Upregulation of C/EBPα Inhibits Suppressive Activity of Myeloid Cells and Potentiates Antitumor Response in Mice and Patients with Cancer

Ayumi Hashimoto, Debashis Sarker, Vikash Reebye, Sheba Jarvis, Mikael H Sodergren, Andrew Kossenkov, Emilio Sanseviero, Nina Raulf, Jenni Vasara, Pinelopi Andrikakou, Tim Meyer, Kai-Wen Huang, Ruth Plummer, Cheng E Chee, Duncan Spalding, Madhava Pai, Shahid Khan, David J Pinato, Rohini Sharma, Bristi Basu, Daniel Palmer, Yuk-Ting Ma, Jeff Evans, Robert Habib, Anna Martirosyan, Naouel Elasri, Adeline Reynaud, John J Rossi, Mark Cobbold, Nagy A Habib, Dmitry I Gabrilovich, Ayumi Hashimoto, Debashis Sarker, Vikash Reebye, Sheba Jarvis, Mikael H Sodergren, Andrew Kossenkov, Emilio Sanseviero, Nina Raulf, Jenni Vasara, Pinelopi Andrikakou, Tim Meyer, Kai-Wen Huang, Ruth Plummer, Cheng E Chee, Duncan Spalding, Madhava Pai, Shahid Khan, David J Pinato, Rohini Sharma, Bristi Basu, Daniel Palmer, Yuk-Ting Ma, Jeff Evans, Robert Habib, Anna Martirosyan, Naouel Elasri, Adeline Reynaud, John J Rossi, Mark Cobbold, Nagy A Habib, Dmitry I Gabrilovich

Abstract

Purpose: To evaluate the mechanisms of how therapeutic upregulation of the transcription factor, CCAAT/enhancer-binding protein alpha (C/EBPα), prevents tumor progression in patients with advanced hepatocellular carcinoma (HCC) and in different mouse tumor models.

Experimental design: We conducted a phase I trial in 36 patients with HCC (NCT02716012) who received sorafenib as part of their standard care, and were given therapeutic C/EBPα small activating RNA (saRNA; MTL-CEBPA) as either neoadjuvant or adjuvant treatment. In the preclinical setting, the effects of MTL-CEBPA were assessed in several mouse models, including BNL-1ME liver cancer, Lewis lung carcinoma (LLC), and colon adenocarcinoma (MC38).

Results: MTL-CEBPA treatment caused radiologic regression of tumors in 26.7% of HCC patients with an underlying viral etiology with 3 complete responders. MTL-CEBPA treatment in those patients caused a marked decrease in peripheral blood monocytic myeloid-derived suppressor cell (M-MDSC) numbers and an overall reduction in the numbers of protumoral M2 tumor-associated macrophages (TAM). Gene and protein analysis of patient leukocytes following treatment showed CEBPA activation affected regulation of factors involved in immune-suppressive activity. To corroborate this observation, treatment of all the mouse tumor models with MTL-CEBPA led to a reversal in the suppressive activity of M-MDSCs and TAMs, but not polymorphonuclear MDSCs (PMN-MDSC). The antitumor effects of MTL-CEBPA in these tumor models showed dependency on T cells. This was accentuated when MTL-CEBPA was combined with checkpoint inhibitors or with PMN-MDSC-targeted immunotherapy.

Conclusions: This report demonstrates that therapeutic upregulation of the transcription factor C/EBPα causes inactivation of immune-suppressive myeloid cells with potent antitumor responses across different tumor models and in cancer patients. MTL-CEBPA is currently being investigated in combination with pembrolizumab in a phase I/Ib multicenter clinical study (NCT04105335).

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

Figures

Figure 1.
Figure 1.
Clinical activity of MTL-CEBPA in advanced HCC patients treated in combination with sorafenib. A, Waterfall plot of patients in phase Ib study showing best percentage (%) change from baseline, with identification of groups that had previously been treated with TKI and those that had HCC of viral etiology. B, Durable responses of patients previously naïve to TKI with HCC of viral etiology. Spider plot in phase Ib patients who had not previously been treated with TKI and had HCC of viral etiology, showing tumor response for target lesions. C, Complete radiologic response of lung metastases following treatment with MTL-CEBPA and sorafenib. Cross-sectional imaging of a patient with baseline imaging on top from June 12, 2018, showing right lung metastases and on bottom from December 31, 2018, showing complete resolution of lung metastases. This patient maintains a complete radiologic response to both liver and lung metastases on last surveillance imaging on March 13, 2020.
Figure 2.
Figure 2.
Effect of MTL-CEBPA treatment of patients with HCC on gene and protein expression in myeloid cells. A, Gene-expression profile was evaluated by NanoString using the human PanCaner IO 360 panel. Heat map of gene-expression upregulated (+1> log2 fold change and above) or downregulated (−1< log2 fold change and below) with a false discovery rate (FDR) of <5% is shown. B, Protein expression profile was evaluated by mass spectroscopy. Proteins with P < 0.05 and absolute log2 fold change > 1 were considered as significantly differentially expressed. Adjusted P values were calculated by correcting for an FDR of <5%.
Figure 3.
Figure 3.
Changes in gene expression in total leukocytes in patients treated with MTL-CEBPA. A, Expression of indicated genes in leukocytes from 12 patients. Gene expression was evaluated by qRT-PCR. Individual results, mean, and standard deviation are shown. P values are calculated using two-sided Student t test. B, the presence of M-MDSCs (CD66b− CD14+ HLA-DR−/loCD15− CD11b+CD38+) and PMN-MDSCs (CD66b+CD14−CD15+CD11b+ LOX1+) among mononuclear cells was analyzed by flow cytometry and represented as frequency of gated cell population at 60K event per 1 × 106 cells (n = 3). C, Total circulating population of monocytes and neutrophils in the same patients was measured as a percentage of total PBMC.
Figure 4.
Figure 4.
The effects of MTL-CEBPA treatment on TAMs in patients with HCC. A, Representative images of a CR patient's biopsies with a complete loss of protumoral M2 macrophages (blue squares: CD68+ CD163+ CD64− cells) are shown in bottom (CR posttreatment) panel when compared with top (CR pretreatment) panel. White squares represent the pan-macrophage population expressing CD68+. Pseudo-color image: created by virtual slides alignment and imported in Halo software for biomarker analysis. B, A heat map of macrophage subsets was set up based on log2 fold change between pre- and posttreatment (cell densities) samples of 3 HCC patients including the CR as shown in A, stable disease (SD), and progressive disease (PD). The macrophage populations analyzed were: pan-macrophage (CD68+ cells), antitumoral M1 macrophages (CD68+CD64+CD163−CD206− cells), activated M1 macrophages (CD68+CD64+CD163−CD206− cells), protumoral M2 macrophages subset 1 (CD68+CD163+CD64− cells), protumoral M2 macrophages subset 2 (CD68+CD206+CD64− cells), protumoral M2 macrophages subset 3 (CD68+CD163+CD206+CD64− cells), and activated M2 subsets characterized by IL10 production.
Figure 5.
Figure 5.
The effect of MTL-CEBPA on tumor growth in mouse tumor models. MTL-CEBPA and control NOV-FLUC were intravenously injected to the tumor-bearing mice at 3 mg/kg twice a week from day 5. A, Kinetic of tumor growth in mice bearing BNL HCC cell line (n = 5). P value was calculated using two-way ANOVA test. B, The presence of the indicated cell population in spleens of NBL tumor–bearing mice presented as percentages (%). C, Tumor volume in NBL tumor–bearing mice treated with MTL-CEBPA and sorafenib after 10 days of treatment. Mean and standard deviation are shown. n = 6 for PBS control- and sorafenib-treated groups, n = 10 for the MTL-CEBPA–treated group, n = 8 for the combination group. P values were calculated in one-way ANOVA test with corrections for multiple comparisons. D, Kinetics of LLC tumor growth (n = 5 per group). P value was calculated using two-way ANOVA test. E, Kinetics of LLC tumor growth in the mice depleted of CD8 T cells and treated with MTL-CEBPA (n = 5 per group). Mean and standard deviation are shown. P values were calculated using two-way ANOVA. F, Kinetics of tumor growth in NOD-SCID mice (n = 4 and 5 per group).
Figure 6.
Figure 6.
Effect of MTL-CEBPA treatment on immune-suppressive function of myeloid cells. A, Suppression of T-cell proliferation by M-MDSCs, macrophages, and PMN-MDSCs isolated from the tumors of the LLC tumor–bearing mice treated with NOV-FLUC or MTL-CEBPA for 2 weeks (n = 4). Mean and standard deviation are shown. P values were calculated using two-sided Student t test. B, Suppression of T-cell proliferation by M-MDSCs and PMN-MDSCs isolated from the spleens of the LLC tumor–bearing mice treated with NOV-FLUC or MTL-CEBPA for 2 weeks (n = 4). Mean and standard deviation are shown. P values were calculated using a two-sided Student t test. C and D, TAMs and PMN-MDSCs were isolated from the tumors of LLC tumor–bearing mice treated with NOV-FLUC or MTL-CEBPA for 2 weeks and used for RNA-seq analysis. C, Pathways predicted to be inhibited (z-score < −2) in TAMs in MTL-CEBPA as compared with NOV-FLUC–treated groups. D, Regulators predicted to be activated (z-score > 2) or inhibited (z-score < −2) in TAMs in MTL-CEBPA as compared with NOV-FLUC–treated group.
Figure 7.
Figure 7.
Therapeutic activity of MTL-CEBPA in combination with checkpoint inhibitors. A, MC38 tumor–bearing mice were treated with MTL-CEBPA or NOV-FLUC control at 5 mg/kg from day 4 (twice a week). Anti–PD-1 antibody was intraperitoneally injected to the mice twice a week at 10 mg/kg. n = 5 per group. Mean and SEM are shown. P values were calculated using two-way ANOVA test. B, LLC tumor–bearing mice were treated with MTL-CEBPA or NOV-FLUC control at 3 mg/kg from day 3 (twice a week). Anti–CTLA-4 antibody was intraperitoneally injected to the mice on days 10, 17, and 24 (100 μg/mouse). Celecoxib was orally given to the mice at 50 mg/kg from day 3 (daily). Mean and SEM (n = 4) are shown. P values were calculated using two-way ANOVA test. C, LLC tumor–bearing mice were treated with MTL-CEBPA or NOV-FLUC (3 mg/kg from day 3, twice a week) in combination with lipofermata (2 mg/kg, twice per day from day 3, subcutaneously). In each experiment, P values were calculated in two-way ANOVA.

References

    1. Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, et al. . Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med 2018;24:541–50.
    1. Veglia F, Perego M, Gabrilovich D. Myeloid-derived suppressor cells coming of age. Nat Immunol 2018;19:108–19.
    1. Newman JR, Keating AE. Comprehensive identification of human bZIP interactions with coiled-coil arrays. Science 2003;300:2097–101.
    1. Avellino R, Delwel R. Expression and regulation of C/EBPalpha in normal myelopoiesis and in malignant transformation. Blood 2017;129:2083–91.
    1. Lourenco AR, Coffer PJ. A tumor suppressor role for C/EBPalpha in solid tumors: more than fat and blood. Oncogene 2017;36:5221–30.
    1. Marigo I, Bosio E, Solito S, Mesa C, Fernandez A, Dolcetti L, et al. . Tumor-induced tolerance and immune suppression depend on the C/EBPbeta transcription factor. Immunity 2010;32:790–802.
    1. Mackert JR, Qu P, Min Y, Johnson PF, Yang L, Lin PC. Dual negative roles of C/EBPalpha in the expansion and protumor functions of MDSCs. Sci Rep 2017;7:14048.
    1. Reebye V, Huang KW, Lin V, Jarvis S, Cutilas P, Dorman S, et al. . Gene activation of CEBPA using saRNA: preclinical studies of the first in human saRNA drug candidate for liver cancer. Oncogene 2018;37:3216–28.
    1. Huan H, Wen X, Chen X, Wu L, Liu W, Habib NA, et al. . C/EBPalpha short-activating RNA suppresses metastasis of hepatocellular carcinoma through inhibiting EGFR/beta-catenin signaling mediated EMT. PLoS One 2016;11:e0153117.
    1. Voutila J, Reebye V, Roberts TC, Protopapa P, Andrikakou P, Blakey DC, et al. . Development and mechanism of small activating RNA targeting CEBPA, a novel therapeutic in clinical trials for liver cancer. Mol Ther 2017;25:2705–14.
    1. Sarker D, Plummer R, Meyer T, Sodergren MH, Basu B, Chee CE, et al. . MTL-CEBPA, a small activating RNA therapeutic upregulating C/EBP-alpha, in patients with advanced liver cancer: a first-in-human, multicenter, open-label, phase I trial. Clin Cancer Res 2020;26:3936–46.
    1. Sarker D, Sodergren M, Plummer ER, Basu B, Meyer T, Huang K-W, et al. . Phase Ib dose escalation and cohort expansion study of the novel myeloid differentiating agent MTL-CEBPA in combination with sorafenib in patients with advanced hepatocellular carcinoma (HCC). J Clin Oncol 2020;38:4601.
    1. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al. . Sorafenib in advanced hepatocellular carcinoma. N Engl J Med 2008;359:378–90.
    1. Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, et al. . Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med 2020;382:1894–905.
    1. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods 2012;9:357–9.
    1. Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-seq data with or without a reference genome. BMC Bioinformatics 2011;12:323.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 2014;15:550.
    1. Team RC. R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria; 2013. Available from: .
    1. Wickham H. ggplot2: Elegant graphics for data analysis. New York: Springer-Verlag; 2016.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Series B Stat Methodol 1995;B, 57:289–300.
    1. Rajeeve V, Vendrell I, Wilkes E, Torbett N, Cutillas PR. Cross-species proteomics reveals specific modulation of signaling in cancer and stromal cells by phosphoinositide 3-kinase (PI3K) inhibitors. Mol Cell Proteomics 2014;13:1457–70.
    1. Casado P, Rodriguez-Prados JC, Cosulich SC, Guichard S, Vanhaesebroeck B, Joel S, et al. . Kinase-substrate enrichment analysis provides insights into the heterogeneity of signaling pathway activation in leukemia cells. Sci Signal 2013;6:rs6.
    1. Larsen MR, Thingholm TE, Jensen ON, Roepstorff P, Jørgensen TJ. Highly selective enrichment of phosphorylated peptides from peptide mixtures using titanium dioxide microcolumns. Mol Cell Proteomics 2005;4:873–86.
    1. Montoya A, Beltran L, Casado P, Rodriguez-Prados JC, Cutillas PR. Characterization of a TiO(2) enrichment method for label-free quantitative phosphoproteomics. Methods (San Diego, Calif) 2011;54:370–8.
    1. Perkins DN, Pappin DJ, Creasy DM, Cottrell JS. Probability-based protein identification by searching sequence databases using mass spectrometry data. Electrophoresis 1999;20:3551–67.
    1. Casado P, Cutillas PR. A self-validating quantitative mass spectrometry method for assessing the accuracy of high-content phosphoproteomic experiments. Mol Cell Proteomics 2011;10:M110 003079.
    1. Cutillas PR, Vanhaesebroeck B. Quantitative profile of five murine core proteomes using label-free functional proteomics. Mol Cell Proteomics 2007;6:1560–73.
    1. Law CW, Chen Y, Shi W, Smyth GK. Voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome biol 2014;15:R29.
    1. Warren S, Danaher P, Mashadi-Hossein A, Skewis L, Wallden B, Ferree S, et al. . Development of gene expression-based biomarkers on the ncounter((R)) platform for immuno-oncology applications. Methods Mol Biol 2020;2055:273–300.
    1. Zelenay S, van der Veen AG, Bottcher JP, Snelgrove KJ, Rogers N, Acton SE, et al. . Cyclooxygenase-dependent tumor growth through evasion of immunity. Cell 2015;162:1257–70.
    1. Efremova M, Rieder D, Klepsch V, Charoentong P, Finotello F, Hackl H, et al. . Targeting immune checkpoints potentiates immunoediting and changes the dynamics of tumor evolution. Nat Commun 2018;9:32.
    1. Veglia F, Tyurin VA, Blasi M, De Leo A, Kossenkov AV, Donthireddy L, et al. . Fatty acid transport protein 2 reprograms neutrophils in cancer. Nature 2019;569:73–8.
    1. Ruffell D, Mourkioti F, Gambardella A, Kirstetter P, Lopez RG, Rosenthal N, et al. . A CREB-C/EBPbeta cascade induces M2 macrophage-specific gene expression and promotes muscle injury repair. Proc Natl Acad Sci U S A 2009;106:17475–80.
    1. Radomska HS, Huettner CS, Zhang P, Cheng T, Scadden DT, Tenen DG. CCAAT/enhancer binding protein alpha is a regulatory switch sufficient for induction of granulocytic development from bipotential myeloid progenitors. Mol Cell Biol 1998;18:4301–14.
    1. Zhang DE, Zhang P, Wang ND, Hetherington CJ, Darlington GJ, Tenen DG. Absence of granulocyte colony-stimulating factor signaling and neutrophil development in CCAAT enhancer binding protein alpha-deficient mice. Proc Natl Acad Sci U S A 1997;94:569–74.
    1. Prima V, Kaliberova LN, Kaliberov S, Curiel DT, Kusmartsev S. COX2/mPGES1/PGE2 pathway regulates PD-L1 expression in tumor-associated macrophages and myeloid-derived suppressor cells. Proc Natl Acad Sci U S A 2017;114:1117–22.
    1. Cao M, Xu Y, Youn JI, Cabrera R, Zhang X, Gabrilovich D, et al. . Kinase inhibitor sorafenib modulates immunosuppressive cell populations in a murine liver cancer model. Lab Invest 2011;91:598–608.
    1. Zhou SL, Zhou ZJ, Hu ZQ, Huang XW, Wang Z, Chen EB, et al. . Tumor-associated neutrophils recruit macrophages and T-regulatory cells to promote progression of hepatocellular carcinoma and resistance to sorafenib. Gastroenterology 2016;150:1646–58e17.
    1. Sachdeva M, Arora SK. Prognostic role of immune cells in hepatocellular carcinoma. EXCLI J 2020;19:718–33.
    1. Inada Y, Mizukoshi E, Seike T, Tamai T, Iida N, Kitahara M, et al. . Characteristics of immune response to tumor-associated antigens and immune cell profile in patients with hepatocellular carcinoma. Hepatology 2019;69:653–65.

Source: PubMed

Подписаться