Myeloid-derived suppressor cell

From Wikipedia, the free encyclopedia

Myeloid-derived suppressor cells (MDSC) are a heterogeneous group of immune cells from the myeloid lineage (a family of cells that originate from bone marrow stem cells).

MDSCs strongly expand in pathological situations such as chronic infections and cancer, as a result of an altered haematopoiesis.[1] MDSCs are discriminated from other myeloid cell types in which they possess strong immunosuppressive activities rather than immunostimulatory properties. Similar to other myeloid cells, MDSCs interact with other immune cell types including T cells, dendritic cells, macrophages and natural killer cells to regulate their functions. Although their mechanisms of action are not clear yet, clinical and experimental evidence has shown that cancer tissues with high infiltration of MDSCs are associated with poor patient prognosis and resistance to therapies.[2][3][4][5] MDSCs can also be detected in the blood. In breast cancer patients, MDSC levels in the blood are about 10-fold higher than normal.[6] The size of the myeloid suppressor compartment is considered to be an important factor in the clinical success or failure of cancer immunotherapy, highlighting the importance of this cell type for human pathophysiology.[7]

It is yet unclear whether MDSCs represent a group of immature myeloid cell types that have stopped their differentiation, or they represent a distinctive myeloid lineage.

Phenotype[]

In mice[]

In mouse models, MDSCs are found as myeloid cells expressing high levels of CD11b (a classical myeloid lineage marker) and GR1 (). The GR1 marker is made up of two cell membrane molecules, Ly6C and , and according to their relative expression levels murine MDSCs are further classified into two subtypes, monocytic and granulocytic. Monocytic MDSCs express high levels of the Ly6C surface marker with low or no expression of the Ly6G marker, while granulocytic MDSCs express Ly6C and high levels of Ly6G. These phenotypes are reminiscent of those from inflammatory monocytes (and hence the term "monocytic MDSC") and granulocytes (for "granulocytic MDSCs), respectively.

In humans[]

Human MDSCs are less characterized, and they are generally defined as myeloid cells expressing CD33, CD14 and low levels of HLA DR. The absence of the human equivalent to the murine GR1 marker makes it difficult to compare murine and human MDSCs. Although they functionally resemble murine MDSCs, their characterization and classification into different subsets remains to be resolved as there is (As of 2012) no international consensus on how human subsets of MDSC should be defined.[8] However, A combination of CD33 and CD15 has been found to identify two major subsets of the MSDC in the peripheral blood of bladder cancer patients into granulocyte-type CD15(high) CD33(low) cells and monocyte-type CD15(low) CD33(high) cells.[9][10]

Production and activity[]

Generally speaking, regardless of whether they are from mice or human, MDSC suppressor function lies in their ability to inhibit T cell proliferation and activation. In healthy individuals, immature myeloid cells formed in the bone marrow differentiate to dendritic cells, macrophages and neutrophils. However, under chronic inflammatory conditions (viral and bacterial infections) or cancer, myeloid differentiation is skewed towards the expansion of MDSCs. These MDSCs infiltrate inflammation sites and tumors, where they stop immune responses by inhibiting T cells and NK cells, for example. MDSCs also accelerate angiogenesis, tumor progression and metastasis through the expression of cytokines and factors such as TGF-beta. Therefore, they have become a key therapeutic target.

MDSC differentiation[]

In humans[]

MDSCs derive from bone marrow precursors usually as the result of a perturbed myeloipoiesis caused by different pathologies. In cancer patients, growing tumours secrete a variety of cytokines and other molecules which are key signals involved in the generation of MDSC. Tumor cell lines overexpressing colony stimulating factors (e.g. G-CSF and GM-CSF) have long been used in in vivo models of MDSC generation. GM-CSF, G-CSF and IL-6 allow the in vitro generation of MDSC that retain their suppressive function in vivo. In addition to CSF, other cytokines such as IL-6, IL-10, VEGF, PGE2 and IL-1 have been implicated in the development and regulation of MDSC.[2][11] The myeloid-differentiation cytokine GM-CSF is a key factor in MDSC production from bone marrow,[12][unreliable medical source?] and it has been shown that the c/EBPβ transcription factor plays a key role in the generation of in vitro bone marrow-derived and in vivo tumor-induced MDSC. Moreover, STAT3 promotes MDSC differentiation and expansion and IRF8 has been suggested to counterbalance MDSC-inducing signals.

In mice[]

Murine MDSCs show two distinct phenotypes which discriminate them into either monocytic MDSCs or granulocytic MDSCs. The relationship between these two subtypes remains controversial, as they closely resemble monocytes and neutrophils respectively. While monocyte and neutrophil differentiation pathways within the bone marrow are antagonistic and dependent on the relative expression of IRF8 and c/EBP transcription factors (and hence there is not a direct precursor-progeny link between these two myeloid cell types), this seems not to be the case for MDSCs. Monocytic MDSCs seem to be precursors of granulocytic subsets demonstrated both in vitro and in vivo.[12][13] This differentiation process is accelerated upon tumour infiltration and possibly driven by the hypoxic tumor microenvironment.

Activity/function[]

MDSCs are immune suppressive and play a role in tumor maintenance and progression. MDSCs also obstruct therapies that seek to treat cancer through both immunotherapy and other non-immune means.[14] MDSC activity was originally described as suppressors of T cells, in particular of CD8+ T-cell responses. The spectrum of action of MDSC activity also encompasses NK cells, dendritic cells and macrophages. Suppressor activity of MDSC is determined by their ability to inhibit the effector function of lymphocytes. Inhibition can be caused by different mechanisms. It is primarily attributed to the effects of the metabolism of L-arginine. Another important factor influencing the activity of MDSC is oppressive ROS.[2][15]

MDSCs can also play a positive regulatory role. It is stated that MMR vaccine stimulates MDSC populations in people taking the vaccine, inhibiting septic inflammation and mortality that is broadly applicable not only to measles, mumps, and rubella, but extends to covid-19 induced cytokine inflammation. This vaccination inducement appears to be neither permanent nor chronic. Despite MDSC's being immunosuppressive in certain instances, the MMR vaccine itself is immunostimulatory.

MDSC inhibitors[]

In addition to host-derived factors, pharmacologic agents also have profound impact on MDSC. Chemotherapeutic agents belonging to different classes have been reported to inhibit MDSC. Although this effect may well be secondary to inhibition of hematopoietic progenitors, there may be grounds for search of selectivity based on long-known differential effects of these agents on immunocompetent cells and macrophages.[2] In 2015, MDSCs were compared to immunogenic myeloid cells highlighting a group of core signaling pathways that control pro-carcinogenic MDSC functions.[16][unreliable medical source?] Many of these pathways are known targets of chemotherapy drugs with strong anti-cancer properties.

As of May 2018 there are no FDA approved drugs developed to target MDSCs but experimental has entered early clinical trials.[17][18]

There is promising evidence for inhibiting Galectin-3 as a therapeutic target to reduce MDSCs.[19][20] In a Phase 1b clinical trial of GR-MD-02 developed by Galectin Therapeutics, investigators observed a significant decrease in the frequency of suppressive myeloid-derived suppressor cells following treatment in responding melanoma patients.[21]

History[]

The term myeloid-derived suppressor cell originated in a 2007 journal article published in Cancer Research by Gabrilovich et al. Publications in 2008 established that there are two subpopulations of MDSC: mononuclear MDSC (M-MDSC) and polymorphonuclear or granulocytic MDSC (PMN-MDSC). M-MDSC are similar to monocytes found in blood, while PMN-MDSC are physically akin to neutrophils.[14]

References[]

  1. ^ Li T, Li X, Chen YH (May 2020). "c-Rel is a myeloid checkpoint for cancer immunotherapy". Nature Cancer. 1 (5): 507–517. doi:10.1038/s43018-020-0061-3. PMC 7808269. PMID 33458695.
  2. ^ Jump up to: a b c d Mantovani A (December 2010). "The growing diversity and spectrum of action of myeloid-derived suppressor cells". European Journal of Immunology. 40 (12): 3317–20. doi:10.1002/eji.201041170. PMID 21110315.
  3. ^ Allavena P, Mantovani A (February 2012). "Immunology in the clinic review series; focus on cancer: tumour-associated macrophages: undisputed stars of the inflammatory tumour microenvironment". Clinical and Experimental Immunology. 167 (2): 195–205. doi:10.1111/j.1365-2249.2011.04515.x. PMC 3278685. PMID 22235995.
  4. ^ Galdiero MR, Bonavita E, Barajon I, Garlanda C, Mantovani A, Jaillon S (November 2013). "Tumor associated macrophages and neutrophils in cancer". Immunobiology. 218 (11): 1402–10. doi:10.1016/j.imbio.2013.06.003. PMID 23891329.
  5. ^ Gabrilovich DI, Ostrand-Rosenberg S, Bronte V (March 2012). "Coordinated regulation of myeloid cells by tumours". Nature Reviews. Immunology. 12 (4): 253–68. doi:10.1038/nri3175. PMC 3587148. PMID 22437938.
  6. ^ Safarzadeh E, Hashemzadeh S, Duijf PH, Mansoori B, Khaze V, Mohammadi A, et al. (April 2019). "Circulating myeloid-derived suppressor cells: An independent prognostic factor in patients with breast cancer". Journal of Cellular Physiology. 234 (4): 3515–3525. doi:10.1002/jcp.26896. PMID 30362521.
  7. ^ Kodach LL, Peppelenbosch MP (August 2021). "Targeting the Myeloid-Derived Suppressor Cell Compartment for Inducing Responsiveness to Immune Checkpoint Blockade Is Best Limited to Specific Subtypes of Gastric Cancers". Gastroenterology. 161 (2): 727. doi:10.1053/j.gastro.2021.03.047. PMID 33798523.
  8. ^ Poschke I, Kiessling R (September 2012). "On the armament and appearances of human myeloid-derived suppressor cells". Clinical Immunology. 144 (3): 250–68. doi:10.1016/j.clim.2012.06.003. PMID 22858650.
  9. ^ Eruslanov E, Neuberger M, Daurkin I, Perrin GQ, Algood C, Dahm P, et al. (March 2012). "Circulating and tumor-infiltrating myeloid cell subsets in patients with bladder cancer". International Journal of Cancer. 130 (5): 1109–19. doi:10.1002/ijc.26123. PMID 21480223.
  10. ^ Crispen PL, Kusmartsev S (December 2019). "Mechanisms of immune evasion in bladder cancer". Cancer Immunology, Immunotherapy. 69 (1): 3–14. doi:10.1007/s00262-019-02443-4. PMC 6949323. PMID 31811337.
  11. ^ Gros A, Turcotte S, Wunderlich JR, Ahmadzadeh M, Dudley ME, Rosenberg SA (October 2012). "Myeloid cells obtained from the blood but not from the tumor can suppress T-cell proliferation in patients with melanoma". Clinical Cancer Research. 18 (19): 5212–23. doi:10.1158/1078-0432.CCR-12-1108. PMC 6374773. PMID 22837179.
  12. ^ Jump up to: a b Liechtenstein T, Perez-Janices N, Gato M, Caliendo F, Kochan G, Blanco-Luquin I, et al. (September 2014). "A highly efficient tumor-infiltrating MDSC differentiation system for discovery of anti-neoplastic targets, which circumvents the need for tumor establishment in mice". Oncotarget. 5 (17): 7843–57. doi:10.18632/oncotarget.2279. PMC 4202165. PMID 25151659.
  13. ^ Youn JI, Kumar V, Collazo M, Nefedova Y, Condamine T, Cheng P, et al. (March 2013). "Epigenetic silencing of retinoblastoma gene regulates pathologic differentiation of myeloid cells in cancer". Nature Immunology. 14 (3): 211–20. doi:10.1038/ni.2526. PMC 3578019. PMID 23354483.
  14. ^ Jump up to: a b Ostrand-Rosenberg, Suzanne (2021-03-04). "Myeloid-Derived Suppressor Cells: Facilitators of Cancer and Obesity-Induced Cancer". Annual Review of Cancer Biology. 5 (1): 17–38. doi:10.1146/annurev-cancerbio-042120-105240. ISSN 2472-3428.
  15. ^ Kusmartsev S, Nefedova Y, Yoder D, Gabrilovich DI (January 2004). "Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species". Journal of Immunology. 172 (2): 989–99. doi:10.4049/jimmunol.172.2.989. PMID 14707072.
  16. ^ Gato-Cañas M, Martinez de Morentin X, Blanco-Luquin I, Fernandez-Irigoyen J, Zudaire I, Liechtenstein T, et al. (September 2015). "A core of kinase-regulated interactomes defines the neoplastic MDSC lineage". Oncotarget. 6 (29): 27160–75. doi:10.18632/oncotarget.4746. PMC 4694980. PMID 26320174.
  17. ^ INmune Bio Initiates Phase I Clinical Trial Of INB03 May 2018
  18. ^ Toor SM, Elkord E (October 2018). "Therapeutic prospects of targeting myeloid-derived suppressor cells and immune checkpoints in cancer". Immunology and Cell Biology. 96 (9): 888–897. doi:10.1111/imcb.12054. PMID 29635843.
  19. ^ Wang T, Chu Z, Lin H, Jiang J, Zhou X, Liang X (June 2014). "Galectin-3 contributes to cisplatin-induced myeloid derived suppressor cells (MDSCs) recruitment in Lewis lung cancer-bearing mice". Molecular Biology Reports. 41 (6): 4069–76. doi:10.1007/s11033-014-3276-5. PMID 24615503.
  20. ^ Blidner AG, Méndez-Huergo SP, Cagnoni AJ, Rabinovich GA (November 2015). "Re-wiring regulatory cell networks in immunity by galectin-glycan interactions". FEBS Letters. 589 (22): 3407–18. doi:10.1016/j.febslet.2015.08.037. PMID 26352298.
  21. ^ Galectin Therapeutics Inc. (2018-09-20). "Positive Preliminary Results from Phase 1b Clinical Trial of GR-MD-02 and KEYTRUDA® in Advanced Melanoma and Expansion of the Trial". GlobeNewswire News Room. Retrieved 2019-03-14.
Retrieved from ""