1,767
Views
1
CrossRef citations to date
0
Altmetric
Original Research

Decreased melanoma CSF-1 secretion by Cannabigerol treatment reprograms regulatory myeloid cells and reduces tumor progression

, , , , , , , , & show all
Article: 2219164 | Received 15 Jan 2023, Accepted 24 May 2023, Published online: 11 Jun 2023

References

  • Galli F, Aguilera JV, Palermo B, Markovic SN, Nisticò P, Signore A. Relevance of immune cell and tumor microenvironment imaging in the new era of immunotherapy. J Exp Clin Cancer Res. 2020;39(1):1–14. doi:10.1186/s13046-020-01586-y.
  • Tcyganov E, Mastio J, Chen E, Gabrilovich DI. Plasticity of myeloid-derived suppressor cells in cancer. Curr Opin Immunol. 2019;51:76–82. doi:10.1016/j.coi.2018.03.009.
  • Monteran L, Ershaid N, Doron H, Zait Y, Scharff Y, Ben-Yosef S, Avivi C, Barshack I, Sonnenblick A, Erez N. Chemotherapy-induced complement signal- ing modulates immunosuppression and metastatic relapse in breast cancer. Nat Commun. 2022;13(1). doi:10.1038/s41467-022-33598-x.
  • Condamine T, Gabrilovich DI. Molecular mechanisms regulating myeloid-derived suppressor cell differentiation and function. Trends Immunol. 2011;32(1):19–25. doi:10.1016/j.it.2010.10.002.
  • Laoui D, Van Overmeire E, De Baetselier P, Van Ginderachter JA, Raes G. Functional relationship between tumor-associated macrophages and macrophage colony-stimulating factor as contributors to cancer progression. Front Immunol. 2014;5(October):1–15. doi:10.3389/fimmu.2014.00489.
  • Greten TF. Does CSF1R Blockade turn into friendly fire? Cancer Cell. 2018;32(5):546–547. doi:10.1016/j.ccell.2017.10.012.
  • Ma T, Renz BW, Ilmer M, Koch D, Yang Y, Werner J, Bazhin AV. Myeloid-derived suppressor cells in solid tumors. Cells. 2022;11(2):310–322. doi:10.3390/cells11020310.
  • Li T, Li X, Zamani A, Wang W, Lee C-N, Li M, Luo G, Eiler E, Sun H, Ghosh S, et al. C-Rel is a myeloid checkpoint for cancer immunotherapy. Nat Cancer. 2020;1(5):507–517. doi:10.1038/s43018-020-0061-3.
  • Geiger R, Rieckmann JC, Wolf T, Basso C, Feng Y, Fuhrer T, Kogadeeva M, Picotti P, Meissner F, Mann M, et al. L-Arginine modulates T cell metabolism and enhances survival and anti-tumor activity article L-Arginine modulates T cell metabolism and enhances survival and anti-tumor activity. Cell. 2016;167(3):829–842.e13. doi:10.1016/j.cell.2016.09.031.
  • Fares CM, Van Allen EM, Drake CG, Allison JP, Hu-Lieskovan S. Mechanisms of Resistance to Immune Checkpoint Blockade: why Does Checkpoint Inhibitor Immunotherapy Not Work for All Patients? Am Soc Clin Oncol Educ B. 2022;39:147–164. doi:10.1200/EDBK_240837.
  • Ries CH, Hoves S, Cannarile MA, Rüttinger D. CSF-1/CSF-1R targeting agents in clinical development for cancer therapy. Curr Opin Pharmacol. 2015;23:45–51. doi:10.1016/j.coph.2015.05.008.
  • Baram L, Peled E, Berman P, Yellin B, Besser E, Benami M, Louria-Hayon I, Lewitus GM, Meiri D. The heterogeneity and complexity of Cannabis extracts as antitumor agents. Oncotarget. 2019;10(41):4091–4106. doi:10.18632/oncotarget.26983.
  • Hinz B, Ramer R. Cannabinoids as anticancer drugs: current status of preclinical research. Br J Cancer. 2022;127(1):1–13. doi:10.1038/s41416-022-01727-4.
  • Di Marzo V, Piscitelli F. The endocannabinoid system and its modulation by phytocannabinoids. Neurotherapeutics. 2015;12(4):692–698. doi:10.1007/s13311-015-0374-6.
  • Lowe H, Toyang N, Steele B, Bryant J, Ngwa W. The endocannabinoid system: a potential target for the treatment of various diseases. Int J Mol Sci. 2021;22(17):22. doi:10.3390/ijms22179472.
  • Jacobson MR, Watts JJ, Boileau I, Tong J, Mizrahi R. A systematic review of phytocannabinoid exposure on the endocannabinoid system: implications for psychosis. Eur Neuropsychopharmacol. 2019;29(3):330–348. doi:10.1016/j.euroneuro.2018.12.014.
  • Straiker A, Wilson S, Corey W, Dvorakova M, Bosquez T, Tracey J, Wilkowski C, Ho K, Wager-Miller J, Mackie K, et al. An evaluation of understudied phytocannabinoids and their effects in two neuronal models. Molecules. 2021;26(17):1–17. doi:10.3390/molecules26175352.
  • Berman P, Sulimani L, Gelfand A, Amsalem K, Lewitus GM, Meiri D. Cannabinoidomics – an analytical approach to understand the effect of medical Cannabis treatment on the endocannabinoid metabolome. Talanta. 2020;219:121336. doi:10.1016/j.talanta.2020.121336.
  • Chakravarti B, Ravi J, Ganju RK. Cannabinoids as therapeutic agents in cancer: current status and future implications. Oncotarget. 2014;5(15):5852–5872. doi:10.18632/oncotarget.2233.
  • Zagzoog A, Mohamed KA, Kim HJJ, Kim ED, Frank CS, Black T, Jadhav PD, Holbrook LA, Laprairie RB. In vitro and in vivo pharmacological activity of minor cannabinoids isolated from Cannabis sativa. Sci Rep. 2020;10(1):1–13. doi:10.1038/s41598-020-77175-y.
  • Elliott DM, Singh N, Nagarkatti M, Nagarkatti PS. Cannabidiol attenuates experimental autoimmune encephalomyelitis model of multiple sclerosis through induction of myeloid- derived suppressor cells. Front Immunol. 2018;9:1–12.
  • Passarelli A, Mannavola F, Stucci LS, Tucci M, Silvestris F. Immune system and melanoma biology: a balance between immunosurveillance and immune escape. Oncotarget. 2017;8(62):106132–106142. doi:10.18632/oncotarget.22190.
  • Berman P, Futoran K, Lewitus GM, Mukha D, Benami M, Shlomi T, Meiri D. A new ESI-LC/MS approach for comprehensive metabolic profiling of phytocannabinoids in Cannabis. Sci Rep. 2018;8(1):1–15. doi:10.1038/s41598-018-32651-4.
  • Galve-Roperh I, Chiurchiù V, Díaz-Alonso J, Bari M, Guzmán M, Maccarrone M. Prog Lipid Res Cannabinoid receptor signaling in progenitor/stem cell proliferation and differentiation. Prog Lipid Res. 2013;52(4):633–650. doi:10.1016/j.plipres.2013.05.004.
  • Bronte V, Brandau S, Chen SH, Colombo MP, Frey AB, Greten TF, Mandruzzato S, Murray PJ, Ochoa A, Ostrand-Rosenberg S, et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun. 2016;7(1):1–10. doi:10.1038/ncomms12150.
  • Clementi E, Cervia D, Di Renzo I, Moscheni C, Bassi MT, Campana L, Martelli C, Catalani E, Giovarelli M, Zecchini S, et al. Nitric oxide generated by tumor-associated macrophages is responsible for cancer resistance to cisplatin and correlated with syntaxin 4 and acid sphingomyelinase inhibition. Front Immunol. 2018;9(May). doi:10.3389/fimmu.2018.01186.
  • Zhou Y, Yoshida S, Kubo Y, Yoshimura T, Kobayashi Y, Nakama T, Yamaguchi M, Ishikawa K, Oshima Y, Ishibashi T. Different distributions of M1 and M2 macrophages in a mouse model of laser-induced choroidal neovascularization. Mol Med Rep. 2017;15(6):3949–3956. doi:10.3892/mmr.2017.6491.
  • Patrinely JR, Baker LX, Davis EJ, Song H, Ye F, Johnson DB. Outcomes after progression of disease with anti – PD-1/PD-L1 therapy for patients with advanced melanoma. Cancer. 2020;126(15):3448–3455. doi:10.1002/cncr.32984.
  • Sockolosky JT, Dougan M, Ingram JR, Chi C, Ho M, Kauke MJ, Ploegh HL, Garcia KC. Durable antitumor responses to CD47 blockade require adaptive immune stimulation. Proc Natl Acad Sci USA. 2016;113(19). doi:10.1073/pnas.1604268113.
  • Fejza A, Polano M, Camicia L, Poletto E, Carobolante G, Toffoli G, Mongiat M, Andreuzzi E. The efficacy of anti-PD-L1 treatment in melanoma is associated with the expression of the ECM molecule EMILIN2. Int J Mol Sci. 2021;22(14):7511–7515. doi:10.3390/ijms22147511.
  • Kalbasi A, Ribas A. Tumor-intrinsic resistance to immune checkpoint blockade. Nat Rev Immunol. 2021;20(1):25–39. doi:10.1038/s41577-019-0218-4.
  • Postow MA, Callahan MK, Wolchok JD. Immune checkpoint blockade in cancer therapy. J Clin Oncol. 2015;33(17):1974–1982. doi:10.1200/JCO.2014.59.4358.
  • Qiu Y, Su M, Liu L, Tang Y, Pan Y, Sun J. Clinical application of cytokines in cancer immunotherapy. Drug Des Devel Ther. 2021;15:2269–2287. doi:10.2147/DDDT.S308578.
  • Yagiz K, Rittling SR. Both cell-surface and secreted CSF-1 expressed by tumor cells metastatic to bone can contribute to osteoclast activation. Exp Cell Res. 2010;315(14):2442–2452. doi:10.1016/j.yexcr.2009.05.002.
  • Ding J, Guo C, Hu P, Chen J, Liu Q, Wu X, Cao Y, Wu J. CSF1 is involved in breast cancer progression through inducing monocyte differentiation and homing. Int J Oncol. 2016;49(5):2064–2074. doi:10.3892/ijo.2016.3680.
  • Nachnani R, Raup-Konsavage WM, Vrana KE. The pharmacological case for Cannabigerol. J Pharmacol Exp Ther. 2021;376(2):204–212. doi:10.1124/jpet.120.000340.
  • Lewis MA, Russo EB, Smith KM, Village W, Republic C, Lewis M. Pharmacological foundations of Cannabis chemovars authors. Planta Med. 2018;84(4):225–233. doi:10.1055/s-0043-122240.
  • Procaccia S, Lewitus GM, Lipson Feder C, Shapira A, Berman P, Meiri D. Cannabis for medical use: versatile plant rather than a single drug. Front Pharmacol. 2022;13(April):1–14. doi:10.3389/fphar.2022.894960.
  • Kumar V, Patel S, Tcyganov E, Gabrilovich DI. The nature of myeloid-derived suppressor cells in the tumor miroenvironment. Trends Immunol. 2017;37(3):208–220. doi:10.1016/j.it.2016.01.004.
  • Youn J-I, Gabrilovich DI. The biology of myeloid-derived suppressor cells: the blessing and the curse of morphological and functional heterogeneity. Eur J Immunol. 2012;40(11):2969–2975. doi:10.1002/eji.201040895.
  • Solito S, Marigo I, Pinton L, Damuzzo V, Mandruzzato S, Bronte V. Myeloid-derived suppressor cell heterogeneity in human cancers. Ann Ny Acad Sci. 2014;1319(1):47–65. doi:10.1111/nyas.12469.
  • Kienzl M, Kargl J, Schicho R. The immune endocannabinoid system of the tumor microenvironment. Int J Mol Sci. 2020;21(23):8929. doi:10.3390/ijms21238929.
  • Qiu C, Yang L, Wang B, Cui L, Li C, Zhuo Y, Zhang L, Zhang S, Zhang Q, Wang X. The role of 2-arachidonoylglycerol in the regulation of the tumor-immune microenvironment in murine models of pancreatic cancer. Biomed Pharmacother. 2019;115(May):108952. doi:10.1016/j.biopha.2019.108952.
  • Elbaz M, Nasser MW, Ravi J, Wani NA, Ahirwar DK, Zhao H, Oghumu S, Satoskar A, Shilo K, Carson WE, et al. Modulation of the tumor microenvironment and inhibition of EGF/EGFR pathway: novel anti-tumor mechanisms of Cannabidiol in breast cancer. Mol Oncol. 2015;9(4):906–919. doi:10.1016/j.molonc.2014.12.010.
  • Davis MP. Cannabinoids for symptom management and cancer therapy: the evidence. J Natl Compr Cancer Netw. 2016;14(7):915–922. doi:10.6004/jnccn.2016.0094.
  • Emerson C Swallowable Cannabigerol tablets for sleep quality in veterans [Internet]. 2022. Available from: https://clinicaltrials.gov/ct2/show/NCT05088018
  • Aviram J, Lewitus GM, Vysotski Y, Amna MA, Ouryvaev A, Procaccia S, Cohen I, Leibovici A, Akria L, Goncharov D, et al. The effectiveness and safety of medical Cannabis for treating cancer related symptoms in oncology patients. Front Pain Res. 2022;3(May):1–14. doi:10.3389/fpain.2022.861037.
  • Xiong X, Chen S, Shen J, You H, Yang H, Yan C, Fang Z, Zhang J, Cai X, Dong X, et al. Cannabis suppresses antitumor immunity by inhibiting JAK/STAT signaling in T cells through CNR2. Signal Transduct Target Ther. 2022;7(1). doi:10.1038/s41392-022-00918-y.
  • Bar-Sela G, Cohen I, Campisi-Pinto S, Lewitus GM, Oz-Ari L, Jehassi A, Peer A, Turgeman I, Vernicova O, Berman P, et al. Cannabis consumption used by cancer patients during immunotherapy correlates with poor clinical outcome. Cancers (Basel). 2020;12(9):12. doi:10.3390/cancers12092447.
  • Taha T, Meiri D, Talhamy S, Wollner M, Peer A, Bar-Sela G. Cannabis impacts tumor response rate to nivolumab in patients with advanced malignancies. Oncologist. 2019;24(4):549–554. doi:10.1634/theoncologist.2018-0383.
  • Hegde VL, Nagarkatti M, Nagarkatti PS. Cannabinoid receptor activation leads to massive mobilization of myeloid-derived suppressor cells with potent immunosuppressive properties. Eur J Immunol. 2010;40(12):3358–3371. doi:10.1002/eji.201040667.
  • Wang X, Zhang J, Hu B, Qian F. High expression of CSF-1R predicts poor prognosis and CSF-1R high tumor-associated macrophages inhibit anti-tumor immunity in colon adenocarcinoma. Front Oncol. 2022;12(April):1–13. doi:10.3389/fonc.2022.850767.
  • Liu H, Zhang H, Shen Z, Lin C, Wang X, Qin J, Qin X, Xu J, Sun Y. Increased expression of CSF-1 Associates with poor prognosis of patients with gastric cancer undergoing gastrectomy. Medicine (Baltimore). 2016;95(9):1–7. doi:10.1097/MD.0000000000002675.