4.7 Article

Mitochondrial Inhibitor Atovaquone Increases Tumor Oxygenation and Inhibits Hypoxic Gene Expression in Patients with Non-Small Cell Lung Cancer

期刊

CLINICAL CANCER RESEARCH
卷 27, 期 9, 页码 2459-2469

出版社

AMER ASSOC CANCER RESEARCH
DOI: 10.1158/1078-0432.CCR-20-4128

关键词

-

类别

资金

  1. University of Oxford
  2. Howat Foundation [C5255/A25069]
  3. CRUK & EPSRC Cancer Imaging Centre in Oxford [C5255/A16466]
  4. CRUK/MRC Oxford Institute for Radiation Oncology [C5255/A23755]
  5. NIHR Oxford Experimental Cancer Medicine Centre
  6. NIHR/HEE Clinical Lectureship [ICA-CL-2016-02-009]
  7. Jean Shanks Foundation/Pathological Society of Great Britain & Ireland Clinical Research Training Fellowship
  8. EPSRC/MRC Centre for Doctoral Training in Systems Approaches to Biomedical Science [EP/G037280/1]
  9. EPSRC Impact Acceleration Account [EP/R511742/1]
  10. CRUK Clinician Scientist Fellowship [C34326/A19590]
  11. National Institutes of Health Research (NIHR) [ICA-CL-2016-02-009] Funding Source: National Institutes of Health Research (NIHR)

向作者/读者索取更多资源

This study presents the first clinical evidence that targeting tumor mitochondrial metabolism can reduce hypoxia and produce relevant antitumor effects at the mRNA level in NSCLC. Repurposing atovaquone for this purpose may improve treatment outcomes for NSCLC patients, as demonstrated by a significant reduction in hypoxic volume and downregulation of hypoxia-regulated genes in atovaquone-treated tumors. No atovaquone-related adverse events were reported, suggesting its potential as a safe and effective treatment option for NSCLC.
Purpose: Tumor hypoxia fuels an aggressive tumor phenotype and confers resistance to anticancer treatments. We conducted a clinical trial to determine whether the antimalarial drug atovaquone, a known mitochondrial inhibitor, reduces hypoxia in non-small cell lung cancer (NSCLC). Patients and Methods: Patients with NSCLC scheduled for surgery were recruited sequentially into two cohorts: cohort 1 received oral atovaquone at the standard clinical dose of 750 mg twice daily, while cohort 2 did not. Primary imaging endpoint was change in tumor hypoxic volume (HV) measured by hypoxia PET-CT. Intercohort comparison of hypoxia gene expression signatures using RNA sequencing from resected tumors was performed. Results: Thirty patients were evaluable for hypoxia PET-CT analysis, 15 per cohort. Median treatment duration was 12 days. Eleven (73.3%) atovaquone-treated patients had meaningful HV reduction, with median change -28% [95% confidence interval (CI), -58.2 to -4.4]. In contrast, median change in untreated patients was +15.5% (95% CI, -6.5 to 35.5). Linear regression estimated the expected mean HV was 55% (95% CI, 24%-74%) lower in cohort 1 compared with cohort 2 (P = 0.004), adjusting for cohort, tumor volume, and baseline HV. A key pharmacodynamics endpoint was reduction in hypoxia-regulated genes, which were significantly downregulated in atovaquone-treated tumors. Data from multiple additional measures of tumor hypoxia and perfusion are presented. No atovaquone-related adverse events were reported. Conclusions: This is the first clinical evidence that targeting tumor mitochondrial metabolism can reduce hypoxia and produce relevant antitumor effects at the mRNA level. Repurposing atovaquone for this purpose may improve treatment outcomes for NSCLC.

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