AI

Revolutionizing Fibrosis Treatment: AI-Driven Discovery of TNIK Inhibitor INS018_055 Unveils New Horizons in Therapeutics

3 Mins read

Idiopathic Pulmonary Fibrosis (IPF) and renal fibrosis present significant challenges in drug development due to their complex pathogenesis and lack of effective treatments. Despite extensive research, potential drug targets, such as TGF-β signaling pathways, have not successfully translated into viable therapies for clinical use. IPF, explained by fibroblast proliferation and extracellular matrix deposition, remains particularly lethal, with limited treatment options like nintedanib and pirfenidone. Renal fibrosis, associated with chronic kidney disease, also lacks specific inhibitors despite its increasing global prevalence. Addressing these unmet clinical needs requires innovative approaches to identify and develop effective anti-fibrotic medicines.

Researchers from multiple institutions, including Insilico Medicine, have identified TNIK as a promising anti-fibrotic target using AI. They have developed INS018_055, a TNIK inhibitor showing favorable drug properties and anti-fibrotic effects across various organs in vivo via different administration routes. The compound also exhibits anti-inflammatory effects, which have been validated in multiple animal studies. Phase I clinical trials confirmed its safety, tolerability, and pharmacokinetics in healthy individuals. This AI-driven drug discovery process, spanning from target identification to clinical validation, took approximately 18 months, demonstrating the efficacy of their approach in addressing unmet medical needs in fibrosis treatment.

The study explores the use of overexpression, knockouts, and mutations to know the relevance of pathways and interactome in a heterogeneous graph walk. It also utilizes matrix factorization and machine learning models to optimize compounds. The study involves using human tissue and clinical trials, with all tissues obtained with informed consent and adherence to HIPAA regulations. Written consent was obtained from humans participating in the clinical trials. The study follows the Declaration of Helsinki. The study mentions the canonical Wnt signaling pathway’s positive regulation, NF-kappaB transcription factor activity, and cellular response to transforming growth factor.

The study utilized predictive AI to identify TNIK as an anti-fibrotic target. An AI-driven drug discovery pipeline, incorporating pathway analysis and multiomics data, generated INS018_055, a TNIK inhibitor. Its anti-fibrotic effects were assessed through various administration routes in vivo and validated for safety in clinical trials with healthy participants. The research involved analyzing multiomics datasets, biological networks, and scientific literature to prioritize potential targets. Experimental conditions, including temperature, humidity, and gas levels, were rigorously controlled, with real-time monitoring during experiments to ensure accuracy.

Utilizing PandaOmics, an AI-driven platform, anti-fibrotic targets were discovered by integrating multiomics datasets, biological network analysis, and text data. TNIK emerged as the top candidate, unrecognized in IPF therapy, with potential implications for fibrosis and aging-related conditions. Transparency analysis revealed its involvement in crucial fibrosis-related processes and tight connection with IPF-associated genes. Single-cell expression data confirmed elevated TNIK expression in fibrotic tissue, particularly in key cell types. Simulation studies demonstrated that TNIK inhibition primarily activates Hippo signaling, suggesting its significance in regulating IPF pathogenesis. These findings underscore TNIK’s promise as a therapeutic target for fibrosis, supported by diverse AI-driven analyses.

In conclusion, researchers leveraging generative AI identified TNIK as a promising anti-fibrotic target, addressing the challenge of limited understanding in fibrotic reprogramming. Small-molecule inhibitor INS018_055 effectively mitigated fibrosis in lung, kidney, and skin models in vitro and in vivo, notably improving lung function in murine lung fibrosis. Preclinical validation and phase I trials demonstrated its safety and tolerability, with ongoing phase II trials for IPF. Integrating AI-driven target discovery and drug design approach offers a swift path to potent anti-fibrotic therapies with potential applications in COVID-19-related complications and chronic kidney disease.


Check out the PaperAll credit for this research goes to the researchers of this project. Also, don’t forget to follow us on Twitter. Join our Telegram Channel, Discord Channel, and LinkedIn Group.

If you like our work, you will love our newsletter..

Don’t Forget to join our 38k+ ML SubReddit

Want to get in front of 1.5 Million AI enthusiasts? Work with us here


Sana Hassan, a consulting intern at Marktechpost and dual-degree student at IIT Madras, is passionate about applying technology and AI to address real-world challenges. With a keen interest in solving practical problems, he brings a fresh perspective to the intersection of AI and real-life solutions.




Source link

Related posts
AI

PRISE: A Unique Machine Learning Method for Learning Multitask Temporal Action Abstractions Using Natural Language Processing (NLP)

2 Mins read
In the domain of sequential decision-making, especially in robotics, agents often deal with continuous action spaces and high-dimensional observations. These difficulties result…
AI

FLUTE: A CUDA Kernel Designed for Fused Quantized Matrix Multiplications to Accelerate LLM Inference

3 Mins read
Large Language Models (LLMs) face deployment challenges due to latency issues caused by memory bandwidth constraints. Researchers use weight-only quantization to address…
AI

Self-Route: A Simple Yet Effective AI Method that Routes Queries to RAG or Long Context LC based on Model Self-Reflection

3 Mins read
Large Language Models (LLMs) have revolutionized the field of natural language processing, allowing machines to understand and generate human language. These models,…

 

 

Leave a Reply

Your email address will not be published. Required fields are marked *