Your browser doesn't support javascript.
loading
De novo generation of multi-target compounds using deep generative chemistry.
Munson, Brenton P; Chen, Michael; Bogosian, Audrey; Kreisberg, Jason F; Licon, Katherine; Abagyan, Ruben; Kuenzi, Brent M; Ideker, Trey.
Affiliation
  • Munson BP; Division of Human Genomics and Precision Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
  • Chen M; Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA.
  • Bogosian A; Division of Human Genomics and Precision Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
  • Kreisberg JF; Division of Human Genomics and Precision Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
  • Licon K; Division of Human Genomics and Precision Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
  • Abagyan R; Division of Human Genomics and Precision Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
  • Kuenzi BM; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92093, USA.
  • Ideker T; Division of Human Genomics and Precision Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
Nat Commun ; 15(1): 3636, 2024 May 06.
Article in En | MEDLINE | ID: mdl-38710699
ABSTRACT
Polypharmacology drugs-compounds that inhibit multiple proteins-have many applications but are difficult to design. To address this challenge we have developed POLYGON, an approach to polypharmacology based on generative reinforcement learning. POLYGON embeds chemical space and iteratively samples it to generate new molecular structures; these are rewarded by the predicted ability to inhibit each of two protein targets and by drug-likeness and ease-of-synthesis. In binding data for >100,000 compounds, POLYGON correctly recognizes polypharmacology interactions with 82.5% accuracy. We subsequently generate de-novo compounds targeting ten pairs of proteins with documented co-dependency. Docking analysis indicates that top structures bind their two targets with low free energies and similar 3D orientations to canonical single-protein inhibitors. We synthesize 32 compounds targeting MEK1 and mTOR, with most yielding >50% reduction in each protein activity and in cell viability when dosed at 1-10 µM. These results support the potential of generative modeling for polypharmacology.
Subject(s)

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Molecular Docking Simulation Limits: Humans Language: En Journal: Nat Commun Journal subject: BIOLOGIA / CIENCIA Year: 2024 Document type: Article Affiliation country: Estados Unidos Country of publication: Reino Unido

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Molecular Docking Simulation Limits: Humans Language: En Journal: Nat Commun Journal subject: BIOLOGIA / CIENCIA Year: 2024 Document type: Article Affiliation country: Estados Unidos Country of publication: Reino Unido