4.7 Article

CXCR4-targeted nanotoxins induce GSDME-dependent pyroptosis in head and neck squamous cell carcinoma

出版社

BMC
DOI: 10.1186/s13046-022-02267-8

关键词

Targeted drug delivery; CXCR4; HNSCC; Pyroptosis; GSDME

类别

资金

  1. EU COST Action (FEDER, Spain) [CA 17140, PI19/01661, PI17/00584]
  2. AEI/FEDER, UE [BIO2016-76063-R]
  3. Agencia Estatal de Investigacion (AEI) [PID2019-105416RB-I00/AEI]
  4. Fondo Europeo de Desarrollo Regional (FEDER) (Spain) [PID2019-105416RB-I00/AEI]
  5. CIBER-BBN (Spain) [CB06/01/1031]
  6. AGAUR (Spain) [2017-SGR-865, 2017SGR-229]
  7. AGAUR (Spain) - European Social Fund (ESF investing in your future) [2020FI_B2 00168, 2018FI_B2_00051]
  8. AECC (Spanish Association of Cancer Research, Spain)
  9. Icrea Academia Award (Spain)
  10. Generalitat de Catalunya (Spain) [PERIS SLT006/17/00093]
  11. Instituto de Salud Carlos III (Spain) - European Social Fund (ESF investing in your future) [CP19/00028]
  12. [PI21/00150]
  13. [PI18/00650]
  14. [PIE15/00028]
  15. [PI15/00378]

向作者/读者索取更多资源

Novel therapeutic strategies are urgently needed for HNSCC patients with therapy resistance. T22-PE24-H6 and T22-DITOX-H6 nanotoxins show promising potential for the treatment of HNSCC by specifically targeting CXCR4-overexpressing cells and triggering caspase-3/GSDME-dependent pyroptosis.
Background Therapy resistance, which leads to the development of loco-regional relapses and distant metastases after treatment, constitutes one of the major problems that head and neck squamous cell carcinoma (HNSCC) patients currently face. Thus, novel therapeutic strategies are urgently needed. Targeted drug delivery to the chemokine receptor 4 (CXCR4) represents a promising approach for HNSCC management. In this context, we have developed the self-assembling protein nanotoxins T22-PE24-H6 and T22-DITOX-H6, which incorporate the de-immunized catalytic domain of Pseudomonas aeruginosa (PE24) exotoxin A and the diphtheria exotoxin (DITOX) domain, respectively. Both nanotoxins contain the T22 peptide ligand to specifically target CXCR4-overexpressing HNSCC cells. In this study, we evaluate the potential use of T22-PE24-H6 and T22-DITOX-H6 nanotoxins for the treatment of HNSCC. Methods T22-PE24-H6 and T22-DITOX-H6 CXCR4-dependent cytotoxic effect was evaluated in vitro in two different HNSCC cell lines. Both nanotoxins cell death mechanisms were assessed in HNSCC cell lines by phase-contrast microscopy, AnnexinV/ propidium iodide (PI) staining, lactate dehydrogenase (LDH) release assays, and western blotting. Nanotoxins antitumor effect in vivo was studied in a CXCR4(+) HNSCC subcutaneous mouse model. Immunohistochemistry, histopathology, and toxicity analyses were used to evaluate both nanotoxins antitumor effect and possible treatment toxicity. GSMDE and CXCR4 expression in HNSCC patient tumor samples was also assessed by immunohistochemical staining. Results First, we found that both nanotoxins exhibit a potent CXCR4-dependent cytotoxic effect in vitro. Importantly, nanotoxin treatment triggered caspase-3/Gasdermin E (GSDME)-mediated pyroptosis. The activation of this alternative cell death pathway that differs from traditional apoptosis, becomes a promising strategy to bypass therapy resistance. In addition, T22-PE24-H6 and T22-DITOX-H6 displayed a potent antitumor effect in the absence of systemic toxicity in a CXCR4(+) subcutaneous HNSCC mouse model. Lastly, GSDME was found to be overexpressed in tumor tissue from HNSCC patients, highlighting the relevance of this strategy. Conclusions Altogether, our results show that T22-PE24-H6 and T22-DITOX-H6 represent a promising therapy for HNSCC patients. Remarkably, this is the first study showing that both nanotoxins are capable of activating caspase-3/GSDME-dependent pyroptosis, opening a novel avenue for HNSCC treatment.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据