miRNA as a Modulator of Immunotherapy and Immune Response in Melanoma
Abstract
:1. Introduction
2. Mechanism of Immunotherapy Resistance in Melanoma
3. miRNAs as Biomarkers to Predict the Response of Melanoma Patients to Immunotherapy
4. Role of miRNAs in the Tumor Microenvironment
4.1. Inflammation
4.2. Cancer-Associated Fibroblasts (CAFs)
4.3. Dendritic Cells (DCs)
4.4. T Cells
4.5. Macrophages
miRNA | Functions in Tumor Immunity | Target Genes | Response to Immunotherapy | Ref. |
---|---|---|---|---|
miR-21 | Reduce activity of T cells, and promote TAM polarization, MDSCs and CAFs | STAT1, SMAD7 | Reduce the response to anti-PD-1 | [75,89,114] |
miR-155 | Enhance antitumor response of CD8+ T cells, activation of M1 macrophages and maturation of DCs Promote the functions of MDSCs Secrete via melanoma EVs to induce differentiation of CAFs Positive regulator of NK cells | STAT5, SHIP1, SOCS1, PTPN2, BACH1, CEBPB, IL7R, BCL6, IL13Rα1, PTEN, KPC1, DC115 | [16,78,79,80,95,96,97,99,106,107,108,116,117] | |
miR-211 | Secrete from melanosome to educate primary fibroblasts to become CAFs | IGF2R | [76] | |
miR-9 | Promote activation and functions of DCs | PCGF6 | [84] | |
miR-128 | Enhance anti-tumor response of DCs | p38 | [83] | |
miR-22 | Reduce the tumor-suppressing role of DCs | p38 | [82] | |
miR-28 | Rescue exhaustive T cells | PD-1 | [88] | |
miR-23a | Negative modulator of CD8+ T cells Inhibit M2 polarization | BLIMP-1, JAK1, STAT6 | [87,110] | |
miR-146 | Reduce immune activation | Reduce the response to anti-PD-1 | [90,91] | |
miR-498 miR-3187-3p | Secrete via melanoma EVs to reduce T cell responses | IFN-γ, PTPRC | [105] | |
miR-192-5p | Regulator of melanoma cells to reduce the cytotoxicity of T cells | [85] | ||
miR-125a-5p | Promote M2 phenotype | KLF13 | [111] | |
miR-125b-5p | Secrete from melanoma EVs to induce TAM phenotypes | LIPA | [112] | |
miR-378 | Induce activation of M1 macrophages | AKT1 | [109] | |
miR-27a | Inhibit M2 polarization | IRF4, PPAR-γ | [110] | |
miR-494 | Enhance the functions of MDSCs | PTEN | [118] | |
let-7e miR-99b miR-100 miR-125a/b miR-146a/b | Secrete from melanoma EVs to induce the polarization and functions of MDSCs | [57] | ||
miR-181a/b | Induce development and functions of NK cells | [119] | ||
miR-34a/c | Induce from melanoma cells to negatively regulate the cytolytic activity of NK cells | ULBP2 | [120] |
4.6. Myeloid-Derived Suppressor Cells (MDSCs)
4.7. Natural Killer Cells (NK Cells)
5. Conclusions and Future Aspects
Author Contributions
Funding
Conflicts of Interest
Abbreviations
FDA | Food and Drug Administration, |
MDSCs | myeloid-derived suppressor cells, |
JAK1 | Janus kinase 1, |
JAK2 | Janus kinase 2, |
B2M | beta-2-microglobulin, |
MHC-I | major histocompatibility complex class 1, |
STAT1 | signal transducer and activator of transcription 1, |
IFNGR1 | interferon gamma receptor 1, |
PTPN2 | protein tyrosine phosphatase non-receptor type 2, |
APLNR | apelin receptor, |
PBRM1 | polybromo 1, |
ARID2 | AT-rich interaction domain 2, |
BRD7 | bromodomain containing 7, |
USP22 | ubiquitin-specific peptidase 22, |
PTEN | phosphatase and tensin homolog, |
CDKN2A | cyclin-dependent kinase inhibitor 2A, |
SK1 | sphingosine kinase-1, |
HLA-I | human leukocyte antigen class I, |
APM | antigen-processing machinery, |
NGFR | nerve growth factor receptor, |
FMT | fecal microbiome transplantation, |
TMB | tumor mutation burden, |
ADAR1 | adenosine deaminase acting on RNA 1, |
ICAM1 | intercellular adhesion molecule 1, |
AUC | area under curve, |
CAFs | cancer-associated fibroblasts, |
BLIMP-1 | B lymphocyte-induced maturation protein-1, |
TIM3 | T cell immunoglobulin domain and mucin domain 3, |
BTLA | B and T lymphocyte associated, |
TAMs | tumor-associated macrophages, |
irAEs | immune-related adverse events, |
DCs | dendritic cells, |
STAT5 | signal transducer and activator of transcription 5, |
SHIP1 | Src homology-2 domain-containing inositol 5-phosphatase 1, |
SOCS1 | suppressor of cytokine signaling 1, |
BACH1 | BTB domain and CNC homolog 1, |
CEBPB | CCAAT enhancer binding protein beta, |
IL7R | interleukin 7 receptor, |
NF-κB | nuclear factor kappa B, |
AP-1 | activator protein 1, |
PTPRC | protein tyrosine phosphatase receptor type C, |
BCL6 | B-cell lymphoma-6 protein, |
IL13Rα1 | interleukin 13 receptor alpha1, |
AKT1 | AKT serine/threonine kinase 1, |
IRAK1 | interleukin 1 receptor-associated kinase 1, |
TRAF6 | TNF receptor-associated factor 6, |
STAT6 | signal transducer and activator of transcription 6, |
IRF4 | interferon regulatory factor 4, |
PPAR-γ | peroxisome proliferator activated receptor gamma, |
KLF13 | Kruppel-like factor 13, |
LIPA | lysosomal acid lipase A, |
EVs | extracellular vesicles, |
SMAD7 | SMAD family member 7, |
IGF2R | insulin-like growth factor 2 receptor, |
VEGFA | vascular endothelial growth factor A, |
FGF2 | fibroblast growth factor 2, |
NLK | Nemo-like kinase, |
ULBP2 | UL16 binding protein 2, |
TAP1 | antigen processing 1, |
HLA-A | major histocompatibility complex class I, A, |
BMDCs | bone marrow-derived dendritic cells, |
cDC1s | conventional DC1s, |
CXCL | C-X-C motif ligand, |
MMPs | matrix metalloproteinases, |
JNK | c-Jun N-terminal kinase, |
MITF | microphthalmia-associated transcription factor, |
TYR | tyrosinase, |
MLANA | Melan-A/Mart-1, |
CTL | cytolytic T lymphocytes, |
TRAF6 | TNFR-associated factor 6, |
IRAK1/2 | Il-1R-associated kinase 1/2. |
References
- Weiss, S.A.; Wolchok, J.D.; Sznol, M. Immunotherapy of Melanoma: Facts and Hopes. Clin. Cancer Res. 2019, 25, 5191–5201. [Google Scholar] [CrossRef] [Green Version]
- Kirkwood, J.M.; Ibrahim, J.G.; Sosman, J.A.; Sondak, V.K.; Agarwala, S.S.; Ernstoff, M.S.; Rao, U. High-dose interferon alfa-2b significantly prolongs relapse-free and overall survival compared with the GM2-KLH/QS-21 vaccine in patients with resected stage IIB-III melanoma: Results of intergroup trial E1694/S9512/C509801. J. Clin. Oncol. 2001, 19, 2370–2380. [Google Scholar] [CrossRef] [PubMed]
- Kirkwood, J.M.; Strawderman, M.H.; Ernstoff, M.S.; Smith, T.J.; Borden, E.C.; Blum, R.H. Interferon alfa-2b adjuvant therapy of high-risk resected cutaneous melanoma: The Eastern Cooperative Oncology Group Trial EST 1684. J. Clin. Oncol. 1996, 14, 7–17. [Google Scholar] [CrossRef]
- Coit, D.G.; Andtbacka, R.; Bichakjian, C.K.; Dilawari, R.A.; Dimaio, D.; Guild, V.; Halpern, A.C.; Hodi, F.S.; Kashani-Sabet, M.; Lange, J.R.; et al. Melanoma. J. Natl. Compr. Cancer Netw. 2009, 7, 250–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 2010, 363, 711–723. [Google Scholar] [CrossRef]
- Mansh, M. Ipilimumab and cancer immunotherapy: A new hope for advanced stage melanoma. Yale J. Biol. Med. 2011, 84, 381–389. [Google Scholar]
- Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Dummer, R.; Smylie, M.; Rutkowski, P.; et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N. Engl. J. Med. 2015, 373, 23–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schachter, J.; Ribas, A.; Long, G.V.; Arance, A.; Grob, J.J.; Mortier, L.; Daud, A.; Carlino, M.S.; McNeil, C.; Lotem, M.; et al. Pembrolizumab versus ipilimumab for advanced melanoma: Final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006). Lancet 2017, 390, 1853–1862. [Google Scholar] [CrossRef]
- Luke, J.J.; Flaherty, K.T.; Ribas, A.; Long, G.V. Targeted agents and immunotherapies: Optimizing outcomes in melanoma. Nat. Rev. Clin. Oncol. 2017, 14, 463–482. [Google Scholar] [CrossRef] [Green Version]
- Herbst, R.S.; Soria, J.C.; Kowanetz, M.; Fine, G.D.; Hamid, O.; Gordon, M.S.; Sosman, J.A.; McDermott, D.F.; Powderly, J.D.; Gettinger, S.N.; et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014, 515, 563–567. [Google Scholar] [CrossRef] [Green Version]
- Hodi, F.S.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Rutkowski, P.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Nivolumab plus ipilimumab or nivolumab alone versus ipilimumab alone in advanced melanoma (CheckMate 067): 4-year outcomes of a multicentre, randomised, phase 3 trial. Lancet Oncol. 2018, 19, 1480–1492. [Google Scholar] [CrossRef]
- Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Rutkowski, P.; Grob, J.J.; Cowey, C.L.; Lao, C.D.; Wagstaff, J.; Schadendorf, D.; Ferrucci, P.F.; et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2017, 377, 1345–1356. [Google Scholar] [CrossRef]
- Paladini, L.; Fabris, L.; Bottai, G.; Raschioni, C.; Calin, G.A.; Santarpia, L. Targeting microRNAs as key modulators of tumor immune response. J. Exp. Clin. Cancer Res. 2016, 35, 103. [Google Scholar] [CrossRef] [Green Version]
- Jorge, N.A.N.; Cruz, J.G.V.; Pretti, M.A.M.; Bonamino, M.H.; Possik, P.A.; Boroni, M. Poor clinical outcome in metastatic melanoma is associated with a microRNA-modulated immunosuppressive tumor microenvironment. J. Transl. Med. 2020, 18, 56. [Google Scholar] [CrossRef]
- Mashima, R. Physiological roles of miR-155. Immunology 2015, 145, 323–333. [Google Scholar] [CrossRef]
- Trotta, R.; Chen, L.; Ciarlariello, D.; Josyula, S.; Mao, C.; Costinean, S.; Yu, L.; Butchar, J.P.; Tridandapani, S.; Croce, C.M.; et al. miR-155 regulates IFN-gamma production in natural killer cells. Blood 2012, 119, 3478–3485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tili, E.; Croce, C.M.; Michaille, J.J. miR-155: On the crosstalk between inflammation and cancer. Int. Rev. Immunol. 2009, 28, 264–284. [Google Scholar] [CrossRef]
- Bai, R.; Chen, N.; Li, L.; Du, N.; Bai, L.; Lv, Z.; Tian, H.; Cui, J. Mechanisms of Cancer Resistance to Immunotherapy. Front. Oncol. 2020, 10, 1290. [Google Scholar] [CrossRef]
- Zaretsky, J.M.; Garcia-Diaz, A.; Shin, D.S.; Escuin-Ordinas, H.; Hugo, W.; Hu-Lieskovan, S.; Torrejon, D.Y.; Abril-Rodriguez, G.; Sandoval, S.; Barthly, L.; et al. Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N. Engl. J. Med. 2016, 375, 819–829. [Google Scholar] [CrossRef]
- Shin, D.S.; Zaretsky, J.M.; Escuin-Ordinas, H.; Garcia-Diaz, A.; Hu-Lieskovan, S.; Kalbasi, A.; Grasso, C.S.; Hugo, W.; Sandoval, S.; Torrejon, D.Y.; et al. Primary Resistance to PD-1 Blockade Mediated by JAK1/2 Mutations. Cancer Discov. 2017, 7, 188–201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Manguso, R.T.; Pope, H.W.; Zimmer, M.D.; Brown, F.D.; Yates, K.B.; Miller, B.C.; Collins, N.B.; Bi, K.; LaFleur, M.W.; Juneja, V.R.; et al. In vivo CRISPR screening identifies Ptpn2 as a cancer immunotherapy target. Nature 2017, 547, 413–418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Patel, S.J.; Sanjana, N.E.; Kishton, R.J.; Eidizadeh, A.; Vodnala, S.K.; Cam, M.; Gartner, J.J.; Jia, L.; Steinberg, S.M.; Yamamoto, T.N.; et al. Identification of essential genes for cancer immunotherapy. Nature 2017, 548, 537–542. [Google Scholar] [CrossRef] [Green Version]
- Li, M.; Xu, Y.; Liang, J.; Lin, H.; Qi, X.; Li, F.; Han, P.; Gao, Y.; Yang, X. USP22 deficiency in melanoma mediates resistance to T cells through IFNgamma-JAK1-STAT1 signal axis. Mol. Ther. 2021, 29, 2108–2120. [Google Scholar] [CrossRef] [PubMed]
- Pan, D.; Kobayashi, A.; Jiang, P.; Ferrari de Andrade, L.; Tay, R.E.; Luoma, A.M.; Tsoucas, D.; Qiu, X.; Lim, K.; Rao, P.; et al. A major chromatin regulator determines resistance of tumor cells to T cell-mediated killing. Science 2018, 359, 770–775. [Google Scholar] [CrossRef] [Green Version]
- Spranger, S.; Bao, R.; Gajewski, T.F. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature 2015, 523, 231–235. [Google Scholar] [CrossRef]
- Trujillo, J.A.; Luke, J.J.; Zha, Y.; Segal, J.P.; Ritterhouse, L.L.; Spranger, S.; Matijevich, K.; Gajewski, T.F. Secondary resistance to immunotherapy associated with beta-catenin pathway activation or PTEN loss in metastatic melanoma. J. Immunother. Cancer 2019, 7, 295. [Google Scholar] [CrossRef] [Green Version]
- Peng, W.; Chen, J.Q.; Liu, C.; Malu, S.; Creasy, C.; Tetzlaff, M.T.; Xu, C.; McKenzie, J.A.; Zhang, C.; Liang, X.; et al. Loss of PTEN Promotes Resistance to T Cell-Mediated Immunotherapy. Cancer Discov. 2016, 6, 202–216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sade-Feldman, M.; Jiao, Y.J.; Chen, J.H.; Rooney, M.S.; Barzily-Rokni, M.; Eliane, J.P.; Bjorgaard, S.L.; Hammond, M.R.; Vitzthum, H.; Blackmon, S.M.; et al. Resistance to checkpoint blockade therapy through inactivation of antigen presentation. Nat. Commun. 2017, 8, 1136. [Google Scholar] [CrossRef]
- Gao, J.; Shi, L.Z.; Zhao, H.; Chen, J.; Xiong, L.; He, Q.; Chen, T.; Roszik, J.; Bernatchez, C.; Woodman, S.E.; et al. Loss of IFN-gamma Pathway Genes in Tumor Cells as a Mechanism of Resistance to Anti-CTLA-4 Therapy. Cell 2016, 167, 397–404.e399. [Google Scholar] [CrossRef] [Green Version]
- Liu, D.; Lin, J.R.; Robitschek, E.J.; Kasumova, G.G.; Heyde, A.; Shi, A.; Kraya, A.; Zhang, G.; Moll, T.; Frederick, D.T.; et al. Evolution of delayed resistance to immunotherapy in a melanoma responder. Nat. Med. 2021, 27, 985–992. [Google Scholar] [CrossRef]
- Imbert, C.; Montfort, A.; Fraisse, M.; Marcheteau, E.; Gilhodes, J.; Martin, E.; Bertrand, F.; Marcellin, M.; Burlet-Schiltz, O.; Peredo, A.G.; et al. Resistance of melanoma to immune checkpoint inhibitors is overcome by targeting the sphingosine kinase-1. Nat. Commun. 2020, 11, 437. [Google Scholar] [CrossRef] [PubMed]
- Anichini, A.; Mortarini, R.; Nonaka, D.; Molla, A.; Vegetti, C.; Montaldi, E.; Wang, X.; Ferrone, S. Association of antigen-processing machinery and HLA antigen phenotype of melanoma cells with survival in American Joint Committee on Cancer stage III and IV melanoma patients. Cancer Res. 2006, 66, 6405–6411. [Google Scholar] [CrossRef] [Green Version]
- Such, L.; Zhao, F.; Liu, D.; Thier, B.; Le-Trilling, V.T.K.; Sucker, A.; Coch, C.; Pieper, N.; Howe, S.; Bhat, H.; et al. Targeting the innate immunoreceptor RIG-I overcomes melanoma-intrinsic resistance to T cell immunotherapy. J. Clin. Investig. 2020, 130, 4266–4281. [Google Scholar] [CrossRef]
- Singh, S.; Xiao, Z.; Bavisi, K.; Roszik, J.; Melendez, B.D.; Wang, Z.; Cantwell, M.J.; Davis, R.E.; Lizee, G.; Hwu, P.; et al. IL-1alpha Mediates Innate and Acquired Resistance to Immunotherapy in Melanoma. J. Immunol. 2021, 206, 1966–1975. [Google Scholar] [CrossRef] [PubMed]
- Fallahi-Sichani, M.; Becker, V.; Izar, B.; Baker, G.J.; Lin, J.R.; Boswell, S.A.; Shah, P.; Rotem, A.; Garraway, L.A.; Sorger, P.K. Adaptive resistance of melanoma cells to RAF inhibition via reversible induction of a slowly dividing de-differentiated state. Mol. Syst. Biol. 2017, 13, 905. [Google Scholar] [CrossRef]
- Boshuizen, J.; Vredevoogd, D.W.; Krijgsman, O.; Ligtenberg, M.A.; Blankenstein, S.; de Bruijn, B.; Frederick, D.T.; Kenski, J.C.N.; Parren, M.; Bruggemann, M.; et al. Reversal of pre-existing NGFR-driven tumor and immune therapy resistance. Nat. Commun. 2020, 11, 3946. [Google Scholar] [CrossRef]
- Dzutsev, A.; Goldszmid, R.S.; Viaud, S.; Zitvogel, L.; Trinchieri, G. The role of the microbiota in inflammation, carcinogenesis, and cancer therapy. Eur. J. Immunol. 2015, 45, 17–31. [Google Scholar] [CrossRef]
- Finlay, B.B.; Goldszmid, R.; Honda, K.; Trinchieri, G.; Wargo, J.; Zitvogel, L. Can we harness the microbiota to enhance the efficacy of cancer immunotherapy? Nat. Rev. Immunol. 2020, 20, 522–528. [Google Scholar] [CrossRef]
- Garrett, W.S. Cancer and the microbiota. Science 2015, 348, 80–86. [Google Scholar] [CrossRef] [Green Version]
- Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Lei, Y.M.; Jabri, B.; Alegre, M.L.; et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015, 350, 1084–1089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Frankel, A.E.; Coughlin, L.A.; Kim, J.; Froehlich, T.W.; Xie, Y.; Frenkel, E.P.; Koh, A.Y. Metagenomic Shotgun Sequencing and Unbiased Metabolomic Profiling Identify Specific Human Gut Microbiota and Metabolites Associated with Immune Checkpoint Therapy Efficacy in Melanoma Patients. Neoplasia 2017, 19, 848–855. [Google Scholar] [CrossRef] [PubMed]
- Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Matson, V.; Fessler, J.; Bao, R.; Chongsuwat, T.; Zha, Y.; Alegre, M.L.; Luke, J.J.; Gajewski, T.F. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018, 359, 104–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Davar, D.; Dzutsev, A.K.; McCulloch, J.A.; Rodrigues, R.R.; Chauvin, J.M.; Morrison, R.M.; Deblasio, R.N.; Menna, C.; Ding, Q.; Pagliano, O.; et al. Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science 2021, 371, 595–602. [Google Scholar] [CrossRef]
- Tray, N.; Weber, J.S.; Adams, S. Predictive Biomarkers for Checkpoint Immunotherapy: Current Status and Challenges for Clinical Application. Cancer Immunol. Res. 2018, 6, 1122–1128. [Google Scholar] [CrossRef] [Green Version]
- Vilain, R.E.; Menzies, A.M.; Wilmott, J.S.; Kakavand, H.; Madore, J.; Guminski, A.; Liniker, E.; Kong, B.Y.; Cooper, A.J.; Howle, J.R.; et al. Dynamic Changes in PD-L1 Expression and Immune Infiltrates Early During Treatment Predict Response to PD-1 Blockade in Melanoma. Clin. Cancer Res. 2017, 23, 5024–5033. [Google Scholar] [CrossRef] [Green Version]
- Teng, M.W.; Ngiow, S.F.; Ribas, A.; Smyth, M.J. Classifying Cancers Based on T-cell Infiltration and PD-L1. Cancer Res. 2015, 75, 2139–2145. [Google Scholar] [CrossRef] [Green Version]
- Snyder, A.; Makarov, V.; Merghoub, T.; Yuan, J.; Zaretsky, J.M.; Desrichard, A.; Walsh, L.A.; Postow, M.A.; Wong, P.; Ho, T.S.; et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 2014, 371, 2189–2199. [Google Scholar] [CrossRef] [Green Version]
- Van Allen, E.M.; Miao, D.; Schilling, B.; Shukla, S.A.; Blank, C.; Zimmer, L.; Sucker, A.; Hillen, U.; Foppen, M.H.G.; Goldinger, S.M.; et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 2015, 350, 207–211. [Google Scholar] [CrossRef] [Green Version]
- Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S.; et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015, 348, 124–128. [Google Scholar] [CrossRef] [Green Version]
- Davoli, T.; Uno, H.; Wooten, E.C.; Elledge, S.J. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science 2017, 355. [Google Scholar] [CrossRef] [Green Version]
- Fattore, L.; Ruggiero, C.F.; Pisanu, M.E.; Liguoro, D.; Cerri, A.; Costantini, S.; Capone, F.; Acunzo, M.; Romano, G.; Nigita, G.; et al. Reprogramming miRNAs global expression orchestrates development of drug resistance in BRAF mutated melanoma. Cell Death Differ. 2019, 26, 1267–1282. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, M.T.; Lin, C.H.; Liu, S.M.; Miyashita, A.; Ihn, H.; Lin, H.; Ng, C.H.; Tsai, J.C.; Chen, M.H.; Tsai, M.S.; et al. miR-524-5p reduces the progression of the BRAF inhibitor-resistant melanoma. Neoplasia 2020, 22, 789–799. [Google Scholar] [CrossRef]
- Nemlich, Y.; Greenberg, E.; Ortenberg, R.; Besser, M.J.; Barshack, I.; Jacob-Hirsch, J.; Jacoby, E.; Eyal, E.; Rivkin, L.; Prieto, V.G.; et al. MicroRNA-mediated loss of ADAR1 in metastatic melanoma promotes tumor growth. J. Clin. Investig. 2013, 123, 2703–2718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Galore-Haskel, G.; Nemlich, Y.; Greenberg, E.; Ashkenazi, S.; Hakim, M.; Itzhaki, O.; Shoshani, N.; Shapira-Fromer, R.; Ben-Ami, E.; Ofek, E.; et al. A novel immune resistance mechanism of melanoma cells controlled by the ADAR1 enzyme. Oncotarget 2015, 6, 28999–29015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bachmann, M.F.; McKall-Faienza, K.; Schmits, R.; Bouchard, D.; Beach, J.; Speiser, D.E.; Mak, T.W.; Ohashi, P.S. Distinct roles for LFA-1 and CD28 during activation of naive T cells: Adhesion versus costimulation. Immunity 1997, 7, 549–557. [Google Scholar] [CrossRef] [Green Version]
- Huber, V.; Vallacchi, V.; Fleming, V.; Hu, X.; Cova, A.; Dugo, M.; Shahaj, E.; Sulsenti, R.; Vergani, E.; Filipazzi, P.; et al. Tumor-derived microRNAs induce myeloid suppressor cells and predict immunotherapy resistance in melanoma. J. Clin. Investig. 2018, 128, 5505–5516. [Google Scholar] [CrossRef] [Green Version]
- Tengda, L.; Shuping, L.; Mingli, G.; Jie, G.; Yun, L.; Weiwei, Z.; Anmei, D. Serum exosomal microRNAs as potent circulating biomarkers for melanoma. Melanoma Res. 2018, 28, 295–303. [Google Scholar] [CrossRef] [PubMed]
- Fridman, W.H.; Pages, F.; Sautes-Fridman, C.; Galon, J. The immune contexture in human tumours: Impact on clinical outcome. Nat. Rev. Cancer 2012, 12, 298–306. [Google Scholar] [CrossRef]
- Martinez-Usatorre, A.; Sempere, L.F.; Carmona, S.J.; Carretero-Iglesia, L.; Monnot, G.; Speiser, D.E.; Rufer, N.; Donda, A.; Zehn, D.; Jandus, C.; et al. MicroRNA-155 Expression Is Enhanced by T-cell Receptor Stimulation Strength and Correlates with Improved Tumor Control in Melanoma. Cancer Immunol. Res. 2019, 7, 1013–1024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nakahara, S.; Fukushima, S.; Okada, E.; Morinaga, J.; Kubo, Y.; Tokuzumi, A.; Matsumoto, S.; Tsuruta-Kadohisa, M.; Kimura, T.; Kuriyama, H.; et al. MicroRNAs that predict the effectiveness of anti-PD-1 therapies in patients with advanced melanoma. J. Dermatol. Sci. 2020, 97, 77–79. [Google Scholar] [CrossRef]
- Bustos, M.A.; Gross, R.; Rahimzadeh, N.; Cole, H.; Tran, L.T.; Tran, K.D.; Takeshima, L.; Stern, S.L.; O’Day, S.; Hoon, D.S.B. A Pilot Study Comparing the Efficacy of Lactate Dehydrogenase Levels Versus Circulating Cell-Free microRNAs in Monitoring Responses to Checkpoint Inhibitor Immunotherapy in Metastatic Melanoma Patients. Cancers 2020, 12, 3361. [Google Scholar] [CrossRef]
- Grenda, A.; Krawczyk, P.; Blach, J.; Chmielewska, I.; Kubiatowski, T.; Kieszko, S.; Wojas-Krawczyk, K.; Kucharczyk, T.; Jarosz, B.; Pasnik, I.; et al. Tissue MicroRNA Expression as a Predictor of Response to Immunotherapy in NSCLC Patients. Front. Oncol. 2020, 10, 563613. [Google Scholar] [CrossRef]
- Fan, J.; Yin, Z.; Xu, J.; Wu, F.; Huang, Q.; Yang, L.; Jin, Y.; Yang, G. Circulating microRNAs predict the response to anti-PD-1 therapy in non-small cell lung cancer. Genomics 2020, 112, 2063–2071. [Google Scholar] [CrossRef]
- Peng, X.X.; Yu, R.; Wu, X.; Wu, S.Y.; Pi, C.; Chen, Z.H.; Zhang, X.C.; Gao, C.Y.; Shao, Y.W.; Liu, L.; et al. Correlation of plasma exosomal microRNAs with the efficacy of immunotherapy in EGFR / ALK wild-type advanced non-small cell lung cancer. J. Immunother. Cancer 2020, 8, e000376. [Google Scholar] [CrossRef] [Green Version]
- Proietti, I.; Skroza, N.; Michelini, S.; Mambrin, A.; Balduzzi, V.; Bernardini, N.; Marchesiello, A.; Tolino, E.; Volpe, S.; Maddalena, P.; et al. BRAF Inhibitors: Molecular Targeting and Immunomodulatory Actions. Cancers 2020, 12, 1823. [Google Scholar] [CrossRef] [PubMed]
- Proietti, I.; Skroza, N.; Bernardini, N.; Tolino, E.; Balduzzi, V.; Marchesiello, A.; Michelini, S.; Volpe, S.; Mambrin, A.; Mangino, G.; et al. Mechanisms of Acquired BRAF Inhibitor Resistance in Melanoma: A Systematic Review. Cancers 2020, 12, 2801. [Google Scholar] [CrossRef] [PubMed]
- Chakraborty, C.; Sharma, A.R.; Sharma, G.; Lee, S.S. The Interplay among miRNAs, Major Cytokines, and Cancer-Related Inflammation. Mol. Ther. Nucleic Acids 2020, 20, 606–620. [Google Scholar] [CrossRef]
- Rupaimoole, R.; Slack, F.J. MicroRNA therapeutics: Towards a new era for the management of cancer and other diseases. Nat. Rev. Drug Discov. 2017, 16, 203–222. [Google Scholar] [CrossRef] [PubMed]
- Arts, N.; Cane, S.; Hennequart, M.; Lamy, J.; Bommer, G.; Van den Eynde, B.; De Plaen, E. microRNA-155, induced by interleukin-1ss, represses the expression of microphthalmia-associated transcription factor (MITF-M) in melanoma cells. PLoS ONE 2015, 10, e0122517. [Google Scholar] [CrossRef] [Green Version]
- Taganov, K.D.; Boldin, M.P.; Chang, K.J.; Baltimore, D. NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc. Natl. Acad. Sci. USA 2006, 103, 12481–12486. [Google Scholar] [CrossRef] [Green Version]
- Hou, J.; Wang, P.; Lin, L.; Liu, X.; Ma, F.; An, H.; Wang, Z.; Cao, X. MicroRNA-146a feedback inhibits RIG-I-dependent Type I IFN production in macrophages by targeting TRAF6, IRAK1, and IRAK2. J. Immunol. 2009, 183, 2150–2158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mann, M.; Mehta, A.; Zhao, J.L.; Lee, K.; Marinov, G.K.; Garcia-Flores, Y.; Lu, L.F.; Rudensky, A.Y.; Baltimore, D. An NF-kappaB-microRNA regulatory network tunes macrophage inflammatory responses. Nat. Commun. 2017, 8, 851. [Google Scholar] [CrossRef] [Green Version]
- Bazzoni, F.; Rossato, M.; Fabbri, M.; Gaudiosi, D.; Mirolo, M.; Mori, L.; Tamassia, N.; Mantovani, A.; Cassatella, M.A.; Locati, M. Induction and regulatory function of miR-9 in human monocytes and neutrophils exposed to proinflammatory signals. Proc. Natl. Acad. Sci. USA 2009, 106, 5282–5287. [Google Scholar] [CrossRef] [Green Version]
- Li, Q.; Zhang, D.; Wang, Y.; Sun, P.; Hou, X.; Larner, J.; Xiong, W.; Mi, J. MiR-21/Smad 7 signaling determines TGF-beta1-induced CAF formation. Sci. Rep. 2013, 3, 2038. [Google Scholar] [CrossRef] [PubMed]
- Dror, S.; Sander, L.; Schwartz, H.; Sheinboim, D.; Barzilai, A.; Dishon, Y.; Apcher, S.; Golan, T.; Greenberger, S.; Barshack, I.; et al. Melanoma miRNA trafficking controls tumour primary niche formation. Nat. Cell Biol. 2016, 18, 1006–1017. [Google Scholar] [CrossRef] [PubMed]
- Shu, S.; Yang, Y.; Allen, C.L.; Maguire, O.; Minderman, H.; Sen, A.; Ciesielski, M.J.; Collins, K.A.; Bush, P.J.; Singh, P.; et al. Metabolic reprogramming of stromal fibroblasts by melanoma exosome microRNA favours a pre-metastatic microenvironment. Sci. Rep. 2018, 8, 12905. [Google Scholar] [CrossRef] [Green Version]
- Zhou, X.; Yan, T.; Huang, C.; Xu, Z.; Wang, L.; Jiang, E.; Wang, H.; Chen, Y.; Liu, K.; Shao, Z.; et al. Melanoma cell-secreted exosomal miR-155-5p induce proangiogenic switch of cancer-associated fibroblasts via SOCS1/JAK2/STAT3 signaling pathway. J. Exp. Clin. Cancer Res. 2018, 37, 242. [Google Scholar] [CrossRef] [Green Version]
- Rodriguez, A.; Vigorito, E.; Clare, S.; Warren, M.V.; Couttet, P.; Soond, D.R.; van Dongen, S.; Grocock, R.J.; Das, P.P.; Miska, E.A.; et al. Requirement of bic/microRNA-155 for normal immune function. Science 2007, 316, 608–611. [Google Scholar] [CrossRef] [Green Version]
- Lu, C.; Huang, X.; Zhang, X.; Roensch, K.; Cao, Q.; Nakayama, K.I.; Blazar, B.R.; Zeng, Y.; Zhou, X. miR-221 and miR-155 regulate human dendritic cell development, apoptosis, and IL-12 production through targeting of p27kip1, KPC1, and SOCS-1. Blood 2011, 117, 4293–4303. [Google Scholar] [CrossRef] [Green Version]
- Sato, K.; Nagayama, H.; Tadokoro, K.; Juji, T.; Takahashi, T.A. Extracellular signal-regulated kinase, stress-activated protein kinase/c-Jun N-terminal kinase, and p38mapk are involved in IL-10-mediated selective repression of TNF-alpha-induced activation and maturation of human peripheral blood monocyte-derived dendritic cells. J. Immunol. 1999, 162, 3865–3872. [Google Scholar]
- Liang, X.; Liu, Y.; Mei, S.; Zhang, M.; Xin, J.; Zhang, Y.; Yang, R. MicroRNA-22 impairs anti-tumor ability of dendritic cells by targeting p38. PLoS ONE 2015, 10, e0121510. [Google Scholar] [CrossRef] [Green Version]
- Liang, X.; Shangguan, W.; Zhang, M.; Mei, S.; Wang, L.; Yang, R. miR-128 enhances dendritic cell-mediated anti-tumor immunity via targeting of p38. Mol. Med. Rep. 2017, 16, 1307–1313. [Google Scholar] [CrossRef] [Green Version]
- Cordeiro, B.; Jeon, P.; Boukhaled, G.M.; Corrado, M.; Lapohos, O.; Roy, D.G.; Williams, K.; Jones, R.G.; Gruenheid, S.; Sagan, S.M.; et al. MicroRNA-9 Fine-Tunes Dendritic Cell Function by Suppressing Negative Regulators in a Cell-Type-Specific Manner. Cell Rep. 2020, 31, 107585. [Google Scholar] [CrossRef] [PubMed]
- Tittarelli, A.; Navarrete, M.; Lizana, M.; Hofmann-Vega, F.; Salazar-Onfray, F. Hypoxic Melanoma Cells Deliver microRNAs to Dendritic Cells and Cytotoxic T Lymphocytes through Connexin-43 Channels. Int. J. Mol. Sci. 2020, 21, 7567. [Google Scholar] [CrossRef] [PubMed]
- Omar, H.A.; El-Serafi, A.T.; Hersi, F.; Arafa, E.A.; Zaher, D.M.; Madkour, M.; Arab, H.H.; Tolba, M.F. Immunomodulatory MicroRNAs in cancer: Targeting immune checkpoints and the tumor microenvironment. FEBS J. 2019, 286, 3540–3557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lin, R.; Chen, L.; Chen, G.; Hu, C.; Jiang, S.; Sevilla, J.; Wan, Y.; Sampson, J.H.; Zhu, B.; Li, Q.J. Targeting miR-23a in CD8+ cytotoxic T lymphocytes prevents tumor-dependent immunosuppression. J. Clin. Investig. 2014, 124, 5352–5367. [Google Scholar] [CrossRef]
- Li, Q.; Johnston, N.; Zheng, X.; Wang, H.; Zhang, X.; Gao, D.; Min, W. miR-28 modulates exhaustive differentiation of T cells through silencing programmed cell death-1 and regulating cytokine secretion. Oncotarget 2016, 7, 53735–53750. [Google Scholar] [CrossRef] [Green Version]
- Xi, J.; Huang, Q.; Wang, L.; Ma, X.; Deng, Q.; Kumar, M.; Zhou, Z.; Li, L.; Zeng, Z.; Young, K.H.; et al. miR-21 depletion in macrophages promotes tumoricidal polarization and enhances PD-1 immunotherapy. Oncogene 2018, 37, 3151–3165. [Google Scholar] [CrossRef]
- Mastroianni, J.; Stickel, N.; Andrlova, H.; Hanke, K.; Melchinger, W.; Duquesne, S.; Schmidt, D.; Falk, M.; Andrieux, G.; Pfeifer, D.; et al. miR-146a Controls Immune Response in the Melanoma Microenvironment. Cancer Res. 2019, 79, 183–195. [Google Scholar] [CrossRef] [Green Version]
- Marschner, D.; Falk, M.; Javorniczky, N.R.; Hanke-Muller, K.; Rawluk, J.; Schmitt-Graeff, A.; Simonetta, F.; Haring, E.; Dicks, S.; Ku, M.; et al. MicroRNA-146a regulates immune-related adverse events caused by immune checkpoint inhibitors. JCI Insight 2020, 5, e132334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ricciuti, B.; Genova, C.; De Giglio, A.; Bassanelli, M.; Dal Bello, M.G.; Metro, G.; Brambilla, M.; Baglivo, S.; Grossi, F.; Chiari, R. Impact of immune-related adverse events on survival in patients with advanced non-small cell lung cancer treated with nivolumab: Long-term outcomes from a multi-institutional analysis. J. Cancer Res. Clin. Oncol. 2019, 145, 479–485. [Google Scholar] [CrossRef] [PubMed]
- Indini, A.; Di Guardo, L.; Cimminiello, C.; Prisciandaro, M.; Randon, G.; De Braud, F.; Del Vecchio, M. Immune-related adverse events correlate with improved survival in patients undergoing anti-PD1 immunotherapy for metastatic melanoma. J. Cancer Res. Clin. Oncol. 2019, 145, 511–521. [Google Scholar] [CrossRef]
- Kaehler, K.C.; Piel, S.; Livingstone, E.; Schilling, B.; Hauschild, A.; Schadendorf, D. Update on immunologic therapy with anti-CTLA-4 antibodies in melanoma: Identification of clinical and biological response patterns, immune-related adverse events, and their management. Semin. Oncol. 2010, 37, 485–498. [Google Scholar] [CrossRef] [PubMed]
- Hsin, J.P.; Lu, Y.; Loeb, G.B.; Leslie, C.S.; Rudensky, A.Y. The effect of cellular context on miR-155-mediated gene regulation in four major immune cell types. Nat. Immunol. 2018, 19, 1137–1145. [Google Scholar] [CrossRef]
- Ji, Y.; Wrzesinski, C.; Yu, Z.; Hu, J.; Gautam, S.; Hawk, N.V.; Telford, W.G.; Palmer, D.C.; Franco, Z.; Sukumar, M.; et al. miR-155 augments CD8+ T-cell antitumor activity in lymphoreplete hosts by enhancing responsiveness to homeostatic gammac cytokines. Proc. Natl. Acad. Sci. USA 2015, 112, 476–481. [Google Scholar] [CrossRef] [Green Version]
- Ekiz, H.A.; Huffaker, T.B.; Grossmann, A.H.; Stephens, W.Z.; Williams, M.A.; Round, J.L.; O’Connell, R.M. MicroRNA-155 coordinates the immunological landscape within murine melanoma and correlates with immunity in human cancers. JCI Insight 2019, 4, e126543. [Google Scholar] [CrossRef]
- Huffaker, T.B.; Lee, S.H.; Tang, W.W.; Wallace, J.A.; Alexander, M.; Runtsch, M.C.; Larsen, D.K.; Thompson, J.; Ramstead, A.G.; Voth, W.P.; et al. Antitumor immunity is defective in T cell-specific microRNA-155-deficient mice and is rescued by immune checkpoint blockade. J. Biol. Chem. 2017, 292, 18530–18541. [Google Scholar] [CrossRef] [Green Version]
- Dudda, J.C.; Salaun, B.; Ji, Y.; Palmer, D.C.; Monnot, G.C.; Merck, E.; Boudousquie, C.; Utzschneider, D.T.; Escobar, T.M.; Perret, R.; et al. MicroRNA-155 is required for effector CD8+ T cell responses to virus infection and cancer. Immunity 2013, 38, 742–753. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Levati, L.; Pagani, E.; Romani, S.; Castiglia, D.; Piccinni, E.; Covaciu, C.; Caporaso, P.; Bondanza, S.; Antonetti, F.R.; Bonmassar, E.; et al. MicroRNA-155 targets the SKI gene in human melanoma cell lines. Pigment. Cell Melanoma Res. 2011, 24, 538–550. [Google Scholar] [CrossRef]
- DiSano, J.A.; Huffnagle, I.; Gowda, R.; Spiegelman, V.S.; Robertson, G.P.; Pameijer, C.R. Loss of miR-155 upregulates WEE1 in metastatic melanoma. Melanoma Res. 2019, 29, 216–219. [Google Scholar] [CrossRef]
- Li, H.; Song, J.B.; Chen, H.X.; Wang, Q.Q.; Meng, L.X.; Li, Y. MiR-155 inhibits proliferation, invasion and migration of melanoma via targeting CBL. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 9525–9534. [Google Scholar] [CrossRef] [PubMed]
- Yin, Q.; Wang, X.; McBride, J.; Fewell, C.; Flemington, E. B-cell receptor activation induces BIC/miR-155 expression through a conserved AP-1 element. J. Biol. Chem. 2008, 283, 2654–2662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, J.; Zhang, P.; Krishna, S.; Wang, J.; Lin, X.; Huang, H.; Xie, D.; Gorentla, B.; Huang, R.; Gao, J.; et al. Unexpected positive control of NFkappaB and miR-155 by DGKalpha and zeta ensures effector and memory CD8+ T cell differentiation. Oncotarget 2016, 7, 33744–33764. [Google Scholar] [CrossRef] [Green Version]
- Vignard, V.; Labbe, M.; Marec, N.; Andre-Gregoire, G.; Jouand, N.; Fonteneau, J.F.; Labarriere, N.; Fradin, D. MicroRNAs in Tumor Exosomes Drive Immune Escape in Melanoma. Cancer Immunol. Res. 2020, 8, 255–267. [Google Scholar] [CrossRef] [Green Version]
- Androulidaki, A.; Iliopoulos, D.; Arranz, A.; Doxaki, C.; Schworer, S.; Zacharioudaki, V.; Margioris, A.N.; Tsichlis, P.N.; Tsatsanis, C. The kinase Akt1 controls macrophage response to lipopolysaccharide by regulating microRNAs. Immunity 2009, 31, 220–231. [Google Scholar] [CrossRef] [Green Version]
- Nazari-Jahantigh, M.; Wei, Y.; Noels, H.; Akhtar, S.; Zhou, Z.; Koenen, R.R.; Heyll, K.; Gremse, F.; Kiessling, F.; Grommes, J.; et al. MicroRNA-155 promotes atherosclerosis by repressing Bcl6 in macrophages. J. Clin. Investig. 2012, 122, 4190–4202. [Google Scholar] [CrossRef] [Green Version]
- Martinez-Nunez, R.T.; Louafi, F.; Sanchez-Elsner, T. The interleukin 13 (IL-13) pathway in human macrophages is modulated by microRNA-155 via direct targeting of interleukin 13 receptor alpha1 (IL13Ralpha1). J. Biol. Chem. 2011, 286, 1786–1794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ruckerl, D.; Jenkins, S.J.; Laqtom, N.N.; Gallagher, I.J.; Sutherland, T.E.; Duncan, S.; Buck, A.H.; Allen, J.E. Induction of IL-4Ralpha-dependent microRNAs identifies PI3K/Akt signaling as essential for IL-4-driven murine macrophage proliferation in vivo. Blood 2012, 120, 2307–2316. [Google Scholar] [CrossRef]
- Ma, S.; Liu, M.; Xu, Z.; Li, Y.; Guo, H.; Ge, Y.; Liu, Y.; Zheng, D.; Shi, J. A double feedback loop mediated by microRNA-23a/27a/24-2 regulates M1 versus M2 macrophage polarization and thus regulates cancer progression. Oncotarget 2016, 7, 13502–13519. [Google Scholar] [CrossRef] [Green Version]
- Banerjee, S.; Cui, H.; Xie, N.; Tan, Z.; Yang, S.; Icyuz, M.; Thannickal, V.J.; Abraham, E.; Liu, G. miR-125a-5p regulates differential activation of macrophages and inflammation. J. Biol. Chem. 2013, 288, 35428–35436. [Google Scholar] [CrossRef] [Green Version]
- Gerloff, D.; Lutzkendorf, J.; Moritz, R.K.C.; Wersig, T.; Mader, K.; Muller, L.P.; Sunderkotter, C. Melanoma-Derived Exosomal miR-125b-5p Educates Tumor Associated Macrophages (TAMs) by Targeting Lysosomal Acid Lipase A (LIPA). Cancers 2020, 12, 464. [Google Scholar] [CrossRef] [Green Version]
- Yang, C.H.; Yue, J.; Pfeffer, S.R.; Handorf, C.R.; Pfeffer, L.M. MicroRNA miR-21 regulates the metastatic behavior of B16 melanoma cells. J. Biol. Chem. 2011, 286, 39172–39178. [Google Scholar] [CrossRef] [Green Version]
- Sahraei, M.; Chaube, B.; Liu, Y.; Sun, J.; Kaplan, A.; Price, N.L.; Ding, W.; Oyaghire, S.; Garcia-Milian, R.; Mehta, S.; et al. Suppressing miR-21 activity in tumor-associated macrophages promotes an antitumor immune response. J. Clin. Investig. 2019, 129, 5518–5536. [Google Scholar] [CrossRef] [Green Version]
- Mathsyaraja, H.; Thies, K.; Taffany, D.A.; Deighan, C.; Liu, T.; Yu, L.; Fernandez, S.A.; Shapiro, C.; Otero, J.; Timmers, C.; et al. CSF1-ETS2-induced microRNA in myeloid cells promote metastatic tumor growth. Oncogene 2015, 34, 3651–3661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, L.; Zhang, J.; Diao, W.; Wang, D.; Wei, Y.; Zhang, C.Y.; Zen, K. MicroRNA-155 and MicroRNA-21 promote the expansion of functional myeloid-derived suppressor cells. J. Immunol. 2014, 192, 1034–1043. [Google Scholar] [CrossRef]
- Chen, S.; Wang, L.; Fan, J.; Ye, C.; Dominguez, D.; Zhang, Y.; Curiel, T.J.; Fang, D.; Kuzel, T.M.; Zhang, B. Host miR155 promotes tumor growth through a myeloid-derived suppressor cell-dependent mechanism. Cancer Res. 2015, 75, 519–531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Y.; Lai, L.; Chen, Q.; Song, Y.; Xu, S.; Ma, F.; Wang, X.; Wang, J.; Yu, H.; Cao, X.; et al. MicroRNA-494 is required for the accumulation and functions of tumor-expanded myeloid-derived suppressor cells via targeting of PTEN. J. Immunol. 2012, 188, 5500–5510. [Google Scholar] [CrossRef] [PubMed]
- Cichocki, F.; Felices, M.; McCullar, V.; Presnell, S.R.; Al-Attar, A.; Lutz, C.T.; Miller, J.S. Cutting edge: microRNA-181 promotes human NK cell development by regulating Notch signaling. J. Immunol. 2011, 187, 6171–6175. [Google Scholar] [CrossRef] [Green Version]
- Heinemann, A.; Zhao, F.; Pechlivanis, S.; Eberle, J.; Steinle, A.; Diederichs, S.; Schadendorf, D.; Paschen, A. Tumor suppressive microRNAs miR-34a/c control cancer cell expression of ULBP2, a stress-induced ligand of the natural killer cell receptor NKG2D. Cancer Res. 2012, 72, 460–471. [Google Scholar] [CrossRef] [Green Version]
- Lazaridou, M.F.; Gonschorek, E.; Massa, C.; Friedrich, M.; Handke, D.; Mueller, A.; Jasinski-Bergner, S.; Dummer, R.; Koelblinger, P.; Seliger, B. Identification of miR-200a-5p targeting the peptide transporter TAP1 and its association with the clinical outcome of melanoma patients. Oncoimmunology 2020, 9, 1774323. [Google Scholar] [CrossRef] [PubMed]
- Chen, Z.; Ma, T.; Huang, C.; Hu, T.; Li, J. The pivotal role of microRNA-155 in the control of cancer. J. Cell Physiol. 2014, 229, 545–550. [Google Scholar] [CrossRef] [PubMed]
miRNA | Sample Source | Expression | Target Genes | Ref. |
---|---|---|---|---|
miR-222 | Tissue | Low in clinical benefit melanoma tissues received anti-CTLA-4 (ipilimumab) | ICAM1 | [55] |
let-7e miR-99b miR-125a miR-125b miR-146b | Plasma EVs | High expression of miRNA cluster reduced the overall survival and progression-free survival of the patients treated with anti-CTLA-4 (ipilimumab) and anti-PD-1 (nivolumab) | [57] | |
miR-106b miR-532-5p | Serum | Decrease in melanoma treated with anti-PD-1 (pembrolizumab) | [58] | |
miR-155 | Peripheral blood | Increase after treatment with anti-PD-1 | PTPN2 | [60] |
miR-16-5p miR-17-5p miR-20a-5p | Serum | High in serum from melanoma responded to anti-PD-1 (nivolumab or pembrolizumab) | [61] | |
miR-1972 miR-4502 | Serum | Increase in non-responders treated with anti-PD-1 (nivolumab or pembrolizumab) | [61] | |
miR-615-3p miR-1234-3p miR-4649-3p | Serum | Decrease in responders received anti-PD-1 (nivolumab or pembrolizumab) and anti-CTLA-4 (ipilimumab) or combination of ipilimumab and nivolumab | [62] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Nguyen, M.-H.T.; Luo, Y.-H.; Li, A.-L.; Tsai, J.-C.; Wu, K.-L.; Chung, P.-J.; Ma, N. miRNA as a Modulator of Immunotherapy and Immune Response in Melanoma. Biomolecules 2021, 11, 1648. https://doi.org/10.3390/biom11111648
Nguyen M-HT, Luo Y-H, Li A-L, Tsai J-C, Wu K-L, Chung P-J, Ma N. miRNA as a Modulator of Immunotherapy and Immune Response in Melanoma. Biomolecules. 2021; 11(11):1648. https://doi.org/10.3390/biom11111648
Chicago/Turabian StyleNguyen, Mai-Huong Thi, Yueh-Hsia Luo, An-Lun Li, Jen-Chieh Tsai, Kun-Lin Wu, Pei-Jung Chung, and Nianhan Ma. 2021. "miRNA as a Modulator of Immunotherapy and Immune Response in Melanoma" Biomolecules 11, no. 11: 1648. https://doi.org/10.3390/biom11111648
APA StyleNguyen, M.-H. T., Luo, Y.-H., Li, A.-L., Tsai, J.-C., Wu, K.-L., Chung, P.-J., & Ma, N. (2021). miRNA as a Modulator of Immunotherapy and Immune Response in Melanoma. Biomolecules, 11(11), 1648. https://doi.org/10.3390/biom11111648