Data availability
DNA sequencing data of TCR discovery screens have been deposited in the National Center for Biotechnology Informationʼs Sequence Read Archive under accession codes PRJNA1068078 (ref. 41), PRJNA1068299 (ref. 42), PRJNA1068301 (ref. 43) and PRJNA1068303 (ref. 44).
References
Ribas, A. & Wolchok, J. D. Cancer immunotherapy using checkpoint blockade. Science359, 1350–1355 (2018).
Article
CAS
PubMed
PubMed Central
Google Scholar
Rohaan, M. W. et al. Tumor-infiltrating lymphocyte therapy or ipilimumab in advanced melanoma. N. Engl. J. Med.387, 2113–2125 (2022).
Article
CAS
PubMed
Google Scholar
Rosenberg, S. A. & Restifo, N. P. Adoptive cell transfer as personalized immunotherapy for human cancer. Science348, 62–68 (2015).
Article
CAS
PubMed
PubMed Central
Google Scholar
Klebanoff, C. A., Chandran, S. S., Baker, B. M., Quezada, S. A. & Ribas, A. T cell receptor therapeutics: immunological targeting of the intracellular cancer proteome. Nat. Rev. Drug Discov.22, 996–1017 (2023).
Article
CAS
PubMed
Google Scholar
Simoni, Y. et al. Bystander CD8+ T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature557, 575–579 (2018).
Article
CAS
PubMed
Google Scholar
Scheper, W. et al. Low and variable tumor reactivity of the intratumoral TCR repertoire in human cancers. Nat. Med.25, 89–94 (2019).
Article
CAS
PubMed
Google Scholar
Schumacher, T. N., Scheper, W. & Kvistborg, P. Cancer neoantigens. Annu. Rev. Immunol.37, 173–200 (2019).
Article
CAS
PubMed
Google Scholar
Schumacher, T. N. & Schreiber, R. D. Neoantigens in cancer immunotherapy. Science348, 69–74 (2015).
Article
CAS
PubMed
Google Scholar
Gros, A. et al. Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat. Med.22, 433–438 (2016).
Article
CAS
PubMed
PubMed Central
Google Scholar
Gubin, M. M. et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature515, 577–581 (2014).
Article
CAS
PubMed
PubMed Central
Google Scholar
Rizvi, N. A. et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science348, 124–128 (2015).
Article
CAS
PubMed
PubMed Central
Google Scholar
Alspach, E. et al. MHC-II neoantigens shape tumour immunity and response to immunotherapy. Nature574, 696–701 (2019).
Article
CAS
PubMed
PubMed Central
Google Scholar
Oh, D. Y. et al. Intratumoral CD4+ T cells mediate anti-tumor cytotoxicity in human bladder cancer. Cell181, 1612–1625 (2020).
Article
CAS
PubMed
PubMed Central
Google Scholar
Tran, E. et al. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science344, 641–645 (2014).
Article
CAS
PubMed
PubMed Central
Google Scholar
Borst, J., Ahrends, T., Babala, N., Melief, C. J. M. & Kastenmuller, W. CD4+ T cell help in cancer immunology and immunotherapy. Nat. Rev. Immunol.18, 635–647 (2018).
Article
CAS
PubMed
Google Scholar
Hu, Z. et al. A cloning and expression system to probe T-cell receptor specificity and assess functional avidity to neoantigens. Blood132, 1911–1921 (2018).
Article
CAS
PubMed
PubMed Central
Google Scholar
Guo, X. Z. et al. Rapid cloning, expression, and functional characterization of paired αβ and γδ T-cell receptor chains from single-cell analysis. Mol. Ther. Methods Clin. Dev.3, 15054 (2016).
Article
PubMed
PubMed Central
Google Scholar
Zong, S. et al. Very rapid cloning, expression and identifying specificity of T-cell receptors for T-cell engineering. PLoS ONE15, e0228112 (2020).
Article
CAS
PubMed
PubMed Central
Google Scholar
Genolet, R. et al. TCR sequencing and cloning methods for repertoire analysis and isolation of tumor-reactive TCRs. Cell Rep. Methods3, 100459 (2023).
Article
CAS
PubMed
PubMed Central
Google Scholar
Fahad, A. S. et al. Immortalization and functional screening of natively paired human T cell receptor repertoires. Protein Eng. Des. Sel.35, gzab034 (2022).
Article
PubMed
PubMed Central
Google Scholar
Spindler, M. J. et al. Massively parallel interrogation and mining of natively paired human TCRαβ repertoires. Nat. Biotechnol.38, 609–619 (2020).
Article
CAS
PubMed
PubMed Central
Google Scholar
Muller, T. R. et al. A T-cell reporter platform for high-throughput and reliable investigation of TCR function and biology. Clin. Transl. Immunol.9, e1216 (2020).
Article
Google Scholar
Fahad, A. S. et al. Cell activation-based screening of natively paired human T cell receptor repertoires. Sci. Rep.13, 8011 (2023).
Article
CAS
PubMed
PubMed Central
Google Scholar
Vazquez-Lombardi, R. et al. High-throughput T cell receptor engineering by functional screening identifies candidates with enhanced potency and specificity. Immunity55, 1953–1966 (2022).
Article
CAS
PubMed
Google Scholar
Cattaneo, C. M. et al. Identification of patient-specific CD4+ and CD8+ T cell neoantigens through HLA-unbiased genetic screens. Nat. Biotechnol.41, 783–787 (2023).
Article
CAS
PubMed
PubMed Central
Google Scholar
Lowery, F. J. et al. Molecular signatures of antitumor neoantigen-reactive T cells from metastatic human cancers. Science375, 877–884 (2022).
Article
CAS
PubMed
PubMed Central
Google Scholar
Gros, A. et al. PD-1 identifies the patient-specific CD8+ tumor-reactive repertoire infiltrating human tumors. J. Clin. Invest.124, 2246–2259 (2014).
Article
CAS
PubMed
PubMed Central
Google Scholar
Arnaud, M. et al. Sensitive identification of neoantigens and cognate TCRs in human solid tumors. Nat. Biotechnol.40, 656–660 (2022).
Article
CAS
PubMed
Google Scholar
Foy, S. P. et al. Non-viral precision T cell receptor replacement for personalized cell therapy. Nature615, 687–696 (2022).
Hilf, N. et al. Actively personalized vaccination trial for newly diagnosed glioblastoma. Nature565, 240–245 (2019).
Article
CAS
PubMed
Google Scholar
Ott, P. A. et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature547, 217–221 (2017).
Article
CAS
PubMed
PubMed Central
Google Scholar
Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol.36, 411–420 (2018).
Article
CAS
PubMed
PubMed Central
Google Scholar
Lefranc, M. P. et al. IMGT®, the international ImMunoGeneTics information system® 25 years on. Nucleic Acids Res.43, D413–D422 (2015).
Article
CAS
PubMed
Google Scholar
Chen, X. & Porter, E. Compositions and methods for T-cell receptor gene assembly. Patent WO2020206238A2 (2020).
Klein, J. C. et al. Multiplex pairwise assembly of array-derived DNA oligonucleotides. Nucleic Acids Res.44, e43 (2016).
Article
PubMed
Google Scholar
Chapuis, A. G. et al. T cell receptor gene therapy targeting WT1 prevents acute myeloid leukemia relapse post-transplant. Nat. Med.25, 1064–1072 (2019).
Article
CAS
PubMed
PubMed Central
Google Scholar
Schmitt, T. M., Greenberg, P. D. & Nguyen, H. N. T cell immunotherapy specific for WT-1. US patent US20160083449A1 (2015).
Borbulevych, O. Y., Santhanagopolan, S. M., Hossain, M. & Baker, B. M. TCRs used in cancer gene therapy cross-react with MART-1/Melan-A tumor antigens via distinct mechanisms. J. Immunol.187, 2453–2463 (2011).
Article
CAS
PubMed
Google Scholar
Johnson, L. A. 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.177, 6548–6559 (2006).
Article
CAS
PubMed
Google Scholar
Kwakkenbos, M. J. et al. Generation of stable monoclonal antibody–producing B cell receptor–positive human memory B cells by genetic programming. Nat. Med.16, 123–128 (2010).
Article
CAS
PubMed
Google Scholar
Moravec, Z. et al. Functional discovery of tumor-reactive T cell receptors by massively parallel library synthesis and screening: validation in CD8 T cells. NCBI Sequence Read Archive https://www.ncbi.nlm.nih.gov/bioproject/PRJNA1068078 (2024).
Moravec, Z. et al. Functional discovery of tumor-reactive T cell receptors by massively parallel library synthesis and screening: validation in CD4 T cells and OVC190 TCR screen. NCBI Sequence Read Archive https://www.ncbi.nlm.nih.gov/bioproject/PRJNA1068299 (2024).
Moravec, Z. et al. Functional discovery of tumor-reactive T cell receptors by massively parallel library synthesis and screening: NKIRTIL063 titration screen. NCBI Sequence Read Archive https://www.ncbi.nlm.nih.gov/bioproject/PRJNA1068301 (2024).
Moravec, Z. et al. Functional discovery of tumor-reactive T cell receptors by massively parallel library synthesis and screening: NKIRTIL063 neoantigen screen. NCBI Sequence Read Archive https://www.ncbi.nlm.nih.gov/bioproject/PRJNA1068303 (2024).
Download references
Acknowledgements
We would like to thank P. Kaptein and J. Urbanus for valuable help with developing the screening technology; S. Ketelaars for bioinformatic support; R. Tissier for support with statistical analyses; the Netherlands Cancer Institute–Antoni van Leeuwenhoek Hospital (NKI-AVL) Flow Cytometry Facility for flow cytometric support; the NKI-AVL Core Facility Molecular Pathology & Biobanking for supplying NKI-AVL Biobank material and laboratory support; and the NKI-AVL Genomics Core Facility for support with next-generation sequencing. This work was supported by the Dutch Cancer Society Young Investigator Grant (no. 2020-1/12977) (to W.S.); ZonMw Translational Research Program 2 (no. 446002001) (to W.S. and J.B.A.G.H.); the Stevin Prize and the Louis-Jeantet Prize (to T.N.S.); National Institutes of Health grants (no. R01CA269898 and no. R37CA273333-01) (to J.W.); the V Foundation (to J.W.); the Mark Foundation ASPIRE award (to J.W.); the Melanoma Research Alliance Young Investigator award (to J.W.); and a generous donation from Florry Vyth (to J.B.A.G.H.). Research at the Netherlands Cancer Institute is supported by institutional grants from the Dutch Cancer Society and the Dutch Ministry of Health, Welfare and Sport.
Author information
Author notes
These authors contributed equally: Yue Zhao, Rhianne Voogd.
Authors and Affiliations
Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
Ziva Moravec, Rhianne Voogd, Brenda Raud, John B.A.G. Haanen, Ton N. Schumacher & Wouter Scheper
RootPath, Inc. (Guangzhou), Guangzhou, China
Yue Zhao, Haiyan Yang, Jiayu Ou & Xi Chen
RootPath, Inc. (US), Watertown, MA, USA
Danielle R. Cook, Seon Kinrot, Benjamin Capra, Jiekun Xuan, Xi Chen & Ely Porter
RootPath, Inc. (Hangzhou), Hangzhou, China
Jiekun Xuan & Xi Chen
Cytotherapy Laboratory, People’s Hospital, Shenzhen, Guangdong, China
Teng Wei & Lili Ren
Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
Dandan Hu
State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
Dandan Hu
Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
Jun Wang
Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
Jun Wang
Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
John B.A.G. Haanen
Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
John B.A.G. Haanen & Ton N. Schumacher
Oncode Institute, Utrecht, The Netherlands
Ton N. Schumacher
Contributions
Z.M., R.V., D.R.C., Y.Z., B.R., B.C., H.Y., J.O., J.X., T.N.S., X.C., E.P. and W.S. designed, performed, analyzed and/or interpreted experiments. Z.M. and S.K. analyzed sequencing data. J.B.A.G.H. and D.H. supplied patient tumor material. T.W. and L.R. supplied patient tumor material and isolated and cultured patient TIL lines. J.W. helped conceptualize the methodology. E.P., X.C. and W.S. wrote the manuscript. All authors reviewed the manuscript.
Corresponding authors
Ethics declarations
Competing interests
D.R.C., Y.Z., B.C., S.K., H.Y., J.O., J.X. and E.P. are employees and stock option holders of RootPath, Inc. or its affiliates. X.C. is a director and shareholder of RootPath, Inc. or its affiliates. J.W. is a paid consultant for RootPath Genomics, Bristol Myers Squibb (Relatlimab Advisory Council) and Hanmi Pharmaceutical and is a founder and equity holder of and a consultant to Remunix, Inc. J.B.A.G.H. is an advisor to Achilles Therapeutics, BioNTech, Instil Bio, Neogene Therapeutics, PokeAcell, Scenic Biotech, T-Knife and Third Rock Ventures; is a recipient of research grant support from BioNTech; and is a stock option holder of Neogene Therapeutics. T.N.S. serves as an advisor for Allogene Therapeutics, Merus, Neogene Therapeutics and Scenic Biotech and is a stockholder in Allogene Therapeutics, Cell Control, Celsius, Merus and Scenic Biotech. T.N.S. is also a venture partner at Third Rock Ventures, all outside the submitted work. W.S. is an advisor to BD Biosciences and Lumicks. The TCR library synthesis method is described in patent application WO2020206238A2, assigned to a subsidiary of RootPath, Inc. (X.C. and E.P.). The other authors declare no competing interests.
Peer review
Peer review information
Nature Biotechnology thanks Michael Birnbaum and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Extended data
Extended Data Fig. 1 Pooled TCR library subcloning and accuracy.
(a, b) Schemes to produce ssDNA CDR3Jα pools (a) and ssDNA CDR3Jβ pools (b). Squares indicate 5′ phosphothioate modification. See Supplementary Table 29 for primer sequences. (c) Complete VCV pools can be PCR-amplified prior to subcloning using the common forward (CF) and reverse (CR) primers. Individual TCRs may be selectively amplified from VCV pools using the common forward primers (CF) and reverse primers targeting their respective Zip barcode sequences. (d) For the proof-of-concept TCR library (553 TCRs), the frequencies of individual CDR3Jα and CDR3Jβ sequences within the commercially synthesized CDR3Jα and CDR3Jβ ssDNA oligo pools and the assembled CDR3Jα-CDR3Jβ product were assessed by deep sequencing. The expected frequency of each CDR3Jα-CDR3Jβ pair was derived by multiplication of the observed frequencies of its respective CDR3Jα and CDR3Jβ sequences in the original, unassembled oligo pools. (e) Relation between Zip and CDR3Jα/CDR3Jβ oligo characteristics and the frequencies of resulting CDR3Jα-CDR3Jβ pairs after hybridization. (f) Relation between Zip and CDR3Jα/CDR3Jβ oligo characteristics and the accuracy of CDR3Jα-CDR3Jβ pairing (‘α-β pairing accuracy’).
Extended Data Fig. 2 ConnA and ConnB orthogonality for murine TRAV and TRBV genes and acquisition accuracies.
(a) Computer-predicted orthogonality of natural (left) and codon-diversified (right) FR3 connector sequences (ConnA) for murine TRAV. (b) Computer-predicted orthogonality of natural (left) and codon-diversified (right) FR3 connector sequences (ConnB) for murine TRBV. (c, d) Acquisition accuracies for TRAV (c) and TRBV (d) genes used in the proof-of-concept TCR library. The average acquisition accuracy for each TRAV/TRBV gene is shown above the bars.
Extended Data Fig. 3 Gating strategies.
(a) Gating strategy for the isolation of CD4+ and/or CD8+ T cells from patient tumor material, for the purpose of identifying patient-derived TCRs by single-cell TCR sequencing (relating to the patient TCR libraries used in Figs. 2 to 4). (b) Gating strategy for the isolation of activated reporter T cells in TCR library screens (relating to Figs. 2b–d, 3a,d and 4a, and Extended Data Figs. 7a–c, and 9b,d. (c) Gating strategy for T cell activation assays (relating to Figs. 3c,e,f and 4b,c, and Extended Data Figs. 7d–f and 9a. CD137 served as activation marker when using primary T cells, and CD69 served as activation marker when using Jurkat cells. (d) Gating strategy for T cell cytotoxicity assays (relating to Figs. 3g and 4d).
Extended Data Fig. 4 Quality control of the NKIRTIL063 TCR library.
(a) Schematic overview of the custom ‘2-reads-on-1-strand’ sequencing method. (b) Overall assembly accuracies and frequencies of TCRs in the fully assembled NKIRTIL063 VCV library (n=935 TCRs). Dots represent individual TCRs. Box plots depict the median, the interquartile range and whiskers extending to minimal and maximal values. (c) Pie chart depicting the overall fraction of NKIRTIL063 VCV molecules with and without sequence errors (taking both assembly accuracy and sequence mutations into account). (d) The fraction of NKIRTIL063 library TCRs successfully expressed at the cell surface of library-transduced cells was determined by isolation of mouse TCRβ+ Jurkat cells by flow cytometry, followed by deep sequencing. NKIRTIL063 library-expressing Jurkat cells that were not subjected to cell sorting were used as reference. Internal control TCRs DMF4 and DMF5 are highlighted for reference.
Extended Data Fig. 5 Quality control of the OVC190 TCR libraries.
(a) Overall assembly accuracies and frequencies of TCRs in the fully assembled OVC190 CD4+ TIL-derived VCV libraries (combined n=1,341 TCRs). Selected TCRs were encoded in replicate, resulting in a total of 2,899 unique sequences that were assembled in two separate reactions (SP1 and SP2). Dots represent individual TCRs. Box plots depict the median, the interquartile range and whiskers extending to minimal and maximal values. (b) Overall assembly accuracies and frequencies of TCRs in the fully assembled OVC190 CD8+ TIL-derived VCV library (n=274 TCRs). (c) Pie chart depicting the overall fraction of OVC190 CD4+ and CD8+ TIL-derived VCV molecules with and without sequence errors (taking both assembly accuracy and sequence mutations into account).
Extended Data Fig. 6 Quality control of the CV19 TCR library.
(a) Overall assembly accuracies and frequencies of TCRs in the fully assembled CV19 TIL-derived VCV libraries (combined n=1,501 TCRs). Sequences were assembled in two separate reactions (SP1 and SP2). Dots represent individual TCRs. Box plots depict the median, the interquartile range and whiskers extending to minimal and maximal values. (b) Pie chart depicting the overall fraction of CV19 VCV molecules with and without sequence errors (taking both assembly accuracy and sequence mutations into account).
Extended Data Fig. 7 Functional screening of intratumoral TCR repertoire of patient CV19.
(a) The SiHa cervical cancer cell line was engineered to express the entire MHC class I haplotype of patient CV19 (A*24:02, A*33:03, B*38:15, B*55:02, C*03:02 and C*12:03). The CV19 TCR library (n=1,501 TCRs) was subsequently expressed in donor T cells and screened against either the unmodified or MHC-modified SiHa line. Fold change represents the relative abundance of TCRs after incubating T cells with unmodified or MHC-modified SiHa cells. TCRs selected for validation are highlighted in red. (b, c) To assess TIL reactivity against HPV-derived antigens, the patient TCR library was screened against K562 cells that were modified to express the patient’s MHC class I alleles as well as the full ORF of either HPV E6 (b) or E7 (c) oncoproteins. Data are depicted as in (a). Two of four selected TCRs (2495 and 362) responded to E7-expressing K562 cells, while no TCRs responded against E6-expressing cells. (d) The reactivity of selected TCRs was validated by amplifying TCRs from the CV19 VCV pool, followed by expression in donor T cells and co-incubation with the indicated cell lines. T cell activation was assessed by measuring CD137 expression using flow cytometry. (e, f) MHC restriction of selected TCRs was assessed by expressing individual patient MHC alleles in SiHa cells and incubating resulting cells with donor T cells engineered to express selected TCRs. T cell activation was assessed by measuring CD137 surface expression. (g) SiHa-reactive TCRs 1007 and 3645, but not E7-specific TCRs 362 and 2495, recognize autologous patient tumor cells. TCR-modified donor T cells were incubated with unmodified, HLA-B*55:02-modified or HLA-B*38:15-modified SiHa cells, or autologous dissociated tumor tissue at the indicated effector to target ratios. Activation of TCR-engineered T cells in measured by IFNγ ELISpot.
Extended Data Fig. 8 Sensitive TCR discovery across variable TCR frequencies and sequence fidelities.
(a) Projection of all NKIRTIL063 screen hit TCRs with confirmed (blue, n=44) and unconfirmed (red, n=36) reactivity (as reported in Fig. 3) onto the quality control data of the NKIRTIL063 TCR library (see Extended Data Fig. 4). (b, c) Comparison between the frequencies (b) and assembly accuracies (c) of the overall NKIRTIL063 TCR library, TCRs with confirmed reactivity and TCRs with unconfirmed reactivity. Box plots depict the median, the interquartile range and whiskers extending to minimal and maximal values. P values were determined using the Kruskal-Wallis test.
Extended Data Fig. 9 Neoantigen specificities of OVC190 TCRs.
(a) TCR-modified CD4+ Jurkat cells were incubated with patient B cells expressing either the mutant or wildtype sequence of the individual minigenes of TMG 5. T cell activation was determined by measuring CD69 expression on T cells by flow cytometry. (b) CD8+ Jurkat cells were transduced with the CD8+ TIL-derived TCR library of patient OVC190 (n=274 unique TCRs). Patient immortalized B cells were transduced with TMGs encoding all expressed non-synonymous mutations (n=61) of the patient’s tumor, combined in pools and used to screen the OVC190 TCR library. Dots represent individual TCRs. Fold change represents the relative abundance of TCRs in cultures with the indicated TMG pools. (c) Flow cytometry analysis of MHC class I (top panels) and MHC class II (bottom panels) expression on CD45+ and CD45- cells within the OVC190 tumor. Fluorescence minus one (FMO) stains with antibody panels that lacked either the panMHC-I or panMHC-II antibody served as negative control. (d) CD8+ Jurkat cells expressing the OVC190 CD8+ TIL-derived TCR library were screened against single cell suspension of OVC190 tumor. Screening the TCR library against tumor cells in the presence of MHC class I blocking antibody (clone W6-32) served as negative control. Data are depicted as in (b).
Extended Data Fig. 10 Patient HC25 tumor-specific TCR identification.
(a) Patient HC25 PD-1+ TIL were sorted by flow cytometry, and subjected to paired single cell RNA and TCR sequencing. NeoTCR4 and NeoTCR8 transcriptional signatures were derived for CD4+ and CD8+ clonotypes, respectively, and clonotypes with the 382 highest scores were gene-synthesized. Individual TCRs were expressed in donor T cells and reactivity to autologous dissociated tumor tissue was assessed by IFNγ ELISpot. Wells with responding TCRs are marked by red asterisks. Red boxes indicate HC25 TCR-independent experimental controls. (b) Selective reactivity of hit TCRs to patient tumor cells, but not non-malignant cells, was validated by incubating TCR-engineered donor T cells with either medium, patient HC25 activated T blasts, or patient HC25 dissociated tumor tissue. T cell activation was assessed by measuring CD137 expression using flow cytometry. Asterisks indicate TCRs with selective tumor-reactivity.
Supplementary information
Supplementary Tables
Supplementary Tables 1–29.
POC TCRsZip sequences. POC a-b pairing. POC a-b uniformity. ConnA sequences. ConnB sequences. POC Va-CDR3Ja. POC Vb-CDR3Jb. NKIRTIL063 Ref. NKIRTIL063 QC. OVC190CD4 SP1 Ref. OVC190CD4 SP2 Ref. OVC190CD4 SP1 QC. OVC190CD4 SP2 QC. OVC190CD8 Ref. OVC190CD8 QC. CV19 SP1 Ref. CV19 SP2 Ref. CV19 SP1 QC. CV19 SP2 QC. HC25 Ref. POC Oligos. NKIRTIL063 Oligos. OVC190CD4-SP1 Oligos. OVC190CD4-SP2 Oligos. OVC190CD8 Oligos. CV19-SP1 Oligos. CV19-SP2 Oligos. Other sequences.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
Reprints and permissions
About this article
Cite this article
Moravec, Z., Zhao, Y., Voogd, R. et al. Discovery of tumor-reactive T cell receptors by massively parallel library synthesis and screening.
Nat Biotechnol (2024). https://doi.org/10.1038/s41587-024-02210-6
Download citation
Received: 26 May 2023
Accepted: 18 March 2024
Published: 23 April 2024
DOI: https://doi.org/10.1038/s41587-024-02210-6
>>> Read full article>>>
Copyright for syndicated content belongs to the linked Source : Nature.com – https://www.nature.com/articles/s41587-024-02210-6