PDF
Abstract
Background: Mounting evidence shows that myeloid-derived suppressor cells (MDSCs) reprogramming can significantly enhance the outcomes of immunotherapy. However, the therapeutic potential of targeting MDSCs alone is limited by persistent immunosuppressive cytokines and cellular crosstalk. In our previous study, we found that novel cryo-thermal therapy (CTT) can drive MDSCs maturation and induce CD4+ T helper type (Th)1-dominant differentiation, improving long-term survival in spontaneous high metastatic mouse models. Considering the established roles of Interleukin (IL)-6 and IL-17A in non-small cell lung cancer (NSCLC) progression and immune evasion, we developed a combination strategy integrating cytokine neutralization with CTT (combination therapy) in LLC1 tumor-bearing mice. Although the combination therapy successfully promoted MDSCs maturation and Th1 differentiation, the underlying mechanistic basis remained unclear.
Methods: The combination therapy was implemented in LLC1 tumor-bearing mice. We then observed its impacts on MDSCs maturation and Th1 differentiation and explored the related mechanisms by examining various aspects including the expression of CD40, the reactive oxygen species (ROS)-nuclear factor-kappa B (NF-κB) pathway, and the induction of tumor necrosis factor-α (TNF-α).
Results: It was observed that the combination therapy increased the expression of CD40 on MDSCs through the ROS-NF-κB pathway-dependent TNF-α induction. This TNF-α-mediated CD40 upregulation facilitated Th1 polarization via CD40L engagement on CD4+ T cells. Our results provided the first mechanistic evidence that autocrine TNF-α production by reprogrammed MDSCs governs CD40 expression following combination therapy.
Conclusion: Our research elucidated the methods and mechanisms of MDSCs reprogramming and offered a promising therapeutic strategy for patients with NSCLC and other types of cancer.
Keywords
CD4-positive T-lymphocytes
/
CD40
/
interleukin-17A
/
interleukin-6
/
myeloid-derived suppressor cells
/
tumour necrosis factor-alpha (TNF-α)
Cite this article
Download citation ▾
Yuankai Hao, Shicheng Wang, Junjun Wang, Zelu Zhang, Yichen Yao, Ke Wang, Ping Liu, Lisa X. Xu.
Reprogrammed MDSCs promote Th1-dominant antitumour response via CD40 induced by autocrine TNF-α after combining cryo-thermal therapy with IL6 and IL17A neutralization.
Clinical and Translational Medicine, 2025, 15(10): e70493 DOI:10.1002/ctm2.70493
| [1] |
Chow A, Perica K, Klebanoff CA, et al. Clinical implications of T cell exhaustion for cancer immunotherapy. Nat Rev Clin Oncol. 2022. 19(12): 775–790.
|
| [2] |
Wang B, Iriguchi S, Waseda M, et al. Generation of hypoimmunogenic T cells from genetically engineered allogeneic human induced pluripotent stem cells. Nat Biomed Eng. 2021. 5(5): 429–440.
|
| [3] |
Kumar V, Patel S, Tcyganov E, et al. The Nature of Myeloid-Derived Suppressor Cells in the Tumor Microenvironment. Trends Immunol. 2016. 37(3): 208–220.
|
| [4] |
Liu NN, Yi CX, Wei LQ, et al. The intratumor mycobiome promotes lung cancer progression via myeloid-derived suppressor cells. Cancer Cell. 2023.; 41(11): 1927–1944.e9.
|
| [5] |
Deepak KGK, Vempati R, Nagaraju GP, et al. Tumor microenvironment: Challenges and opportunities in targeting metastasis of triple negative breast cancer. Pharmacol Res. 2020. 153: 104683.
|
| [6] |
Xu S, Wang Q, Ma W. Cytokines and soluble mediators as architects of tumor microenvironment reprogramming in cancer therapy. Cytokine & Growth Factor Reviews. 2024. 76: 12–21.
|
| [7] |
Propper DJ, Balkwill FR. Harnessing cytokines and chemokines for cancer therapy. Nat Rev Clin Oncol. 2022. 19(4): 237–253.
|
| [8] |
Lee JH, Massagué J. TGF-β in developmental and fibrogenic EMTs. Semin Cancer Biol. 2022. 86(2): 136–145.
|
| [9] |
Tao L, Qiu J, Slavin S, et al. Recruited T cells promote the bladder cancer metastasis via up-regulation of the estrogen receptor β/IL-1/c-MET signals. Cancer Lett. 2018. 28(430): 215–223.
|
| [10] |
Li S, Cong X, Gao H, et al. Tumor-associated neutrophils induce EMT by IL-17a to promote migration and invasion in gastric cancer cells. J Exp Clin Cancer Res. 2019. 38(1): 6.
|
| [11] |
Weber R, Groth C, Lasser S, et al. IL-6 as a major regulator of MDSC activity and possible target for cancer immunotherapy. Cell Immunol. 2021. 359: 104254.
|
| [12] |
LaMarche NM, Hegde S, Park MD, et al. An IL-4 signaling axis in bone marrow drives pro-tumorigenic myelopoiesis. Nature. 2024. 625(7993): 166–174.
|
| [13] |
Qiao J, Liu Z, Dong C. Targeting Tumors with IL-10 Prevents Dendritic Cell-Mediated CD8+ T Cell Apoptosis. Cancer Cell. 2019. 35(6): 901–915.
|
| [14] |
Shen Y, Lu C, Song Z, et al. Ursodeoxycholic acid reduces anti-tumor immunosuppression by inducing CHIP-mediated TGF-β degradation. Nat Commun. 2022. 13(1): 3419.
|
| [15] |
Xu S, Wang Q, Ma W. Cytokines and soluble mediators as architects of tumor microenvironment reprogramming in cancer therapy. Cytokine Growth Factor Rev. 2024. 76: 12–21.
|
| [16] |
Propper DJ, Balkwill FR. Harnessing cytokines and chemokines for cancer therapy. Nat Rev Clin Oncol. 2022. 19(4): 237–253.
|
| [17] |
Xie Y, Liu P, Xu LX. A novel thermal treatment modality for controlling breast tumor growth and progression. Annu Int Conf IEEE Eng Med Biol Soc. 2012. 5703–6.
|
| [18] |
Zhu J, Lou Y, Liu P, et al. Tumor-related HSP70 released after cryo-thermal therapy targeted innate immune initiation in the anti-tumor immune response. Int J Hyperthermia. 2020. 37(1): 843–853.
|
| [19] |
Peng P, Lou Y, Wang J, et al. Th1-Dominant CD4+ T Cells Orchestrate Endogenous Systematic Anti-tumor Immune Memory After Cryo-Thermal Therapy. Front Immunol. 2022. 13: 944115.
|
| [20] |
Peng P, Hu H, Liu P, et al. Neoantigen-specific CD4+ T-cell response is critical for the therapeutic efficacy of cryo-thermal therapy. J Immunother Cancer. 2020. 8(2): e000421.
|
| [21] |
He K, Liu P, Xu LX. The cryo-thermal therapy eradicated melanoma in mice by eliciting CD4+ T-cell-mediated anti-tumor memory immune response. Cell Death Dis. 2017. 8(3): e2703.
|
| [22] |
Zheng J, Zou J, Lou Y. et al. Combining multi-mode thermal therapy with IL-6 and IL-17A neutralization amplifies antitumor immunity to facilitate long-term survival in LLC1-bearing mice. Med-X. 2024. 2: 3.
|
| [23] |
Hailemichael Y, Johnson DH, Abdel-Wahab N, et al. Interleukin-6 blockade abrogates immunotherapy toxicity and promotes tumor immunity. Cancer Cell. 2022. 40(5): 509–523.
|
| [24] |
Liu C, Liu R, Wang B, et al. Blocking IL-17A enhances tumor response to anti-PD-1 immunotherapy in microsatellite stable colorectal cancer. J Immunother Cancer. 2021. 9(1): e001895.
|
| [25] |
Ugel S, De Sanctis F, Mandruzzato S, et al. Tumor-induced myeloid deviation: when myeloid-derived suppressor cells meet tumor-associated macrophages. J Clin Invest. 2015. 125(9): 3365–76.
|
| [26] |
Ferris ST, Durai V, Wu R, et al. cDC1 prime and are licensed by CD4 T cells to induce anti-tumor immunity. Nature. 2020. 584(7822): 624–629.
|
| [27] |
Shannar A, Sarwar MS, Dave PD, et al. Cyproheptadine inhibits in vitro and in vivo lung metastasis and drives metabolic rewiring. Mol Biol Rep. 2024. 51(1): 1139.
|
| [28] |
Liner AG, van Gogh M, Roblek M, et al. Non-redundant roles of the CCR1 and CCR2 chemokine axes in monocyte recruitment during lung metastasis. Neoplasia. 2025. 59: 101089.
|
| [29] |
Chen J, Sun HW, Yang YY, et al. Reprogramming immunosuppressive myeloid cells by activated T cells promotes the response to anti-PD-1 therapy in colorectal cancer. Signal Transduct Target Ther. 2021. 6(1): 4.
|
| [30] |
Foreman TW, Nelson CE, Sallin MA, et al. CD30 co-stimulation drives differentiation of protective T cells during Mycobacterium tuberculosis infection. J Exp Med. 2023. 220(8): e20222090.
|
| [31] |
Savai R, Schermuly RT, Pullamsetti SS, et al. A combination hybrid-based vaccination/adoptive cellular therapy to prevent tumor growth by involvement of T cells. Cancer Res. 2007. 67(11): 5443–53.
|
| [32] |
Spinetti T, Spagnuolo L, Mottas I, et al. TLR7-based cancer immunotherapy decreases intratumoral myeloid-derived suppressor cells and blocks their immunosuppressive function. Oncoimmunology. 2016.: 5(11).
|
| [33] |
Dai S, Zeng H, Liu Z, et al. Intratumoral CXCL13+CD8+T cell infiltration determines poor clinical outcomes and immunoevasive contexture in patients with clear cell renal cell carcinoma. J Immunother Cancer. 2021. 9(2): e001823.
|
| [34] |
Sia JK, Bizzell E, Madan-Lala R, et al. Engaging the CD40-CD40L pathway augments T-helper cell responses and improves control of Mycobacterium tuberculosis infection. PLoS Pathog. 2017. 13(8): e1006530.
|
| [35] |
Pesce B, Ribeiro CH, Larrondo M, et al. TNF-α Affects Signature Cytokines of Th1 and Th17 T Cell Subsets through Differential Actions on TNFR1 and TNFR2. Int J Mol Sci. 2022. 23(16): 9306.
|
| [36] |
Jr BRD, MA T, Sasse S, et al. Th1 cytokines TNF-α and IFN-γ promote corticosteroid resistance in developing human airway smooth muscle. Am J Physiol Lung Cell Mol Physiol. 2019. 16(1): L71-L81.
|
| [37] |
Li Q, Cheng Y, Zhang S, et al. TRPV4-induced Müller cell gliosis and TNF-α elevation-mediated retinal ganglion cell apoptosis in glaucomatous rats via JAK2/STAT3/NF-κB pathway. J Neuroinflammation. 2021. 18(1): 271.
|
| [38] |
Kuo HC, Chen SL, Chiu SC, et al. Tolerized Microglia Protect Neurons Against Endotoxin-Induced TNF-α Production via an LBP-Dependent Intracellular p38 MAPK Signaling Pathway. Inflammation. 2023. 46(5): 2011–2023.
|
| [39] |
Jung E, Ou S, Ahn SS, et al. The JNK-EGR1 signaling axis promotes TNF-α-induced endothelial differentiation of human mesenchymal stem cells via VEGFR2 expression. Cell Death Differ. 2023. 30(2): 356–368.
|
| [40] |
Qin F, Chen G, Yu KN, et al. Golgi Phosphoprotein 3 Mediates Radiation-Induced Bystander Effect via ERK/EGR1/TNF-α Signal Axis. Antioxidants (Basel). 2022. 11(11): 2172.
|
| [41] |
Somensi N, Brum PO, de M, et al. Extracellular HSP70 Activates ERK1/2, NF-kB and Pro-Inflammatory Gene Transcription Through Binding with RAGE in A549 Human Lung Cancer Cells. Cell Physiol Biochem. 2017. 42(6): 2507–2522.
|
| [42] |
Velez A, DeMaio A, Sterman D. Cryoablation and immunity in non-small cell lung cancer: a new era of cryo-immunotherapy. Front Immunol. 2023. 14: 1203539.
|
| [43] |
Khilwani R, Singh S. Traversing through the Mechanistic Event Analysis in IL-6 and IL-17 Signaling for a New Therapeutic Paradigm in NSCLC. Int J Mol Sci. 2024. 25(2): 1216.
|
| [44] |
Khilwani R, Singh S. Systems Biology and Cytokines Potential Role in Lung Cancer Immunotherapy Targeting Autophagic Axis. Biomedicines. 2023. 11(10): 2706.
|
| [45] |
Shill MC, Biswas B, Kamal S, et al. Screening of plasma IL-6 and IL-17 in Bangladeshi lung cancer patients. Heliyon. 2023. 9(10): e20471.
|
| [46] |
Peng P, Lou Y, Wang S, et al. Activated NK cells reprogram MDSCs via NKG2D-NKG2DL and IFN-γ to modulate anti-tumor T-cell response after cryo-thermal therapy. J Immunother Cancer. 2022. 10(12): e005769.
|
| [47] |
You J, Wang S, Zhu Y, et al. Natural Killer Cells Reprogram Myeloid-Derived Suppressor Cells to Induce TNF-α Release via NKG2D-Ligand Interaction after Cryo-Thermal Therapy. Int J Mol Sci. 2024. 25(10): 5151.
|
| [48] |
Shen Y, Liu P, Zhang A, et al. Study on tumor microvasculature damage induced by alternate cooling and heating. Ann Biomed Eng. 2008. 36(8): 1409–19.
|
| [49] |
Brighton TA, Khot A, Harrison SJ, et al. Randomized, Double-Blind, Placebo-Controlled, Multicenter Study of Siltuximab in High-Risk Smoldering Multiple Myeloma. Clin Cancer Res. 2019. 25(13): 3772–3775.
|
| [50] |
Li S, Na R, Li X, et al. Targeting interleukin-17 enhances tumor response to immune checkpoint inhibitors in colorectal cancer. Biochim Biophys Acta Rev Cancer. 2022. 1877(4): 188758.
|
| [51] |
Nada H, Sivaraman A, Lu Q, et al. Perspective for Discovery of Small Molecule IL-6 Inhibitors through Study of Structure-Activity Relationships and Molecular Docking. J Med Chem. 2023.66(7): 4417–4433.
|
| [52] |
Fauny M, Moulin D, D'Amico F, et al. Paradoxical gastrointestinal effects of interleukin-17 blockers. Ann Rheum Dis. 2020. 79(9): 1132–1138.
|
RIGHTS & PERMISSIONS
2025 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.