miRNA-Based Therapeutics in the Era of Immune-Checkpoint Inhibitors
Abstract
:1. Introduction
2. miRNA Dysregulation in Cancer
3. Response Prediction to Immune-Checkpoint Inhibitors by miRNAs
3.1. Non-Small Cell Lung Cancer (NSCLC)
3.2. Gastric and Esophageal Cancer
3.3. Melanoma
4. miRNAs as Therapeutic Adjuvant for Immune-Checkpoint Inhibitors
4.1. Clinical Trials Using miRNA Based Approaches
4.2. Preclinical Investigations Using miRNA-Based Approaches
4.2.1. Mesothelioma
4.2.2. Melanoma
4.2.3. NSCLC
4.2.4. Glioblastoma
4.2.5. Head and Neck Squamous Cell Carcinoma (HNSCC)
4.2.6. Breast Cancer
4.2.7. Lymphoma
4.2.8. Prevention of Immune-Related Adverse Events
5. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Lee, R.C.; Feinbaum, R.L.; Ambros, V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993, 75, 843–854. [Google Scholar] [CrossRef]
- Wightman, B.; Ha, I.; Ruvkun, G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell 1993, 75, 855–862. [Google Scholar] [CrossRef]
- Reinhart, B.J.; Slack, F.J.; Basson, M.; Pasquinelli, A.E.; Bettinger, J.C.; Rougvie, A.E.; Horvitz, H.R.; Ruvkun, G. The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans. Nature 2000, 403, 901–906. [Google Scholar] [CrossRef] [PubMed]
- Lau, N.C.; Lim, L.P.; Weinstein, E.G.; Bartel, D.P. An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans. Science 2001, 294, 858–862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lagos-Quintana, M.; Rauhut, R.; Lendeckel, W.; Tuschl, T. Identification of novel genes coding for small expressed RNAs. Science 2001, 294, 853–858. [Google Scholar] [CrossRef] [Green Version]
- Kozomara, A.; Birgaoanu, M.; Griffiths-Jones, S. miRBase: From microRNA sequences to function. Nucleic Acids Res. 2019, 47, D155–D162. [Google Scholar] [CrossRef]
- Bhaskaran, M.; Mohan, M. MicroRNAs: History, biogenesis, and their evolving role in animal development and disease. Vet. Pathol. 2014, 51, 759–774. [Google Scholar] [CrossRef] [Green Version]
- Stavast, C.J.; Erkeland, S.J. The Non-Canonical Aspects of MicroRNAs: Many Roads to Gene Regulation. Cells 2019, 8, 1465. [Google Scholar] [CrossRef] [Green Version]
- Ranade, A.R.; Cherba, D.; Sridhar, S.; Richardson, P.; Webb, C.; Paripati, A.; Bowles, B.; Weiss, G.J. MicroRNA 92a-2*: A biomarker predictive for chemoresistance and prognostic for survival in patients with small cell lung cancer. J. Thorac. Oncol. 2010, 5, 1273–1278. [Google Scholar] [CrossRef] [Green Version]
- Kjersem, J.B.; Ikdahl, T.; Lingjaerde, O.C.; Guren, T.; Tveit, K.M.; Kure, E.H. Plasma microRNAs predicting clinical outcome in metastatic colorectal cancer patients receiving first-line oxaliplatin-based treatment. Mol. Oncol. 2014, 8, 59–67. [Google Scholar] [CrossRef]
- Rinnerthaler, G.; Gampenrieder, S.P.; Hackl, H.; Steiner, M.; Monzo-Fuentes, C.; Melchardt, T.; Magnes, T.; Huemer, F.; Westphal, T.; Hufnagl, C.; et al. Low Expression of miR-20a-5p Predicts Benefit to Bevacizumab in Metastatic Breast Cancer Patients Treated within the TANIA Phase III Trial. J. Clin. Med. 2020, 9, 1663. [Google Scholar] [CrossRef] [PubMed]
- Hansen, T.F.; Carlsen, A.L.; Heegaard, N.H.; Sorensen, F.B.; Jakobsen, A. Changes in circulating microRNA-126 during treatment with chemotherapy and bevacizumab predicts treatment response in patients with metastatic colorectal cancer. Br. J. Cancer 2015, 112, 624–629. [Google Scholar] [CrossRef] [PubMed]
- Halvorsen, A.R.; Sandhu, V.; Sprauten, M.; Flote, V.G.; Kure, E.H.; Brustugun, O.T.; Helland, A. Circulating microRNAs associated with prolonged overall survival in lung cancer patients treated with nivolumab. Acta Oncol. 2018, 57, 1225–1231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boeri, M.; Milione, M.; Proto, C.; Signorelli, D.; Lo Russo, G.; Galeone, C.; Verri, C.; Mensah, M.; Centonze, G.; Martinetti, A.; et al. Circulating miRNAs and PD-L1 Tumor Expression Are Associated with Survival in Advanced NSCLC Patients Treated with Immunotherapy: A Prospective Study. Clin. Cancer Res. 2019, 25, 2166–2173. [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]
- Cimmino, A.; Calin, G.A.; Fabbri, M.; Iorio, M.V.; Ferracin, M.; Shimizu, M.; Wojcik, S.E.; Aqeilan, R.I.; Zupo, S.; Dono, M.; et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc. Natl. Acad. Sci. USA 2005, 102, 13944–13949. [Google Scholar] [CrossRef] [Green Version]
- Pekarsky, Y.; Croce, C.M. Role of miR-15/16 in CLL. Cell Death Differ. 2015, 22, 6–11. [Google Scholar] [CrossRef] [Green Version]
- Lovat, F.; Fassan, M.; Gasparini, P.; Rizzotto, L.; Cascione, L.; Pizzi, M.; Vicentini, C.; Balatti, V.; Palmieri, D.; Costinean, S.; et al. miR-15b/16-2 deletion promotes B-cell malignancies. Proc. Natl. Acad. Sci. USA 2015, 112, 11636–11641. [Google Scholar] [CrossRef] [Green Version]
- Calin, G.A.; Sevignani, C.; Dumitru, C.D.; Hyslop, T.; Noch, E.; Yendamuri, S.; Shimizu, M.; Rattan, S.; Bullrich, F.; Negrini, M.; et al. Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc. Natl. Acad. Sci. USA 2004, 101, 2999–3004. [Google Scholar] [CrossRef] [Green Version]
- Volinia, S.; Calin, G.A.; Liu, C.G.; Ambs, S.; Cimmino, A.; Petrocca, F.; Visone, R.; Iorio, M.; Roldo, C.; Ferracin, M.; et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc. Natl. Acad. Sci. USA 2006, 103, 2257–2261. [Google Scholar] [CrossRef] [Green Version]
- Han, L.; Witmer, P.D.; Casey, E.; Valle, D.; Sukumar, S. DNA methylation regulates MicroRNA expression. Cancer Biol. Ther. 2007, 6, 1284–1288. [Google Scholar] [CrossRef]
- Saito, Y.; Jones, P.A. Epigenetic activation of tumor suppressor microRNAs in human cancer cells. Cell Cycle 2006, 5, 2220–2222. [Google Scholar] [CrossRef] [PubMed]
- Peng, Y.; Croce, C.M. The role of MicroRNAs in human cancer. Signal. Transduct. Target. Ther. 2016, 1, 15004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Correia de Sousa, M.; Gjorgjieva, M.; Dolicka, D.; Sobolewski, C.; Foti, M. Deciphering miRNAs’ Action through miRNA Editing. Int. J. Mol. Sci. 2019, 20, 6249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eisenberg, E.; Levanon, E.Y. A-to-I RNA editing—immune protector and transcriptome diversifier. Nat. Rev. Genet. 2018, 19, 473–490. [Google Scholar] [CrossRef] [PubMed]
- Tan, M.H.; Li, Q.; Shanmugam, R.; Piskol, R.; Kohler, J.; Young, A.N.; Liu, K.I.; Zhang, R.; Ramaswami, G.; Ariyoshi, K.; et al. Dynamic landscape and regulation of RNA editing in mammals. Nature 2017, 550, 249–254. [Google Scholar] [CrossRef]
- Paz-Yaacov, N.; Bazak, L.; Buchumenski, I.; Porath, H.T.; Danan-Gotthold, M.; Knisbacher, B.A.; Eisenberg, E.; Levanon, E.Y. Elevated RNA Editing Activity Is a Major Contributor to Transcriptomic Diversity in Tumors. Cell Rep. 2015, 13, 267–276. [Google Scholar] [CrossRef] [Green Version]
- Han, L.; Diao, L.; Yu, S.; Xu, X.; Li, J.; Zhang, R.; Yang, Y.; Werner, H.M.J.; Eterovic, A.K.; Yuan, Y.; et al. The Genomic Landscape and Clinical Relevance of A-to-I RNA Editing in Human Cancers. Cancer Cell 2015, 28, 515–528. [Google Scholar] [CrossRef] [Green Version]
- Gassner, F.J.; Zaborsky, N.; Buchumenski, I.; Levanon, E.Y.; Gatterbauer, M.; Schubert, M.; Rauscher, S.; Hebenstreit, D.; Nadeu, F.; Campo, E.; et al. RNA editing contributes to epitranscriptome diversity in chronic lymphocytic leukemia. Leukemia 2020. [Google Scholar] [CrossRef]
- Warf, M.B.; Shepherd, B.A.; Johnson, W.E.; Bass, B.L. Effects of ADARs on small RNA processing pathways in C. elegans. Genome Res. 2012, 22, 1488–1498. [Google Scholar] [CrossRef] [Green Version]
- Shoshan, E.; Mobley, A.K.; Braeuer, R.R.; Kamiya, T.; Huang, L.; Vasquez, M.E.; Salameh, A.; Lee, H.J.; Kim, S.J.; Ivan, C.; et al. Reduced adenosine-to-inosine miR-455-5p editing promotes melanoma growth and metastasis. Nat. Cell Biol. 2015, 17, 311–321. [Google Scholar] [CrossRef] [PubMed]
- Dyck, L.; Mills, K.H.G. Immune checkpoints and their inhibition in cancer and infectious diseases. Eur. J. Immunol. 2017, 47, 765–779. [Google Scholar] [CrossRef] [PubMed]
- Huemer, F.; Leisch, M.; Geisberger, R.; Melchardt, T.; Rinnerthaler, G.; Zaborsky, N.; Greil, R. Combination Strategies for Immune-Checkpoint Blockade and Response Prediction by Artificial Intelligence. Int. J. Mol. Sci. 2020, 21, 2856. [Google Scholar] [CrossRef] [PubMed]
- Hamid, O.; Robert, C.; Daud, A.; Hodi, F.S.; Hwu, W.J.; Kefford, R.; Wolchok, J.D.; Hersey, P.; Joseph, R.; Weber, J.S.; et al. Five-year survival outcomes for patients with advanced melanoma treated with pembrolizumab in KEYNOTE-001. Ann. Oncol. 2019, 30, 582–588. [Google Scholar] [CrossRef]
- Mok, T.S.K.; Wu, Y.L.; Kudaba, I.; Kowalski, D.M.; Cho, B.C.; Turna, H.Z.; Castro, G., Jr.; Srimuninnimit, V.; Laktionov, K.K.; Bondarenko, I.; et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): A randomised, open-label, controlled, phase 3 trial. Lancet 2019, 393, 1819–1830. [Google Scholar] [CrossRef]
- Burtness, B.; Harrington, K.J.; Greil, R.; Soulieres, D.; Tahara, M.; de Castro, G., Jr.; Psyrri, A.; Baste, N.; Neupane, P.; Bratland, A.; et al. Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): A randomised, open-label, phase 3 study. Lancet 2019, 394, 1915–1928. [Google Scholar] [CrossRef]
- Brahmer, J.; Reckamp, K.L.; Baas, P.; Crino, L.; Eberhardt, W.E.; Poddubskaya, E.; Antonia, S.; Pluzanski, A.; Vokes, E.E.; Holgado, E.; et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2015, 373, 123–135. [Google Scholar] [CrossRef] [Green Version]
- Borghaei, H.; Paz-Ares, L.; Horn, L.; Spigel, D.R.; Steins, M.; Ready, N.E.; Chow, L.Q.; Vokes, E.E.; Felip, E.; Holgado, E.; et al. Nivolumab versus Docetaxel in Advanced Nonsquamous Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2015, 373, 1627–1639. [Google Scholar] [CrossRef]
- Reck, M.; Rodriguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csoszi, T.; Fulop, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2016, 375, 1823–1833. [Google Scholar] [CrossRef] [Green Version]
- Herbst, R.S.; Giaccone, G.; de Marinis, F.; Reinmuth, N.; Vergnenegre, A.; Barrios, C.H.; Morise, M.; Felip, E.; Andric, Z.; Geater, S.; et al. Atezolizumab for First-Line Treatment of PD-L1-Selected Patients with NSCLC. N. Engl. J. Med. 2020, 383, 1328–1339. [Google Scholar] [CrossRef]
- Gandhi, L.; Rodriguez-Abreu, D.; Gadgeel, S.; Esteban, E.; Felip, E.; De Angelis, F.; Domine, M.; Clingan, P.; Hochmair, M.J.; Powell, S.F.; et al. Pembrolizumab plus Chemotherapy in Metastatic Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 378, 2078–2092. [Google Scholar] [CrossRef] [PubMed]
- Paz-Ares, L.; Luft, A.; Vicente, D.; Tafreshi, A.; Gumus, M.; Mazieres, J.; Hermes, B.; Cay Senler, F.; Csoszi, T.; Fulop, A.; et al. Pembrolizumab plus Chemotherapy for Squamous Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 379, 2040–2051. [Google Scholar] [CrossRef] [PubMed]
- Socinski, M.A.; Jotte, R.M.; Cappuzzo, F.; Orlandi, F.; Stroyakovskiy, D.; Nogami, N.; Rodriguez-Abreu, D.; Moro-Sibilot, D.; Thomas, C.A.; Barlesi, F.; et al. Atezolizumab for First-Line Treatment of Metastatic Nonsquamous NSCLC. N. Engl. J. Med. 2018, 378, 2288–2301. [Google Scholar] [CrossRef]
- Hellmann, M.D.; Ciuleanu, T.E.; Pluzanski, A.; Lee, J.S.; Otterson, G.A.; Audigier-Valette, C.; Minenza, E.; Linardou, H.; Burgers, S.; Salman, P.; et al. Nivolumab plus Ipilimumab in Lung Cancer with a High Tumor Mutational Burden. N. Engl. J. Med. 2018, 378, 2093–2104. [Google Scholar] [CrossRef]
- Georges, S.A.; Biery, M.C.; Kim, S.Y.; Schelter, J.M.; Guo, J.; Chang, A.N.; Jackson, A.L.; Carleton, M.O.; Linsley, P.S.; Cleary, M.A.; et al. Coordinated regulation of cell cycle transcripts by p53-Inducible microRNAs, miR-192 and miR-215. Cancer Res. 2008, 68, 10105–10112. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Xie, J.; Xu, X.; Wang, J.; Ao, F.; Wan, Y.; Zhu, Y. MicroRNA-548 down-regulates host antiviral response via direct targeting of IFN-lambda1. Protein Cell 2013, 4, 130–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shukuya, T.; Ghai, V.; Amann, J.M.; Okimoto, T.; Shilo, K.; Kim, T.K.; Wang, K.; Carbone, D.P. Circulating MicroRNAs and Extracellular Vesicle-Containing MicroRNAs as Response Biomarkers of Anti-programmed Cell Death Protein 1 or Programmed Death-Ligand 1 Therapy in NSCLC. J. Thorac. Oncol. 2020. [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]
- Lei, T.; Zhu, Y.; Jiang, C.; Wang, Y.; Fu, J.; Fan, Z.; Qin, H. MicroRNA-320 was downregulated in non-small cell lung cancer and inhibited cell proliferation, migration and invasion by targeting fatty acid synthase. Mol. Med. Rep. 2016, 14, 1255–1262. [Google Scholar] [CrossRef]
- Fortunato, O.; Borzi, C.; Milione, M.; Centonze, G.; Conte, D.; Boeri, M.; Verri, C.; Moro, M.; Facchinetti, F.; Andriani, F.; et al. Circulating mir-320a promotes immunosuppressive macrophages M2 phenotype associated with lung cancer risk. Int. J. Cancer 2019, 144, 2746–2761. [Google Scholar] [CrossRef] [Green Version]
- Zhu, Y.; Zhang, S.; Li, Z.; Wang, H.; Li, Z.; Hu, Y.; Chen, H.; Zhang, X.; Cui, L.; Zhang, J.; et al. miR-125b-5p and miR-99a-5p downregulate human gammadelta T-cell activation and cytotoxicity. Cell Mol. Immunol. 2019, 16, 112–125. [Google Scholar] [CrossRef]
- Sestini, S.; Boeri, M.; Marchiano, A.; Pelosi, G.; Galeone, C.; Verri, C.; Suatoni, P.; Sverzellati, N.; La Vecchia, C.; Sozzi, G.; et al. Circulating microRNA signature as liquid-biopsy to monitor lung cancer in low-dose computed tomography screening. Oncotarget 2015, 6, 32868–32877. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Genova, C.; Coco, S.; Rossi, G.; Longo, L.; Chiorino, G.; Ostano, P.; Guana, F.; Metro, G.; Baglivo, S.; Ludovini, V.; et al. An exosomal miRNA signature as predictor of benefit from immune checkpoint inhibitors in non-small cell lung cancer. ESMO Virtual Congr. 2020. [Google Scholar] [CrossRef]
- Fuchs, C.S.; Doi, T.; Jang, R.W.; Muro, K.; Satoh, T.; Machado, M.; Sun, W.; Jalal, S.I.; Shah, M.A.; Metges, J.P.; et al. Safety and Efficacy of Pembrolizumab Monotherapy in Patients With Previously Treated Advanced Gastric and Gastroesophageal Junction Cancer: Phase 2 Clinical KEYNOTE-059 Trial. JAMA Oncol. 2018, 4, e180013. [Google Scholar] [CrossRef] [PubMed]
- Le, D.T.; Durham, J.N.; Smith, K.N.; Wang, H.; Bartlett, B.R.; Aulakh, L.K.; Lu, S.; Kemberling, H.; Wilt, C.; Luber, B.S.; et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017, 357, 409–413. [Google Scholar] [CrossRef] [Green Version]
- Kato, K.; Cho, B.C.; Takahashi, M.; Okada, M.; Lin, C.Y.; Chin, K.; Kadowaki, S.; Ahn, M.J.; Hamamoto, Y.; Doki, Y.; et al. Nivolumab versus chemotherapy in patients with advanced oesophageal squamous cell carcinoma refractory or intolerant to previous chemotherapy (ATTRACTION-3): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019, 20, 1506–1517. [Google Scholar] [CrossRef]
- Kojima, T.; Shah, M.A.; Muro, K.; Francois, E.; Adenis, A.; Hsu, C.H.; Doi, T.; Moriwaki, T.; Kim, S.B.; Lee, S.H.; et al. Randomized Phase III KEYNOTE-181 Study of Pembrolizumab Versus Chemotherapy in Advanced Esophageal Cancer. J. Clin. Oncol. 2020. [Google Scholar] [CrossRef]
- Moehler, M.; Shitara, K.; Garrido, M.; Salman, P.; Shen, L.; Wyrwicz, L.; Yamaguchi, K.; Skoczylas, T.; Campos Bragagnoli, A.; Liu, T.; et al. LBA6_PR—Nivolumab (nivo) plus chemotherapy (chemo) versus chemo as first-line (1L) treatment for advanced gastric cancer/gastroesophageal junction cancer (GC/GEJC)/esophageal adenocarcinoma (EAC): First results of the CheckMate 649 study. Ann. Oncol. 2020, 31 (Suppl. S4). [Google Scholar] [CrossRef]
- Kato, K.; Sun, J.; Shah, M.A.; Enzinger, P.C.; Adenis, A.; Doi, T.; Kojima, T.; Metges, J.; Li, Z.; Kim, S.; et al. LBA8_PR—Pembrolizumab plus chemotherapy versus chemotherapy as first-line therapy in patients with advanced esophageal cancer: The phase 3 KEYNOTE-590 study. Ann. Oncol. 2020, 31 (Suppl. S4), S1142–S1215. [Google Scholar] [CrossRef]
- Kelly, R.J.; Ajani, J.A.; Kuzdzal, J.; Zander, T.; Van Cutsem, E.; Piessen, G.; Mendez, G.; Feliciano, J.L.; Motoyama, S.; Lièvre, A.; et al. LBA9_PR—Adjuvant nivolumab in resected esophageal or gastroesophageal junction cancer (EC/GEJC) following neoadjuvant chemoradiation therapy (CRT): First results of the CheckMate 577 study. Ann. Oncol. 2020, 31 (Suppl. S4), S1142–S1215. [Google Scholar] [CrossRef]
- Kudo, T.; Hamamoto, Y.; Kato, K.; Ura, T.; Kojima, T.; Tsushima, T.; Hironaka, S.; Hara, H.; Satoh, T.; Iwasa, S.; et al. Nivolumab treatment for oesophageal squamous-cell carcinoma: An open-label, multicentre, phase 2 trial. Lancet Oncol. 2017, 18, 631–639. [Google Scholar] [CrossRef]
- Sudo, K.; Kato, K.; Matsuzaki, J.; Takizawa, S.; Aoki, Y.; Shoji, H.; Iwasa, S.; Honma, Y.; Takashima, A.; Sakamoto, H.; et al. Identification of serum microRNAs predicting the response of esophageal squamous-cell carcinoma to nivolumab. Jpn. J. Clin. Oncol. 2020, 50, 114–121. [Google Scholar] [CrossRef]
- Kang, Y.K.; Boku, N.; Satoh, T.; Ryu, M.H.; Chao, Y.; Kato, K.; Chung, H.C.; Chen, J.S.; Muro, K.; Kang, W.K.; et al. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017, 390, 2461–2471. [Google Scholar] [CrossRef]
- Miyamoto, T.; Kato, K.; Matsuzaki, J.; Takizawa, S.; Sudo, K.; Shoji, H.; Iwasa, S.; Honma, Y.; Takashima, A.; Okita, N.; et al. 182—Identification of serum microRNAs predicting the response to nivolumab in patients with advanced gastric cancer. Ann. Oncol. 2018, 29 (Suppl. S10), x1–x10. [Google Scholar] [CrossRef]
- 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] [PubMed]
- 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]
- Robert, C.; Schachter, J.; Long, G.V.; Arance, A.; Grob, J.J.; Mortier, L.; Daud, A.; Carlino, M.S.; McNeil, C.; Lotem, M.; et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2015, 372, 2521–2532. [Google Scholar] [CrossRef] [PubMed]
- Gutzmer, R.; Stroyakovskiy, D.; Gogas, H.; Robert, C.; Lewis, K.; Protsenko, S.; Pereira, R.P.; Eigentler, T.; Rutkowski, P.; Demidov, L.; et al. Atezolizumab, vemurafenib, and cobimetinib as first-line treatment for unresectable advanced BRAF(V600) mutation-positive melanoma (IMspire150): Primary analysis of the randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2020, 395, 1835–1844. [Google Scholar] [CrossRef]
- National Comprehensive Cancer Network. Cutaneous Melanoma (Version 4.2020). Available online: https://www.nccn.org/professionals/physician_gls/pdf/cutaneous_melanoma.pdf (accessed on 21 November 2020).
- Weber, J.; Mandala, M.; Del Vecchio, M.; Gogas, H.J.; Arance, A.M.; Cowey, C.L.; Dalle, S.; Schenker, M.; Chiarion-Sileni, V.; Marquez-Rodas, I.; et al. Adjuvant Nivolumab versus Ipilimumab in Resected Stage III or IV Melanoma. N. Engl. J. Med. 2017, 377, 1824–1835. [Google Scholar] [CrossRef]
- Eggermont, A.M.M.; Blank, C.U.; Mandala, M.; Long, G.V.; Atkinson, V.; Dalle, S.; Haydon, A.; Lichinitser, M.; Khattak, A.; Carlino, M.S.; et al. Adjuvant Pembrolizumab versus Placebo in Resected Stage III Melanoma. N. Engl. J. Med. 2018, 378, 1789–1801. [Google Scholar] [CrossRef]
- 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]
- Van Laar, R.; Lincoln, M.; Van Laar, B. Development and validation of a plasma-based melanoma biomarker suitable for clinical use. Br. J. Cancer 2018, 118, 857–866. [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]
- Sozzi, G.; Boeri, M.; Rossi, M.; Verri, C.; Suatoni, P.; Bravi, F.; Roz, L.; Conte, D.; Grassi, M.; Sverzellati, N.; et al. Clinical utility of a plasma-based miRNA signature classifier within computed tomography lung cancer screening: A correlative MILD trial study. J. Clin. Oncol. 2014, 32, 768–773. [Google Scholar] [CrossRef] [PubMed]
- Mollaei, H.; Safaralizadeh, R.; Rostami, Z. MicroRNA replacement therapy in cancer. J. Cell Physiol. 2019, 234, 12369–12384. [Google Scholar] [CrossRef] [PubMed]
- Beg, M.S.; Brenner, A.J.; Sachdev, J.; Borad, M.; Kang, Y.K.; Stoudemire, J.; Smith, S.; Bader, A.G.; Kim, S.; Hong, D.S. Phase I study of MRX34, a liposomal miR-34a mimic, administered twice weekly in patients with advanced solid tumors. Investig. New Drugs 2017, 35, 180–188. [Google Scholar] [CrossRef] [PubMed]
- van Zandwijk, N.; Pavlakis, N.; Kao, S.C.; Linton, A.; Boyer, M.J.; Clarke, S.; Huynh, Y.; Chrzanowska, A.; Fulham, M.J.; Bailey, D.L.; et al. Safety and activity of microRNA-loaded minicells in patients with recurrent malignant pleural mesothelioma: A first-in-man, phase 1, open-label, dose-escalation study. Lancet Oncol. 2017, 18, 1386–1396. [Google Scholar] [CrossRef]
- Seto, A.G.; Beatty, X.; Lynch, J.M.; Hermreck, M.; Tetzlaff, M.; Duvic, M.; Jackson, A.L. Cobomarsen, an oligonucleotide inhibitor of miR-155, co-ordinately regulates multiple survival pathways to reduce cellular proliferation and survival in cutaneous T-cell lymphoma. Br. J. Haematol. 2018, 183, 428–444. [Google Scholar] [CrossRef] [Green Version]
- Anastasiadou, E.; Seto, A.; Beatty, X.; Hermreck, M.; Gilles, M.E.; Stroopinsky, D.; Pinter-Brown, L.C.; Pestano, L.; Marchese, C.; Avigan, D.; et al. Cobomarsen, an oligonucleotide inhibitor of miR-155, slows DLBCL tumor cell growth in vitro and in vivo. Clin. Cancer Res. 2020. [Google Scholar] [CrossRef]
- Wright, K. FDA Approves Nivolumab Plus Ipilimumab for Previously Untreated Unresectable Malignant Pleural Mesothelioma. Oncology (Williston Park) 2020, 34, 502–503. [Google Scholar] [CrossRef]
- Kao, S.C.; Cheng, Y.Y.; Williams, M.; Kirschner, M.B.; Madore, J.; Lum, T.; Sarun, K.H.; Linton, A.; McCaughan, B.; Klebe, S.; et al. Tumor Suppressor microRNAs Contribute to the Regulation of PD-L1 Expression in Malignant Pleural Mesothelioma. J. Thorac. Oncol. 2017, 12, 1421–1433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jia, X.; Li, X.; Shen, Y.; Miao, J.; Liu, H.; Li, G.; Wang, Z. MiR-16 regulates mouse peritoneal macrophage polarization and affects T-cell activation. J. Cell Mol. Med. 2016, 20, 1898–1907. [Google Scholar] [CrossRef] [PubMed] [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] [PubMed] [Green Version]
- 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]
- 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]
- Zhang, P.F.; Pei, X.; Li, K.S.; Jin, L.N.; Wang, F.; Wu, J.; Zhang, X.M. Circular RNA circFGFR1 promotes progression and anti-PD-1 resistance by sponging miR-381-3p in non-small cell lung cancer cells. Mol. Cancer 2019, 18, 179. [Google Scholar] [CrossRef] [Green Version]
- Chen, I.X.; Chauhan, V.P.; Posada, J.; Ng, M.R.; Wu, M.W.; Adstamongkonkul, P.; Huang, P.; Lindeman, N.; Langer, R.; Jain, R.K. Blocking CXCR4 alleviates desmoplasia, increases T-lymphocyte infiltration, and improves immunotherapy in metastatic breast cancer. Proc. Natl. Acad. Sci. USA 2019, 116, 4558–4566. [Google Scholar] [CrossRef] [Green Version]
- Seo, Y.D.; Jiang, X.; Sullivan, K.M.; Jalikis, F.G.; Smythe, K.S.; Abbasi, A.; Vignali, M.; Park, J.O.; Daniel, S.K.; Pollack, S.M.; et al. Mobilization of CD8(+) T Cells via CXCR4 Blockade Facilitates PD-1 Checkpoint Therapy in Human Pancreatic Cancer. Clin. Cancer Res. 2019, 25, 3934–3945. [Google Scholar] [CrossRef] [Green Version]
- Reardon, D.A.; Brandes, A.A.; Omuro, A.; Mulholland, P.; Lim, M.; Wick, A.; Baehring, J.; Ahluwalia, M.S.; Roth, P.; Bahr, O.; et al. Effect of Nivolumab vs Bevacizumab in Patients With Recurrent Glioblastoma: The CheckMate 143 Phase 3 Randomized Clinical Trial. JAMA Oncol. 2020, 6, 1003–1010. [Google Scholar] [CrossRef]
- Wei, J.; Wang, F.; Kong, L.Y.; Xu, S.; Doucette, T.; Ferguson, S.D.; Yang, Y.; McEnery, K.; Jethwa, K.; Gjyshi, O.; et al. miR-124 inhibits STAT3 signaling to enhance T cell-mediated immune clearance of glioma. Cancer Res. 2013, 73, 3913–3926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wei, J.; Nduom, E.K.; Kong, L.Y.; Hashimoto, Y.; Xu, S.; Gabrusiewicz, K.; Ling, X.; Huang, N.; Qiao, W.; Zhou, S.; et al. MiR-138 exerts anti-glioma efficacy by targeting immune checkpoints. Neuro Oncol. 2016, 18, 639–648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, L.; Yu, H.; Yi, S.; Peng, X.; Su, P.; Xiao, Z.; Liu, R.; Tang, A.; Li, X.; Liu, F.; et al. The tumor suppressor miR-138-5p targets PD-L1 in colorectal cancer. Oncotarget 2016, 7, 45370–45384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ferris, R.L.; Blumenschein, G., Jr.; Fayette, J.; Guigay, J.; Colevas, A.D.; Licitra, L.; Harrington, K.; Kasper, S.; Vokes, E.E.; Even, C.; et al. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N. Engl. J. Med. 2016, 375, 1856–1867. [Google Scholar] [CrossRef] [PubMed]
- Cohen, E.E.W.; Soulieres, D.; Le Tourneau, C.; Dinis, J.; Licitra, L.; Ahn, M.J.; Soria, A.; Machiels, J.P.; Mach, N.; Mehra, R.; et al. Pembrolizumab versus methotrexate, docetaxel, or cetuximab for recurrent or metastatic head-and-neck squamous cell carcinoma (KEYNOTE-040): A randomised, open-label, phase 3 study. Lancet 2019, 393, 156–167. [Google Scholar] [CrossRef]
- Yu, D.; Liu, X.; Han, G.; Liu, Y.; Zhao, X.; Wang, D.; Bian, X.; Gu, T.; Wen, L. The let-7 family of microRNAs suppresses immune evasion in head and neck squamous cell carcinoma by promoting PD-L1 degradation. Cell Commun. Signal. 2019, 17, 173. [Google Scholar] [CrossRef] [Green Version]
- Schmid, P.; Adams, S.; Rugo, H.S.; Schneeweiss, A.; Barrios, C.H.; Iwata, H.; Dieras, V.; Hegg, R.; Im, S.A.; Shaw Wright, G.; et al. Atezolizumab and Nab-Paclitaxel in Advanced Triple-Negative Breast Cancer. N. Engl. J. Med. 2018, 379, 2108–2121. [Google Scholar] [CrossRef]
- Schmid, P.; Rugo, H.S.; Adams, S.; Schneeweiss, A.; Barrios, C.H.; Iwata, H.; Dieras, V.; Henschel, V.; Molinero, L.; Chui, S.Y.; et al. Atezolizumab plus nab-paclitaxel as first-line treatment for unresectable, locally advanced or metastatic triple-negative breast cancer (IMpassion130): Updated efficacy results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2020, 21, 44–59. [Google Scholar] [CrossRef]
- Zhang, M.; Gao, D.; Shi, Y.; Wang, Y.; Joshi, R.; Yu, Q.; Liu, D.; Alotaibi, F.; Zhang, Y.; Wang, H.; et al. miR-149-3p reverses CD8(+) T-cell exhaustion by reducing inhibitory receptors and promoting cytokine secretion in breast cancer cells. Open Biol. 2019, 9, 190061. [Google Scholar] [CrossRef] [Green Version]
- Huang, Z.; Gan, J.; Long, Z.; Guo, G.; Shi, X.; Wang, C.; Zang, Y.; Ding, Z.; Chen, J.; Zhang, J.; et al. Targeted delivery of let-7b to reprogramme tumor-associated macrophages and tumor infiltrating dendritic cells for tumor rejection. Biomaterials 2016, 90, 72–84. [Google Scholar] [CrossRef]
- Zheng, Z.; Sun, R.; Zhao, H.J.; Fu, D.; Zhong, H.J.; Weng, X.Q.; Qu, B.; Zhao, Y.; Wang, L.; Zhao, W.L. MiR155 sensitized B-lymphoma cells to anti-PD-L1 antibody via PD-1/PD-L1-mediated lymphoma cell interaction with CD8+T cells. Mol. Cancer 2019, 18, 54. [Google Scholar] [CrossRef] [PubMed]
- 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]
Author | Year | Tumor Type | Patients | ICI | ICI Therapy Line | miRNAs | Validation | Outcome |
---|---|---|---|---|---|---|---|---|
Halvorsen et al. [13] | 2018 | NSCLC (whole plasma) | 20 | anti-PD-1 | ≥2nd line | miR-215-5p , miR-411-3p (NA), miR-493-5p , miR-494-3p , miR-495-3p , miR-548j-5p , miR-93-3p | yes | OS |
Peng et al. [48] | 2019 | NSCLC (plasma EV) | 16 | anti-PD-1, anti-PD-L1 | ≥1st line | miR-320d , miR-320c , miR-320b , miR-125-5p | no | response |
Boeri et al. [14] | 2019 | NSCLC (whole plasma) | 111 | anti-PD-1, anti-PD-L1, anti-PD-L1+ anti-CTLA-4 | ≥1st line | miR-101-3p, miR-106a-5p, miR-126-5p, miR-133a, miR-140-3p, miR-140-5p, miR-142-3p, miR-145-5p, miR-148a-3p, miR-15b-5p, miR-16-5p, miR-17-5p, miR-197-3p, miR-19b-3p, miR-21-5p, miR-221-3p, miR-28-3p, miR-30b-5p, miR-30c-5p, miR-320a, miR-451a, miR-486-5p, miR-660-5p, miR-92a-3p (miRNA ratios were used to obtain miRNA signatures and in turn to calculate the MSC risk: low, intermediate and high [75]) | no | response, PFS, OS |
Genova et al. [53] | 2020 | NSCLC (plasma EV) | 174 | anti-PD-1 | ≥2nd line | miR-208a-5p ,miR-574-5p | yes | OS |
Shukuya et al. [47] | 2020 | NSCLC (whole plasma and plasma EV) | 29 | anti-PD-1, anti-PD-L1 | NA | miR-548am-5p , miR-200a-3p , miR-4707-3p , miR-335-3p , miR-429-3p , miR-200b-3p , miR-191-3p , miR-1277-3p , miR-200c-3p , miR-28-5p , miR-3120-3p , miR-152-3p , miR-335-5p , miR-199a-1-3p , miR-22-5p , miR-30e-3p , miR-33a-5p , miR-556-5p , miR-21-3p , miR-30d-3p , miR-130b-5p , miR-24-1-3p , miR-3138-3p , miR-548ax-5p , miR-6791-3p , miR-1287-5p , miR-3074-5p , miR-103a-1-3p , miR-21-5p , miR-130b-3p , miR-186-5p , miR-660-3p , miR-1246-5p , miR-1296-5p , miR-4707-3p , miR-1229-3p , miR-874-3p , miR-378c-5p , miR-1468-5p | yes | response |
Sudo et al. [62] | 2020 | ESSC (whole plasma) | 19 | anti-PD-1 | ≥2nd line | miR-1233-5p , miR-6885-5p , miR-4698 , miR-128-2-5p | no | response |
Miyamoto et al. [64] | 2018 | GC (whole plasma and plasma EV) | 20 | anti-PD-1 | ≥3rd line | two miRNAs termed “miR-A” and “miR-B” (not further specified by authors) | no | response, PFS |
Huber et al. [15] | 2018 | Melanoma (whole plasma and plasma EV) | 49 | anti-PD-1, anti-CTLA-4 | ≥1st line | let-7e , miR-125a , miR-99b , miR-146b , miR-125b | no | OS, PFS |
Galore-Haskel et al. [74] | 2015 | Melanoma (tumor tissue) | 13 | anti-CTLA-4 | ≥1st line | miR-222 | yes | response |
miRNA | Tumor Type | Experimental Setting | Outcome | Author |
---|---|---|---|---|
miR-16 | Healthy tissue | Mouse model | M1 macrophage differentiation, T cell activation and downregulation of PD-L1 following viral miR-16 transfection | Jia et al. [83] |
Mesothelioma | Humans (phase I) | 3% PR, 55% SD with a miR-16 based mimic | Van Zandwijk. [78] | |
miR-21 | Melanoma | Mouse model | Inhibition of M2 macrophage differentiation and increased tumor cell killing in combination with ICI in miR-21 deficient mice | Xi et al. [84] |
miR-28 | Melanoma | Mouse model | Restoration of T cell function and increased cytokine production following transfection with a miR-28 mimic | Li et al. [86] |
miR-34a | Advanced tumors | Humans (Phase I) | 2% PR, 12% SD with a liposomal miR-34a mimic | Beg et al. [77] |
miR-124 | Glioblastoma | Mouse model | Increased IFN-γ production by T cells following treatment with a miR-124 mimic | Wei et al. [92] |
miR-138 | Glioblastoma | Mouse model | Decreased expression of T cell exhaustion markers following treatment with a miR-138 mimic | Wei et al. [93] |
CRC | Mouse model | PD-L1 downregulation by miR-138-5p | Zhao et al. [94] | |
miR-146a | Melanoma | Mouse model | Increased IFN-γ expression levels in miR-146a deficient mice Increased ICI sensitivity with combined miR-146a antagomiR treatment | Mastroianni et al. [85] |
miR-149-3 | TNBC | Mouse model | Reversal of T cell exhaustion and increased secretion of effector cytokines after transfection with a miR-149-3 mimic | Zhang et al. [100] |
miR-155 | Melanoma | Mouse model | miR-155 triggers M1 macrophage differentiation | Huffaker et al. [87] |
DLBCL | In vitro | Increased sensitivity to ICI after transfection with a miR-155 mimic | Zheng et al. [102] | |
miR-381-3p | NSCLC | Mouse model | Decreased CXCR4 expression and increased ICI sensitivity by miR-381-3p | Zhang et al. [88] |
let-7a/b | HNSCC | Mouse model | CD8+ T cell infiltration and cytokine production by combining let-7a/b mimics with ICI | Yu et al. [97] |
TNBC | Mouse model | Reversal of tumor-suppressive properties of TAMs following delivery of let-7b mimics | Huang et al. [101] |
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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Huemer, F.; Leisch, M.; Geisberger, R.; Zaborsky, N.; Greil, R. miRNA-Based Therapeutics in the Era of Immune-Checkpoint Inhibitors. Pharmaceuticals 2021, 14, 89. https://doi.org/10.3390/ph14020089
Huemer F, Leisch M, Geisberger R, Zaborsky N, Greil R. miRNA-Based Therapeutics in the Era of Immune-Checkpoint Inhibitors. Pharmaceuticals. 2021; 14(2):89. https://doi.org/10.3390/ph14020089
Chicago/Turabian StyleHuemer, Florian, Michael Leisch, Roland Geisberger, Nadja Zaborsky, and Richard Greil. 2021. "miRNA-Based Therapeutics in the Era of Immune-Checkpoint Inhibitors" Pharmaceuticals 14, no. 2: 89. https://doi.org/10.3390/ph14020089
APA StyleHuemer, F., Leisch, M., Geisberger, R., Zaborsky, N., & Greil, R. (2021). miRNA-Based Therapeutics in the Era of Immune-Checkpoint Inhibitors. Pharmaceuticals, 14(2), 89. https://doi.org/10.3390/ph14020089