Low-Dose Radiotherapy Reverses Tumor Immune Desertification and Resistance to Immunotherapy

Fernanda G Herrera, Catherine Ronet, Maria Ochoa de Olza, David Barras, Isaac Crespo, Massimo Andreatta, Jesus Corria-Osorio, Aodrenn Spill, Fabrizio Benedetti, Raphael Genolet, Angela Orcurto, Martina Imbimbo, Eleonora Ghisoni, Blanca Navarro Rodrigo, Dominik R Berthold, Apostolos Sarivalasis, Khalil Zaman, Rafael Duran, Clarisse Dromain, John Prior, Niklaus Schaefer, Jean Bourhis, Georgia Dimopoulou, Zoi Tsourti, Marius Messemaker, Thomas Smith, Sarah E Warren, Periklis Foukas, Sylvie Rusakiewicz, Mikaël J Pittet, Stefan Zimmermann, Christine Sempoux, Urania Dafni, Alexandre Harari, Lana E Kandalaft, Santiago J Carmona, Denarda Dangaj Laniti, Melita Irving, George Coukos, Fernanda G Herrera, Catherine Ronet, Maria Ochoa de Olza, David Barras, Isaac Crespo, Massimo Andreatta, Jesus Corria-Osorio, Aodrenn Spill, Fabrizio Benedetti, Raphael Genolet, Angela Orcurto, Martina Imbimbo, Eleonora Ghisoni, Blanca Navarro Rodrigo, Dominik R Berthold, Apostolos Sarivalasis, Khalil Zaman, Rafael Duran, Clarisse Dromain, John Prior, Niklaus Schaefer, Jean Bourhis, Georgia Dimopoulou, Zoi Tsourti, Marius Messemaker, Thomas Smith, Sarah E Warren, Periklis Foukas, Sylvie Rusakiewicz, Mikaël J Pittet, Stefan Zimmermann, Christine Sempoux, Urania Dafni, Alexandre Harari, Lana E Kandalaft, Santiago J Carmona, Denarda Dangaj Laniti, Melita Irving, George Coukos

Abstract

Developing strategies to inflame tumors is critical for increasing response to immunotherapy. Here, we report that low-dose radiotherapy (LDRT) of murine tumors promotes T-cell infiltration and enables responsiveness to combinatorial immunotherapy in an IFN-dependent manner. Treatment efficacy relied upon mobilizing both adaptive and innate immunity and depended on both cytotoxic CD4+ and CD8+ T cells. LDRT elicited predominantly CD4+ cells with features of exhausted effector cytotoxic cells, with a subset expressing NKG2D and exhibiting proliferative capacity, as well as a unique subset of activated dendritic cells expressing the NKG2D ligand RAE1. We translated these findings to a phase I clinical trial administering LDRT, low-dose cyclophosphamide, and immune checkpoint blockade to patients with immune-desert tumors. In responsive patients, the combinatorial treatment triggered T-cell infiltration, predominantly of CD4+ cells with Th1 signatures. Our data support the rational combination of LDRT with immunotherapy for effectively treating low T cell-infiltrated tumors. SIGNIFICANCE: Low-dose radiation reprogrammed the tumor microenvironment of tumors with scarce immune infiltration and together with immunotherapy induced simultaneous mobilization of innate and adaptive immunity, predominantly CD4+ effector T cells, to achieve tumor control dependent on NKG2D. The combination induced important responses in patients with metastatic immune-cold tumors.This article is highlighted in the In This Issue feature, p. 1.

©2021 The Authors; Published by the American Association for Cancer Research.

Figures

Figure 1.
Figure 1.
LDRT induces immune-cell infiltration in orthotopic ID8 tumors. A, Treatment schema of mice engrafted with intraperitoneal ID8 ovarian tumors. Arrow represents administration of LD-WART (1 Gy). B and C, NanoString analysis of LD-WART treated versus control tumors. MSigDB pathways (B) and intratumoral levels of cytokines and chemokines (C) are displayed as heat maps. Red, upregulated; blue, downregulated. D, mRNA levels of intratumoral cytokines and chemokines. E, Heat map of cell density changes in tumors based on NanoString analysis. The heat map legend applies to B, C, and E. F, Flow cytometry quantification of TILs. G, CD8+:Foxp3+ cell ratio by mIF imaging five days after LD-WART. H, Flow cytometry quantification of CD8+ TILs in control mice and mice subjected to IFNα receptor blockade or IFNγ depletion. Symbols represent individual tumors and bars the mean. Data are representative of three independent experiments and are presented as mean ± SEM. *, P ≤ 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; Student unpaired t test.
Figure 2.
Figure 2.
Metronomic radiotherapy enables combinatorial immunotherapy. A, Schema of in vivo study evaluating treatment by LD-WART (1 Gy) versus CIM versus RACIM. B, Left, waterfall plot representing the percentage change in tumor BLI levels at day 20 for mice treated in the different groups (RACIM, n = 36 mice; control, n = 41; CIM, n = 26; LD-WART, n = 24). Complete response (CR), PR (at least 30% decrease in BLI from baseline), SD, PD (at least 20% increase in BLI from baseline). Right, tumor growth curves evaluated by BLI. C, Kaplan–Meier analysis in representative mice treated in six different experiments (RACIM, n = 80 mice; control, n = 92; CIM, n = 30; LD-WART, n = 24). P values were determined by a one-sided log-rank Mantel–Cox test. D, Heat map of cell density changes in tumors based on NanoString analysis. E, mIF imaging reveals immune-cell infiltration in tumors at cycle 2, day 5 (20× magnification; DAPI nuclear counterstaining; images are representative of n = 5 mice/group). Number of cells per HPF plotted as mean ± SEM; P was calculated using unpaired two-tailed Student t tests. F–H, Immune-cell phenotypes evaluated on single-cell suspensions of control, RACIM, or RACIM one component, ID8 tumors (n = 5–7 mice per group). Kaplan–Meier analyses of overall survival following RACIM in the absence of anti–PD-1 (F) or anti-CTLA4 antibody, CP (G), or anti-CD40 agonist antibody (H) for n = 10 mice per group. P values were determined by a one-sided log-rank Mantel–Cox test. I, mRNA levels of Tnfa and Ifng in differently treated ID8 tumors. In vivo data are representative of three independent experiments. *, P ≤ 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.
Figure 3.
Figure 3.
Low-dose irradiation and combinatorial immunotherapy expands tumor-rejecting CD4+ and CD8+ TILs exhibiting states of activation and exhaustion. A, UMAP plots of tumor lymphocyte scRNA-seq data (n = 3 tumors/treatment, n = 4 tumors pooled for control, all collected on day 5 of cycle 2). Left, reference map for all groups. Right, contour plots reveal cell density/group. Supervised T-cell state classification by TILPRED identifies functional T-cell subsets: Tpex, Tex, TEM, early activated (EA), Th1, Tfh, Treg, and naïve-like T cells. B, Fold change in T-cell subsets following RACIM versus CIM. C, Cord diagram of the Jaccard similarity coefficient shows the relative number of common TCRs shared between CD4+ T-cell subsets following RACIM. D, Violin plots representing the expression of various activation and cytotoxicity markers in CD4+ T-cell subsets. E, CD4+ T cells expressing indicated genes across subsets and their corresponding average expression (size of dot indicates the percentage of cells in each subset; expression intensity is indicated by color). F, Pseudotime trajectory analysis of CD4_Tpex, CD4_Tex, and Th1 clusters identified by unsupervised single-cell analysis. G, Left, SPICE graphic representing flow-cytometric analysis of GzmB and cytokine production by CD4+ TCF1−PD1+ TILs. Right, bar plots representing cytokine production by CD4+ TCF1−PD-1+ TOX+ cells after PMA/ionomycin or anti-CD3/anti-CD28 TCR stimulation. H, Kaplan–Meier analysis of overall survival of RACIM-treated mice, depleted or not of CD4+ T cells. P values determined by a one-sided log-rank Mantel–Cox test. I, The percentage of CD8+ T cells expressing indicated genes across subsets and their corresponding average expression (as in E). J, Violin plots showing expression of Ifng, Gzmb, and Prf1 in CD8+ T cells following CIM vs. RACIM. K, Bar plots representing the most clonally expanded CD8+ T-cell clonotypes (by TCR-seq) following RACIM treatment (TCRs in all three tumors: #1; in individual tumors: #2–4). L, Cord diagram of the Jaccard similarity coefficient shows the relative number of common TCRs between CD8+ T-cell subsets following RACIM. M, Left, SPICE graphic representing flow cytometric analysis of GzmB and cytokine production by CD8+PD-1+TCF1− TILs. Right: bar plots representing cytokine production by CD8+ TCF1−PD-1+TOX+ cells after PMA/ionomycin or anti-CD3/anti-CD28 TCR stimulation. N, Kaplan–Meier analysis of overall survival of RACIM-treated mice depleted or not of CD8+ T cells. P values were determined by a one-sided log-rank Mantel–Cox test. Data are representative of n = 3 biologically independent experiments (n = 5–10). *, P ≤ 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.
Figure 4.
Figure 4.
RACIM reprograms tumor APCs and enlists NKG2D as a key signal. A, UMAP plots of intratumoral myeloid cell scRNA-seq data (n = 3 tumors/treatment, n = 4 tumors pooled for control, all collected on day 5 of cycle 2). Left, reference map for all groups. Red, DCs; blue, monocytes; green, macrophages. Right, 29 myeloid states among groups. B, Fold change in myeloid cell subsets for RACIM versus CIM. C, Quantification of DC clusters among groups. D, Rose plot of differentially expressed genes corresponding to DC clusters among groups. E and F, Violin plots showing expression of Batf3 (E) and H2k1 and H2d1 (MHC-I; F) transcripts in cDC1 cells among groups. G and H, Kaplan–Meier analysis of control versus RACIM in Batf3−/− mice (G), and in wild-type (WT) mice (H) in the presence of fingolimod (FTY-720) treatment. P values were determined by a one-sided log-rank Mantel–Cox test. I, Heat map showing expression of the most representative genes for clusters 9, 19, and 21. Gene expression was normalized to median expression value per gene across all clusters shown in the heat map. J, Percentage of cells expressing Rae1, Ulbp1, H60b, and H60c, and average expression in the myeloid compartment by scRNA-seq (size of dot indicates the percentage of cells in each subset; expression level is indicated by color). K, RAE1 expression on intratumoral CD11b+CD11c+MHC-II+ cells determined by flow-cytometric analysis on day 5 of cycle 2. L, Left, mIF imaging reveals RAE1 expression (red) by CD11b+ cells (yellow; 20× magnification; DAPI nuclear counterstaining; representative of n = 5 mice/group). Right, number of CD11b+RAE+ cells per HPF plotted as mean ± SD; P was calculated using unpaired two-tailed Student t tests. M and N, The percentage of CD4+ and CD8+ exhausted T cells expressing NKG2D at the transcriptional (M and N, Klrk1 gene by scRNA-seq analysis) and protein levels (O and P, flow cytometry analysis) on day 5 of cycle 2. Q and R, The percentage of intratumoral Ki-67+proliferating CD4+TCF1+PD-1+ (Q) and CD8+TCF1−PD-1+ (R) cells upon RACIM on day 5 of cycle 2. S–T, NKG2D expression on intratumoral CD4+TCF1−PD-1+ (S) and CD8+TCF1−PD1+ (T) T cells determined by flow cytometry on day 5 of cycle 2 in control or RACIM or RACIM without ICB-treated tumors. U, RACIM survival with NKG2D blockade. P values were determined by a one-sided log-rank Mantel–Cox test. Data are representative of two to three independent experiments (n = 5–10 mice/group). Unless otherwise indicated, statistical analysis was performed using Student unpaired t test; error bars represent mean ± standard deviation. *, P ≤ 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.
Figure 5.
Figure 5.
Low-dose irradiation plus ICB induces responses in advanced human immune-desert tumors. A, Therapeutic schema of the phase I RACIN study. B, Spider plot depicts the percentage change in the sum of targeted irradiated metastases compared with baseline. C, Swimmer plot depicts patients' response to RACIN over time; each bar, one patient; light orange, time on combination treatment; green, time on maintenance treatment; cohort 1: 0.5 Gy, cohort 2: 1 Gy; orange triangles, completed treatment; asterisk, treatment termination due to toxicity or progression; black circles, death. iRECIST v1.1 was used to indicate PR (iPR, green diamond), SD (iSD, light blue square), confirmed progressive disease (iCPD, maroon circle), or unconfirmed (iUPD, maroon empty circle). D,68Ga-PSMA PET/CT images of irradiated tumors (white arrows) before and after treatment from a patient with metastatic castration-resistant prostate cancer having SD according to Prostate Cancer Clinical Trials Working Group 3 (PCWG3) but an important response on 68Ga-PSMA PET/CT images. Changes in PSA tumor marker. Progression observed outside the irradiated areas 24 weeks after treatment initiation. E,18FDG-PET/CT images of irradiated tumors (white arrows) before and after treatment from a patient with high-grade serous ovarian carcinoma having by iRECIST iPD, but an important response on 18FDG-PET/CT imaging. Changes in the CA125 tumor marker. Progression outside the irradiated areas 24 weeks after treatment initiation. F, CT images of irradiated tumors (white arrows and circles) before and after treatment from a patient with gallbladder cancer having PR by iRECIST and 70% reduction from baseline in targeted irradiated lesions. Changes in the CA 19-9 tumor marker. Progression outside the irradiated areas 17 weeks after treatment initiation. G, Anatomic location of irradiated target and nontargeted lesions in responder patients and the anatomical location of tumor recurrence (D2, second dorsal vertebrae; D12, dorsal 12; R, right; L, left; LN, lymph node; liver segments identified with roman numbers III, IV, and V).
Figure 6.
Figure 6.
Effect of low-dose irradiation on tumor immune landscape. A, TILs before and after LDRT revealed by mIF imaging in two representative responding tumors. Left, representative mIF images (20× magnification; CK, pancytokeratin); right, quantification of CD4+ and CD8+ cells. B, Scatter plot showing differential gene expression between baseline and post-irradiation biopsy in responding (x-axis) versus nonresponding tumors (y-axis). The log2 of the fold change in median gene expression (log2FC) is shown (positive values indicate upregulation post-LDRT). Genes displaying a significant change (unadjusted P < 0.05) are color-coded as shown in the legend. C, Line plots showing the progression of immune gene signature scores from baseline to post-LDRT biopsies in responding versus nonresponding tumors. D, Scatter plot showing differential immune signature score analysis between baseline and post-irradiation biopsy in responding human tumors (x-axis) versus responding mouse (RACIM) ID8 tumors (y-axis; top) and between nonresponding human versus nonresponding (CIM) mouse tumors (bottom). E, NanoString GeoMx analysis of intraepithelial tumor immune infiltrates vs. tumor stroma immune infiltrates in responding versus nonresponding tumors. The log2 of the fold change in the median of the signature score (log2FC) is shown. Immune signature score displaying a significant change (unadjusted P < 0.05) is color-coded as depicted in the legend of B. F, Comparison of TCR CDR3 diversity by clonality, Shannon diversity entropy, Gini coefficient, richness, and shared frequency in three patients with responding tumors.

References

    1. Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert Let al. . PD1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014;515:568–71.
    1. Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MSet al. . Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014;515:563–7.
    1. Herrera FG, Bourhis J, Coukos G. Radiotherapy combination opportunities leveraging immunity for the next oncology practice. CA Cancer J Clin 2017;67:65–85.
    1. Formenti SC, Rudqvist NP, Golden E, Cooper B, Wennerberg E, Lhuillier Cet al. . Radiotherapy induces responses of lung cancer to CTLA4 blockade. Nat Med 2018;24:1845–51.
    1. Deng L, Liang H, Xu M, Yang X, Burnette B, Arina Aet al. . STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity 2014;41:843–52.
    1. Matsumura S, Wang B, Kawashima N, Braunstein S, Badura M, Cameron TOet al. . Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells. J Immunol 2008;181:3099–107.
    1. Lugade AA, Moran JP, Gerber SA, Rose RC, Frelinger JG, Lord EM. Local radiation therapy of B16 melanoma tumors increases the generation of tumor antigen-specific effector cells that traffic to the tumor. J Immunol 2005;174:7516–23.
    1. Vanpouille-Box C, Alard A, Aryankalayil MJ, Sarfraz Y, Diamond JM, Schneider RJet al. . DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun 2017;8:15618.
    1. Burnette BC, Liang H, Lee Y, Chlewicki L, Khodarev NN, Weichselbaum RRet al. . The efficacy of radiotherapy relies upon induction of type I interferon-dependent innate and adaptive immunity. Cancer Res 2011;71:2488–96.
    1. Demaria S, Kawashima N, Yang AM, Devitt ML, Babb JS, Allison JPet al. . Immune-mediated inhibition of metastases after treatment with local radiation and CTLA4 blockade in a mouse model of breast cancer. Clin Cancer Res 2005;11:728–34.
    1. Deng L, Liang H, Burnette B, Beckett M, Darga T, Weichselbaum RRet al. . Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest 2014;124:687–95.
    1. Twyman-Saint Victor C, Rech AJ, Maity A, Rengan R, Pauken KE, Stelekati Eet al. . Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 2015;520:373–7.
    1. Dovedi SJ, Adlard AL, Lipowska-Bhalla G, McKenna C, Jones S, Cheadle EJet al. . Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. Cancer Res 2014;74:5458–68.
    1. Postow MA, Callahan MK, Barker CA, Yamada Y, Yuan J, Kitano Set al. . Immunologic correlates of the abscopal effect in a patient with melanoma. N Engl J Med 2012;366:925–31.
    1. Klug F, Prakash H, Huber PE, Seibel T, Bender N, Halama Net al. . Low-dose irradiation programs macrophage differentiation to an iNOS(+)/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell 2013;24:589–602.
    1. Barsoumian HB, Ramapriyan R, Younes AI, Caetano MS, Menon H, Comeaux NIet al. . Low-dose radiation treatment enhances systemic antitumor immune responses by overcoming the inhibitory stroma. J Immunother Cancer 2020;8:e000537.
    1. Fyles AW, Dembo AJ, Bush RS, Levin W, Manchul LA, Pringle JFet al. . Analysis of complications in patients treated with abdomino-pelvic radiation therapy for ovarian carcinoma. Int J Radiat Oncol Biol Phys 1992;22:847–51.
    1. Herrera FG, Irving M, Kandalaft LE, Coukos G. Rational combinations of immunotherapy with radiotherapy in ovarian cancer. Lancet Oncol 2019;20:e417–e33.
    1. Kunos CA, Sill MW, Buekers TE, Walker JL, Schilder JM, Yamada SDet al. . Low-dose abdominal radiation as a docetaxel chemosensitizer for recurrent epithelial ovarian cancer: a phase I study of the Gynecologic Oncology Group. Gynecol Oncol 2011;120:224–8.
    1. Ngoi NYL, Heong VYM, Kumarakulasinghe NB, Phyu PS, Peh EY, Lim SEet al. . Phase I study of low dose whole abdominal radiation therapy (LDWART) in combination with weekly paclitaxel (wP) for platinum resistant ovarian cancer (PROC). Ann Oncol 2019;30:v422.
    1. Yang G, Li W, Jiang H, Liang X, Zhao Y, Yu Det al. . Low-dose radiation may be a novel approach to enhance the effectiveness of cancer therapeutics. Int J Cancer 2016;139:2157–68.
    1. Roby KF, Taylor CC, Sweetwood JP, Cheng Y, Pace JL, Tawfik Oet al. . Development of a syngeneic mouse model for events related to ovarian cancer. Carcinogenesis 2000;21:585–91.
    1. Dangaj D, Bruand M, Grimm AJ, Ronet C, Barras D, Duttagupta PAet al. . Cooperation between constitutive and inducible chemokines enables T cell engraftment and immune attack in solid tumors. Cancer Cell 2019;35:885–900.
    1. Crozat K, Guiton R, Contreras V, Feuillet V, Dutertre CA, Ventre Eet al. . The XC chemokine receptor 1 is a conserved selective marker of mammalian cells homologous to mouse CD8alpha+ dendritic cells. J Exp Med 2010;207:1283–92.
    1. Dorner BG, Dorner MB, Zhou X, Opitz C, Mora A, Guttler Set al. . Selective expression of the chemokine receptor XCR1 on cross-presenting dendritic cells determines cooperation with CD8+ T cells. Immunity 2009;31:823–33.
    1. Vonderheide RH, Glennie MJ. Agonistic CD40 antibodies and cancer therapy. Clin Cancer Res 2013;19:1035–43.
    1. Le DT, Jaffee EM. Regulatory T-cell modulation using cyclophosphamide in vaccine approaches: a current perspective. Cancer Res 2012;72:3439–44.
    1. Li S, Simoni Y, Zhuang S, Ma S, Chee J, Redwood Aet al. . Unsuccessful checkpoint blockade and vaccination therapy despite expansion of tumor neoantigen-specific CD8 T cells in the Lewis lung carcinoma (LLC) model. J Immunol 2020;204:165.13.
    1. Zhong W, Myers JS, Wang F, Wang K, Lucas J, Rosfjord Eet al. . Comparison of the molecular and cellular phenotypes of common mouse syngeneic models with human tumors. BMC Genomics 2020;21:2.
    1. Andreatta M, Corria-Osorio J, Müller S, Cubas R, Coukos G, Carmona SJ. Interpretation of T cell states from single-cell transcriptomics data using reference atlases. Nat Commun 2021;12:2965.
    1. Siddiqui I, Schaeuble K, Chennupati V, Fuertes Marraco SA, Calderon-Copete S, Pais Ferreira Det al. . Intratumoral Tcf1(+)PD1(+)CD8(+) T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity 2019;50:195–211.
    1. Miller BC, Sen DR, Al Abosy R, Bi K, Virkud YV, LaFleur MWet al. . Subsets of exhausted CD8(+) T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol 2019;20:326–36.
    1. Alfei F, Kanev K, Hofmann M, Wu M, Ghoneim HE, Roelli Pet al. . TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection. Nature 2019;571:265–9.
    1. Utzschneider DT, Charmoy M, Chennupati V, Pousse L, Ferreira DP, Calderon-Copete Set al. . T cell factor 1-expressing memory-like CD8(+) T cells sustain the immune response to chronic viral infections. Immunity 2016;45:415–27.
    1. Zilionis R, Engblom C, Pfirschke C, Savova V, Zemmour D, Saatcioglu HDet al. . Single-cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity 2019;50:1317–34.
    1. De Palma M, Lewis CE. Macrophage regulation of tumor responses to anticancer therapies. Cancer Cell 2013;23:277–86.
    1. Painter MW, Davis S, Hardy RR, Mathis D, Benoist C, Immunological Genome Project C. Transcriptomes of the B and T lineages compared by multiplatform microarray profiling. J Immunol 2011;186:3047–57.
    1. Maier B, Leader AM, Chen ST, Tung N, Chang C, LeBerichel Jet al. . A conserved dendritic-cell regulatory program limits antitumour immunity. Nature 2020;580:257–62.
    1. Zhang L, Li Z, Skrzypczynska KM, Fang Q, Zhang W, O'Brien SAet al. . Single-cell analyses inform mechanisms of myeloid-targeted therapies in colon cancer. Cell 2020;181:442–59.
    1. Hildner K, Edelson BT, Purtha WE, Diamond M, Matsushita H, Kohyama Met al. . Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells in cytotoxic T cell immunity. Science 2008;322:1097–100.
    1. Sanchez-Paulete AR, Cueto FJ, Martinez-Lopez M, Labiano S, Morales-Kastresana A, Rodriguez-Ruiz MEet al. . Cancer immunotherapy with immunomodulatory anti-CD137 and anti-PD-1 monoclonal antibodies requires BATF3-dependent dendritic cells. Cancer Discov 2016;6:71–9.
    1. Broz ML, Binnewies M, Boldajipour B, Nelson AE, Pollack JL, Erle DJet al. . Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell 2014;26:638–52.
    1. Grajales-Reyes GE, Iwata A, Albring J, Wu X, Tussiwand R, Kc Wet al. . Batf3 maintains autoactivation of Irf8 for commitment of a CD8alpha(+) conventional DC clonogenic progenitor. Nat Immunol 2015;16:708–17.
    1. Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature 2015;523:231–5.
    1. Binnewies M, Mujal AM, Pollack JL, Combes AJ, Hardison EA, Barry KCet al. . Unleashing type-2 dendritic cells to drive protective antitumor CD4(+) T cell immunity. Cell 2019;177:556–71.
    1. Antonczyk A, Krist B, Sajek M, Michalska A, Piaszyk-Borychowska A, Plens-Galaska Met al. . Direct inhibition of IRF-dependent transcriptional regulatory mechanisms associated with disease. Front Immunol 2019;10:1176.
    1. Diefenbach A, Tomasello E, Lucas M, Jamieson AM, Hsia JK, Vivier Eet al. . Selective associations with signaling proteins determine stimulatory versus costimulatory activity of NKG2D. Nat Immunol 2002;3:1142–9.
    1. Maasho K, Opoku-Anane J, Marusina AI, Coligan JE, Borrego F. NKG2D is a costimulatory receptor for human naive CD8+ T cells. J Immunol 2005;174:4480–4.
    1. Hellmann MD, Paz-Ares L, Bernabe Caro R, Zurawski B, Kim SW, Carcereny Costa Eet al. . Nivolumab plus ipilimumab in advanced non-small-cell lung cancer. N Engl J Med 2019;381:2020–31.
    1. Zhao X, Suryawanshi S, Hruska M, Feng Y, Wang X, Shen Jet al. . Assessment of nivolumab benefit-risk profile of a 240-mg flat dose relative to a 3-mg/kg dosing regimen in patients with advanced tumors. Ann Oncol 2017;28:2002–8.
    1. Motz GT, Santoro SP, Wang LP, Garrabrant T, Lastra RR, Hagemann ISet al. . Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nat Med 2014;20:607–15.
    1. Elicin O, Callaway S, Prior JO, Bourhis J, Ozsahin M, Herrera FG. [(18)F]FDG-PET standard uptake value as a metabolic predictor of bone marrow response to radiation: impact on acute and late hematological toxicity in cervical cancer patients treated with chemoradiation therapy. Int J Radiat Oncol Biol Phys 2014;90:1099–107.
    1. Seymour L, Bogaerts J, Perrone A, Ford R, Schwartz LH, Mandrekar Set al. . iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol 2017;18:e143–e52.
    1. Zollinger DR, Lingle SE, Sorg K, Beechem JM, Merritt CR. GeoMx RNA assay: high multiplex, digital, spatial analysis of RNA in FFPE tissue. Methods Mol Biol 2020;2148:331–45.
    1. Bindea G, Mlecnik B, Tosolini M, Kirilovsky A, Waldner M, Obenauf ACet al. . Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity 2013;39:782–95.
    1. Fairfax BP, Taylor CA, Watson RA, Nassiri I, Danielli S, Fang Het al. . Peripheral CD8(+) T cell characteristics associated with durable responses to immune checkpoint blockade in patients with metastatic melanoma. Nat Med 2020;26:193–9.
    1. Lutsiak ME, Semnani RT, De Pascalis R, Kashmiri SV, Schlom J, Sabzevari H. Inhibition of CD4(+)25+ T regulatory cell function implicated in enhanced immune response by low-dose cyclophosphamide. Blood 2005;105:2862–8.
    1. Moynihan KD, Opel CF, Szeto GL, Tzeng A, Zhu EF, Engreitz JMet al. . Eradication of large established tumors in mice by combination immunotherapy that engages innate and adaptive immune responses. Nat Med 2016;22:1402–10.
    1. Brown DM, Lee S, Garcia-Hernandez Mde L, Swain SL. Multifunctional CD4 cells expressing gamma interferon and perforin mediate protection against lethal influenza virus infection. J Virol 2012;86:6792–803.
    1. Crawford A, Angelosanto JM, Kao C, Doering TA, Odorizzi PM, Barnett BEet al. . Molecular and transcriptional basis of CD4(+) T cell dysfunction during chronic infection. Immunity 2014;40:289–302.
    1. Peeters LM, Vanheusden M, Somers V, Van Wijmeersch B, Stinissen P, Broux Bet al. . Cytotoxic CD4+ T cells drive multiple sclerosis progression. Front Immunol 2017;8:1160.
    1. Quezada SA, Simpson TR, Peggs KS, Merghoub T, Vider J, Fan Xet al. . Tumor-reactive CD4(+) T cells develop cytotoxic activity and eradicate large established melanoma after transfer into lymphopenic hosts. J Exp Med 2010;207:637–50.
    1. Xie Y, Akpinarli A, Maris C, Hipkiss EL, Lane M, Kwon EKet al. . Naive tumor-specific CD4(+) T cells differentiated in vivo eradicate established melanoma. J Exp Med 2010;207:651–67.
    1. Kitano S, Tsuji T, Liu C, Hirschhorn-Cymerman D, Kyi C, Mu Zet al. . Enhancement of tumor-reactive cytotoxic CD4+ T cell responses after ipilimumab treatment in four advanced melanoma patients. Cancer Immunol Res 2013;1:235–44.
    1. Oh DY, Kwek SS, Raju SS, Li T, McCarthy E, Chow Eet al. . Intratumoral CD4(+) T cells mediate anti-tumor cytotoxicity in human bladder cancer. Cell 2020;181:1612–25.
    1. Hirschhorn-Cymerman D, Budhu S, Kitano S, Liu C, Zhao F, Zhong Het al. . Induction of tumoricidal function in CD4+ T cells is associated with concomitant memory and terminally differentiated phenotype. J Exp Med 2012;209:2113–26.
    1. Qui HZ, Hagymasi AT, Bandyopadhyay S, St Rose MC, Ramanarasimhaiah R, Menoret Aet al. . CD134 plus CD137 dual costimulation induces Eomesodermin in CD4 T cells to program cytotoxic Th1 differentiation. J Immunol 2011;187:3555–64.
    1. Serroukh Y, Gu-Trantien C, Hooshiar Kashani B, Defrance M, Vu Manh TP, Azouz Aet al. . The transcription factors Runx3 and ThPOK cross-regulate acquisition of cytotoxic function by human Th1 lymphocytes. Elife 2018;7:e30496.
    1. Oja AE, Vieira Braga FA, Remmerswaal EB, Kragten NA, Hertoghs KM, Zuo Jet al. . The transcription factor hobit identifies human cytotoxic CD4(+) T cells. Front Immunol 2017;8:325.
    1. Sledzinska A, Vila de Mucha M, Bergerhoff K, Hotblack A, Demane DF, Ghorani Eet al. . Regulatory T cells restrain interleukin-2- and Blimp-1-dependent acquisition of cytotoxic function by CD4(+) T cells. Immunity 2020;52:151–66.
    1. Scheper W, Kelderman S, Fanchi LF, Linnemann C, Bendle G, de Rooij MAJet al. . Low and variable tumor reactivity of the intratumoral TCR repertoire in human cancers. Nat Med 2019;25:89–94.
    1. van der Leun AM, Thommen DS, Schumacher TN. CD8(+) T cell states in human cancer: insights from single-cell analysis. Nat Rev Cancer 2020;20:218–32.
    1. Beltra JC, Manne S, Abdel-Hakeem MS, Kurachi M, Giles JR, Chen Zet al. . Developmental relationships of four exhausted CD8(+) T cell subsets reveals underlying transcriptional and epigenetic landscape control mechanisms. Immunity 2020;52:825–41.
    1. Kallies A, Zehn D, Utzschneider DT. Precursor exhausted T cells: key to successful immunotherapy? Nat Rev Immunol 2020;20:128–36.
    1. Kurtulus S, Madi A, Escobar G, Klapholz M, Nyman J, Christian Eet al. . Checkpoint blockade immunotherapy induces dynamic changes in PD1(−)CD8(+) tumor-infiltrating T cells. Immunity 2019;50:181–94.
    1. Khan O, Giles JR, McDonald S, Manne S, Ngiow SF, Patel KPet al. . TOX transcriptionally and epigenetically programs CD8(+) T cell exhaustion. Nature 2019;571:211–8.
    1. Scott AC, Dundar F, Zumbo P, Chandran SS, Klebanoff CA, Shakiba Met al. . TOX is a critical regulator of tumour-specific T cell differentiation. Nature 2019;571:270–4.
    1. Markiewicz MA, Carayannopoulos LN, Naidenko OV, Matsui K, Burack WR, Wise ELet al. . Costimulation through NKG2D enhances murine CD8+ CTL function: similarities and differences between NKG2D and CD28 costimulation. J Immunol 2005;175:2825–33.
    1. Groh V, Smythe K, Dai Z, Spies T. Fas-ligand-mediated paracrine T cell regulation by the receptor NKG2D in tumor immunity. Nat Immunol 2006;7:755–62.
    1. Groh V, Bruhl A, El-Gabalawy H, Nelson JL, Spies T. Stimulation of T cell autoreactivity by anomalous expression of NKG2D and its MIC ligands in rheumatoid arthritis. Proc Natl Acad Sci U S A 2003;100:9452–7.
    1. Garcia-Chagollan M, Jave-Suarez LF, Haramati J, Bueno-Topete MR, Aguilar-Lemarroy A, Estrada-Chavez Cet al. . An approach to the immunophenotypic features of circulating CD4(+)NKG2D(+) T cells in invasive cervical carcinoma. J Biomed Sci 2015;22:91.
    1. Lazarova M, Steinle A. Impairment of NKG2D-mediated tumor immunity by TGF-beta. Front Immunol 2019;10:2689.
    1. Groh V, Wu J, Yee C, Spies T. Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation. Nature 2002;419:734–8.
    1. Salih HR, Rammensee HG, Steinle A. Cutting edge: down-regulation of MICA on human tumors by proteolytic shedding. J Immunol 2002;169:4098–102.
    1. Paczulla AM, Rothfelder K, Raffel S, Konantz M, Steinbacher J, Wang Het al. . Absence of NKG2D ligands defines leukaemia stem cells and mediates their immune evasion. Nature 2019;572:254–9.
    1. Zhang J, Larrocha PS, Zhang B, Wainwright D, Dhar P, Wu JD. Antibody targeting tumor-derived soluble NKG2D ligand sMIC provides dual co-stimulation of CD8 T cells and enables sMIC(+) tumors respond to PD1/PD-L1 blockade therapy. J Immunother Cancer 2019;7:223.
    1. Ruocco MG, Pilones KA, Kawashima N, Cammer M, Huang J, Babb JSet al. . Suppressing T cell motility induced by anti-CTLA4 monotherapy improves antitumor effects. J Clin Invest 2012;122:3718–30.
    1. Stojanovic A, Correia MP, Cerwenka A. The NKG2D/NKG2DL axis in the crosstalk between lymphoid and myeloid cells in health and disease. Front Immunol 2018;9:827.
    1. Ma HS, Poudel B, Torres ER, Sidhom JW, Robinson TM, Christmas Bet al. . A CD40 agonist and PD1 antagonist antibody reprogram the microenvironment of nonimmunogenic tumors to allow T-cell-mediated anticancer activity. Cancer Immunol Res 2019;7:428–42.
    1. Beatty GL, Chiorean EG, Fishman MP, Saboury B, Teitelbaum UR, Sun Wet al. . CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science 2011;331:1612–6.
    1. Vonderheide RH. CD40 agonist antibodies in cancer immunotherapy. Annu Rev Med 2020;71:47–58.
    1. Antonarakis ES, Piulats JM, Gross-Goupil M, Goh J, Ojamaa K, Hoimes CJet al. . Pembrolizumab for treatment-refractory metastatic castration-resistant prostate cancer: multicohort, open-label phase II KEYNOTE-199 study. J Clin Oncol 2020;38:395–405.
    1. Kandalaft LE, Odunsi K, Coukos G. Immunotherapy in ovarian cancer: are we there yet? J Clin Oncol 2019;37:2460–71.
    1. Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring ADet al. . PD1 blockade in tumors with mismatch-repair deficiency. N Engl J Med 2015;372:2509–20.
    1. Papadopoulos KP, Johnson ML, Lockhart AC, Moore K, Falchook GS, Formenti SCet al. . First-in-human study of cemiplimab alone or in combination with radiotherapy and/or low-dose cyclophosphamide in patients with advanced malignancies. Clin Cancer Res 2020;26:1025–33.
    1. Heylmann D, Bauer M, Becker H, van Gool S, Bacher N, Steinbrink Ket al. . Human CD4+CD25+ regulatory T cells are sensitive to low dose cyclophosphamide: implications for the immune response. PLoS One 2013;8:e83384.
    1. Scurr M, Pembroke T, Bloom A, Roberts D, Thomson A, Smart Ket al. . Low-dose cyclophosphamide induces antitumor T-cell responses, which associate with survival in metastatic colorectal cancer. Clin Cancer Res 2017;23:6771–80.
    1. Liakou CI, Kamat A, Tang DN, Chen H, Sun J, Troncoso Pet al. . CTLA4 blockade increases IFNgamma-producing CD4+ICOShi cells to shift the ratio of effector to regulatory T cells in cancer patients. Proc Natl Acad Sci U S A 2008;105:14987–92.
    1. Wei SC, Anang NAS, Sharma R, Andrews MC, Reuben A, Levine JHet al. . Combination anti-CTLA4 plus anti-PD-1 checkpoint blockade utilizes cellular mechanisms partially distinct from monotherapies. Proc Natl Acad Sci U S A 2019;116:22699–709.
    1. Pavlick AC, Sunandana C, Stein C, Madden K, Kannan R, Escano Cet al. . Phase II study of low-dose cyclophosphamide and ipilimumab in metastatic melanoma. J Clin Oncol 2014;32:e20025.
    1. Sinha P, Clements VK, Fulton AM, Ostrand-Rosenberg S. Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells. Cancer Res 2007;67:4507–13.
    1. Baratelli F, Lin Y, Zhu L, Yang SC, Heuze-Vourc'h N, Zeng Get al. . Prostaglandin E2 induces FOXP3 gene expression and T regulatory cell function in human CD4+ T cells. J Immunol 2005;175:1483–90.
    1. Lanitis E, Rota G, Kosti P, Ronet C, Spill A, Seijo Bet al. . Optimized gene engineering of murine CAR-T cells reveals the beneficial effects of IL-15 coexpression. J Exp Med 2021;218:e20192203.
    1. Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini JLet al. . Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med 2007;13:54–61.
    1. Tabula Muris Consortium. Overall coordination, Logistical coordination, Organ collection and processing, Library preparation and sequencing, Computational data analysis, et al.Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature 2018;562:367–72.
    1. Newman AM, Steen CB, Liu CL, Gentles AJ, Chaudhuri AA, Scherer Fet al. . Determining cell type abundance and expression from bulk tissues with digital cytometry. Nat Biotechnol 2019;37:773–82.
    1. Roederer M, Nozzi JL, Nason MC. SPICE: exploration and analysis of post-cytometric complex multivariate datasets. Cytometry A 2011;79:167–74.
    1. Festing MF. On determining sample size in experiments involving laboratory animals. Lab Anim 2018;52:341–50.
    1. Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM IIIet al. . Comprehensive integration of single-cell data. Cell 2019;177:1888–902.
    1. Borcherding N, Bormann NL, Kraus G. scRepertoire: an R-based toolkit for single-cell immune receptor analysis. F1000Res 2020;9:47.
    1. Waltman L, van Eck NJ. A smart local moving algorithm for large-scale modularity-based community detection. Eur Phys J B 2013;86:471.
    1. Aran D, Looney AP, Liu L, Wu E, Fong V, Hsu Aet al. . Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat Immunol 2019;20:163–72.
    1. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha Set al. . STAR: ultrafast universal RNA-seq aligner. Bioinformatics 2013;29:15–21.
    1. Anders S, Pyl PT, Huber W. HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics 2015;31:166–9.

Source: PubMed

3
Předplatit