1. Academic Validation
  2. Connectivity mapping-based identification of pharmacological inhibitor targeting HDAC6 in aggressive pancreatic ductal adenocarcinoma

Connectivity mapping-based identification of pharmacological inhibitor targeting HDAC6 in aggressive pancreatic ductal adenocarcinoma

  • NPJ Precis Oncol. 2024 Mar 7;8(1):66. doi: 10.1038/s41698-024-00562-5.
Pranita Atri 1 Ashu Shah 1 Gopalakrishnan Natarajan 1 Satyanarayana Rachagani 1 Sanchita Rauth 1 Koelina Ganguly 1 Joseph Carmicheal 1 Dario Ghersi 2 Jesse L Cox 3 Lynette M Smith 4 Maneesh Jain 1 Sushil Kumar 1 Moorthy P Ponnusamy 1 Parthasarathy Seshacharyulu 5 Surinder K Batra 6 7 8
Affiliations

Affiliations

  • 1 Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
  • 2 School of Interdisciplinary Informatics, College of Information Science and Technology, University of Nebraska at Omaha, Omaha, NE, USA.
  • 3 Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA.
  • 4 Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE, USA.
  • 5 Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA. [email protected].
  • 6 Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA. [email protected].
  • 7 Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA. [email protected].
  • 8 Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA. [email protected].
Abstract

Pancreatic ductal adenocarcinoma (PDAC) remains highly lethal due to limited therapeutic options and expensive/burdensome drug discovery processes. Utilizing genomic-data-driven Connectivity Mapping (CMAP) to identify a drug closer to real-world PC targeting may improve pancreatic Cancer (PC) patient outcomes. Initially, we mapped CMAP data to gene expression from 106 PC patients, identifying nine negatively connected drugs. These drugs were further narrowed down using a similar analysis for PC cell lines, human tumoroids, and patient-derived xenografts datasets, where ISOX emerged as the most potent agent to target PC. We used human and mouse syngeneic PC cells, human and mouse tumoroids, and in vivo mice to assess the ability of ISOX alone and in combination with 5FU to inhibit tumor growth. Global transcriptomic and pathway analysis of the ISOX-LINCS signature identified HDAC 6/cMyc as the target axis for ISOX. Specifically, we discovered that genetic and pharmacological targeting of HDAC 6 affected non-histone protein cMyc acetylation, leading to cMyc instability, thereby disrupting PC growth and metastasis by affecting Cancer stemness. Finally, KrasG12D harboring tumoroids and mice responded effectively against ISOX and 5FU treatment by enhancing survival and controlling metastasis incidence. Overall, our data validate ISOX as a new drug to treat advanced PC patients without toxicity to normal cells. Our study supports the clinical utility of ISOX along with 5FU in future PC clinical trials.

Figures
Products