Eliminating Radiation Resistance of Non-Small Cell Lung Cancer by Dihydroartemisinin Through Abrogating Immunity Escaping and Promoting Radiation Sensitivity by Inhibiting PD-L1 Expression

Radiation resistance is linked to immune escaping and radiation sensitivity. In this study, we found that the PD-L1 expressions of non-killed tumor cells in NSCLC were enhanced after radiotherapy, and dihydroartemisinin (DHA) could synergistically enhance the antitumor effect of radiotherapy in NSCLC. A total of 48 NSCLC patients with sufficient tumor tissues for further analyses were enrolled. The PD-L1 expressions of NSCLC were evaluated by immunohistochemistry. Cell apoptosis was measured by flow cytometry, and the relationship between the PD-L1 expression and radiation resistance was investigated in patient specimens, xenograft model, and cell lines. First, the results indicate that the PD-L1 expression of NSCLC was positively related with the radiation resistance. Second, we found that DHA could eliminate the radiation resistance and synergistically enhance the antitumor effect of radiotherapy in the NSCLC cells lines and xenograft model. Finally, mechanistically, DHA could inhibit the PD-L1 expression to avoid immune escaping by inhibiting TGF-β, PI3K/Akt, and STAT3 signaling pathways. In addition, DHA could activate TRIM21 and regulate the EMT-related proteins by inhibiting the PD-L1 so as to enhance the radiation sensitivity and eliminate radiation resistance to NSCLC. Collectively, this study established a basis for the rational design of integrated radiotherapy and DHA for the treatment of NSCLC.

[1]  K. Ozato,et al.  TRIM21 Dysfunction Enhances Aberrant B-Cell Differentiation in Autoimmune Pathogenesis , 2020, Frontiers in Immunology.

[2]  E. Zervos,et al.  Emerging Role of Immune Checkpoint Blockade in Pancreatic Cancer , 2018, International journal of molecular sciences.

[3]  U. Ricardi,et al.  Locally-advanced non-small cell lung cancer: shall immunotherapy be a new chance? , 2018, Journal of thoracic disease.

[4]  R. Weichselbaum,et al.  JAK2 Inhibitor SAR302503 Abrogates PD-L1 Expression and Targets Therapy-Resistant Non–small Cell Lung Cancers , 2018, Molecular Cancer Therapeutics.

[5]  Yuhchyau Chen,et al.  Radiation alters PD-L1/NKG2D ligand levels in lung cancer cells and leads to immune escape from NK cell cytotoxicity via IL-6-MEK/Erk signaling pathway , 2017, Oncotarget.

[6]  T. Jiang,et al.  Combined Radiotherapy and Anti–PD‐L1 Antibody Synergistically Enhances Antitumor Effect in Non–Small Cell Lung Cancer , 2017, Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer.

[7]  S. Fujita,et al.  PD-L1 Expression in Patients with Non-small Cell Lung Cancer. , 2017, Anticancer research.

[8]  T. Burns,et al.  The next generation of immunotherapy: keeping lung cancer in check , 2017, Journal of Hematology & Oncology.

[9]  R. Irby,et al.  TRIM21 is a novel regulator of Par-4 in colon and pancreatic cancer cells , 2016, Cancer biology & therapy.

[10]  J. Larkin,et al.  Immunotherapy Combined or Sequenced With Targeted Therapy in the Treatment of Solid Tumors: Current Perspectives. , 2016, Journal of the National Cancer Institute.

[11]  A. Jemal,et al.  Cancer statistics in China, 2015 , 2016, CA: a cancer journal for clinicians.

[12]  Hui Li,et al.  Dihydroartemisinin as a Putative STAT3 Inhibitor, Suppresses the Growth of Head and Neck Squamous Cell Carcinoma by Targeting Jak2/STAT3 Signaling , 2016, PloS one.

[13]  H. Rodemann,et al.  Phosphatidylinositol 3-kinase/Akt signaling as a key mediator of tumor cell responsiveness to radiation. , 2015, Seminars in cancer biology.

[14]  C. Simone,et al.  Novel radiotherapy approaches for lung cancer: combining radiation therapy with targeted and immunotherapies. , 2015, Translational lung cancer research.

[15]  H. Feng,et al.  Dihydroartemisinin exhibits anti-glioma stem cell activity through inhibiting p-AKT and activating caspase-3. , 2014, Die Pharmazie.

[16]  I. Stratford,et al.  Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. , 2014, Cancer research.

[17]  F. Hirsch,et al.  MiR-200c overexpression is associated with better efficacy of EGFR-TKIs in non-small cell lung cancer patients with EGFR wild-type , 2014, Oncotarget.

[18]  M. Fukumoto,et al.  AKT-mediated enhanced aerobic glycolysis causes acquired radioresistance by human tumor cells. , 2014, Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology.

[19]  R. Weichselbaum,et al.  Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. , 2014, The Journal of clinical investigation.

[20]  Andy H. Choi,et al.  Current Perspectives , 2013, Journal of dental research.

[21]  Tongsheng Chen,et al.  Ionizing Radiation Potentiates Dihydroartemisinin-Induced Apoptosis of A549 Cells via a Caspase-8-Dependent Pathway , 2013, PloS one.

[22]  M. Fukumoto,et al.  Cyclin D1 overexpression perturbs DNA replication and induces replication-associated DNA double-strand breaks in acquired radioresistant cells , 2013, Cell cycle.

[23]  Ying Li Qinghaosu (artemisinin): Chemistry and pharmacology , 2012, Acta Pharmacologica Sinica.

[24]  R. Hallett,et al.  Artemisinin-Resistant Malaria: Research Challenges, Opportunities, and Public Health Implications , 2012, The American journal of tropical medicine and hygiene.

[25]  T. Efferth,et al.  A radiosensitizing effect of artesunate in glioblastoma cells is associated with a diminished expression of the inhibitor of apoptosis protein survivin. , 2012, Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology.

[26]  S. Fu,et al.  Selective Radiosensitization of Human Cervical Cancer Cells and Normal Cells by Artemisinin Through the Abrogation of Radiation-Induced G2 Block , 2012, International Journal of Gynecologic Cancer.

[27]  X. Pang,et al.  Artesunate enhances radiosensitivity of human non-small cell lung cancer A549 cells via increasing NO production to induce cell cycle arrest at G2/M phase. , 2011, International immunopharmacology.

[28]  T. Wellems,et al.  The threat of artemisinin-resistant malaria. , 2011, The New England journal of medicine.

[29]  N G Burnet,et al.  Guidelines for preclinical and early phase clinical assessment of novel radiosensitisers , 2011, British Journal of Cancer.

[30]  Y. Kuwahara,et al.  Acquired radioresistance of human tumor cells by DNA-PK/AKT/GSK3β-mediated cyclin D1 overexpression , 2010, Oncogene.

[31]  R. Kong,et al.  Dihydroartemisinin inactivates NF-kappaB and potentiates the anti-tumor effect of gemcitabine on pancreatic cancer both in vitro and in vivo. , 2010, Cancer letters.

[32]  Ao Li,et al.  Dihydroartemisinin improves the efficiency of chemotherapeutics in lung carcinomas in vivo and inhibits murine Lewis lung carcinoma cell line growth in vitro , 2010, Cancer Chemotherapy and Pharmacology.

[33]  Haiyang Xie,et al.  Interaction of B7‐H1 on intrahepatic cholangiocarcinoma cells with PD‐1 on tumor‐infiltrating T cells as a mechanism of immune evasion , 2009, Journal of surgical oncology.

[34]  W. Oyen,et al.  The PI3-K/AKT-Pathway and Radiation Resistance Mechanisms in Non-small Cell Lung Cancer , 2009, Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer.

[35]  Tongsheng Chen,et al.  Dihydroartemisinin (DHA) induces caspase-3-dependent apoptosis in human lung adenocarcinoma ASTC-a-1 cells , 2009, Journal of Biomedical Science.

[36]  Hui Wang,et al.  Experimental Therapy of Hepatoma with Artemisinin and Its Derivatives: In vitro and In vivo Activity, Chemosensitization, and Mechanisms of Action , 2008, Clinical Cancer Research.

[37]  F. Jin,et al.  The role of calcium, P38 MAPK in dihydroartemisinin-induced apoptosis of lung cancer PC-14 cells , 2008, Cancer Chemotherapy and Pharmacology.

[38]  S. Fan,et al.  Dihydroartemisinin is an inhibitor of ovarian cancer cell growth , 2007, Acta Pharmacologica Sinica.

[39]  Woong Nam,et al.  Effects of artemisinin and its derivatives on growth inhibition and apoptosis of oral cancer cells , 2007, Head & neck.

[40]  S. J. Kim,et al.  Dihydroartemisinin enhances radiosensitivity of human glioma cells in vitro , 2006, Journal of Cancer Research and Clinical Oncology.

[41]  P. O’Neill,et al.  A medicinal chemistry perspective on artemisinin and related endoperoxides. , 2004, Journal of medicinal chemistry.

[42]  S. Meshnick,et al.  Artemisinin: mechanisms of action, resistance and toxicity. , 2002, International journal for parasitology.

[43]  J. Yewdell,et al.  Identification of human cancers deficient in antigen processing , 1993, The Journal of experimental medicine.

[44]  P. Harari,et al.  Radiation and new molecular agents, part II: targeting HDAC, HSP90, IGF-1R, PI3K, and Ras. , 2006, Seminars in radiation oncology.