Boosting Clear Cell Renal Carcinoma-Specific Drug Discovery Using a Deep Learning Algorithm and Single-Cell Analysis

Int J Mol Sci. 2024 Apr 8;25(7):4134. doi: 10.3390/ijms25074134.

Abstract

Clear cell renal carcinoma (ccRCC), the most common subtype of renal cell carcinoma, has the high heterogeneity of a highly complex tumor microenvironment. Existing clinical intervention strategies, such as target therapy and immunotherapy, have failed to achieve good therapeutic effects. In this article, single-cell transcriptome sequencing (scRNA-seq) data from six patients downloaded from the GEO database were adopted to describe the tumor microenvironment (TME) of ccRCC, including its T cells, tumor-associated macrophages (TAMs), endothelial cells (ECs), and cancer-associated fibroblasts (CAFs). Based on the differential typing of the TME, we identified tumor cell-specific regulatory programs that are mediated by three key transcription factors (TFs), whilst the TF EPAS1/HIF-2α was identified via drug virtual screening through our analysis of ccRCC's protein structure. Then, a combined deep graph neural network and machine learning algorithm were used to select anti-ccRCC compounds from bioactive compound libraries, including the FDA-approved drug library, natural product library, and human endogenous metabolite compound library. Finally, five compounds were obtained, including two FDA-approved drugs (flufenamic acid and fludarabine), one endogenous metabolite, one immunology/inflammation-related compound, and one inhibitor of DNA methyltransferase (N4-methylcytidine, a cytosine nucleoside analogue that, like zebularine, has the mechanism of inhibiting DNA methyltransferase). Based on the tumor microenvironment characteristics of ccRCC, five ccRCC-specific compounds were identified, which would give direction of the clinical treatment for ccRCC patients.

Keywords: EPAS1/HIF-2α; ccRCC; deep learning algorithm; single-cell RNA sequencing; specific drug discovery; tumor microenvironment heterogeneity.

MeSH terms

  • Algorithms
  • Antimetabolites
  • Carcinoma, Renal Cell* / drug therapy
  • DNA
  • DNA Modification Methylases
  • Deep Learning*
  • Drug Discovery
  • Endothelial Cells
  • Humans
  • Kidney Neoplasms* / drug therapy
  • Single-Cell Analysis
  • Tumor Microenvironment

Substances

  • Antimetabolites
  • DNA Modification Methylases
  • DNA