1
|
Wilczok D, Zhavoronkov A. Progress, Pitfalls, and Impact of AI-Driven Clinical Trials. Clin Pharmacol Ther 2024. [PMID: 39722473 DOI: 10.1002/cpt.3542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024]
Abstract
Since the deep learning revolution of the early 2010s, significant efforts and billions of dollars have been invested in applying artificial intelligence (AI) to drug discovery and development (AIDD). However, despite high expectations, few AI-discovered or AI-designed drugs have entered human clinical trials, and none have achieved clinical approval. In this perspective, we examine the challenges impeding progress and highlight opportunities to harness AI's potential in transforming drug discovery and development.
Collapse
Affiliation(s)
| | - Alex Zhavoronkov
- Insilico Medicine US Inc, Cambridge, Massachusetts, USA
- Insilico Medicine AI Ltd, Abu Dhabi, UAE
- Insilico Medicine Hong Kong Ltd, Hong Kong, SAR, China
| |
Collapse
|
2
|
Wu Y, Ma L, Li X, Yang J, Rao X, Hu Y, Xi J, Tao L, Wang J, Du L, Chen G, Liu S. The role of artificial intelligence in drug screening, drug design, and clinical trials. Front Pharmacol 2024; 15:1459954. [PMID: 39679365 PMCID: PMC11637864 DOI: 10.3389/fphar.2024.1459954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/11/2024] [Indexed: 12/17/2024] Open
Abstract
The role of computational tools in drug discovery and development is becoming increasingly important due to the rapid development of computing power and advancements in computational chemistry and biology, improving research efficiency and reducing the costs and potential risks of preclinical and clinical trials. Machine learning, especially deep learning, a subfield of artificial intelligence (AI), has demonstrated significant advantages in drug discovery and development, including high-throughput and virtual screening, ab initio design of drug molecules, and solving difficult organic syntheses. This review summarizes AI technologies used in drug discovery and development, including their roles in drug screening, design, and solving the challenges of clinical trials. Finally, it discusses the challenges of drug discovery and development based on AI technologies, as well as potential future directions.
Collapse
Affiliation(s)
- Yuyuan Wu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Lijing Ma
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xinyi Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jingpeng Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xinyu Rao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yiru Hu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jingyi Xi
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Lin Tao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jianjun Wang
- Department of Respiratory Medicine of Affiliated Hospital, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Lailing Du
- Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Gongxing Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Shuiping Liu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
García-Vázquez FA. Artificial intelligence and porcine breeding. Anim Reprod Sci 2024; 269:107538. [PMID: 38926001 DOI: 10.1016/j.anireprosci.2024.107538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024]
Abstract
Livestock management is evolving into a new era, characterized by the analysis of vast quantities of data (Big Data) collected from both traditional breeding methods and new technologies such as sensors, automated monitoring system, and advanced analytics. Artificial intelligence (A-In), which refers to the capability of machines to mimic human intelligence, including subfields like machine learning and deep learning, is playing a pivotal role in this transformation. A wide array of A-In techniques, successfully employed in various industrial and scientific contexts, are now being integrated into mainstream livestock management practices. In the case of swine breeding, while traditional methods have yielded considerable success, the increasing amount of information requires the adoption of new technologies such as A-In to drive productivity, enhance animal welfare, and reduce environmental impact. Current findings suggest that these techniques have the potential to match or exceed the performance of traditional methods, often being more scalable in terms of efficiency and sustainability within the breeding industry. This review provides insights into the application of A-In in porcine breeding, from the perspectives of both sows (including welfare and reproductive management) and boars (including semen quality and health), and explores new approaches which are already being applied in other species.
Collapse
Affiliation(s)
- Francisco A García-Vázquez
- Departamento de Fisiología, Facultad de Veterinaria, Campus de Excelencia Mare Nostrum, Universidad de Murcia, Murcia 30100, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), Murcia, Spain.
| |
Collapse
|
4
|
Zhang Y, Mastouri M, Zhang Y. Accelerating drug discovery, development, and clinical trials by artificial intelligence. MED 2024; 5:1050-1070. [PMID: 39173629 DOI: 10.1016/j.medj.2024.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/21/2024] [Accepted: 07/25/2024] [Indexed: 08/24/2024]
Abstract
Artificial intelligence (AI) has profoundly advanced the field of biomedical research, which also demonstrates transformative capacity for innovation in drug development. This paper aims to deliver a comprehensive analysis of the progress in AI-assisted drug development, particularly focusing on small molecules, RNA, and antibodies. Moreover, this paper elucidates the current integration of AI methodologies within the industrial drug development framework. This encompasses a detailed examination of the industry-standard drug development process, supplemented by a review of medications presently undergoing clinical trials. Conclusively, the paper tackles a predominant obstacle within the AI pharmaceutical sector: the absence of AI-conceived drugs receiving approval. This paper also advocates for the adoption of large language models and diffusion models as a viable strategy to surmount this challenge. This review not only underscores the significant potential of AI in drug discovery but also deliberates on the challenges and prospects within this dynamically progressing field.
Collapse
Affiliation(s)
- Yilun Zhang
- College of Science, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong, China; School of Medicine, The Chinese University of Hong Kong (Shenzhen), Shenzhen, Guangdong, China
| | - Mohamed Mastouri
- College of Science, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong, China
| | - Yang Zhang
- College of Science, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong, China.
| |
Collapse
|
5
|
Guo Q, Fu B, Tian Y, Xu S, Meng X. Recent progress in artificial intelligence and machine learning for novel diabetes mellitus medications development. Curr Med Res Opin 2024; 40:1483-1493. [PMID: 39083361 DOI: 10.1080/03007995.2024.2387187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 07/29/2024] [Indexed: 08/02/2024]
Abstract
Diabetes mellitus, stemming from either insulin resistance or inadequate insulin secretion, represents a complex ailment that results in prolonged hyperglycemia and severe complications. Patients endure severe ramifications such as kidney disease, vision impairment, cardiovascular disorders, and susceptibility to infections, leading to significant physical suffering and imposing substantial socio-economic burdens. This condition has evolved into an increasingly severe health crisis. There is an urgent need to develop new treatments with improved efficacy and fewer adverse effects to meet clinical demands. However, novel drug development is costly, time-consuming, and often associated with side effects and suboptimal efficacy, making it a major challenge. Artificial Intelligence (AI) and Machine Learning (ML) have revolutionized drug development across its comprehensive lifecycle, spanning drug discovery, preclinical studies, clinical trials, and post-market surveillance. These technologies have significantly accelerated the identification of promising therapeutic candidates, optimized trial designs, and enhanced post-approval safety monitoring. Recent advances in AI, including data augmentation, interpretable AI, and integration of AI with traditional experimental methods, offer promising strategies for overcoming the challenges inherent in AI-based drug discovery. Despite these advancements, there exists a notable gap in comprehensive reviews detailing AI and ML applications throughout the entirety of developing medications for diabetes mellitus. This review aims to fill this gap by evaluating the impact and potential of AI and ML technologies at various stages of diabetes mellitus drug development. It does that by synthesizing current research findings and technological advances so as to effectively control diabetes mellitus and mitigate its far-reaching social and economic impacts. The integration of AI and ML promises to revolutionize diabetes mellitus treatment strategies, offering hope for improved patient outcomes and reduced healthcare burdens worldwide.
Collapse
Affiliation(s)
- Qi Guo
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, P. R. China
| | - Bo Fu
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, P. R. China
| | - Yuan Tian
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, P. R. China
| | - Shujun Xu
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, P. R. China
| | - Xin Meng
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, P. R. China
| |
Collapse
|
6
|
Aribindi K, Liu GY, Albertson TE. Emerging pharmacological options in the treatment of idiopathic pulmonary fibrosis (IPF). Expert Rev Clin Pharmacol 2024; 17:817-835. [PMID: 39192604 PMCID: PMC11441789 DOI: 10.1080/17512433.2024.2396121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/20/2024] [Indexed: 08/29/2024]
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis (IPF) is a progressive-fibrosing lung disease with a median survival of less than 5 years. Currently, two agents, pirfenidone and nintedanib are approved for this disease, and both have been shown to reduce the rate of decline in lung function in patients with IPF. However, both have significant adverse effects and neither completely arrest the decline in lung function. AREAS COVERED Thirty experimental agents with unique mechanisms of action that are being evaluated for the treatment of IPF are discussed. These agents work through various mechanisms of action, these include inhibition of transcription nuclear factor k-B on fibroblasts, reduced expression of metalloproteinase 7, the generation of more lysophosphatidic acids, blocking the effects of transforming growth factor ß, and reducing reactive oxygen species as examples of some unique mechanisms of action of these agents. EXPERT OPINION New drug development has the potential to expand the treatment options available in the treatment of IPF patients. It is expected that the adverse drug effect profiles will be more favorable than current agents. It is further anticipated that these new agents or combinations of agents will arrest the fibrosis, not just slow the fibrotic process.
Collapse
Affiliation(s)
- Katyayini Aribindi
- Department of Internal Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, University of California Davis, School of Medicine, Sacramento, CA, USA
- Department of Medicine, Department of Veterans Affairs Northern California Health Care System, Mather, CA, USA
| | - Gabrielle Y Liu
- Department of Internal Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, University of California Davis, School of Medicine, Sacramento, CA, USA
| | - Timothy E Albertson
- Department of Internal Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, University of California Davis, School of Medicine, Sacramento, CA, USA
- Department of Medicine, Department of Veterans Affairs Northern California Health Care System, Mather, CA, USA
| |
Collapse
|
7
|
Sinha S, McLaren E, Mullick M, Singh S, Boland BS, Ghosh P. FORWARD: A Learning Framework for Logical Network Perturbations to Prioritize Targets for Drug Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.602603. [PMID: 39071297 PMCID: PMC11275938 DOI: 10.1101/2024.07.16.602603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Despite advances in artificial intelligence (AI), target-based drug development remains a costly, complex and imprecise process. We introduce F.O.R.W.A.R.D [ Framework for Outcome-based Research and Drug Development ], a network-based target prioritization approach and test its utility in the challenging therapeutic area of Inflammatory Bowel Diseases (IBD), which is a chronic condition of multifactorial origin. F.O.R.W.A.R.D leverages real-world outcomes, using a machine-learning classifier trained on transcriptomic data from seven prospective randomized clinical trials involving four drugs. It establishes a molecular signature of remission as the therapeutic goal and computes, by integrating principles of network connectivity, the likelihood that a drug's action on its target(s) will induce the remission-associated genes. Benchmarking F.O.R.W.A.R.D against 210 completed clinical trials on 52 targets showed a perfect predictive accuracy of 100%. The success of F.O.R.W.A.R.D was achieved despite differences in targets, mechanisms, and trial designs. F.O.R.W.A.R.D-driven in-silico phase '0' trials revealed its potential to inform trial design, justify re-trialing failed drugs, and guide early terminations. With its extendable applications to other therapeutic areas and its iterative refinement with emerging trials, F.O.R.W.A.R.D holds the promise to transform drug discovery by generating foresight from hindsight and impacting research and development as well as human-in-the-loop clinical decision-making.
Collapse
|
8
|
Vidovszky AA, Fisher CK, Loukianov AD, Smith AM, Tramel EW, Walsh JR, Ross JL. Increasing acceptance of AI-generated digital twins through clinical trial applications. Clin Transl Sci 2024; 17:e13897. [PMID: 39039704 PMCID: PMC11263130 DOI: 10.1111/cts.13897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 07/24/2024] Open
Abstract
Today's approach to medicine requires extensive trial and error to determine the proper treatment path for each patient. While many fields have benefited from technological breakthroughs in computer science, such as artificial intelligence (AI), the task of developing effective treatments is actually getting slower and more costly. With the increased availability of rich historical datasets from previous clinical trials and real-world data sources, one can leverage AI models to create holistic forecasts of future health outcomes for an individual patient in the form of an AI-generated digital twin. This could support the rapid evaluation of intervention strategies in silico and could eventually be implemented in clinical practice to make personalized medicine a reality. In this work, we focus on uses for AI-generated digital twins of clinical trial participants and contend that the regulatory outlook for this technology within drug development makes it an ideal setting for the safe application of AI-generated digital twins in healthcare. With continued research and growing regulatory acceptance, this path will serve to increase trust in this technology and provide momentum for the widespread adoption of AI-generated digital twins in clinical practice.
Collapse
|
9
|
Livne M, Miftahutdinov Z, Tutubalina E, Kuznetsov M, Polykovskiy D, Brundyn A, Jhunjhunwala A, Costa A, Aliper A, Aspuru-Guzik A, Zhavoronkov A. nach0: multimodal natural and chemical languages foundation model. Chem Sci 2024; 15:8380-8389. [PMID: 38846388 PMCID: PMC11151847 DOI: 10.1039/d4sc00966e] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/26/2024] [Indexed: 06/09/2024] Open
Abstract
Large Language Models (LLMs) have substantially driven scientific progress in various domains, and many papers have demonstrated their ability to tackle complex problems with creative solutions. Our paper introduces a new foundation model, nach0, capable of solving various chemical and biological tasks: biomedical question answering, named entity recognition, molecular generation, molecular synthesis, attributes prediction, and others. nach0 is a multi-domain and multi-task encoder-decoder LLM pre-trained on unlabeled text from scientific literature, patents, and molecule strings to incorporate a range of chemical and linguistic knowledge. We employed instruction tuning, where specific task-related instructions are utilized to fine-tune nach0 for the final set of tasks. To train nach0 effectively, we leverage the NeMo framework, enabling efficient parallel optimization of both base and large model versions. Extensive experiments demonstrate that our model outperforms state-of-the-art baselines on single-domain and cross-domain tasks. Furthermore, it can generate high-quality outputs in molecular and textual formats, showcasing its effectiveness in multi-domain setups.
Collapse
Affiliation(s)
- Micha Livne
- NVIDIA 2788 San Tomas Expressway Santa Clara 95051 CA USA
| | - Zulfat Miftahutdinov
- Insilico Medicine Canada Inc. 3710-1250 René-Lévesque West Montreal Quebec Canada
| | - Elena Tutubalina
- Insilico Medicine Hong Kong Ltd. Unit 310, 3/F, Building 8W, Phase 2, Hong Kong Science Park, Pak Shek Kok New Territories Hong Kong
| | - Maksim Kuznetsov
- Insilico Medicine Canada Inc. 3710-1250 René-Lévesque West Montreal Quebec Canada
| | - Daniil Polykovskiy
- Insilico Medicine Canada Inc. 3710-1250 René-Lévesque West Montreal Quebec Canada
| | - Annika Brundyn
- NVIDIA 2788 San Tomas Expressway Santa Clara 95051 CA USA
| | | | - Anthony Costa
- NVIDIA 2788 San Tomas Expressway Santa Clara 95051 CA USA
| | - Alex Aliper
- Insilico Medicine AI Ltd. Level 6, Unit 08, Block A, IRENA HQ Building, Masdar City Abu Dhabi United Arab Emirates
| | - Alán Aspuru-Guzik
- University of Toronto Lash Miller Building 80 St. George Street Toronto Ontario Canada
| | - Alex Zhavoronkov
- Insilico Medicine Hong Kong Ltd. Unit 310, 3/F, Building 8W, Phase 2, Hong Kong Science Park, Pak Shek Kok New Territories Hong Kong
| |
Collapse
|
10
|
Jacobsen LM, Cuthbertson D, Bundy BN, Atkinson MA, Moore W, Haller MJ, Russell WE, Gitelman SE, Herold KC, Redondo MJ, Sims EK, Wherrett DK, Moran A, Pugliese A, Gottlieb PA, Sosenko JM, Ismail HM. Early Metabolic Endpoints Identify Persistent Treatment Efficacy in Recent-Onset Type 1 Diabetes Immunotherapy Trials. Diabetes Care 2024; 47:1048-1055. [PMID: 38621411 PMCID: PMC11294635 DOI: 10.2337/dc24-0171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/18/2024] [Indexed: 04/17/2024]
Abstract
OBJECTIVE Mixed-meal tolerance test-stimulated area under the curve (AUC) C-peptide at 12-24 months represents the primary end point for nearly all intervention trials seeking to preserve β-cell function in recent-onset type 1 diabetes. We hypothesized that participant benefit might be detected earlier and predict outcomes at 12 months posttherapy. Such findings would support shorter trials to establish initial efficacy. RESEARCH DESIGN AND METHODS We examined data from six Type 1 Diabetes TrialNet immunotherapy randomized controlled trials in a post hoc analysis and included additional stimulated metabolic indices beyond C-peptide AUC. We partitioned the analysis into successful and unsuccessful trials and analyzed the data both in the aggregate as well as individually for each trial. RESULTS Among trials meeting their primary end point, we identified a treatment effect at 3 and 6 months when using C-peptide AUC (P = 0.030 and P < 0.001, respectively) as a dynamic measure (i.e., change from baseline). Importantly, no such difference was seen in the unsuccessful trials. The use of C-peptide AUC as a 6-month dynamic measure not only detected treatment efficacy but also suggested long-term C-peptide preservation (R2 for 12-month C-peptide AUC adjusted for age and baseline value was 0.80, P < 0.001), and this finding supported the concept of smaller trial sizes down to 54 participants. CONCLUSIONS Early dynamic measures can identify a treatment effect among successful immune therapies in type 1 diabetes trials with good long-term prediction and practical sample size over a 6-month period. While external validation of these findings is required, strong rationale and data exist in support of shortening early-phase clinical trials.
Collapse
Affiliation(s)
- Laura M. Jacobsen
- Department of Pediatrics, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL
| | - David Cuthbertson
- Health Informatics Institute, University of South Florida, Tampa, FL
| | - Brian N. Bundy
- Health Informatics Institute, University of South Florida, Tampa, FL
| | - Mark A. Atkinson
- Department of Pediatrics, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL
| | - Wayne Moore
- Pediatric Endocrinology, Children’s Mercy Hospital/University of Missouri-Kansas City Mercy, Kansas City, MO
| | - Michael J. Haller
- Department of Pediatrics, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL
| | | | | | | | - Maria J. Redondo
- Baylor College of Medicine, Texas Children’s Hospital, Houston, TX
| | - Emily K. Sims
- Department of Pediatrics, Indiana University, Indianapolis, IN
| | - Diane K. Wherrett
- Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Antoinette Moran
- Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Alberto Pugliese
- Department of Diabetes Immunology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA
| | - Peter A. Gottlieb
- Barbara Davis Center, University of Colorado School of Medicine, Aurora, CO
| | - Jay M. Sosenko
- Division of Endocrinology, University of Miami, Miami, FL
| | - Heba M. Ismail
- Baylor College of Medicine, Texas Children’s Hospital, Houston, TX
| | | |
Collapse
|
11
|
Singh H, Nim DK, Randhawa AS, Ahluwalia S. Integrating clinical pharmacology and artificial intelligence: potential benefits, challenges, and role of clinical pharmacologists. Expert Rev Clin Pharmacol 2024; 17:381-391. [PMID: 38340012 DOI: 10.1080/17512433.2024.2317963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/08/2024] [Indexed: 02/12/2024]
Abstract
INTRODUCTION The integration of artificial intelligence (AI) into clinical pharmacology could be a potential approach for accelerating drug discovery and development, improving patient care, and streamlining medical research processes. AREAS COVERED We reviewed the current state of AI applications in clinical pharmacology, focusing on drug discovery and development, precision medicine, pharmacovigilance, and other ventures. Key AI applications in clinical pharmacology are examined, including machine learning, natural language processing, deep learning, and reinforcement learning etc. Additionally, the evolving role of clinical pharmacologists, ethical considerations, and challenges in implementing AI in clinical pharmacology are discussed. EXPERT OPINION The AI could be instrumental in accelerating drug discovery, predicting drug safety and efficacy, and optimizing clinical trial designs. It can play a vital role in precision medicine by helping in personalized drug dosing, treatment selection, and predicting drug response based on genetic, clinical, and environmental factors. The role of AI in pharmacovigilance, such as signal detection and adverse event prediction, is also promising. The collaboration between clinical pharmacologists and AI experts also poses certain ethical and practical challenges. Clinical pharmacologists can be instrumental in shaping the future of AI-driven clinical pharmacology and contribute to the improvement of healthcare systems.
Collapse
Affiliation(s)
- Harmanjit Singh
- Department of Pharmacology, Government Medical College & Hospital, Chandigarh, India
| | | | | | | |
Collapse
|
12
|
Ren F, Aliper A, Chen J, Zhao H, Rao S, Kuppe C, Ozerov IV, Zhang M, Witte K, Kruse C, Aladinskiy V, Ivanenkov Y, Polykovskiy D, Fu Y, Babin E, Qiao J, Liang X, Mou Z, Wang H, Pun FW, Torres-Ayuso P, Veviorskiy A, Song D, Liu S, Zhang B, Naumov V, Ding X, Kukharenko A, Izumchenko E, Zhavoronkov A. A small-molecule TNIK inhibitor targets fibrosis in preclinical and clinical models. Nat Biotechnol 2024:10.1038/s41587-024-02143-0. [PMID: 38459338 DOI: 10.1038/s41587-024-02143-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 01/16/2024] [Indexed: 03/10/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an aggressive interstitial lung disease with a high mortality rate. Putative drug targets in IPF have failed to translate into effective therapies at the clinical level. We identify TRAF2- and NCK-interacting kinase (TNIK) as an anti-fibrotic target using a predictive artificial intelligence (AI) approach. Using AI-driven methodology, we generated INS018_055, a small-molecule TNIK inhibitor, which exhibits desirable drug-like properties and anti-fibrotic activity across different organs in vivo through oral, inhaled or topical administration. INS018_055 possesses anti-inflammatory effects in addition to its anti-fibrotic profile, validated in multiple in vivo studies. Its safety and tolerability as well as pharmacokinetics were validated in a randomized, double-blinded, placebo-controlled phase I clinical trial (NCT05154240) involving 78 healthy participants. A separate phase I trial in China, CTR20221542, also demonstrated comparable safety and pharmacokinetic profiles. This work was completed in roughly 18 months from target discovery to preclinical candidate nomination and demonstrates the capabilities of our generative AI-driven drug-discovery pipeline.
Collapse
Affiliation(s)
- Feng Ren
- Insilico Medicine Shanghai Ltd., Shanghai, China
- Insilico Medicine AI Limited, Abu Dhabi, UAE
| | - Alex Aliper
- Insilico Medicine AI Limited, Abu Dhabi, UAE
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Jian Chen
- Department of Clinical Pharmacology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Heng Zhao
- Insilico Medicine Shanghai Ltd., Shanghai, China
| | - Sujata Rao
- Insilico Medicine US Inc., New York, NY, USA
| | - Christoph Kuppe
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
- Department of Nephrology, University Clinic RWTH Aachen, Aachen, Germany
| | - Ivan V Ozerov
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Man Zhang
- Insilico Medicine Shanghai Ltd., Shanghai, China
| | - Klaus Witte
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Chris Kruse
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong SAR, China
| | | | - Yan Ivanenkov
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong SAR, China
| | | | - Yanyun Fu
- Insilico Medicine Shanghai Ltd., Shanghai, China
| | | | - Junwen Qiao
- Insilico Medicine Shanghai Ltd., Shanghai, China
| | - Xing Liang
- Insilico Medicine Shanghai Ltd., Shanghai, China
| | - Zhenzhen Mou
- Insilico Medicine Shanghai Ltd., Shanghai, China
| | - Hui Wang
- Insilico Medicine Shanghai Ltd., Shanghai, China
| | - Frank W Pun
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Pedro Torres-Ayuso
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, PA, USA
| | | | - Dandan Song
- Department of Clinical Pharmacology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Sang Liu
- Insilico Medicine Shanghai Ltd., Shanghai, China
| | - Bei Zhang
- Insilico Medicine Shanghai Ltd., Shanghai, China
| | - Vladimir Naumov
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Xiaoqiang Ding
- Division of Nephrology, Zhongshan Hospital Shanghai Medical College, Fudan University, Shanghai, China
| | - Andrey Kukharenko
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Evgeny Izumchenko
- Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Alex Zhavoronkov
- Insilico Medicine AI Limited, Abu Dhabi, UAE.
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong SAR, China.
- Insilico Medicine US Inc., New York, NY, USA.
- Insilico Medicine Canada Inc, Montreal, Quebec, Canada.
| |
Collapse
|
13
|
Gayathiri E, Prakash P, Kumaravel P, Jayaprakash J, Ragunathan MG, Sankar S, Pandiaraj S, Thirumalaivasan N, Thiruvengadam M, Govindasamy R. Computational approaches for modeling and structural design of biological systems: A comprehensive review. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 185:17-32. [PMID: 37821048 DOI: 10.1016/j.pbiomolbio.2023.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 08/14/2023] [Accepted: 08/27/2023] [Indexed: 10/13/2023]
Abstract
The convergence of biology and computational science has ushered in a revolutionary era, revolutionizing our understanding of biological systems and providing novel solutions to global problems. The field of genetic engineering has facilitated the manipulation of genetic codes, thus providing opportunities for the advancement of innovative disease therapies and environmental enhancements. The emergence of bio-molecular simulation represents a significant advancement in this particular field, as it offers the ability to gain microscopic insights into molecular-level biological processes over extended periods. Biomolecular simulation plays a crucial role in advancing our comprehension of organismal mechanisms by establishing connections between molecular structures, interactions, and biological functions. The field of computational biology has demonstrated its significance in deciphering intricate biological enigmas through the utilization of mathematical models and algorithms. The process of decoding the human genome has resulted in the advancement of therapies for a wide range of genetic disorders, while the simulation of biological systems contributes to the identification of novel pharmaceutical compounds. The potential of biomolecular simulation and computational biology is vast and limitless. As the exploration of the underlying principles that govern living organisms progresses, the potential impact of this understanding on cancer treatment, environmental restoration, and other domains is anticipated to be transformative. This review examines the notable advancements achieved in the field of computational biology, emphasizing its potential to revolutionize the comprehension and enhancement of biological systems.
Collapse
Affiliation(s)
- Ekambaram Gayathiri
- Department of Plant Biology and Plant Biotechnology, Guru Nanak College (Autonomous), Chennai, 42, Tamil Nadu, India
| | - Palanisamy Prakash
- Department of Botany, Periyar University, Periyar Palkalai Nagar, Salem, 636011, Tamil Nadu, India
| | - Priya Kumaravel
- Department of Biotechnology, St. Joseph College (Arts & Science), Kovur, Chennai, Tamil Nadu, India
| | - Jayanthi Jayaprakash
- Department of Advanced Zoology and Biotechnology, Guru Nanak College, Chennai, Tamil Nadu, India
| | | | - Sharmila Sankar
- Department of Advanced Zoology and Biotechnology, Guru Nanak College, Chennai, Tamil Nadu, India
| | - Saravanan Pandiaraj
- Department of Self-Development Skills, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Natesan Thirumalaivasan
- Department of Periodontics, Saveetha Dental College, and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMTAS), Chennai, 600077, Tamil Nadu, India
| | - Muthu Thiruvengadam
- Department of Applied Bioscience, College of Life and Environmental Sciences, Konkuk University, Seoul, 05029, South Korea
| | - Rajakumar Govindasamy
- Department of Orthodontics, Saveetha Dental College and Hospitals, Saveetha University, Chennai, India.
| |
Collapse
|