promiscuous gene expression in medullary thymic epithelial cells mirrors the peripheral self. Nat Immunol. 2001;2(11):1032–9.PubMedCrossRef Derbinski J, et al. Promiscuous gene expression in medullary thymic epithelial cells mirrors the peripheral self. Nat Immunol. 2001;2(11):1032–9.PubMedCrossRef
63.
go back to reference Aleksic M, et al. Different affinity windows for virus and cancer-specific T-cell receptors: Implications for therapeutic strategies. Eur J Immunol. 2012;42(12):3174–9.PubMedPubMedCentralCrossRef Aleksic M, et al. Different affinity windows for virus and cancer-specific T-cell receptors: Implications for therapeutic strategies. Eur J Immunol. 2012;42(12):3174–9.PubMedPubMedCentralCrossRef
64.
65.
go back to reference Öhlén C, et al. Expression of a tolerizing tumor antigen in peripheral tissue does not preclude recovery of high-affinity CD8<sup>+</sup> T cells or CTL immunotherapy of tumors expressing the antigen. J Immunol. 2001;166(4):2863–70.PubMedCrossRef Öhlén C, et al. Expression of a tolerizing tumor antigen in peripheral tissue does not preclude recovery of high-affinity CD8<sup>+</sup> T cells or CTL immunotherapy of tumors expressing the antigen. J Immunol. 2001;166(4):2863–70.PubMedCrossRef
66.
go back to reference Johnson LA, et al. Gene transfer of tumor-reactive TCR confers both high avidity and tumor reactivity to nonreactive peripheral blood mononuclear cells and tumor-infiltrating lymphocytes. J Immunol. 2006;177(9):6548–59.PubMedCrossRef Johnson LA, et al. Gene transfer of tumor-reactive TCR confers both high avidity and tumor reactivity to nonreactive peripheral blood mononuclear cells and tumor-infiltrating lymphocytes. J Immunol. 2006;177(9):6548–59.PubMedCrossRef
67.
go back to reference Davis JL, et al. Development of human anti-murine T-cell receptor antibodies in both responding and nonresponding patients enrolled in TCR gene therapy trials. Clin Cancer Res. 2010;16(23):5852–61.PubMedPubMedCentralCrossRef Davis JL, et al. Development of human anti-murine T-cell receptor antibodies in both responding and nonresponding patients enrolled in TCR gene therapy trials. Clin Cancer Res. 2010;16(23):5852–61.PubMedPubMedCentralCrossRef
68.
70.
go back to reference Li Y, et al. Directed evolution of human T-cell receptors with picomolar affinities by phage display. Nat Biotechnol. 2005;23(3):349–54.PubMedCrossRef Li Y, et al. Directed evolution of human T-cell receptors with picomolar affinities by phage display. Nat Biotechnol. 2005;23(3):349–54.PubMedCrossRef
72.
73.
go back to reference Bendle GM, et al. Lethal graft-versus-host disease in mouse models of T cell receptor gene therapy. Nat Med. 2010;16(5):565–70 1p following 570.PubMedCrossRef Bendle GM, et al. Lethal graft-versus-host disease in mouse models of T cell receptor gene therapy. Nat Med. 2010;16(5):565–70 1p following 570.PubMedCrossRef
74.
go back to reference Cohen CJ, et al. Enhanced antitumor activity of murine-human hybrid T-cell receptor (TCR) in human lymphocytes is associated with improved pairing and TCR/CD3 stability. Cancer Res. 2006;66(17):8878–86.PubMedPubMedCentralCrossRef Cohen CJ, et al. Enhanced antitumor activity of murine-human hybrid T-cell receptor (TCR) in human lymphocytes is associated with improved pairing and TCR/CD3 stability. Cancer Res. 2006;66(17):8878–86.PubMedPubMedCentralCrossRef
76.
go back to reference Voss R-H, et al. Molecular design of the Cαβ interface favors specific pairing of introduced TCRαβ in human T cells. J Immunol. 2008;180(1):391–401.PubMedCrossRef Voss R-H, et al. Molecular design of the Cαβ interface favors specific pairing of introduced TCRαβ in human T cells. J Immunol. 2008;180(1):391–401.PubMedCrossRef
78.
go back to reference Govers C, et al. TCRs genetically linked to CD28 and CD3ε do not mispair with endogenous TCR chains and mediate enhanced T cell persistence and anti-melanoma activity. J Immunol. 2014;193(10):5315–26.PubMedCrossRef Govers C, et al. TCRs genetically linked to CD28 and CD3ε do not mispair with endogenous TCR chains and mediate enhanced T cell persistence and anti-melanoma activity. J Immunol. 2014;193(10):5315–26.PubMedCrossRef
79.
go back to reference Okamoto S, et al. Improved expression and reactivity of transduced tumor-specific TCRs in human lymphocytes by specific silencing of endogenous TCR. Cancer Res. 2009;69(23):9003–11.PubMedCrossRef Okamoto S, et al. Improved expression and reactivity of transduced tumor-specific TCRs in human lymphocytes by specific silencing of endogenous TCR. Cancer Res. 2009;69(23):9003–11.PubMedCrossRef
80.
go back to reference Ochi T, et al. Novel adoptive T-cell immunotherapy using a WT1-specific TCR vector encoding silencers for endogenous TCRs shows marked antileukemia reactivity and safety. Blood. 2011;118(6):1495–503.PubMedCrossRef Ochi T, et al. Novel adoptive T-cell immunotherapy using a WT1-specific TCR vector encoding silencers for endogenous TCRs shows marked antileukemia reactivity and safety. Blood. 2011;118(6):1495–503.PubMedCrossRef
82.
go back to reference Torikai H, et al. A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric-antigen-receptor and eliminate expression of endogenous TCR. Blood. 2012;119(24):5697–705.PubMedPubMedCentralCrossRef Torikai H, et al. A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric-antigen-receptor and eliminate expression of endogenous TCR. Blood. 2012;119(24):5697–705.PubMedPubMedCentralCrossRef
83.
go back to reference Berdien B, et al. TALEN-mediated editing of endogenous T-cell receptors facilitates efficient reprogramming of T lymphocytes by lentiviral gene transfer. Gene Ther. 2014;21:539.PubMedCrossRef Berdien B, et al. TALEN-mediated editing of endogenous T-cell receptors facilitates efficient reprogramming of T lymphocytes by lentiviral gene transfer. Gene Ther. 2014;21:539.PubMedCrossRef
85.
go back to reference Yang L, et al. Generation of functional antigen-specific T cells in defined genetic backgrounds by retrovirus-mediated expression of TCR cDNAs in hematopoietic precursor cells. Proc Natl Acad Sci U S A. 2002;99(9):6204–9.PubMedPubMedCentralCrossRef Yang L, et al. Generation of functional antigen-specific T cells in defined genetic backgrounds by retrovirus-mediated expression of TCR cDNAs in hematopoietic precursor cells. Proc Natl Acad Sci U S A. 2002;99(9):6204–9.PubMedPubMedCentralCrossRef
86.
go back to reference van der Veken LT, et al. αβ T-cell receptor engineered γδ T cells mediate effective antileukemic reactivity. Cancer Res. 2006;66(6):3331–7.PubMedCrossRef van der Veken LT, et al. αβ T-cell receptor engineered γδ T cells mediate effective antileukemic reactivity. Cancer Res. 2006;66(6):3331–7.PubMedCrossRef
87.
go back to reference Rosenberg SA. Of mice, not men: no evidence for graft-versus-host disease in humans receiving T-cell receptor–transduced autologous T cells. Mol Ther. 2010;18(10):1744–5.PubMedPubMedCentralCrossRef Rosenberg SA. Of mice, not men: no evidence for graft-versus-host disease in humans receiving T-cell receptor–transduced autologous T cells. Mol Ther. 2010;18(10):1744–5.PubMedPubMedCentralCrossRef
88.
go back to reference Bunse M, et al. RNAi-mediated TCR knockdown prevents autoimmunity in mice caused by mixed TCR dimers following TCR gene transfer. Mol Ther. 2014;22(11):1983–91.PubMedPubMedCentralCrossRef Bunse M, et al. RNAi-mediated TCR knockdown prevents autoimmunity in mice caused by mixed TCR dimers following TCR gene transfer. Mol Ther. 2014;22(11):1983–91.PubMedPubMedCentralCrossRef
89.
90.
go back to reference Nayersina R, et al. HLA A2 restricted cytotoxic T lymphocyte responses to multiple hepatitis B surface antigen epitopes during hepatitis B virus infection. J Immunol. 1993;150(10):4659–71.PubMed Nayersina R, et al. HLA A2 restricted cytotoxic T lymphocyte responses to multiple hepatitis B surface antigen epitopes during hepatitis B virus infection. J Immunol. 1993;150(10):4659–71.PubMed
91.
go back to reference Mizukoshi E, et al. Cellular immune responses to the hepatitis B virus polymerase. J Immunol. 2004;173(9):5863–71.PubMedCrossRef Mizukoshi E, et al. Cellular immune responses to the hepatitis B virus polymerase. J Immunol. 2004;173(9):5863–71.PubMedCrossRef
92.
go back to reference Ressing ME, et al. Differential binding of viral peptides to HLA-A2 alleles. Implications for human papillomavirus type 16 E7 peptide-based vaccination against cervical carcinoma. Eur J Immunol. 1999;29(4):1292–303.PubMedCrossRef Ressing ME, et al. Differential binding of viral peptides to HLA-A2 alleles. Implications for human papillomavirus type 16 E7 peptide-based vaccination against cervical carcinoma. Eur J Immunol. 1999;29(4):1292–303.PubMedCrossRef
93.
go back to reference Robbins PF, et al. A mutated beta-catenin gene encodes a melanoma-specific antigen recognized by tumor infiltrating lymphocytes. J Exp Med. 1996;183(3):1185–92.PubMedCrossRef Robbins PF, et al. A mutated beta-catenin gene encodes a melanoma-specific antigen recognized by tumor infiltrating lymphocytes. J Exp Med. 1996;183(3):1185–92.PubMedCrossRef
94.
go back to reference Echchakir H, et al. A point mutation in the alpha-actinin-4 gene generates an antigenic peptide recognized by autologous cytolytic T lymphocytes on a human lung carcinoma. Cancer Res. 2001;61(10):4078–83.PubMed Echchakir H, et al. A point mutation in the alpha-actinin-4 gene generates an antigenic peptide recognized by autologous cytolytic T lymphocytes on a human lung carcinoma. Cancer Res. 2001;61(10):4078–83.PubMed
95.
go back to reference Gaudin C, et al. A hsp70-2 mutation recognized by CTL on a human renal cell carcinoma. J Immunol. 1999;162(3):1730–8.PubMed Gaudin C, et al. A hsp70-2 mutation recognized by CTL on a human renal cell carcinoma. J Immunol. 1999;162(3):1730–8.PubMed
96.
go back to reference De Backer O, et al. Characterization of the GAGE genes that are expressed in various human cancers and in normal testis. Cancer Res. 1999;59(13):3157–65.PubMed De Backer O, et al. Characterization of the GAGE genes that are expressed in various human cancers and in normal testis. Cancer Res. 1999;59(13):3157–65.PubMed
97.
go back to reference Chomez P, et al. An overview of the MAGE gene family with the identification of all human members of the family. Cancer Res. 2001;61(14):5544–51.PubMed Chomez P, et al. An overview of the MAGE gene family with the identification of all human members of the family. Cancer Res. 2001;61(14):5544–51.PubMed
98.
go back to reference Brichard V, et al. The tyrosinase gene codes for an antigen recognized by autologous cytolytic T lymphocytes on HLA-A2 melanomas. J Exp Med. 1993;178(2):489–95.PubMedCrossRef Brichard V, et al. The tyrosinase gene codes for an antigen recognized by autologous cytolytic T lymphocytes on HLA-A2 melanomas. J Exp Med. 1993;178(2):489–95.PubMedCrossRef
99.
go back to reference Duffy MJ. Carcinoembryonic antigen as a marker for colorectal cancer: is it clinically useful? Clin Chem. 2001;47(4):624–30.PubMed Duffy MJ. Carcinoembryonic antigen as a marker for colorectal cancer: is it clinically useful? Clin Chem. 2001;47(4):624–30.PubMed
102.
104.
go back to reference Therasse P, et al. New guidelines to evaluate the response to treatment in solid tumors. J Natl Cancer Inst. 2000;92(3):205–16.PubMedCrossRef Therasse P, et al. New guidelines to evaluate the response to treatment in solid tumors. J Natl Cancer Inst. 2000;92(3):205–16.PubMedCrossRef
105.
go back to reference Johnson LA, et al. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood. 2009;114(3):535–46.PubMedPubMedCentralCrossRef Johnson LA, et al. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood. 2009;114(3):535–46.PubMedPubMedCentralCrossRef
106.
go back to reference Chodon T, et al. Adoptive transfer of MART-1 T-cell receptor transgenic lymphocytes and dendritic cell vaccination in patients with metastatic melanoma. Clin Cancer Res. 2014;20(9):2457–65.PubMedPubMedCentralCrossRef Chodon T, et al. Adoptive transfer of MART-1 T-cell receptor transgenic lymphocytes and dendritic cell vaccination in patients with metastatic melanoma. Clin Cancer Res. 2014;20(9):2457–65.PubMedPubMedCentralCrossRef
107.
go back to reference Yang S, et al. Development of optimal bicistronic lentiviral vectors facilitates high-level TCR gene expression and robust tumor cell recognition. Gene Ther. 2008;15(21):1411–23.PubMedPubMedCentralCrossRef Yang S, et al. Development of optimal bicistronic lentiviral vectors facilitates high-level TCR gene expression and robust tumor cell recognition. Gene Ther. 2008;15(21):1411–23.PubMedPubMedCentralCrossRef
108.
go back to reference Robbins PF, et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J Clin Oncol. 2011;29(7):917–24.PubMedPubMedCentralCrossRef Robbins PF, et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J Clin Oncol. 2011;29(7):917–24.PubMedPubMedCentralCrossRef
109.
go back to reference Chen YT, et al. A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening. Proc Natl Acad Sci U S A. 1997;94(5):1914–8.PubMedPubMedCentralCrossRef Chen YT, et al. A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening. Proc Natl Acad Sci U S A. 1997;94(5):1914–8.PubMedPubMedCentralCrossRef
110.
go back to reference van Baren N, et al. Genes encoding tumor-specific antigens are expressed in human myeloma cells. Blood. 1999;94(4):1156–64.PubMed van Baren N, et al. Genes encoding tumor-specific antigens are expressed in human myeloma cells. Blood. 1999;94(4):1156–64.PubMed
111.
go back to reference van Rhee F, et al. NY-ESO-1 is highly expressed in poor-prognosis multiple myeloma and induces spontaneous humoral and cellular immune responses. Blood. 2005;105(10):3939–44.PubMedPubMedCentralCrossRef van Rhee F, et al. NY-ESO-1 is highly expressed in poor-prognosis multiple myeloma and induces spontaneous humoral and cellular immune responses. Blood. 2005;105(10):3939–44.PubMedPubMedCentralCrossRef
112.
go back to reference Jungbluth AA, et al. Monophasic and biphasic synovial sarcomas abundantly express cancer/testis antigen NY-ESO-1 but not MAGE-A1 or CT7. Int J Cancer. 2001;94(2):252–6.PubMedCrossRef Jungbluth AA, et al. Monophasic and biphasic synovial sarcomas abundantly express cancer/testis antigen NY-ESO-1 but not MAGE-A1 or CT7. Int J Cancer. 2001;94(2):252–6.PubMedCrossRef
113.
go back to reference Parkhurst MR, et al. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol Ther. 2011;19(3):620–6.PubMedCrossRef Parkhurst MR, et al. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol Ther. 2011;19(3):620–6.PubMedCrossRef
114.
115.
go back to reference Kageyama S, et al. Adoptive transfer of MAGE-A4 T-cell receptor gene-transduced lymphocytes in patients with recurrent esophageal cancer. Clin Cancer Res. 2015;21(10):2268–77.PubMedCrossRef Kageyama S, et al. Adoptive transfer of MAGE-A4 T-cell receptor gene-transduced lymphocytes in patients with recurrent esophageal cancer. Clin Cancer Res. 2015;21(10):2268–77.PubMedCrossRef
116.
go back to reference Robbins PF, et al. A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response. Clin Cancer Res. 2015;21(5):1019–27.PubMedCrossRef Robbins PF, et al. A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response. Clin Cancer Res. 2015;21(5):1019–27.PubMedCrossRef
117.
go back to reference Rapoport AP, et al. NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat Med. 2015;21(8):914–21.PubMedPubMedCentralCrossRef Rapoport AP, et al. NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat Med. 2015;21(8):914–21.PubMedPubMedCentralCrossRef
118.
go back to reference Tawara I, et al. Safety and persistence of WT1-specific T-cell receptor gene−transduced lymphocytes in patients with AML and MDS. Blood. 2017;130(18):1985–94.PubMedCrossRef Tawara I, et al. Safety and persistence of WT1-specific T-cell receptor gene−transduced lymphocytes in patients with AML and MDS. Blood. 2017;130(18):1985–94.PubMedCrossRef
119.
go back to reference Linette GP, et al. Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma. Blood. 2013;122(6):863–71.PubMedPubMedCentralCrossRef Linette GP, et al. Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma. Blood. 2013;122(6):863–71.PubMedPubMedCentralCrossRef
120.
go back to reference Cameron BJ, et al. Identification of a titin-derived HLA-A1-presented peptide as a cross-reactive target for engineered MAGE A3-directed T cells. Sci Transl Med. 2013;5(197):197ra103.PubMedPubMedCentralCrossRef Cameron BJ, et al. Identification of a titin-derived HLA-A1-presented peptide as a cross-reactive target for engineered MAGE A3-directed T cells. Sci Transl Med. 2013;5(197):197ra103.PubMedPubMedCentralCrossRef
121.
122.
go back to reference Boulter JM, et al. Stable, soluble T-cell receptor molecules for crystallization and therapeutics. Protein Eng. 2003;16(9):707–11.PubMedCrossRef Boulter JM, et al. Stable, soluble T-cell receptor molecules for crystallization and therapeutics. Protein Eng. 2003;16(9):707–11.PubMedCrossRef
123.
go back to reference Withoff S, et al. Bi-specific antibody therapy for the treatment of cancer. Curr Opin Mol Ther. 2001;3(1):53–62.PubMed Withoff S, et al. Bi-specific antibody therapy for the treatment of cancer. Curr Opin Mol Ther. 2001;3(1):53–62.PubMed
124.
go back to reference McCormack E, et al. Bi-specific TCR-anti CD3 redirected T-cell targeting of NY-ESO-1- and LAGE-1-positive tumors. Cancer Immunol Immunother. 2013;62(4):773–85.PubMedCrossRef McCormack E, et al. Bi-specific TCR-anti CD3 redirected T-cell targeting of NY-ESO-1- and LAGE-1-positive tumors. Cancer Immunol Immunother. 2013;62(4):773–85.PubMedCrossRef
125.
go back to reference Bossi G, et al. ImmTAC-redirected tumour cell killing induces and potentiates antigen cross-presentation by dendritic cells. Cancer Immunol Immunother. 2014;63(5):437–48.PubMedCrossRef Bossi G, et al. ImmTAC-redirected tumour cell killing induces and potentiates antigen cross-presentation by dendritic cells. Cancer Immunol Immunother. 2014;63(5):437–48.PubMedCrossRef
126.
127.
go back to reference Card KF, et al. A soluble single-chain T-cell receptor IL-2 fusion protein retains MHC-restricted peptide specificity and IL-2 bioactivity. Cancer Immunol Immunother. 2004;53(4):345–57.PubMedCrossRef Card KF, et al. A soluble single-chain T-cell receptor IL-2 fusion protein retains MHC-restricted peptide specificity and IL-2 bioactivity. Cancer Immunol Immunother. 2004;53(4):345–57.PubMedCrossRef
128.
go back to reference Belmont HJ, et al. Potent antitumor activity of a tumor-specific soluble TCR/IL-2 fusion protein. Clin Immunol. 2006;121(1):29–39.PubMedCrossRef Belmont HJ, et al. Potent antitumor activity of a tumor-specific soluble TCR/IL-2 fusion protein. Clin Immunol. 2006;121(1):29–39.PubMedCrossRef
129.
go back to reference Wen J, et al. Targeting activity of a TCR/IL-2 fusion protein against established tumors. Cancer Immunol Immunother. 2008;57(12):1781–94.PubMedCrossRef Wen J, et al. Targeting activity of a TCR/IL-2 fusion protein against established tumors. Cancer Immunol Immunother. 2008;57(12):1781–94.PubMedCrossRef
130.
go back to reference Fishman MN, et al. Phase I trial of ALT-801, an interleukin-2/T-cell receptor fusion protein targeting p53 (aa264–272)/HLA-A*0201 complex, in patients with advanced malignancies. Clin Cancer Res. 2011;17(24):7765–75.PubMedPubMedCentralCrossRef Fishman MN, et al. Phase I trial of ALT-801, an interleukin-2/T-cell receptor fusion protein targeting p53 (aa264–272)/HLA-A*0201 complex, in patients with advanced malignancies. Clin Cancer Res. 2011;17(24):7765–75.PubMedPubMedCentralCrossRef
131.
go back to reference Mosquera LA, et al. In vitro and in vivo characterization of a novel antibody-like single-chain TCR human IgG1 fusion protein. J Immunol. 2005;174(7):4381–8.PubMedCrossRef Mosquera LA, et al. In vitro and in vivo characterization of a novel antibody-like single-chain TCR human IgG1 fusion protein. J Immunol. 2005;174(7):4381–8.PubMedCrossRef
132.
go back to reference Weidle UH, Georges G, Tiefenthaler G. TCR-MHC/peptide interaction: prospects for new anti-tumoral agents. Cancer Genomics Proteomics. 2014;11(6):267–77.PubMed Weidle UH, Georges G, Tiefenthaler G. TCR-MHC/peptide interaction: prospects for new anti-tumoral agents. Cancer Genomics Proteomics. 2014;11(6):267–77.PubMed
133.
go back to reference Zhu X, et al. Visualization of p53<sub>264–272</sub>/HLA-A*0201 complexes naturally presented on tumor cell surface by a multimeric soluble single-chain T cell receptor. J Immunol. 2006;176(5):3223–32.PubMedCrossRef Zhu X, et al. Visualization of p53<sub>264–272</sub>/HLA-A*0201 complexes naturally presented on tumor cell surface by a multimeric soluble single-chain T cell receptor. J Immunol. 2006;176(5):3223–32.PubMedCrossRef
134.
go back to reference Sykulev Y, et al. Evidence that a single peptide–MHC complex on a target cell can elicit a cytolytic T cell response. Immunity. 1996;4(6):565–71.PubMedCrossRef Sykulev Y, et al. Evidence that a single peptide–MHC complex on a target cell can elicit a cytolytic T cell response. Immunity. 1996;4(6):565–71.PubMedCrossRef
135.
go back to reference Huang J, et al. A single peptide-major histocompatibility complex ligand triggers digital cytokine secretion in CD4+ T cells. Immunity. 2013;39(5):846–57.PubMedCrossRef Huang J, et al. A single peptide-major histocompatibility complex ligand triggers digital cytokine secretion in CD4+ T cells. Immunity. 2013;39(5):846–57.PubMedCrossRef
136.
go back to reference Irvine DJ, et al. Direct observation of ligand recognition by T cells. Nature. 2002;419(6909):845–9.PubMedCrossRef Irvine DJ, et al. Direct observation of ligand recognition by T cells. Nature. 2002;419(6909):845–9.PubMedCrossRef
137.
go back to reference Weber S, et al. Specific low-affinity recognition of major histocompatibility complex plus peptide by soluble T-cell receptor. Nature. 1992;356(6372):793–6.PubMedCrossRef Weber S, et al. Specific low-affinity recognition of major histocompatibility complex plus peptide by soluble T-cell receptor. Nature. 1992;356(6372):793–6.PubMedCrossRef
138.
go back to reference Watanabe K, et al. Target antigen density governs the efficacy of anti–CD20-CD28-CD3 ζ chimeric antigen receptor–modified effector CD8<sup>+</sup> T cells. J Immunol. 2015;194(3):911–20.PubMedCrossRef Watanabe K, et al. Target antigen density governs the efficacy of anti–CD20-CD28-CD3 ζ chimeric antigen receptor–modified effector CD8<sup>+</sup> T cells. J Immunol. 2015;194(3):911–20.PubMedCrossRef
139.
go back to reference Stone JD, et al. A sensitivity scale for targeting T cells with chimeric antigen receptors (CARs) and bispecific T-cell Engagers (BiTEs). OncoImmunology. 2012;1(6):863–73.PubMedPubMedCentralCrossRef Stone JD, et al. A sensitivity scale for targeting T cells with chimeric antigen receptors (CARs) and bispecific T-cell Engagers (BiTEs). OncoImmunology. 2012;1(6):863–73.PubMedPubMedCentralCrossRef
140.
go back to reference Morgan RA, et al. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. 2010;18(4):843–51.PubMedPubMedCentralCrossRef Morgan RA, et al. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. 2010;18(4):843–51.PubMedPubMedCentralCrossRef
141.
go back to reference Kawakami Y, et al. Cloning of the gene coding for a shared human melanoma antigen recognized by autologous T cells infiltrating into tumor. Proc Natl Acad Sci. 1994;91(9):3515–9.PubMedCrossRefPubMedCentral Kawakami Y, et al. Cloning of the gene coding for a shared human melanoma antigen recognized by autologous T cells infiltrating into tumor. Proc Natl Acad Sci. 1994;91(9):3515–9.PubMedCrossRefPubMedCentral
142.
go back to reference Kawakami Y, et al. Identification of a human melanoma antigen recognized by tumor-infiltrating lymphocytes associated with in vivo tumor rejection. Proc Natl Acad Sci. 1994;91(14):6458–62.PubMedCrossRefPubMedCentral Kawakami Y, et al. Identification of a human melanoma antigen recognized by tumor-infiltrating lymphocytes associated with in vivo tumor rejection. Proc Natl Acad Sci. 1994;91(14):6458–62.PubMedCrossRefPubMedCentral
143.
go back to reference Uslu U, et al. Combining a chimeric antigen receptor and a conventional T-cell receptor to generate T cells expressing two additional receptors (TETARs) for a multi-hit immunotherapy of melanoma. Exp Dermatol. 2016;25(11):872–9.PubMedCrossRef Uslu U, et al. Combining a chimeric antigen receptor and a conventional T-cell receptor to generate T cells expressing two additional receptors (TETARs) for a multi-hit immunotherapy of melanoma. Exp Dermatol. 2016;25(11):872–9.PubMedCrossRef
144.
go back to reference Miller AM, et al. Leveraging TCR affinity in adoptive immunotherapy against shared tumor/self-antigens. Cancer Immun Res. 2019;7(1):40–9.CrossRef Miller AM, et al. Leveraging TCR affinity in adoptive immunotherapy against shared tumor/self-antigens. Cancer Immun Res. 2019;7(1):40–9.CrossRef
145.
go back to reference Solal-Céligny P. Safety of rituximab maintenance therapy in follicular lymphomas. Leuk Res. 2006;30:S16–21.PubMedCrossRef Solal-Céligny P. Safety of rituximab maintenance therapy in follicular lymphomas. Leuk Res. 2006;30:S16–21.PubMedCrossRef
147.
go back to reference van der Bruggen P, et al. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science. 1991;254(5038):1643–7.PubMedCrossRef van der Bruggen P, et al. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science. 1991;254(5038):1643–7.PubMedCrossRef
148.
go back to reference Groeper C, et al. Cancer/testis antigen expression and specific cytotoxic T lymphocyte responses in non small cell lung cancer. Int J Cancer. 2007;120(2):337–43.PubMedCrossRef Groeper C, et al. Cancer/testis antigen expression and specific cytotoxic T lymphocyte responses in non small cell lung cancer. Int J Cancer. 2007;120(2):337–43.PubMedCrossRef
149.
go back to reference Brichard VG, Lejeune D. Cancer immunotherapy targeting tumour-specific antigens: towards a new therapy for minimal residual disease. Expert Opin Biol Ther. 2008;8(7):951–68.PubMedCrossRef Brichard VG, Lejeune D. Cancer immunotherapy targeting tumour-specific antigens: towards a new therapy for minimal residual disease. Expert Opin Biol Ther. 2008;8(7):951–68.PubMedCrossRef
151.
go back to reference Monte M, et al. MAGE-A tumor antigens target p53 transactivation function through histone deacetylase recruitment and confer resistance to chemotherapeutic agents. Proc Natl Acad Sci U S A. 2006;103(30):11160–5.PubMedPubMedCentralCrossRef Monte M, et al. MAGE-A tumor antigens target p53 transactivation function through histone deacetylase recruitment and confer resistance to chemotherapeutic agents. Proc Natl Acad Sci U S A. 2006;103(30):11160–5.PubMedPubMedCentralCrossRef
152.
go back to reference Kim J, et al. The clinical significance of MAGEA3 expression in pancreatic cancer. Int J Cancer. 2006;118(9):2269–75.PubMedCrossRef Kim J, et al. The clinical significance of MAGEA3 expression in pancreatic cancer. Int J Cancer. 2006;118(9):2269–75.PubMedCrossRef
153.
go back to reference Suyama T, et al. Expression of cancer/testis antigens in prostate cancer is associated with disease progression. Prostate. 2010;70(16):1778–87.PubMedPubMedCentral Suyama T, et al. Expression of cancer/testis antigens in prostate cancer is associated with disease progression. Prostate. 2010;70(16):1778–87.PubMedPubMedCentral
155.
go back to reference Bos R, et al. Balancing between antitumor efficacy and autoimmune pathology in T-cell–mediated targeting of carcinoembryonic antigen. Cancer Res. 2008;68(20):8446–55.PubMedCrossRef Bos R, et al. Balancing between antitumor efficacy and autoimmune pathology in T-cell–mediated targeting of carcinoembryonic antigen. Cancer Res. 2008;68(20):8446–55.PubMedCrossRef
156.
go back to reference Palmer DC, et al. Effective tumor treatment targeting a melanoma/melanocyte-associated antigen triggers severe ocular autoimmunity. Proc Natl Acad Sci. 2008;105(23):8061–6.PubMedCrossRefPubMedCentral Palmer DC, et al. Effective tumor treatment targeting a melanoma/melanocyte-associated antigen triggers severe ocular autoimmunity. Proc Natl Acad Sci. 2008;105(23):8061–6.PubMedCrossRefPubMedCentral
158.
159.
go back to reference Li J, et al. Chimeric antigen receptor T cell (CAR-T) immunotherapy for solid tumors: lessons learned and strategies for moving forward. J Hematol Oncol. 2018;11(1):22.PubMedPubMedCentralCrossRef Li J, et al. Chimeric antigen receptor T cell (CAR-T) immunotherapy for solid tumors: lessons learned and strategies for moving forward. J Hematol Oncol. 2018;11(1):22.PubMedPubMedCentralCrossRef
163.
164.
go back to reference Vatakis DN, et al. Introduction of exogenous T-cell receptors into human hematopoietic progenitors results in exclusion of endogenous T-cell receptor expression. Mol Ther. 2013;21(5):1055–63.PubMedPubMedCentralCrossRef Vatakis DN, et al. Introduction of exogenous T-cell receptors into human hematopoietic progenitors results in exclusion of endogenous T-cell receptor expression. Mol Ther. 2013;21(5):1055–63.PubMedPubMedCentralCrossRef
165.
go back to reference Lei F, et al. <em>In vivo</em> programming of tumor antigen-specific T lymphocytes from pluripotent stem cells to promote cancer immunosurveillance. Cancer Res. 2011;71(14):4742–7.PubMedCrossRef Lei F, et al. <em>In vivo</em> programming of tumor antigen-specific T lymphocytes from pluripotent stem cells to promote cancer immunosurveillance. Cancer Res. 2011;71(14):4742–7.PubMedCrossRef
166.
go back to reference Zeh HJ, et al. High avidity CTLs for two self-antigens demonstrate superior in vitro and in vivo antitumor efficacy. J Immunol. 1999;162(2):989–94.PubMed Zeh HJ, et al. High avidity CTLs for two self-antigens demonstrate superior in vitro and in vivo antitumor efficacy. J Immunol. 1999;162(2):989–94.PubMed
167.
go back to reference Derby M, et al. High-avidity CTL exploit two complementary mechanisms to provide better protection against viral infection than low-avidity CTL. J Immunol. 2001;166(3):1690–7.PubMedCrossRef Derby M, et al. High-avidity CTL exploit two complementary mechanisms to provide better protection against viral infection than low-avidity CTL. J Immunol. 2001;166(3):1690–7.PubMedCrossRef
169.
go back to reference Presotto D, et al. Fine-tuning of optimal TCR signaling in tumor-redirected CD8 T cells by distinct TCR affinity-mediated mechanisms. Front Immunol. 2017;8:1564.PubMedPubMedCentralCrossRef Presotto D, et al. Fine-tuning of optimal TCR signaling in tumor-redirected CD8 T cells by distinct TCR affinity-mediated mechanisms. Front Immunol. 2017;8:1564.PubMedPubMedCentralCrossRef
170.
go back to reference Zhong S, et al. T-cell receptor affinity and avidity defines antitumor response and autoimmunity in T-cell immunotherapy. Proc Natl Acad Sci. 2013;110(17):6973–8.PubMedCrossRefPubMedCentral Zhong S, et al. T-cell receptor affinity and avidity defines antitumor response and autoimmunity in T-cell immunotherapy. Proc Natl Acad Sci. 2013;110(17):6973–8.PubMedCrossRefPubMedCentral
171.
172.
go back to reference Schmid DA, et al. Evidence for a TCR affinity threshold delimiting maximal CD8 T cell function. J Immunol. 2010;184(9):4936–46.PubMedCrossRef Schmid DA, et al. Evidence for a TCR affinity threshold delimiting maximal CD8 T cell function. J Immunol. 2010;184(9):4936–46.PubMedCrossRef
175.
go back to reference Wong SBJ, Bos R, Sherman LA. Tumor-specific CD4<sup>+</sup> T cells render the tumor environment permissive for infiltration by low-avidity CD8<sup>+</sup> T cells. J Immunol. 2008;180(5):3122–31.PubMedCrossRef Wong SBJ, Bos R, Sherman LA. Tumor-specific CD4<sup>+</sup> T cells render the tumor environment permissive for infiltration by low-avidity CD8<sup>+</sup> T cells. J Immunol. 2008;180(5):3122–31.PubMedCrossRef
176.
go back to reference Morgan DJ, et al. Activation of low avidity CTL specific for a self epitope results in tumor rejection but not autoimmunity. J Immunol. 1998;160(2):643–51.PubMed Morgan DJ, et al. Activation of low avidity CTL specific for a self epitope results in tumor rejection but not autoimmunity. J Immunol. 1998;160(2):643–51.PubMed
178.
go back to reference Garrido F, Cabrera T, Aptsiauri N. “Hard” and “soft” lesions underlying the HLA class I alterations in cancer cells: Implications for immunotherapy. Int J Cancer. 2010;127(2):249–56.PubMed Garrido F, Cabrera T, Aptsiauri N. “Hard” and “soft” lesions underlying the HLA class I alterations in cancer cells: Implications for immunotherapy. Int J Cancer. 2010;127(2):249–56.PubMed
179.
go back to reference Liu B, Song Y, Liu D. Recent development in clinical applications of PD-1 and PD-L1 antibodies for cancer immunotherapy. J Hematol Oncol. 2017;10(1):174.PubMedPubMedCentralCrossRef Liu B, Song Y, Liu D. Recent development in clinical applications of PD-1 and PD-L1 antibodies for cancer immunotherapy. J Hematol Oncol. 2017;10(1):174.PubMedPubMedCentralCrossRef
Metadata
Title
Targeting cancers through TCR-peptide/MHC interactions
Authors
Qinghua He
Xianhan Jiang
Xinke Zhou
Jinsheng Weng
Publication date
01-12-2019
Publisher
BioMed Central
Keywords
Melanoma
Melanoma
Published in
Journal of Hematology & Oncology / Issue 1/2019
Electronic ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-019-0812-8

Other articles of this Issue 1/2019

Journal of Hematology & Oncology 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine