Peripheral gene signatures reveal distinct cancer patient immunotypes with therapeutic implications for autologous DC-based vaccines

Michal Hensler, Jana Rakova, Lenka Kasikova, Tereza Lanickova, Josef Pasulka, Peter Holicek, Marek Hraska, Tereza Hrnciarova, Pavla Kadlecova, Andreu Schoenenberger, Klara Sochorova, Daniela Rozkova, Ludek Sojka, Jana Drozenova, Jan Laco, Rudolf Horvath, Michal Podrazil, Guo Hongyan, Tomas Brtnicky, Michal J Halaska, Lukas Rob, Ales Ryska, An Coosemans, Ignace Vergote, Abhishek D Garg, David Cibula, Jirina Bartunkova, Radek Spisek, Jitka Fucikova, Michal Hensler, Jana Rakova, Lenka Kasikova, Tereza Lanickova, Josef Pasulka, Peter Holicek, Marek Hraska, Tereza Hrnciarova, Pavla Kadlecova, Andreu Schoenenberger, Klara Sochorova, Daniela Rozkova, Ludek Sojka, Jana Drozenova, Jan Laco, Rudolf Horvath, Michal Podrazil, Guo Hongyan, Tomas Brtnicky, Michal J Halaska, Lukas Rob, Ales Ryska, An Coosemans, Ignace Vergote, Abhishek D Garg, David Cibula, Jirina Bartunkova, Radek Spisek, Jitka Fucikova

Abstract

Dendritic cells (DCs) have received considerable attention as potential targets for the development of novel cancer immunotherapies. However, the clinical efficacy of DC-based vaccines remains suboptimal, largely reflecting local and systemic immunosuppression at baseline. An autologous DC-based vaccine (DCVAC) has recently been shown to improve progression-free survival and overall survival in randomized clinical trials enrolling patients with lung cancer (SLU01, NCT02470468) or ovarian carcinoma (SOV01, NCT02107937), but not metastatic castration-resistant prostate cancer (SP005, NCT02111577), despite a good safety profile across all cohorts. We performed biomolecular and cytofluorometric analyses on peripheral blood samples collected prior to immunotherapy from 1000 patients enrolled in these trials, with the objective of identifying immunological biomarkers that may improve the clinical management of DCVAC-treated patients. Gene signatures reflecting adaptive immunity and T cell activation were associated with favorable disease outcomes and responses to DCVAC in patients with prostate and lung cancer, but not ovarian carcinoma. By contrast, the clinical benefits of DCVAC were more pronounced among patients with ovarian carcinoma exhibiting reduced expression of T cell-associated genes, especially those linked to TH2-like signature and immunosuppressive regulatory T (TREG) cells. Clinical responses to DCVAC were accompanied by signs of antitumor immunity in the peripheral blood. Our findings suggest that circulating signatures of antitumor immunity may provide a useful tool for monitoring the potency of autologous DC-based immunotherapy.

Keywords: Cancer immunotherapy; anti-PD-1; circulating biomarkers; epithelial ovarian carcinoma; metastatic castrate-resistant prostate cancer; non-small cell lung carcinoma.

Conflict of interest statement

IV declares consulting for AstraZeneca, Clovis Oncology Inc., Carrick Therapeutics, Deciphera Pharmaceuticals, Elevar Therapeutics, F. Hoffmann-La Roche Ltd, Genmab, GSK, Immunogen Inc., Jazzpharma, Mersana, Millennium Pharmaceuticals, MSD, Novocure, Octimet Oncology NV, Oncoinvent AS, Sotio a.s., Verastem Oncology, Zentalis; contracted research for: Oncoinvent AS, Genmab; and research funding from Amgen and Roche. RS and JB are minority shareholders of Sotio. ADG received fees for consultancy, lectures or services from Boehringer Ingelheim (Germany), Miltenyi Biotec (Germany), Isoplexis (USA) and Novigenix (Switzerland). AR declares advisory services and invited lectures for Amgen, AstraZeneca, BMS, Eli-Lilly, Janssen-Cilag, MSD, and Roche. AC is a contracted researcher for Oncoinvent AS and Novocure and a consultant for Sotio Biotech a.s. MH, JR, LK, TL, JF, PH, MH, TH, PK, KS, DR, LS, JB, RS, and JF are employees of Sotio a.s. The other authors declare no conflicts of interest.

© 2022 Sotio Biotech. Published with license by Taylor & Francis Group, LLC.

Figures

Figure 1.
Figure 1.
High expression of CD8A in peripheral blood is correlated with favorable prognosis and response to DCVAC in mCRPC patients in SP005. (a) Unsupervised hierarchical clustering of 804 mCRPC patients in SP005 based on the expression of 93 genes classified into clusters related to B cells, cytotoxicity, DCs, immune populations, immunosuppression, NK cells, T cell activation, and TH1 and TH2 signatures. (b, c) OS of 260 patients from the SOC arm (b) and 544 patients from the DCVAC arm (c) following stratification by unsupervised hierarchical clustering into low and high inflammatory clusters. (d, e) Direct comparison of OS of SOC and DCVAC patients following stratification by unsupervised hierarchical clustering into low (d) and high inflammatory clusters. (f) OS of 804 mCRPC patients stratified by the median CD8A expression and study arm. Survival curves were estimated using the Kaplan–Meier method and differences between groups were evaluated using the log-rank test. The numbers of patients at risk and p values are reported.
Figure 2.
Figure 2.
High expression gene signatures associated with B cells, CD8A, cytotoxicity, and DCs is correlated with favorable prognosis and response to DCVAC in NSCLC patients in SLU01. (a) Unsupervised hierarchical clustering of 103 NSCLC patients in SLU01 based on the expression of 93 genes classified into clusters related to B cells, cytotoxicity, DCs, immune populations, immunosuppression, NK cells, T cell activation, and TH1 and TH2 signatures. (b, c) OS of 35 patients from the SOC arm (b) and 68 patients from the DCVAC arm (c) following stratification by unsupervised hierarchical clustering into low and high inflammatory clusters. (d, e) Direct comparison of OS of SOC and DCVAC patients following stratification by unsupervised hierarchical clustering into low (d) and high inflammatory clusters (e). (f) OS of 103 NSCLC patients stratified by the median expression of genes associated with B cell signature, CD8A expression, cytotoxicity, and DCs, and study arm. Survival curves were estimated using the Kaplan–Meier method, and differences between groups were evaluated using the log-rank test. The numbers of patients at risk and p values are reported.
Figure 3.
Figure 3.
Low expression of genes associated with immunosuppression and TH2 signature is correlated with an improved response to DCVAC in EOC patients in SOV01. (a) Unsupervised hierarchical clustering of 93 EOC patients in SOV01 based on the expression of 93 genes classified into clusters related to B cells, cytotoxicity, DCs, immune populations, immunosuppression, NK cells, T cell activation, and TH1 and TH2 signatures. (b, c) PFS of 28 patients from the SOC arm (b) and 65 patients from the DCVAC arm (c) following stratification by unsupervised hierarchical clustering into low and high inflammatory clusters. (d, e) Direct comparison of PFS of SOC and DCVAC patients following stratification by unsupervised hierarchical clustering into low (d) and high inflammatory clusters (e). (f) PFS of 93 EOC patients upon stratification by the median expression of genes associated with B cell signature, CD3E, immunosuppression, and TH2 signature, and study arm. Survival curves were estimated using the Kaplan–Meier method, and differences between groups were evaluated using the log-rank test. The numbers of patients at risk and p values are reported.
Figure 4.
Figure 4.
High frequency of regulatory T cells in peripheral blood of EOC patients is associated with poor response to DCVAC therapy. (a) Heat map and (b) relative expression levels of the differentially expressed genes (DEGs) ARG1, FOXP3, IL6, IL13, PDCD1, TGFB1, TIGIT and TNFA in pre-treatment peripheral blood samples among mCRPC, NSCLC, and EOC patients in SP005, SLU01, and SOV01. (c) Relative expression levels of immunostimulatory (CD8A, GNLY, GZMA, GZMB, IFNG, IL12A, PRF1, TBX21) and immunosuppressive (FOXP3, HAVCR2, IDO1, IL10, LAG3, PDCD1, TGFB1, TIGIT) gene signatures in mCRPC, NSCLC and EOC patients in SP005, SLU01, and SOV01. (d, e) OS of 804 mCRPC (d) and 103 NSCLC (e) patients following stratification by the median expression of the immunostimulatory-like gene signature and study arm. (f) PFS of 93 EOC patients following stratification by the median expression of the immunosuppressive-like gene signature and study arm. Survival curves were estimated using the Kaplan–Meier method, and differences between groups were evaluated using the log-rank test. The numbers of patients at risk and p values are reported. (g) Representative dot plots for CD4+CD25+FoxP3+ regulatory T cells in low and high EOC patients in SOV01. (h) PFS of EOC patients treated with SOC or DCVAC stratified by the median percentage of CD4+CD25+FoxP3+ regulatory T cells in peripheral blood. Survival curves were estimated using the Kaplan–Meier method, and differences between groups were evaluated using the log-rank test. (i) Percentage of CD8+ T cells in peripheral blood of SOC FoxP3Lo, SOC FoxP3Hi, DCVAC FoxP3Lo and DCVAC FoxP3Hi patients prior and post DCVAC therapy. Statistical significance was calculated by the Wilcoxon test. p values are indicated.

References

    1. Galluzzi L, Chan TA, Kroemer G, Wolchok JD, Lopez-Soto A.. The hallmarks of successful anticancer immunotherapy. Sci Transl Med. 2018;10(459). doi:10.1126/scitranslmed.aat7807.
    1. Garon EB, Rizvi NA, Hui R, Leighl N, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 2015;372(21):2018–14. doi:10.1056/NEJMoa1501824.
    1. Ansell SM, Lesokhin AM, Borrello I, Halwani A, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med. 2015;372(4):311–319. doi:10.1056/NEJMoa1411087.
    1. Huang AC, Postow MA, Orlowski RJ, Mick R, et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature. 2017;545(7652):60–65. doi:10.1038/nature22079.
    1. Hellmann MD, Callahan MK, Awad MM, Calvo E, et al. Tumor Mutational Burden and Efficacy of Nivolumab Monotherapy and in Combination with Ipilimumab in Small-Cell Lung Cancer. Cancer Cell. 2018;33(5):853–61 e4. doi:10.1016/j.ccell.2018.04.001.
    1. Bassez A, Vos H, Van Dyck L, Floris G, et al. A single-cell map of intratumoral changes during anti-PD1 treatment of patients with breast cancer. Nat Med. 2021;27(5):820–832. doi:10.1038/s41591-021-01323-8.
    1. Wculek SK, Cueto FJ, Mujal AM, Melero I, Krummel MF, Sancho D. Dendritic cells in cancer immunology and immunotherapy. Nat Rev Immunol. 2020;20(1):7–24. doi:10.1038/s41577-019-0210-z.
    1. Garg AD, Coulie PG, Van den Eynde BJ, Agostinis P. Integrating Next-Generation Dendritic Cell Vaccines into the Current Cancer Immunotherapy Landscape. Trends Immunol. 2017;38(8):577–593. doi:10.1016/j.it.2017.05.006.
    1. Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discov. 2019;18(3):197–218. doi:10.1038/s41573-018-0007-y.
    1. Vitale I, Shema E, Loi S, Galluzzi L. Intratumoral heterogeneity in cancer progression and response to immunotherapy. Nat Med. 2021;27(2):212–224. doi:10.1038/s41591-021-01233-9.
    1. Sprooten J, Vankerckhoven A, Vanmeerbeek I, Borras DM, et al. Peripherally-driven myeloid NFkB and IFN/ISG responses predict malignancy risk, survival, and immunotherapy regime in ovarian cancer. J Immunother Cancer. 2021;9(11):e003609. doi:10.1136/jitc-2021-003609.
    1. Nixon AB, Schalper KA, Jacobs I, Potluri S, Wang IM, Fleener C. Peripheral immune-based biomarkers in cancer immunotherapy: can we realize their predictive potential? J Immunother Cancer. 2019;7(1):325. doi:10.1186/s40425-019-0799-2.
    1. Sprooten J, Coosemans A, Garg AD. A first-in-class, non-invasive, immunodynamic biomarker approach for precision immuno-oncology. Oncoimmunology. 2022;11(1):2024692. doi:10.1080/2162402X.2021.2024692.
    1. Havel JJ, Chowell D, Chan TA. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat Rev Cancer. 2019;19(3):133–150. doi:10.1038/s41568-019-0116-x.
    1. Scher HI, Graf RP, Schreiber NA, Jayaram A, et al. Assessment of the validity of nuclear-localized androgen receptor splice variant 7 in circulating tumor cells as a predictive biomarker for castration-resistant prostate cancer. JAMA Oncol. 2018;4(9):1179–1186. doi:10.1001/jamaoncol.2018.1621.
    1. Wang Z, Duan J, Cai S, Han M, et al. Assessment of blood tumor mutational burden as a potential biomarker for immunotherapy in patients with non-small cell lung cancer with use of a next-generation sequencing cancer gene panel. JAMA Oncol. 2019;5(5):696–702. doi:10.1001/jamaoncol.2018.7098.
    1. Weiss GJ, Beck J, Braun DP, Bornemann-Kolatzki K, et al. Tumor cell-free DNA copy number instability predicts therapeutic response to immunotherapy. Clin Cancer Res. 2017;23(17):5074–5081. doi:10.1158/1078-0432.CCR-17-0231.
    1. Hong X, Sullivan RJ, Kalinich M, Kwan TT, et al. Molecular signatures of circulating melanoma cells for monitoring early response to immune checkpoint therapy. Proc Natl Acad Sci U S A. 2018;115(10):2467–2472. doi:10.1073/pnas.1719264115.
    1. Lee EY, Kulkarni RP. Circulating biomarkers predictive of tumor response to cancer immunotherapy. Expert Rev Mol Diagn. 2019;19(10):895–904. doi:10.1080/14737159.2019.1659728.
    1. Indini A, Rijavec E, Grossi F. Circulating biomarkers of response and toxicity of immunotherapy in advanced non-small cell lung cancer (NSCLC): a comprehensive review. Cancers (Basel). 2021;13(8):1794. doi:10.3390/cancers13081794.
    1. Zemanova M, Cernovska M, Havel L, Bartek T, et al. Autologous dendritic cell-based immunotherapy (DCVAC/LuCa) and carboplatin/paclitaxel in advanced non-small cell lung cancer: a randomized, open-label, phase I/II trial. Cancer Treat Res Commun. 2021;28:100427. doi:10.1016/j.ctarc.2021.100427.
    1. Rob L, Cibula D, Knapp P, Mallmann P, et al. Safety and efficacy of dendritic cell-based immunotherapy DCVAC/OvCa added to first-line chemotherapy (carboplatin plus paclitaxel) for epithelial ovarian cancer: a phase 2, open-label, multicenter, randomized trial. J Immunother Cancer. 2022;10(1):e003190. doi:10.1136/jitc-2021-003190.
    1. Fucikova J, Hensler M, Kasikova L, Lanickova T, et al. An autologous dendritic cell vaccine promotes anticancer immunity in ovarian cancer patients with low mutational burden and cold tumors. Clin Cancer Res. pp.OF1–OF13. 2022. doi:10.1158/1078-0432.CCR-21-4413
    1. Vogelzang NJ, Beer TM, Gerritsen W, Oudard S, et al. Efficacy and safety of autologous dendritic cell-based immunotherapy, docetaxel, and prednisone vs placebo in patients with metastatic castration-resistant prostate cancer: the VIABLE Phase 3 randomized clinical trial. JAMA Oncol. 2022;8:546. doi:10.1001/jamaoncol.2021.7298.
    1. Fucikova J, Moserova I, Truxova I, Hermanova I, et al. High hydrostatic pressure induces immunogenic cell death in human tumor cells. Int J Cancer. 2014;135(5):1165–1177. doi:10.1002/ijc.28766.
    1. Fucikova J, Rozkova D, Ulcova H, Budinsky V, et al. Poly I: c-activated dendritic cells that were generated in CellGro for use in cancer immunotherapy trials. J Transl Med. 2011;9:223. doi:10.1186/1479-5876-9-223.
    1. Gu Z, Eils R, Schlesner M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics. 2016;32(18):2847–2849. doi:10.1093/bioinformatics/btw313.
    1. Wu T, Hu E, Xu S, Chen M, et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation (N Y). 2021;2(3):100141.
    1. Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol. 2020;20(11):651–668. doi:10.1038/s41577-020-0306-5.
    1. Dall’Olio FG, Marabelle A, Caramella C, Garcia C, et al. Tumour burden and efficacy of immune-checkpoint inhibitors. Nat Rev Clin Oncol. 2022;19(2):75–90. doi:10.1038/s41571-021-00564-3.
    1. Rosenberg JE, Hoffman-Censits J, Powles T, van der Heijden MS, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet. 2016;387(10031):1909–1920. doi:10.1016/S0140-6736(16)00561-4.
    1. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366(26):2443–2454. doi:10.1056/NEJMoa1200690.
    1. Monk BJ, Colombo N, Oza AM, Fujiwara K, et al. Chemotherapy with or without avelumab followed by avelumab maintenance versus chemotherapy alone in patients with previously untreated epithelial ovarian cancer (JAVELIN Ovarian 100): an open-label, randomised, phase 3 trial. Lancet Oncol. 2021;22(9):1275–1289. doi:10.1016/S1470-2045(21)00342-9.
    1. Beer TM, Kwon ED, Drake CG, Fizazi K, et al. Randomized, double-blind, phase iii trial of ipilimumab versus placebo in asymptomatic or minimally symptomatic patients with metastatic chemotherapy-naive castration-resistant prostate cancer. J Clin Oncol. 2017;35(1):40–47. doi:10.1200/JCO.2016.69.1584.
    1. Hollingsworth RE, Jansen K. Turning the corner on therapeutic cancer vaccines. NPJ Vaccines. 2019;4:7. doi:10.1038/s41541-019-0103-y.
    1. Fucikova J, Palova-Jelinkova L, Bartunkova J, Spisek R. Induction of tolerance and immunity by dendritic cells: mechanisms and clinical applications. Front Immunol. 2019;10:2393. doi:10.3389/fimmu.2019.02393.
    1. Sprooten J, Ceusters J, Coosemans A, Agostinis P, et al. Trial watch: dendritic cell vaccination for cancer immunotherapy. Oncoimmunology. 2019;8(11):e1638212. doi:10.1080/2162402X.2019.1638212.
    1. Vacchelli E, Vitale I, Eggermont A, Fridman WH, et al. Trial watch: dendritic cell-based interventions for cancer therapy. Oncoimmunology. 2013;2(10):e25771. doi:10.4161/onci.25771.
    1. Pol J, Bloy N, Buque A, Eggermont A, et al. Trial watch: peptide-based anticancer vaccines. Oncoimmunology. 2015;4(4):e974411. doi:10.4161/2162402X.2014.974411.
    1. Salmon H, Idoyaga J, Rahman A, Leboeuf M, et al. Expansion and activation of CD103(+) Dendritic cell progenitors at the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. Immunity. 2016;44(4):924–938. doi:10.1016/j.immuni.2016.03.012.
    1. Merad M, Sathe P, Helft J, Miller J, Mortha A. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu Rev Immunol. 2013;31:563–604. doi:10.1146/annurev-immunol-020711-074950.
    1. Garg AD, Vandenberk L, Koks C, Verschuere T, et al. Dendritic cell vaccines based on immunogenic cell death elicit danger signals and T cell-driven rejection of high-grade glioma. Sci Transl Med. 2016;8(328):328ra27. doi:10.1126/scitranslmed.aae0105.
    1. Sarivalasis A, Boudousquie C, Balint K, Stevenson BJ, et al. A Phase I/II trial comparing autologous dendritic cell vaccine pulsed either with personalized peptides (PEP-DC) or with tumor lysate (OC-DC) in patients with advanced high-grade ovarian serous carcinoma. J Transl Med. 2019;17(1):391. doi:10.1186/s12967-019-02133-w.
    1. Charles J, Chaperot L, Hannani D, Bruder Costa J, et al. An innovative plasmacytoid dendritic cell line-based cancer vaccine primes and expands antitumor T-cells in melanoma patients in a first-in-human trial. Oncoimmunology. 2020;9(1):1738812. doi:10.1080/2162402X.2020.1738812.
    1. Cibula D, Rob L, Mallmann P, Knapp P, et al. Dendritic cell-based immunotherapy (DCVAC/OvCa) combined with second-line chemotherapy in platinum-sensitive ovarian cancer (SOV02): a randomized, open-label, phase 2 trial. Gynecol Oncol. 2021;162:652–660. doi:10.1016/j.ygyno.2021.07.003.
    1. Coosemans A, Decoene J, Baert T, Laenen A, et al. Immunosuppressive parameters in serum of ovarian cancer patients change during the disease course. Oncoimmunology. 2016;5(4):e1111505. doi:10.1080/2162402X.2015.1111505.
    1. De Bruyn C, Ceusters J, Landolfo C, Baert T, et al. Neo-adjuvant chemotherapy reduces, and surgery increases immunosuppression in first-line treatment for ovarian cancer. Cancers (Basel). 2021;13(23):5899. doi:10.3390/cancers13235899.
    1. Liang B, Workman C, Lee J, Chew C, et al. Regulatory T cells inhibit dendritic cells by lymphocyte activation gene-3 engagement of MHC class II. J Immunol. 2008;180(9):5916–5926. doi:10.4049/jimmunol.180.9.5916.
    1. Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T, et al. CTLA-4 control over Foxp3+ regulatory T cell function. Science. 2008;322(5899):271–275. doi:10.1126/science.1160062.
    1. Deaglio S, Dwyer KM, Gao W, Friedman D, et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med. 2007;204(6):1257–1265. doi:10.1084/jem.20062512.
    1. Campbell DJ, Koch MA. Phenotypical and functional specialization of FOXP3+ regulatory T cells. Nat Rev Immunol. 2011;11(2):119–130. doi:10.1038/nri2916.
    1. Rochman Y, Spolski R, Leonard WJ. New insights into the regulation of T cells by gamma(c) family cytokines. Nat Rev Immunol. 2009;9(7):480–490. doi:10.1038/nri2580.
    1. Kandalaft LE, Odunsi K, Coukos G. Immunotherapy in ovarian cancer: are we there yet? J Clin Oncol. 2019;37(27):2460–2471. doi:10.1200/JCO.19.00508.
    1. Fucikova J, Palova-Jelinkova L, Klapp V, Holicek P, et al. Immunological control of ovarian carcinoma by chemotherapy and targeted anticancer agents. Trends Cancer. 2022;8:426–444. doi:10.1016/j.trecan.2022.01.010.
    1. Petroni G, Buque A, Zitvogel L, Kroemer G, Galluzzi L. Immunomodulation by targeted anticancer agents. Cancer Cell. 2021;39(3):310–345. doi:10.1016/j.ccell.2020.11.009.
    1. Stewart RA, Pilie PG, Yap TA. Development of PARP and immune-checkpoint inhibitor combinations. Cancer Res. 2018;78(24):6717–6725. doi:10.1158/0008-5472.CAN-18-2652.
    1. Bol KF, Schreibelt G, Gerritsen WR, de Vries IJ, Figdor CG. Dendritic cell-based immunotherapy: state of the art and beyond. Clin Cancer Res. 2016;22(8):1897–1906. doi:10.1158/1078-0432.CCR-15-1399.
    1. Kroemer G, Galassi C, Zitvogel L, Galluzzi L. Immunogenic cell stress and death. Nat Immunol. 2022;23(4):487–500. doi:10.1038/s41590-022-01132-2.
    1. Coukos G, Tanyi J, Kandalaft LE. Opportunities in immunotherapy of ovarian cancer. Ann Oncol. 2016;27 Suppl 1:i11–i5. doi:10.1093/annonc/mdw084.

Source: PubMed

3
S'abonner