Differential combination immunotherapy requirements for inflamed (warm) tumors versus T cell excluded (cool) tumors: engage, expand, enable, and evolve

Kellsye P Fabian, Michelle R Padget, Rika Fujii, Jeffrey Schlom, James W Hodge, Kellsye P Fabian, Michelle R Padget, Rika Fujii, Jeffrey Schlom, James W Hodge

Abstract

Background: Different types of tumors have varying susceptibility to immunotherapy and hence require different treatment strategies; these cover a spectrum ranging from 'hot' tumors or those with high mutational burden and immune infiltrates that are more amenable to targeting to 'cold' tumors that are more difficult to treat due to the fewer targetable mutations and checkpoint markers. We hypothesized that an effective anti-tumor response requires multiple agents that would (1) engage the immune response and generate tumor-specific effector cells; (2) expand the number and breadth of the immune effector cells; (3) enable the anti-tumor activity of these immune cells in the tumor microenvironment; and (4) evolve the tumor response to widen immune effector repertoire.

Methods: A hexatherapy combination was designed and administered to MC38-CEA (warm) and 4T1 (cool) murine tumor models. The hexatherapy regimen was composed of adenovirus-based vaccine and IL-15 (interleukin-15) superagonist (N-803) to engage the immune response; anti-OX40 and anti-4-1BB to expand effector cells; anti-PD-L1 (anti-programmed death-ligand 1) to enable anti-tumor activity; and docetaxel to promote antigen spread. Primary and metastatic tumor growth inhibition were measured. The generation of anti-tumor immune effector cells was analyzed using flow cytometry, ELISpot (enzyme-linked immunospot), and RNA analysis.

Results: The MC38-CEA and 4T1 tumor models have differential sensitivities to the combination treatments. In the 'warm' MC38-CEA, combinations with two to five agents resulted in moderate therapeutic benefit while the hexatherapy regimen outperformed all these combinations. On the other hand, the hexatherapy regimen was required in order to decrease the primary and metastatic tumor burden in the 'cool' 4T1 model. In both models, the hexatherapy regimen promoted CD4+ and CD8+ T cell proliferation and activity. Furthermore, the hexatherapy regimen induced vaccine-specific T cells and stimulated antigen cascade. The hexatherapy regimen also limited the immunosuppressive T cell and myeloid derived suppressor cell populations, and also decreased the expression of exhaustion markers in T cells in the 4T1 model.

Conclusion: The hexatherapy regimen is a strategic combination of immuno-oncology agents that can engage, expand, enable, and evolve the immune response and can provide therapeutic benefits in both MC38-CEA (warm) and 4T1 (cool) tumor models.

Trial registration: ClinicalTrials.gov NCT03493945.

Keywords: cytokines; immunomodulation; immunotherapy; translational medical research; vaccination.

Conflict of interest statement

Competing interests: None declared.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY. Published by BMJ.

Figures

Figure 1
Figure 1
MC38-CEA colorectal carcinoma has an immune-inflamed phenotype compared with 4T1 breast carcinoma. (A, B) Female C57BL/6-CEA-Tg mice (8–12 weeks old; n=5) were implanted with 3×105 MC38-CEA cells on the flank and female Balb/c mice (8–12 weeks old) were implanted with 5×104 4T1 cells on the mammary fat pad. Fourteen to fifteen days after tumor implantation, the tumors were harvested and analyzed via (A) flow cytometry and (B) immunofluorescence staining for CD8+ T cell infiltration. (C, D) RNA was isolated from three MC38-CEA and three 4T1 tumor explants harvested 28 days post-tumor implantation and the immune-related transcriptome for each tumor was analyzed using the nCounter PanCancer Immune Profiling Panel. Heatmap showing select genes with data presented as fold change values compared with housekeeping genes suite of that particular tumor sample on scale of 0 (light blue) to (C) 800 (red) and (D) 3500 (red). All genes reported are significantly different. CEA, carcinoembryonic antigen; Tg, transgenic.
Figure 2
Figure 2
The hexatherapy treatment regimen results in enhanced therapeutic effects in the ‘warm’ MC38-CEA model. (A) Female C57BL/6-CEA-Tg mice (8–12 weeks old) were inoculated s.c. on the flank with 3×105 MC38-CEA cells on day 0. Ad-CEA (1×1010 VP) was administered s.c. on days 7, 14, and 21; 1 µg N-803 s.c. on days 14 and 21; 100 µg anti-OX40 and 20 µg anti-4-1BB i.p. on days 7, 14, and 21; 200 µg anti-PD-L1 i.p. on days 14 and 21; and 500 µg docetaxel on day 21. In this hexatherapy regimen, the IO agents were grouped into four modalities: Ad-CEA+N-803, OX40+4-1BB, PD-L1, and docetaxel. (B, C) The single modality treatments were cumulatively combined and used to treat MC38-CEA tumor-bearing mice (n=6–8/group) using the schedule described above. Tumor volumes were monitored; (B) tumor growth curve and (C) mean tumor volumes on day 25 were plotted. Another set of MC38-CEA tumor-bearing mice (n=6–10/group) were treated with the hexatherapy regimen or with corresponding empty adenoviral vector and antibody isotype. Tumor volumes on day 28 were plotted (B inset). (D, E) MC38-CEA tumor-bearing female C57BL/6-CEA-Tg mice (n=8–10/group) were treated with the hexatherapy regimen or the hexatherapy regimen minus one treatment modality. Tumor volumes were monitored; (D) tumor growth curve and (E) mean tumor volumes on day 26 were plotted. (F, G) The hexatherapy regimen was administered to CD8+ T cell-depleted MC38-CEA tumor-bearing female C57BL/6 mice (n=6–10). (F) Tumor volumes on day 28 and (G) survival were monitored. The same untreated and hexatherapy-treated groups are presented in figure 2B inset and figure 2F. (H) For each treatment combination tested in the MC38-CEA model, the percentage of mice with tumor volume <300 mm3 was calculated and plotted against the number of IO agents received. Meta-analysis of three to four independent experiments is shown. Statistical test: Analysis of variance with Tukey’s post hoc test. Error bars, SEM. *p<0.05; **p<0.01; ***p<0.001; ****p<0.0001. CEA, carcinoembryonic antigen; IO, immuno-oncology; i.p., intraperitoneal; s.c., subcutaneous; Tg, transgenic; VP, viral particle.
Figure 3
Figure 3
Treatment with hexatherapy induces enhanced CD4+ and CD8+ T cell activity in the ‘warm’ MC38-CEA tumor model. In a separate experiment, MC38-CEA tumor-bearing mice (n=6–10/group) were treated as in figure 2A. Three spleens from each animal cohort were collected on day 28 (7 days after the last treatment). (A–D) Splenocytes from different treatment groups were stimulated in vitro with 1 µg/mL plate-bound CD3 antibody for 4 hours. Intracellular expression of Ki67 and IFNγ in (A, B) CD4+ T cells and (C, D) CD8+ T cells was analyzed by flow cytometry as frequency of CD4+ and CD8+ T cells, respectively. (E–I) 1×106 splenocytes were stimulated with H2-Kb-restricted peptide epitopes for (E, F) CEA, (G) gp70 (p15E), the neo-epitopes (H) JAK1 and (I) Ptgfr, and HIV-gag. Antigen-specific IFN-γ production was measured via ELISpot. HIV-gag values were subtracted from the values obtained with the other antigens to normalize the data. Statistical test: Analysis of variance with Tukey’s post hoc test. Error bars, SEM. *p<0.05; **, p<0.01; ***p<0.001; ****p<0.0001. CEA, carcinoembryonic antigen; IFN, interferon; PD-L1, programmed death-ligand 1.
Figure 4
Figure 4
The hexatherapy regimen results in enhanced therapeutic effects associated with expanded effector immune cell populations in the ‘cool’ 4T1 tumor model. (A) Female Balb/c mice (8–12 weeks old) were implanted with 5×104 4T1 cells on the mammary fat pad on day 0. Ad-Twist (1×1010 VP) was administered s.c. on days 7, 14, and 21; 1 μg N-803 s.c. on days 14 and 21; 100 µg anti-OX40 and 20 µg anti-4-1BB i.p. on days 7, 14, and 21; 200 µg anti-PD-L1 i.p. on days 14 and 21; and 500 µg docetaxel on day 21. In this hexatherapy regimen, the IO agents were grouped into four modalities: Ad-CEA+N-803, OX40+4-1BB, PD-L1, and docetaxel. (B–D) 4T1 tumor-bearing mice were treated with the hexatherapy regimen or hexatherapy regimen minus one treatment modality (n=18–20/group). (B) Primary tumor volumes were monitored and (C) mean tumor volumes on day 25 were plotted. On day 28 post-tumor implantation the lungs (n=13–15/group) were collected from the different animal treatment cohorts. (D) Single cell suspension samples of lungs were incubated in complete RPMI supplemented with 6 µM 6-thioguanine for 10–12 days, after which clonogenic metastatic cell colonies were enumerated. Another set of 4T1 tumor-bearing female Balb/c mice (n=8–10/group) were treated as described above. (E–H). On day 28, splenocytes from the different treatment groups were harvested and stimulated in vitro with 1 µg/mL plate-bound CD3 antibody overnight. Intracellular expression of Ki67 and IFN-γ in (E, F) CD4+ T cells and (G, H) CD8+ T cells was analyzed by flow cytometry as frequency of CD4+ and CD8+ T cells, respectively. (I, J) The splenocytes were also stimulated in vitro with H2-Kd-restricted peptide epitopes for (I) Twist, (J) AH1 and β-gal. Antigen-specific IFN-γ production was measured via ELISpot. β-gal values were subtracted from the values obtained with the other antigens to normalize the data. Statistical tests: Analysis of variance with Tukey’s post hoc test for group analyses. Student’s t-test for comparing two groups. Error bars, SEM. *p<0.05; **p<0.01; ***p<0.001; ****p<0.0001. CEA, carcinoembryonic antigen; IFN, interferon; IO, immuno-oncology; PD-L1, programmed death-ligand 1; s.c., subcutaneous; VP, viral particle.
Figure 5
Figure 5
Treatment with the hexatherapy regimen results in tumor-infiltrating T cells that are more proliferative and less exhausted in the ‘cool’ 4T1 tumor model. Primary tumors (n=4–5/group) from figure 4B, C were collected on day 28 post-tumor implantation (7 days after the last treatment). (A–C) Flow cytometry was performed to determine (A) the frequency of FoxP3neg CD3+CD4+ T cells in the CD45+ tumor-infiltrating immune population, (B) the frequency of Ki67+ expression in the FoxP3neg CD4+ T cells, and the (C) frequency of CTLA-4 and PD-1 expression in the CD4+ T cells. (D) Likewise, flow cytometry was performed to determine the frequency of CD3+CD8+ T cells in the CD45+ compartment. (E) Immunofluorescence staining of CD8+ T cells (green) and PECAM1+ cells (red) was performed on untreated and hexatherapy regimen-treated tumors to further elucidate T cell infiltration. (F, G) Flow cytometry was done to assess (F) the frequency of Ki67+ expression, and the (G) frequency of CTLA-4 and PD-1 expression in the CD8+ T cells. Statistical test: Analysis of variance with Tukey’s post hoc test. Error bars, SEM. *p<0.05; **p<0.01; ***p<0.001; ****p<0.0001. PD-1, programmed cell death protein-1; PECAM1+, platelet endothelial cell adhesion molecule-1 positive.
Figure 6
Figure 6
The hexatherapy treatment regimen reduces PMN-MDSC and improves effector T cell-to-Treg cell ratios in the 4T1 tumor microenvironment. (A–C) Primary tumors (n=4–5/group) from figure 4B, C were collected on day 28 post-tumor implantation (7 days after the last treatment) and were assessed through flow cytometry for frequency of (A) PMN-MDSC (CD11b+Ly6G+Ly6Clo), (B) M-MDSC (CD11b+Ly6G−Ly6Chigh), and (C) Tregs (FoxP3+CD4+ T cells) in the CD45+ population. (D, E) Flow cytometric analysis was used to determine the (D) CD4-to-Treg ratio and (E) CD8-to-Treg ratio in the tumor. Analysis of variance with Tukey’s post hoc test. Error bars, SEM. *p<0.05; **p<0.01; ***p<0.001; ****p<0.0001. M-MDSC, mononuclear myeloid derived suppressor cell; PD-L1, programmed death-ligand 1; PMN-MDSC, polymorphonuclear MDSC; Treg, regulatory T cell.
Figure 7
Figure 7
Treatment with the hexatherapy regimen results in enhanced therapeutic effects associated with expanded effector immune cell populations in the 4T1 tumor model. (A) For each treatment combination tested in the 4T1 tumor model, the percentage of mice with tumor volume 3 was calculated and plotted against the number of IO agents received. Meta-analysis of three independent experiments is shown. (B) 4T1 tumor-bearing female Balb/c mice (8–12 weeks old) were treated with hexatherapy regimen or single IO agent as described in figure 4A. Two to three tumor samples from each animal cohort were harvested on day 28 post-tumor implantation and were used to analyze the immune-related transcriptome using the nCounter PanCancer Immune Profiling Panel. Heatmap showing select genes with data presented as fold change on scale of −2 (blue) to +2 (red) relative to the gene expression in the untreated tumors. Statistical test: Student’s t-test. *p<0.05. IO, immuno-oncology; PD-1, programmed cell death protein-1; PD-L1, programmed death-ligand 1.

References

    1. Maleki Vareki S High and low mutational burden tumors versus immunologically hot and cold tumors and response to immune checkpoint inhibitors. J Immunother Cancer 2018;6:157. 10.1186/s40425-018-0479-7
    1. Havel JJ, Chowell D, Chan TA. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat Rev Cancer 2019;19:133–50. 10.1038/s41568-019-0116-x
    1. Tumeh PC, Harview CL, Yearley JH, et al. . Pd-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014;515:568–71. 10.1038/nature13954
    1. Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol 2020;20:651–68. 10.1038/s41577-020-0306-5
    1. Greiner JW, Zeytin H, Anver MR, et al. . Vaccine-based therapy directed against carcinoembryonic antigen demonstrates antitumor activity on spontaneous intestinal tumors in the absence of autoimmunity. Cancer Res 2002;62:6944.
    1. Kass E, Schlom J, Thompson J, et al. . Induction of protective host immunity to carcinoembryonic antigen (CEA), a self-antigen in CEA transgenic mice, by immunizing with a recombinant vaccinia-CEA virus. Cancer Res 1999;59:676–83.
    1. Knudson KM, Hicks KC, Alter S, et al. . Mechanisms involved in IL-15 superagonist enhancement of anti-PD-L1 therapy. J Immunother Cancer 2019;7:82. 10.1186/s40425-019-0551-y
    1. Kwilas AR, Ardiani A, Dirmeier U, et al. . A poxviral-based cancer vaccine the transcription factor twist inhibits primary tumor growth and metastases in a model of metastatic breast cancer and improves survival in a spontaneous prostate cancer model. Oncotarget 2015;6:28194–210. 10.18632/oncotarget.4442
    1. Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discov 2019;18:197–218. 10.1038/s41573-018-0007-y
    1. Gatti-Mays ME, Redman JM, Collins JM, et al. . Cancer vaccines: enhanced immunogenic modulation through therapeutic combinations. Hum Vaccin Immunother 2017;13:2561–74. 10.1080/21645515.2017.1364322
    1. Xu W, Jones M, Liu B, et al. . Efficacy and mechanism-of-action of a novel superagonist interleukin-15: interleukin-15 receptor αSu/Fc fusion complex in syngeneic murine models of multiple myeloma. Cancer Res 2013;73:3075–86. 10.1158/0008-5472.CAN-12-2357
    1. Kim PS, Kwilas AR, Xu W, et al. . Il-15 superagonist/IL-15RαSushi-Fc fusion complex (IL-15SA/IL-15RαSu-Fc; ALT-803) markedly enhances specific subpopulations of NK and memory CD8+ T cells, and mediates potent anti-tumor activity against murine breast and colon carcinomas. Oncotarget 2016;7:16130–45. 10.18632/oncotarget.7470
    1. Knudson KM, Hodge JW, Schlom J, et al. . Rationale for IL-15 superagonists in cancer immunotherapy. Expert Opin Biol Ther 2020;20:705–9. 10.1080/14712598.2020.1738379
    1. Sanmamed MF, Pastor F, Rodriguez A, et al. . Agonists of co-stimulation in cancer immunotherapy directed against CD137, OX40, GITR, CD27, CD28, and ICOS. Semin Oncol 2015;42:640–55. 10.1053/j.seminoncol.2015.05.014
    1. Marin-Acevedo JA, Dholaria B, Soyano AE, et al. . Next generation of immune checkpoint therapy in cancer: new developments and challenges. J Hematol Oncol 2018;11:39. 10.1186/s13045-018-0582-8
    1. Garnett CT, Schlom J, Hodge JW. Combination of docetaxel and recombinant vaccine enhances T-cell responses and antitumor activity: effects of docetaxel on immune enhancement. Clin Cancer Res 2008;14:3536–44. 10.1158/1078-0432.CCR-07-4025
    1. Hodge JW, Garnett CT, Farsaci B, et al. . Chemotherapy-Induced immunogenic modulation of tumor cells enhances killing by cytotoxic T lymphocytes and is distinct from immunogenic cell death. Int J Cancer 2013;133:624–36. 10.1002/ijc.28070
    1. Robbins PF, Kantor JA, Salgaller M, et al. . Transduction and expression of the human carcinoembryonic antigen gene in a murine colon carcinoma cell line. Cancer Res 1991;51:3657–62.
    1. Pulaski BA, Ostrand-Rosenberg S. Mouse 4T1 breast tumor model. Curr Protoc Immunol 2001;Chapter 20:Unit 20.2. 10.1002/0471142735.im2002s39
    1. Abdul Sater H, Marté JL, Donahue RN, et al. . Neoadjuvant PROSTVAC prior to radical prostatectomy enhances T-cell infiltration into the tumor immune microenvironment in men with prostate cancer. J Immunother Cancer 2020;8:e000655. 10.1136/jitc-2020-000655
    1. Lee KL, Benz SC, Hicks KC, et al. . Efficient tumor clearance and diversified immunity through neoepitope vaccines and combinatorial immunotherapy. Cancer Immunol Res 2019;7:1359–70. 10.1158/2326-6066.CIR-18-0620
    1. Mosely SIS, Prime JE, Sainson RCA, et al. . Rational selection of syngeneic preclinical tumor models for immunotherapeutic drug discovery. Cancer Immunol Res 2017;5:29–41. 10.1158/2326-6066.CIR-16-0114
    1. Shrimali RK, Ahmad S, Verma V, et al. . Concurrent PD-1 blockade negates the effects of OX40 agonist antibody in combination immunotherapy through inducing T-cell apoptosis. Cancer Immunol Res 2017;5:755–66. 10.1158/2326-6066.CIR-17-0292
    1. Messenheimer DJ, Jensen SM, Afentoulis ME, et al. . Timing of PD-1 blockade is critical to effective combination immunotherapy with Anti-OX40. Clin Cancer Res 2017;23:6165–77. 10.1158/1078-0432.CCR-16-2677
    1. Ardiani A, Gameiro SR, Palena C, et al. . Vaccine-mediated immunotherapy directed against a transcription factor driving the metastatic process. Cancer Res 2014;74:1945–57. 10.1158/0008-5472.CAN-13-2045
    1. Mall C, Sckisel GD, Proia DA, et al. . Repeated PD-1/PD-L1 monoclonal antibody administration induces fatal xenogeneic hypersensitivity reactions in a murine model of breast cancer. Oncoimmunology 2016;5:e1075114. 10.1080/2162402X.2015.1075114
    1. Chester C, Fritsch K, Kohrt HE. Natural killer cell immunomodulation: targeting activating, inhibitory, and co-stimulatory receptor signaling for cancer immunotherapy. Front Immunol 2015;6:601. 10.3389/fimmu.2015.00601
    1. Ahmadzadeh M, Johnson LA, Heemskerk B, et al. . Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 2009;114:1537–44. 10.1182/blood-2008-12-195792
    1. Gabrilovich DI Myeloid-Derived suppressor cells. Cancer Immunol Res 2017;5:3–8. 10.1158/2326-6066.CIR-16-0297
    1. Semeraro M, Adam J, Stoll G, et al. . The ratio of CD8+/FOXP3 T lymphocytes infiltrating breast tissues predicts the relapse of ductal carcinoma in situ. Oncoimmunology 2016;5:e1218106. 10.1080/2162402X.2016.1218106
    1. Asano Y, Kashiwagi S, Goto W, et al. . Tumour-Infiltrating CD8 to FOXP3 lymphocyte ratio in predicting treatment responses to neoadjuvant chemotherapy of aggressive breast cancer. Br J Surg 2016;103:845–54. 10.1002/bjs.10127
    1. Vaddepally RK, Kharel P, Pandey R, et al. . Review of indications of FDA-approved immune checkpoint inhibitors per NCCN guidelines with the level of evidence. Cancers 2020;12:738. 10.3390/cancers12030738
    1. Jenkins RW, Barbie DA, Flaherty KT. Mechanisms of resistance to immune checkpoint inhibitors. Br J Cancer 2018;118:9–16. 10.1038/bjc.2017.434
    1. Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med 2017;377:2500–1. 10.1056/NEJMc1713444
    1. Tang F, Zheng P. Tumor cells versus host immune cells: whose PD-L1 contributes to PD-1/PD-L1 blockade mediated cancer immunotherapy? Cell Biosci 2018;8:34. 10.1186/s13578-018-0232-4
    1. Zhang Y, Sun Z, Mao X, et al. . Impact of mismatch-repair deficiency on the colorectal cancer immune microenvironment. Oncotarget 2017;8:85526–36. 10.18632/oncotarget.20241
    1. Pagès F, Mlecnik B, Marliot F, et al. . International validation of the consensus immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet 2018;391:2128–39. 10.1016/S0140-6736(18)30789-X
    1. Thomas J, Leal A, Overman MJ. Clinical development of immunotherapy for deficient mismatch repair colorectal cancer. Clin Colorectal Cancer 2020;19:73–81. 10.1016/j.clcc.2020.02.002
    1. Schmid P, Adams S, Rugo HS, et al. . Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N Engl J Med 2018;379:2108–21. 10.1056/NEJMoa1809615
    1. Uno T, Takeda K, Kojima Y, et al. . Eradication of established tumors in mice by a combination antibody-based therapy. Nat Med 2006;12:693–8. 10.1038/nm1405
    1. Bonaventura P, Shekarian T, Alcazer V, et al. . Cold tumors: a therapeutic challenge for immunotherapy. Front Immunol 2019;10:168. 10.3389/fimmu.2019.00168
    1. Szekely B, Bossuyt V, Li X, et al. . Immunological differences between primary and metastatic breast cancer. Ann Oncol 2018;29:2232–9. 10.1093/annonc/mdy399
    1. Ramakrishnan R, Assudani D, Nagaraj S, et al. . Chemotherapy enhances tumor cell susceptibility to CTL-mediated killing during cancer immunotherapy in mice. J Clin Invest 2010;120:1111–24. 10.1172/JCI40269
    1. Kuo PT, Zeng Z, Salim N, et al. . The role of CXCR3 and its chemokine ligands in skin disease and cancer. Front Med 2018;5:271. 10.3389/fmed.2018.00271
    1. Binnewies M, Roberts EW, Kersten K, et al. . Understanding the tumor immune microenvironment (time) for effective therapy. Nat Med 2018;24:541–50. 10.1038/s41591-018-0014-x
    1. Paluskievicz CM, Cao X, Abdi R, et al. . T regulatory cells and priming the suppressive tumor microenvironment. Front Immunol 2019;10:2453. 10.3389/fimmu.2019.02453
    1. Spitzer MH, Carmi Y, Reticker-Flynn NE, et al. . Systemic immunity is required for effective cancer immunotherapy. Cell 2017;168:e15:487–502. 10.1016/j.cell.2016.12.022
    1. Stucci S, Palmirotta R, Passarelli A, et al. . Immune-Related adverse events during anticancer immunotherapy: pathogenesis and management. Oncol Lett 2017;14:5671–80. 10.3892/ol.2017.6919
    1. Liu J, Blake SJ, Smyth MJ, et al. . Improved mouse models to assess tumour immunity and irAEs after combination cancer immunotherapies. Clin Transl Immunology 2014;3:e22. 10.1038/cti.2014.18
    1. Liu J, Blake SJ, Harjunpää H, et al. . Assessing immune-related adverse events of efficacious combination immunotherapies in preclinical models of cancer. Cancer Res 2016;76:5288–301. 10.1158/0008-5472.CAN-16-0194
    1. Hicks KC, Knudson KM, Lee KL, et al. . Cooperative immune-mediated mechanisms of the HDAC inhibitor entinostat, an IL15 superagonist, and a cancer vaccine effectively synergize as a novel cancer therapy. Clin Cancer Res 2020;26:704–16. 10.1158/1078-0432.CCR-19-0727
    1. Redman JM, Steinberg SM, Gulley JL. Quick efficacy seeking trial (QuEST1): a novel combination immunotherapy study designed for rapid clinical signal assessment metastatic castration-resistant prostate cancer. J Immunother Cancer 2018;6:91. 10.1186/s40425-018-0409-8

Source: PubMed

3
Abonnieren