[en] BACKGROUND: Loss of Ambra1 (autophagy and beclin 1 regulator 1), a multifunctional scaffold protein, promotes the formation of nevi and contributes to several phases of melanoma development. The suppressive functions of Ambra1 in melanoma are mediated by negative regulation of cell proliferation and invasion; however, evidence suggests that loss of Ambra1 may also affect the melanoma microenvironment. Here, we investigate the possible impact of Ambra1 on antitumor immunity and response to immunotherapy. METHODS: This study was performed using an Ambra1-depleted Braf(V600E) /Pten(-/) (-) genetically engineered mouse (GEM) model of melanoma, as well as GEM-derived allografts of Braf(V600E) /Pten(-/) (-) and Braf(V600E) /Pten(-/) (-)/Cdkn2a(-/) (-) tumors with Ambra1 knockdown. The effects of Ambra1 loss on the tumor immune microenvironment (TIME) were analyzed using NanoString technology, multiplex immunohistochemistry, and flow cytometry. Transcriptome and CIBERSORT digital cytometry analyses of murine melanoma samples and human melanoma patients (The Cancer Genome Atlas) were applied to determine the immune cell populations in null or low-expressing AMBRA1 melanoma. The contribution of Ambra1 on T-cell migration was evaluated using a cytokine array and flow cytometry. Tumor growth kinetics and overall survival analysis in Braf(V600E) /Pten(-/) (-)/Cdkn2a(-/) (-) mice with Ambra1 knockdown were evaluated prior to and after administration of a programmed cell death protein-1 (PD-1) inhibitor. RESULTS: Loss of Ambra1 was associated with altered expression of a wide range of cytokines and chemokines as well as decreased infiltration of tumors by regulatory T cells, a subpopulation of T cells with potent immune-suppressive properties. These changes in TIME composition were associated with the autophagic function of Ambra1. In the Braf(V600E) /Pten(-/) (-)/Cdkn2a(-/) (-) model inherently resistant to immune checkpoint blockade, knockdown of Ambra1 led to accelerated tumor growth and reduced overall survival, but at the same time conferred sensitivity to anti-PD-1 treatment. CONCLUSIONS: This study shows that loss of Ambra1 affects the TIME and the antitumor immune response in melanoma, highlighting new functions of Ambra1 in the regulation of melanoma biology.
Disciplines :
Sciences du vivant: Multidisciplinaire, généralités & autres
Auteur, co-auteur :
Frias, Alex
Di Leo, Luca
Antoranz, Asier
Nazerai, Loulieta
Carretta, Marco
Bodemeyer, Valérie
Pagliuca, Chiara
Dahl, Christina
Claps, Giuseppina
Mandelli, Giulio Eugenio
Andhari, Madhavi Dipak
PIRES PACHECO, Maria Irene ; University of Luxembourg > Faculty of Science, Technology and Medicine (FSTM) > Department of Life Sciences and Medicine (DLSM)
SAUTER, Thomas ; University of Luxembourg > Faculty of Science, Technology and Medicine (FSTM) > Department of Life Sciences and Medicine (DLSM)
Shain AH, Bastian BC. From melanocytes to melanomas. Nat Rev Cancer 2016;16:345-58.
Shain AH, Joseph NM, Yu R, et al. Genomic and transcriptomic analysis reveals incremental disruption of key signaling pathways during melanoma evolution. Cancer Cell 2018;34:45-55.
Luke JJ, Flaherty KT, Ribas A, et al. Targeted agents and immunotherapies: optimizing outcomes in melanoma. Nat Rev Clin Oncol 2017;14:463-82.
Huang AC, Zappasodi R. A decade of checkpoint blockade immunotherapy in melanoma: understanding the molecular basis for immune sensitivity and resistance. Nat Immunol 2022;23:660-70.
Robert C. A decade of immune-checkpoint inhibitors in cancer therapy. Nat Commun 2020;11:3801.
Carlino MS, Larkin J, Long GV. Immune checkpoint inhibitors in melanoma. Lancet 2021;398:1002-14.
Morad G, Helmink BA, Sharma P, et al. Hallmarks of response, resistance, and toxicity to immune checkpoint blockade. Cell 2021;184:5309-37.
Barnes TA, Amir E. Hype or hope: the prognostic value of infiltrating immune cells in cancer. Br J Cancer 2017;117:451-60.
Binnewies M, Roberts EW, Kersten K, et al. Understanding the tumor immune microenvironment (time) for effective therapy. Nat Med 2018;24:541-50.
Di Leo L, Bodemeyer V, Bosisio FM, et al. Loss of ambra1 promotes melanoma growth and invasion. Nat Commun 2021;12:2550.
Cianfanelli V, De Zio D, Di Bartolomeo S, et al. Ambra1 at a glance. J Cell Sci 2015;128:2003-8.
Cianfanelli V, Fuoco C, Lorente M, et al. AMBRA1 links autophagy to cell proliferation and tumorigenesis by promoting c-myc dephosphorylation and degradation. Nat Cell Biol 2015;17:20-30.
Maiani E, Milletti G, Nazio F, et al. AMBRA1 regulates cyclin D to guard S-phase entry and genomic integrity. Nature 2021;592:799-803.
Fimia GM, Stoykova A, Romagnoli A, et al. Ambra1 regulates autophagy and development of the nervous system. Nature 2007;447:1121-5.
Di Bartolomeo S, Corazzari M, Nazio F, et al. The dynamic interaction of AMBRA1 with the dynein motor complex regulates mammalian autophagy. J Cell Biol 2010;191:155-68.
Strappazzon F, Di Rita A, Cianfanelli V, et al. Prosurvival AMBRA1 turns into a proapoptotic BH3-like protein during mitochondrial apoptosis. Autophagy 2016;12:963-75.
Strappazzon F, Nazio F, Corrado M, et al. AMBRA1 is able to induce mitophagy via LC3 binding, regardless of parkin and p62/SQSTM1. Cell Death Differ 2015;22:419-32.
Bolognesi MM, Manzoni M, Scalia CR, et al. Multiplex staining by sequential immunostaining and antibody removal on routine tissue sections. J Histochem Cytochem 2017;65:431-44.
Tiberti M, Di Leo L, Vistesen MV, et al. The cancermuts software package for the prioritization of missense cancer variants: a case study of AMBRA1 in melanoma. Cell Death Dis 2022;13:872.
Di Leo L, Vegliante R, Ciccarone F, et al. Forcing ATGL expression in hepatocarcinoma cells imposes glycolytic rewiring through PPAR-/p300-mediated acetylation of p53. Oncogene 2019;38:1860-75.
Vandesompele J, De Preter K, Pattyn F, et al. Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol 2002;3:RESEARCH0034.
Väremo L, Nielsen J, Nookaew I. Enriching the gene set analysis of genome-wide data by incorporating directionality of gene expression and combining statistical hypotheses and methods. Nucleic Acids Res 2013;41:4378-91.
Chen B, Khodadoust MS, Liu CL, et al. Profiling tumor infiltrating immune cells with CIBERSORT. Methods Mol Biol 2018;1711:243-59.
Hu H, Miao YR, Jia LH, et al. AnimalTFDB 3.0: a comprehensive resource for annotation and prediction of animal transcription factors. Nucleic Acids Res 2019;47:D33-8.
Riaz N, Havel JJ, Makarov V, et al. Tumor and microenvironment evolution during immunotherapy with nivolumab. Cell 2017;171:934-49.
Antoranz A, Van Herck Y, Bolognesi MM, et al. Mapping the immune landscape in metastatic melanoma reveals localized cell-cell interactions that predict immunotherapy response. Cancer Res 2022;82:3275-90.
Wiedemann GM, Knott MML, Vetter VK, et al. Cancer cell-derived IL-1 induces CCL22 and the recruitment of regulatory T cells. Oncoimmunology 2016;5:e1175794.
Serrels A, Lund T, Serrels B, et al. Nuclear FAK controls chemokine transcription, tregs, and evasion of anti-tumor immunity. Cell 2015;163:160-73.
Ouyang Z, Wu H, Li L, et al. Regulatory T cells in the immunotherapy of melanoma. Tumour Biol 2016;37:77-85.
Jacobs JFM, Nierkens S, Figdor CG, et al. Regulatory T cells in melanoma: the final hurdle towards effective immunotherapy? Lancet Oncol 2012;13:e32-42.
Huang L, Guo Y, Liu S, et al. Targeting regulatory T cells for immunotherapy in melanoma. Mol Biomed 2021;2:11.
Meeth K, Wang JX, Micevic G, et al. The YUMM lines: a series of congenic mouse melanoma cell lines with defined genetic alterations. Pigment Cell Melanoma Res 2016;29:590-7.
Homet Moreno B, Zaretsky JM, Garcia-Diaz A, et al. Response to programmed cell death-1 blockade in a murine melanoma syngeneic model requires costimulation, CD4, and CD8 T cells. Cancer Immunol Res 2016;4:845-57.
Lelliott EJ, Cullinane C, Martin CA, et al. A novel immunogenic mouse model of melanoma for the preclinical assessment of combination targeted and immune-based therapy. Sci Rep 2019;9:1225.
Wang J, Perry CJ, Meeth K, et al. Uv-Induced somatic mutations elicit a functional T cell response in the YUMMER1.7 mouse melanoma model. Pigment Cell Melanoma Res 2017;30:428-35.
Sharma P, Hu-Lieskovan S, Wargo JA, et al. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 2017;168:707-23.
Arce Vargas F, Furness AJS, Solomon I, et al. Fc-optimized anti-CD25 depletes tumor-infiltrating regulatory T cells and synergizes with PD-1 blockade to eradicate established tumors. Immunity 2017;46:577-86.
Arce Vargas F, Furness AJS, Litchfield K, et al. Fc effector function contributes to the activity of human anti-CTLA-4 antibodies. Cancer Cell 2018;33:649-63.
Solomon I, Amann M, Goubier A, et al. CD25-Treg-depleting antibodies preserving IL-2 signaling on effector T cells enhance effector activation and antitumor immunity. Nat Cancer 2020;1:1153-66.
Harris J. Autophagy and IL-1 family cytokines. Front Immunol 2013;4:83.
Mgrditchian T, Arakelian T, Paggetti J, et al. Targeting autophagy inhibits melanoma growth by enhancing NK cells infiltration in a CCL5-dependent manner. Proc Natl Acad Sci U S A 2017;114:E9271-9.
Monkkonen T, Debnath J. Inflammatory signaling cascades and autophagy in cancer. Autophagy 2018;14:190-8.
Crian TO, Plantinga TS, van de Veerdonk FL, et al. Inflammasome-independent modulation of cytokine response by autophagy in human cells. PLoS One 2011;6:e18666.
Criollo A, Chereau F, Malik SA, et al. Autophagy is required for the activation of NFB. Cell Cycle 2012;11:194-9.
Di Leo L, De Zio D. AMBRA1 has an impact on melanoma development beyond autophagy. Autophagy 2021;17:1802-3.
Sumimoto H, Imabayashi F, Iwata T, et al. The BRAF-MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells. J Exp Med 2006;203:1651-6.
Hamarsheh S, Groß O, Brummer T, et al. Immune modulatory effects of oncogenic KRAS in cancer. Nat Commun 2020;11:5439.
Li X, Lyu Y, Li J, et al. AMBRA1 and its role as a target for anticancer therapy. Front Oncol 2022;12:946086.