WNT2 blockade augments antitumor immunity by attenuating myeloid-derived suppressor cells in colorectal cancer

Cheng Cui, Tian-Tian Zhang, Qian Lin, Tu-Xiong Huang, En-Yu Rao, Ji-Hui Du, Li Fu

MEDCOMM - Oncology ›› 2024, Vol. 3 ›› Issue (4) : e70004.

PDF
MEDCOMM - Oncology ›› 2024, Vol. 3 ›› Issue (4) : e70004. DOI: 10.1002/mog2.70004
ORIGINAL ARTICLE

WNT2 blockade augments antitumor immunity by attenuating myeloid-derived suppressor cells in colorectal cancer

Author information +
History +

Abstract

Colorectal cancer (CRC) ranks as one of the most common malignancies worldwide. Myeloid-derived suppressor cells (MDSCs) represent an immunosuppressive heterogeneous population of immature monocytes and granulocytes constituting a major obstacle for CRC therapy. Previous studies demonstrated that WNT2 is enriched in tumor microenvironment (TME), promoting CRC progression. However, the role of WNT2 in regulating MDSCs to facilitate CRC progression remains largely unexplored. Our analysis of The Cancer Genome Atlas (TCGA) database and blood samples from 50 primary and recurrent CRC patients revealed a positive correlation between WNT2 expression and MDSCs abundance. Treatment with recombinant WNT2 protein significantly enhanced the accumulation and immunosuppressive function of MDSCs in vitro. Conversely, anti-WNT2 monoclonal antibody remarkably reduced the percentage and functional activity of MDSCs in CRC tumor-bearing mice. Mechanistic analyses further demonstrated that WNT2 mediates MDSCs activities through the p38 MAPK/Akt pathway. Collectively, our findings not only highlight the pivotal role of WNT2 in CRC progression by enhancing MDSCs activities within the TME, but also provide evidence that WNT2 levels and MDSCs abundance in peripheral blood could serve as predictive biomarkers for early diagnosis and prognosis of CRC patients.

Keywords

antitumor immunity / colorectal cancer / myeloid-derived suppressor cells / WNT2

Cite this article

Download citation ▾
Cheng Cui, Tian-Tian Zhang, Qian Lin, Tu-Xiong Huang, En-Yu Rao, Ji-Hui Du, Li Fu. WNT2 blockade augments antitumor immunity by attenuating myeloid-derived suppressor cells in colorectal cancer. MEDCOMM - Oncology, 2024, 3(4): e70004 https://doi.org/10.1002/mog2.70004

References

[1]
SungH, FerlayJ, SiegelRL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209-249.
CrossRef Google scholar
[2]
XieY-H, ChenY-X, FangJ-Y. Comprehensive review of targeted therapy for colorectal cancer. Signal Transduct Target Ther. 2020;5(1):22.
CrossRef Google scholar
[3]
GabrilovichDI. Myeloid-derived suppressor cells. Cancer Immunol Res. 2017;5(1):3-8.
CrossRef Google scholar
[4]
NoyR, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity. 2014;41(1):49-61.
CrossRef Google scholar
[5]
CuiC, LanP, FuL. The role of myeloid-derived suppressor cells in gastrointestinal cancer. Cancer Commun. 2021;41(6):442-471.
CrossRef Google scholar
[6]
GabrilovichDI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9(3):162-174.
CrossRef Google scholar
[7]
BronteV, Apolloni E, CabrelleA, et al. Identification of a CD11b+/Gr-1+/CD31+myeloid progenitor capable of activating or suppressing CD8+T cells. Blood. 2000;96(12):3838-3846.
CrossRef Google scholar
[8]
KumarV, PatelS, TcyganovE, Gabrilovich DI. The nature of myeloid-derived suppressor cells in the tumor microenvironment. Trends Immunol. 2016;37(3):208-220.
CrossRef Google scholar
[9]
StiffA, TrikhaP, Mundy-BosseB, et al. Nitric oxide production by myeloid-derived suppressor cells plays a role in impairing Fc receptor-mediated natural killer cell function. Clin Cancer Res. 2018;24(8):1891-1904.
CrossRef Google scholar
[10]
CorzoCA, CotterMJ, ChengP, et al. Mechanism regulating reactive oxygen species in tumor-induced myeloid-derived suppressor cells. J Immunol. 2009;182(9):5693-5701.
CrossRef Google scholar
[11]
RodriguezPC, Ernstoff MS, HernandezC, et al. Arginase I-producing myeloid-derived suppressor cells in renal cell carcinoma are a subpopulation of activated granulocytes. Cancer Res. 2009;69(4):1553-1560.
CrossRef Google scholar
[12]
BaniyashM. Myeloid-derived suppressor cells as intruders and targets: clinical implications in cancer therapy. Cancer Immunol Immunother. 2016;65(7):857-867.
CrossRef Google scholar
[13]
KarakashevaTA, Waldron TJ, EruslanovE, et al. CD38-expressing myeloid-derived suppressor cells promote tumor growth in a murine model of esophageal cancer. Cancer Res. 2015;75(19):4074-4085.
CrossRef Google scholar
[14]
KarakashevaTA, Dominguez GA, HashimotoA, et al. CD38+M-MDSC expansion characterizes a subset of advanced colorectal cancer patients. JCI Insight. 2018;3(6):1-8.
CrossRef Google scholar
[15]
KalluriR. The biology and function of fibroblasts in cancer. Nat Rev Cancer. 2016;16(9):582-598.
CrossRef Google scholar
[16]
LoefflerM. Targeting tumor-associated fibroblasts improves cancer chemotherapy by increasing intratumoral drug uptake. J Clin Invest. 2006;116(7):1955-1962.
CrossRef Google scholar
[17]
ValkenburgKC, De Groot AE, PientaKJ. Targeting the tumour stroma to improve cancer therapy. Nat Rev Clin Oncol. 2018;15(6):366-381.
CrossRef Google scholar
[18]
Gok YavuzB, Gunaydin G, GedikME, et al. Cancer associated fibroblasts sculpt tumour microenvironment by recruiting monocytes and inducing immunosuppressive PD-1+TAMs. Sci Rep. 2019;9(1):3172.
CrossRef Google scholar
[19]
CostaA, Kieffer Y, Scholer-DahirelA, et al. Fibroblast heterogeneity and immunosuppressive environment in human breast cancer. Cancer Cell. 2018;33(3):463-479.e10.
CrossRef Google scholar
[20]
FuL, ZhangC, ZhangL-Y, et al. Wnt2 secreted by tumour fibroblasts promotes tumour progression in oesophageal cancer by activation of the Wnt/β-catenin signalling pathway. Gut. 2011;60(12):1635-1643.
CrossRef Google scholar
[21]
KramerN, Schmöllerl J, UngerC, et al. Autocrine WNT2 signaling in fibroblasts promotes colorectal cancer progression. Oncogene. 2017;36(39):5460-5472.
CrossRef Google scholar
[22]
LoganCY, NusseR. The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol. 2004;20:781-810.
CrossRef Google scholar
[23]
RodriguezPC, Quiceno DG, ZabaletaJ, et al. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res. 2004;64(16):5839-5849.
CrossRef Google scholar
[24]
GabrilovichDI. The dawn of myeloid-derived suppressor cells: identification of arginase I as the mechanism of immune suppression. Cancer Res. 2021;81(15):3953-3955.
CrossRef Google scholar
[25]
NagarajS, YounJ-I, WeberH, et al. Anti-inflammatory triterpenoid blocks immune suppressive function of MDSCs and improves immune response in cancer. Clin Cancer Res. 2010;16(6):1812-1823.
CrossRef Google scholar
[26]
XiangH, RamilCP, HaiJ, et al. Cancer-associated fibroblasts promote immunosuppression by inducing ROS-generating monocytic MDSCs in lung squamous cell carcinoma. Cancer Immunol Res. 2020;8(4):436-450.
CrossRef Google scholar
[27]
ZhangS, MaX, ZhuC, LiuL, WangG, Yuan X. The role of myeloid-derived suppressor cells in patients with solid tumors: a meta-analysis. PLoS One. 2016;11(10):e0164514.
CrossRef Google scholar
[28]
LimagneE, Euvrard R, ThibaudinM, et al. Accumulation of MDSC and Th17 cells in patients with metastatic colorectal cancer predicts the efficacy of a FOLFOX-bevacizumab drug treatment regimen. Cancer Res. 2016;76(18):5241-5252.
CrossRef Google scholar
[29]
ViderB-Z, ZimberA, ChastreE, et al. Evidence for the involvement of the Wnt 2 gene in human colorectal cancer. Oncogene. 1996;12(1):153-158.
[30]
PeinadoH, Lavotshkin S, LydenD. The secreted factors responsible for pre-metastatic niche formation: old sayings and new thoughts. Sem Cancer Biol. 2011;21:139-146.
CrossRef Google scholar
[31]
GilesAJ, ReidCM, EvansJD, et al. Activation of hematopoietic stem/progenitor cells promotes immunosuppression within the pre-metastatic niche. Cancer Res. 2016;76(6):1335-1347.
CrossRef Google scholar
[32]
HuangB, PanP-Y, LiQ, et al. Gr-1+CD115+immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host. Cancer Res. 2006;66(2):1123-1131.
CrossRef Google scholar
[33]
RaberPL, Thevenot P, SierraR, et al. Subpopulations of myeloid-derived suppressor cells impair T cell responses through independent nitric oxide-related pathways. Int J Cancer. 2014;134(12):2853-2864.
CrossRef Google scholar
[34]
ZosoA, MazzaEMC, BicciatoS, et al. Human fibrocytic myeloid-derived suppressor cells express IDO and promote tolerance via Treg-cell expansion. Eur J Immunol. 2014;44(11):3307-3319.
CrossRef Google scholar
[35]
DouglassSM, FaneME, SansevieroE, et al. Myeloid-derived suppressor cells are a major source of Wnt5A in the melanoma microenvironment and depend on Wnt5A for full suppressive activity. Cancer Res. 2021;81(3):658-670.
CrossRef Google scholar
[36]
JungY-S, ParkJ-I. Wnt signaling in cancer: therapeutic targeting of Wnt signaling beyond β-catenin and the destruction complex. Exp Mol Med. 2020;52(2):183-191.
CrossRef Google scholar
[37]
DeVitoNC, Sturdivant M, ThievanthiranB, et al. Pharmacological Wnt ligand inhibition overcomes key tumor-mediated resistance pathways to anti-PD-1 immunotherapy. Cell Rep. 2021;35(5):109071.
CrossRef Google scholar
[38]
HaoY, ChungCK, GuZ, et al. Combinatorial therapeutic approaches of photodynamic therapy and immune checkpoint blockade for colon cancer treatment. Mol Biomed. 2022;3(1):26.
CrossRef Google scholar
[39]
LiQ, LiZ, LuoT, ShiH. Targeting the PI3K/AKT/mTOR and RAF/MEK/ERK pathways for cancer therapy. Mol Biomed. 2022;3(1):47.
CrossRef Google scholar
[40]
ChiuW-C, FangP-T, LeeY-C, et al. DNA repair protein Rad51 induces tumor growth and metastasis in esophageal squamous cell carcinoma via a p38/Akt-dependent pathway. Ann Surg Oncol. 2020;27:2090-2101.
CrossRef Google scholar
[41]
ZhangX, ZhangW, ChenF, Lu Z. Combined effect of chrysin and apigenin on inhibiting the development and progression of colorectal cancer by suppressing the activity of P38-MAPK/AKT pathway. IUBMB Life. 2021;73(5):774-783.
CrossRef Google scholar
[42]
SeimonTA, WangY, HanS, et al. Macrophage deficiency of p38αMAPK promotes apoptosis and plaque necrosis in advanced atherosclerotic lesions in mice. J Clin Invest. 2009;119(4):886-898.
[43]
CaverzasioJ, ManenD. Essential role of Wnt3a-mediated activation of mitogen-activated protein kinase p38 for the stimulation of alkaline phosphatase activity and matrix mineralization in C3H10T1/2 mesenchymal cells. Endocrinology. 2007;148(11):5323-5330.
CrossRef Google scholar
[44]
CuadradoA, Nebreda AR. Mechanisms and functions of p38 MAPK signalling. Biochem J. 2010;429(3):403-417.
CrossRef Google scholar
[45]
WahyudiLD, JeongJ, YangH, Kim JH. Amentoflavone-induced oxidative stress activates NF-E2-related factor 2 via the p38 MAP kinase-AKT pathway in human keratinocytes. Int J Biochem Cell Biol. 2018;99:100-108.
CrossRef Google scholar
[46]
CaiZ, TianY, LiJ, et al. Peimine ameliorates pulmonary fibrosis via the inhibition of M2-type macrophage polarization through the suppression of P38/Akt/STAT6 signals. Biosci Rep. 2022;42(10):BSR20220986.
CrossRef Google scholar
[47]
CuiC, LuH, HuiQ, et al. A preliminary investigation of the toxic effects of benzylpenicilloic acid. Food Chem Toxicol. 2018;111:567-577.
CrossRef Google scholar
[48]
CuiC, BaX, HolmesMAJJ-AR. Prevalence and characterization of mecC MRSA in bovine bulk tank milk in Great Britain. JAC Antimicrob Resist. 2021;3(1):dlaa125.
CrossRef Google scholar

RIGHTS & PERMISSIONS

2024 2024 The Author(s). MedComm – Oncology published by John Wiley & Sons Australia, Ltd on behalf of Sichuan International Medical Exchange & Promotion Association (SCIMEA).
PDF

Accesses

Citations

Detail

Sections
Recommended

/