1
|
Pike CM, Levi JA, Boone LA, Peddibhotla S, Johnson J, Zwarycz B, Bunger MK, Thelin W, Boazak EM. High-throughput assay for predicting diarrhea risk using a 2D human intestinal stem cell-derived model. Toxicol In Vitro 2025; 106:106040. [PMID: 40086646 DOI: 10.1016/j.tiv.2025.106040] [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: 10/09/2024] [Revised: 01/29/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Gastrointestinal toxicities (GITs) in clinical trials often lead to dose-limitations that reduce drug efficacy and delay treatment optimization. Preclinical animal models do not accurately replicate human physiology, leaving few options for early detection of GITs, such as diarrhea, before human studies. Chemotherapeutic agents, known to cause clinical diarrhea, frequently target mitotic cells. Therefore, we hypothesized a model utilizing proliferative cell populations derived from human intestinal crypts would predict clinical diarrhea occurrence with high accuracy. Here, we describe the development of a diarrhea prediction assay utilizing RepliGut® Planar, a primary intestinal stem cell-derived platform. To evaluate the ability of this model to predict clinical diarrhea risk, we assessed toxicity of 30 marketed drugs by measuring cell proliferation (EdU incorporation), cell abundance (nuclei quantification), and barrier formation (TEER) in cells derived from three human donors. Dose response curves were generated for each drug, and the IC15 to Cmax ratio was used to identify a threshold for assay positivity. This model accurately predicted diarrhea potential, achieving an accuracy of 91 % for proliferation, 90 % for abundance, and 88 % for barrier formation. In vitro toxicity screening using primary proliferative cells may reduce clinical diarrhea and ultimately lead to safer and more effective treatments for patients.
Collapse
|
2
|
Lee CJ, Nam Y, Rim YA, Ju JH. Advanced Animal Replacement Testing Strategies Using Stem Cell and Organoids. Int J Stem Cells 2025; 18:107-125. [PMID: 40064522 PMCID: PMC12122249 DOI: 10.15283/ijsc24118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 06/02/2025] Open
Abstract
The increasing ethical concerns and regulatory restrictions surrounding animal testing have accelerated the development of advanced in vitro models that more accurately replicate human physiology. Among these, stem cell-based systems and organoids have emerged as revolutionary tools, providing ethical, scalable, and physiologically relevant alternatives. This review explores the key trends and driving factors behind the adoption of these models, such as technological advancements, the principles of the 3Rs (Replacement, Reduction, and Refinement), and growing regulatory support from agencies like the OECD and FDA. It also delves into the development and application of various model systems, including 3D reconstructed tissues, induced pluripotent stem cell-derived cells, and microphysiological systems, highlighting their potential to replace animal models in toxicity evaluation, disease modeling, and drug development. A critical aspect of implementing these models is ensuring robust quality control protocols to enhance reproducibility and standardization, which is necessary for gaining regulatory acceptance. Additionally, we discuss advanced strategies for assessing toxicity and efficacy, focusing on organ-specific evaluation methods and applications in diverse fields such as pharmaceuticals, cosmetics, and food safety. Despite existing challenges related to scalability, standardization, and regulatory alignment, these innovative models represent a transformative step towards reducing animal use and improving the relevance and reliability of preclinical testing outcomes.
Collapse
Affiliation(s)
- Chang-Jin Lee
- Department of Biomedical Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Catholic iPSC Research Center, CiSTEM Laboratory, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Yipscell Inc, Seoul, Korea
| | - Yoojun Nam
- Yipscell Inc, Seoul, Korea
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, Korea
| | - Yeri Alice Rim
- Department of Biomedical Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Catholic iPSC Research Center, CiSTEM Laboratory, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Division of Rheumatology, Department of Internal Medicine, Institute of Medical Science, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ji Hyeon Ju
- Catholic iPSC Research Center, CiSTEM Laboratory, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Yipscell Inc, Seoul, Korea
- Division of Rheumatology, Department of Internal Medicine, Institute of Medical Science, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
3
|
Zheng S, Zhou Z, Ji J, Liu Y, Jiao X, Li X, Shen Y, Hong H, Han X. M2 macrophage-targeted metal-polyphenol networks (MPNs) for OPN siRNA delivery and idiopathic pulmonary fibrosis therapy. J Control Release 2025; 383:113862. [PMID: 40383161 DOI: 10.1016/j.jconrel.2025.113862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 05/11/2025] [Accepted: 05/16/2025] [Indexed: 05/20/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) exhibits extremely high mortality rates. Targeted therapy, which utilizes specific drugs or other substances to identify and attack specific molecular targets in the lesion, holds promise as a potent means of treating IPF. M2 macrophages have been shown to express high levels of osteopontin (OPN) early in the onset of IPF and sustain this high expression to promote the progression of IPF. Intervention in OPN expression can effectively impede the development of fibrosis. While the technology for targeting proteins with siRNA has become increasingly mature, the targeted delivery of siRNA to resident M2 macrophages in the lungs remains challenging. In this study, we developed an engineered self-assembling OPN siRNA carrier complex based on a metal-polyphenol network (luteolin-Zr) and PEG conjugated with an M2 macrophage-targeting peptide (Pery-PEG-M2), termed siOPN@LuZ-M2, for the treatment of pulmonary fibrosis. Consequently, significant therapeutic effects were observed in both bleomycin-induced pulmonary fibrosis mouse models and human precision-cut lung slices (hPCLS) models. Importantly, luteolin, which is slowly released from siOPN@LuZ-M2 within cells, can gradually accumulate in fibrotic lung tissue, exerting an anti-inflammatory effect and further enhancing the treatment of IPF. It is worth mentioning that siOPN@LuZ-M2 can be labeled with 89Zr, allowing for the detection of its in vivo distribution and metabolic behavior via PET-CT. This study presents a promising new image-guided molecular targeting strategy for the treatment of fibrosis.
Collapse
Affiliation(s)
- Shudan Zheng
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Zhenghao Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Medical School of Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Jie Ji
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Yuxin Liu
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Xiaodan Jiao
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Medical School of Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Xiaoyang Li
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Medical School of Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Yi Shen
- Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Hao Hong
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Medical School of Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu 210093, China.
| | - Xiaodong Han
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu 210093, China.
| |
Collapse
|
4
|
Rayasingh AR, Manivannan V. Palladium(II) and platinum(II) complexes of disubstituted imidazo[1,5- a]pyridine and imidazolylpyridine: coordination chemistry, versatile catalysis, and biophysical study. Dalton Trans 2025; 54:7741-7752. [PMID: 40259763 DOI: 10.1039/d5dt00346f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
Pincer-type mono- and poly-nuclear Pd(II) and Pt(II) complexes bearing imidazo[1,5-a]pyridine and imidazolylpyridine moieties were synthesized and characterized using several spectroscopic methods. Determination of molecular structures of these complexes using single crystal X-ray diffraction studies revealed a distorted square planar geometry around the bivalent palladium and platinum in all the complexes. These Pd(II) complexes displayed high catalytic activity in various reactions, such as the Suzuki-Miyaura cross-coupling reaction, transfer hydrogenation reaction, and alkyne homocoupling. The experimental results matched well with the theoretical data of all catalysts. Substantial deviations in the catalytic activity were observed by changing the co-ligand, binding mode of the ligand and the number of metal centres. Under optimal conditions, the Suzuki cross-coupling and transfer hydrogenation reactions were successfully accomplished with a wide range of functional groups by taking only 0.1 mol% of tetranuclear Pd(II) complex (5) as the catalyst. Intermediates in the Suzuki coupling reaction were also detected using mass spectroscopy. Among the studied complexes, the tetranuclear palladium complex exhibited the highest catalytic activity. Further, Pd(II) complexes were tested in a model reaction of the homocoupling of phenylacetylene, and diphenylbutadiyne was produced in excellent yield. Additionally, the interactions of all the complexes with calf thymus DNA (CT-DNA) and bovine serum albumin (BSA) were investigated using electronic spectroscopy. Absorption study showed minor groove binding of DNA with these complexes, while intercalative binding through displacement of ethidium bromide (EB) in EB-DNA was observed in all the complexes, quenching the fluorescence intensity. The complexes also displayed high binding affinity toward BSA, as confirmed by emission, synchronous fluorescence, and steady-state fluorescence anisotropy measurements. Moreover, the pharmacokinetic properties of two bioactive compounds (3s and 3t) obtained from the Suzuki coupling reaction were calculated, and to evaluate their activity as leukotriene A4 hydrolase (LTA4H) inhibitor, these molecules were docked with human LTA4H enzyme.
Collapse
Affiliation(s)
| | - Vadivelu Manivannan
- Department of Chemistry, Indian Institute of Technology, Guwahati, Assam 781039, India.
| |
Collapse
|
5
|
Koutroumpa NM, Tsoumanis A, Sarimveis H, Lynch I, Melagraki G, Afantitis A. Prediction of blood-brain barrier and Caco-2 permeability through the Enalos Cloud Platform: combining contrastive learning and atom-attention message passing neural networks. J Cheminform 2025; 17:68. [PMID: 40325398 PMCID: PMC12051285 DOI: 10.1186/s13321-025-01007-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 03/30/2025] [Indexed: 05/07/2025] Open
Abstract
In this study, we introduce a novel approach for predicting two key drug properties, blood-brain barrier (BBB) permeability and human intestinal absorption via Caco-2 permeability. Our methodology centers around a specialized neural network, the atom transformer-based Message Passing Neural Network (MPNN), which we have combined with contrastive learning techniques to enhance the process of representing and embedding molecular structures for more accurate property prediction. These innovative models focus on predicting BBB and Caco-2 permeability -two critical factors in drug absorption and distribution- which fall under the broader scope of ADMET (absorption, distribution, metabolism, excretion, and toxicity) properties. The models are readily accessible online through the Enalos Cloud Platform which offers a user-friendly, AI-powered, ready-to-use web service that significantly streamlines the drug design process, enabling users to easily predict and understand the behavior of potential drug compounds within the human body.Scientific Contribution Our study combines an atom-attention Message Passing Neural Network (AA-MPNN) with contrastive learning (CL), which significantly improves predictive accuracy. Our model leverages self-supervised learning to expand the chemical space used in training and self-attention mechanisms to focus on critical molecular features, enhancing both model accuracy and interpretability. Additionally, the ready-to-use web service based on our model democratizes access to predictive tools for the scientific and regulatory communities.
Collapse
Affiliation(s)
- Nikoletta-Maria Koutroumpa
- NovaMechanics Ltd, 1070, Nicosia, Cyprus
- School of Chemical Engineering, National Technical University of Athens, 157 80, Athens, Greece
- Entelos Institute, 6059, Larnaca, Cyprus
| | | | - Haralambos Sarimveis
- School of Chemical Engineering, National Technical University of Athens, 157 80, Athens, Greece
| | - Iseult Lynch
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Georgia Melagraki
- Division of Physical Sciences & Applications, Hellenic Military Academy, 166 73, Vari, Greece
| | - Antreas Afantitis
- NovaMechanics Ltd, 1070, Nicosia, Cyprus.
- Entelos Institute, 6059, Larnaca, Cyprus.
- NovaMechanics MIKE, 185 45, Piraeus, Greece.
| |
Collapse
|
6
|
Yang X, Artibani M, Jin Y, Aggarwal A, Zhang Y, Muñoz‐Galvan S, Mikhailova E, Rai L, Mukherjee N, Kumar RK, Albukhari A, Ma S, Zhou L, Ahmed AA, Bayley H. 3D Microtumors Representing Ovarian Cancer Minimal Residual Disease Respond to the Fatty Acid Oxidation Inhibitor Perhexiline. Adv Healthc Mater 2025; 14:e2404072. [PMID: 39924751 PMCID: PMC12118330 DOI: 10.1002/adhm.202404072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 01/13/2025] [Indexed: 02/11/2025]
Abstract
The poor survival of ovarian cancer patients is linked to their high likelihood of relapse. In spite of full apparent macroscopic clearance, tumor recurrences arise from cells that are resistant to primary chemotherapy in the form of minimal residual disease (MRD). MRD exhibits distinct molecular drivers from bulk cancer and therefore necessitates alternative therapeutic strategies. However, there is a lack of 3D models that faithfully recapitulate MRD ex vivo for therapy development. This study constructs microfluidics-based 3D microtumors to generate a clinically-relevant model for ovarian cancer MRD. The microtumors recapitulate the non-genetic heterogeneity of ovarian cancer, capturing the "Oxford Classic" five molecular signatures. Gene expression in the 3D microtumors aligns closely with MRD from ovarian cancer patients and features the upregulation of fatty acid metabolism genes. Finally, the MRD 3D microtumors respond to the approved fatty acid oxidation inhibitor, perhexiline, demonstrating their utility in drug discovery. This system might be used as a drug-testing platform for the discovery of novel MRD-specific therapies in ovarian cancer.
Collapse
Affiliation(s)
- Xingyun Yang
- Department of ChemistryUniversity of OxfordOxfordOX1 3TAUK
| | - Mara Artibani
- Ovarian Cancer Cell LaboratoryMRC Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordOX3 9DSUK
- Nuffield Department of Women's & Reproductive HealthUniversity of OxfordOxfordOX3 9DUUK
| | - Yongcheng Jin
- Department of ChemistryUniversity of OxfordOxfordOX1 3TAUK
| | - Aneesh Aggarwal
- Ovarian Cancer Cell LaboratoryMRC Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordOX3 9DSUK
- Nuffield Department of Women's & Reproductive HealthUniversity of OxfordOxfordOX3 9DUUK
| | - Yujia Zhang
- Department of ChemistryUniversity of OxfordOxfordOX1 3TAUK
- Institute of Electrical and MicroengineeringÉcole Polytechnique Fédérale de LausanneLausanne1015Switzerland
| | - Sandra Muñoz‐Galvan
- Ovarian Cancer Cell LaboratoryMRC Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordOX3 9DSUK
- Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaAvda Manuel SiurotSeville41013Spain
| | | | - Lena Rai
- Ovarian Cancer Cell LaboratoryMRC Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordOX3 9DSUK
- Nuffield Department of Women's & Reproductive HealthUniversity of OxfordOxfordOX3 9DUUK
| | | | - Ravinash Krishna Kumar
- Department of ChemistryUniversity of OxfordOxfordOX1 3TAUK
- Department of Infectious DiseaseImperial College LondonSouth KensingtonLondonSW7 2AZUK
| | - Ashwag Albukhari
- Biochemistry DepartmentFaculty of ScienceKing Abdulaziz UniversityJeddah21589Saudi Arabia
| | - Shaohua Ma
- Tsinghua Shenzhen International Graduate School (SIGS)Tsinghua UniversityShenzhen518055China
- Tsinghua‐Berkeley Shenzhen Institute (TBSI)Tsinghua UniversityShenzhen518055China
| | - Linna Zhou
- Department of ChemistryUniversity of OxfordOxfordOX1 3TAUK
- Ludwig Institute for Cancer ResearchNuffield Department of MedicineUniversity of OxfordOxfordOX3 7DQUK
| | - Ahmed Ashour Ahmed
- Ovarian Cancer Cell LaboratoryMRC Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordOX3 9DSUK
- Nuffield Department of Women's & Reproductive HealthUniversity of OxfordOxfordOX3 9DUUK
| | - Hagan Bayley
- Department of ChemistryUniversity of OxfordOxfordOX1 3TAUK
| |
Collapse
|
7
|
Li Y, Lai D, Li R, Chen H, Huang X, Ning J. A Biomarker Signature-Guided Clinical Trial Design for Precision Medicine. Stat Med 2025; 44:e70103. [PMID: 40405471 DOI: 10.1002/sim.70103] [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: 10/08/2024] [Revised: 03/16/2025] [Accepted: 04/10/2025] [Indexed: 05/24/2025]
Abstract
Targeted cancer therapies aim to effectively treat patients with specific biomarker profiles. Nevertheless, these therapies may not always precisely hit their intended targets, leading to uncertainty about the specific subset of patients who will benefit. To address this uncertainty, the identification of sensitive patient subsets in clinical trials becomes crucial. Our proposed phase IIB/III clinical trial design seeks to pinpoint a biomarker signature with precision, ensuring the accurate identification of patients who will respond to a specific treatment. This approach allows for the selective enrollment of sensitive patients to maximize benefits for trial participants. We incorporate Bayesian methodology to facilitate response-adaptive randomization, enhancing the likelihood that each participant receives his/her optimal treatment. Furthermore, our design uses inverse-probability-of-treatment-weighted analysis to avoid selection bias and control for the type I error rate. The evaluation of this trial design is based on four criteria: the statistical power, response rate of all patients participating in the current trial, their individual loss, and probabilities of receiving their optimal treatment for both current trial participants and future patients. Simulations demonstrate the proposed design's potential for maximizing trial participants' benefits with little sacrifice on statistical power. Its key advantages include an improved overall response rate within the trial and a higher percentage of patients receiving the optimal treatment.
Collapse
Affiliation(s)
- Yuan Li
- Department of Biostatistics and Data Science, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Biometrics, Supernus Pharmaceuticals, Inc., Rockville, MD, USA
| | - Dejian Lai
- Department of Biostatistics and Data Science, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ruosha Li
- Department of Biostatistics and Data Science, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Han Chen
- Department of Epidemiology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xuelin Huang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Ning
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
8
|
Hanitrarimalala V, Prgomet Z, Hedhammar M, Tassidis H, Wingren AG. In vitro 3D modeling of colorectal cancer: the pivotal role of the extracellular matrix, stroma and immune modulation. Front Genet 2025; 16:1545017. [PMID: 40376304 PMCID: PMC12078225 DOI: 10.3389/fgene.2025.1545017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 04/23/2025] [Indexed: 05/18/2025] Open
Abstract
Colorectal cancer (CRC) is a leading global cancer with high mortality, especially in metastatic cases, with limited therapeutic options. The tumor microenvironment (TME), a network comprising various immune cells, stromal cells and extracellular (ECM) components plays a crucial role in influencing tumor progression and therapy outcome. The genetic heterogeneity of CRC and the complex TME complicates the development of effective, personalized treatment strategies. The prognosis has slowly improved during the past decades, but metastatic CRC (mCRC) is common among patients and is still associated with low survival. The therapeutic options for CRC differ from those for mCRC and include surgery (mostly for CRC), chemotherapy, growth factor receptor signaling pathway targeting, as well as immunotherapy. Malignant CRC cells are established in the TME, which varies depending on the primary or metastatic site. Herein, we review the role and interactions of several ECM components in 3D models of CRC and mCRC tumor cells, with an emphasis on how the TME affects tumor growth and treatment. This comprehensive summary provides support for the development of 3D models that mimic the interactions within the TME, which will be essential for the development of novel anticancer therapies.
Collapse
Affiliation(s)
- Veroniaina Hanitrarimalala
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
- Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - Zdenka Prgomet
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
- Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - My Hedhammar
- KTH Royal Institute of Technology, Division of Protein Technology, Stockholm, Sweden
| | - Helena Tassidis
- Department of Bioanalysis, Faculty of Natural Sciences, Kristianstad University, Kristianstad, Sweden
| | - Anette Gjörloff Wingren
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
- Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden
- Department of Bioanalysis, Faculty of Natural Sciences, Kristianstad University, Kristianstad, Sweden
| |
Collapse
|
9
|
Garg A, Ramamurthi N, Das SS. Addressing Imbalanced Classification Problems in Drug Discovery and Development Using Random Forest, Support Vector Machine, AutoGluon-Tabular, and H2O AutoML. J Chem Inf Model 2025; 65:3976-3989. [PMID: 40230275 DOI: 10.1021/acs.jcim.5c00023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
The classification models built on class imbalanced data sets tend to prioritize the accuracy of the majority class, and thus, the minority class generally has a higher misclassification rate. Different techniques are available to address the class imbalance in classification models and can be categorized as data-level, algorithm-level, and hybrid methods. But to the best of our knowledge, an in-depth analysis of the performance of these techniques against the class ratio is not available in the literature. We have addressed these shortcomings in this study and have performed a detailed analysis of the performance of four different techniques to address imbalanced class distribution using machine learning (ML) methods and AutoML tools. To carry out our study, we have selected four such techniques─(a) threshold optimization using (i) GHOST and (ii) the area under the precision-recall curve (AUPR) curve, (b) internal balancing method of AutoML and class-weight of machine learning methods, and (c) data balancing using SMOTETomek─and generated 27 data sets considering nine different class ratios (i.e., the ratio of the positive class and total samples) from three data sets that belong to the drug discovery and development field. We have employed random forest (RF) and support vector machine (SVM) as representatives of ML classifier and AutoGluon-Tabular (version 0.6.1) and H2O AutoML (version 3.40.0.4) as representatives of AutoML tools. The important findings of our studies are as follows: (i) there is no effect of threshold optimization on ranking metrics such as AUC and AUPR, but AUC and AUPR get affected by class-weighting and SMOTTomek; (ii) for ML methods RF and SVM, significant percentage improvement up to 375, 33.33, and 450 over all the data sets can be achieved, respectively, for F1 score, MCC, and balanced accuracy, which are suitable for performance evaluation of imbalanced data sets; (iii) for AutoML libraries AutoGluon-Tabular and H2O AutoML, significant percentage improvement up to 383.33, 37.25, and 533.33 over all the data sets can be achieved, respectively, for F1 score, MCC, and balanced accuracy; (iv) the general pattern of percentage improvement in balanced accuracy is that the percentage improvement increases when the class ratio is systematically decreased from 0.5 to 0.1; in the case of F1 score and MCC, maximum improvement is achieved at the class ratio of 0.3; (v) for both ML and AutoML with balancing, it is observed that any individual class-balancing technique does not outperform all other methods on a significantly higher number of data sets based on F1 score; (vi) the three external balancing techniques combined outperformed the internal balancing methods of the ML and AutoML; (vii) AutoML tools perform as good as the ML models and in some cases perform even better for handling imbalanced classification when applied with imbalance handling techniques. In summary, exploration of multiple data balancing techniques is recommended for classifying imbalanced data sets to achieve optimal performance as neither of the external techniques nor the internal techniques outperform others significantly. The results are specific to the ML methods and AutoML libraries used in this study, and for generalization, a study can be carried out considering a sizable number of ML methods and AutoML libraries.
Collapse
Affiliation(s)
- Ayush Garg
- TCS Research (Life Sciences Division), Tata Consultancy Services Limited, Noida 201303, India
| | - Narayanan Ramamurthi
- TCS Research (Life Sciences Division), Tata Consultancy Services Limited, Chennai 600113, India
| | - Shyam Sundar Das
- TCS Research (Life Sciences Division), Tata Consultancy Services Limited, Kolkata 700160, India
| |
Collapse
|
10
|
Qu Y, Li T, Liu Z, Tong W, Li D. DICTrank Is a Reliable Dataset for Cardiotoxicity Prediction Using Machine Learning Methods. Chem Res Toxicol 2025; 38:647-655. [PMID: 40146530 DOI: 10.1021/acs.chemrestox.4c00428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Drug-induced cardiotoxicity (DICT) is a significant challenge in drug development and public health. DICT can arise from various mechanisms; New Approach Methods (NAMs), including quantitative structure-activity relationships (QSARs), have been extensively developed to predict DICT based solely on individual mechanisms (e.g., hERG-related cardiotoxicity) due to the availability of datasets limited to specific mechanisms. While these efforts have significantly contributed to our understanding of cardiotoxicity, DICT assessment remains challenging, suggesting that approaches focusing on isolated mechanisms may not provide a comprehensive evaluation. To address this, we previously developed DICTrank, the largest dataset for assessing overall cardiotoxicity liability in humans based on FDA drug labels. In this study, we evaluated the utility of DICTrank for QSAR modeling using five machine learning methods─Logistic Regression (LR), K-Nearest Neighbors, Support Vector Machines, Random Forest (RF), and extreme gradient boosting (XGBoost)─which vary in algorithmic complexity and explainability. To reflect real-world scenarios, models were trained on drugs approved before and within 2005 to predict the DICT risk of those approved thereafter. While we observed no clear association between prediction performance and model complexity, LR and XGBoost achieved the best results with DICTrank. Additionally, our significant-feature analyses with RF and XGBoost models provided novel insights into DICT mechanisms, revealing that drug properties associated with descriptors such as "structural and topological", "polarizability", and "electronegativity" contributed significantly to DICT. Moreover, we found that model performance varied by therapeutic category, suggesting the need to tailor models accordingly. In conclusion, our study demonstrated the robustness and reliability of DICTrank for cardiotoxicity prediction in humans using machine learning methods.
Collapse
Affiliation(s)
- Yanyan Qu
- US Food and Drug Administration, National Center for Toxicological Research, Jefferson, Arkansas 72079, United States
- University of Arkansas at Little Rock and University of Arkansas for Medical Sciences Joint Bioinformatics Program, Little Rock, Arkansas 72204, United States
| | - Ting Li
- US Food and Drug Administration, National Center for Toxicological Research, Jefferson, Arkansas 72079, United States
| | - Zhichao Liu
- Integrative Toxicology, Nonclinical Drug Safety, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Weida Tong
- US Food and Drug Administration, National Center for Toxicological Research, Jefferson, Arkansas 72079, United States
| | - Dongying Li
- US Food and Drug Administration, National Center for Toxicological Research, Jefferson, Arkansas 72079, United States
| |
Collapse
|
11
|
Evangelista D, Nelson E, Skyner R, Tehan B, Bernetti M, Roberti M, Bolognesi ML, Bottegoni G. Application of Deep Learning to Predict the Persistence, Bioaccumulation, and Toxicity of Pharmaceuticals. J Chem Inf Model 2025; 65:3248-3261. [PMID: 40178174 PMCID: PMC12004513 DOI: 10.1021/acs.jcim.4c02293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/14/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
This study investigates the application of a deep learning (DL) model, specifically a message-passing neural network (MPNN) implemented through Chemprop, to predict the persistence, bioaccumulation, and toxicity (PBT) characteristics of compounds, with a focus on pharmaceuticals. We employed a clustering strategy to provide a fair assessment of the model performances. By applying the generated model to a set of pharmaceutically relevant molecules, we aim to highlight potential PBT chemicals and extract PBT-relevant substructures. These substructures can serve as structural flags, alerting drug designers to potential environmental issues from the earliest stages of the drug discovery process. Incorporating these findings into pharmaceutical development workflows is expected to drive significant advancements in creating more environmentally friendly drug candidates while preserving their therapeutic efficacy.
Collapse
Affiliation(s)
- Dominga Evangelista
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum—University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Elliot Nelson
- OMass
Therapeutics, Building 4000, Chancellor Court, John Smith Dr, Oxford Business Park,
ARC, Oxford OX4 2GX, United Kingdom
| | - Rachael Skyner
- OMass
Therapeutics, Building 4000, Chancellor Court, John Smith Dr, Oxford Business Park,
ARC, Oxford OX4 2GX, United Kingdom
| | - Ben Tehan
- OMass
Therapeutics, Building 4000, Chancellor Court, John Smith Dr, Oxford Business Park,
ARC, Oxford OX4 2GX, United Kingdom
| | - Mattia Bernetti
- Department
of Biomolecular Sciences, University of
Urbino, Urbino 60129, Italy
- Computational
and Chemical Biology, Fondazione Istituto
Italiano di Tecnologia, Via Morego 30, Genova 16163, Italy
| | - Marinella Roberti
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum—University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Maria Laura Bolognesi
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum—University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Giovanni Bottegoni
- Department
of Biomolecular Sciences, University of
Urbino, Urbino 60129, Italy
- Department
of Pharmacy, University of Birmingham, Edgbaston B15 2TT, Birmingham, United
Kingdom
| |
Collapse
|
12
|
Varela MT, Dias GG, de Oliveira LFN, de Oliveira RG, Aguiar FD, Nogueira JP, Cruz LR, Dias LC. Spirocyclic compounds as innovative tools in drug discovery for medicinal chemists. Eur J Med Chem 2025; 287:117368. [PMID: 39952099 DOI: 10.1016/j.ejmech.2025.117368] [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: 11/29/2024] [Revised: 01/17/2025] [Accepted: 01/26/2025] [Indexed: 02/17/2025]
Abstract
The occurrence of spirocyclic motifs in clinical candidates and approved drugs is on the rise. This is related to the improvement of drug-like properties that can be achieved by introducing this sp3-rich system into bioactive compounds. Given the increasing number of synthetic methodologies and building blocks available, spirocycles are becoming widely accessible to medicinal chemists. From restricting conformation to induce a better fit with the target, to modulation of physicochemical and pharmacokinetic properties, spirocycles are being used to address several challenges in drug discovery. This review covers general aspects of the chemistry of spirocycles, highlighting some key strategies for their preparation. As reported in publications over the past five years, we demonstrate that, beyond the exploration of structure-activity relationships (SAR) in medicinal chemistry, the use of spirocycles is an attractive approach for enhancing properties such as potency, selectivity, physicochemistry, and pharmacokinetics.
Collapse
Affiliation(s)
- Marina T Varela
- Universidade Estadual de Campinas (UNICAMP), Instituto de Química, Rua Monteiro Lobato 270, 13083-862, Campinas, Brazil
| | - Gleiston G Dias
- Universidade Estadual de Campinas (UNICAMP), Instituto de Química, Rua Monteiro Lobato 270, 13083-862, Campinas, Brazil
| | - Luiz Fernando N de Oliveira
- Universidade Estadual de Campinas (UNICAMP), Instituto de Química, Rua Monteiro Lobato 270, 13083-862, Campinas, Brazil
| | - Ramon G de Oliveira
- Universidade Estadual de Campinas (UNICAMP), Instituto de Química, Rua Monteiro Lobato 270, 13083-862, Campinas, Brazil
| | - Francielle D Aguiar
- Universidade Estadual de Campinas (UNICAMP), Instituto de Química, Rua Monteiro Lobato 270, 13083-862, Campinas, Brazil
| | - João Pedro Nogueira
- Universidade Estadual de Campinas (UNICAMP), Instituto de Química, Rua Monteiro Lobato 270, 13083-862, Campinas, Brazil
| | - Luiza R Cruz
- Universidade Estadual de Campinas (UNICAMP), Instituto de Química, Rua Monteiro Lobato 270, 13083-862, Campinas, Brazil; Drugs for Neglected Diseases Initiative, Rua São José, 70, 20010-020, Rio de Janeiro, Brazil.
| | - Luiz C Dias
- Universidade Estadual de Campinas (UNICAMP), Instituto de Química, Rua Monteiro Lobato 270, 13083-862, Campinas, Brazil.
| |
Collapse
|
13
|
Goulooze SC, Muliaditan M, Franzese RC, Mantero A, Visser SAG, Melhem M, Post TM, Rathi C, Struemper H. Tutorial on Conditional Simulations With a Tumor Size-Overall Survival Model to Support Oncology Drug Development. CPT Pharmacometrics Syst Pharmacol 2025; 14:640-650. [PMID: 39985154 PMCID: PMC12001264 DOI: 10.1002/psp4.70003] [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: 09/18/2024] [Revised: 12/19/2024] [Accepted: 02/03/2025] [Indexed: 02/24/2025] Open
Abstract
The gold standard for regulatory approval in oncology is overall survival (OS). Because OS data are initially limited, early drug development decisions are often based on early efficacy endpoints, such as objective response rate and progression-free survival. Tumor size (TS)-OS models provide a framework to support decision-making on potential late-stage success based on early readouts, through leveraging TS data with limited follow-up and treatment-agnostic TS-OS link functions, to predict longer-term OS. Conditional simulations (also known as Bayesian forecasting) with TS-OS models can be used to simulate long-term OS outcomes for an ongoing study, conditional on the available TS and OS data at interim data cuts of the same study. This tutorial provides a comprehensive overview of the steps involved in using such conditional simulations to support better informed drug development decisions in oncology. The tutorial covers the selection of the TS-OS framework model; applying the TS-OS model to the interim data; performing conditional simulations; generating relevant output; as well as correct interpretation and communication of the output for decision making.
Collapse
Affiliation(s)
| | | | - Richard C. Franzese
- Clinical Pharmacology Modeling & SimulationGSKUpper ProvidencePennsylvaniaUSA
| | | | - Sandra A. G. Visser
- Clinical Pharmacology Modeling & SimulationGSKUpper ProvidencePennsylvaniaUSA
| | - Murad Melhem
- Clinical Pharmacology Modeling & SimulationGSKWalthamMassachusettsUSA
| | | | - Chetan Rathi
- Clinical Pharmacology Modeling & SimulationGSKWalthamMassachusettsUSA
| | - Herbert Struemper
- Clinical Pharmacology Modeling & SimulationGSKDurhamNorth CarolinaUSA
| |
Collapse
|
14
|
Shepherd J. Biomimetic Approaches in the Development of Optimised 3D Culture Environments for Drug Discovery in Cardiac Disease. Biomimetics (Basel) 2025; 10:204. [PMID: 40277603 PMCID: PMC12024959 DOI: 10.3390/biomimetics10040204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/09/2025] [Accepted: 03/21/2025] [Indexed: 04/26/2025] Open
Abstract
Cardiovascular disease remains the leading cause of death worldwide, yet despite massive investment in drug discovery, the progress of cardiovascular drugs from lab to clinic remains slow. It is a complex, costly pathway from drug discovery to the clinic and failure becomes more expensive as a drug progresses along this pathway. The focus has begun to shift to optimisation of in vitro culture methodologies, not only because these must be undertaken are earlier on in the drug discovery pathway, but also because the principles of the 3Rs have become embedded in national and international legislation and regulation. Numerous studies have shown myocyte cell behaviour to be much more physiologically relevant in 3D culture compared to 2D culture, highlighting the advantages of using 3D-based models, whether microfluidic or otherwise, for preclinical drug screening. This review aims to provide an overview of the challenges in cardiovascular drug discovery, the limitations of traditional routes, and the successes in the field of preclinical models for cardiovascular drug discovery. It focuses on the particular role biomimicry can play, but also the challenges around implementation within commercial drug discovery.
Collapse
Affiliation(s)
- Jenny Shepherd
- School of Engineering, University of Leicester, Leicester LE1 7RH, UK
| |
Collapse
|
15
|
Yang Y, Liu X, Xi D, Zhang Y, Gao X, Xu K, Liu H, Fang M. Precision Imaging of Biothiols in Live Cells and Treatment Evaluation during the Development of Liver Injury via a Near-Infrared Fluorescent Probe. CHEMICAL & BIOMEDICAL IMAGING 2025; 3:169-179. [PMID: 40151820 PMCID: PMC11937986 DOI: 10.1021/cbmi.4c00048] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 01/03/2025]
Abstract
In this study, a biothiol-sensitive near-infrared (NIR) fluorescent sensor, BDP-CYS, based on a coumarin-hemicyanine skeleton, was designed and developed based on thiol-halogen SNAr nucleophilic substitution. BDP-CYS was successfully implemented to ratiometrically monitor endogenous and exogenous Cys, Hcy, and GSH in living cells as well as to distinguish between normal and cancer cells. Furthermore, the probe was utilized to detect changes of biothiols in drug-induced hepatotoxicity and evaluate the treatment effectiveness of diabetes-associated liver injury in vivo. The advantages of BDP-CYS's Cys, Hcy, and GSH include practical sensitivity, high selectivity, rapidity of reaction, and stability across a range of pH and temperature conditions, thus introducing a new tool to better understand the roles of biothiols in oxidative stress.
Collapse
Affiliation(s)
- Yinshuang Yang
- School
of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Xiaolan Liu
- School
of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Deyang Xi
- Department
of Infectious Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Yibin Zhang
- College
of Chemistry and Chemical Engineering, Yangtze
Normal University, Fuling, Chongqing 408000, PR China
| | - Xiucai Gao
- Department
of Medical Imaging, The Affiliated No. 3
Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Kai Xu
- School
of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Haiying Liu
- Department
of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Mingxi Fang
- School
of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| |
Collapse
|
16
|
Makarov DM, Ksenofontov AA, Budkov YA. Consensus Modeling for Predicting Chemical Binding to Transthyretin as the Winning Solution of the Tox24 Challenge. Chem Res Toxicol 2025; 38:392-399. [PMID: 39969008 DOI: 10.1021/acs.chemrestox.4c00421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
The utilization of predictive methodologies for the assessment of toxicological properties represents an alternative approach that facilitates the identification of safe compounds while concurrently reducing the financial costs associated with the process. The objective of the Tox24 Challenge was to assess the progress in computational methods for predicting the activity of chemical binding to transthyretin (TTR). In order to fulfill the requirements of this task, the data set, measured by the Environmental Protection Agency, consisted of 1512 chemical substances of diverse nature. This paper describes the model that won the Tox24 Challenge and the steps taken for its further improvement. The Transformer convolutional neural network (CNN) model achieved the best performance as a standalone solution. Meanwhile, a multitask model built on a graph CNN, trained using 11 additional acute systemic toxicity data sets with increased weighting on the TTR binding activity, showed comparable results on the blind test set. The winning solution was a consensus model consisting of two catBoost models with OEstate and Mold2 descriptor sets, as well as two transformer-based models. The improvement of this solution involved adding a fifth model based on multitask learning using the graph CNN method, which led to a reduction in RMSE on the blind test set to 20.3%. The winning model was developed using the OCHEM web platform and is available online at https://ochem.eu/article/162082.
Collapse
Affiliation(s)
- Dmitriy M Makarov
- G. A. Krestov Institute of Solution Chemistry of the Russian Academy of Sciences, Ivanovo 153045, Russia
| | - Alexander A Ksenofontov
- G. A. Krestov Institute of Solution Chemistry of the Russian Academy of Sciences, Ivanovo 153045, Russia
| | - Yury A Budkov
- G. A. Krestov Institute of Solution Chemistry of the Russian Academy of Sciences, Ivanovo 153045, Russia
- Laboratory of Computational Physics, HSE University, Tallinskaya st. 34, Moscow 123458, Russia
- School of Applied Mathematics, HSE University, Tallinskaya st. 34, Moscow 123458, Russia
| |
Collapse
|
17
|
Chowdhury S, Rajaganapathy S, Sun L, Wang L, Yang P, Cerhan JR, Zong N. SensitiveCancerGPT: Leveraging Generative Large Language Model on Structured Omics Data to Optimize Drug Sensitivity Prediction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640661. [PMID: 40060567 PMCID: PMC11888479 DOI: 10.1101/2025.02.27.640661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Objective The fast accumulation of vast pharmacogenomics data of cancer cell lines provide unprecedented opportunities for drug sensitivity prediction (DSP), a crucial prerequisite for the advancement of precision oncology. Recently, Generative Large Language Models (LLM) have demonstrated performance and generalization prowess across diverse tasks in the field of natural language processing (NLP). However, the structured format of the pharmacogenomics data poses challenge for the utility of LLM in DSP. Therefore, the objective of this study is multi-fold: to adapt prompt engineering for structured pharmacogenomics data toward optimizing LLM's DSP performance, to evaluate LLM's generalization in real-world DSP scenarios, and to compare LLM's DSP performance against that of state-of-the-science baselines. Methods We systematically investigated the capability of the Generative Pre-trained Transformer (GPT) as a DSP model on four publicly available benchmark pharmacogenomics datasets, which are stratified by five cancer tissue types of cell lines and encompass both oncology and non-oncology drugs. Essentially, the predictive landscape of GPT is assessed for effectiveness on the DSP task via four learning paradigms: zero-shot learning, few-shot learning, fine-tuning and clustering pretrained embeddings. To facilitate GPT in seamlessly processing the structured pharmacogenomics data, domain-specific novel prompt engineering is employed by implementing three prompt templates (i.e., Instruction, Instruction-Prefix, Cloze) and integrating pharmacogenomics-related features into the prompt. We validated GPT's performance in diverse real-world DSP scenarios: cross-tissue generalization, blind tests, and analyses of drug-pathway associations and top sensitive/resistant cell lines. Furthermore, we conducted a comparative evaluation of GPT against multiple Transformer-based pretrained models and existing DSP baselines. Results Extensive experiments on the pharmacogenomics datasets across the five tissue cohorts demonstrate that fine-tuning GPT yields the best DSP performance (28% F1 increase, p-value= 0.0003) followed by clustering pretrained GPT embeddings (26% F1 increase, p-value= 0.0005), outperforming GPT in-context learning (i.e., few-shot). However, GPT in the zero-shot setting had a big F1 gap, resulting in the worst performance. Within the scope of prompt engineering, performance enhancement was achieved by directly instructing GPT about the DSP task and resorting to a concise context format (i.e., instruction-prefix), leading to F1 performance gain of 22% (p-value=0.02); while incorporation of drug-cell line prompt context derived from genomics and/or molecular features further boosted F1 score by 2%. Compared to state-of-the-science DSP baselines, GPT significantly asserted superior mean F1 performance (16% gain, p-value<0.05) on the GDSC dataset. In the cross-tissue analysis, GPT showcased comparable generalizability to the within-tissue performances on the GDSC and PRISM datasets, while statistically significant F1 performance improvements on the CCLE (8%, p-value=0.001) and DrugComb (19%, p-value=0.009) datasets. Evaluation on the challenging blind tests suggests GPT's competitiveness on the CCLE and DrugComb datasets compared to random splitting. Furthermore, analyses of the drug-pathway associations and log probabilities provided valuable insights that align with previous DSP findings. Conclusion The diverse experiment setups and in-depth analysis underscore the importance of generative LLM, such as GPT, as a viable in silico approach to guide precision oncology. Availability https://github.com/bioIKEA/SensitiveCancerGPT.
Collapse
Affiliation(s)
- Shaika Chowdhury
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN
| | | | - Lichao Sun
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN
- Lehigh University, Bethlehem, PA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN
| | - Ping Yang
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - James R Cerhan
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Nansu Zong
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN
| |
Collapse
|
18
|
Zhang PZ, Ballard J, Esquivel Fagiani F, Smith D, Gibson C, Yu X. Large-Scale Compartmental Model-Based Study of Preclinical Pharmacokinetic Data and Its Impact on Compound Triaging in Drug Discovery. Mol Pharm 2025; 22:1230-1240. [PMID: 39960135 DOI: 10.1021/acs.molpharmaceut.4c00813] [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] [Indexed: 03/04/2025]
Abstract
Reliable and robust human dose prediction plays a pivotal role in drug discovery. The prediction of human dose requires proper modeling of preclinical intravenous (IV) pharmacokinetic (PK) data, which is usually achieved either through noncompartmental analysis (NCA) or compartmental analysis. While NCA is straightforward, it loses valuable information about the shape of the PK curves. In contrast, compartmental analysis offers a more comprehensive interpretation but poses challenges in scaling up for high-throughput applications in discovery. To address this challenge, we developed computational frameworks, termed compartmental PK (CPK) and automated dose prediction (ADP), to enable automated compartmental model-based IV PK data modeling, translation, and simulation for human dose prediction in compound triaging and optimization. With CPK and ADP, we analyzed compounds with data collected at the MRL between 2013 and 2023 to quantitatively characterize the impact of different PK modeling and simulation methods on human dose prediction. Our study revealed that despite minimal impact on estimating animal PK parameters, different methods significantly impacted predicted human dose, exposure, and Cmax, driven more by different simulation assumptions than by the PK modeling itself. CPK-ADP therefore enables us to efficiently perform complex human dose predictions on a large scale while integrating the latest and best information available on absorption, distribution, and clearance to support decision-making in discovery.
Collapse
Affiliation(s)
- Peter Zhiping Zhang
- Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics (PDMB), MRL, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Jeanine Ballard
- Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics (PDMB), MRL, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Facundo Esquivel Fagiani
- Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics (PDMB), MRL, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Dustin Smith
- Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics (PDMB), MRL, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Christopher Gibson
- Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics (PDMB), MRL, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Xiang Yu
- Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics (PDMB), MRL, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| |
Collapse
|
19
|
Bashir B, Vishwas S, Gupta G, Paudel KR, Dureja H, Kumar P, Cho H, Sugandhi VV, Kumbhar PS, Disouza J, Dhanasekaran M, Goh BH, Gulati M, Dua K, Singh SK. Does drug repurposing bridge the gaps in management of Parkinson's disease? Unravelling the facts and fallacies. Ageing Res Rev 2025; 105:102693. [PMID: 39961372 DOI: 10.1016/j.arr.2025.102693] [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: 05/03/2024] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 02/21/2025]
Abstract
Repurposing the existing drugs for the management of both common and rare diseases is increasingly appealing due to challenges such as high attrition rates, the economy, and the slow pace of discovering new drugs. Drug repurposing involves the utilization of existing medications to treat diseases for which they were not originally intended. Despite encountering scientific and economic challenges, the pharmaceutical industry is intrigued by the potential to uncover new indications for medications. Medication repurposing is applicable across different stages of drug development, with the greatest potential observed when the drug has undergone prior safety testing. In this review, strategies for repurposing drugs for Parkinson's disease (PD) are outlined, a neurodegenerative disorder predominantly impacting dopaminergic neurons in the substantia nigra pars compacta region. PD is a debilitating neurodegenerative condition marked by an amalgam of motor and non-motor symptoms. Despite the availability of certain symptomatic treatments, particularly targeting motor symptoms, there remains a lack of established drugs capable of modifying the clinical course of PD, leading to its unchecked progression. Although standard drug discovery initiatives focusing on treatments that relieve diseases have yielded valuable understanding into the underlying mechanisms of PD, none of the numerous promising candidates identified in preclinical studies have successfully transitioned into clinically effective medications. Due to the substantial expenses associated with drug discovery endeavors, it is understandable that there has been a notable shift towards drug reprofiling strategies. Assessing the efficacy of an existing medication offers the additional advantage of circumventing the requirement for preclinical safety assessments and formulation enhancements, consequently streamlining the process and reducing both the duration of time and financial investments involved in bringing a treatment to clinical fruition. Furthermore, repurposed drugs may benefit from lower rates of failure, presenting an additional potential advantage. Various strategies for repurposing drugs are available to researchers in the field of PD. Some of these strategies have demonstrated effectiveness in identifying appropriate drugs for clinical trials, thereby providing validation for such strategies. This review provides an overview of the diverse strategies employed for drug reprofiling from approaches that place emphasis on single-gene transcriptional investigations to comprehensive epidemiological correlation analysis. Additionally, instances of previous or current research endeavors employing each strategy have been discussed. For the strategies that have not been yet implemented in PD research, their strategic efficacy is demonstrated using examples involving other disorders. In this review, we assess the safety and efficacy potential of prominent candidates repurposed as potential treatments for modifying the course of PD undergoing advanced clinical trials.
Collapse
Affiliation(s)
- Bushra Bashir
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Keshav Raj Paudel
- Centre of Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2007, Australia
| | - Harish Dureja
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India; Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana 124001, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Ghudda, Punjab, India
| | - Hyunah Cho
- College of Pharmacy & Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Vrashabh V Sugandhi
- College of Pharmacy & Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Popat S Kumbhar
- Department of Pharmaceutics, Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Kolhapur, Maharashtra, 416113, India.
| | - John Disouza
- Bombay Institute of Pharmacy and Research, Dombivli, Mumbai, Maharashtra, 421 203, India..
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University Auburn, AL 36849, USA
| | - Bey Hing Goh
- Sunway Biofunctional Molecules Discovery Centre (SBMDC), School of Medical and Life Sciences, Sunway University, Sunway, Malaysia
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India; Sunway Biofunctional Molecules Discovery Centre (SBMDC), School of Medical and Life Sciences, Sunway University, Sunway, Malaysia.
| |
Collapse
|
20
|
Elbouhi M, Ouabane M, Tabti K, Badaoui H, Abdessadak O, El Alaouy MA, Elkamel K, Lakhlifi T, Sbai A, Ajana MA, Bouachrine M. Computational evaluation of 1,2,3-triazole-based VEGFR-2 inhibitors: anti-angiogenesis potential and pharmacokinetic assessment. J Biomol Struct Dyn 2025; 43:2549-2559. [PMID: 38193897 DOI: 10.1080/07391102.2023.2301686] [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: 04/01/2023] [Accepted: 11/20/2023] [Indexed: 01/10/2024]
Abstract
The vascular endothelial growth factor (VEGF) and its cell surface receptor, as well as the human VEGFR-2 domain kinase, are some of the signaling pathways that have received the most attention in this field. This study aimed to identify novel molecules as VEGFR-2 inhibitors using 3D-QSAR modeling based on 1,2,3-triazole. Docking studies and dynamic simulations were performed to analyze novel interactions with the inhibitors and validate the molecular docking, dynamic simulations, and ADMET analyses. The optimized CoMSIA/SEH model showed good statistical results, and molecular docking and molecular dynamics simulations demonstrated stability of M3 ligand with the receptor and provided insight into ligand-receptor interactions. The newly developed compounds performed well in ADMET evaluations and showed promising results using Lipinski's rule of five, suggesting that the molecule M3 could be a useful anti-angiogenesis agent. In conclusion, this study provides insights into the structure-activity relationship of VEGFR-2 inhibitors and identifies M3 as a potential new anti-angiogenesis drug. The methodology used in this study can be applied to other similar drug targets to discover new and potent inhibitors.
Collapse
Affiliation(s)
- Mhamed Elbouhi
- Molecular Chemistry and Natural Substances Laboratory (MCNSL), Department of Chemistry, Faculty of Science, Moulay Ismail University, Meknes, Morocco
| | - Mohamed Ouabane
- Molecular Chemistry and Natural Substances Laboratory (MCNSL), Department of Chemistry, Faculty of Science, Moulay Ismail University, Meknes, Morocco
| | - Kamal Tabti
- Molecular Chemistry and Natural Substances Laboratory (MCNSL), Department of Chemistry, Faculty of Science, Moulay Ismail University, Meknes, Morocco
| | - Hassan Badaoui
- Molecular Chemistry and Natural Substances Laboratory (MCNSL), Department of Chemistry, Faculty of Science, Moulay Ismail University, Meknes, Morocco
| | - Oumayma Abdessadak
- Molecular Chemistry and Natural Substances Laboratory (MCNSL), Department of Chemistry, Faculty of Science, Moulay Ismail University, Meknes, Morocco
| | - Moulay Ahfid El Alaouy
- Molecular Chemistry and Natural Substances Laboratory (MCNSL), Department of Chemistry, Faculty of Science, Moulay Ismail University, Meknes, Morocco
| | - Khalid Elkamel
- Molecular Chemistry and Natural Substances Laboratory (MCNSL), Department of Chemistry, Faculty of Science, Moulay Ismail University, Meknes, Morocco
| | - Tahar Lakhlifi
- Molecular Chemistry and Natural Substances Laboratory (MCNSL), Department of Chemistry, Faculty of Science, Moulay Ismail University, Meknes, Morocco
| | - Abdelouahid Sbai
- Molecular Chemistry and Natural Substances Laboratory (MCNSL), Department of Chemistry, Faculty of Science, Moulay Ismail University, Meknes, Morocco
| | - Mohammed Aziz Ajana
- Molecular Chemistry and Natural Substances Laboratory (MCNSL), Department of Chemistry, Faculty of Science, Moulay Ismail University, Meknes, Morocco
| | - Mohammed Bouachrine
- Molecular Chemistry and Natural Substances Laboratory (MCNSL), Department of Chemistry, Faculty of Science, Moulay Ismail University, Meknes, Morocco
- Higher School of Technology (EST Khenifra), Sultan Moulay Slimane University, Beni-Mellal, Morocco
| |
Collapse
|
21
|
Guan H, Chen J, Yin H, Feng X, Liu C, Liu S, Li J, Li J, Cao Y, Ma C. A UHPLC-MS/MS Method Reveals the Pharmacokinetics of Deacetyl Asperulosidic Acid Methyl Ester in Rats. Biomed Chromatogr 2025; 39:e70001. [PMID: 39917783 DOI: 10.1002/bmc.70001] [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: 10/08/2024] [Revised: 12/08/2024] [Accepted: 01/03/2025] [Indexed: 05/08/2025]
Abstract
In the current study, a simple ultra-high performance liquid chromatography-tandem mass spectrometry method was developed and fully validated for the quantitation of deacetyl asperulosidic acid methyl ester in rat plasma. The plasma sample was precipitated with acetonitrile and then separated on the Waters ACQUITY UPLC HSS T3 column. The mobile phases, water and acetonitrile, were added with 0.1% formic acid. The mass spectrometry detection was performed in negative-ion multiple reaction monitoring. In the range of 1-1000 ng/mL, the linearity meets the requirements with correlation coefficient more than 0.99. The parameters of accuracy, precision, carryover, matrix effect, extraction recovery, stability, and dilution integrity are within accepted ranges. The validated method has been successfully used for pharmacokinetic study of deacetyl asperulosidic acid methyl ester in rats. After oral administration, deacetyl asperulosidic acid methyl ester was quickly absorbed into blood and reached the maximum plasma drug concentration of 4047.49 ng/mL at 2 h. The half-life of deacetyl asperulosidic acid methyl ester is 5.6 h, which suggests that it has a moderate metabolic process. Since the absolute bioavailability of deacetyl asperulosidic acid methyl ester is only 3.74%, its gastrointestinal stability, first-pass effect, and transmembrane properties remain to be studied.
Collapse
Affiliation(s)
- Huida Guan
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jian Chen
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hao Yin
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xia Feng
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chang Liu
- Department of Chinese Medicine Authentication, Faculty of Pharmacy, Naval Medical University, People's Liberation Army Navy, Shanghai, China
| | - Shanshan Liu
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiacheng Li
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingchu Li
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yongbing Cao
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chao Ma
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
22
|
Palacharla VRC, Nirogi R, Kumar N, Nandakumar K. Determination of Intrinsic Clearance and Fraction Unbound in Human Liver Microsomes and In Vitro-In Vivo Extrapolation of Human Hepatic Clearance for Marketed Central Nervous System Drugs. Eur J Drug Metab Pharmacokinet 2025; 50:119-135. [PMID: 39724218 DOI: 10.1007/s13318-024-00931-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 12/28/2024]
Abstract
OBJECTIVE The objective of this study was to determine the apparent intrinsic clearance (Clint, app) and fraction unbound in human liver microsomes (fu, mic) of 86 marketed central nervous system (CNS) drugs and to predict the in vivo hepatic blood clearance (CLh, b). METHODS Clint, app in human liver microsomes (HLM) was determined by substrate depletion, and fu, mic was determined by equilibrium dialysis. The relationship between lipophilicity (logP) and unbound intrinsic clearance (Clint, u) was explored using the Biopharmaceutical Drug Disposition Classification System (BDDCS) and Extended Clearance Classification System (ECCS). The predicted hepatic blood clearance by direct scaling, conventional method and Poulin method using well-stirred (WS) and parallel-tube (PT) models were compared with observed values. RESULTS The Clint, app in HLM ranged from < 5.8 to 477 µl/min/mg. The fu, mic in HLM ranged from 0.02 to 1.0. The scaled Clint values ranged from < 5 to 4496 ml/min/kg. The metabolic rate increased with an increase in logP (logP ≥ 2.5) of the CNS compounds. The direct scaling and Poulin methods showed comparable results based on the percentage of clearance predictions within a two-fold error. The conventional method resulted in under-predictions of Clint, in vivo or CLh, b using the WS or PT models. The Poulin method is favored over the other methods based on the statistical parameters. CONCLUSIONS Experimental Clint, app and fu, mic for 86 CNS compounds were successfully determined, and the scaled clearance was used to predict the hepatic blood clearance of 34 drugs. The success of prospective clearance predictions using HLM is expected to be high for most of the lipophilic BDDCS class 1 and class 2 and ECCS class 2 CNS compounds. The Poulin method resulted in more accurate predictions falling within a two-fold error of the observed values using the WS or PT models.
Collapse
Affiliation(s)
| | - Ramakrishna Nirogi
- Suven Life Sciences Limited, Serene Chambers, Road # 7, Hyderabad, 500034, India.
| | - Nitesh Kumar
- National Institute of Pharmaceutical Education and Research, Vaishali, Hajipur, Bihar, India
- Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Krishnadas Nandakumar
- Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
23
|
Chandel S, Parashar B, Ali SA, Sharma S. Predictive cavity and binding site identification: Techniques and applications. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2025; 103:43-63. [PMID: 40175054 DOI: 10.1016/bs.apha.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Strategies for recognizing predictive cavities and binding site identification are decisive for drug discovery, molecular docking, and tracing protein-ligand interactions. The two major approaches experimental and computational strive for prognosticating and distinguishing protein's binding sites. Profuse diminutive molecules are associated with the binding sites and influence normal biological functioning. The various structure-based strategies such as molecular dynamics, docking simulations, algorithms for pocket identification, and homology modeling are covered under computational techniques, where they propound the exhaustive comprehension of possible binding pockets hinge on the structure of protein and its physiochemical properties. The various sequence-based approaches rely on the homogeneousness of the sequence and machine learning replicas edified on already known protein and ligand composites to anticipate the interactive sites of novel proteins. The high-resolution structural identification and foot printing of protein to map the confirmational changes attributable to ligand and binding sites can be identified through diverse experimental methods such as NMR spectroscopy, mass spectrometry, and x-ray crystallography. These techniques are pivotal for drug discovery and designing, as the efficiency and specificity of ligands can be amplified through virtual screening and structural-based drug designing. Moreover, the ongoing developments in this domain promise to drive the revolution and efficiency in drug discovery and research in molecular biology.
Collapse
Affiliation(s)
- Shilpa Chandel
- Faculty of Pharmaceutical Sciences, The ICFAI University, Himachal Pradesh, India; Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India.
| | - Bharat Parashar
- Divine International College of Pharmacy, Gwalior, Madhya Pradesh, India
| | - Syed Atif Ali
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Shailesh Sharma
- Amar Shaheed Baba Ajit Singh Jujhar Singh Memorial College of Pharmacy, Bela, Punjab, India
| |
Collapse
|
24
|
Sur S, Nimesh H. Challenges and limitations of computer-aided drug design. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2025; 103:415-428. [PMID: 40175052 DOI: 10.1016/bs.apha.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Molecular Modelling in Drug Designing or Computer Aided Drug Designing (CADD) plays a significant role in new drug identification in the current world. However, it has sensitivity challenges and limitation because theoretical models involve assumption and approximations Computational models are not very accurate, some of the major challenges that face these models include the following. These include, for instance, molecular-docking or molecular-dynamics-simulation models which may not represent an accurate biological system and thus the predictions will be wrong. CADD depends on the availability of accurate, high-quality structural information for target proteins and ligand. Unfortunately, there are instances when experimental structures are not available, and homology models are employed, which can be imprecise. The computational cost is another drawback; only high accuracy simulations call for huge amounts of computational power and time well-suited for screening a multitude of agents. Moreover, they have weaknesses in determining pharmacokinetic and toxicity patterns of compounds that influence drug performance and effectiveness. In other words, even though CADD greatly helps drug discovery, it is still constrained by experimental validation to solve its drawbacks and optimize its foretelling.
Collapse
Affiliation(s)
- Souvik Sur
- Research and Development Center, Teerthanker Mahaveer University, Moradabad, Uttar Pradesh, India.
| | - Hemlata Nimesh
- Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh, India
| |
Collapse
|
25
|
Ambreen S, Umar M, Noor A, Jain H, Ali R. Advanced AI and ML frameworks for transforming drug discovery and optimization: With innovative insights in polypharmacology, drug repurposing, combination therapy and nanomedicine. Eur J Med Chem 2025; 284:117164. [PMID: 39721292 DOI: 10.1016/j.ejmech.2024.117164] [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: 09/27/2024] [Revised: 11/24/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024]
Abstract
Artificial Intelligence (AI) and Machine Learning (ML) are transforming drug discovery by overcoming traditional challenges like high costs, time-consuming, and frequent failures. AI-driven approaches streamline key phases, including target identification, lead optimization, de novo drug design, and drug repurposing. Frameworks such as deep neural networks (DNNs), convolutional neural networks (CNNs), and deep reinforcement learning (DRL) models have shown promise in identifying drug targets, optimizing delivery systems, and accelerating drug repurposing. Generative adversarial networks (GANs) and variational autoencoders (VAEs) aid de novo drug design by creating novel drug-like compounds with desired properties. Case studies, such as DDR1 kinase inhibitors designed using generative models and CDK20 inhibitors developed via structure-based methods, highlight AI's ability to produce highly specific therapeutics. Models like SNF-CVAE and DeepDR further advance drug repurposing by uncovering new therapeutic applications for existing drugs. Advanced ML algorithms enhance precision in predicting drug efficacy, toxicity, and ADME-Tox properties, reducing development costs and improving drug-target interactions. AI also supports polypharmacology by optimizing multi-target drug interactions and enhances combination therapy through predictions of drug synergies and antagonisms. In nanomedicine, AI models like CURATE.AI and the Hartung algorithm optimize personalized treatments by predicting toxicological risks and real-time dosing adjustments with high accuracy. Despite its potential, challenges like data quality, model interpretability, and ethical concerns must be addressed. High-quality datasets, transparent models, and unbiased algorithms are essential for reliable AI applications. As AI continues to evolve, it is poised to revolutionize drug discovery and personalized medicine, advancing therapeutic development and patient care.
Collapse
Affiliation(s)
- Subiya Ambreen
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Mohammad Umar
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Aaisha Noor
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Himangini Jain
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Ruhi Ali
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India.
| |
Collapse
|
26
|
Yousaf MA, Michel M, Khan ATA, Noreen M, Bano S. Repurposing doxycycline for the inhibition of monkeypox virus DNA polymerase: a comprehensive computational study. In Silico Pharmacol 2025; 13:27. [PMID: 39958784 PMCID: PMC11825436 DOI: 10.1007/s40203-025-00307-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 01/17/2025] [Indexed: 02/18/2025] Open
Abstract
The global spread of monkeypox, caused by the double-stranded DNA monkeypox virus (MPXV), has underscored the urgent need for effective antiviral treatments. In this study, we aim to identify a potent inhibitor for MPXV DNA polymerase (DNAP), a critical enzyme in the virus replication process. Using a computational drug repurposing approach, we performed a virtual screening of 1615 FDA-approved drugs based on drug-likeness and molecular docking against DNAP. Among these, 1430 compounds met Lipinski's rule of five for drug-likeness, with Doxycycline emerging as the most promising competitive inhibitor, binding strongly to the DNAP active site with a binding affinity of - 9.3 kcal/mol. This interaction involved significant hydrogen bonds, electrostatic interactions, and hydrophobic contacts, with Doxycycline demonstrating a stronger affinity than established antivirals for smallpox, including Cidofovir, Brincidofovir, and Tecovirimat. Stability and flexibility analyses through a 200 ns molecular dynamics simulation and normal mode analysis confirmed the robustness of Doxycycline binding to DNAP. Overall, our results suggest Doxycycline as a promising candidate for monkeypox treatment, though additional experimental and clinical studies are needed to confirm its therapeutic potential and clinical utility. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-025-00307-7.
Collapse
Affiliation(s)
- Muhammad Abrar Yousaf
- Section of Biology and Genetics, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Maurice Michel
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Abeedha Tu-Allah Khan
- School of Biological Sciences, Faculty of Life-Sciences, University of the Punjab, Lahore, Pakistan
- Department of Biological Sciences, Faculty of Allied Health Sciences, Superior University, Lahore, Pakistan
| | - Misbah Noreen
- Department of Biological Sciences, Virtual University of Pakistan, Lahore, Pakistan
- Department of Wildlife and Ecology, University of Veterinary and Animal Sciences, Ravi Campus, Pattoki, Pakistan
| | - Saddia Bano
- Department of Biological Sciences, Virtual University of Pakistan, Lahore, Pakistan
| |
Collapse
|
27
|
Warapande V, Meng F, Bozan A, Graff DE, Fromer JC, Mughal K, Mohideen FK, Shivangi, Paruchuri S, Johnston ML, Sharma P, Crea TR, Rudraraju RS, George A, Folvar C, Nelson AM, Neiditch MB, Zimmerman MD, Coley CW, Freundlich JS. Identification of Antituberculars with Favorable Potency and Pharmacokinetics through Structure-Based and Ligand-Based Modeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.636334. [PMID: 39974961 PMCID: PMC11838534 DOI: 10.1101/2025.02.03.636334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Drug discovery is inherently challenged by a multiple criteria decision making problem. The arduous path from hit discovery through lead optimization and preclinical candidate selection necessitates the evolution of a plethora of molecular properties. In this study, we focus on the hit discovery phase while beginning to address multiple criteria critical to the development of novel therapeutics to treat Mycobacterium tuberculosis infection. We develop a hybrid structure- and ligand-based pipeline for nominating diverse inhibitors targeting the β-ketoacyl synthase KasA by employing a Bayesian optimization-guided docking method and an ensemble model for compound nominations based on machine learning models for in vitro antibacterial efficacy, as characterized by minimum inhibitory concentration (MIC), and mouse pharmacokinetic (PK) plasma exposure. The application of our pipeline to the Enamine HTS library of 2.1M molecules resulted in the selection of 93 compounds, the experimental validation of which revealed exceptional PK (41%) and MIC (19%) success rates. Twelve compounds meet hit-like criteria in terms of MIC and PK profile and represent promising seeds for future drug discovery programs.
Collapse
|
28
|
Bowling PE, Broderick DR, Herbert JM. Convergent Protocols for Computing Protein-Ligand Interaction Energies Using Fragment-Based Quantum Chemistry. J Chem Theory Comput 2025; 21:951-966. [PMID: 39745995 PMCID: PMC11950710 DOI: 10.1021/acs.jctc.4c01429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Fragment-based quantum chemistry methods offer a means to sidestep the steep nonlinear scaling of electronic structure calculations so that large molecular systems can be investigated using high-level methods. Here, we use fragmentation to compute protein-ligand interaction energies in systems with several thousand atoms, using a new software platform for managing fragment-based calculations that implements a screened many-body expansion. Convergence tests using a minimal-basis semiempirical method (HF-3c) indicate that two-body calculations, with single-residue fragments and simple hydrogen caps, are sufficient to reproduce interaction energies obtained using conventional supramolecular electronic structure calculations, to within 1 kcal/mol at about 1% of the computational cost. We also demonstrate that the HF-3c results are illustrative of trends obtained with density functional theory in basis sets up to augmented quadruple-ζ quality. Strategic deployment of fragmentation facilitates the use of converged biomolecular model systems alongside high-quality electronic structure methods and basis sets, bringing ab initio quantum chemistry to systems of hitherto unimaginable size. This will be useful for generation of high-quality training data for machine learning applications.
Collapse
Affiliation(s)
- Paige E. Bowling
- Biophysics Graduate Program, The Ohio State University, Columbus, Ohio 43210 USA
- Department of Chemistry & Biochemistry, The Ohio State University, Columbus, Ohio 43210 USA
| | - Dustin R. Broderick
- Department of Chemistry & Biochemistry, The Ohio State University, Columbus, Ohio 43210 USA
| | - John M. Herbert
- Biophysics Graduate Program, The Ohio State University, Columbus, Ohio 43210 USA
- Department of Chemistry & Biochemistry, The Ohio State University, Columbus, Ohio 43210 USA
| |
Collapse
|
29
|
Chen J, Lin A, Jiang A, Qi C, Liu Z, Cheng Q, Yuan S, Luo P. Computational frameworks transform antagonism to synergy in optimizing combination therapies. NPJ Digit Med 2025; 8:44. [PMID: 39828791 PMCID: PMC11743742 DOI: 10.1038/s41746-025-01435-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025] Open
Abstract
While drug combinations are increasingly important in disease treatment, predicting their therapeutic interactions remains challenging. This review systematically analyzes computational methods for predicting drug combination effects through multi-omics data integration. We comprehensively assess key algorithms including DrugComboRanker and AuDNNsynergy, and evaluate integration approaches encompassing kernel regression and graph networks. The review elucidates artificial intelligence applications in predicting drug synergistic and antagonistic effects.
Collapse
Affiliation(s)
- Jinghong Chen
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Chang Qi
- Vienna University of Technology, Institute of Logic and Computation, Vienna, Austria
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Shuofeng Yuan
- Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China.
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong, Hong Kong, China.
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
30
|
Tripathi R, Kumar P. Identification of CXCR4 inhibitory activity in natural compounds using cheminformatics-guided machine learning algorithms. Integr Biol (Camb) 2025; 17:zyaf004. [PMID: 39985292 DOI: 10.1093/intbio/zyaf004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 10/22/2024] [Accepted: 02/11/2025] [Indexed: 02/24/2025]
Abstract
Neurodegenerative disorders are characterised by progressive damage to neurons that leads to cognitive impairment and motor dysfunction. Current treatment options focus only on symptom management and palliative care, without addressing their root cause. In our previous study, we reported the upregulation of the CXC motif chemokine receptor 4 (CXCR4), in Alzheimer's disease (ad) and Parkinson's disease (PD). We reached this conclusion by analysing gene expression patterns of ad and PD patients, compared to healthy individuals of similar age. We used RNA sequencing data from Gene Expression Omnibus to carry out this analysis. Herein, we aim to identify natural compounds that have potential inhibitory activity against CXCR4 through cheminformatics-guided machine learning, to aid drug discovery for neurodegenerative disorders, especially ad and PD. Natural compounds are gaining prominence in the treatment of neurodegenerative disorders due to their biocompatibility and potential neuroprotective properties, including their ability to modulate CXCR4 expression. Recent advances in artificial intelligence (AI) and machine learning (ML) algorithms have opened new avenues for drug discovery research across various therapeutic areas, including neurodegenerative disorders. We aim to produce an ML model using cheminformatics-guided machine learning algorithms using data of compounds with known CXCR4 activity, retrieved from the Binding Database, to analyse various physicochemical attributes of natural compounds obtained from the COCONUT Database and predict their inhibitory activity against CXCR4. Insight Box This work extends our previous study published in Integrative Biology (DOI: 10.1093/intbio/zyad012). We aim to demonstrate the effectiveness of AI and ML in identifying potential treatment options for Alzheimer's and Parkinson's diseases. By analysing vast amounts of data and identifying patterns that may not be apparent to human researchers, AI-powered systems can provide valuable insight into potential treatment options that may have been overlooked through traditional research methods. Our study underscores the significance of interdisciplinary collaboration between computational and experimental scientists in drug discovery and in developing a robust pipeline to identify potential leads for drug development.
Collapse
Affiliation(s)
- Rahul Tripathi
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University, Shahabad Daulatpur, Bawana Road, Delhi 110042, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University, Shahabad Daulatpur, Bawana Road, Delhi 110042, India
| |
Collapse
|
31
|
Zaki YH, Gomha SM, Farag B, Zaki ME, Hussein AM. Synthesis, characterization, and in silico studies of substituted 2,3-dihydro-1,3,4-thiadiazole derivatives. RESULTS IN CHEMISTRY 2025; 13:101977. [DOI: 10.1016/j.rechem.2024.101977] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025] Open
|
32
|
Loiodice S, D'Acquisto F, Drinkenburg P, Suojanen C, Llorca PM, Manji HK. Neuropsychiatric drug development: Perspectives on the current landscape, opportunities and potential future directions. Drug Discov Today 2025; 30:104255. [PMID: 39615745 DOI: 10.1016/j.drudis.2024.104255] [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: 10/09/2024] [Revised: 11/15/2024] [Accepted: 11/26/2024] [Indexed: 12/09/2024]
Abstract
Mental health represents a major challenge to our societies. One key difficulty associated with neuropsychiatric drug development is the lack of connection between the underlying biology and the disease. Nevertheless, there is growing optimism in this field with recent drug approvals (the first in decades) and renewed interest from pharmaceutical companies and investors. Here we review some of the most promising drug discovery and development endeavors currently deployed by industry. We also present elements illustrating the renewed interest from key stakeholders in neuropsychiatric drug development and provide potential future directions in this field.
Collapse
Affiliation(s)
| | - Fulvio D'Acquisto
- William Harvey Research Institute, School of Medicine and Dentistry, Queen Mary University of London, London, UK; School of Life and Health Science, University of Roehampton, London, UK
| | - Pim Drinkenburg
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands
| | - Christian Suojanen
- Broadreach Global LLC, Miami, FL, USA; European Brain Council, Brussels, Belgium
| | - Pierre-Michel Llorca
- Department of Psychiatry, CHU Clermont-Ferrand, University of Clermont Auvergne, CNRS, Clermont Auvergne INP, Institut Pascal (UMR 6602), Clermont-Ferrand, France; Fondation FondaMental, Créteil, France
| | - Husseini K Manji
- Oxford University, Oxford, UK; Yale University, New Haven, CT, USA; UK Government Mental Health Mission, London, UK
| |
Collapse
|
33
|
Gomha SM, Riyadh SM, Farag B, Al-Hussain SA, Zaki MEA, Mohamed MA. Green synthesis of hydrazono-thiazolones using vitamin B1 and their antibacterial implications. GREEN CHEMISTRY LETTERS AND REVIEWS 2024; 17. [DOI: 10.1080/17518253.2024.2380746] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 07/12/2024] [Indexed: 05/14/2025]
Affiliation(s)
- Sobhi M. Gomha
- Department of Chemistry, Faculty of Science, Islamic University of Madinah, Madinah, Saudi Arabia
| | - Sayed M. Riyadh
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| | - Basant Farag
- Department of Chemistry, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Sami A. Al-Hussain
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Magdi E. A. Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Mahmoud A. Mohamed
- Technology of Textile Department, Faculty of Technology and Education, Beni-Suef University, Beni-Suef, Egypt
- Chemistry Department, Faculty of Science and Humanity study-Afif, Shaqra University, Saudi Arabia
| |
Collapse
|
34
|
Gadaleta D, Serrano-Candelas E, Ortega-Vallbona R, Colombo E, Garcia de Lomana M, Biava G, Aparicio-Sánchez P, Roncaglioni A, Gozalbes R, Benfenati E. Comprehensive benchmarking of computational tools for predicting toxicokinetic and physicochemical properties of chemicals. J Cheminform 2024; 16:145. [PMID: 39726044 DOI: 10.1186/s13321-024-00931-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 11/11/2024] [Indexed: 12/28/2024] Open
Abstract
Ensuring the safety of chemicals for environmental and human health involves assessing physicochemical (PC) and toxicokinetic (TK) properties, which are crucial for absorption, distribution, metabolism, excretion, and toxicity (ADMET). Computational methods play a vital role in predicting these properties, given the current trends in reducing experimental approaches, especially those that involve animal experimentation. In the present manuscript, twelve software tools implementing Quantitative Structure-Activity Relationship (QSAR) models were selected for the prediction of 17 relevant PC and TK properties. A total of 41 validation datasets were collected from the literature, curated and used for assessing the models' external predictivity, emphasizing the performance of the models inside the applicability domain. Overall, the results confirmed the adequate predictive performance of the majority of the selected tools, with models for PC properties (R2 average = 0.717) generally outperforming those for TK properties (R2 average = 0.639 for regression, average balanced accuracy = 0.780 for classification). Notably, several of the tools evaluated exhibited good predictivity across different properties and were identified as recurring optimal choices. Moreover, a systematic analysis of the chemical space covered by the external validation datasets confirmed the validity of the collected results for relevant chemical categories (e.g., drugs and industrial chemicals), further increasing the confidence in the overall evaluation. The best performing models were ultimately suggested for each investigated property and proposed as robust computational tools for high-throughput assessment of highly relevant chemical properties. SCIENTIFIC CONTRIBUTION: The present manuscript provides an overview of the state-of-the-art available computational tools for predicting the PC and TK properties of chemicals. The results here offer valuable guidance to researchers, regulatory authorities, and the industry in identifying robust computational tools suitable for predicting relevant chemical properties in the context of chemical design, toxicity and environmental fate assessment.
Collapse
Affiliation(s)
- Domenico Gadaleta
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| | - Eva Serrano-Candelas
- ProtoQSAR SL, CEEI (Centro Europeo de Empresas Innovadoras), 46980, Paterna, Valencia, Spain
| | - Rita Ortega-Vallbona
- ProtoQSAR SL, CEEI (Centro Europeo de Empresas Innovadoras), 46980, Paterna, Valencia, Spain
| | - Erika Colombo
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marina Garcia de Lomana
- Bayer AG, Machine Learning Research, Research & Development, Pharmaceuticals, Leverkusen, Germany
| | - Giada Biava
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Pablo Aparicio-Sánchez
- ProtoQSAR SL, CEEI (Centro Europeo de Empresas Innovadoras), 46980, Paterna, Valencia, Spain
- Spanish National Cancer Research Center (CNIO), Experimental Therapeutics Programme, Madrid, Spain
| | - Alessandra Roncaglioni
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Rafael Gozalbes
- ProtoQSAR SL, CEEI (Centro Europeo de Empresas Innovadoras), 46980, Paterna, Valencia, Spain
- Moldrug AI Systems SL, c/Olimpia Arozena Torres 45, 46018, Valencia, Spain
| | - Emilio Benfenati
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
35
|
Li P, Hua L, Ma Z, Hu W, Liu Y, Zhu J. Conformalized Graph Learning for Molecular ADMET Property Prediction and Reliable Uncertainty Quantification. J Chem Inf Model 2024; 64:8705-8717. [PMID: 39571080 DOI: 10.1021/acs.jcim.4c01139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Drug discovery and development is a complex and costly process, with a substantial portion of the expense dedicated to characterizing the absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties of new drug candidates. While the advent of deep learning and molecular graph neural networks (GNNs) has significantly enhanced in silico ADMET prediction capabilities, reliably quantifying prediction uncertainty remains a critical challenge. The performance of GNNs is influenced by both the volume and the quality of the data. Hence, determining the reliability and extent of a prediction is as crucial as achieving accurate predictions, especially for out-of-domain (OoD) compounds. This paper introduces a novel GNN model called conformalized fusion regression (CFR). CFR combined a GNN model with a joint mean-quantile regression loss and an ensemble-based conformal prediction (CP) method. Through rigorous evaluation across various ADMET tasks, we demonstrate that our framework provides accurate predictions, reliable probability calibration, and high-quality prediction intervals, outperforming existing uncertainty quantification methods.
Collapse
Affiliation(s)
- Peiyao Li
- Department of Computer Science and Technology, Tsinghua University, Beijing 100084, China
- Molecular Science, BeiGene (Beijing) Inc., Beijing 102206, China
| | - Lan Hua
- Molecular Science, BeiGene (Beijing) Inc., Beijing 102206, China
| | - Zhechao Ma
- Department of Computer Science and Technology, Hefei University of Technology, Hefei 230009, China
| | - Wenbo Hu
- Department of Computer Science and Technology, Hefei University of Technology, Hefei 230009, China
| | - Ye Liu
- Molecular Science, BeiGene (Beijing) Inc., Beijing 102206, China
| | - Jun Zhu
- Department of Computer Science and Technology, Tsinghua University, Beijing 100084, China
| |
Collapse
|
36
|
Schofield C, Sarrigiannidis S, Moran‐Horowich A, Jackson E, Rodrigo‐Navarro A, van Agtmael T, Cantini M, Dalby MJ, Salmeron‐Sanchez M. An In Vitro Model of the Blood-Brain Barrier for the Investigation and Isolation of the Key Drivers of Barriergenesis. Adv Healthc Mater 2024; 13:e2303777. [PMID: 39101628 PMCID: PMC11670300 DOI: 10.1002/adhm.202303777] [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: 10/30/2023] [Revised: 07/24/2024] [Indexed: 08/06/2024]
Abstract
The blood-brain barrier (BBB) tightly regulates substance transport between the bloodstream and the brain. Models for the study of the physiological processes affecting the BBB, as well as predicting the permeability of therapeutic substances for neurological and neurovascular pathologies, are highly desirable. Existing models, such as Transwell utilizing-models, do not mimic the extracellular environment of the BBB with their stiff, semipermeable, non-biodegradable membranes. To help overcome this, we engineered electrospun membranes from poly L-lactic acid in combination with a nanometric coating of poly(ethyl acrylate) (PEA) that drives fibrillogenesis of fibronectin, facilitating the synergistic presentation of both growth factors and integrin binding sites. Compared to commercial semi-porous membranes, these membranes significantly improve the expression of BBB-related proteins in brain endothelial cells. PEA-coated membranes in combination with different growth factors and extracellular protein coatings reveal nerve growth factor (NGF) and fibroblast growth factor (FGF-2) caused formation of better barriers in vitro. This BBB model offers a robust platform for studying key biochemical factors influencing barrier formation that marries the simplicity of the Transwell model with the highly tunable electrospun PEA-fibronectin membranes. This enables the generation of high-throughput drug permeability models without the need of complicated co-culture conditions.
Collapse
Affiliation(s)
- Christina Schofield
- Centre for the Cellular MicroenvironmentUniversity of GlasgowGlasgowG11 6EWUK
| | | | | | - Emma Jackson
- Centre for the Cellular MicroenvironmentUniversity of GlasgowGlasgowG11 6EWUK
| | | | - Tom van Agtmael
- School of Cardiovascular and Metabolic HealthUniversity of GlasgowGlasgowG12 8TAUK
| | - Marco Cantini
- Centre for the Cellular MicroenvironmentUniversity of GlasgowGlasgowG11 6EWUK
| | - Matthew J. Dalby
- Centre for the Cellular MicroenvironmentUniversity of GlasgowGlasgowG11 6EWUK
| | - Manuel Salmeron‐Sanchez
- Centre for the Cellular MicroenvironmentUniversity of GlasgowGlasgowG11 6EWUK
- Institute for Bioengineering of Catalonia (IBEC)The Barcelona Institute for Science and Technology (BIST)Barcelona08028Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA)BarcelonaSpain
| |
Collapse
|
37
|
Ma L, Yan Y, Dai S, Shao D, Yi S, Wang J, Li J, Yan J. Research on prediction of human oral bioavailability of drugs based on improved deep forest. J Mol Graph Model 2024; 133:108851. [PMID: 39232489 DOI: 10.1016/j.jmgm.2024.108851] [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: 06/06/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024]
Abstract
Human oral bioavailability is a crucial factor in drug discovery. In recent years, researchers have constructed a variety of different prediction models. However, given the limited size of human oral bioavailability data sets, the challenge of making accurate predictions with small sample sizes has become a critical issue in the field. The deep forest model, with its adaptively determinable number of cascade levels, can perform exceptionally well even on small-scale data. However, the original deep forest suffers unbalanced multi-grained scanning process and premature stopping of cascade forest training. In this paper, we propose a human oral bioavailability predict method based on an improved deep forest, called balanced multi-grained scanning mapping cascade forest (bgmc-forest). Firstly, the mordred descriptor method is selected to feature extraction, then enhanced features are obtained by the improved balanced multi-grained scanning, which solves the problem of missing features at both ends. And finally, the prediction results are obtained by feature mapping cascaded forests, which is based on principal component analysis and cascade forests, ensures the effectiveness of the cascade forest. The superiority of the model constructed in this paper is demonstrated through comparative experiments, while the effectiveness of the improved module is verified through ablation experiments. Finally the decision-making process of the model is explained by the shapley additive explanations interpretation algorithm.
Collapse
Affiliation(s)
- Lei Ma
- Kunming University of Science and Technology, Kunming, CN 650500, China
| | - Yukun Yan
- Kunming University of Science and Technology, Kunming, CN 650500, China
| | - Shaoxing Dai
- Kunming University of Science and Technology, Kunming, CN 650500, China
| | - Dangguo Shao
- Kunming University of Science and Technology, Kunming, CN 650500, China.
| | - Sanli Yi
- Kunming University of Science and Technology, Kunming, CN 650500, China
| | - Jiawei Wang
- Kunming University of Science and Technology, Kunming, CN 650500, China
| | - Jingtao Li
- Kunming University of Science and Technology, Kunming, CN 650500, China
| | - Jiangkai Yan
- Kunming University of Science and Technology, Kunming, CN 650500, China
| |
Collapse
|
38
|
Camara Dit Pinto S, Cherkaoui J, Ghosh D, Cazaubon V, Benzeroual KE, Levine SM, Cherkaoui M, Sood GK, Anandasabapathy S, Dhingra S, Vierling JM, Gallo NR. A virtual scalable model of the Hepatic Lobule for acetaminophen hepatotoxicity prediction. NPJ Digit Med 2024; 7:340. [PMID: 39604584 PMCID: PMC11603025 DOI: 10.1038/s41746-024-01349-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 11/16/2024] [Indexed: 11/29/2024] Open
Abstract
Addressing drug-induced liver injury is crucial in drug development, often causing Phase III trial failures and market withdrawals. Traditional animal models fail to predict human liver toxicity accurately. Virtual twins of human organs present a promising solution. We introduce the Virtual Hepatic Lobule, a foundational element of the Living Liver, a multi-scale liver virtual twin. This model integrates blood flow dynamics and an acetaminophen-induced injury model to predict hepatocyte injury patterns specific to patients. By incorporating metabolic zonation, our predictions align with clinical zonal hepatotoxicity observations. This methodology advances the development of a human liver virtual twin, aiding in the prediction and validation of drug-induced liver injuries.
Collapse
Affiliation(s)
- Stelian Camara Dit Pinto
- Department of Computer Science, Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY, USA
| | - Jalal Cherkaoui
- Department of Computer Science, Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY, USA
- Institut National des Sciences Appliquées, Lyon, France
| | - Debarshi Ghosh
- Department of Computer Science, Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY, USA
| | - Valentine Cazaubon
- Department of Computer Science, Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY, USA
| | - Kenza E Benzeroual
- Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA
| | | | - Mohammed Cherkaoui
- Department of Computer Science, Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY, USA
| | - Gagan K Sood
- Division of Gastroenterology, Baylor College of Medicine, Houston, TX, USA
| | | | - Sadhna Dhingra
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - John M Vierling
- Departments of Medicine and Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Nicolas R Gallo
- Department of Computer Science, Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY, USA.
- Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA.
| |
Collapse
|
39
|
Kreutz A, Chang X, Hogberg HT, Wetmore BA. Advancing understanding of human variability through toxicokinetic modeling, in vitro-in vivo extrapolation, and new approach methodologies. Hum Genomics 2024; 18:129. [PMID: 39574200 PMCID: PMC11580331 DOI: 10.1186/s40246-024-00691-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/01/2024] [Indexed: 11/25/2024] Open
Abstract
The merging of physiology and toxicokinetics, or pharmacokinetics, with computational modeling to characterize dosimetry has led to major advances for both the chemical and pharmaceutical research arenas. Driven by the mutual need to estimate internal exposures where in vivo data generation was simply not possible, the application of toxicokinetic modeling has grown exponentially in the past 30 years. In toxicology the need has been the derivation of quantitative estimates of toxicokinetic and toxicodynamic variability to evaluate the suitability of the tenfold uncertainty factor employed in risk assessment decision-making. Consideration of a host of physiologic, ontogenetic, genetic, and exposure factors are all required for comprehensive characterization. Fortunately, the underlying framework of physiologically based toxicokinetic models can accommodate these inputs, in addition to being amenable to capturing time-varying dynamics. Meanwhile, international interest in advancing new approach methodologies has fueled the generation of in vitro toxicity and toxicokinetic data that can be applied in in vitro-in vivo extrapolation approaches to provide human-specific risk-based information for historically data-poor chemicals. This review will provide a brief introduction to the structure and evolution of toxicokinetic and physiologically based toxicokinetic models as they advanced to incorporate variability and a wide range of complex exposure scenarios. This will be followed by a state of the science update describing current and emerging experimental and modeling strategies for population and life-stage variability, including the increasing application of in vitro-in vivo extrapolation with physiologically based toxicokinetic models in pharmaceutical and chemical safety research. The review will conclude with case study examples demonstrating novel applications of physiologically based toxicokinetic modeling and an update on its applications for regulatory decision-making. Physiologically based toxicokinetic modeling provides a sound framework for variability evaluation in chemical risk assessment.
Collapse
Affiliation(s)
- Anna Kreutz
- Inotiv, 601 Keystone Park Drive, Suite 200, Morrisville, NC, 27560, USA.
- Oak Ridge Institute for Science and Education, Oak Ridge, TN, 37830, USA.
| | - Xiaoqing Chang
- Inotiv, 601 Keystone Park Drive, Suite 200, Morrisville, NC, 27560, USA
| | | | - Barbara A Wetmore
- Office of Research and Development, Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| |
Collapse
|
40
|
Rao M, Nassiri V, Srivastava S, Yang A, Brar S, McDuffie E, Sachs C. Artificial Intelligence and Machine Learning Models for Predicting Drug-Induced Kidney Injury in Small Molecules. Pharmaceuticals (Basel) 2024; 17:1550. [PMID: 39598459 PMCID: PMC11597314 DOI: 10.3390/ph17111550] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/09/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND/OBJECTIVES Drug-Induced Kidney Injury (DIKI) presents a significant challenge in drug development, often leading to clinical-stage failures. The early prediction of DIKI risk can improve drug safety and development efficiency. Existing models tend to focus on physicochemical properties alone, often overlooking drug-target interactions crucial for DIKI. This study introduces an AI/ML (artificial intelligence/machine learning) model that integrates both physicochemical properties and off-target interactions to enhance DIKI prediction. METHODS We compiled a dataset of 360 FDA-classified compounds (231 non-nephrotoxic and 129 nephrotoxic) and predicted 6064 off-target interactions, 59% of which were validated in vitro. We also calculated 55 physicochemical properties for these compounds. Machine learning (ML) models were developed using four algorithms: Ridge Logistic Regression (RLR), Support Vector Machine (SVM), Random Forest (RF), and Neural Network (NN). These models were then combined into an ensemble model for enhanced performance. RESULTS The ensemble model achieved an ROC-AUC of 0.86, with a sensitivity and specificity of 0.79 and 0.78, respectively. The key predictive features included 38 off-target interactions and physicochemical properties such as the number of metabolites, polar surface area (PSA), pKa, and fraction of Sp3-hybridized carbons (fsp3). These features effectively distinguished DIKI from non-DIKI compounds. CONCLUSIONS The integrated model, which combines both physicochemical properties and off-target interaction data, significantly improved DIKI prediction accuracy compared to models that rely on either data type alone. This AI/ML model provides a promising early screening tool for identifying compounds with lower DIKI risk, facilitating safer drug development.
Collapse
Affiliation(s)
- Mohan Rao
- Preclinical and Clinical Pharmacology and Chemistry, Neurocrine Biosciences, San Diego, CA 92130, USA (C.S.)
| | - Vahid Nassiri
- Open Analytics NV, Jupiterstraat 20, 2600 Antwerp, Belgium;
| | - Sanjay Srivastava
- Preclinical and Clinical Pharmacology and Chemistry, Neurocrine Biosciences, San Diego, CA 92130, USA (C.S.)
| | - Amy Yang
- Preclinical and Clinical Pharmacology and Chemistry, Neurocrine Biosciences, San Diego, CA 92130, USA (C.S.)
| | - Satjit Brar
- Preclinical and Clinical Pharmacology and Chemistry, Neurocrine Biosciences, San Diego, CA 92130, USA (C.S.)
| | - Eric McDuffie
- Preclinical and Clinical Pharmacology and Chemistry, Neurocrine Biosciences, San Diego, CA 92130, USA (C.S.)
| | - Clifford Sachs
- Preclinical and Clinical Pharmacology and Chemistry, Neurocrine Biosciences, San Diego, CA 92130, USA (C.S.)
| |
Collapse
|
41
|
Hublikar M, Kadu V, Edake N, Raut D, Shirame S, Ahmed MZ, Makam P, Ahmad MS, Meshram RJ, Bhosale R. Design, Synthesis, Anti-Cancer, Anti-Inflammatory and In Silico Studies of 3-Substituted-2-Oxindole Derivatives. Chem Biodivers 2024; 21:e202400844. [PMID: 39078869 DOI: 10.1002/cbdv.202400844] [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: 04/07/2024] [Accepted: 07/29/2024] [Indexed: 09/25/2024]
Abstract
This study focuses on the design and synthesis of 3-substituted-2-oxindole derivatives aimed at developing dual-active molecules with anti-cancer and anti-inflammatory properties. The molecules were designed with diverse structural and functional features while adhering to Lipinski, Veber, and Leeson criteria. Physicochemical properties were assessed using SWISSADME to ensure drug-likeness and favourable pharmacokinetics. Multistep synthetic procedures were employed for molecule synthesis. In vitro evaluations confirmed the dual activity of the derivatives, with specific emphasis on the significance of dialkyl aminomethyl substitutions for potency against various cell lines. 4 a exhibited GI50 value 3.00E-05 against MDA-MB-231, 4 b has shown GI50 value 2E-05 against MDA-MB-231, 4 c has shown GI50 value 6E-05 against VERO, 4 d has shown GI50 value 8E-05 each against both the MDA-MB-231 and MCF-7 and 4 e has shown GI50 values 2E-05 and 5E-05 each against both the MCF-7 and VERO. The analysis indicates that compounds 3 c (71.19 %), 3 e (66.84 %), and 3 g (63.04 %) exhibited significant anti-inflammatory activity. Additionally, in silico binding free energy analysis and interaction studies revealed significant correlations between in vitro and computational data, identifying compounds 4 d, 4 e, 3 b, 3 i, and 3 e as promising candidates. Key residues such as Glu917, Cys919, Lys920, Glu850, Lys838, and Asp1046 were found to play critical roles in ligand binding and kinase inhibition, providing valuable insights for designing potent VEGFR2 inhibitors. The Quantum Mechanics-based Independent Gradient Model analysis further highlighted the electronic interaction landscape, showing larger attractive peaks and higher electron density gradients for compounds 4 d and 4 e compared to Sunitinib, suggesting stronger and more diverse attractive forces. These findings support the potential of these compounds for further development and optimization in anticancer drug design.
Collapse
Affiliation(s)
- Mahesh Hublikar
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Solapur University, Solapur, Maharashtra, 413255, India
| | - Vikas Kadu
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Solapur University, Solapur, Maharashtra, 413255, India
| | - Nagesh Edake
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Solapur University, Solapur, Maharashtra, 413255, India
| | - Dattatraya Raut
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Solapur University, Solapur, Maharashtra, 413255, India
| | - Sachin Shirame
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Solapur University, Solapur, Maharashtra, 413255, India
| | - Mahammad Z Ahmed
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Parameshwar Makam
- Department of Chemistry, School of Applied and Life Sciences, Uttaranchal University, Arcadia Grant, P.O. Chandanwari, Premnagar, Dehradun, Uttarakhand, 248007, India
| | - Md Sibgatullah Ahmad
- Bioinformatics Centre, Savitribai Phule Pune University, Pune, Maharashtra, 248007, India
| | - Rohan J Meshram
- Bioinformatics Centre, Savitribai Phule Pune University, Pune, Maharashtra, 248007, India
| | - Raghunath Bhosale
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Solapur University, Solapur, Maharashtra, 413255, India
| |
Collapse
|
42
|
Sharma R, Banerjee S, Sharma R. Role of Mandukparni (Centella asiatica Linn Urban) in neurological disorders: Evidence from ethnopharmacology and clinical studies to network enrichment analysis. Neurochem Int 2024; 180:105865. [PMID: 39307460 DOI: 10.1016/j.neuint.2024.105865] [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: 12/17/2023] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
Centella asiatica Linn Urban (C. asiatica), aka Mandukparni, is one of the flagship herbs used in traditional medicines to effectively manage neurological problems. Although this plant has a wealth of comprehensive preclinical pharmacological profiles, further clinical research and execution of its molecular mode of action are still required. We searched electronic databases (Google Scholar, SciFinder, MEDLINE, Scopus, EMBASE, Science Direct, and PubMed) using relevant key words to retrieve information pertaining to C. asiatica till June 2023 and performed network pharmacology to understand the mechanism related to their neurobiological roles. This study extensively analyses its pharmacological properties, nutritional profile, ethnomedical uses, safety, and mechanistic role in treating neurological and neurodegenerative disorders. Additionally, a network pharmacology study was performed which suggests that its phytomolecules are involved in different neuroactive ligand-receptor pathways, glial cell differentiation, gliogenesis, and astrocyte differentiation. Hopefully, this report will lead to a paradigm shift in medical practice, research, and the creation of phytopharmaceuticals derived from C. asiatica that target the central nervous system.
Collapse
Affiliation(s)
- Ruchi Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India.
| | - Subhadip Banerjee
- Medicinal Plant Innovation Centre, School of Integrative Medicine, Mae Fah Luang University, Chiang Rai, 57100, Thailand.
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India.
| |
Collapse
|
43
|
Driche EH, Badji B, Mathieu F, Zitouni A. In-vitro antibacterial and antibiofilm activities and in-silico analysis of a potent cyclic peptide from a novel Streptomyces sp. strain RG-5 against antibiotic-resistant and biofilm-forming pathogenic bacteria. Arch Microbiol 2024; 206:450. [PMID: 39476249 DOI: 10.1007/s00203-024-04174-2] [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: 08/19/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 11/10/2024]
Abstract
The proliferation of multidrug-resistant and biofilm-forming pathogenic bacteria poses a serious threat to public health. The limited effectiveness of current antibiotics motivates the search for new antibacterial compounds. In this study, a novel strain, RG-5, was isolated from desert soil. This strain exhibited potent antibacterial and antibiofilm properties against multidrug-resistant and biofilm-forming pathogenic bacteria. Through phenotypical characterizations, 16S rRNA gene sequence and phylogenetic analysis, the strain was identified as Streptomyces pratensis with 99.8% similarity. The active compound, RG5-1, was extracted, purified by reverse phase silica column HPLC, identified by ESI-MS spectrometry, and confirmed by 1H and 13C NMR analysis as 2,5-Piperazinedione, 3,6-bis(2-methylpropyl), belonging to cyclic peptides. This compound showed interesting minimum inhibitory concentrations (MICs) of 04 to 15 µg/mL and minimum biofilm inhibitory concentrations (MBICs 50%) of ½ MIC against the tested bacteria. Its molecular mechanism of action was elucidated through a molecular docking study against five drug-protein targets. The results demonstrated that the compound RG5-1 has a strong affinity and interaction patterns with glucosamine-6-phosphate synthase at - 6.0 kcal/mol compared to reference inhibitor (- 5.4 kcal/mol), medium with penicillin-binding protein 1a (- 6.1 kcal/mol), and LasR regulator protein of quorum sensing (- 5.4 kcal/mol), confirming its antibacterial and antibiofilm activities. The compound exhibited minimal toxicity and favorable physicochemical and pharmacological properties. This is the first report that describes its production from Streptomyces, its activities against biofilm-forming and multidrug-resistant bacteria, and its mechanism of action. These findings indicate that 2,5-piperazinedione, 3,6-bis(2-methylpropyl) has the potential to be a promising lead compound in the treatment of antibiotic-resistant and biofilm-forming pathogens.
Collapse
Affiliation(s)
- El-Hadj Driche
- Laboratory of Molecular Biology, Genomics and Bioinformatics (LBMGB), Department of Biology, Faculty of Nature and Life Sciences (SNV), Hassiba Benbouali University of Chlef, Ouled Fares, Chlef, 02180, Algeria.
- Laboratory of Biology of Microbial Systems (LBSM), Higher Normal School of Kouba B.P. 92, Kouba, Alger, 16050, Algeria.
| | - Boubekeur Badji
- Laboratory of Biology of Microbial Systems (LBSM), Higher Normal School of Kouba B.P. 92, Kouba, Alger, 16050, Algeria
| | - Florence Mathieu
- Chemical Engineering Laboratory, LGC, UMR 5503 (CNRS/INPT/UPS), University of Toulouse, Toulouse, France
| | - Abdelghani Zitouni
- Laboratory of Biology of Microbial Systems (LBSM), Higher Normal School of Kouba B.P. 92, Kouba, Alger, 16050, Algeria
| |
Collapse
|
44
|
Guengerich FP. Roles of Individual Human Cytochrome P450 Enzymes in Drug Metabolism. Pharmacol Rev 2024; 76:1104-1132. [PMID: 39054072 PMCID: PMC11549934 DOI: 10.1124/pharmrev.124.001173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/28/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024] Open
Abstract
Our knowledge of the roles of individual cytochrome P450 (P450) enzymes in drug metabolism has developed considerably in the past 30 years, and this base has been of considerable use in avoiding serious issues with drug interactions and issues due to variations. Some newer approaches are being considered for "phenotyping" metabolism reactions with new drug candidates. Endogenous biomarkers are being used for noninvasive estimation of levels of individual P450 enzymes. There is also the matter of some remaining "orphan" P450s, which have yet to be assigned reactions. Practical problems that continue in drug development include predicting drug-drug interactions, predicting the effects of polymorphic and other P450 variations, and evaluating interspecies differences in drug metabolism, particularly in the context of "metabolism in safety testing" regulatory issues ["disproportionate (human) metabolites"]. SIGNIFICANCE STATEMENT: Cytochrome P450 enzymes are the major catalysts involved in drug metabolism. The characterization of their individual roles has major implications in drug development and clinical practice.
Collapse
Affiliation(s)
- F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
45
|
Aburidi M, Marcia R. Optimal Transport Based Graph Kernels for Drug Property Prediction. IEEE OPEN JOURNAL OF ENGINEERING IN MEDICINE AND BIOLOGY 2024; 6:152-157. [PMID: 39906265 PMCID: PMC11793854 DOI: 10.1109/ojemb.2024.3480708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 09/12/2024] [Accepted: 10/09/2024] [Indexed: 02/06/2025] Open
Abstract
Objective: The development of pharmaceutical agents relies heavily on optimizing their pharmacodynamics, pharmacokinetics, and toxicological properties, collectively known as ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity). Accurate assessment of these properties during the early stages of drug development is challenging due to resource-intensive experimental evaluation and limited comprehensive data availability. To overcome these obstacles, there has been a growing reliance on computational and predictive tools, leveraging recent advancements in machine learning and graph-based methodologies. This study presents an innovative approach that harnesses the power of optimal transport (OT) theory to construct three graph kernels for predicting drug ADMET properties. This approach involves the use of graph matching to create a similarity matrix, which is subsequently integrated into a predictive model. Results: Through extensive evaluations on 19 distinct ADMET datasets, the potential of this methodology becomes evident. The OT-based graph kernels exhibits exceptional performance, outperforming state-of-the-art graph deep learning models in 9 out of 19 datasets, even surpassing the most impactful Graph Neural Network (GNN) that excels in 4 datasets. Furthermore, they are very competitive in 2 additional datasets. Conclusion: Our proposed novel class of OT-based graph kernels not only demonstrates a high degree of effectiveness and competitiveness but also, in contrast to graph neural networks, offers interpretability, adaptability and generalizability across multiple datasets.
Collapse
Affiliation(s)
- Mohammed Aburidi
- Department of Applied MathematicsUniversity of California MercedMercedCA95348USA
| | - Roummel Marcia
- Department of Applied MathematicsUniversity of California MercedMercedCA95348USA
| |
Collapse
|
46
|
Li F, Youn J, Millsop C, Tagkopoulos I. Predicting clinical trial success for Clostridium difficile infections based on preclinical data. Front Artif Intell 2024; 7:1487335. [PMID: 39444663 PMCID: PMC11496251 DOI: 10.3389/frai.2024.1487335] [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: 08/27/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
Preclinical models are ubiquitous and essential for drug discovery, yet our understanding of how well they translate to clinical outcomes is limited. In this study, we investigate the translational success of treatments for Clostridium difficile infection from animal models to human patients. Our analysis shows that only 36% of the preclinical and clinical experiment pairs result in translation success. Univariate analysis shows that the sustained response endpoint is correlated with translation failure (SRC = -0.20, p-value = 1.53 × 10-54), and explainability analysis of multi-variate random forest models shows that both sustained response endpoint and subject age are negative predictors of translation success. We have developed a recommendation system to help plan the right preclinical study given factors such as drug dosage, bacterial dosage, and preclinical/clinical endpoint. With an accuracy of 0.76 (F1 score of 0.71) and by using only 7 features (out of 68 total), the proposed system boosts translational efficiency by 25%. The method presented can extend to any disease and can serve as a preclinical to clinical translation decision support system to accelerate drug discovery and de-risk clinical outcomes.
Collapse
Affiliation(s)
- Fangzhou Li
- Department of Computer Science, University of California, Davis, Davis, CA, United States
- Genome Center, University of California, Davis, Davis, CA, United States
- USDA/NSF AI Institute for Next Generation Food Systems, University of California, Davis, Davis, CA, United States
| | - Jason Youn
- Department of Computer Science, University of California, Davis, Davis, CA, United States
- Genome Center, University of California, Davis, Davis, CA, United States
- USDA/NSF AI Institute for Next Generation Food Systems, University of California, Davis, Davis, CA, United States
| | - Christian Millsop
- Department of Computer Science, University of California, Davis, Davis, CA, United States
- Genome Center, University of California, Davis, Davis, CA, United States
| | - Ilias Tagkopoulos
- Department of Computer Science, University of California, Davis, Davis, CA, United States
- Genome Center, University of California, Davis, Davis, CA, United States
- USDA/NSF AI Institute for Next Generation Food Systems, University of California, Davis, Davis, CA, United States
| |
Collapse
|
47
|
Zhang Y, Gao J, Xu Y, Liu J, Huang S, Li G, Yao B, Sun Z, Wang X. Investigation of cytochrome P450 inhibitory properties of deoxyshikonin, a bioactive compound from Lithospermum erythrorhizon Sieb. et Zucc. Phytother Res 2024; 38:4855-4864. [PMID: 36317387 DOI: 10.1002/ptr.7664] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 09/26/2022] [Accepted: 10/20/2022] [Indexed: 06/16/2023]
Abstract
Deoxyshikonin, a natural naphthoquinone compound extracted from Lithospermum erythrorhizon Sieb. et Zucc (Boraginaceae), has a wide range of pharmacological activities, including anti-tumor, anti-bacterial and wound healing effects. However, the inhibitory effect of deoxyshikonin on cytochrome P450 (CYP) remains unclear. This study investigated the potential inhibitory effects of deoxyshikonin on CYP1A2, 2B1/6, 2C9/11, 2D1/6, 2E1 and 3A2/4 enzymes in human and rat liver microsomes (HLMs and RLMs) by the cocktail approach in vitro. The single-point inactivation experiment showed that deoxyshikonin presented no time-dependent inhibition on CYP activities in HLMs and RLMs. Enzyme inhibition kinetics indicated that in HLMs, deoxyshikonin was not only a competitive inhibitor of CYP1A2 and 2E1, but also a mixed inhibitor of CYP2B6, 2C9, 2D6 and 3A4, with Ki of 2.21, 1.78, 1.68, 0.20, 4.08 and 0.44 μM, respectively. In RLMs, deoxyshikonin not only competitively inhibited CYP2B1 and 2E1, but also exhibited mixed inhibition on CYP1A2, 2C11, 2D1 and 3A2, with Ki values of no more than 18.66 μM. In conclusion, due to the low Ki values of deoxythiokonin on CYP enzymes in HLMs, this may lead to drug-drug interactions (DDI) and potential toxicity.
Collapse
Affiliation(s)
- Yuanjin Zhang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, People's Republic of China
| | - Jing Gao
- The College of Life Sciences, Northwest University, Xi'an, People's Republic of China
| | - Yuan Xu
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, People's Republic of China
| | - Jie Liu
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, People's Republic of China
| | - Shengbo Huang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, People's Republic of China
| | - Guihong Li
- Southern Medical University Affiliated Fengxian Hospital, Shanghai, People's Republic of China
| | - Bingyi Yao
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, People's Republic of China
| | - Zhenliang Sun
- Southern Medical University Affiliated Fengxian Hospital, Shanghai, People's Republic of China
| | - Xin Wang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, People's Republic of China
| |
Collapse
|
48
|
Lu J, Zhao J, Xie D, Ding J, Yu Q, Wang T. Use of a PK/PD Model to Select Cetagliptin Dosages for Patients with Type 2 Diabetes in Phase 3 Trials. Clin Pharmacokinet 2024; 63:1463-1476. [PMID: 39367290 DOI: 10.1007/s40262-024-01427-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND Cetagliptin is a novel dipeptidyl peptidase-4 (DPP-4) inhibitor developed for the treatment of patients with type 2 diabetes (T2D). Several phase 1 studies have been conducted in China. Modelling and simulation were used to obtain cetagliptin dose for phase 3 trials in T2D patients. METHODS A pharmacokinetic (PK)/pharmacodynamic (PD) model and model-based analysis of the relationship between hemoglobin A1c (HbA1c) and dosage was explored to guide dose selection of cetagliptin for phase 3 trials. The PK/PD data were derived from four phase 1 clinical studies, and sitagliptin 100 mg was employed as a positive control in studies 1, 3, and 4. RESULTS The PK profiles of cetagliptin were well described by a two-compartment model with first-order absorption, saturated efflux, and first-order elimination. The final PD model was a sigmoid maximum inhibitory efficacy (Emax) model with the Hill coefficient. The final model accurately captured cetagliptin PK/PD, demonstrated by goodness-of-fit plots. Based on weighted average inhibition (WAI), the relationship between HbA1c and dose was well displayed. Cetagliptin 50 mg once daily or above as monotherapy or as add-on therapy appeared more effective in HbA1c reduction than sitagliptin 100 mg. Cetagliptin 50 mg or 100 mg once daily was selected as the dose for phase 3 trials of cetagliptin in T2D patients. CONCLUSIONS The PK/PD model supports dose selection of cetagliptin for phase 3 trials. A model‑informed approach can be used to replace a dose-finding trial and accelerate cetagliptin's development.
Collapse
Affiliation(s)
- Jinmiao Lu
- CGeneTech (Suzhou, China) Co., Ltd., 218 Xinghu Street, Suzhou, 215123, China.
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China.
| | - Jiahong Zhao
- CGeneTech (Suzhou, China) Co., Ltd., 218 Xinghu Street, Suzhou, 215123, China
| | - Daosheng Xie
- Beijing Noahpharm Medical Technology Co., Ltd., Beijing, China
| | - Juping Ding
- CGeneTech (Suzhou, China) Co., Ltd., 218 Xinghu Street, Suzhou, 215123, China
| | - Qiang Yu
- CGeneTech (Suzhou, China) Co., Ltd., 218 Xinghu Street, Suzhou, 215123, China
| | - Tong Wang
- CGeneTech (Suzhou, China) Co., Ltd., 218 Xinghu Street, Suzhou, 215123, China.
| |
Collapse
|
49
|
Banerjee C, Tripathy D, Kumar D, Chakraborty J. Monoamine oxidase and neurodegeneration: Mechanisms, inhibitors and natural compounds for therapeutic intervention. Neurochem Int 2024; 179:105831. [PMID: 39128624 DOI: 10.1016/j.neuint.2024.105831] [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: 05/13/2024] [Revised: 07/26/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Mammalian flavoenzyme Monoamine oxidase (MAO) resides on the outer mitochondrial membrane (OMM) and it is involved in the metabolism of different monoamine neurotransmitters in brain. During MAO mediated oxidative deamination of relevant substrates, H2O2 is released as a catalytic by-product, thus serving as a major source of reactive oxygen species (ROS). Under normal conditions, MAO mediated ROS is reported to propel the functioning of mitochondrial electron transport chain and phasic dopamine release. However, due to its localization onto mitochondria, sudden elevation in its enzymatic activity could directly impact the form and function of the organelle. For instance, in the case of Parkinson's disease (PD) patients who are on l-dopa therapy, the enzyme could be a concurrent source of extensive ROS production in the presence of uncontrolled substrate (dopamine) availability, thus further impacting the health of surviving neurons. It is worth mentioning that the expression of the enzyme in different brain compartments increases with age. Moreover, the involvement of MAO in the progression of neurological disorders such as PD, Alzheimer's disease and depression has been extensively studied in recent times. Although the usage of available synthetic MAO inhibitors has been instrumental in managing these conditions, the associated complications have raised significant concerns lately. Natural products have served as a major source of lead molecules in modern-day drug discovery; however, there is still no FDA-approved MAO inhibitor which is derived from natural sources. In this review, we have provided a comprehensive overview of MAO and how the enzyme system is involved in the pathogenesis of different age-associated neuropathologic conditions. We further discussed the applications and drawbacks of the long-term usage of presently available synthetic MAO inhibitors. Additionally, we have highlighted the prospect and worth of natural product derived molecules in addressing MAO associated complications.
Collapse
Affiliation(s)
- Chayan Banerjee
- Cell Biology and Physiology Division, CSIR- Indian Institute of Chemical Biology, Kolkata, 700032, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Debasmita Tripathy
- Department of Zoology, Netaji Nagar College for Women, Kolkata, 700092, India
| | - Deepak Kumar
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, 700032, India.
| | - Joy Chakraborty
- Cell Biology and Physiology Division, CSIR- Indian Institute of Chemical Biology, Kolkata, 700032, India.
| |
Collapse
|
50
|
Shah P, Siramshetty VB, Mathé E, Xu X. Developing Robust Human Liver Microsomal Stability Prediction Models: Leveraging Inter-Species Correlation with Rat Data. Pharmaceutics 2024; 16:1257. [PMID: 39458588 PMCID: PMC11510424 DOI: 10.3390/pharmaceutics16101257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/03/2024] [Accepted: 09/19/2024] [Indexed: 10/28/2024] Open
Abstract
Objectives: Pharmacokinetic issues were the leading cause of drug attrition, accounting for approximately 40% of all cases before the turn of the century. To this end, several high-throughput in vitro assays like microsomal stability have been developed to evaluate the pharmacokinetic profiles of compounds in the early stages of drug discovery. At NCATS, a single-point rat liver microsomal (RLM) stability assay is used as a Tier I assay, while human liver microsomal (HLM) stability is employed as a Tier II assay. We experimentally screened and collected data on over 30,000 compounds for RLM stability and over 7000 compounds for HLM stability. Although HLM stability screening provides valuable insights, the increasing number of hits generated, along with the time- and resource-intensive nature of the assay, highlights the need for alternative strategies. One promising approach is leveraging in silico models trained on these experimental datasets. Methods: We describe the development of an HLM stability prediction model using our in-house HLM stability dataset. Results: Employing both classical machine learning methods and advanced techniques, such as neural networks, we achieved model accuracies exceeding 80%. Moreover, we validated our model using external test sets and found that our models are comparable to some of the best models in literature. Additionally, the strong correlation observed between our RLM and HLM data was further reinforced by the fact that our HLM model performance improved when using RLM stability predictions as an input descriptor. Conclusions: The best model along with a subset of our dataset (PubChem AID: 1963597) has been made publicly accessible on the ADME@NCATS website for the benefit of the greater drug discovery community. To the best of our knowledge, it is the largest open-source model of its kind and the first to leverage cross-species data.
Collapse
|