Immune-awakening revealed by peripheral T cell dynamics after one cycle of immunotherapy

Sara Valpione, Elena Galvani, Joshua Tweedy, Piyushkumar A Mundra, Antonia Banyard, Philippa Middlehurst, Jeff Barry, Sarah Mills, Zena Salih, John Weightman, Avinash Gupta, Gabriela Gremel, Franziska Baenke, Nathalie Dhomen, Paul C Lorigan, Richard Marais, Sara Valpione, Elena Galvani, Joshua Tweedy, Piyushkumar A Mundra, Antonia Banyard, Philippa Middlehurst, Jeff Barry, Sarah Mills, Zena Salih, John Weightman, Avinash Gupta, Gabriela Gremel, Franziska Baenke, Nathalie Dhomen, Paul C Lorigan, Richard Marais

Abstract

Our understanding of how checkpoint inhibitors (CPI) affect T cell evolution is incomplete, limiting our ability to achieve full clinical benefit from these drugs. Here we analyzed peripheral T cell populations after one cycle of CPI and identified a dynamic awakening of the immune system revealed by T cell evolution in response to treatment. We sequenced T cell receptors (TCR) in plasma cell-free DNA (cfDNA) and peripheral blood mononuclear cells (PBMC) and performed phenotypic analysis of peripheral T cell subsets from metastatic melanoma patients treated with CPI. We found that early peripheral T cell turnover and TCR repertoire dynamics identified which patients would respond to treatment. Additionally, the expansion of a subset of immune-effector peripheral T cells we call TIE cells correlated with response. These events are prognostic and occur within 3 weeks of starting immunotherapy, raising the potential for monitoring patients responses using minimally invasive liquid biopsies."

Conflict of interest statement

Conflict of Interest: RM is a consultant for Pfizer and has a drug discovery programme with Basilea Pharmaceutica. PL serves as paid advisor/speaker for Bristol-Myers Squibb, Merck Sharp and Dohme, Roche, Novartis, Amgen, Pierre Fabre, Nektar, Melagenix. PL reports travel support from Bristol-Myers Squibb and Merck Sharp and Dohme, and receives research support from Bristol-Myers Squibb. AG received honoraria and consultancy fees from BMS and Novartis.

Figures

Extended Data Fig. 1. Schematic summarizing T…
Extended Data Fig. 1. Schematic summarizing T cell maturation and life-cycle.
a Pro-T cells undergo sequential somatic recombination of their T cell Receptor β (TCR) loci in attempts to generate functional TCR with unique CDR3 antigen binding regions. Cells that fail to generate a functional TCRβ at the first attempt can recombine their second TCR allele, but cells which fail to produce a functional TCR at the end of the process (crossed red box) are eliminated (β-selection) and their DNA, which encodes the CDR3unique regions, enters the blood as circulating cell-free DNA (cfDNA). Surviving cells retain the T cell receptor excision circle (TREC) generated during TCR locus rearrangement as an episome in the nucleus. The TREC does not replicate so is diluted during subsequent cell divisions. b T cells with a functional TCR undergo positive and negative selection (+/- selection) for HLA and self-antigen recognition. TheCDR3 DNA from T cells eliminated during this step is released into the blood. c Naive T cells enter the circulation as early thymic emigrants (ETE). d T cells primed by antigen presenting cells (APC) in the lymphatic system undergo clonal expansion, which dilutes the TREC amongst the daughter cells. e T cell homeostasis is maintained by subsequent contraction (turnover cycles), releasing further CDR3 DNA into the blood.
Extended Data Fig. 2. Gating strategy for…
Extended Data Fig. 2. Gating strategy for the identification of T cell subsets in peripheral blood of melanoma patients.
Multiparametric fluorescence activated cell sorting analysis using the indicated gates. a Lymphocyte gate on side scatter/forward scatter; b single cell gate to exclude doublets; clive gating to exclude dead cells from subsequent gates; dCD3+ gate for T cells; e,f CD4+ and CD8+ gates for “helper” and “killer” T cell subsets, CD8 was detected with a PE-Cy7 labelled antibody for the Treg panel (e) and with a FITC labelled antibody for the T maturation panel (f); gCD4+/CD25+/CD127-/low regulatory T cells (Treg); h naive (top left) and memory (bottom right) gates total T cells; i ETE (top) and CD31-naive (bottom) gates for naive T cells; j naive (top left) and memory (bottom right) gates for CD8+ T cells; k CD8+ memory T cell subsets, the left bottom subset (CCR7-/CD27-) represents the TIE cells.
Extended Data Fig. 3. Clonal relatedness in…
Extended Data Fig. 3. Clonal relatedness in tumor infiltrating T cells and PBMC.
a Clonal relatedness changes in PBMC-private and TIL-private TCR pools; comparison of week 3 (W3)CDR3 clonal relatedness in patients with progressive disease (PD, n=11 patients) and disease control at week 12 (DC, n=7 patients) in the PBMC-private (P=0.724, median=0.6x10-6 and 0.6x10-6, respectively; two-sided Mann-Whitney U test) and TIL-private pools (P=0.246, median= 0.5x10-4 and 0.8x10-5, respectively; two-sided Mann-Whitney U test). Dot represents one patient; green indicates DC; orange indicates PD; error bar is standard deviation.
Extended Data Fig. 4. Identification of T…
Extended Data Fig. 4. Identification of TIE in CPI-treated patient PBMC.
a Comparison of differential abundance of TIE in CD8+ memory T cells in the PBMC of The Christie NHS Foundation Trust patients with best response progressive disease (PD, orange, n=14) and disease control (DC, green, n=16) at T0 (n=30, light shade) and week 9 (W9; n=10, dark shade; PD, n=4, DC, n=6). Differences over time were not significant for PD (median=15.2 and 35.5; P=0.375; two-sided Wilcoxon test) or DC (median=7.9 and 24; P=0.219; two-sided Wilcoxon test); PD vs DC patient values did not differ at T0 (P=0.275; two-sided Mann-Whitney U test) or W9 (P=0.762; two-sided Mann-Whitney U test). b Distributions of marker intensities of the T cell surface markers in the 20 cell populations (clusters) for PBMC from a published cohort3 (n=20 patients). Cluster 5 was identified as the TIE subset. Blue densities are calculated over all the cells and serve as a reference and red densities represent marker expression for cells in a given cluster. Arrows highlight the TIE subset. c T-stochastic neighbor embedding of single cell profiles (dots) performed in an external cohort3 using the T cell surface markers CD3, CD4, CD8, CD45RA, CD45RO, CCR7 and CD27; different colors are attributed by clustering. Arrow highlights the TIE subset. d Comparison of the differential abundance of the TIE cluster in the PBMC from a published cohort3 of patients with PD (orange, n=9) or DC (green, n=11) at pre-treatment (light shade, n=20; PD, n=9; DC, n=11) and at week 12 (W12, dark shade, n=20) on treatment with pembrolizumab or nivolumab in the external cohort. Horizontal bars indicate the differences over time for the PD (median at T0=5.9 and W12=9.1; P=0.164; two-sided Wilcoxon test) or DC patients (median at T0=3.8 and W12=3.3; P=0.831; two-sided Wilcoxon test), and difference in the two response groups at T0 or W12 (P=0.37 and P=0.201, respectively; two-sided Mann-Whitney U test). Light and dark orange indicate PD for T0 and W9-W12, respectively, light and dark green indicate DC for T0 and W9-W12, respectively; n represents patients; ns means not significant P values; error bars are standard deviation.
Extended Data Fig. 5. Characterization of T…
Extended Data Fig. 5. Characterization of TIE in PBMC.
Analysis of published cohort of PBMC single cell data from reference #27. a Violin plots of the expression level of selected phenotypic and transcriptomic features of the clusters identifying peripheral T cell subsets (n=7488 single cells), the cluster with TIE phenotype is indicated in red; the plots represent the density probability, the area shapes reflect the data distribution; horizontal lines represent the minima and maxima values; central dots represent the medians. Overall minima, mean and maxima values: surface CD3=0, 0.3785, 4.1396; surface CD8a=0, 0.96327, 6.21476; surface CD45RA=0, 0.8161, 4.8508; surface CD45RO=0, 0.6628, 4.6468; surface CD197/CCR7=0, 0.8961, 5.7975; surface CD69=0, 0.5219, 4.2200; surface CD279=0, 0.09787, 3.84886; surface CD25=0, 0.08653, 4.00428; surface TIGIT=0, 0.4663, 4.2381; surface CD155=0, 0.4850, 4.6679; surface CD40=0, 0.6003, 5.5083; surface CD154=0, 0.4062, 3.8159; surface CD357=0, 0.1193, 4.0316; LGALS2=0, 0.561, 6.089; TYROBP=0, 1.337, 6.662; FCN1=0, 1.290, 6.789; CST3=0, 1.404, 6.504; LST1=0, 1.042, 6.097; LYZ=0, 1.775, 6.859. b T-SNE plot showing the clusters identified by means of the antibody derived tags (ADT) targeted to surface markers (n=7488 single cells); the black arrow indicates the cluster with TIE phenotype. c Plot showing the proportion of cells with the TIE phenotype from the same published cohort after standard in vitro culture (CTRL, n=3 sorted healthy donor peripheral blood CD8+ naïve T cell samples in standard culture) or following stimulation with anti-CD3/anti-CD27 Dynabeads (STIM, n=3 sorted healthy donor peripheral blood CD8+ naïve T cell samples after stimulation) (P=0.0267, two-sided paired t test, two degrees of freedom) andd Volcano plot representing the transcriptomic differential expression of the cells with the TIE phenotype in PBMC presented in a (n=7488 single cells) or expanded from naive CD8+ T cells from the experiment presented inc(n=12217 single cells; two-sided Wilcoxon test with Bonferroni correction for multiple comparisons).
Extended Data Fig. 6. Expression of Ki-67…
Extended Data Fig. 6. Expression of Ki-67 and PD-1 in peripheral TIE cells before and after 1 cycle of CPI.
a Expression of Ki67 and PD1 in the TIE subset as measured by FACS in n=5 frozen samples of PBMC from The Christie NHS Foundation Trust metastatic melanoma patients treated with CPI, at pre-treatment (T0) and after 1 cycle of CPI (W3); horizontal line indicates median; error bar indicates standard deviation. The small sample size did not allow statistical comparison of the outcome groups.
Figure 1. CPI induced peripheral TCR repertoire…
Figure 1. CPI induced peripheral TCR repertoire divergence.
a Graph showing early thymic emigrants in pre-treated patients’ blood (% ETET0 relative to total naive T cells; determined by FACS) relative to age (P=0.002, linear regression R2=-0.17; n=50). b Levels of ETE in pre-treatment (T0) and week 3 (W3) of CPI in paired patient samples (P=0.274, two-sided Wilcoxon test, n=50). c TREC (T cell receptor excision circle) concentration relative to genomic DNA was measured by droplet digital PCR in sorted CD3+ peripheral T cells at T0 (median 0.5x10-3) and W3 (median 0.1x10-2)(P=0.129, two-sided Wilcoxon test, n=17).d Tumor infiltrating T lymphocyte (TIL) CDR3sequences also present in peripheral PBMC and cfDNA for one patient at T0 and W3. See Table 1 for specific DNA sequence; tot = total. e Venn diagram showing unique predicted productive CDR3 sequences in PBMC and TIL for patient #01 at T0 (Supplementary Table 1). Numbers show unique nucleotide sequence counts for PBMC-private (pink), TIL-private (brown) and tePBMC (tumor emigrant PBMC; intersection, orange) pools. fClonal relatedness (the proportion of amino acids sequences that are related by maximum edit distance=3) for CDR3 in the PBMC-private pool, tePBMC and TIL-private pools at T0. Horizontal lines: comparison of clonal relatedness between PBMC-private and TIL-private TCR sequences at T0; ***: P=0.003; n=18, two-sided Wilcoxon test; median=0.4x10-6 and 0.4x10-3, respectively; ****: P<0.0001; n=18, two-sided Wilcoxon test; median=0.4x10-6 and 0.2x10-2, respectively.g Clonal relatedness (maximum edit distance=3 amino acids) for CDR3 sequence in PBMC TCR pools at T0 and W3. Comparison between the clonal relatedness of PBMC-private TCR of patients with progressive disease (PD, orange, n=11, median=4.3x10-5 and 5.6x10-5, respectively) or disease control (DC, green, n=7, median=4.0x10-5 and 8.0x10-5, respectively) after 12 weeks of treatment; ns: not significant (P=0.413 and P=0.999, two-sided Wilcoxon test) and between the clonal relatedness of tePBMC TCR of patients with PD (n=11) or DC (n=7); ns: not significant (P=0.638; two-sided Wilcoxon test; median=0.002 and 0.0008, respectively); *: P=0.031; n=7, two-sided Wilcoxon test; median=0.0017 and 0.0007, respectively). Dot is one patient; line is median; error bar is standard deviation; connecting line is paired samples; ns indicates not significant P values, n represents patients.
Figure 2. CPI induced peripheral T cell…
Figure 2. CPI induced peripheral T cell turnover.
a Venn diagram showing unique predicted productiveCDR3 sequences in PBMC (left, pink), PBMC/cfDNA-shared pool (intersection, purple) and cfDNA (right, blue) for patient #27 at T0 (Supplementary Table 2).b Total number of CDR3 clones at T0 (pink) and W3 (purple) in the PBMC/cfDNA-shared pool (P=0.010, two-sided Wilcoxon test).c Clonal relatedness (maximum edit distance=3 amino acids) for CDR3 in the PBMC-private pool, PBMC/cfDNA-shared pool and cfDNA-private pools at T0. Horizontal lines: comparison of clonal relatedness between PBMC-private and cfDNA-private TCR sequences T0 (P<0.0001; two-sided Wilcoxon test; median was 0.3x10-3 and 0.01, respectively); comparison of clonal relatedness between PBMC-private and PBMC/cfDNA shared TCR sequences T0 (P<0.0001, two-sided Wilcoxon test; median was 0.3x10-3 and 0.06). d Clonal relatedness of CDR3 sequence in PBMC/cfDNA-shared pool at T0 and W3 for patients with progressive disease (the number of patients is 12, PD, orange) or disease control (the number of patients was 16, DC, green) at week 12. Comparison (horizontal lines) of clonal relatedness between: T0 PBMC/cfDNA-shared pool TCR sequences for patients with PD or DC (P=0.623, two-sided Mann-Whitney U test; median is 0.04 and 0.08); W3 PBMC/cfDNA-shared pool TCR sequences for patients with PD or DC (P=0.026; two-sided Mann-Whitney U test; median=0.06 and 0.03); T0 and W3 for the PBMC/cfDNA-shared pool TCR sequences for patients with PD (P=0.733; the number of patients was 12, two-sided Wilcoxon test; median=0.04 and 0.06) or DC (P=0.039; n=16, Wilcoxon test; median=0.08 and 0.03).e Pre-treatment TCR rearrangement efficiency score (REST0) of rearranged CDR3 in healthy donors (HD) and patients on CPI in PBMC (P=0.445; median 0.83 and 0.81; the number of HD was 77 batches and the number of patients was 29; two-sided Mann-Whitney U test) and cfDNA (P=0.09, median 0.44 and 0.62; n=3 and 28; two-sided Mann-Whitney U test). Comparisons (horizontal lines) between: HD PBMC and cfDNA REST0 (P<0.0001, two-sided Mann-Whitney U test); matched samples of patients’ PBMC and cfDNA RES T0 (P<0.0001, Wilcoxon test). f ΔW3RES (change in RES from T0 to W3) according to response group at week 12; **: P=0.008, two-sided Wilcoxon test, median was 0.001 and 0.08; *: P=0.037, two-sided Mann-Whitney U test, median=0.02 and 0.08. Total number of melanoma patients equal to 28; dot is one patient; error bar is standard deviation; connecting line is paired samples; horizontal line is median; T0 indicates pre-treatment; W3 indicates week 3; ns is not significant; * indicates P=0.05-0.01; **** indicates P<0.0001.
Figure 3. Identification of T IE cells.
Figure 3. Identification of TIE cells.
a Correlation between TIE cell abundance (ΔW3TIE) and changes in cfDNA RES (ΔW3RES: RESW3-REST0,) at W3 relative to T0 (P=0.001; linear regression R2=0.34, n=28 patients); dotted line is the linear regression line. b Similarity matrix ofTCR sequences in cfDNA and peripheral CD8+ T cell subsets. TCM, TIE Tnaive and ETE similarity with cfDNA in 6 patients at T0 (median=0.026, 0.045, 0.004 and 0.003, respectively; P=0.0013; Friedman analysis of variance, Friedman statistics=15.64; patient #16 naive subset not assessed) and W3 (median=0.043, 0.136, 0 and 0.003, respectively; P<0.0001; Friedman analysis of variance, Friedman statistics=23.05; patient #16 naive subset not assessed).c Clonality (Gini coefficient) in peripheral CD8+ T cell subsets. TIE subset clonality relative to other subsets at baseline (T0, TIE median clonality=0.46) and after the first cycle of CPI (W3, TIE median clonality=0.61) in 6 matched patient samples (P=0.0006 and 0.0002, Friedman analysis of variance Friedman analysis of variance, Friedman statistics=12.6 and 13.08; patient #16 naive subset not assessed); error bar is standard deviation. The small sample size did not allow the comparison between responders (#16-18) and patients who progressed (#12,19,29); horizontal line is median; error bar is standard deviation.d Graph showing the frequency of pre-treatment TILCDR3 sequences in patient #12 sorted peripheral CD8+ T cell subsets at T0 and W3. Dot in a and c is a single patient.
Figure 4. T IE cells infiltrate tumors…
Figure 4. TIE cells infiltrate tumors that respond to immunotherapy.
T-SNE (t distributed stochastic neighbor embedding) plots of biopsy cell clusters according to T cell surface markers in (a) melanoma (n=18 patient samples), (b) renal cell carcinoma (RCC, n=3 patient samples), (c) glioblastoma (GMB, n=3 patient samples), and (d) non cancerous tonsils (n=4 patient samples). Samples for this analysis were from reference #24. Black arrows highlight the clusters with the TIE phenotype.
Figure 5. Peripheral T IE cell expansion…
Figure 5. Peripheral TIE cell expansion in response to first-line CPI.
a ΔW3TIE in patients with best response PD (orange, n=14, median=-0.58%) or best response DC (green, n=16, median=10.04%; ***: P=0.0007, two-sided Mann-Whitney U test) in the training set. Arrow: patient #20. b ΔW3TIE in patients receiving anti-PD1 monotherapy (αPD1, n=18, median=1.35%) or combination of ipilimumab plus nivolumab (I+N, n=12, median=10.84%; P=0.2, two-sided Mann-Whitney U test). c Variations over time (days from treatment initiation) of variant allele frequency (VAF) of mutantNRASQ61R measured in circulating tumor DNA by droplet digital PCR over the course of CPI treatment for patient #20 (arrow in a). Blood collection ceased after day 90 due to patient complications leading to patient death. d Receiver operating curve showing the sensitivity and false positive rate (1-specificity) of ΔW3TIE values in identifying the patients that will achieve disease control; *: maximum accuracy (cut-off=+0.8%).e Kaplan-Meier survival curves for patients with TIE expansion ≥0.8% (pink, n=12; median survival not reached) at W3 compared to patients without TIE expansion <0.8% (blue, n=18; median survival=9.6 months; P=0.013; log rank test), in the training set.f ΔW3TIE in patients with best response PD (orange, n=3, median=-1.3%) or best response DC (green, n=17, median=3.3%; *: P=0.019, two-sided Mann-Whitney U test) in the validation set.g Kaplan-Meier survival curves for patients with TIE expansion≥0.8% (pink, n=15; median survival not reached) at W3 compared to patients without TIE expansion≥0.8% (blue, n=5; median survival=4.2 months; two-sided log rank test, P=0.003), in the validation set. OS is overall survival; dot represents one patient; n represents patients; horizontal line is median; error bar is standard deviation; dotted vertical line is landmark at week 3.
Figure 6. Expansion of a peripheral regulatory…
Figure 6. Expansion of a peripheral regulatory T cell subset associated with toxicity.
a Graph showing expansion of TIE at W3 (ΔW3TIE) in patients with no ≥grade 3 toxicity (median=1.6; n=33) or ≥grade 3 toxicity (median=3.72; P=0.347; n=17; two-sided Mann-Whitney U test). Line, median; error bar, standard deviation. Orange=PD, green=DC, dot=single patient, triangles=single agent anti-PD1, squares=combination ipilimumab+nivolumab. b Expansion of CD3+/CD4+/CD8-/CD25+/CD127-/low Treg cells (Extended Data Figure 2g) at W3 (ΔW3Treg) according to toxicity at any time between 2 weeks and 6 months (P<0.0001; n=50, two-sided linear regression analysis, R2=0.29). Horizontal line is the median; error bar is standard deviation; orange represents PD; green represents DC; triangle represents one patient treated with single agent anti-PD1 drug; square represents one patient treated with combination ipilimumab+nivolumab; n represents patients; dotted line is linear regression line; ns is not significant; T0 is pre-treatment; W3 is week 3.
Figure 7. TCR repertoire evolution after immune-stimulation.
Figure 7. TCR repertoire evolution after immune-stimulation.
a Changes in CDR3 clonality (Δclonality, Gini coefficient) and diversity (Δdiversity, Renyi index, alpha=1) in peripheral T cells from T0 to W1-2 in healthy donors who received anti-viral vaccination (n=25 healthy donor samples). b Changes inCDR3 clonality (Δclonality, Gini coefficient) and diversity (Δdiversity, Renyi index, alpha=1) in peripheral T cells from T0 to W3 in the training cohort (The Christie NHS Foundation Trust) of advanced melanoma patients receiving first line anti-PD1 based immunotherapy (n=17 patients) who progressed (n=9 patients) or responded (n=8 patients) at week 12.c Δclonality (Gini coefficient) and Δdiversity (Renyi index, alpha=1) in peripheral T cells from T0 to W3 in the validation cohort (The Christie NHS Foundation Trust, Huang et al. and Amaria et al.cohort n=12 patients, n=4 patients and n=11 patients, respectively) of advanced melanoma patients who progressed (n=11 patients) or responded (n=16 patients) at week 12 of anti-PD1 based treatment. Dot is one healthy donor or patient; maroon represents healthy donors in the vaccination cohort; orange represents patients who progressed after 12 weeks of immunotherapy; green represents patients who achieved disease control after 12 weeks of immunotherapy; dotted line is the linear regression line.

References

    1. Badovinac VP, Porter BB, Harty JT. Programmed contraction of CD8(+) T cells after infection. Nat Immunol. 2002;3:619–626. doi: 10.1038/ni804.
    1. Ugurel S, et al. Survival of patients with advanced metastatic melanoma: the impact of novel therapies-update 2017. Eur J Cancer. 2017;83:247–257. doi: 10.1016/j.ejca.2017.06.028.
    1. Wykes MN, Lewin SR. Immune checkpoint blockade in infectious diseases. Nat Rev Immunol. 2018;18:91–104. doi: 10.1038/nri.2017.112.
    1. Goldszmid RS, Dzutsev A, Trinchieri G. Host immune response to infection and cancer: unexpected commonalities. Cell Host Microbe. 2014;15:295–305. doi: 10.1016/j.chom.2014.02.003.
    1. Vance RE, Eichberg MJ, Portnoy DA, Raulet DH. Listening to each other: Infectious disease and cancer immunology. Sci Immunol. 2017;2 doi: 10.1126/sciimmunol.aai9339.
    1. Dunn GP, Old LJ, Schreiber RD. The three Es of cancer immunoediting. Annu Rev Immunol. 2004;22:329–360. doi: 10.1146/annurev.immunol.22.012703.104803.
    1. Huang AC, et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature. 2017;545:60–65. doi: 10.1038/nature22079.
    1. Krieg C, et al. High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat Med. 2018;24:144–153. doi: 10.1038/nm.4466.
    1. Jacquelot N, et al. Predictors of responses to immune checkpoint blockade in advanced melanoma. Nat Commun. 2017;8 doi: 10.1038/s41467-017-00608-2. 592.
    1. Huang AC, et al. A single dose of neoadjuvant PD-1 blockade predicts clinical outcomes in resectable melanoma. Nat Med. 2019;25:454–461. doi: 10.1038/s41591-019-0357-y.
    1. Hozumi N, Tonegawa S. Evidence for somatic rearrangement of immunoglobulin genes coding for variable and constant regions. Proc Natl Acad Sci U S A. 1976;73:3628–3632.
    1. Schatz DG, Baltimore D. Uncovering the V(D)J recombinase. Cell. 2004;116 S103–106, 102 p following S106.
    1. Janeway CA, Jr, T P, Walport M, et al. Immunobiology: The Immune System in Health and Disease. 5th edition. Garland Science; 2001.
    1. Kohler S, Thiel A. Life after the thymus: CD31+ and CD31- human naive CD4+ T-cell subsets. Blood. 2009;113:769–774. doi: 10.1182/blood-2008-02-139154.
    1. Steinmann GG, Klaus B, Muller-Hermelink HK. The involution of the ageing human thymic epithelium is independent of puberty. A morphometric study. Scand J Immunol. 1985;22:563–575.
    1. Geenen V, et al. Quantification of T cell receptor rearrangement excision circles to estimate thymic function: an important new tool for endocrine-immune physiology. J Endocrinol. 2003;176:305–311.
    1. Mangul SMI, Yang HT, Strauli N, Montoya D, Rotman J, Van Der Wey W, Ronas JR, Statz B, Zelikovsky A, Spreafico R. Profiling adaptive immune repertoires across multiple human tissues by RNA Sequencing. bioRxiv. 2016 089235.
    1. Amaria RN, et al. Neoadjuvant immune checkpoint blockade in high-risk resectable melanoma. Nat Med. 2018;24:1649–1654. doi: 10.1038/s41591-018-0197-1.
    1. Coffey D. LymphoSeq: Analyze high-throughput sequencing of T and B cell receptors. R package version 1.4.1. 2017
    1. Alves Sousa AP, et al. Comprehensive Analysis of TCR-beta Repertoire in Patients with Neurological Immune-mediated Disorders. Sci Rep. 2019;9 doi: 10.1038/s41598-018-36274-7. 344.
    1. Radziewicz H, Uebelhoer L, Bengsch B, Grakoui A. Memory CD8+ T cell differentiation in viral infection: a cell for all seasons. World J Gastroenterol. 2007;13:4848–4857. doi: 10.3748/wjg.v13.i36.4848.
    1. Mahnke YD, Brodie TM, Sallusto F, Roederer M, Lugli E. The who's who of T-cell differentiation: human memory T-cell subsets. Eur J Immunol. 2013;43:2797–2809. doi: 10.1002/eji.201343751.
    1. Ribas A, et al. PD-1 Blockade Expands Intratumoral Memory T Cells. Cancer Immunol Res. 2016;4:194–203. doi: 10.1158/2326-6066.CIR-15-0210.
    1. Greenplate AR, et al. Computational Immune Monitoring Reveals Abnormal Double-Negative T Cells Present across Human Tumor Types. Cancer Immunol Res. 2019;7:86–99. doi: 10.1158/2326-6066.CIR-17-0692.
    1. Gremel G, et al. Distinct subclonal tumour responses to therapy revealed by circulating cell-free DNA. Ann Oncol. 2016;27:1959–1965. doi: 10.1093/annonc/mdw278.
    1. Robert C, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372:320–330. doi: 10.1056/NEJMoa1412082.
    1. Peterson VM, et al. Multiplexed quantification of proteins and transcripts in single cells. Nat Biotechnol. 2017;35:936–939. doi: 10.1038/nbt.3973.
    1. Venken K, et al. Natural naive CD4+CD25+CD127low regulatory T cell (Treg) development and function are disturbed in multiple sclerosis patients: recovery of memory Treg homeostasis during disease progression. J Immunol. 2008;180:6411–6420.
    1. Herati RS, et al. Successive annual influenza vaccination induces a recurrent oligoclonotypic memory response in circulating T follicular helper cells. Sci Immunol. 2017;2 doi: 10.1126/sciimmunol.aag2152.
    1. DeWitt WS, et al. Dynamics of the cytotoxic T cell response to a model of acute viral infection. J Virol. 2015;89:4517–4526. doi: 10.1128/JVI.03474-14.
    1. Amaria RN, et al. Neoadjuvant immune checkpoint blockade in high-risk resectable melanoma. Nat Med. 2018 doi: 10.1038/s41591-018-0197-1.
    1. Martin MD, Badovinac VP. Defining Memory CD8 T Cell. Front Immunol. 2018;9:2692. doi: 10.3389/fimmu.2018.02692.
    1. Tomiyama H, Takata H, Matsuda T, Takiguchi M. Phenotypic classification of human CD8+ T cells reflecting their function: inverse correlation between quantitative expression of CD27 and cytotoxic effector function. Eur J Immunol. 2004;34:999–1010. doi: 10.1002/eji.200324478.
    1. Rossi JF, Ceballos P, Lu ZY. Immune precision medicine for cancer: a novel insight based on the efficiency of immune effector cells. Cancer Commun (Lond) 2019;39:34. doi: 10.1186/s40880-019-0379-3.
    1. Yost KE, et al. Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat Med. 2019;25:1251–1259. doi: 10.1038/s41591-019-0522-3.
    1. Cha E, et al. Improved survival with T cell clonotype stability after anti-CTLA-4 treatment in cancer patients. Sci Transl Med. 2014;6 doi: 10.1126/scitranslmed.3008211. 238ra270.
    1. Robert L, et al. CTLA4 blockade broadens the peripheral T-cell receptor repertoire. Clin Cancer Res. 2014;20:2424–2432. doi: 10.1158/1078-0432.CCR-13-2648.
    1. Wieland A, et al. T cell receptor sequencing of activated CD8 T cells in the blood identifies tumor-infiltrating clones that expand after PD-1 therapy and radiation in a melanoma patient. Cancer Immunol Immunother. 2018;67:1767–1776. doi: 10.1007/s00262-018-2228-7.
    1. Wei SC, et al. Distinct Cellular Mechanisms Underlie Anti-CTLA-4 and Anti-PD-1 Checkpoint Blockade. Cell. 2017;170:1120–1133 e1117. doi: 10.1016/j.cell.2017.07.024.
    1. Fritsch RD, et al. Stepwise differentiation of CD4 memory T cells defined by expression of CCR7 and CD27. J Immunol. 2005;175:6489–6497.
    1. Hendriks J, Xiao Y, Borst J. CD27 promotes survival of activated T cells and complements CD28 in generation and establishment of the effector T cell pool. J Exp Med. 2003;198:1369–1380. doi: 10.1084/jem.20030916.
    1. Britschgi MR, Link A, Lissandrin TK, Luther SA. Dynamic modulation of CCR7 expression and function on naive T lymphocytes in vivo. J Immunol. 2008;181:7681–7688.
    1. Larbi A, Fulop T. From "truly naive" to "exhausted senescent" T cells: when markers predict functionality. Cytometry A. 2014;85:25–35. doi: 10.1002/cyto.a.22351.
    1. Sallusto F, et al. Switch in chemokine receptor expression upon TCR stimulation reveals novel homing potential for recently activated T cells. Eur J Immunol. 1999;29:2037–2045. doi: 10.1002/(SICI)1521-4141(199906)29:06<2037::AID-IMMU2037>;2-V.
    1. Geginat J, Lanzavecchia A, Sallusto F. Proliferation and differentiation potential of human CD8+ memory T-cell subsets in response to antigen or homeostatic cytokines. Blood. 2003;101:4260–4266. doi: 10.1182/blood-2002-11-3577.
    1. Valpione S, et al. Plasma total cell-free DNA (cfDNA) is a surrogate biomarker for tumour burden and a prognostic biomarker for survival in metastatic melanoma patients. Eur J Cancer. 2018;88:1–9. doi: 10.1016/j.ejca.2017.10.029.
    1. Falci C, et al. Immune senescence and cancer in elderly patients: results from an exploratory study. Exp Gerontol. 2013;48:1436–1442. doi: 10.1016/j.exger.2013.09.011.
    1. Richardson MW, et al. Analysis of telomere length and thymic output in fast and slow/non-progressors with HIV infection. Biomed Pharmacother. 2000;54:21–31.
    1. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30:2114–2120. doi: 10.1093/bioinformatics/btu170.
    1. Dobin A, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21. doi: 10.1093/bioinformatics/bts635.
    1. Thapa DR, et al. Longitudinal analysis of peripheral blood T cell receptor diversity in patients with systemic lupus erythematosus by next-generation sequencing. Arthritis Res Ther. 2015;17:132. doi: 10.1186/s13075-015-0655-9.
    1. Spreafico R, et al. A circulating reservoir of pathogenic-like CD4+ T cells shares a genetic and phenotypic signature with the inflamed synovial micro-environment. Ann Rheum Dis. 2016;75:459–465. doi: 10.1136/annrheumdis-2014-206226.
    1. Nowicka M, et al. CyTOF workflow: differential discovery in high-throughput high-dimensional cytometry datasets. F1000Res. 2017;6:748. doi: 10.12688/f1000research.11622.2.
    1. Gribov A, et al. SEURAT: visual analytics for the integrated analysis of microarray data. BMC Med Genomics. 2010;3:21. doi: 10.1186/1755-8794-3-21.
    1. Kotecha N, Krutzik PO, Irish JM. Web-based analysis and publication of flow cytometry experiments. Curr Protoc Cytom. 2010;Chapter 10:Unit10–17. doi: 10.1002/0471142956.cy1017s53.

Source: PubMed

3
订阅