HOMEPRODUCTSCOMPANYCONTACTFAQResearchDictionaryPharmaSign Up FREE or Login

Sensitizing tumors to anti-PD-1 therapy by promoting NK and CD8+ T cells via pharmacological activation of FOXO3.

AbstractBACKGROUND:
Stimulating antitumor immunity by blocking programmed death-1 (PD-1) or its ligand (programmed death-ligand 1 (PD-L1) is a promising antitumor therapy. However, numerous patients respond poorly to PD-1/PD-L1 blockade. Unresponsiveness to immune-checkpoint blockade (ICB) can cast significant challenges to the therapeutic options for patients with hard-to-treat tumors. There is an unmet clinical need to establish new therapeutic approaches for mitigating ICB unresponsiveness in patients. In this study, we investigated the efficacy and role of low-dose antineoplastic agent SN-38 or metformin in sensitizing unresponsive tumors to respond to ICB therapy.
METHODS:
We assessed the significant pathological relationships between PD-L1 and FOXO3 expression and between PD-L1 and c-Myc or STAT3 expression in patients with various tumors. We determined the efficacy of low-dose SN-38 or metformin in sensitizing unresponsive tumors to respond to anti-PD-1 therapy in a syngeneic tumor system. We deciphered novel therapeutic mechanisms underlying the SN-38 and anti-PD-1 therapy-mediated engagement of natural killer (NK) or CD8+ T cells to infiltrate tumors and boost antitumor immunity.
RESULTS:
We showed that PD-L1 protein level was inversely associated with FOXO3 protein level in patients with ovarian, breast, and hepatocellular tumors. Low-dose SN-38 or metformin abrogated PD-L1 protein expression, promoted FOXO3 protein level, and significantly increased the animal survival rate in syngeneic mouse tumor models. SN-38 or metformin sensitized unresponsive tumors responding to anti-PD-1 therapy by engaging NK or CD8+ T cells to infiltrate the tumor microenvironment (TME) and secret interferon-γ and granzyme B to kill tumors. SN-38 suppressed the levels of c-Myc and STAT3 proteins, which controlled PD-L1 expression. FOXO3 was essential for SN38-mediated PD-L1 suppression. The expression of PD-L1 was compellingly linked to that of c-Myc or STAT3 in patients with the indicated tumors.
CONCLUSION:
We show that SN-38 or metformin can boost antitumor immunity in the TME by inhibiting c-Myc and STAT3 through FOXO3 activation. These results may provide novel insight into ameliorating patient response to overarching immunotherapy for tumors.
AuthorsYoung Min Chung, Pragya P Khan, Hong Wang, Wen-Bin Tsai, Yanli Qiao, Bo Yu, James W Larrick, Mickey C-T Hu
JournalJournal for immunotherapy of cancer (J Immunother Cancer) Vol. 9 Issue 12 (12 2021) ISSN: 2051-1426 [Electronic] England
PMID34887262 (Publication Type: Journal Article, Research Support, N.I.H., Extramural, Research Support, Non-U.S. Gov't, Research Support, U.S. Gov't, Non-P.H.S.)
Copyright© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.
Chemical References
  • B7-H1 Antigen
  • CD274 protein, human
  • FOXO3 protein, human
  • Forkhead Box Protein O3
  • Immune Checkpoint Inhibitors
  • PDCD1 protein, human
  • Programmed Cell Death 1 Receptor
  • Topoisomerase I Inhibitors
  • Irinotecan
Topics
  • Animals
  • Apoptosis
  • B7-H1 Antigen (antagonists & inhibitors, metabolism)
  • Breast Neoplasms (drug therapy, immunology, metabolism, pathology)
  • CD8-Positive T-Lymphocytes (immunology)
  • Carcinoma, Hepatocellular (drug therapy, immunology, metabolism, pathology)
  • Cell Proliferation
  • Female
  • Forkhead Box Protein O3 (genetics, metabolism)
  • Gene Expression Regulation, Neoplastic (drug effects)
  • Humans
  • Immune Checkpoint Inhibitors (pharmacology)
  • Immunotherapy
  • Irinotecan (pharmacology)
  • Killer Cells, Natural (immunology)
  • Liver Neoplasms (drug therapy, immunology, metabolism, pathology)
  • Mice
  • Mice, Inbred C57BL
  • Ovarian Neoplasms (drug therapy, immunology, metabolism, pathology)
  • Prognosis
  • Programmed Cell Death 1 Receptor (antagonists & inhibitors)
  • Topoisomerase I Inhibitors (pharmacology)
  • Tumor Cells, Cultured
  • Tumor Microenvironment

Join CureHunter, for free Research Interface BASIC access!

Take advantage of free CureHunter research engine access to explore the best drug and treatment options for any disease. Find out why thousands of doctors, pharma researchers and patient activists around the world use CureHunter every day.
Realize the full power of the drug-disease research graph!


Choose Username:
Email:
Password:
Verify Password:
Enter Code Shown: