A Complex Metabolic Network Confers Immunosuppressive Functions to Myeloid-Derived Suppressor Cells (MDSCs) within the Tumour Microenvironment
Abstract
:1. Introduction
2. Ontogeny, Phenotype, and Main Characteristics of MDSCs
3. Metabolic Mechanisms of Immune Suppression by MDSCs
3.1. Glucose Metabolism
3.2. Amino Acid Metabolism
3.3. Lipid Metabolism
4. MDSC Targeting as a Therapeutic Approach in Cancer
4.1. Current MDSC-Targeting Approaches
- MDSC depletion. The use of conventional chemotherapy drugs such as gemcitabine, 5-fluoruracil, paclitaxel, and doxorubicin demonstrated the ability of these compounds to restore the anti-tumour immune response by controlling the accumulation of MDSCs in both tumour-bearing mice [92] and patients with cancer [60,61]. More recently, “peptibodies” consisting of S100A9-derived peptides conjugated to antibody Fc fragments have exhibited the ability to regulate MDSC expansion in tumour-bearing mice [210], and the use of antibodies recognising CD33 (a surface marker expressed by human MDSCs) conjugated with ozogamicin was able to affect myeloid cells accumulation in patients with acute promyelocytic leukaemia [211]. Therefore, this myeloid-cell-targeting approach may be exploited in solid cancer settings to block MDSCs. Ongoing clinical trials are also evaluating the therapeutic effects of modulating growth factors such as G-CSF (NCT02961257) or M-CSF (NCT02880371, NCT02554812, and NCT02777710, Table 2) to improve the effectiveness of cancer therapy by preventing MDSC expansion and accumulation.
- Inhibition of MDSC recruitment to the tumour site. Interrupting the chemokine and chemokine receptor axis is a rational strategy to prevent MDSC trafficking. Indeed, therapeutic blockade of CCL2-CCR2 interaction using CCL2 neutralising antibodies or a CCR2 antagonist as well as blocking the heterotypic CXCL5-CXCR2 signalling circuit produces promising results in several preclinical cancer models by affecting MDSC accumulation and restoring anti-tumour immunity [212,213,214]. Interestingly, low doses of methyltransferase and histone deacetylase inhibitors affect MDSC accumulation in lung premetastatic niches by affecting CCR2- and CXCR2-dependent pathways [215]. Currently, ongoing clinical trials are testing the therapeutic impact of targeting the CC or CXC axis to block MDSC trafficking as a single therapy (NCT01349036) or in combination with either chemotherapy (NCT02370238) or ICI-based therapy (NCT03177187 and NCT03161431, Table 2).
- Blocking MDSC differentiation into mature suppressive cells. One promising therapeutic approach depends upon the conversion of MDSCs into non-suppressive elements of the TME. Early studies demonstrated that all trans-retinoic acid (ATRA), a derivative of vitamin A, possesses the potential to force the differentiation of MDSCs into mature granulocytes and upregulate glutathione synthase (GSS), one of the enzymes required for glutathione synthesis [216]. As the enforced expression of IRF8, either directly or indirectly, may be a potential strategy to favour MDSC reprogramming, several strategies are able to sustain IRF8-dependent signatures such as STAT3 inhibition [217]. Sorafenib, a multikinase inhibitor, was able to reverse the immunosuppressive cytokine profile in tumour-infiltrating myeloid cells, thus promoting them to elicit a more robust anti-tumour immune response [218]. A phase II clinical trial is currently investigating the impact of ATRA in combination with ICI-based therapy to decrease the immunosuppressive activity of MDSCs in patients with melanoma compared to that in patients treated with immunotherapy alone (NCT02403778).
- Inhibition of MDSC-suppressive activity. Impeding MDSC immunosuppression by targeting key enzymes, signal transduction molecules, or transcription factors involved in the MDSC metabolic network is likely the most effective strategy to enforce cancer immunotherapy. Here, we report only some example strategies based on pivotal interfering metabolism-associated molecules that have not been discussed in detail in the previous sections.
4.2. New Perspectives in MDSC-Targeting Approaches
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Garraway, L.A.; Lander, E.S. Lessons from the cancer genome. Cell 2013, 153, 17–37. [Google Scholar] [CrossRef] [Green Version]
- Zahir, N.; Sun, R.; Gallahan, D.; Gatenby, R.A.; Curtis, C. Characterizing the ecological and evolutionary dynamics of cancer. Nat. Genet. 2020, 52, 759–767. [Google Scholar] [CrossRef]
- Keenan, T.E.; Burke, K.P.; Van Allen, E.M. Genomic correlates of response to immune checkpoint blockade. Nat. Med. 2019, 25, 389–402. [Google Scholar] [CrossRef] [PubMed]
- Luksza, M.; Riaz, N.; Makarov, V.; Balachandran, V.P.; Hellmann, M.D.; Solovyov, A.; Rizvi, N.A.; Merghoub, T.; Levine, A.J.; Chan, T.A.; et al. A neoantigen fitness model predicts tumour response to checkpoint blockade immunotherapy. Nature 2017, 551, 517–520. [Google Scholar] [CrossRef] [PubMed]
- Balachandran, V.P.; Luksza, M.; Zhao, J.N.; Makarov, V.; Moral, J.A.; Remark, R.; Herbst, B.; Askan, G.; Bhanot, U.; Senbabaoglu, Y.; et al. Identification of unique neoantigen qualities in long-term survivors of pancreatic cancer. Nature 2017, 551, 512–516. [Google Scholar] [CrossRef]
- Pylayeva-Gupta, Y.; Lee, K.E.; Hajdu, C.H.; Miller, G.; Bar-Sagi, D. Oncogenic Kras-induced GM-CSF production promotes the development of pancreatic neoplasia. Cancer Cell 2012, 21, 836–847. [Google Scholar] [CrossRef] [Green Version]
- Vitale, I.; Manic, G.; Coussens, L.M.; Kroemer, G.; Galluzzi, L. Macrophages and Metabolism in the Tumor Microenvironment. Cell Metab. 2019, 30, 36–50. [Google Scholar] [CrossRef]
- Calcinotto, A.; Spataro, C.; Zagato, E.; Di Mitri, D.; Gil, V.; Crespo, M.; De Bernardis, G.; Losa, M.; Mirenda, M.; Pasquini, E.; et al. IL-23 secreted by myeloid cells drives castration-resistant prostate cancer. Nature 2018, 559, 363–369. [Google Scholar] [CrossRef] [PubMed]
- Bronte, V.; Cingarlini, S.; Marigo, I.; De Santo, C.; Gallina, G.; Dolcetti, L.; Ugel, S.; Peranzoni, E.; Mandruzzato, S.; Zanovello, P. Leukocyte infiltration in cancer creates an unfavorable environment for antitumor immune responses: A novel target for therapeutic intervention. Immunol. Investig. 2006, 35, 327–357. [Google Scholar] [CrossRef]
- Murciano-Goroff, Y.R.; Warner, A.B.; Wolchok, J.D. The future of cancer immunotherapy: Microenvironment-targeting combinations. Cell Res. 2020, 30, 507–519. [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]
- Hegde, P.S.; Chen, D.S. Top 10 Challenges in Cancer Immunotherapy. Immunity 2020, 52, 17–35. [Google Scholar] [CrossRef] [PubMed]
- Bindea, G.; Mlecnik, B.; Tosolini, M.; Kirilovsky, A.; Waldner, M.; Obenauf, A.C.; Angell, H.; Fredriksen, T.; Lafontaine, L.; Berger, A.; et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity 2013, 39, 782–795. [Google Scholar] [CrossRef] [Green Version]
- Chen, D.S.; Mellman, I. Elements of cancer immunity and the cancer-immune set point. Nature 2017, 541, 321–330. [Google Scholar] [CrossRef] [PubMed]
- McGranahan, N.; Furness, A.J.; Rosenthal, R.; Ramskov, S.; Lyngaa, R.; Saini, S.K.; Jamal-Hanjani, M.; Wilson, G.A.; Birkbak, N.J.; Hiley, C.T.; et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016, 351, 1463–1469. [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]
- Yarchoan, M.; Johnson, B.A., 3rd; Lutz, E.R.; Laheru, D.A.; Jaffee, E.M. Targeting neoantigens to augment antitumour immunity. Nat. Rev. Cancer 2017, 17, 209–222. [Google Scholar] [CrossRef]
- 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] [PubMed] [Green Version]
- Galon, J.; Bruni, D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat. Rev. Drug Discov. 2019, 18, 197–218. [Google Scholar] [CrossRef]
- De Olza, M.O.; Rodrigo, B.N.; Zimmermann, S.; Coukos, G. Turning up the heat on non-immunoreactive tumours: Opportunities for clinical development. Lancet Oncol. 2020, 21, e419–e430. [Google Scholar] [CrossRef]
- Joyce, J.A.; Fearon, D.T. T cell exclusion, immune privilege, and the tumor microenvironment. Science 2015, 348, 74–80. [Google Scholar] [CrossRef] [Green Version]
- Schurch, C.M.; Bhate, S.S.; Barlow, G.L.; Phillips, D.J.; Noti, L.; Zlobec, I.; Chu, P.; Black, S.; Demeter, J.; McIlwain, D.R.; et al. Coordinated Cellular Neighborhoods Orchestrate Antitumoral Immunity at the Colorectal Cancer Invasive Front. Cell 2020, 182, 1341–1359.e19. [Google Scholar] [CrossRef] [PubMed]
- Bagaev, A.; Kotlov, N.; Nomie, K.; Svekolkin, V.; Gafurov, A.; Isaeva, O.; Osokin, N.; Kozlov, I.; Frenkel, F.; Gancharova, O.; et al. Conserved pan-cancer microenvironment subtypes predict response to immunotherapy. Cancer Cell 2021, 39, 845–865.e7. [Google Scholar] [CrossRef] [PubMed]
- Huang, Y.; Kim, B.Y.S.; Chan, C.K.; Hahn, S.M.; Weissman, I.L.; Jiang, W. Improving immune-vascular crosstalk for cancer immunotherapy. Nat. Rev. Immunol. 2018, 18, 195–203. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Li, S.; Wang, Q.; Jin, Z.; Shao, W.; Gao, Y.; Li, L.; Lin, K.; Zhu, L.; Wang, H.; et al. Tumor immunological phenotype signature-based high-throughput screening for the discovery of combination immunotherapy compounds. Sci. Adv. 2021, 7, eabd7851. [Google Scholar] [CrossRef] [PubMed]
- De Sanctis, F.; Ugel, S.; Facciponte, J.; Facciabene, A. The dark side of tumor-associated endothelial cells. Semin. Immunol. 2018, 35, 35–47. [Google Scholar] [CrossRef]
- Gettinger, S.; Choi, J.; Hastings, K.; Truini, A.; Datar, I.; Sowell, R.; Wurtz, A.; Dong, W.; Cai, G.; Melnick, M.A.; et al. Impaired HLA Class I Antigen Processing and Presentation as a Mechanism of Acquired Resistance to Immune Checkpoint Inhibitors in Lung Cancer. Cancer Discov. 2017, 7, 1420–1435. [Google Scholar] [CrossRef] [Green Version]
- Busse, D.; de la Rosa, M.; Hobiger, K.; Thurley, K.; Flossdorf, M.; Scheffold, A.; Hofer, T. Competing feedback loops shape IL-2 signaling between helper and regulatory T lymphocytes in cellular microenvironments. Proc. Natl. Acad. Sci. USA 2010, 107, 3058–3063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pandiyan, P.; Zheng, L.; Ishihara, S.; Reed, J.; Lenardo, M.J. CD4+CD25+Foxp3+ regulatory T cells induce cytokine deprivation-mediated apoptosis of effector CD4+ T cells. Nat. Immunol. 2007, 8, 1353–1362. [Google Scholar] [CrossRef]
- Wang, H.; Franco, F.; Tsui, Y.C.; Xie, X.; Trefny, M.P.; Zappasodi, R.; Mohmood, S.R.; Fernandez-Garcia, J.; Tsai, C.H.; Schulze, I.; et al. CD36-mediated metabolic adaptation supports regulatory T cell survival and function in tumors. Nat. Immunol. 2020, 21, 298–308. [Google Scholar] [CrossRef]
- Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Bronte, V. Coordinated regulation of myeloid cells by tumours. Nat. Rev. Immunol. 2012, 12, 253–268. [Google Scholar] [CrossRef] [Green Version]
- Ugel, S.; Cane, S.; De Sanctis, F.; Bronte, V. Monocytes in the Tumor Microenvironment. Annu. Rev. Pathol. 2021, 16, 93–122. [Google Scholar] [CrossRef]
- Shojaei, F.; Wu, X.; Malik, A.K.; Zhong, C.; Baldwin, M.E.; Schanz, S.; Fuh, G.; Gerber, H.P.; Ferrara, N. Tumor refractoriness to anti-VEGF treatment is mediated by CD11b+Gr1+ myeloid cells. Nat. Biotechnol. 2007, 25, 911–920. [Google Scholar] [CrossRef]
- Shojaei, F.; Wu, X.; Zhong, C.; Yu, L.; Liang, X.H.; Yao, J.; Blanchard, D.; Bais, C.; Peale, F.V.; van Bruggen, N.; et al. Bv8 regulates myeloid-cell-dependent tumour angiogenesis. Nature 2007, 450, 825–831. [Google Scholar] [CrossRef]
- Cui, T.X.; Kryczek, I.; Zhao, L.; Zhao, E.; Kuick, R.; Roh, M.H.; Vatan, L.; Szeliga, W.; Mao, Y.; Thomas, D.G.; et al. Myeloid-derived suppressor cells enhance stemness of cancer cells by inducing microRNA101 and suppressing the corepressor CtBP2. Immunity 2013, 39, 611–621. [Google Scholar] [CrossRef] [Green Version]
- Lim, J.Y.; Kim, T.W.; Ryu, D.B.; Park, S.S.; Lee, S.E.; Kim, B.S.; Min, C.K. Myeloma-Secreted Galectin-1 Potently Interacts with CD304 on Monocytic Myeloid-Derived Suppressor Cells. Cancer Immunol. Res. 2021, 9, 503–513. [Google Scholar] [CrossRef] [PubMed]
- Talmadge, J.E.; Gabrilovich, D.I. History of myeloid-derived suppressor cells. Nat. Rev. Cancer 2013, 13, 739–752. [Google Scholar] [CrossRef]
- Trovato, R.; Cane, S.; Petrova, V.; Sartoris, S.; Ugel, S.; De Sanctis, F. The Engagement Between MDSCs and Metastases: Partners in Crime. Front. Oncol. 2020, 10, 165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khadge, S.; Cole, K.; Talmadge, J.E. Myeloid derived suppressor cells and the release of micro-metastases from dormancy. Clin. Exp. Metastasis 2021, 38, 279–293. [Google Scholar] [CrossRef] [PubMed]
- De Sanctis, F.; Bronte, V.; Ugel, S. Tumor-Induced Myeloid-Derived Suppressor Cells. Microbiol. Spectr. 2016, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Veglia, F.; Sanseviero, E.; Gabrilovich, D.I. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat. Rev. Immunol. 2021, 21, 485–498. [Google Scholar] [CrossRef]
- Peranzoni, E.; Zilio, S.; Marigo, I.; Dolcetti, L.; Zanovello, P.; Mandruzzato, S.; Bronte, V. Myeloid-derived suppressor cell heterogeneity and subset definition. Curr. Opin. Immunol. 2010, 22, 238–244. [Google Scholar] [CrossRef]
- Veglia, F.; Perego, M.; Gabrilovich, D. Myeloid-derived suppressor cells coming of age. Nat. Immunol. 2018, 19, 108–119. [Google Scholar] [CrossRef] [PubMed]
- Bronte, V.; Brandau, S.; Chen, S.H.; Colombo, M.P.; Frey, A.B.; Greten, T.F.; Mandruzzato, S.; Murray, P.J.; Ochoa, A.; Ostrand-Rosenberg, S.; et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat. Commun. 2016, 7, 12150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bronte, V.; Apolloni, E.; Cabrelle, A.; Ronca, R.; Serafini, P.; Zamboni, P.; Restifo, N.P.; Zanovello, P. Identification of a CD11b(+)/Gr-1(+)/CD31(+) myeloid progenitor capable of activating or suppressing CD8(+) T cells. Blood 2000, 96, 3838–3846. [Google Scholar] [CrossRef] [PubMed]
- Movahedi, K.; Guilliams, M.; Van den Bossche, J.; Van den Bergh, R.; Gysemans, C.; Beschin, A.; De Baetselier, P.; Van Ginderachter, J.A. Identification of discrete tumor-induced myeloid-derived suppressor cell subpopulations with distinct T cell-suppressive activity. Blood 2008, 111, 4233–4244. [Google Scholar] [CrossRef]
- Dolcetti, L.; Peranzoni, E.; Ugel, S.; Marigo, I.; Fernandez Gomez, A.; Mesa, C.; Geilich, M.; Winkels, G.; Traggiai, E.; Casati, A.; et al. Hierarchy of immunosuppressive strength among myeloid-derived suppressor cell subsets is determined by GM-CSF. Eur. J. Immunol. 2010, 40, 22–35. [Google Scholar] [CrossRef] [PubMed]
- Alshetaiwi, H.; Pervolarakis, N.; McIntyre, L.L.; Ma, D.; Nguyen, Q.; Rath, J.A.; Nee, K.; Hernandez, G.; Evans, K.; Torosian, L.; et al. Defining the emergence of myeloid-derived suppressor cells in breast cancer using single-cell transcriptomics. Sci. Immunol. 2020, 5, eaay6017. [Google Scholar] [CrossRef]
- Condamine, T.; Dominguez, G.A.; Youn, J.I.; Kossenkov, A.V.; Mony, S.; Alicea-Torres, K.; Tcyganov, E.; Hashimoto, A.; Nefedova, Y.; Lin, C.; et al. Lectin-type oxidized LDL receptor-1 distinguishes population of human polymorphonuclear myeloid-derived suppressor cells in cancer patients. Sci. Immunol. 2016, 1, aaf8943. [Google Scholar] [CrossRef] [Green Version]
- Solito, S.; Marigo, I.; Pinton, L.; Damuzzo, V.; Mandruzzato, S.; Bronte, V. Myeloid-derived suppressor cell heterogeneity in human cancers. Ann. N. Y. Acad. Sci. 2014, 1319, 47–65. [Google Scholar] [CrossRef]
- Mandruzzato, S.; Brandau, S.; Britten, C.M.; Bronte, V.; Damuzzo, V.; Gouttefangeas, C.; Maurer, D.; Ottensmeier, C.; van der Burg, S.H.; Welters, M.J.; et al. Toward harmonized phenotyping of human myeloid-derived suppressor cells by flow cytometry: Results from an interim study. Cancer Immunol. Immunother. 2016, 65, 161–169. [Google Scholar] [CrossRef] [PubMed]
- Ugel, S.; De Sanctis, F.; Mandruzzato, S.; Bronte, V. Tumor-induced myeloid deviation: When myeloid-derived suppressor cells meet tumor-associated macrophages. J. Clin. Investig. 2015, 125, 3365–3376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Solito, S.; Pinton, L.; De Sanctis, F.; Ugel, S.; Bronte, V.; Mandruzzato, S.; Marigo, I. Methods to Measure MDSC Immune Suppressive Activity In Vitro and In Vivo. Curr. Protoc. Immunol. 2019, 124, e61. [Google Scholar] [CrossRef] [Green Version]
- Wang, L.; Chang, E.W.; Wong, S.C.; Ong, S.M.; Chong, D.Q.; Ling, K.L. Increased myeloid-derived suppressor cells in gastric cancer correlate with cancer stage and plasma S100A8/A9 proinflammatory proteins. J. Immunol. 2013, 190, 794–804. [Google Scholar] [CrossRef]
- Solito, S.; Falisi, E.; Diaz-Montero, C.M.; Doni, A.; Pinton, L.; Rosato, A.; Francescato, S.; Basso, G.; Zanovello, P.; Onicescu, G.; et al. A human promyelocytic-like population is responsible for the immune suppression mediated by myeloid-derived suppressor cells. Blood 2011, 118, 2254–2265. [Google Scholar] [CrossRef] [Green Version]
- Gabitass, R.F.; Annels, N.E.; Stocken, D.D.; Pandha, H.A.; Middleton, G.W. Elevated myeloid-derived suppressor cells in pancreatic, esophageal and gastric cancer are an independent prognostic factor and are associated with significant elevation of the Th2 cytokine interleukin-13. Cancer Immunol. Immunother. 2011, 60, 1419–1430. [Google Scholar] [CrossRef] [Green Version]
- Trovato, R.; Fiore, A.; Sartori, S.; Cane, S.; Giugno, R.; Cascione, L.; Paiella, S.; Salvia, R.; De Sanctis, F.; Poffe, O.; et al. Immunosuppression by monocytic myeloid-derived suppressor cells in patients with pancreatic ductal carcinoma is orchestrated by STAT3. J. Immunother. Cancer 2019, 7, 255. [Google Scholar] [CrossRef]
- Weide, B.; Martens, A.; Hassel, J.C.; Berking, C.; Postow, M.A.; Bisschop, K.; Simeone, E.; Mangana, J.; Schilling, B.; Di Giacomo, A.M.; et al. Baseline Biomarkers for Outcome of Melanoma Patients Treated with Pembrolizumab. Clin. Cancer Res. 2016, 22, 5487–5496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peranzoni, E.; Ingangi, V.; Masetto, E.; Pinton, L.; Marigo, I. Myeloid Cells as Clinical Biomarkers for Immune Checkpoint Blockade. Front. Immunol. 2020, 11, 1590. [Google Scholar] [CrossRef]
- Walter, S.; Weinschenk, T.; Stenzl, A.; Zdrojowy, R.; Pluzanska, A.; Szczylik, C.; Staehler, M.; Brugger, W.; Dietrich, P.Y.; Mendrzyk, R.; et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat. Med. 2012, 18, 1254–1261. [Google Scholar] [CrossRef] [PubMed]
- Welters, M.J.; van der Sluis, T.C.; van Meir, H.; Loof, N.M.; van Ham, V.J.; van Duikeren, S.; Santegoets, S.J.; Arens, R.; de Kam, M.L.; Cohen, A.F.; et al. Vaccination during myeloid cell depletion by cancer chemotherapy fosters robust T cell responses. Sci. Transl. Med. 2016, 8, 334ra352. [Google Scholar] [CrossRef]
- Bost, P.; De Sanctis, F.; Cane, S.; Ugel, S.; Donadello, K.; Castellucci, M.; Eyal, D.; Fiore, A.; Anselmi, C.; Barouni, R.M.; et al. Deciphering the state of immune silence in fatal COVID-19 patients. Nat. Commun. 2021, 12, 1428. [Google Scholar] [CrossRef]
- Consonni, F.M.; Porta, C.; Marino, A.; Pandolfo, C.; Mola, S.; Bleve, A.; Sica, A. Myeloid-Derived Suppressor Cells: Ductile Targets in Disease. Front. Immunol. 2019, 10, 949. [Google Scholar] [CrossRef] [PubMed]
- Haverkamp, J.M.; Smith, A.M.; Weinlich, R.; Dillon, C.P.; Qualls, J.E.; Neale, G.; Koss, B.; Kim, Y.; Bronte, V.; Herold, M.J.; et al. Myeloid-derived suppressor activity is mediated by monocytic lineages maintained by continuous inhibition of extrinsic and intrinsic death pathways. Immunity 2014, 41, 947–959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fiore, A.; Ugel, S.; De Sanctis, F.; Sandri, S.; Fracasso, G.; Trovato, R.; Sartoris, S.; Solito, S.; Mandruzzato, S.; Vascotto, F.; et al. Induction of immunosuppressive functions and NF-kappaB by FLIP in monocytes. Nat. Commun. 2018, 9, 5193. [Google Scholar] [CrossRef] [PubMed]
- Adamo, A.; Frusteri, C.; Pallotta, M.T.; Pirali, T.; Sartoris, S.; Ugel, S. Moonlighting Proteins Are Important Players in Cancer Immunology. Front. Immunol. 2020, 11, 613069. [Google Scholar] [CrossRef]
- Musiu, C.; Caligola, S.; Fiore, A.; Lamolinara, A.; Frusteri, C.; Del Pizzo, F.D.; De Sanctis, F.; Canè, S.; Adamo, A.; Hofer, F.; et al. Fatal cytokine release syndrome by an aberrant FLIP/STAT3 axis. Cell Death Differ. 2021, 28, 1–19. [Google Scholar] [CrossRef]
- Hamilton, J.A. Colony-stimulating factors in inflammation and autoimmunity. Nat. Rev. Immunol. 2008, 8, 533–544. [Google Scholar] [CrossRef]
- Wu, W.C.; Sun, H.W.; Chen, H.T.; Liang, J.; Yu, X.J.; Wu, C.; Wang, Z.; Zheng, L. Circulating hematopoietic stem and progenitor cells are myeloid-biased in cancer patients. Proc. Natl. Acad. Sci. USA 2014, 111, 4221–4226. [Google Scholar] [CrossRef] [Green Version]
- Condamine, T.; Mastio, J.; Gabrilovich, D.I. Transcriptional regulation of myeloid-derived suppressor cells. J. Leukoc. Biol. 2015, 98, 913–922. [Google Scholar] [CrossRef]
- Marigo, I.; Bosio, E.; Solito, S.; Mesa, C.; Fernandez, A.; Dolcetti, L.; Ugel, S.; Sonda, N.; Bicciato, S.; Falisi, E.; et al. Tumor-induced tolerance and immune suppression depend on the C/EBPbeta transcription factor. Immunity 2010, 32, 790–802. [Google Scholar] [CrossRef]
- Chalmin, F.; Ladoire, S.; Mignot, G.; Vincent, J.; Bruchard, M.; Remy-Martin, J.P.; Boireau, W.; Rouleau, A.; Simon, B.; Lanneau, D.; et al. Membrane-associated Hsp72 from tumor-derived exosomes mediates STAT3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells. J. Clin. Investig. 2010, 120, 457–471. [Google Scholar] [CrossRef]
- Veglia, F.; Hashimoto, A.; Dweep, H.; Sanseviero, E.; De Leo, A.; Tcyganov, E.; Kossenkov, A.; Mulligan, C.; Nam, B.; Masters, G.; et al. Analysis of classical neutrophils and polymorphonuclear myeloid-derived suppressor cells in cancer patients and tumor-bearing mice. J. Exp. Med. 2021, 218, e20201803. [Google Scholar] [CrossRef]
- Strauss, L.; Sangaletti, S.; Consonni, F.M.; Szebeni, G.; Morlacchi, S.; Totaro, M.G.; Porta, C.; Anselmo, A.; Tartari, S.; Doni, A.; et al. RORC1 Regulates Tumor-Promoting “Emergency” Granulo-Monocytopoiesis. Cancer Cell 2015, 28, 253–269. [Google Scholar] [CrossRef] [Green Version]
- Netherby, C.S.; Messmer, M.N.; Burkard-Mandel, L.; Colligan, S.; Miller, A.; Gomez, E.C.; Wang, J.; Nemeth, M.J.; Abrams, S.I. The Granulocyte Progenitor Stage Is a Key Target of IRF8-Mediated Regulation of Myeloid-Derived Suppressor Cell Production. J. Immunol. 2017, 198, 4129–4139. [Google Scholar] [CrossRef] [Green Version]
- Waight, J.D.; Netherby, C.; Hensen, M.L.; Miller, A.; Hu, Q.; Liu, S.; Bogner, P.N.; Farren, M.R.; Lee, K.P.; Liu, K.; et al. Myeloid-derived suppressor cell development is regulated by a STAT/IRF-8 axis. J. Clin. Investig. 2013, 123, 4464–4478. [Google Scholar] [CrossRef]
- Kwok, I.; Becht, E.; Xia, Y.; Ng, M.; Teh, Y.C.; Tan, L.; Evrard, M.; Li, J.L.Y.; Tran, H.T.N.; Tan, Y.; et al. Combinatorial Single-Cell Analyses of Granulocyte-Monocyte Progenitor Heterogeneity Reveals an Early Uni-potent Neutrophil Progenitor. Immunity 2020, 53, 303–318.e5. [Google Scholar] [CrossRef]
- Kortylewski, M.; Kujawski, M.; Wang, T.; Wei, S.; Zhang, S.; Pilon-Thomas, S.; Niu, G.; Kay, H.; Mule, J.; Kerr, W.G.; et al. Inhibiting Stat3 signaling in the hematopoietic system elicits multicomponent antitumor immunity. Nat. Med. 2005, 11, 1314–1321. [Google Scholar] [CrossRef] [PubMed]
- Hu, X.; Bardhan, K.; Paschall, A.V.; Yang, D.; Waller, J.L.; Park, M.A.; Nayak-Kapoor, A.; Samuel, T.A.; Abrams, S.I.; Liu, K. Deregulation of apoptotic factors Bcl-xL and Bax confers apoptotic resistance to myeloid-derived suppressor cells and contributes to their persistence in cancer. J. Biol. Chem. 2013, 288, 19103–19115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Corzo, C.A.; Cotter, M.J.; Cheng, P.; Cheng, F.; Kusmartsev, S.; Sotomayor, E.; Padhya, T.; McCaffrey, T.V.; McCaffrey, J.C.; Gabrilovich, D.I. Mechanism regulating reactive oxygen species in tumor-induced myeloid-derived suppressor cells. J. Immunol. 2009, 182, 5693–5701. [Google Scholar] [CrossRef] [PubMed]
- Vasquez-Dunddel, D.; Pan, F.; Zeng, Q.; Gorbounov, M.; Albesiano, E.; Fu, J.; Blosser, R.L.; Tam, A.J.; Bruno, T.; Zhang, H.; et al. STAT3 regulates arginase-I in myeloid-derived suppressor cells from cancer patients. J. Clin. Investig. 2013, 123, 1580–1589. [Google Scholar] [CrossRef] [Green Version]
- Sinha, P.; Okoro, C.; Foell, D.; Freeze, H.H.; Ostrand-Rosenberg, S.; Srikrishna, G. Proinflammatory S100 proteins regulate the accumulation of myeloid-derived suppressor cells. J. Immunol. 2008, 181, 4666–4675. [Google Scholar] [CrossRef] [Green Version]
- Cheng, P.; Corzo, C.A.; Luetteke, N.; Yu, B.; Nagaraj, S.; Bui, M.M.; Ortiz, M.; Nacken, W.; Sorg, C.; Vogl, T.; et al. Inhibition of dendritic cell differentiation and accumulation of myeloid-derived suppressor cells in cancer is regulated by S100A9 protein. J. Exp. Med. 2008, 205, 2235–2249. [Google Scholar] [CrossRef]
- Weber, R.; Umansky, V. Fighting infant infections with myeloid-derived suppressor cells. J. Clin. Investig. 2019, 129, 4080–4082. [Google Scholar] [CrossRef] [Green Version]
- Zhou, H.; Jiang, M.; Yuan, H.; Ni, W.; Tai, G. Dual roles of myeloid-derived suppressor cells induced by Toll-like receptor signaling in cancer. Oncol. Lett. 2021, 21, 149. [Google Scholar] [CrossRef] [PubMed]
- Sangaletti, S.; Talarico, G.; Chiodoni, C.; Cappetti, B.; Botti, L.; Portararo, P.; Gulino, A.; Consonni, F.M.; Sica, A.; Randon, G.; et al. SPARC Is a New Myeloid-Derived Suppressor Cell Marker Licensing Suppressive Activities. Front. Immunol. 2019, 10, 1369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Porta, C.; Consonni, F.M.; Morlacchi, S.; Sangaletti, S.; Bleve, A.; Totaro, M.G.; Larghi, P.; Rimoldi, M.; Tripodo, C.; Strauss, L.; et al. Tumor-Derived Prostaglandin E2 Promotes p50 NF-kappaB-Dependent Differentiation of Monocytic MDSCs. Cancer Res. 2020, 80, 2874–2888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Halaby, M.J.; Hezaveh, K.; Lamorte, S.; Ciudad, M.T.; Kloetgen, A.; MacLeod, B.L.; Guo, M.; Chakravarthy, A.; Medina, T.D.S.; Ugel, S.; et al. GCN2 drives macrophage and MDSC function and immunosuppression in the tumor microenvironment. Sci. Immunol. 2019, 4, eaax8189. [Google Scholar] [CrossRef]
- Corzo, C.A.; Condamine, T.; Lu, L.; Cotter, M.J.; Youn, J.I.; Cheng, P.; Cho, H.I.; Celis, E.; Quiceno, D.G.; Padhya, T.; et al. HIF-1alpha regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J. Exp. Med. 2010, 207, 2439–2453. [Google Scholar] [CrossRef] [PubMed]
- Trikha, P.; Plews, R.L.; Stiff, A.; Gautam, S.; Hsu, V.; Abood, D.; Wesolowski, R.; Landi, I.; Mo, X.; Phay, J.; et al. Targeting myeloid-derived suppressor cells using a novel adenosine monophosphate-activated protein kinase (AMPK) activator. Oncoimmunology 2016, 5, e1214787. [Google Scholar] [CrossRef] [Green Version]
- Youn, J.I.; Kumar, V.; Collazo, M.; Nefedova, Y.; Condamine, T.; Cheng, P.; Villagra, A.; Antonia, S.; McCaffrey, J.C.; Fishman, M.; et al. Epigenetic silencing of retinoblastoma gene regulates pathologic differentiation of myeloid cells in cancer. Nat. Immunol. 2013, 14, 211–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ugel, S.; Peranzoni, E.; Desantis, G.; Chioda, M.; Walter, S.; Weinschenk, T.; Ochando, J.C.; Cabrelle, A.; Mandruzzato, S.; Bronte, V. Immune tolerance to tumor antigens occurs in a specialized environment of the spleen. Cell Rep. 2012, 2, 628–639. [Google Scholar] [CrossRef]
- Wynn, T.A.; Chawla, A.; Pollard, J.W. Macrophage biology in development, homeostasis and disease. Nature 2013, 496, 445–455. [Google Scholar] [CrossRef] [PubMed]
- Cane, S.; Ugel, S.; Trovato, R.; Marigo, I.; De Sanctis, F.; Sartoris, S.; Bronte, V. The Endless Saga of Monocyte Diversity. Front. Immunol. 2019, 10, 1786. [Google Scholar] [CrossRef] [Green Version]
- Marigo, I.; Trovato, R.; Hofer, F.; Ingangi, V.; Desantis, G.; Leone, K.; De Sanctis, F.; Ugel, S.; Cane, S.; Simonelli, A.; et al. Disabled Homolog 2 Controls Prometastatic Activity of Tumor-Associated Macrophages. Cancer Discov. 2020, 10, 1758–1773. [Google Scholar] [CrossRef]
- Kumar, V.; Cheng, P.; Condamine, T.; Mony, S.; Languino, L.R.; McCaffrey, J.C.; Hockstein, N.; Guarino, M.; Masters, G.; Penman, E.; et al. CD45 Phosphatase Inhibits STAT3 Transcription Factor Activity in Myeloid Cells and Promotes Tumor-Associated Macrophage Differentiation. Immunity 2016, 44, 303–315. [Google Scholar] [CrossRef] [Green Version]
- Highfill, S.L.; Cui, Y.; Giles, A.J.; Smith, J.P.; Zhang, H.; Morse, E.; Kaplan, R.N.; Mackall, C.L. Disruption of CXCR2-mediated MDSC tumor trafficking enhances anti-PD1 efficacy. Sci. Transl. Med. 2014, 6, 237ra67. [Google Scholar] [CrossRef] [PubMed]
- Cassetta, L.; Pollard, J.W. Targeting macrophages: Therapeutic approaches in cancer. Nat. Rev. Drug Discov. 2018, 17, 887–904. [Google Scholar] [CrossRef]
- Centuori, S.M.; Trad, M.; LaCasse, C.J.; Alizadeh, D.; Larmonier, C.B.; Hanke, N.T.; Kartchner, J.; Janikashvili, N.; Bonnotte, B.; Larmonier, N.; et al. Myeloid-derived suppressor cells from tumor-bearing mice impair TGF-beta-induced differentiation of CD4+CD25+FoxP3+ Tregs from CD4+CD25-FoxP3- T cells. J. Leukoc. Biol. 2012, 92, 987–997. [Google Scholar] [CrossRef] [Green Version]
- Sinha, P.; Clements, V.K.; Bunt, S.K.; Albelda, S.M.; Ostrand-Rosenberg, S. Cross-talk between myeloid-derived suppressor cells and macrophages subverts tumor immunity toward a type 2 response. J. Immunol. 2007, 179, 977–983. [Google Scholar] [CrossRef]
- Li, H.; Han, Y.; Guo, Q.; Zhang, M.; Cao, X. Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells through membrane-bound TGF-beta 1. J. Immunol. 2009, 182, 240–249. [Google Scholar] [CrossRef] [Green Version]
- Dardalhon, V.; Anderson, A.C.; Karman, J.; Apetoh, L.; Chandwaskar, R.; Lee, D.H.; Cornejo, M.; Nishi, N.; Yamauchi, A.; Quintana, F.J.; et al. Tim-3/galectin-9 pathway: Regulation of Th1 immunity through promotion of CD11b+Ly-6G+ myeloid cells. J. Immunol. 2010, 185, 1383–1392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Limagne, E.; Richard, C.; Thibaudin, M.; Fumet, J.D.; Truntzer, C.; Lagrange, A.; Favier, L.; Coudert, B.; Ghiringhelli, F. Tim-3/galectin-9 pathway and mMDSC control primary and secondary resistances to PD-1 blockade in lung cancer patients. Oncoimmunology 2019, 8, e1564505. [Google Scholar] [CrossRef] [PubMed]
- Noman, M.Z.; Desantis, G.; Janji, B.; Hasmim, M.; Karray, S.; Dessen, P.; Bronte, V.; Chouaib, S. PD-L1 is a novel direct target of HIF-1alpha, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J. Exp. Med. 2014, 211, 781–790. [Google Scholar] [CrossRef] [PubMed]
- Warburg, O. On the origin of cancer cells. Science 1956, 123, 309–314. [Google Scholar] [CrossRef] [PubMed]
- Li, W.; Tanikawa, T.; Kryczek, I.; Xia, H.; Li, G.; Wu, K.; Wei, S.; Zhao, L.; Vatan, L.; Wen, B.; et al. Aerobic Glycolysis Controls Myeloid-Derived Suppressor Cells and Tumor Immunity via a Specific CEBPB Isoform in Triple-Negative Breast Cancer. Cell Metab. 2018, 28, 87–103.e6. [Google Scholar] [CrossRef] [Green Version]
- Cai, T.T.; Ye, S.B.; Liu, Y.N.; He, J.; Chen, Q.Y.; Mai, H.Q.; Zhang, C.X.; Cui, J.; Zhang, X.S.; Busson, P.; et al. LMP1-mediated glycolysis induces myeloid-derived suppressor cell expansion in nasopharyngeal carcinoma. PLoS Pathog. 2017, 13, e1006503. [Google Scholar] [CrossRef]
- Hammami, I.; Chen, J.; Murschel, F.; Bronte, V.; De Crescenzo, G.; Jolicoeur, M. Immunosuppressive activity enhances central carbon metabolism and bioenergetics in myeloid-derived suppressor cells in vitro models. BMC Cell Biol. 2012, 13, 18. [Google Scholar] [CrossRef] [Green Version]
- Christofk, H.R.; Vander Heiden, M.G.; Harris, M.H.; Ramanathan, A.; Gerszten, R.E.; Wei, R.; Fleming, M.D.; Schreiber, S.L.; Cantley, L.C. The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature 2008, 452, 230–233. [Google Scholar] [CrossRef]
- Palsson-McDermott, E.M.; Curtis, A.M.; Goel, G.; Lauterbach, M.A.; Sheedy, F.J.; Gleeson, L.E.; van den Bosch, M.W.; Quinn, S.R.; Domingo-Fernandez, R.; Johnston, D.G.; et al. Pyruvate kinase M2 regulates Hif-1alpha activity and IL-1beta induction and is a critical determinant of the warburg effect in LPS-activated macrophages. Cell Metab. 2015, 21, 65–80. [Google Scholar] [CrossRef] [Green Version]
- Lizak, B.; Szarka, A.; Kim, Y.; Choi, K.S.; Nemeth, C.E.; Marcolongo, P.; Benedetti, A.; Banhegyi, G.; Margittai, E. Glucose Transport and Transporters in the Endomembranes. Int. J. Mol. Sci. 2019, 20, 5898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reinfeld, B.I.; Madden, M.Z.; Wolf, M.M.; Chytil, A.; Bader, J.E.; Patterson, A.R.; Sugiura, A.; Cohen, A.S.; Ali, A.; Do, B.T.; et al. Cell-programmed nutrient partitioning in the tumour microenvironment. Nature 2021, 593, 282–288. [Google Scholar] [CrossRef] [PubMed]
- Saxton, R.A.; Sabatini, D.M. mTOR Signaling in Growth, Metabolism, and Disease. Cell 2017, 168, 960–976. [Google Scholar] [CrossRef] [Green Version]
- Baumann, T.; Dunkel, A.; Schmid, C.; Schmitt, S.; Hiltensperger, M.; Lohr, K.; Laketa, V.; Donakonda, S.; Ahting, U.; Lorenz-Depiereux, B.; et al. Regulatory myeloid cells paralyze T cells through cell-cell transfer of the metabolite methylglyoxal. Nat. Immunol. 2020, 21, 555–566. [Google Scholar] [CrossRef] [PubMed]
- Ippolito, L.; Morandi, A.; Giannoni, E.; Chiarugi, P. Lactate: A Metabolic Driver in the Tumour Landscape. Trends Biochem. Sci. 2019, 44, 153–166. [Google Scholar] [CrossRef] [PubMed]
- Andrejeva, G.; Rathmell, J.C. Similarities and Distinctions of Cancer and Immune Metabolism in Inflammation and Tumors. Cell Metab. 2017, 26, 49–70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Burgos, E.S.; Vetticatt, M.J.; Schramm, V.L. Recycling nicotinamide. The transition-state structure of human nicotinamide phosphoribosyltransferase. J. Am. Chem. Soc. 2013, 135, 3485–3493. [Google Scholar] [CrossRef] [Green Version]
- Travelli, C.; Consonni, F.M.; Sangaletti, S.; Storto, M.; Morlacchi, S.; Grolla, A.A.; Galli, U.; Tron, G.C.; Portararo, P.; Rimassa, L.; et al. Nicotinamide Phosphoribosyltransferase Acts as a Metabolic Gate for Mobilization of Myeloid-Derived Suppressor Cells. Cancer Res. 2019, 79, 1938–1951. [Google Scholar] [CrossRef] [Green Version]
- Chen, A.N.; Luo, Y.; Yang, Y.H.; Fu, J.T.; Geng, X.M.; Shi, J.P.; Yang, J. Lactylation, a Novel Metabolic Reprogramming Code: Current Status and Prospects. Front. Immunol. 2021, 12, 688910. [Google Scholar] [CrossRef] [PubMed]
- Dichtl, S.; Lindenthal, L.; Zeitler, L.; Behnke, K.; Schlosser, D.; Strobl, B.; Scheller, J.; El Kasmi, K.C.; Murray, P.J. Lactate and IL6 define separable paths of inflammatory metabolic adaptation. Sci. Adv. 2021, 7, eabg3505. [Google Scholar] [CrossRef]
- Brand, A.; Singer, K.; Koehl, G.E.; Kolitzus, M.; Schoenhammer, G.; Thiel, A.; Matos, C.; Bruss, C.; Klobuch, S.; Peter, K.; et al. LDHA-Associated Lactic Acid Production Blunts Tumor Immunosurveillance by T and NK Cells. Cell Metab. 2016, 24, 657–671. [Google Scholar] [CrossRef] [Green Version]
- Schneiderhan, W.; Scheler, M.; Holzmann, K.H.; Marx, M.; Gschwend, J.E.; Bucholz, M.; Gress, T.M.; Seufferlein, T.; Adler, G.; Oswald, F. CD147 silencing inhibits lactate transport and reduces malignant potential of pancreatic cancer cells in in vivo and in vitro models. Gut 2009, 58, 1391–1398. [Google Scholar] [CrossRef] [PubMed]
- Li, H.; Myeroff, L.; Smiraglia, D.; Romero, M.F.; Pretlow, T.P.; Kasturi, L.; Lutterbaugh, J.; Rerko, R.M.; Casey, G.; Issa, J.P.; et al. SLC5A8, a sodium transporter, is a tumor suppressor gene silenced by methylation in human colon aberrant crypt foci and cancers. Proc. Natl. Acad. Sci. USA 2003, 100, 8412–8417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vijayan, D.; Young, A.; Teng, M.W.L.; Smyth, M.J. Targeting immunosuppressive adenosine in cancer. Nat. Rev. Cancer 2017, 17, 709–724. [Google Scholar] [CrossRef] [PubMed]
- Young, A.; Mittal, D.; Stagg, J.; Smyth, M.J. Targeting cancer-derived adenosine: New therapeutic approaches. Cancer Discov. 2014, 4, 879–888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stagg, J.; Divisekera, U.; Duret, H.; Sparwasser, T.; Teng, M.W.; Darcy, P.K.; Smyth, M.J. CD73-deficient mice have increased antitumor immunity and are resistant to experimental metastasis. Cancer Res. 2011, 71, 2892–2900. [Google Scholar] [CrossRef] [Green Version]
- Chiu, D.K.; Tse, A.P.; Xu, I.M.; Di Cui, J.; Lai, R.K.; Li, L.L.; Koh, H.Y.; Tsang, F.H.; Wei, L.L.; Wong, C.M.; et al. Hypoxia inducible factor HIF-1 promotes myeloid-derived suppressor cells accumulation through ENTPD2/CD39L1 in hepatocellular carcinoma. Nat. Commun. 2017, 8, 517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ryzhov, S.; Novitskiy, S.V.; Goldstein, A.E.; Biktasova, A.; Blackburn, M.R.; Biaggioni, I.; Dikov, M.M.; Feoktistov, I. Adenosinergic regulation of the expansion and immunosuppressive activity of CD11b+Gr1+ cells. J. Immunol. 2011, 187, 6120–6129. [Google Scholar] [CrossRef] [Green Version]
- Won, W.J.; Deshane, J.S.; Leavenworth, J.W.; Oliva, C.R.; Griguer, C.E. Metabolic and functional reprogramming of myeloid-derived suppressor cells and their therapeutic control in glioblastoma. Cell Stress 2019, 3, 47–65. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Wang, L.; Chen, X.; Li, L.; Li, Y.; Ping, Y.; Huang, L.; Yue, D.; Zhang, Z.; Wang, F.; et al. CD39/CD73 upregulation on myeloid-derived suppressor cells via TGF-beta-mTOR-HIF-1 signaling in patients with non-small cell lung cancer. Oncoimmunology 2017, 6, e1320011. [Google Scholar] [CrossRef] [Green Version]
- Ryzhov, S.V.; Pickup, M.W.; Chytil, A.; Gorska, A.E.; Zhang, Q.; Owens, P.; Feoktistov, I.; Moses, H.L.; Novitskiy, S.V. Role of TGF-beta signaling in generation of CD39+CD73+ myeloid cells in tumors. J. Immunol. 2014, 193, 3155–3164. [Google Scholar] [CrossRef] [Green Version]
- Li, L.; Wang, L.; Li, J.; Fan, Z.; Yang, L.; Zhang, Z.; Zhang, C.; Yue, D.; Qin, G.; Zhang, T.; et al. Metformin-Induced Reduction of CD39 and CD73 Blocks Myeloid-Derived Suppressor Cell Activity in Patients with Ovarian Cancer. Cancer Res. 2018, 78, 1779–1791. [Google Scholar] [CrossRef] [Green Version]
- Kepp, O.; Bezu, L.; Yamazaki, T.; Di Virgilio, F.; Smyth, M.J.; Kroemer, G.; Galluzzi, L. ATP and cancer immunosurveillance. EMBO J. 2021, 40, e108130. [Google Scholar] [CrossRef]
- Rittiner, J.E.; Korboukh, I.; Hull-Ryde, E.A.; Jin, J.; Janzen, W.P.; Frye, S.V.; Zylka, M.J. AMP is an adenosine A1 receptor agonist. J. Biol. Chem. 2012, 287, 5301–5309. [Google Scholar] [CrossRef] [Green Version]
- Bianchi, G.; Vuerich, M.; Pellegatti, P.; Marimpietri, D.; Emionite, L.; Marigo, I.; Bronte, V.; Di Virgilio, F.; Pistoia, V.; Raffaghello, L. ATP/P2X7 axis modulates myeloid-derived suppressor cell functions in neuroblastoma microenvironment. Cell Death Dis. 2014, 5, e1135. [Google Scholar] [CrossRef] [Green Version]
- Sorrentino, C.; Miele, L.; Porta, A.; Pinto, A.; Morello, S. Myeloid-derived suppressor cells contribute to A2B adenosine receptor-induced VEGF production and angiogenesis in a mouse melanoma model. Oncotarget 2015, 6, 27478–27489. [Google Scholar] [CrossRef] [Green Version]
- Iannone, R.; Miele, L.; Maiolino, P.; Pinto, A.; Morello, S. Blockade of A2b adenosine receptor reduces tumor growth and immune suppression mediated by myeloid-derived suppressor cells in a mouse model of melanoma. Neoplasia 2013, 15, 1400–1409. [Google Scholar] [CrossRef] [Green Version]
- Pallotta, M.T.; Rossini, S.; Suvieri, C.; Coletti, A.; Orabona, C.; Macchiarulo, A.; Volpi, C.; Grohmann, U. Indoleamine 2,3-dioxygenase 1 (IDO1): An up-to-date overview of an eclectic immunoregulatory enzyme. FEBS J. 2021, 6. [Google Scholar] [CrossRef]
- Clement, C.C.; D’Alessandro, A.; Thangaswamy, S.; Chalmers, S.; Furtado, R.; Spada, S.; Mondanelli, G.; Ianni, F.; Gehrke, S.; Gargaro, M.; et al. 3-hydroxy-L-kynurenamine is an immunomodulatory biogenic amine. Nat. Commun. 2021, 12, 4447. [Google Scholar] [CrossRef] [PubMed]
- Mellor, A.L.; Munn, D.H. IDO expression by dendritic cells: Tolerance and tryptophan catabolism. Nat. Rev. Immunol. 2004, 4, 762–774. [Google Scholar] [CrossRef] [PubMed]
- Fiore, A.; Murray, P.J. Tryptophan and indole metabolism in immune regulation. Curr. Opin. Immunol. 2021, 70, 7–14. [Google Scholar] [CrossRef]
- Godin-Ethier, J.; Hanafi, L.A.; Piccirillo, C.A.; Lapointe, R. Indoleamine 2,3-dioxygenase expression in human cancers: Clinical and immunologic perspectives. Clin. Cancer Res. 2011, 17, 6985–6991. [Google Scholar] [CrossRef] [Green Version]
- Munn, D.H.; Mellor, A.L. Indoleamine 2,3 dioxygenase and metabolic control of immune responses. Trends Immunol. 2013, 34, 137–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pinton, L.; Solito, S.; Damuzzo, V.; Francescato, S.; Pozzuoli, A.; Berizzi, A.; Mocellin, S.; Rossi, C.R.; Bronte, V.; Mandruzzato, S. Activated T cells sustain myeloid-derived suppressor cell-mediated immune suppression. Oncotarget 2016, 7, 1168–1184. [Google Scholar] [CrossRef] [Green Version]
- Grohmann, U.; Puccetti, P. The Coevolution of IDO1 and AhR in the Emergence of Regulatory T-Cells in Mammals. Front. Immunol. 2015, 6, 58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Opitz, C.A.; Litzenburger, U.M.; Sahm, F.; Ott, M.; Tritschler, I.; Trump, S.; Schumacher, T.; Jestaedt, L.; Schrenk, D.; Weller, M.; et al. An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature 2011, 478, 197–203. [Google Scholar] [CrossRef]
- Smith, C.; Chang, M.Y.; Parker, K.H.; Beury, D.W.; DuHadaway, J.B.; Flick, H.E.; Boulden, J.; Sutanto-Ward, E.; Soler, A.P.; Laury-Kleintop, L.D.; et al. IDO is a nodal pathogenic driver of lung cancer and metastasis development. Cancer Discov. 2012, 2, 722–735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Platten, M.; Nollen, E.A.A.; Rohrig, U.F.; Fallarino, F.; Opitz, C.A. Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond. Nat. Rev. Drug Discov. 2019, 18, 379–401. [Google Scholar] [CrossRef]
- Mondanelli, G.; Ugel, S.; Grohmann, U.; Bronte, V. The immune regulation in cancer by the amino acid metabolizing enzymes ARG and IDO. Curr. Opin. Pharmacol. 2017, 35, 30–39. [Google Scholar] [CrossRef]
- Long, G.V.; Dummer, R.; Hamid, O.; Gajewski, T.F.; Caglevic, C.; Dalle, S.; Arance, A.; Carlino, M.S.; Grob, J.J.; Kim, T.M.; et al. Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO-301/KEYNOTE-252): A phase 3, randomised, double-blind study. Lancet Oncol. 2019, 20, 1083–1097. [Google Scholar] [CrossRef]
- Ortiz-Meoz, R.F.; Wang, L.; Matico, R.; Rutkowska-Klute, A.; De la Rosa, M.; Bedard, S.; Midgett, R.; Strohmer, K.; Thomson, D.; Zhang, C.; et al. Characterization of Apo-Form Selective Inhibition of Indoleamine 2,3-Dioxygenase*. Chembiochem 2021, 22, 516–522. [Google Scholar] [CrossRef] [PubMed]
- Serafini, M.; Torre, E.; Aprile, S.; Grosso, E.D.; Gesu, A.; Griglio, A.; Colombo, G.; Travelli, C.; Paiella, S.; Adamo, A.; et al. Discovery of Highly Potent Benzimidazole Derivatives as Indoleamine 2,3-Dioxygenase-1 (IDO1) Inhibitors: From Structure-Based Virtual Screening to in Vivo Pharmacodynamic Activity. J. Med. Chem. 2020, 63, 3047–3065. [Google Scholar] [CrossRef] [PubMed]
- Fallarino, F.; Grohmann, U.; You, S.; McGrath, B.C.; Cavener, D.R.; Vacca, C.; Orabona, C.; Bianchi, R.; Belladonna, M.L.; Volpi, C.; et al. The combined effects of tryptophan starvation and tryptophan catabolites down-regulate T cell receptor zeta-chain and induce a regulatory phenotype in naive T cells. J. Immunol. 2006, 176, 6752–6761. [Google Scholar] [CrossRef] [PubMed]
- Sharma, M.D.; Hou, D.Y.; Liu, Y.; Koni, P.A.; Metz, R.; Chandler, P.; Mellor, A.L.; He, Y.; Munn, D.H. Indoleamine 2,3-dioxygenase controls conversion of Foxp3+ Tregs to TH17-like cells in tumor-draining lymph nodes. Blood 2009, 113, 6102–6111. [Google Scholar] [CrossRef] [Green Version]
- Van de Velde, L.A.; Guo, X.J.; Barbaric, L.; Smith, A.M.; Oguin, T.H., 3rd; Thomas, P.G.; Murray, P.J. Stress Kinase GCN2 Controls the Proliferative Fitness and Trafficking of Cytotoxic T Cells Independent of Environmental Amino Acid Sensing. Cell Rep. 2016, 17, 2247–2258. [Google Scholar] [CrossRef] [Green Version]
- Ricklefs, F.L.; Alayo, Q.; Krenzlin, H.; Mahmoud, A.B.; Speranza, M.C.; Nakashima, H.; Hayes, J.L.; Lee, K.; Balaj, L.; Passaro, C.; et al. Immune evasion mediated by PD-L1 on glioblastoma-derived extracellular vesicles. Sci. Adv. 2018, 4, eaar2766. [Google Scholar] [CrossRef] [Green Version]
- Pallotta, M.T.; Orabona, C.; Volpi, C.; Vacca, C.; Belladonna, M.L.; Bianchi, R.; Servillo, G.; Brunacci, C.; Calvitti, M.; Bicciato, S.; et al. Indoleamine 2,3-dioxygenase is a signaling protein in long-term tolerance by dendritic cells. Nat. Immunol. 2011, 12, 870–878. [Google Scholar] [CrossRef] [Green Version]
- Orabona, C.; Pallotta, M.T.; Grohmann, U. Different partners, opposite outcomes: A new perspective of the immunobiology of indoleamine 2,3-dioxygenase. Mol. Med. 2012, 18, 834–842. [Google Scholar] [CrossRef]
- Iacono, A.; Pompa, A.; De Marchis, F.; Panfili, E.; Greco, F.A.; Coletti, A.; Orabona, C.; Volpi, C.; Belladonna, M.L.; Mondanelli, G.; et al. Class IA PI3Ks regulate subcellular and functional dynamics of IDO1. EMBO Rep. 2020, 21, e49756. [Google Scholar] [CrossRef]
- Orabona, C.; Pallotta, M.T.; Volpi, C.; Fallarino, F.; Vacca, C.; Bianchi, R.; Belladonna, M.L.; Fioretti, M.C.; Grohmann, U.; Puccetti, P. SOCS3 drives proteasomal degradation of indoleamine 2,3-dioxygenase (IDO) and antagonizes IDO-dependent tolerogenesis. Proc. Natl. Acad. Sci. USA 2008, 105, 20828–20833. [Google Scholar] [CrossRef] [Green Version]
- Lim, Y.J.; Foo, T.C.; Yeung, A.W.S.; Tu, X.; Ma, Y.; Hawkins, C.L.; Witting, P.K.; Jameson, G.N.L.; Terentis, A.C.; Thomas, S.R. Human Indoleamine 2,3-Dioxygenase 1 Is an Efficient Mammalian Nitrite Reductase. Biochemistry 2019, 58, 974–986. [Google Scholar] [CrossRef] [PubMed]
- Bhutia, Y.D.; Babu, E.; Ganapathy, V. Interferon-gamma induces a tryptophan-selective amino acid transporter in human colonic epithelial cells and mouse dendritic cells. Biochim. Biophys. Acta 2015, 1848, 453–462. [Google Scholar] [CrossRef] [Green Version]
- Silk, J.D.; Lakhal, S.; Laynes, R.; Vallius, L.; Karydis, I.; Marcea, C.; Boyd, C.A.; Cerundolo, V. IDO induces expression of a novel tryptophan transporter in mouse and human tumor cells. J. Immunol. 2011, 187, 1617–1625. [Google Scholar] [CrossRef] [Green Version]
- Rodriguez, P.C.; Ochoa, A.C.; Al-Khami, A.A. Arginine Metabolism in Myeloid Cells Shapes Innate and Adaptive Immunity. Front. Immunol. 2017, 8, 93. [Google Scholar] [CrossRef] [Green Version]
- Bronte, V.; Zanovello, P. Regulation of immune responses by L-arginine metabolism. Nat. Rev. Immunol. 2005, 5, 641–654. [Google Scholar] [CrossRef]
- Murray, P.J.; Allen, J.E.; Biswas, S.K.; Fisher, E.A.; Gilroy, D.W.; Goerdt, S.; Gordon, S.; Hamilton, J.A.; Ivashkiv, L.B.; Lawrence, T.; et al. Macrophage activation and polarization: Nomenclature and experimental guidelines. Immunity 2014, 41, 14–20. [Google Scholar] [CrossRef] [Green Version]
- Marigo, I.; Zilio, S.; Desantis, G.; Mlecnik, B.; Agnellini, A.H.; Ugel, S.; Sasso, M.S.; Qualls, J.E.; Kratochvill, F.; Zanovello, P.; et al. T Cell Cancer Therapy Requires CD40-CD40L Activation of Tumor Necrosis Factor and Inducible Nitric-Oxide-Synthase-Producing Dendritic Cells. Cancer Cell 2016, 30, 651. [Google Scholar] [CrossRef]
- Rodriguez, P.C.; Zea, A.H.; DeSalvo, J.; Culotta, K.S.; Zabaleta, J.; Quiceno, D.G.; Ochoa, J.B.; Ochoa, A.C. L-arginine consumption by macrophages modulates the expression of CD3 zeta chain in T lymphocytes. J. Immunol. 2003, 171, 1232–1239. [Google Scholar] [CrossRef] [Green Version]
- Rodriguez, P.C.; Quiceno, D.G.; Ochoa, A.C. L-arginine availability regulates T-lymphocyte cell-cycle progression. Blood 2007, 109, 1568–1573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Sanctis, F.; Sandri, S.; Ferrarini, G.; Pagliarello, I.; Sartoris, S.; Ugel, S.; Marigo, I.; Molon, B.; Bronte, V. The emerging immunological role of post-translational modifications by reactive nitrogen species in cancer microenvironment. Front. Immunol. 2014, 5, 69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Molon, B.; Ugel, S.; Del Pozzo, F.; Soldani, C.; Zilio, S.; Avella, D.; De Palma, A.; Mauri, P.; Monegal, A.; Rescigno, M.; et al. Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells. J. Exp. Med. 2011, 208, 1949–1962. [Google Scholar] [CrossRef]
- Mondanelli, G.; Bianchi, R.; Pallotta, M.T.; Orabona, C.; Albini, E.; Iacono, A.; Belladonna, M.L.; Vacca, C.; Fallarino, F.; Macchiarulo, A.; et al. A Relay Pathway between Arginine and Tryptophan Metabolism Confers Immunosuppressive Properties on Dendritic Cells. Immunity 2017, 46, 233–244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bozkus, C.C.; Elzey, B.D.; Crist, S.A.; Ellies, L.G.; Ratliff, T.L. Expression of Cationic Amino Acid Transporter 2 Is Required for Myeloid-Derived Suppressor Cell-Mediated Control of T Cell Immunity. J. Immunol. 2015, 195, 5237–5250. [Google Scholar] [CrossRef]
- Katzenelenbogen, Y.; Sheban, F.; Yalin, A.; Yofe, I.; Svetlichnyy, D.; Jaitin, D.A.; Bornstein, C.; Moshe, A.; Keren-Shaul, H.; Cohen, M.; et al. Coupled scRNA-Seq and Intracellular Protein Activity Reveal an Immunosuppressive Role of TREM2 in Cancer. Cell 2020, 182, 872–885.e19. [Google Scholar] [CrossRef]
- Cluntun, A.A.; Lukey, M.J.; Cerione, R.A.; Locasale, J.W. Glutamine Metabolism in Cancer: Understanding the Heterogeneity. Trends Cancer 2017, 3, 169–180. [Google Scholar] [CrossRef] [Green Version]
- Daye, D.; Wellen, K.E. Metabolic reprogramming in cancer: Unraveling the role of glutamine in tumorigenesis. Semin. Cell Dev. Biol. 2012, 23, 362–369. [Google Scholar] [CrossRef]
- Hammami, I.; Chen, J.; Bronte, V.; DeCrescenzo, G.; Jolicoeur, M. L-glutamine is a key parameter in the immunosuppression phenomenon. Biochem. Biophys. Res. Commun. 2012, 425, 724–729. [Google Scholar] [CrossRef] [PubMed]
- Leone, R.D.; Zhao, L.; Englert, J.M.; Sun, I.M.; Oh, M.H.; Sun, I.H.; Arwood, M.L.; Bettencourt, I.A.; Patel, C.H.; Wen, J.; et al. Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion. Science 2019, 366, 1013–1021. [Google Scholar] [CrossRef]
- Oh, M.H.; Sun, I.H.; Zhao, L.; Leone, R.D.; Sun, I.M.; Xu, W.; Collins, S.L.; Tam, A.J.; Blosser, R.L.; Patel, C.H.; et al. Targeting glutamine metabolism enhances tumor-specific immunity by modulating suppressive myeloid cells. J. Clin. Investig. 2020, 130, 3865–3884. [Google Scholar] [CrossRef] [PubMed]
- Ostrand-Rosenberg, S. Myeloid-derived suppressor cells: More mechanisms for inhibiting antitumor immunity. Cancer Immunol. Immunother. 2010, 59, 1593–1600. [Google Scholar] [CrossRef] [Green Version]
- Srivastava, M.K.; Sinha, P.; Clements, V.K.; Rodriguez, P.; Ostrand-Rosenberg, S. Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer Res. 2010, 70, 68–77. [Google Scholar] [CrossRef] [Green Version]
- Procaccini, C.; Garavelli, S.; Carbone, F.; Di Silvestre, D.; La Rocca, C.; Greco, D.; Colamatteo, A.; Lepore, M.T.; Russo, C.; De Rosa, G.; et al. Signals of pseudo-starvation unveil the amino acid transporter SLC7A11 as key determinant in the control of Treg cell proliferative potential. Immunity 2021, 54, 1543–1560.e6. [Google Scholar] [CrossRef]
- Cramer, S.L.; Saha, A.; Liu, J.; Tadi, S.; Tiziani, S.; Yan, W.; Triplett, K.; Lamb, C.; Alters, S.E.; Rowlinson, S.; et al. Systemic depletion of L-cyst(e)ine with cyst(e)inase increases reactive oxygen species and suppresses tumor growth. Nat. Med. 2017, 23, 120–127. [Google Scholar] [CrossRef]
- Jiang, X.; Stockwell, B.R.; Conrad, M. Ferroptosis: Mechanisms, biology and role in disease. Nat. Rev. Mol. Cell Biol. 2021, 22, 266–282. [Google Scholar] [CrossRef]
- Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef] [Green Version]
- Zhu, H.; Klement, J.D.; Lu, C.; Redd, P.S.; Yang, D.; Smith, A.D.; Poschel, D.B.; Zou, J.; Liu, D.; Wang, P.G.; et al. Asah2 Represses the p53-Hmox1 Axis to Protect Myeloid-Derived Suppressor Cells from Ferroptosis. J. Immunol. 2021, 206, 1395–1404. [Google Scholar] [CrossRef]
- Ugolini, A.; Tyurin, V.A.; Tyurina, Y.Y.; Tcyganov, E.N.; Donthireddy, L.; Kagan, V.E.; Gabrilovich, D.I.; Veglia, F. Polymorphonuclear myeloid-derived suppressor cells limit antigen cross-presentation by dendritic cells in cancer. JCI Insight 2020, 5, e138581. [Google Scholar] [CrossRef] [PubMed]
- Zeitler, L.; Fiore, A.; Meyer, C.; Russier, M.; Zanella, G.; Suppmann, S.; Gargaro, M.; Sidhu, S.S.; Seshagiri, S.; Ohnmacht, C.; et al. Anti-ferroptotic mechanism of IL4i1-mediated amino acid metabolism. eLife 2021, 10, e64806. [Google Scholar] [CrossRef]
- Bader, J.E.; Voss, K.; Rathmell, J.C. Targeting Metabolism to Improve the Tumor Microenvironment for Cancer Immunotherapy. Mol. Cell 2020, 78, 1019–1033. [Google Scholar] [CrossRef] [PubMed]
- Hossain, F.; Al-Khami, A.A.; Wyczechowska, D.; Hernandez, C.; Zheng, L.; Reiss, K.; Valle, L.D.; Trillo-Tinoco, J.; Maj, T.; Zou, W.; et al. Inhibition of Fatty Acid Oxidation Modulates Immunosuppressive Functions of Myeloid-Derived Suppressor Cells and Enhances Cancer Therapies. Cancer Immunol. Res. 2015, 3, 1236–1247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Al-Khami, A.A.; Zheng, L.; Del Valle, L.; Hossain, F.; Wyczechowska, D.; Zabaleta, J.; Sanchez, M.D.; Dean, M.J.; Rodriguez, P.C.; Ochoa, A.C. Exogenous lipid uptake induces metabolic and functional reprogramming of tumor-associated myeloid-derived suppressor cells. Oncoimmunology 2017, 6, e1344804. [Google Scholar] [CrossRef]
- Thevenot, P.T.; Sierra, R.A.; Raber, P.L.; Al-Khami, A.A.; Trillo-Tinoco, J.; Zarreii, P.; Ochoa, A.C.; Cui, Y.; Del Valle, L.; Rodriguez, P.C. The stress-response sensor chop regulates the function and accumulation of myeloid-derived suppressor cells in tumors. Immunity 2014, 41, 389–401. [Google Scholar] [CrossRef] [Green Version]
- Yan, D.; Wang, H.W.; Bowman, R.L.; Joyce, J.A. STAT3 and STAT6 Signaling Pathways Synergize to Promote Cathepsin Secretion from Macrophages via IRE1alpha Activation. Cell Rep. 2016, 16, 2914–2927. [Google Scholar] [CrossRef] [Green Version]
- Hetz, C. The unfolded protein response: Controlling cell fate decisions under ER stress and beyond. Nat. Rev. Mol. Cell Biol. 2012, 13, 89–102. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.; Cubillos-Ruiz, J.R. Endoplasmic reticulum stress signals in the tumour and its microenvironment. Nat. Rev. Cancer 2021, 21, 71–88. [Google Scholar] [CrossRef]
- Cubillos-Ruiz, J.R.; Silberman, P.C.; Rutkowski, M.R.; Chopra, S.; Perales-Puchalt, A.; Song, M.; Zhang, S.; Bettigole, S.E.; Gupta, D.; Holcomb, K.; et al. ER Stress Sensor XBP1 Controls Anti-tumor Immunity by Disrupting Dendritic Cell Homeostasis. Cell 2015, 161, 1527–1538. [Google Scholar] [CrossRef] [Green Version]
- Shoulders, M.D.; Ryno, L.M.; Genereux, J.C.; Moresco, J.J.; Tu, P.G.; Wu, C.; Yates, J.R., 3rd; Su, A.I.; Kelly, J.W.; Wiseman, R.L. Stress-independent activation of XBP1s and/or ATF6 reveals three functionally diverse ER proteostasis environments. Cell Rep. 2013, 3, 1279–1292. [Google Scholar] [CrossRef] [Green Version]
- West, A.P.; Khoury-Hanold, W.; Staron, M.; Tal, M.C.; Pineda, C.M.; Lang, S.M.; Bestwick, M.; Duguay, B.A.; Raimundo, N.; MacDuff, D.A.; et al. Mitochondrial DNA stress primes the antiviral innate immune response. Nature 2015, 520, 553–557. [Google Scholar] [CrossRef] [Green Version]
- Alicea-Torres, K.; Sanseviero, E.; Gui, J.; Chen, J.; Veglia, F.; Yu, Q.; Donthireddy, L.; Kossenkov, A.; Lin, C.; Fu, S.; et al. Immune suppressive activity of myeloid-derived suppressor cells in cancer requires inactivation of the type I interferon pathway. Nat. Commun. 2021, 12, 1717. [Google Scholar] [CrossRef] [PubMed]
- Yan, D.; Adeshakin, A.O.; Xu, M.; Afolabi, L.O.; Zhang, G.; Chen, Y.H.; Wan, X. Lipid Metabolic Pathways Confer the Immunosuppressive Function of Myeloid-Derived Suppressor Cells in Tumor. Front. Immunol. 2019, 10, 1399. [Google Scholar] [CrossRef] [PubMed]
- Murdocca, M.; De Masi, C.; Pucci, S.; Mango, R.; Novelli, G.; Di Natale, C.; Sangiuolo, F. LOX-1 and cancer: An indissoluble liaison. Cancer Gene Ther. 2021, 1, 1–11. [Google Scholar] [CrossRef]
- Chai, E.; Zhang, L.; Li, C. LOX-1+ PMN-MDSC enhances immune suppression which promotes glioblastoma multiforme progression. Cancer Manag. Res. 2019, 11, 7307–7315. [Google Scholar] [CrossRef] [Green Version]
- Pencheva, N.; Buss, C.G.; Posada, J.; Merghoub, T.; Tavazoie, S.F. Broad-spectrum therapeutic suppression of metastatic melanoma through nuclear hormone receptor activation. Cell 2014, 156, 986–1001. [Google Scholar] [CrossRef] [Green Version]
- Tavazoie, M.F.; Pollack, I.; Tanqueco, R.; Ostendorf, B.N.; Reis, B.S.; Gonsalves, F.C.; Kurth, I.; Andreu-Agullo, C.; Derbyshire, M.L.; Posada, J.; et al. LXR/ApoE Activation Restricts Innate Immune Suppression in Cancer. Cell 2018, 172, 825–840.e18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yan, D.; Yang, Q.; Shi, M.; Zhong, L.; Wu, C.; Meng, T.; Yin, H.; Zhou, J. Polyunsaturated fatty acids promote the expansion of myeloid-derived suppressor cells by activating the JAK/STAT3 pathway. Eur. J. Immunol. 2013, 43, 2943–2955. [Google Scholar] [CrossRef] [PubMed]
- Adeshakin, A.O.; Liu, W.; Adeshakin, F.O.; Afolabi, L.O.; Zhang, M.; Zhang, G.; Wang, L.; Li, Z.; Lin, L.; Cao, Q.; et al. Regulation of ROS in myeloid-derived suppressor cells through targeting fatty acid transport protein 2 enhanced anti-PD-L1 tumor immunotherapy. Cell Immunol. 2021, 362, 104286. [Google Scholar] [CrossRef]
- Veglia, F.; Tyurin, V.A.; Blasi, M.; De Leo, A.; Kossenkov, A.V.; Donthireddy, L.; To, T.K.J.; Schug, Z.; Basu, S.; Wang, F.; et al. Fatty acid transport protein 2 reprograms neutrophils in cancer. Nature 2019, 569, 73–78. [Google Scholar] [CrossRef] [PubMed]
- Ugel, S.; Delpozzo, F.; Desantis, G.; Papalini, F.; Simonato, F.; Sonda, N.; Zilio, S.; Bronte, V. Therapeutic targeting of myeloid-derived suppressor cells. Curr. Opin. Pharmacol. 2009, 9, 470–481. [Google Scholar] [CrossRef]
- Wang, Y.; Jia, A.; Bi, Y.; Wang, Y.; Liu, G. Metabolic Regulation of Myeloid-Derived Suppressor Cell Function in Cancer. Cells 2020, 9, 1011. [Google Scholar] [CrossRef] [Green Version]
- Qin, H.; Lerman, B.; Sakamaki, I.; Wei, G.; Cha, S.C.; Rao, S.S.; Qian, J.; Hailemichael, Y.; Nurieva, R.; Dwyer, K.C.; et al. Generation of a new therapeutic peptide that depletes myeloid-derived suppressor cells in tumor-bearing mice. Nat. Med. 2014, 20, 676–681. [Google Scholar] [CrossRef]
- Lancet, J.E.; Moseley, A.B.; Coutre, S.E.; DeAngelo, D.J.; Othus, M.; Tallman, M.S.; Litzow, M.R.; Komrokji, R.S.; Erba, H.P.; Appelbaum, F.R. A phase 2 study of ATRA, arsenic trioxide, and gemtuzumab ozogamicin in patients with high-risk APL (SWOG 0535). Blood Adv. 2020, 4, 1683–1689. [Google Scholar] [CrossRef] [Green Version]
- Li, X.; Yao, W.; Yuan, Y.; Chen, P.; Li, B.; Li, J.; Chu, R.; Song, H.; Xie, D.; Jiang, X.; et al. Targeting of tumour-infiltrating macrophages via CCL2/CCR2 signalling as a therapeutic strategy against hepatocellular carcinoma. Gut 2017, 66, 157–167. [Google Scholar] [CrossRef]
- Chun, E.; Lavoie, S.; Michaud, M.; Gallini, C.A.; Kim, J.; Soucy, G.; Odze, R.; Glickman, J.N.; Garrett, W.S. CCL2 Promotes Colorectal Carcinogenesis by Enhancing Polymorphonuclear Myeloid-Derived Suppressor Cell Population and Function. Cell Rep. 2015, 12, 244–257. [Google Scholar] [CrossRef] [Green Version]
- Wang, G.; Lu, X.; Dey, P.; Deng, P.; Wu, C.C.; Jiang, S.; Fang, Z.; Zhao, K.; Konaparthi, R.; Hua, S.; et al. Targeting YAP-Dependent MDSC Infiltration Impairs Tumor Progression. Cancer Discov. 2016, 6, 80–95. [Google Scholar] [CrossRef] [Green Version]
- Lu, Z.; Zou, J.; Li, S.; Topper, M.J.; Tao, Y.; Zhang, H.; Jiao, X.; Xie, W.; Kong, X.; Vaz, M.; et al. Epigenetic therapy inhibits metastases by disrupting premetastatic niches. Nature 2020, 579, 284–290. [Google Scholar] [CrossRef] [PubMed]
- Nefedova, Y.; Fishman, M.; Sherman, S.; Wang, X.; Beg, A.A.; Gabrilovich, D.I. Mechanism of all-trans retinoic acid effect on tumor-associated myeloid-derived suppressor cells. Cancer Res. 2007, 67, 11021–11028. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nefedova, Y.; Nagaraj, S.; Rosenbauer, A.; Muro-Cacho, C.; Sebti, S.M.; Gabrilovich, D.I. Regulation of dendritic cell differentiation and antitumor immune response in cancer by pharmacologic-selective inhibition of the janus-activated kinase 2/signal transducers and activators of transcription 3 pathway. Cancer Res. 2005, 65, 9525–9535. [Google Scholar] [CrossRef] [Green Version]
- Edwards, J.P.; Emens, L.A. The multikinase inhibitor sorafenib reverses the suppression of IL-12 and enhancement of IL-10 by PGE(2) in murine macrophages. Int. Immunopharmacol. 2010, 10, 1220–1228. [Google Scholar] [CrossRef] [Green Version]
- Deng, Y.; Yang, J.; Luo, F.; Qian, J.; Liu, R.; Zhang, D.; Yu, H.; Chu, Y. mTOR-mediated glycolysis contributes to the enhanced suppressive function of murine tumor-infiltrating monocytic myeloid-derived suppressor cells. Cancer Immunol. Immunother. 2018, 67, 1355–1364. [Google Scholar] [CrossRef] [PubMed]
- Meng, G.; Li, B.; Chen, A.; Zheng, M.; Xu, T.; Zhang, H.; Dong, J.; Wu, J.; Yu, D.; Wei, J. Targeting aerobic glycolysis by dichloroacetate improves Newcastle disease virus-mediated viro-immunotherapy in hepatocellular carcinoma. Br. J. Cancer 2020, 122, 111–120. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Shen, L.; Liu, Q.; Hou, L.; Huang, L. Inhibiting PI3 kinase-gamma in both myeloid and plasma cells remodels the suppressive tumor microenvironment in desmoplastic tumors. J. Control. Release 2019, 309, 173–180. [Google Scholar] [CrossRef]
- Hirsch, E.; Katanaev, V.L.; Garlanda, C.; Azzolino, O.; Pirola, L.; Silengo, L.; Sozzani, S.; Mantovani, A.; Altruda, F.; Wymann, M.P. Central role for G protein-coupled phosphoinositide 3-kinase gamma in inflammation. Science 2000, 287, 1049–1053. [Google Scholar] [CrossRef]
- Parker, K.H.; Beury, D.W.; Ostrand-Rosenberg, S. Myeloid-Derived Suppressor Cells: Critical Cells Driving Immune Suppression in the Tumor Microenvironment. Adv. Cancer Res. 2015, 128, 95–139. [Google Scholar] [CrossRef] [Green Version]
- Wong, A.L.A.; Hirpara, J.L.; Pervaiz, S.; Eu, J.Q.; Sethi, G.; Goh, B.C. Do STAT3 inhibitors have potential in the future for cancer therapy? Expert Opin. Investig. Drugs 2017, 26, 883–887. [Google Scholar] [CrossRef] [Green Version]
- Reilley, M.J.; McCoon, P.; Cook, C.; Lyne, P.; Kurzrock, R.; Kim, Y.; Woessner, R.; Younes, A.; Nemunaitis, J.; Fowler, N.; et al. STAT3 antisense oligonucleotide AZD9150 in a subset of patients with heavily pretreated lymphoma: Results of a phase 1b trial. J. Immunother. Cancer 2018, 6, 119. [Google Scholar] [CrossRef] [Green Version]
- Chen, H.M.; Ma, G.; Gildener-Leapman, N.; Eisenstein, S.; Coakley, B.A.; Ozao, J.; Mandeli, J.; Divino, C.; Schwartz, M.; Sung, M.; et al. Myeloid-Derived Suppressor Cells as an Immune Parameter in Patients with Concurrent Sunitinib and Stereotactic Body Radiotherapy. Clin. Cancer Res. 2015, 21, 4073–4085. [Google Scholar] [CrossRef] [Green Version]
- Khan, M.W.; Saadalla, A.; Ewida, A.H.; Al-Katranji, K.; Al-Saoudi, G.; Giaccone, Z.T.; Gounari, F.; Zhang, M.; Frank, D.A.; Khazaie, K. The STAT3 inhibitor pyrimethamine displays anti-cancer and immune stimulatory effects in murine models of breast cancer. Cancer Immunol. Immunother. 2018, 67, 13–23. [Google Scholar] [CrossRef] [PubMed]
- Li, H.; Liu, A.; Zhao, Z.; Xu, Y.; Lin, J.; Jou, D.; Li, C. Fragment-based drug design and drug repositioning using multiple ligand simultaneous docking (MLSD): Identifying celecoxib and template compounds as novel inhibitors of signal transducer and activator of transcription 3 (STAT3). J. Med. Chem. 2011, 54, 5592–5596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bronte, V.; Ugel, S.; Tinazzi, E.; Vella, A.; De Sanctis, F.; Cane, S.; Batani, V.; Trovato, R.; Fiore, A.; Petrova, V.; et al. Baricitinib restrains the immune dysregulation in patients with severe COVID-19. J. Clin. Investig. 2020, 130, 6409–6416. [Google Scholar] [CrossRef] [PubMed]
- Proietti, E.; Rossini, S.; Grohmann, U.; Mondanelli, G. Polyamines and Kynurenines at the Intersection of Immune Modulation. Trends Immunol. 2020, 41, 1037–1050. [Google Scholar] [CrossRef]
- Alexander, E.T.; Minton, A.; Peters, M.C.; Phanstiel, O.T.; Gilmour, S.K. A novel polyamine blockade therapy activates an anti-tumor immune response. Oncotarget 2017, 8, 84140–84152. [Google Scholar] [CrossRef] [Green Version]
- Alexander, E.T.; Mariner, K.; Donnelly, J.; Phanstiel, O.t.; Gilmour, S.K. Polyamine Blocking Therapy Decreases Survival of Tumor-Infiltrating Immunosuppressive Myeloid Cells and Enhances the Antitumor Efficacy of PD-1 Blockade. Mol. Cancer Ther. 2020, 19, 2012–2022. [Google Scholar] [CrossRef]
- Steggerda, S.M.; Bennett, M.K.; Chen, J.; Emberley, E.; Huang, T.; Janes, J.R.; Li, W.; MacKinnon, A.L.; Makkouk, A.; Marguier, G.; et al. Inhibition of arginase by CB-1158 blocks myeloid cell-mediated immune suppression in the tumor microenvironment. J. Immunother. Cancer 2017, 5, 101. [Google Scholar] [CrossRef]
- Serafini, P.; Meckel, K.; Kelso, M.; Noonan, K.; Califano, J.; Koch, W.; Dolcetti, L.; Bronte, V.; Borrello, I. Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function. J. Exp. Med. 2006, 203, 2691–2702. [Google Scholar] [CrossRef]
- Lin, S.; Wang, J.; Wang, L.; Wen, J.; Guo, Y.; Qiao, W.; Zhou, J.; Xu, G.; Zhi, F. Phosphodiesterase-5 inhibition suppresses colonic inflammation-induced tumorigenesis via blocking the recruitment of MDSC. Am. J. Cancer Res. 2017, 7, 41–52. [Google Scholar]
- Califano, J.A.; Khan, Z.; Noonan, K.A.; Rudraraju, L.; Zhang, Z.; Wang, H.; Goodman, S.; Gourin, C.G.; Ha, P.K.; Fakhry, C.; et al. Tadalafil augments tumor specific immunity in patients with head and neck squamous cell carcinoma. Clin. Cancer Res. 2015, 21, 30–38. [Google Scholar] [CrossRef] [Green Version]
- Hassel, J.C.; Jiang, H.; Bender, C.; Winkler, J.; Sevko, A.; Shevchenko, I.; Halama, N.; Dimitrakopoulou-Strauss, A.; Haefeli, W.E.; Jager, D.; et al. Tadalafil has biologic activity in human melanoma. Results of a pilot trial with Tadalafil in patients with metastatic Melanoma (TaMe). Oncoimmunology 2017, 6, e1326440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nayak-Kapoor, A.; Hao, Z.; Sadek, R.; Dobbins, R.; Marshall, L.; Vahanian, N.N.; Jay Ramsey, W.; Kennedy, E.; Mautino, M.R.; Link, C.J.; et al. Phase Ia study of the indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor navoximod (GDC-0919) in patients with recurrent advanced solid tumors. J. Immunother. Cancer 2018, 6, 61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jung, K.H.; LoRusso, P.; Burris, H.; Gordon, M.; Bang, Y.J.; Hellmann, M.D.; Cervantes, A.; Ochoa de Olza, M.; Marabelle, A.; Hodi, F.S.; et al. Phase I Study of the Indoleamine 2,3-Dioxygenase 1 (IDO1) Inhibitor Navoximod (GDC-0919) Administered with PD-L1 Inhibitor (Atezolizumab) in Advanced Solid Tumors. Clin. Cancer Res. 2019, 25, 3220–3228. [Google Scholar] [CrossRef] [Green Version]
- Veltman, J.D.; Lambers, M.E.; van Nimwegen, M.; Hendriks, R.W.; Hoogsteden, H.C.; Aerts, J.G.; Hegmans, J.P. COX-2 inhibition improves immunotherapy and is associated with decreased numbers of myeloid-derived suppressor cells in mesothelioma. Celecoxib influences MDSC function. BMC Cancer 2010, 10, 464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rodriguez, P.C.; Hernandez, C.P.; Quiceno, D.; Dubinett, S.M.; Zabaleta, J.; Ochoa, J.B.; Gilbert, J.; Ochoa, A.C. Arginase I in myeloid suppressor cells is induced by COX-2 in lung carcinoma. J. Exp. Med. 2005, 202, 931–939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stadtmauer, E.A.; Fraietta, J.A.; Davis, M.M.; Cohen, A.D.; Weber, K.L.; Lancaster, E.; Mangan, P.A.; Kulikovskaya, I.; Gupta, M.; Chen, F.; et al. CRISPR-engineered T cells in patients with refractory cancer. Science 2020, 367, eaba7365. [Google Scholar] [CrossRef]
- Knott, G.J.; Doudna, J.A. CRISPR-Cas guides the future of genetic engineering. Science 2018, 361, 866–869. [Google Scholar] [CrossRef] [Green Version]
- Sonda, N.; Simonato, F.; Peranzoni, E.; Cali, B.; Bortoluzzi, S.; Bisognin, A.; Wang, E.; Marincola, F.M.; Naldini, L.; Gentner, B.; et al. miR-142-3p prevents macrophage differentiation during cancer-induced myelopoiesis. Immunity 2013, 38, 1236–1249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kaczanowska, S.; Beury, D.W.; Gopalan, V.; Tycko, A.K.; Qin, H.; Clements, M.E.; Drake, J.; Nwanze, C.; Murgai, M.; Rae, Z.; et al. Genetically engineered myeloid cells rebalance the core immune suppression program in metastasis. Cell 2021, 184, 2033–2052.e21. [Google Scholar] [CrossRef]
- Sasso, M.S.; Lollo, G.; Pitorre, M.; Solito, S.; Pinton, L.; Valpione, S.; Bastiat, G.; Mandruzzato, S.; Bronte, V.; Marigo, I.; et al. Low dose gemcitabine-loaded lipid nanocapsules target monocytic myeloid-derived suppressor cells and potentiate cancer immunotherapy. Biomaterials 2016, 96, 47–62. [Google Scholar] [CrossRef]
- Zhang, Y.; Bush, X.; Yan, B.; Chen, J.A. Gemcitabine nanoparticles promote antitumor immunity against melanoma. Biomaterials 2019, 189, 48–59. [Google Scholar] [CrossRef] [PubMed]
- Pinton, L.; Magri, S.; Masetto, E.; Vettore, M.; Schibuola, I.; Ingangi, V.; Marigo, I.; Matha, K.; Benoit, J.P.; Della Puppa, A.; et al. Targeting of immunosuppressive myeloid cells from glioblastoma patients by modulation of size and surface charge of lipid nanocapsules. J. Nanobiotechnol. 2020, 18, 31. [Google Scholar] [CrossRef]
- Wan, D.; Yang, Y.; Liu, Y.; Cun, X.; Li, M.; Xu, S.; Zhao, W.; Xiang, Y.; Qiu, Y.; Yu, Q.; et al. Sequential depletion of myeloid-derived suppressor cells and tumor cells with a dual-pH-sensitive conjugated micelle system for cancer chemoimmunotherapy. J. Control. Release 2020, 317, 43–56. [Google Scholar] [CrossRef]
- Burslem, G.M.; Crews, C.M. Proteolysis-Targeting Chimeras as Therapeutics and Tools for Biological Discovery. Cell 2020, 181, 102–114. [Google Scholar] [CrossRef]
- Weng, G.; Shen, C.; Cao, D.; Gao, J.; Dong, X.; He, Q.; Yang, B.; Li, D.; Wu, J.; Hou, T. PROTAC-DB: An online database of PROTACs. Nucleic Acids Res. 2021, 49, D1381–D1387. [Google Scholar] [CrossRef] [PubMed]
- Raedler, L.A. Revlimid (Lenalidomide) Now FDA Approved as First-Line Therapy for Patients with Multiple Myeloma. Am. Health Drug Benefits 2016, 9, 140–143. [Google Scholar] [PubMed]
- Mougiakakos, D.; Bach, C.; Bottcher, M.; Beier, F.; Rohner, L.; Stoll, A.; Rehli, M.; Gebhard, C.; Lischer, C.; Eberhardt, M.; et al. The IKZF1-IRF4/IRF5 Axis Controls Polarization of Myeloma-Associated Macrophages. Cancer Immunol. Res. 2021, 9, 265–278. [Google Scholar] [CrossRef]
- Mullard, A. Targeted protein degraders crowd into the clinic. Nat. Rev. Drug Discov. 2021, 20, 247–250. [Google Scholar] [CrossRef] [PubMed]
- Kim, K.; Skora, A.D.; Li, Z.; Liu, Q.; Tam, A.J.; Blosser, R.L.; Diaz, L.A., Jr.; Papadopoulos, N.; Kinzler, K.W.; Vogelstein, B.; et al. Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cells. Proc. Natl. Acad. Sci. USA 2014, 111, 11774–11779. [Google Scholar] [CrossRef] [Green Version]
- Davis, R.J.; Moore, E.C.; Clavijo, P.E.; Friedman, J.; Cash, H.; Chen, Z.; Silvin, C.; Van Waes, C.; Allen, C. Anti-PD-L1 Efficacy Can Be Enhanced by Inhibition of Myeloid-Derived Suppressor Cells with a Selective Inhibitor of PI3Kdelta/gamma. Cancer Res. 2017, 77, 2607–2619. [Google Scholar] [CrossRef] [Green Version]
- Weber, R.; Fleming, V.; Hu, X.; Nagibin, V.; Groth, C.; Altevogt, P.; Utikal, J.; Umansky, V. Myeloid-Derived Suppressor Cells Hinder the Anti-Cancer Activity of Immune Checkpoint Inhibitors. Front. Immunol. 2018, 9, 1310. [Google Scholar] [CrossRef] [Green Version]
M-MDSCs | PMN-MDSCs | Early-MDSCs | |
---|---|---|---|
CD45 | + | + | + |
CD11b | + | + | + |
CD33 | + | + | + |
HLA-DR | low/− | − | − |
IL4Rα/CD124 | + | + | ND |
CD34 | + | ND | + |
CD84 | + | + | ND |
DR5 | + | + | ND |
CD14 | + | - | ND |
CD15 | − | + | ND |
Lin (CD3, CD19, CD20, CD56) | ND | ND | − |
CXCR1 | + | low | ND |
CD66b | − | + | ND |
CD117 | ND | ND | + |
CD38 | + | + | ND |
PD-L1/2 | + | ND | ND |
LOX1 | ND | + | ND |
FATP2 | ND | + | ND |
FSC | low | low | low |
SSC | low | high | high |
NCT | Drug | Target | Combination Therapy | Phase | Effect | Tumour Type |
---|---|---|---|---|---|---|
NCT02961257 | Granulocyte colony-stimulating factor (G-CSF) | Growth factor modulation | Prednisone, cabazitaxel | III | MDSC depletion | Prostate cancer, metastatic |
NCT02880371 | ARRY-382 (cFMS tyrosine kinase inhibitor) | CSF1R inhibitor | Pembrolizumab | II | MDSC depletion | Advanced solid tumours |
NCT02554812 | PD 0360324 (M-CSF mAb) | CSF1R inhibitor | Utomilumab, PF-04518600, avelumab, CMP-001 | II | MDSC depletion | Locally advanced or metastatic solid tumours |
NCT02777710 | Pexidartinib (CSF-1R TKI) | CSF1R inhibitor | Durvalumab | I | MDSC depletion | Colorectal cancer, pancreatic cancer, metastatic cancer, and advanced cancer |
NCT01349036 | Pexidartinib hydrochloride (PLX3397) | binds to and inhibits phosphorylation of KIT, CSF1R and FLT3 | II | MDSC trafficking | Recurrent glioblastoma | |
NCT02370238 | Reparixin | CXCR1/2 inhibitor | Paclitaxel | II | MDSC trafficking | Metastatic triple-negative breast cancer |
NCT03177187 | AZD5069 | CXCR2 antagonist | Enzalutamide | II | MDSC trafficking | Metastatic castration-resistant prostate cancer |
NCT03161431 | SX-682 | CXCR1/2 inhibitor | Pembrolizumab | I | MDSC recruitment | Metastatic melanoma |
NCT02403778 | ATRA | Retinoic acid receptor inhibitor | Ipilimumab | II | Inhibition of MDSC immunosuppression | Advanced melanoma |
NCT02637531 | IPI-549 (Eganelisib) | PI3K-gamma inhibitor | Nivolumab | I | MDSC reprogramming and decreased immunosuppression | Advanced solid tumours |
NCT03961698 | IPI-549 (Eganelisib) | PI3K-gamma inhibitor | Atezolizumab, nab-paclitaxel, bevacizumab | II | Inhibition of MDSC immunosuppression | Triple-negative breast cancer and renal cell carcinoma |
NCT01839604 | AZD9150 | STAT3 antisense oligonucleotide | I | Inhibition of MDSC immunosuppression | Advanced/metastatic hepatocellular carcinoma | |
NCT01112397 | AZD1480 | JAK1/2 inhibitor | I | Inhibition of MDSC immunosuppression | Advanced solid tumours | |
NCT01423058 | Momelotinib (CYT387) | JAK1/2 inhibitor | I/II | Inhibition of MDSC immunosuppression | Myeloproliferative neoplasms | |
NCT03427866 | Ruxolitinib | JAK1/2 inhibitor | II | Inhibition of MDSC immunosuppression | Myelofibrosis | |
NCT01594723 | LY2784544 | JAK2 inhibitor | II | Inhibition of MDSC immunosuppression | Myeloproliferative neoplasms | |
NCT02055781 | Pacritinib | JAK inhibitor | III | Inhibition of MDSC immunosuppression | Myeloproliferative neoplasms | |
NCT03315026 | Siltuximab | IL-6 inhibitor | II | Inhibition of MDSC immunosuppression | Multiple myeloma or systemic amyloidosis | |
NCT02997956 | Tocilizumab | IL-6 inhibitor | II | Inhibition of MDSC immunosuppression | Hepatocellular carcinoma | |
NCT02903914 | CB-1158 (INCB001158) | Arginase inhibitor | Pembrolizumab | I/II | Inhibition of MDSC immunosuppression | Advanced/metastatic solid tumors |
NCT02544880 | Tadalafil | PDE-5 inhibitor | Pembrolizumab | I | Inhibition of MDSC functions | Head and neck squamous cell carcinoma, head and neck cancer |
EudraCT-No:2011-003273-28 | Tadalafil | PDE-5 inhibitor | Metastatic melanoma | |||
NCT02048709 | Navoximod (GDC-0919) | IDO1 inhibitor | I | Solid tumour | ||
NCT02471846 | Navoximod (GDC-0919) | IDO1 inhibitor | Atezolizumab | I | Advanced/metastatic solid tumours | |
NCT04471415 | Sirpiglenastat (DRP-104) | Glutamin antagonist | Atezolizumab | I/II | Advanced solid tumours | |
NCT03026140 | Celecoxib | COX-2 inhibitor | Nivolumab, ipilimumab | II | Inhibition of MDSC accumulation | Colon carcinoma |
NCT04188119 | Aspirin | COX-2 inhibitor | Avelumab, lansoprazole | II | Inhibition of MDSC accumulation | Triple negative breast cancer |
NCT04348747 | Celecoxib | COX-2 inhibitor | Rintatolimod | II | Inhibition of MDSC accumulation | Metastatic breast cancer |
NCT03245489 | Acetylsalicylic acid | COX-2 inhibitor | Pembrolizumab, clopidogrel | I | Inhibition of MDSC accumulation | Head and neck squamous cell carcinoma |
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
Hofer, F.; Di Sario, G.; Musiu, C.; Sartoris, S.; De Sanctis, F.; Ugel, S. A Complex Metabolic Network Confers Immunosuppressive Functions to Myeloid-Derived Suppressor Cells (MDSCs) within the Tumour Microenvironment. Cells 2021, 10, 2700. https://doi.org/10.3390/cells10102700
Hofer F, Di Sario G, Musiu C, Sartoris S, De Sanctis F, Ugel S. A Complex Metabolic Network Confers Immunosuppressive Functions to Myeloid-Derived Suppressor Cells (MDSCs) within the Tumour Microenvironment. Cells. 2021; 10(10):2700. https://doi.org/10.3390/cells10102700
Chicago/Turabian StyleHofer, Francesca, Gianna Di Sario, Chiara Musiu, Silvia Sartoris, Francesco De Sanctis, and Stefano Ugel. 2021. "A Complex Metabolic Network Confers Immunosuppressive Functions to Myeloid-Derived Suppressor Cells (MDSCs) within the Tumour Microenvironment" Cells 10, no. 10: 2700. https://doi.org/10.3390/cells10102700
APA StyleHofer, F., Di Sario, G., Musiu, C., Sartoris, S., De Sanctis, F., & Ugel, S. (2021). A Complex Metabolic Network Confers Immunosuppressive Functions to Myeloid-Derived Suppressor Cells (MDSCs) within the Tumour Microenvironment. Cells, 10(10), 2700. https://doi.org/10.3390/cells10102700