T-cell CX3CR1 expression as a dynamic blood-based biomarker of response to immune checkpoint inhibitors

Takayoshi Yamauchi, Toshifumi Hoki, Takaaki Oba, Vaibhav Jain, Hongbin Chen, Kristopher Attwood, Sebastiano Battaglia, Saby George, Gurkamal Chatta, Igor Puzanov, Carl Morrison, Kunle Odunsi, Brahm H Segal, Grace K Dy, Marc S Ernstoff, Fumito Ito, Takayoshi Yamauchi, Toshifumi Hoki, Takaaki Oba, Vaibhav Jain, Hongbin Chen, Kristopher Attwood, Sebastiano Battaglia, Saby George, Gurkamal Chatta, Igor Puzanov, Carl Morrison, Kunle Odunsi, Brahm H Segal, Grace K Dy, Marc S Ernstoff, Fumito Ito

Abstract

Immune checkpoint inhibitors (ICI) have revolutionized treatment for various cancers; however, durable response is limited to only a subset of patients. Discovery of blood-based biomarkers that reflect dynamic change of the tumor microenvironment, and predict response to ICI, will markedly improve current treatment regimens. Here, we investigate CX3C chemokine receptor 1 (CX3CR1), a marker of T-cell differentiation, as a predictive correlate of response to ICI therapy. Successful treatment of tumor-bearing mice with ICI increases the frequency and T-cell receptor clonality of the peripheral CX3CR1+CD8+ T-cell subset that includes an enriched repertoire of tumor-specific and tumor-infiltrating CD8+ T cells. Furthermore, an increase in the frequency of the CX3CR1+ subset in circulating CD8+ T cells early after initiation of anti-PD-1 therapy correlates with response and survival in patients with non-small cell lung cancer. Collectively, these data support T-cell CX3CR1 expression as a blood-based dynamic early on-treatment predictor of response to ICI therapy.

Conflict of interest statement

Igor Puzanov: Amgen consultant. Marc S. Ernstoff receives clinical trial support from Merck, Bristol Myers Squibb, Alkermes, EMD Serono and serves on a BMS DMSC and consultant for ImmuNext, Alkermes, EMD Serono, Merck, and BMS. Kunle Odunsi co-founder of Tactiva Therapeutics and receives research support from Astra Zeneca and Tessaro. Saby George reports consulting fees from and advisory roles for Pfizer, Exelixis, Bristol-Myers Squibb, Sanofi/Genzyme, Genentech, Bayer, Corvus, EMD Serono, Seattle Genetics/Astellas, Eisai, Aveo, and Merck, and institutional grant support from Bristol-Myers Squibb, Novartis, Bayer, Pfizer, Merck, Seattle Genetics/Astellas, Eisai, Calithera Biosciences, Immunomedics, Corvus Pharmaceuticals, Surface Oncology, and Agensys. The remaining authors have no competing interests.

Figures

Fig. 1. Effective immune checkpoint inhibitor therapy…
Fig. 1. Effective immune checkpoint inhibitor therapy correlates with the increased frequency of circulating CX3CR1+ CD8+ T cells.
a Experimental scheme of treatment with immune checkpoint inhibitors (ICI). b Individual tumor growth and survival curves in MC38 and CT26 tumor-bearing mice treated with isotype antibody (Ab) (NT) or anti-PD-L1 Ab and anti-CTLA-4 Ab (ICI). c, d Representative flow cytometry plots and data panel showing the frequency of CX3CR1+ cells among CD8+ T cells (c) and tetramer (Tet)+ CD8+ T cells (d) in peripheral blood (PB) of MC38 and CT26 tumor-bearing mice in different treatments as indicated. Numbers denote percent CX3CR1+ cells. The gating strategy is presented in Supplementary Fig. 1. PB was harvested 2 weeks after initiation of the treatment. n = 5 mice in all groups (bd). e, f Frequency of the PB CX3CR1+ subset among CD8+ T cells (e), tumor growth curves (mean) and survival curves (f) in CT26-bearing mice in different treatment groups as indicated. n = 9 mice (NT), 6 mice (anti-CTLA-4 Ab), 6 mice (anti-PD-L1 Ab), and 5 mice (combo). g Frequency of the PB CX3CR1+ subset among CD8+ T cells in MC38 tumor-bearing mice treated with isotype control Ab (NT) or anti-PD-L1 Ab. n = 7 mice (NT) and 6 mice (anti-PD-L1 Ab). h Tumor growth curves (mean) and survival curves in MC38-bearing mice in different treatment groups as indicated. n = 5 mice in all groups. Data shown in bh are representative of two independent experiments. PB was harvested 1 week after initiation of the treatment (e, g). Arrows indicate initiation of treatment (b, f, h). P-values were determined by a log-rank (Mantel–Cox) test (b, f, h), a two-tailed Mann–Whitney U-test (c, d, g) or Kruskal–Wallis with Dunn’s multiple comparisons (e). Data in f, h are presented as mean ± SEM. Box plots: dot, single PB; hinges, 25th and 75th percentiles; middle line, median; whiskers, minimum to maximum value (ce, g). Source data are provided as a Source Data file.
Fig. 2. Phenotypic analysis of peripheral CD8…
Fig. 2. Phenotypic analysis of peripheral CD8+ T cells in mice treated with immune checkpoint inhibitor (ICI) therapy.
a Gating strategy for phenotypic analysis of three subsets (CD27lo CX3CR1−, CD27hi CX3CR1−, and CX3CR1+) of peripheral CD8+ T cells in mice. b, c Mice bearing 10-day established CT26 tumors were treated with anti-PD-L1 antibody (Ab) and anti-CTLA-4 Ab every 3 days and every other day, respectively. Peripheral blood (PB) (b) and spleen (c) were harvested 2 weeks after initiation of the treatment. Representative flow-cytometric plots of three subsets (CD27lo CX3CR1−, CD27hi CX3CR1−, and CX3CR1+) of PB (b) and splenic (c) CD8+ T cells are shown. Data panels show frequency among CD8+ T cells. NS, not significant, *P < 0.05, **P < 0.005, ***P < 0.0001, by one-way repeated measures ANOVA with Tukey’s multiple comparisons (b, c). n = 9 mice in all groups (b, c). Data shown b, c are representative of two independent experiments. Source data are provided as a Source Data file.
Fig. 3. Phenotypic analysis of PB CX3CR1…
Fig. 3. Phenotypic analysis of PB CX3CR1+ CD8+ T cells before and during ICI therapy.
ac CT26 (ac) or MC38 (c) tumor-bearing mice were treated with anti-PD-L1 Ab and anti-CTLA-4 antibody (Ab) every 3 days and every other day, respectively. Peripheral blood (PB) was obtained before and 1, 2, and 3 weeks after the initiation of the treatment. Gating strategy: all cells > size lymphocytes > singlets > live > CD3+ CD8+ > CD27, CX3CR1. a Ki-67 expression of CD27lo CX3CR1− (green), CD27hi CX3CR1− (blue), and CX3CR1+ (red) CD8+ T cells in PB 2 weeks after ICI therapy. Numbers denote percent Ki-67+ cells. The data panel shows the median fluorescence intensity (MFI) of Ki-67+ cells in each subset. n = 4 mice in all groups. b Box and whiskers plots showing MFI of Ki-67+ (blue) and frequency (red) of the CX3CR1+ subset in CD8+ T cells (upper) and Tet+ CD8+ T cells (lower) at days 0, 7, 14, and 21 in PB. n = 3 mice (day 0) and n = 4 mice (days 7, 14, and 21). a, b Data shown are representative of two independent experiments. c Frequency of PB Tet+ CD8+ T cells in the CD27lo CX3CR1− (green), CD27hi CX3CR1− (blue), and CX3CR1+ (red) subsets before and 1, 2, and 3 weeks after the initiation of the treatment in CT26 (upper) and MC38 (lower) tumor-bearing mice. For CT26 tumor-bearing mice, n = 31 (day 0), n = 18 (day 7), n = 9 (day 14), and n = 9 (day 21) derived from three independent experiments. For MC38 tumor-bearing mice, n = 14 (day 0), n = 29 (day 7), n = 19 (day 14), and n = 8 (day 21) derived from three independent experiments. P-values were determined by one-way repeated measures ANOVA with Tukey’s multiple comparisons (a, c). Box plots: dot, single PB; hinges, 25th and 75th percentiles; middle line, median; whiskers, minimum to maximum value (ac). Source data are provided as a Source Data file.
Fig. 4. Effective ICI therapy induces a…
Fig. 4. Effective ICI therapy induces a high degree of TCR sequence similarity and clonality between tumor-infiltrating CD8+ T cells and peripheral CX3CR1+ CD8+ T cells.
ad MC38 tumor-bearing mice were treated with anti-CTLA-4 antibody (Ab) and anti-PD-L1 Ab. Three subsets of splenic CD8+ T cells determined by CD27 and CX3CR1 expression (CD27lo CX3CR1−, CD27hi CX3CR1−, and CX3CR1+), and CD8+ tumor-infiltrating lymphocytes (TILs) were isolated 2 weeks after the initiation of the treatment for TCR repertoire and clonality analysis. a TCR repertoire overlap by Morisita’s index (left) and representative pairwise scatter plots of the frequency of TCRβ CDR3 amino acid (AA) sequences between each subset of splenic CD8+ T cells and CD8+ TILs (right). n = 3 independent experiments. b TCR clonality analysis of three subsets of splenic CD8+ T cells and CD8+ TILs by top sequence plot (left), Gini index (center), and Lorenz curve (right). The most abundant 100 AA sequences are colored while other less frequent clones are in purple in the top sequence plot. n = 3 independent experiments for Gini index. The top sequence plot and Lorenz curve are representative of three independent experiments. c Representative overlapped weighted TCR repertoire dendrograms by ImmunoMap analysis between three subsets of splenic CD8+ T cells (blue) and CD8+ TILs (red). The distance of the branch ends represents sequence distance, and the size of circles denotes the frequency of sequence. The data shown are representative of three independent experiments. d Number of dominant motifs within top 100 productive sequences shared between three subsets of splenic CD8+ T cells (blue) and CD8+ TILs (red) in (c). The data table shows the number of dominant motifs shared between three subsets of splenic CD8+ T cells and CD8+ TILs from three independent experiments. One-way repeated-measures ANOVA with Tukey’s multiple comparisons (a, b). Values are mean ± SEM. Source data are provided as a Source Data file.
Fig. 5. Expansion of the CX3CR1 +…
Fig. 5. Expansion of the CX3CR1+ subset in PB CD8+ T cells correlates with response to anti-PD-1 therapy and better survival in patients with NSCLC.
a Gating strategy for identifying CX3CR1+ CD8+ T cells in peripheral mononuclear blood cells. Cells were first gated for lymphocytes (SSC-A vs. FSC-A) and for singlets (FSC-H vs. FSC-A). b Overall survival (OS) of patients with high (n = 18) and low (n = 18) pretreatment frequency of the CX3CR1+ subset in PB CD8+ T cells. Cut-points by median baseline frequency of the CX3CR1+ subset in PB CD8+ T cells. c The largest % change of the CX3CR1+ subset in PB CD8+ T cells from baseline by the given time point in responders (CR/PR: n = 13) and non-responders (SD/PD: n = 23) of 36 NSCLC patients treated with anti-PD-1 therapy. CR/PR: complete and partial response, SD/PD: stable and progressive disease. P-values were calculated by a two-tailed Mann–Whitney U-test. Values are median ± SEM. d Percent change of the CX3CR1+ subset in PB CD8+ T cells from baseline (CX3CR1 score) in responders and non-responders. e Objective response rate (ORR) for high and low PD-L1 tumor proportion score (TPS) and PB CX3CR1 score at 3, 6, 9, and 12 weeks. ORR was analyzed by Fisher’s exact test. f Progression-free survival (PFS) and OS for high vs. low CX3CR1 score. P-values were calculated by a log-rank (Mantel–Cox) test (b, f). NS, not significant. Source data are provided as a Source Data file.

References

    1. Hodi FS, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 2010;363:711–723. doi: 10.1056/NEJMoa1003466.
    1. Topalian SL, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012;366:2443–2454. doi: 10.1056/NEJMoa1200690.
    1. Brahmer JR, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 2012;366:2455–2465. doi: 10.1056/NEJMoa1200694.
    1. Lesterhuis WJ, et al. Dynamic versus static biomarkers in cancer immune checkpoint blockade: unravelling complexity. Nat. Rev. Drug Discov. 2017;16:264–272. doi: 10.1038/nrd.2016.233.
    1. Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat. Rev. Cancer. 2016;16:275–287. doi: 10.1038/nrc.2016.36.
    1. Nishino M, Ramaiya NH, Hatabu H, Hodi FS. Monitoring immune-checkpoint blockade: response evaluation and biomarker development. Nat. Rev. Clin. Oncol. 2017;14:655–668. doi: 10.1038/nrclinonc.2017.88.
    1. Havel JJ, Chowell D, Chan TA. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat. Rev. Cancer. 2019;19:133–150. doi: 10.1038/s41568-019-0116-x.
    1. Herbst RS, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–567. doi: 10.1038/nature14011.
    1. Powles T, et al. MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature. 2014;515:558–562. doi: 10.1038/nature13904.
    1. Tumeh PC, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:568–571. doi: 10.1038/nature13954.
    1. Rizvi NA, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:124–128. doi: 10.1126/science.aaa1348.
    1. Horn L, et al. Nivolumab versus docetaxel in previously treated patients with advanced non-small-cell lung cancer: two-year outcomes from two randomized, open-label, phase III trials (CheckMate 017 and CheckMate 057) J. Clin. Oncol. 2017;35:3924–3933. doi: 10.1200/JCO.2017.74.3062.
    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. Taube JM, et al. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin. Cancer Res. 2014;20:5064–5074. doi: 10.1158/1078-0432.CCR-13-3271.
    1. Riaz, N. et al. Tumor and microenvironment evolution during immunotherapy with nivolumab. Cell171, 934–949.e16 (2017).
    1. Vilain RE, et al. Dynamic changes in PD-L1 expression and immune infiltrates early during treatment predict response to PD-1 blockade in melanoma. Clin. Cancer Res. 2017;23:5024–5033. doi: 10.1158/1078-0432.CCR-16-0698.
    1. Chen PL, et al. Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade. Cancer Discov. 2016;6:827–837. doi: 10.1158/-15-1545.
    1. Anagnostou V, et al. Evolution of neoantigen landscape during immune checkpoint blockade in non-small cell lung cancer. Cancer Discov. 2017;7:264–276. doi: 10.1158/-16-0828.
    1. Chen G, et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature. 2018;560:382–386. doi: 10.1038/s41586-018-0392-8.
    1. Gandara DR, et al. Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab. Nat. Med. 2018;24:1441–1448. doi: 10.1038/s41591-018-0134-3.
    1. Khagi Y, et al. Hypermutated circulating tumor DNA: correlation with response to checkpoint inhibitor-based immunotherapy. Clin. Cancer Res. 2017;23:5729–5736. doi: 10.1158/1078-0432.CCR-17-1439.
    1. Valpione S, et al. Immune-awakening revealed by peripheral T cell dynamics after one cycle of immunotherapy. Nat. Cancer. 2020;1:210–221. doi: 10.1038/s43018-019-0022-x.
    1. Cescon DW, Bratman SV, Chan SM, Siu LL. Circulating tumor DNA and liquid biopsy in oncology. Nat. Cancer. 2020;1:276–290. doi: 10.1038/s43018-020-0043-5.
    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. Kamphorst AO, et al. Proliferation of PD-1+ CD8 T cells in peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc. Natl Acad. Sci. USA. 2017;114:4993–4998. doi: 10.1073/pnas.1705327114.
    1. Kim KH, et al. The first-week proliferative response of peripheral blood PD-1(+)CD8(+) T cells predicts the response to anti-PD-1 therapy in solid tumors. Clin. Cancer Res. 2019;25:2144–2154. doi: 10.1158/1078-0432.CCR-18-1449.
    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. Liakou CI, et al. CTLA-4 blockade increases IFNgamma-producing CD4+ICOShi cells to shift the ratio of effector to regulatory T cells in cancer patients. Proc. Natl Acad. Sci. USA. 2008;105:14987–14992. doi: 10.1073/pnas.0806075105.
    1. Bottcher JP, et al. Functional classification of memory CD8(+) T cells by CX3CR1 expression. Nat. Commun. 2015;6:8306. doi: 10.1038/ncomms9306.
    1. Gerlach C, et al. The chemokine receptor CX3CR1 defines three antigen-experienced CD8 T cell subsets with distinct roles in immune surveillance and homeostasis. Immunity. 2016;45:1270–1284. doi: 10.1016/j.immuni.2016.10.018.
    1. Gordon CL, et al. Induction and maintenance of CX3CR1− intermediate peripheral memory CD8(+) T cells by persistent viruses and vaccines. Cell Rep. 2018;23:768–782. doi: 10.1016/j.celrep.2018.03.074.
    1. Wallin JJ, et al. Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nat. Commun. 2016;7:12624. doi: 10.1038/ncomms12624.
    1. Yan Y, et al. CX3CR1 identifies PD-1 therapy-responsive CD8+ T cells that withstand chemotherapy during cancer chemoimmunotherapy. JCI Insight. 2018;3:e97828. doi: 10.1172/jci.insight.97828.
    1. Blank C, et al. PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell receptor (TCR) transgenic CD8+ T cells. Cancer Res. 2004;64:1140–1145. doi: 10.1158/0008-5472.CAN-03-3259.
    1. Ahn E, et al. Role of PD-1 during effector CD8 T cell differentiation. Proc. Natl Acad. Sci. USA. 2018;115:4749–4754. doi: 10.1073/pnas.1718217115.
    1. Yadav M, et al. Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing. Nature. 2014;515:572–576. doi: 10.1038/nature14001.
    1. Huang AY, et al. The immunodominant major histocompatibility complex class I-restricted antigen of a murine colon tumor derives from an endogenous retroviral gene product. Proc. Natl Acad. Sci. USA. 1996;93:9730–9735. doi: 10.1073/pnas.93.18.9730.
    1. Mosely SI, et al. Rational selection of syngeneic preclinical tumor models for immunotherapeutic drug discovery. Cancer Immunol. Res. 2017;5:29–41. doi: 10.1158/2326-6066.CIR-16-0114.
    1. Oba T, et al. Overcoming primary and acquired resistance to anti-PD-L1 therapy by induction and activation of tumor-residing cDC1s. Nat. Commun. 2020;11:5415. doi: 10.1038/s41467-020-19192-z.
    1. Wei SC, et al. Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade. Cell. 2017;170:1120–1133. doi: 10.1016/j.cell.2017.07.024.
    1. Mikucki ME, et al. Non-redundant requirement for CXCR3 signalling during tumoricidal T-cell trafficking across tumour vascular checkpoints. Nat. Commun. 2015;6:7458. doi: 10.1038/ncomms8458.
    1. von Andrian UH, Mempel TR. Homing and cellular traffic in lymph nodes. Nat. Rev. Immunol. 2003;3:867–878. doi: 10.1038/nri1222.
    1. Yamauchi T, et al. CX3CR1−CD8+ T cells are critical in antitumor efficacy, but functionally suppressed in the tumor microenvironment. JCI Insight. 2020;5:e133920. doi: 10.1172/jci.insight.133920.
    1. Gros A, et al. Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat. Med. 2016;22:433–438. doi: 10.1038/nm.4051.
    1. Morisita M. Measuring of the dispersion of individuals and analysis of the distributional patterns. Mem. Fac. Sci. Kyushu Univ. Ser. E. 1959;2:5–23.
    1. Glanville J, et al. Identifying specificity groups in the T cell receptor repertoire. Nature. 2017;547:94–98. doi: 10.1038/nature22976.
    1. Dash P, et al. Quantifiable predictive features define epitope-specific T cell receptor repertoires. Nature. 2017;547:89–93. doi: 10.1038/nature22383.
    1. Sidhom JW, et al. ImmunoMap: a bioinformatics tool for T-cell repertoire analysis. Cancer Immunol. Res. 2018;6:151–162. doi: 10.1158/2326-6066.CIR-17-0114.
    1. Conroy JM, et al. Analytical validation of a next-generation sequencing assay to monitor immune responses in solid tumors. J. Mol. Diagn. 2018;20:95–109. doi: 10.1016/j.jmoldx.2017.10.001.
    1. Seymour L, et al. iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol. 2017;18:e143–e152. doi: 10.1016/S1470-2045(17)30074-8.
    1. Garon EB, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N. Engl. J. Med. 2015;372:2018–2028. doi: 10.1056/NEJMoa1501824.
    1. Herbst RS, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1−positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2016;387:1540–1550. doi: 10.1016/S0140-6736(15)01281-7.
    1. Youden WJ. Index for rating diagnostic tests. Cancer. 1950;3:32–35. doi: 10.1002/1097-0142(1950)3:1<32::AID-CNCR2820030106>;2-3.
    1. Lim C, et al. Biomarker testing and time to treatment decision in patients with advanced nonsmall-cell lung cancer. Ann. Oncol. 2015;26:1415–1421. doi: 10.1093/annonc/mdv208.
    1. Green MR, et al. Molecular testing prior to first-line therapy in patients with stage IV nonsquamous non-small cell lung cancer (NSCLC): a survey of U.S. medical oncologists. J. Clin. Oncol. 2014;32:8097–8097. doi: 10.1200/jco.2014.32.15_suppl.8097.
    1. Wang W, et al. Biomarkers on melanoma patient T cells associated with ipilimumab treatment. J. Transl. Med. 2012;10:146. doi: 10.1186/1479-5876-10-146.
    1. Zappasodi R, et al. Non-conventional inhibitory CD4(+)Foxp3(-)PD-1(hi) T cells as a biomarker of immune checkpoint blockade activity. Cancer Cell. 2018;33:1017–1032. doi: 10.1016/j.ccell.2018.05.009.
    1. Guo X, et al. Global characterization of T cells in non-small-cell lung cancer by single-cell sequencing. Nat. Med. 2018;24:978–985. doi: 10.1038/s41591-018-0045-3.
    1. Han J, et al. TCR repertoire diversity of peripheral PD-1(+)CD8(+) T cells predicts clinical outcomes after immunotherapy in patients with non-small cell lung cancer. Cancer Immunol. Res. 2020;8:146–154. doi: 10.1158/2326-6066.CIR-19-0398.
    1. Wu TD, et al. Peripheral T cell expansion predicts tumour infiltration and clinical response. Nature. 2020;579:274–278. doi: 10.1038/s41586-020-2056-8.
    1. Fairfax BP, et al. Peripheral CD8(+) T cell characteristics associated with durable responses to immune checkpoint blockade in patients with metastatic melanoma. Nat. Med. 2020;26:193–199. doi: 10.1038/s41591-019-0734-6.
    1. Larkin J, et al. Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med. 2019;381:1535–1546. doi: 10.1056/NEJMoa1910836.
    1. Hellmann MD, et al. Nivolumab plus ipilimumab in advanced non-small-cell lung cancer. N. Engl. J. Med. 2019;381:2020–2031. doi: 10.1056/NEJMoa1910231.
    1. Duraiswamy J, Kaluza KM, Freeman GJ, Coukos G. Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors. Cancer Res. 2013;73:3591–3603. doi: 10.1158/0008-5472.CAN-12-4100.
    1. Binder DC, Schreiber H. Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors—letter. Cancer Res. 2014;74:632. doi: 10.1158/0008-5472.CAN-13-2216.
    1. Binder DC, et al. Antigen-specific bacterial vaccine combined with anti-PD-L1 rescues dysfunctional endogenous T cells to reject long-established cancer. Cancer Immunol. Res. 2013;1:123–133. doi: 10.1158/2326-6066.CIR-13-0058.
    1. Subudhi SK, et al. Clonal expansion of CD8 T cells in the systemic circulation precedes development of ipilimumab-induced toxicities. Proc. Natl Acad. Sci. USA. 2016;113:11919–11924. doi: 10.1073/pnas.1611421113.
    1. Carlson CS, et al. Using synthetic templates to design an unbiased multiplex PCR assay. Nat. Commun. 2013;4:2680. doi: 10.1038/ncomms3680.
    1. Coffey, D. LymphoSeq: Analyze High-Throughput Sequencing of T and B Cell Receptors. R Package Version 1140 (2019).
    1. Pielou EC. Species-diversity and pattern-diversity in the study of ecological succession. J. Theor. Biol. 1966;10:370–383. doi: 10.1016/0022-5193(66)90133-0.
    1. Sauerbrei W, Taube SE, McShane LM, Cavenagh MM, Altman DG. Reporting recommendations for tumor marker prognostic studies (REMARK): an abridged explanation and elaboration. J. Natl Cancer Inst. 2018;110:803–811. doi: 10.1093/jnci/djy088.
    1. Dako. PD-L1 IHC 22C3 pharmDx: Non-Squamous Non-Small Cell Lung Cancer [Interpretation Manual] (2020).
    1. DeLong ER, DeLong DM, Clarke-Pearson DL. Comparing the areas under two or more correlated receiver operating characteristic curves: a nonparametric approach. Biometrics. 1988;44:837–845. doi: 10.2307/2531595.

Source: PubMed

3
Prenumerera