1
|
Elsaman T, Mohamed MA, Elderdery AY, Alsrhani A, Alzahrani B, Ghanem HB, Mills J, Rayzah M, Alzerwi NAN, Al-sultan A, Idrees B, Rayzah F. Pharmacophore-based virtual screening and in silico investigations of small molecule library for discovery of human hepatic ketohexokinase inhibitors for the treatment of fructose metabolic disorders. Front Pharmacol 2025; 16:1531512. [PMID: 40260383 PMCID: PMC12009819 DOI: 10.3389/fphar.2025.1531512] [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: 11/20/2024] [Accepted: 03/14/2025] [Indexed: 04/23/2025] Open
Abstract
Introduction Excessive fructose consumption is a significant driver of metabolic disorders, including obesity, diabetes, non-alcoholic fatty liver disease and non-alcoholic steatohepatitis primarily by promoting insulin resistance and fat accumulation. Ketohexokinase C (KHK-C), a pivotal enzyme in fructose metabolism, catalyzes the phosphorylation of fructose to fructose-1-phosphate, initiating a cascade of downstream metabolic processes. In contrast to glucose metabolism, KHK-C lacks negative feedback regulation, allowing the continuous phosphorylation of fructose, which leads to heightened levels of glucose, glycogen, and triglycerides in the bloodstream and liver. While targeting KHK-C offers a promising therapeutic avenue, no drugs have yet been approved for clinical use. Pfizer's PF-06835919 has progressed to phase II trials, demonstrating a reduction in liver fat and improved insulin sensitivity, while Eli Lilly's LY-3522348 also shows significant potential. Nonetheless, there remains a critical need for the development of novel KHK-C inhibitors that offer improved pharmacokinetics, enhanced efficacy, and superior safety profiles. Methods In the present study, a comprehensive computational strategy was employed to screen 460,000 compounds from the National Cancer Institute library for potential KHK-C inhibitors. Initially, pharmacophore-based virtual screening was used to identify potential hits, followed by multi-level molecular docking, binding free energy estimation, pharmacokinetic analysis, and molecular dynamics (MD) simulations to further evaluate the compounds. This multi-step approach aimed to identify compounds with strong binding affinity, favorable pharmacokinetic profiles, and high potential for efficacy as KHK-C inhibitors. Results Ten compounds exhibited docking scores ranging from -7.79 to -9.10 kcal/mol, surpassing those of the compounds currently undergoing clinical trials, PF-06835919 (-7.768 kcal/mol) and LY-3522348 (-6.54 kcal/mol). Their calculated binding free energies ranged from -57.06 to -70.69 kcal/mol, further demonstrating their superiority over PF-06835919 (-56.71 kcal/mol) and LY-3522348 (-45.15 kcal/mol). ADMET profiling refined the selection to five compounds (1, 2, and 4-6), and molecular dynamics simulations identified compound 2 as the most stable and promising candidate compared to the clinical candidate PF-06835919. Conclusion These findings highlight compound 2 as a potent KHK-C inhibitor with predicted pharmacokinetics and toxicity profiles supporting its potential for treating fructose-driven metabolic disorders, warranting further validation.
Collapse
Affiliation(s)
- Tilal Elsaman
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Saudi Arabia
| | - Magdi Awadalla Mohamed
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Saudi Arabia
| | - Abozer Y. Elderdery
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Abdullah Alsrhani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Badr Alzahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Heba Bassiony Ghanem
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Jeremy Mills
- School of Medicine, Pharmacy and Biomedical Sciences, Uinversity of Portsmouth, Portsmouth, United Kingdom
| | - Musaed Rayzah
- Department of Surgery, College of Medicine, Majmaah University, Al Majma’ah, Saudi Arabia
| | - Nasser A. N. Alzerwi
- Department of Surgery, College of Medicine, Majmaah University, Al Majma’ah, Saudi Arabia
| | - Afnan Al-sultan
- Department of Surgery, King Saud Medical City, Riyadh, Saudi Arabia
| | - Bandar Idrees
- Department of Surgery, Prince Sultan Military Medical City, As Sulimaniyah, Saudi Arabia
| | - Fares Rayzah
- Department of Surgery, Aseer Central Hospital, Abha, Saudi Arabia
| |
Collapse
|
2
|
Yoo J, Jang W, Shin WH. From part to whole: AI-driven progress in fragment-based drug discovery. Curr Opin Struct Biol 2025; 91:102995. [PMID: 39970579 DOI: 10.1016/j.sbi.2025.102995] [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: 09/30/2024] [Revised: 12/23/2024] [Accepted: 01/14/2025] [Indexed: 02/21/2025]
Abstract
Fragment-based drug discovery is a technique that finds potent binding fragments to the binding hotspots and makes them a hit compound. The combination of fragments allows us to explore the large chemical space. Thus, it becomes an effective methodology for identifying lead compounds. Three concepts have been introduced to make the fragments into the compound: growing, merging, and linking. Recently, growing and merging techniques using AI have significantly improved the accuracy and efficiency of molecular design. In this review, recent techniques such as VAE, reinforcement learning, and SE(3)-equivariant models will be discussed. These methods enable precise molecular structure exploration and optimization. Additionally, we address techniques utilizing diffusion models, language models, and deep evolutionary learning. We also introduce linker optimization methods using reinforcement learning and deep learning-based models. This progress of fragment-based drug discovery methods with AI opens the possibility of discovering the vast chemical space with high efficiency.
Collapse
Affiliation(s)
- Jinhyeok Yoo
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, 02708, Republic of Korea
| | - Wonkyeong Jang
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, 02708, Republic of Korea
| | - Woong-Hee Shin
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, 02708, Republic of Korea.
| |
Collapse
|
3
|
Tiwari K, Kumar B, Tiwari A, Dhamija P, Vardhan G, Dehade A, Kumar V. In Silico Analysis of Saroglitazar and Ferulic Acid Binding to Human Ketohexokinase: Implications for Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Cureus 2025; 17:e79437. [PMID: 40130107 PMCID: PMC11931454 DOI: 10.7759/cureus.79437] [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] [Accepted: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease renamed as metabolic dysfunction-associated steatotic liver disease (MASLD) and a global health issue that causes excessive liver fat deposition without alcohol usage. Basic fatty liver to non-alcoholic steatohepatitis can lead to liver fibrosis, cirrhosis, and hepatocellular carcinoma. Role of research is vital due to the multifaceted, complex pathophysiology and the increasing incidence of a sedentary lifestyle. Computational network pharmacology, docking and dynamics studies of saroglitazar and ferulic acid with human ketohexokinase (KHK) were conducted to propose potential MASLD management. METHOD Utilized computational methodologies were utilized to examine binding interactions of saroglitazar (compound identifier (CID): 60151560) and ferulic acid (CID: 445858) with human ketohexokinase (KHK: P50053, Protein Data Bank (PDB) ID: 6W0W). Active site analysis was done by using the Conserved Domain Database (CDD) server (Collaborative Drug Discovery, Burlingame, California) and BIOVIA Discovery Studio 2019 (Dassault Systèmes, Vélizy-Villacoublay, France). The best PDB complex was used for molecular dynamics simulation and trajectory analysis on 100 ns, and functional associations were checked based on network analysis using the Search Tool for Interactions of Chemicals (STITCH) server (STITCH Consortium (EMBL), Heidelberg, Germany). RESULTS Human ketohexokinase (KHK) protein (UniProt ID: P50053) was obtained. Additional KHK PDB Structure (6W0W) was retrieved for docking calculation. PubChem Database 2 Structure-Data File (SDF) files (National Center for Biotechnology Information (NCBI), U.S. National Library of Medicine, Bethesda, Maryland), ferulic acid (CID: 445858) and saroglitazar (CID: 60151560) were used as ligands. Active site residues were identified using the CDD server and BIOVIA Discovery Studio 2019. Further, identified active site residues, i.e., Arg108, Trp225, Glu227, Gly229, Ala230, Pro246, Pro247, Val250, Thr253, Gly257, Cys282, Gly286, and Cys289 were used as potential active site for docking. D. E. Shaw Research Molecular Dynamics (DESMOND, Schrödinger, Inc., New York) was used for molecular dynamics simulation and trajectory analysis equilibrated after 40 ns in best-docked complex (saroglitazar (CID: 60151560) and KHK; binding energy: -21 kcal/mol). CONCLUSION The study shows that saroglitazar and ferulic acid are potent KHK inhibitors for metabolic diseases, including MASLD, suggesting multi-target treatments.
Collapse
Affiliation(s)
- Kalpana Tiwari
- Pharmacology, Institute of Medical Science, Banaras Hindu University, Varanasi, IND
| | - Brijesh Kumar
- Pharmacology and Therapeutics, Institute of Medical Science, Banaras Hindu University, Varanasi, IND
| | - Anurag Tiwari
- Gastroenterology, Institute of Medical Science, Banaras Hindu University, Varanasi, IND
| | - Puneet Dhamija
- Clinical Pharmacology, All India Institute of Medical Sciences, Rishikesh, IND
| | - Gyan Vardhan
- Pharmacology, All India Institute of Medical Sciences, Rishikesh, IND
| | - Amol Dehade
- Pharmacology, Institute of Medical Science, Banaras Hindu University, Varanasi, IND
| | - Vinay Kumar
- School of Biotechnology, Center for Bioinformatics, Institute of Medical Science, Banaras Hindu University, Varanasi, IND
| |
Collapse
|
4
|
Elsaman T, Mohamed MA. Examining Prenylated Xanthones as Potential Inhibitors Against Ketohexokinase C Isoform for the Treatment of Fructose-Driven Metabolic Disorders: An Integrated Computational Approach. Pharmaceuticals (Basel) 2025; 18:126. [PMID: 39861187 PMCID: PMC11768319 DOI: 10.3390/ph18010126] [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: 01/05/2025] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Fructose-driven metabolic disorders, such as obesity, non-alcoholic fatty liver disease (NAFLD), dyslipidemia, and type 2 diabetes, are significant global health challenges. Ketohexokinase C (KHK-C), a key enzyme in fructose metabolism, is a promising therapeutic target. α-Mangostin, a naturally occurring prenylated xanthone, has been identified as an effective KHK-C inhibitor, prompting exploration of its analogs for enhanced efficacy. This study aimed to identify α-Mangostin analogs with improved inhibitory properties against KHK-C to address these disorders. Methods: A library of 1383 analogs was compiled from chemical databases and the literature. Molecular docking, binding free energy calculations, pharmacokinetic assessments, molecular dynamics simulations, and quantum mechani-cal analyses were used to screen and evaluate the compounds. α-Mangostin's binding affinity (37.34 kcal/mol) served as the benchmark. Results: Sixteen analogs demonstrated binding affinities superior to α-Mangostin (from -45.51 to -61.3 kcal/mol), LY-3522348 (-45.36 kcal/mol), and reported marine-derived inhibitors (from -22.74 to -51.83 kcal/mol). Hits 7, 8, 9, 13, and 15 not only surpassed these benchmarks in binding affinity, but also exhibited superior pharmacokinetic properties compared to α-Mangostin, LY-3522348, and marine-derived inhibitors, indicating strong in vivo potential. Among these, hit 8 emerged as the best performer, achieving a binding free energy of -61.30 kcal/mol, 100% predicted oral absorption, enhanced metabolic stability, and stable molecular dynamics. Conclusions: Hit 8 emerged as the most promising candidate due to its superior binding affinity, favorable pharmacokinetics, and stable interactions with KHK-C. These findings highlight its potential for treating fructose-driven metabolic disorders, warranting further experimental validation.
Collapse
Affiliation(s)
- Tilal Elsaman
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia
| | - Magdi Awadalla Mohamed
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia
| |
Collapse
|
5
|
Heine N, Weber A, Pautsch A, Gottschling D, Uphues I, Bauer M, Ebenhoch R, Magarkar A, Nosse B, Kley JT. Discovery of BI-9787, a potent zwitterionic ketohexokinase inhibitor with oral bioavailability. Bioorg Med Chem Lett 2024; 112:129930. [PMID: 39179180 DOI: 10.1016/j.bmcl.2024.129930] [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: 06/04/2024] [Revised: 08/01/2024] [Accepted: 08/15/2024] [Indexed: 08/26/2024]
Abstract
Fructose metabolism by ketohexokinase (KHK) is implicated in a variety of metabolic disorders. KHK inhibition is a potential therapeutic strategy for the treatment of diseases including diabetes, non-alcoholic fatty liver disease, and non-alcoholic steatohepatitis. The first small-molecule KHK-inhibitors have entered clinical trials, but it remains unclear if systemic inhibition of KHK by small-molecules will eventually benefit patients. Here we report the discovery of BI-9787, a potent, zwitterionic KHK inhibitor characterized by high permeability and favorable oral rat pharmacokinetics. BI-9787 was identified by optimizing chemical starting points generated via a ligand-based virtual screening of Boehringer's virtual library of synthetically accessible compounds (BICLAIM). It serves as a high-quality in vitro and in vivo tool compound for investigating the role of fructose metabolism in disease.
Collapse
Affiliation(s)
- Niklas Heine
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Medicinal Chemistry, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany
| | - Alexander Weber
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Medicinal Chemistry, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany
| | - Alexander Pautsch
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Medicinal Chemistry, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany
| | - Dirk Gottschling
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Medicinal Chemistry, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany
| | - Ingo Uphues
- Boehringer Ingelheim Pharma GmbH & Co. KG, Cardiometabolic Diseases Research, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany
| | - Margit Bauer
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Medicinal Chemistry, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany
| | - Rebecca Ebenhoch
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Medicinal Chemistry, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany
| | - Aniket Magarkar
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Medicinal Chemistry, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany
| | - Bernd Nosse
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Medicinal Chemistry, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany; Boehringer Ingelheim International GmbH, Business Development & Licensing, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany
| | - Jörg Thomas Kley
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Medicinal Chemistry, Birkendorfer Straße 65, 88397 Biberach an der Riß, Germany.
| |
Collapse
|
6
|
Alturki MS. Exploring Marine-Derived Compounds: In Silico Discovery of Selective Ketohexokinase (KHK) Inhibitors for Metabolic Disease Therapy. Mar Drugs 2024; 22:455. [PMID: 39452863 PMCID: PMC11509851 DOI: 10.3390/md22100455] [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: 09/06/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/26/2024] Open
Abstract
The increasing prevalence of metabolic diseases, including nonalcoholic fatty liver disease (NAFLD), obesity, and type 2 diabetes, poses significant global health challenges. Ketohexokinase (KHK), an enzyme crucial in fructose metabolism, is a potential therapeutic target due to its role in these conditions. This study focused on the discovery of selective KHK inhibitors using in silico methods. We employed structure-based drug design (SBDD) and ligand-based drug design (LBDD) approaches, beginning with molecular docking to identify promising compounds, followed by induced-fit docking (IFD), molecular mechanics generalized Born and surface area continuum solvation (MM-GBSA), and molecular dynamics (MD) simulations to validate binding affinities. Additionally, shape-based screening was conducted to assess structural similarities. The findings highlight several potential inhibitors with favorable ADMET profiles, offering promising candidates for further development in the treatment of fructose-related metabolic disorders.
Collapse
Affiliation(s)
- Mansour S Alturki
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| |
Collapse
|
7
|
Durham TB, Hao J, Spinazze P, Stack DR, Toth JL, Massey S, Mbofana CT, Johnston RD, Lineswala JP, Wrobleski A, Mínguez JM, Perez C, Smith DL, Lamar J, Leon R, Corkins C, Durbin J, Tung F, Guo S, Linder RJ, Yumibe N, Wang W, MacKrell J, Antonellis M, Mascaro B. Identification of LY3522348: A Highly Selective and Orally Efficacious Ketohexokinase Inhibitor. J Med Chem 2023; 66:15960-15976. [PMID: 37992274 DOI: 10.1021/acs.jmedchem.3c01410] [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] [Indexed: 11/24/2023]
Abstract
The identification of clinical candidate LY3522348 (compound 23) is described. LY3522348 is a highly selective, oral dual inhibitor of human ketohexokinase isoforms C and A (hKHK-C, hKHK-A). Optimization began with highly efficient (S)-2-(2-methylazetidin-1-yl)-6-(1H-pyrazol-4-yl)-4-(trifluoromethyl)nicotinonitrile (3). Efforts focused on developing absorption, distribution, metabolism, potency, and in vitro safety profiles to support oral QD dosing in patients. Structure-based design leveraged vectors for substitution of the pyrazole ring, which provided an opportunity to interact with several different proximal amino acid residues in the protein. LY3522348 displayed a robust pharmacodynamic response in a mouse model of fructose metabolism and was advanced into clinical trials.
Collapse
Affiliation(s)
- Timothy B Durham
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - Junliang Hao
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - Patrick Spinazze
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - Douglas R Stack
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - James L Toth
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - Steven Massey
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - Curren T Mbofana
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - Richard D Johnston
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - Jayana P Lineswala
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - Aaron Wrobleski
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - Jose Miguel Mínguez
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly SA, Avenida de la Industria 30, 28108 Alcobendas, Madrid, Spain
| | - Carlos Perez
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly SA, Avenida de la Industria 30, 28108 Alcobendas, Madrid, Spain
| | - Daryl L Smith
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - Jason Lamar
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - Rebecca Leon
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
- Molecular Pharmacology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - Christopher Corkins
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
- Molecular Pharmacology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - Jim Durbin
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
- Structural Biology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - Frances Tung
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
- Structural Biology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - Sherry Guo
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
- Structural Biology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - Ryan J Linder
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
- Molecular Innovation Hub, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - Nathan Yumibe
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
- ADME, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - Wei Wang
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
- Toxicology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - James MacKrell
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
- Diabetes and Metabolic Research, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - Meghan Antonellis
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
- Diabetes and Metabolic Research, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| | - Bethany Mascaro
- Discovery Chemistry Research and Technology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
- Diabetes and Metabolic Research, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
| |
Collapse
|
8
|
Zhu G, Li J, Lin X, Zhang Z, Hu T, Huo S, Li Y. Discovery of a Novel Ketohexokinase Inhibitor with Improved Drug Distribution in Target Tissue for the Treatment of Fructose Metabolic Disease. J Med Chem 2023; 66:13501-13515. [PMID: 37766386 DOI: 10.1021/acs.jmedchem.3c00715] [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: 09/29/2023]
Abstract
Excessive fructose absorption and its subsequent metabolisms are implicated in nonalcoholic fatty liver disease, obesity, and insulin resistance in humans. Ketohexokinase (KHK) is a primary enzyme involved in fructose metabolism via the conversion of fructose to fructose-1-phosphate. KHK inhibition might be a potential approach for the treatment of metabolic disorders. Herein, a series of novel KHK inhibitors were designed, synthesized, and evaluated. Among them, compound 14 exhibited more potent activity than PF-06835919 based on the rat KHK inhibition assay in vivo, and higher drug distribution concentration in the liver. Its good absorption, distribution, metabolism, and excretion and pharmacokinetic properties make it a promising clinical candidate.
Collapse
Affiliation(s)
- Guodong Zhu
- TuoJie Biotech (Shanghai) Co., Ltd., Shanghai 201206, P. R. China
| | - Jiao Li
- TuoJie Biotech (Shanghai) Co., Ltd., Shanghai 201206, P. R. China
| | - Xiaoyan Lin
- TuoJie Biotech (Shanghai) Co., Ltd., Shanghai 201206, P. R. China
| | - Zhen Zhang
- TuoJie Biotech (Shanghai) Co., Ltd., Shanghai 201206, P. R. China
| | - Tao Hu
- TuoJie Biotech (Shanghai) Co., Ltd., Shanghai 201206, P. R. China
| | - Shuhua Huo
- TuoJie Biotech (Shanghai) Co., Ltd., Shanghai 201206, P. R. China
| | - Yunfei Li
- TuoJie Biotech (Shanghai) Co., Ltd., Shanghai 201206, P. R. China
| |
Collapse
|
9
|
Park SH, Helsley RN, Fadhul T, Willoughby JLS, Noetzli L, Tu HC, Solheim MH, Fujisaka S, Pan H, Dreyfuss JM, Bons J, Rose J, King CD, Schilling B, Lusis AJ, Pan C, Gupta M, Kulkarni RN, Fitzgerald K, Kern PA, Divanovic S, Kahn CR, Softic S. Fructose induced KHK-C can increase ER stress independent of its effect on lipogenesis to drive liver disease in diet-induced and genetic models of NAFLD. Metabolism 2023; 145:155591. [PMID: 37230214 PMCID: PMC10752375 DOI: 10.1016/j.metabol.2023.155591] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 05/04/2023] [Accepted: 05/10/2023] [Indexed: 05/27/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a liver manifestation of metabolic syndrome, and is estimated to affect one billion individuals worldwide. An increased intake of a high-fat diet (HFD) and sugar-sweetened beverages are risk-factors for NAFLD development, but how their combined intake promotes progression to a more severe form of liver injury is unknown. Here we show that fructose metabolism via ketohexokinase (KHK) C isoform leads to unresolved endoplasmic reticulum (ER) stress when coupled with a HFD intake. Conversely, a liver-specific knockdown of KHK in mice consuming fructose on a HFD is adequate to improve the NAFLD activity score and exert a profound effect on the hepatic transcriptome. Overexpression of KHK-C in cultured hepatocytes is sufficient to induce ER stress in fructose free media. Upregulation of KHK-C is also observed in mice with genetically induced obesity or metabolic dysfunction, whereas KHK knockdown in these mice improves metabolic function. Additionally, in over 100 inbred strains of male or female mice hepatic KHK expression correlates positively with adiposity, insulin resistance, and liver triglycerides. Similarly, in 241 human subjects and their controls, hepatic Khk expression is upregulated in early, but not late stages of NAFLD. In summary, we describe a novel role of KHK-C in triggering ER stress, which offers a mechanistic understanding of how the combined intake of fructose and a HFD propagates the development of metabolic complications.
Collapse
Affiliation(s)
- Se-Hyung Park
- Department of Pediatrics, Division of Pediatric Gastroenterology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Robert N Helsley
- Department of Pediatrics, Division of Pediatric Gastroenterology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Taghreed Fadhul
- Department of Pediatrics, Division of Pediatric Gastroenterology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | | | - Leila Noetzli
- Alnylam Pharmaceuticals Inc., Cambridge, MA 02142, USA
| | - Ho-Chou Tu
- Alnylam Pharmaceuticals Inc., Cambridge, MA 02142, USA
| | - Marie H Solheim
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, 50931 Cologne, Germany
| | - Shiho Fujisaka
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; First Department of Internal Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Hui Pan
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Jonathan M Dreyfuss
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Joanna Bons
- Proteomics and Aging Center, Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Jacob Rose
- Proteomics and Aging Center, Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Christina D King
- Proteomics and Aging Center, Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Birgit Schilling
- Proteomics and Aging Center, Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Aldons J Lusis
- Department of Medicine/Division of Cardiology, Department of Human Genetics, A2-237 Center for the Health Sciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Calvin Pan
- Department of Medicine/Division of Cardiology, Department of Human Genetics, A2-237 Center for the Health Sciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Manoj Gupta
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Rohit N Kulkarni
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215, USA
| | | | - Philip A Kern
- Department of Medicine, Division of Endocrinology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - C Ronald Kahn
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Samir Softic
- Department of Pediatrics, Division of Pediatric Gastroenterology, University of Kentucky College of Medicine, Lexington, KY 40536, USA; Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, USA.
| |
Collapse
|
10
|
Helsley RN, Park SH, Vekaria HJ, Sullivan PG, Conroy LR, Sun RC, Romero MDM, Herrero L, Bons J, King CD, Rose J, Meyer JG, Schilling B, Kahn CR, Softic S. Ketohexokinase-C regulates global protein acetylation to decrease carnitine palmitoyltransferase 1a-mediated fatty acid oxidation. J Hepatol 2023; 79:25-42. [PMID: 36822479 PMCID: PMC10679901 DOI: 10.1016/j.jhep.2023.02.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/25/2023]
Abstract
BACKGROUND & AIMS The consumption of sugar and a high-fat diet (HFD) promotes the development of obesity and metabolic dysfunction. Despite their well-known synergy, the mechanisms by which sugar worsens the outcomes associated with a HFD are largely elusive. METHODS Six-week-old, male, C57Bl/6 J mice were fed either chow or a HFD and were provided with regular, fructose- or glucose-sweetened water. Moreover, cultured AML12 hepatocytes were engineered to overexpress ketohexokinase-C (KHK-C) using a lentivirus vector, while CRISPR-Cas9 was used to knockdown CPT1α. The cell culture experiments were complemented with in vivo studies using mice with hepatic overexpression of KHK-C and in mice with liver-specific CPT1α knockout. We used comprehensive metabolomics, electron microscopy, mitochondrial substrate phenotyping, proteomics and acetylome analysis to investigate underlying mechanisms. RESULTS Fructose supplementation in mice fed normal chow and fructose or glucose supplementation in mice fed a HFD increase KHK-C, an enzyme that catalyzes the first step of fructolysis. Elevated KHK-C is associated with an increase in lipogenic proteins, such as ACLY, without affecting their mRNA expression. An increase in KHK-C also correlates with acetylation of CPT1α at K508, and lower CPT1α protein in vivo. In vitro, KHK-C overexpression lowers CPT1α and increases triglyceride accumulation. The effects of KHK-C are, in part, replicated by a knockdown of CPT1α. An increase in KHK-C correlates negatively with CPT1α protein levels in mice fed sugar and a HFD, but also in genetically obese db/db and lipodystrophic FIRKO mice. Mechanistically, overexpression of KHK-C in vitro increases global protein acetylation and decreases levels of the major cytoplasmic deacetylase, SIRT2. CONCLUSIONS KHK-C-induced acetylation is a novel mechanism by which dietary fructose augments lipogenesis and decreases fatty acid oxidation to promote the development of metabolic complications. IMPACT AND IMPLICATIONS Fructose is a highly lipogenic nutrient whose negative consequences have been largely attributed to increased de novo lipogenesis. Herein, we show that fructose upregulates ketohexokinase, which in turn modifies global protein acetylation, including acetylation of CPT1a, to decrease fatty acid oxidation. Our findings broaden the impact of dietary sugar beyond its lipogenic role and have implications on drug development aimed at reducing the harmful effects attributed to sugar metabolism.
Collapse
Affiliation(s)
- Robert N Helsley
- Department of Pediatrics and Gastroenterology, University of Kentucky, Lexington, KY, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA; Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA; Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Se-Hyung Park
- Department of Pediatrics and Gastroenterology, University of Kentucky, Lexington, KY, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Hemendra J Vekaria
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
| | - Patrick G Sullivan
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
| | - Lindsey R Conroy
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Ramon C Sun
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA; Department of Biochemistry & Molecular Biology, University of Florida, Gainesville, FL, USA; Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - María Del Mar Romero
- School of Pharmacy, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, 08028, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Laura Herrero
- School of Pharmacy, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, 08028, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Joanna Bons
- Chemistry & Mass Spectrometry, Buck Institute for Research on Aging, Novato, CA, USA
| | - Christina D King
- Chemistry & Mass Spectrometry, Buck Institute for Research on Aging, Novato, CA, USA
| | - Jacob Rose
- Chemistry & Mass Spectrometry, Buck Institute for Research on Aging, Novato, CA, USA
| | - Jesse G Meyer
- Chemistry & Mass Spectrometry, Buck Institute for Research on Aging, Novato, CA, USA; Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Birgit Schilling
- Chemistry & Mass Spectrometry, Buck Institute for Research on Aging, Novato, CA, USA
| | - C Ronald Kahn
- Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Samir Softic
- Department of Pediatrics and Gastroenterology, University of Kentucky, Lexington, KY, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA; Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Park SH, Helsley RN, Fadhul T, Willoughby JL, Noetzli L, Tu HC, Solheim MH, Fujisaka S, Pan H, Dreyfuss JM, Bons J, Rose J, King CD, Schilling B, Lusis AJ, Pan C, Gupta M, Kulkarni RN, Fitzgerald K, Kern PA, Divanovic S, Kahn CR, Softic S. Fructose Induced KHK-C Increases ER Stress and Modulates Hepatic Transcriptome to Drive Liver Disease in Diet-Induced and Genetic Models of NAFLD. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.27.525605. [PMID: 36747758 PMCID: PMC9900898 DOI: 10.1101/2023.01.27.525605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a liver manifestation of metabolic syndrome, and is estimated to affect one billion individuals worldwide. An increased intake of a high-fat diet (HFD) and sugar-sweetened beverages are risk-factors for NAFLD development, but how their combined intake promotes progression to a more severe form of liver injury is unknown. Here we show that fructose metabolism via ketohexokinase (KHK) C isoform increases endoplasmic reticulum (ER) stress in a dose dependent fashion, so when fructose is coupled with a HFD intake it leads to unresolved ER stress. Conversely, a liver-specific knockdown of KHK in C57BL/6J male mice consuming fructose on a HFD is adequate to improve the NAFLD activity score and exert a profound effect on the hepatic transcriptome. Overexpression of KHK-C in cultured hepatocytes is sufficient to induce ER stress in fructose free media. Upregulation of KHK-C is also observed in genetically obesity ob/ob, db/db and lipodystrophic FIRKO male mice, whereas KHK knockdown in these mice improves metabolic function. Additionally, in over 100 inbred strains of male or female mice hepatic KHK expression correlates positively with adiposity, insulin resistance, and liver triglycerides. Similarly, in 241 human subjects and their controls, hepatic Khk expression is upregulated in early, but not late stages of NAFLD. In summary, we describe a novel role of KHK-C in triggering ER stress, which offers a mechanistic understanding of how the combined intake of fructose and a HFD propagates the development of metabolic complications.
Collapse
Affiliation(s)
- Se-Hyung Park
- Department of Pediatrics, Division of Pediatric Gastroenterology, University of Kentucky College of Medicine, Lexington, KY. 40536
| | - Robert N. Helsley
- Department of Pediatrics, Division of Pediatric Gastroenterology, University of Kentucky College of Medicine, Lexington, KY. 40536
| | - Taghreed Fadhul
- Department of Pediatrics, Division of Pediatric Gastroenterology, University of Kentucky College of Medicine, Lexington, KY. 40536
| | | | | | - Ho-Chou Tu
- Alnylam Pharmaceuticals Inc., Cambridge, MA. 02142
| | - Marie H. Solheim
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA. 02215
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, 50931 Cologne, Germany
| | - Shiho Fujisaka
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA. 02215
- First Department of Internal Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Hui Pan
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Jonathan M. Dreyfuss
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Joanna Bons
- Proteomics and Aging Center, Buck Institute for Research on Aging, Novato, CA 94945
| | - Jacob Rose
- Proteomics and Aging Center, Buck Institute for Research on Aging, Novato, CA 94945
| | - Christina D. King
- Proteomics and Aging Center, Buck Institute for Research on Aging, Novato, CA 94945
| | - Birgit Schilling
- Proteomics and Aging Center, Buck Institute for Research on Aging, Novato, CA 94945
| | - Aldons J. Lusis
- Department of Medicine/Division of Cardiology, Department of Human Genetics, A2-237 Center for the Health Sciences, University of California, Los Angeles, Los Angeles, CA USA
| | - Calvin Pan
- Department of Medicine/Division of Cardiology, Department of Human Genetics, A2-237 Center for the Health Sciences, University of California, Los Angeles, Los Angeles, CA USA
| | - Manoj Gupta
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center; Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215
| | - Rohit N. Kulkarni
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center; Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215
| | | | - Philip A. Kern
- Department of Medicine, Division of Endocrinology, University of Kentucky College of Medicine, Lexington, KY. 40536
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229
| | - C. Ronald Kahn
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA. 02215
| | - Samir Softic
- Department of Pediatrics, Division of Pediatric Gastroenterology, University of Kentucky College of Medicine, Lexington, KY. 40536
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA. 02215
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY. 40536
| |
Collapse
|
12
|
Kibou Z, Aissaoui N, Daoud I, Seijas JA, Vázquez-Tato MP, Klouche Khelil N, Choukchou-Braham N. Efficient Synthesis of 2-Aminopyridine Derivatives: Antibacterial Activity Assessment and Molecular Docking Studies. Molecules 2022; 27:3439. [PMID: 35684377 PMCID: PMC9182143 DOI: 10.3390/molecules27113439] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/22/2022] [Accepted: 05/23/2022] [Indexed: 11/16/2022] Open
Abstract
A new and suitable multicomponent one-pot reaction was developed for the synthesis of 2-amino-3-cyanopyridine derivatives. BACKGROUND This synthesis was demonstrated by the efficient and easy access to a variety of substituted 2-aminopyridines using enaminones as key precursors under solvent-free conditions. METHODS A range of spectroscopic techniques was used to determine and confirm the chemical structures (FTIR, 1H NMR, 13C NMR). The antimicrobial potency of synthesized compounds (2a-d) was tested using disk diffusion assays, and the Minimum Inhibitory Concentration (MIC) for the active compounds was determined against a panel of microorganisms, including Gram-positive and Gram-negative bacteria and yeasts. Moreover, a docking analysis was conducted by Molecular Operating Environment (MOE) software to provide supplementary information about the potential, as well as an ADME-T prediction to describe the pharmacokinetic properties of the best compound and its toxicity. RESULTS The results of the antimicrobial activity indicated that compound 2c showed the highest activity against Gram-positive bacteria, particularly S. aureus and B. subtilis whose MIC values were 0.039 ± 0.000 µg·mL-1. The results of the theoretical study of compound 2c were in line with the experimental data and exhibited excellent antibacterial potential. CONCLUSIONS On the basis of the obtained results, compound 2c can be used as an antibacterial agent model with high antibacterial potency.
Collapse
Affiliation(s)
- Zahira Kibou
- Laboratoire de Catalyse et Synthèse en Chimie Organique, Faculté des Sciences, Université de Tlemcen, BP 119, Tlemcen 13000, Algeria;
- Faculté des Sciences et de la Technologie, Université de Ain Témouchent, BP 284, Ain Témouchent 46000, Algeria
| | - Nadia Aissaoui
- Laboratory of the Sustainable Management of Natural Resources in Arid and Semi Aridareas, University Center Salhi Ahmed Naama, BP 66, Naama 45000, Algeria;
- Department of Biology, Faculty of Nature and Life, Earth and Universe Sciences, University of Tlemcen, Tlemcen 13000, Algeria
| | - Ismail Daoud
- Département des Sciences de la Matière, Université de Mohamed Khider, BP 145 RP, Biskra 07000, Algerie;
- Laboratory of Natural and Bio-Active Substances, Faculty of Sciences, University of Tlemcen, BP 119, Tlemcen 13000, Algeria
| | - Julio A. Seijas
- Departamento de Química Orgánica, Facultad de Ciencias, Universidad de Santiago de Compostela, A da, Alfonso X El Sabio s/n, 27002 Lugo, Spain;
| | - María Pilar Vázquez-Tato
- Departamento de Química Orgánica, Facultad de Ciencias, Universidad de Santiago de Compostela, A da, Alfonso X El Sabio s/n, 27002 Lugo, Spain;
| | - Nihel Klouche Khelil
- Laboratory of Applied Microbiology in Food, Biomedical and Environment (LAMAABE), Department of Biology, Faculty of Nature and Life, Earth and Universe Sciences, University of Tlemcen, Tlemcen 13000, Algeria;
- Laboratory of Experimental Surgery, Medical Faculty, University of Tlemcen, Tlemcen 13000, Algeria
| | - Noureddine Choukchou-Braham
- Laboratoire de Catalyse et Synthèse en Chimie Organique, Faculté des Sciences, Université de Tlemcen, BP 119, Tlemcen 13000, Algeria;
| |
Collapse
|
13
|
Li F, Xing G, Cousineau C, Clemens S, Mofikoya M, Kim MY, Zhang JY, Zhang Y, Raha N. Development and validation of a HILIC-MS/MS method for the quantitation of fructose in human urine in support of clinical programs. J Pharm Biomed Anal 2022; 208:114462. [PMID: 34798390 DOI: 10.1016/j.jpba.2021.114462] [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: 04/08/2021] [Revised: 10/20/2021] [Accepted: 11/02/2021] [Indexed: 10/19/2022]
Abstract
In a previous publication [1], a 20-minute UPLC®-MS/MS method, employing a surrogate analyte approach, was developed and validated to measure fructose and sorbitol, as mechanistic biomarkers, in human plasma to support first-in-human (FIH) studies. Different from plasma which maintains its homeostasis, urine has no such homeostasis mechanisms [2], therefore it is expected to be able to accommodate more changes. Here we describe the development and validation of a LC-MS/MS method for the quantiation of fructose in human urine to support clinical trials. A hydrophilic interaction chromatography (HILIC) method using an Asahipak NH2P-50 column (Shodex, 4.6 × 250 mm, 5 µm) was developed. Acetone precipitation was utilized to extract fructose from urine. For validation, stable isotope-labeled 13C6-fructose was used as the surrogate analyte for fructose in the preparation of calibration curves. QCs were prepared using both the surrogate analyte (13C6-fructose) and the authentic analyte (fructose). Difficulties were encountered for post-extraction stability experiments especially for authentic fructose QCs at low concentrations. Extensive troubleshooting revealed that fructose's chromatography improved as the column aged. As a result, the response factor of fructose increased over time for low concentration samples, leading to failed post-extraction stability experiments. A column cleaning procedure was implemented to ensure consistency in chromatography performance. The HILIC-MS/MS method was successfully validated and applied to analyze clinical samples with a 91% overall run passing rate.
Collapse
Affiliation(s)
- Fumin Li
- PPD Lab, 3230 Deming Way, Middleton, WI 53562, USA
| | - Gang Xing
- Internal Medicine Research Unit Pfizer, Inc, 610 Main Street, Cambridge, MA 02139, USA
| | | | - Sara Clemens
- PPD Lab, 3230 Deming Way, Middleton, WI 53562, USA
| | | | | | - Jenny Yanhua Zhang
- Clinical Biomarker Sciences Group Precision Medicine Early Clinical Development Pfizer Inc, 610 Main Street, Cambridge, MA 02139, USA
| | - Yizhong Zhang
- Clinical Assay Group Clinical Pharmacology Global Product Development Pfizer, Inc, 445 Eastern Point Road, Groton, CT 06340, USA
| | - Nancy Raha
- Clinical Assay Group Clinical Pharmacology Global Product Development Pfizer, Inc, 445 Eastern Point Road, Groton, CT 06340, USA.
| |
Collapse
|
14
|
Bence KK, Birnbaum MJ. Metabolic drivers of non-alcoholic fatty liver disease. Mol Metab 2021; 50:101143. [PMID: 33346069 PMCID: PMC8324696 DOI: 10.1016/j.molmet.2020.101143] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/02/2020] [Accepted: 12/11/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The incidence of non-alcoholic fatty liver disease (NAFLD) is rapidly increasing worldwide parallel to the global obesity epidemic. NAFLD encompasses a range of liver pathologies and most often originates from metabolically driven accumulation of fat in the liver, or non-alcoholic fatty liver (NAFL). In a subset of NAFL patients, the disease can progress to non-alcoholic steatohepatitis (NASH), which is a more severe form of liver disease characterized by hepatocyte injury, inflammation, and fibrosis. Significant progress has been made over the past decade in our understanding of NASH pathogenesis, but gaps remain in our mechanistic knowledge of the precise metabolic triggers for disease worsening. SCOPE OF REVIEW The transition from NAFL to NASH likely involves a complex constellation of multiple factors intrinsic and extrinsic to the liver. This review focuses on early metabolic events in the establishment of NAFL and initial stages of NASH. We discuss the association of NAFL with obesity as well as the role of adipose tissue in disease progression and highlight early metabolic drivers implicated in the pathological transition from hepatic fat accumulation to steatohepatitis. MAJOR CONCLUSIONS The close association of NAFL with features of metabolic syndrome highlight plausible mechanistic roles for adipose tissue health and the release of lipotoxic lipids, hepatic de novo lipogenesis (DNL), and disruption of the intestinal barrier in not only the initial establishment of hepatic steatosis, but also in mediating disease progression. Human genetic variants linked to NASH risk to date are heavily biased toward genes involved in the regulation of lipid metabolism, providing compelling support for the hypothesis that NASH is fundamentally a metabolic disease.
Collapse
Affiliation(s)
- Kendra K Bence
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development, and Medical, Cambridge, MA, USA.
| | - Morris J Birnbaum
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development, and Medical, Cambridge, MA, USA
| |
Collapse
|
15
|
Jiang H, Lin Q, Ma L, Luo S, Jiang X, Fang J, Lu Z. Fructose and fructose kinase in cancer and other pathologies. J Genet Genomics 2021; 48:531-539. [PMID: 34326012 DOI: 10.1016/j.jgg.2021.06.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 12/14/2022]
Abstract
Fructose metabolism and fructose kinase KHK-C/A are key factors in the development of lipid oversynthesis-promoted metabolic disorders and cancer. Here, we summarize and discuss the current knowledge about the specific features of fructose metabolism and the distinct roles of KHK-C and KHK-A in metabolic liver diseases and their relevant metabolic disorders and cancer, and we highlight the specific protein kinase activity of KHK-A in tumor development. In addition, different approaches that have been used to inhibit KHK and the exploration of KHK inhibitors in clinical treatment are introduced.
Collapse
Affiliation(s)
- Hongfei Jiang
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao 266061, China
| | - Qian Lin
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao 266061, China
| | - Leina Ma
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao 266061, China
| | - Shudi Luo
- Zhejiang University Cancer Center, Hangzhou 310029, China
| | - Xiaoming Jiang
- Zhejiang University Cancer Center, Hangzhou 310029, China
| | - Jing Fang
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao 266061, China.
| | - Zhimin Lu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China; Zhejiang University Cancer Center, Hangzhou 310029, China.
| |
Collapse
|
16
|
Federico A, Rosato V, Masarone M, Torre P, Dallio M, Romeo M, Persico M. The Role of Fructose in Non-Alcoholic Steatohepatitis: Old Relationship and New Insights. Nutrients 2021; 13:1314. [PMID: 33923525 PMCID: PMC8074203 DOI: 10.3390/nu13041314] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/12/2021] [Accepted: 04/14/2021] [Indexed: 12/22/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) represents the result of hepatic fat overload not due to alcohol consumption and potentially evolving to advanced fibrosis, cirrhosis, and hepatocellular carcinoma. Fructose is a naturally occurring simple sugar widely used in food industry linked to glucose to form sucrose, largely contained in hypercaloric food and beverages. An increasing amount of evidence in scientific literature highlighted a detrimental effect of dietary fructose consumption on metabolic disorders such as insulin resistance, obesity, hepatic steatosis, and NAFLD-related fibrosis as well. An excessive fructose consumption has been associated with NAFLD development and progression to more clinically severe phenotypes by exerting various toxic effects, including increased fatty acid production, oxidative stress, and worsening insulin resistance. Furthermore, some studies in this context demonstrated even a crucial role in liver cancer progression. Despite this compelling evidence, the molecular mechanisms by which fructose elicits those effects on liver metabolism remain unclear. Emerging data suggest that dietary fructose may directly alter the expression of genes involved in lipid metabolism, including those that increase hepatic fat accumulation or reduce hepatic fat removal. This review aimed to summarize the current understanding of fructose metabolism on NAFLD pathogenesis and progression.
Collapse
Affiliation(s)
- Alessandro Federico
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.D.); (M.R.)
| | - Valerio Rosato
- Internal Medicine and Hepatology Division, Department of Medicine, Surgery and Odontostomatology, “Scuola Medica Salernitana”, University of Salerno, 84084 Salerno, Italy; (V.R.); (M.M.); (P.T.); (M.P.)
- Liver Unit, Ospedale Evangelico Betania, 80147 Naples, Italy
| | - Mario Masarone
- Internal Medicine and Hepatology Division, Department of Medicine, Surgery and Odontostomatology, “Scuola Medica Salernitana”, University of Salerno, 84084 Salerno, Italy; (V.R.); (M.M.); (P.T.); (M.P.)
| | - Pietro Torre
- Internal Medicine and Hepatology Division, Department of Medicine, Surgery and Odontostomatology, “Scuola Medica Salernitana”, University of Salerno, 84084 Salerno, Italy; (V.R.); (M.M.); (P.T.); (M.P.)
| | - Marcello Dallio
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.D.); (M.R.)
| | - Mario Romeo
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.D.); (M.R.)
| | - Marcello Persico
- Internal Medicine and Hepatology Division, Department of Medicine, Surgery and Odontostomatology, “Scuola Medica Salernitana”, University of Salerno, 84084 Salerno, Italy; (V.R.); (M.M.); (P.T.); (M.P.)
| |
Collapse
|
17
|
Xie Z, Yang X, Duan Y, Han J, Liao C. Small-Molecule Kinase Inhibitors for the Treatment of Nononcologic Diseases. J Med Chem 2021; 64:1283-1345. [PMID: 33481605 DOI: 10.1021/acs.jmedchem.0c01511] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Great successes have been achieved in developing small-molecule kinase inhibitors as anticancer therapeutic agents. However, kinase deregulation plays essential roles not only in cancer but also in almost all major disease areas. Accumulating evidence has revealed that kinases are promising drug targets for different diseases, including cancer, autoimmune diseases, inflammatory diseases, cardiovascular diseases, central nervous system disorders, viral infections, and malaria. Indeed, the first small-molecule kinase inhibitor for treatment of a nononcologic disease was approved in 2011 by the U.S. FDA. To date, 10 such inhibitors have been approved, and more are in clinical trials for applications other than cancer. This Perspective discusses a number of kinases and their small-molecule inhibitors for the treatment of diseases in nononcologic therapeutic fields. The opportunities and challenges in developing such inhibitors are also highlighted.
Collapse
Affiliation(s)
- Zhouling Xie
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Xiaoxiao Yang
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Yajun Duan
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Jihong Han
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Chenzhong Liao
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| |
Collapse
|
18
|
Foley TL, Burchett W, Chen Q, Flanagan ME, Kapinos B, Li X, Montgomery JI, Ratnayake AS, Zhu H, Peakman MC. Selecting Approaches for Hit Identification and Increasing Options by Building the Efficient Discovery of Actionable Chemical Matter from DNA-Encoded Libraries. SLAS DISCOVERY 2021; 26:263-280. [PMID: 33412987 DOI: 10.1177/2472555220979589] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Over the past 20 years, the toolbox for discovering small-molecule therapeutic starting points has expanded considerably. Pharmaceutical researchers can now choose from technologies that, in addition to traditional high-throughput knowledge-based and diversity screening, now include the screening of fragment and fragment-like libraries, affinity selection mass spectrometry, and selection against DNA-encoded libraries (DELs). Each of these techniques has its own unique combination of advantages and limitations that makes them more, or less, suitable for different target classes or discovery objectives, such as desired mechanism of action. Layered on top of this are the constraints of the drug-hunters themselves, including budgets, timelines, and available platform capacity; each of these can play a part in dictating the hit identification strategy for a discovery program. In this article, we discuss some of the factors that we use to govern our building of a hit identification roadmap for a program and describe the increasing role that DELs are playing in our discovery strategy. Furthermore, we share our learning during our initial exploration of DEL and highlight the approaches we have evolved to maximize the value returned from DEL selections. Topics addressed include the optimization of library design and production, reagent validation, data analysis, and hit confirmation. We describe how our thinking in these areas has led us to build a DEL platform that has begun to deliver tractable matter to our global discovery portfolio.
Collapse
Affiliation(s)
| | | | - Qiuxia Chen
- Lead Generation Unit, HitGen Inc., Chengdu, Shuangliu District, China
| | | | | | - Xianyang Li
- Lead Generation Unit, HitGen Inc., Chengdu, Shuangliu District, China
| | | | | | - Hongyao Zhu
- Simulation and Modelling Sciences, Pfizer Inc., Groton, CT, USA
| | | |
Collapse
|
19
|
Skenderian S, Park G, Jang C. Organismal Fructose Metabolism in Health and Non-Alcoholic Fatty Liver Disease. BIOLOGY 2020; 9:E405. [PMID: 33218081 PMCID: PMC7698815 DOI: 10.3390/biology9110405] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 02/07/2023]
Abstract
NAFLD has alarmingly increased, yet FDA-approved drugs are still lacking. An excessive intake of fructose, especially in liquid form, is a dietary risk factor of NAFLD. While fructose metabolism has been studied for decades, it is still controversial how fructose intake can cause NAFLD. It has long been believed that fructose metabolism solely happens in the liver and accordingly, numerous studies have investigated liver fructose metabolism using primary hepatocytes or liver cell lines in culture. While cultured cells are useful for studying detailed signaling pathways and metabolism in a cell-autonomous manner, it is equally important to understand fructose metabolism at the whole-body level in live organisms. In this regard, recent in vivo studies using genetically modified mice and stable isotope tracing have tremendously expanded our understanding of the complex interaction between fructose-catabolizing organs and gut microbiota. Here, we discuss how the aberrant distribution of fructose metabolism between organs and gut microbiota can contribute to NAFLD. We also address potential therapeutic interventions of fructose-elicited NAFLD.
Collapse
Affiliation(s)
- Shea Skenderian
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA;
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA;
| | - Grace Park
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA;
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA;
| |
Collapse
|
20
|
Horst R, Farley KA, Kormos BL, Withka JM. NMR spectroscopy: the swiss army knife of drug discovery. JOURNAL OF BIOMOLECULAR NMR 2020; 74:509-519. [PMID: 32617727 DOI: 10.1007/s10858-020-00330-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 06/25/2020] [Indexed: 06/11/2023]
Abstract
Nuclear magnetic resonance (NMR) spectroscopy has evolved into a powerful tool within drug discovery over the last two decades. While traditionally being used by medicinal chemists for small molecule structure elucidation, it can also be a valuable tool for the identification of small molecules that bind to drug targets, for the characterization of target-ligand interactions and for hit-to-lead optimization. Here, we describe how NMR spectroscopy is integrated into the Pfizer drug discovery pipeline and how we utilize this approach to identify and validate initial hits and generate leads.
Collapse
Affiliation(s)
- Reto Horst
- Medicinal Sciences, Pfizer, Eastern Point Road, Groton, CT, 06340, USA.
| | - Kathleen A Farley
- Medicinal Sciences, Pfizer, Eastern Point Road, Groton, CT, 06340, USA
| | - Bethany L Kormos
- Medicinal Sciences, Pfizer, 610 Main St., Cambridge, MA, 02139, USA
| | - Jane M Withka
- Medicinal Sciences, Pfizer, 610 Main St., Cambridge, MA, 02139, USA
| |
Collapse
|
21
|
Futatsugi K, Smith AC, Tu M, Raymer B, Ahn K, Coffey SB, Dowling MS, Fernando DP, Gutierrez JA, Huard K, Jasti J, Kalgutkar AS, Knafels JD, Pandit J, Parris KD, Perez S, Pfefferkorn JA, Price DA, Ryder T, Shavnya A, Stock IA, Tsai AS, Tesz GJ, Thuma BA, Weng Y, Wisniewska HM, Xing G, Zhou J, Magee TV. Discovery of PF-06835919: A Potent Inhibitor of Ketohexokinase (KHK) for the Treatment of Metabolic Disorders Driven by the Overconsumption of Fructose. J Med Chem 2020; 63:13546-13560. [PMID: 32910646 DOI: 10.1021/acs.jmedchem.0c00944] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Increased fructose consumption and its subsequent metabolism have been implicated in metabolic disorders such as nonalcoholic fatty liver disease and steatohepatitis (NAFLD/NASH) and insulin resistance. Ketohexokinase (KHK) converts fructose to fructose-1-phosphate (F1P) in the first step of the metabolic cascade. Herein we report the discovery of a first-in-class KHK inhibitor, PF-06835919 (8), currently in phase 2 clinical trials. The discovery of 8 was built upon our originally reported, fragment-derived lead 1 and the recognition of an alternative, rotated binding mode upon changing the ribose-pocket binding moiety from a pyrrolidinyl to an azetidinyl ring system. This new binding mode enabled efficient exploration of the vector directed at the Arg-108 residue, leading to the identification of highly potent 3-azabicyclo[3.1.0]hexane acetic acid-based KHK inhibitors by combined use of parallel medicinal chemistry and structure-based drug design.
Collapse
Affiliation(s)
- Kentaro Futatsugi
- Pfizer Inc. Medicine Design, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Aaron C Smith
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Meihua Tu
- Pfizer Inc. Medicine Design, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Brian Raymer
- Pfizer Inc. Medicine Design, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Kay Ahn
- Pfizer Inc. Internal Medicine Research Unit, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Steven B Coffey
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Matthew S Dowling
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Dilinie P Fernando
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jemy A Gutierrez
- Pfizer Inc. Internal Medicine Research Unit, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Kim Huard
- Pfizer Inc. Medicine Design, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Jayasankar Jasti
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Amit S Kalgutkar
- Pfizer Inc. Medicine Design, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - John D Knafels
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jayvardhan Pandit
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kevin D Parris
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Sylvie Perez
- Pfizer Inc. Internal Medicine Research Unit, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Jeffrey A Pfefferkorn
- Pfizer Inc. Internal Medicine Research Unit, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - David A Price
- Pfizer Inc. Medicine Design, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Tim Ryder
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Andre Shavnya
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Ingrid A Stock
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Andy S Tsai
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Gregory J Tesz
- Pfizer Inc. Internal Medicine Research Unit, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Benjamin A Thuma
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Yan Weng
- Pfizer Inc. Medicine Design, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Hanna M Wisniewska
- Pfizer Inc. Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Gang Xing
- Pfizer Inc. Internal Medicine Research Unit, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Jun Zhou
- Pfizer Inc. Drug Safety R&D, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Thomas V Magee
- Pfizer Inc. Medicine Design, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
22
|
Attia SL, Softic S, Mouzaki M. Evolving Role for Pharmacotherapy in NAFLD/NASH. Clin Transl Sci 2020; 14:11-19. [PMID: 32583961 PMCID: PMC7877845 DOI: 10.1111/cts.12839] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/04/2020] [Indexed: 12/13/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a highly prevalent, dynamic disease that occurs across the age spectrum and can lead to cirrhosis and hepatocellular carcinoma. There are currently no US Food and Drug Administration (FDA) approved treatments for NAFLD; however, this is a field of active research. This review summarizes emerging pharmacotherapies for the treatment of adult and pediatric NAFLD. Investigated pharmacotherapies predominantly target bile acid signaling, insulin resistance, and lipid handling within the liver. Three drugs have gone on to phase III trials for which results are available. Of those, obeticholic acid is the single agent that demonstrates promise according to the interim analyses of the REGENERATE trial. Obeticholic acid showed reduction of fibrosis in adults with nonalcoholic steatohepatitis (NASH) taking 25 mg daily for 18 months (n = 931, reduction in fibrosis in 25% vs. 12% placebo, P < 0.01). Ongoing phase III trials include REGENERATE and MAESTRO‐NASH, which investigates thyroid hormone receptor‐β agonist MGL‐3196. Outcomes of promising phase II trials in adults with NASH are also available and those have investigated agents, including the fibroblast growth factor (FGF)19 analogue NGM282, the GLP1 agonist liraglutide, the FGF21 analogue Pegbelfermin, the sodium glucose co‐transporter 2 inhibitor Empagliflozin, the ketohexokinase inhibitor PF‐06835919, the acetyl‐coenzyme A carboxylase inhibitor GS‐0976, and the chemokine receptor antagonist Cenicriviroc. Completed and ongoing clinical trials emphasize the need for a more nuanced understanding of the phenotypes of subgroups within NAFLD that may respond to an individualized approach to pharmacotherapy.
Collapse
Affiliation(s)
- Suzanna L Attia
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Kentucky College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Samir Softic
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Kentucky College of Medicine, University of Kentucky, Lexington, Kentucky, USA.,Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, USA.,Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Marialena Mouzaki
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
23
|
Jang C, Wada S, Yang S, Gosis B, Zeng X, Zhang Z, Shen Y, Lee G, Arany Z, Rabinowitz JD. The small intestine shields the liver from fructose-induced steatosis. Nat Metab 2020; 2:586-593. [PMID: 32694791 PMCID: PMC8020332 DOI: 10.1038/s42255-020-0222-9] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 05/20/2020] [Indexed: 02/06/2023]
Abstract
Per capita fructose consumption has increased 100-fold over the last century1. Epidemiological studies suggest that excessive fructose consumption, and especially consumption of sweet drinks, is associated with hyperlipidaemia, non-alcoholic fatty liver disease, obesity and diabetes2-7. Fructose metabolism begins with its phosphorylation by the enzyme ketohexokinase (KHK), which exists in two alternatively spliced forms8. The more active isozyme, KHK-C, is expressed most strongly in the liver, but also substantially in the small intestine9,10 where it drives dietary fructose absorption and conversion into other metabolites before fructose reaches the liver11-13. It is unclear whether intestinal fructose metabolism prevents or contributes to fructose-induced lipogenesis and liver pathology. Here we show that intestinal fructose catabolism mitigates fructose-induced hepatic lipogenesis. In mice, intestine-specific KHK-C deletion increases dietary fructose transit to the liver and gut microbiota and sensitizes mice to fructose's hyperlipidaemic effects and hepatic steatosis. In contrast, intestine-specific KHK-C overexpression promotes intestinal fructose clearance and decreases fructose-induced lipogenesis. Thus, intestinal fructose clearance capacity controls the rate at which fructose can be safely ingested. Consistent with this, we show that the same amount of fructose is more strongly lipogenic when drunk than eaten, or when administered as a single gavage, as opposed to multiple doses spread over 45 min. Collectively, these data demonstrate that fructose induces lipogenesis when its dietary intake rate exceeds the intestinal clearance capacity. In the modern context of ready food availability, the resulting fructose spillover drives metabolic syndrome. Slower fructose intake, tailored to intestinal capacity, can mitigate these consequences.
Collapse
Affiliation(s)
- Cholsoon Jang
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA.
| | - Shogo Wada
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven Yang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bridget Gosis
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xianfeng Zeng
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Zhaoyue Zhang
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Yihui Shen
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Gina Lee
- Meyer Cancer Center and Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Zoltan Arany
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Joshua D Rabinowitz
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
24
|
Ershova AI, Ievlev MY, Maksimova VN, Belikov MY, Ershov OV. Synthesis, Solution and Solid‐State Fluorescence of 2‐(
N
‐cycloamino)cinchomeronic Dinitrile Derivatives. ChemistrySelect 2020. [DOI: 10.1002/slct.202001710] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Anastasia I. Ershova
- Department of Organic and Pharmaceutical ChemistryUlyanov Chuvash State University Cheboksary 428015 Chuvash Republic Russia
| | - Mikhail Yu. Ievlev
- Department of Organic and Pharmaceutical ChemistryUlyanov Chuvash State University Cheboksary 428015 Chuvash Republic Russia
| | - Veronika N. Maksimova
- Department of Organic and Pharmaceutical ChemistryUlyanov Chuvash State University Cheboksary 428015 Chuvash Republic Russia
| | - Mikhail Yu. Belikov
- Department of Organic and Pharmaceutical ChemistryUlyanov Chuvash State University Cheboksary 428015 Chuvash Republic Russia
| | - Oleg V. Ershov
- Department of Organic and Pharmaceutical ChemistryUlyanov Chuvash State University Cheboksary 428015 Chuvash Republic Russia
| |
Collapse
|
25
|
Buziau AM, Schalkwijk CG, Stehouwer CDA, Tolan DR, Brouwers MCGJ. Recent advances in the pathogenesis of hereditary fructose intolerance: implications for its treatment and the understanding of fructose-induced non-alcoholic fatty liver disease. Cell Mol Life Sci 2020; 77:1709-1719. [PMID: 31713637 PMCID: PMC11105038 DOI: 10.1007/s00018-019-03348-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 10/02/2019] [Accepted: 10/16/2019] [Indexed: 12/31/2022]
Abstract
Hereditary fructose intolerance (HFI) is a rare inborn disease characterized by a deficiency in aldolase B, which catalyzes the cleavage of fructose 1,6-bisphosphate and fructose 1-phosphate (Fru 1P) to triose molecules. In patients with HFI, ingestion of fructose results in accumulation of Fru 1P and depletion of ATP, which are believed to cause symptoms, such as nausea, vomiting, hypoglycemia, and liver and kidney failure. These sequelae can be prevented by a fructose-restricted diet. Recent studies in aldolase B-deficient mice and HFI patients have provided more insight into the pathogenesis of HFI, in particular the liver phenotype. Both aldolase B-deficient mice (fed a very low fructose diet) and HFI patients (treated with a fructose-restricted diet) displayed greater intrahepatic fat content when compared to controls. The liver phenotype in aldolase B-deficient mice was prevented by reduction in intrahepatic Fru 1P concentrations by crossing these mice with mice deficient for ketohexokinase, the enzyme that catalyzes the synthesis of Fru 1P. These new findings not only provide a potential novel treatment for HFI, but lend insight into the pathogenesis of fructose-induced non-alcoholic fatty liver disease (NAFLD), which has raised to epidemic proportions in Western society. This narrative review summarizes the most recent advances in the pathogenesis of HFI and discusses the implications for the understanding and treatment of fructose-induced NAFLD.
Collapse
Affiliation(s)
- Amée M Buziau
- Division of Endocrinology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Laboratory for Metabolism and Vascular Medicine, Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - Casper G Schalkwijk
- Laboratory for Metabolism and Vascular Medicine, Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - Coen D A Stehouwer
- Laboratory for Metabolism and Vascular Medicine, Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
- Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Dean R Tolan
- Department of Biology, Boston University, Boston, MA, USA.
| | - Martijn C G J Brouwers
- Division of Endocrinology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands.
- Laboratory for Metabolism and Vascular Medicine, Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands.
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands.
| |
Collapse
|
26
|
Romero FA, Jones CT, Xu Y, Fenaux M, Halcomb RL. The Race to Bash NASH: Emerging Targets and Drug Development in a Complex Liver Disease. J Med Chem 2020; 63:5031-5073. [PMID: 31930920 DOI: 10.1021/acs.jmedchem.9b01701] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is a severe form of nonalcoholic fatty liver disease (NAFLD) characterized by liver steatosis, inflammation, and hepatocellular damage. NASH is a serious condition that can progress to cirrhosis, liver failure, and hepatocellular carcinoma. The association of NASH with obesity, type 2 diabetes mellitus, and dyslipidemia has led to an emerging picture of NASH as the liver manifestation of metabolic syndrome. Although diet and exercise can dramatically improve NASH outcomes, significant lifestyle changes can be challenging to sustain. Pharmaceutical therapies could be an important addition to care, but currently none are approved for NASH. Here, we review the most promising targets for NASH treatment, along with the most advanced therapeutics in development. These include targets involved in metabolism (e.g., sugar, lipid, and cholesterol metabolism), inflammation, and fibrosis. Ultimately, combination therapies addressing multiple aspects of NASH pathogenesis are expected to provide benefit for patients.
Collapse
Affiliation(s)
- F Anthony Romero
- Terns Pharmaceuticals, 1065 E. Hillsdale Blvd., Suite 100, Foster City, California 94404, United States
| | - Christopher T Jones
- Terns Pharmaceuticals, 1065 E. Hillsdale Blvd., Suite 100, Foster City, California 94404, United States
| | - Yingzi Xu
- Terns Pharmaceuticals, 1065 E. Hillsdale Blvd., Suite 100, Foster City, California 94404, United States
| | - Martijn Fenaux
- Terns Pharmaceuticals, 1065 E. Hillsdale Blvd., Suite 100, Foster City, California 94404, United States
| | - Randall L Halcomb
- Terns Pharmaceuticals, 1065 E. Hillsdale Blvd., Suite 100, Foster City, California 94404, United States
| |
Collapse
|
27
|
Jarvis A, Ouvry G. Essential ingredients for rational drug design. Bioorg Med Chem Lett 2019; 29:126674. [PMID: 31521476 DOI: 10.1016/j.bmcl.2019.126674] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/03/2019] [Accepted: 09/04/2019] [Indexed: 01/09/2023]
Abstract
This short review focuses on three aspects of rational drug design that we consider of utmost importance: the conformation of small molecules in solid form, the conformation of small molecules in solution and lesser studied interactions in protein-ligand complexes. Using examples from recent literature, we will illustrate these different aspects and how they have contributed to the discovery of potent modulators.
Collapse
Affiliation(s)
- Ashley Jarvis
- Evotec (U.K.) Ltd., 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, United Kingdom
| | - Gilles Ouvry
- Evotec (U.K.) Ltd., 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, United Kingdom.
| |
Collapse
|
28
|
Helal CJ, Bundesmann M, Hammond S, Holmstrom M, Klug-McLeod J, Lefker BA, McLeod D, Subramanyam C, Zakaryants O, Sakata S. Quick Building Blocks (QBB): An Innovative and Efficient Business Model To Speed Medicinal Chemistry Analog Synthesis. ACS Med Chem Lett 2019; 10:1104-1109. [PMID: 31413793 DOI: 10.1021/acsmedchemlett.9b00205] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/05/2019] [Indexed: 12/18/2022] Open
Abstract
Many pharmaceutical companies have invested millions of dollars in establishing internal chemical stores to provide reliable access to large numbers of building blocks (BB) for the synthesis of new molecules, especially for the timely design and execution of parallel (library) synthesis. Recognizing budget and logistical limitations, we required a more economically scalable process to provide diverse BB. We disclose a novel business partnership that achieves the goals of just-in-time, economical access to commercial BB that increases chemical space coverage and accelerates the synthesis of new drug candidates. We believe that this model can be of benefit to companies of all sizes that are engaged in drug discovery by reducing cost, increasing diversity of analog molecules in a time-conscious manner, and reducing BB inventory. More efficient use of BB by customers may allow commercial vendors to devote a greater portion of their resources to preparing novel BB that increase chemical diversity as opposed to resynthesizing out-of-stock compounds that are inaccessible within company compound collections.
Collapse
Affiliation(s)
- Christopher J. Helal
- Worldwide Research and Development, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Mark Bundesmann
- Worldwide Research and Development, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Susan Hammond
- Worldwide Research and Development, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Melissa Holmstrom
- Worldwide Research and Development, Pfizer Inc., 10770 Science Center Drive, San Diego, California 92121, United States
| | - Jacquelyn Klug-McLeod
- Worldwide Research and Development, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Bruce A. Lefker
- Worldwide Research and Development, Pfizer Inc., 1 Portland Square, Cambridge, Massachusetts 02139, United States
| | - Dale McLeod
- Worldwide Research and Development, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Chakrapani Subramanyam
- Worldwide Research and Development, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Oleg Zakaryants
- MilliporeSigma, 6000 N. Teutonia Avenue, Milwaukee, Wisconsin 53209, United States
| | - Sylvie Sakata
- Worldwide Research and Development, Pfizer Inc., 10770 Science Center Drive, San Diego, California 92121, United States
| |
Collapse
|
29
|
Esler WP, Bence KK. Metabolic Targets in Nonalcoholic Fatty Liver Disease. Cell Mol Gastroenterol Hepatol 2019; 8:247-267. [PMID: 31004828 PMCID: PMC6698700 DOI: 10.1016/j.jcmgh.2019.04.007] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/09/2019] [Accepted: 04/11/2019] [Indexed: 12/18/2022]
Abstract
The prevalence and diagnosis of nonalcoholic fatty liver disease (NAFLD) is on the rise worldwide and currently has no FDA-approved pharmacotherapy. The increase in disease burden of NAFLD and a more severe form of this progressive liver disease, nonalcoholic steatohepatitis (NASH), largely mirrors the increase in obesity and type 2 diabetes (T2D) and reflects the hepatic manifestation of an altered metabolic state. Indeed, metabolic syndrome, defined as a constellation of obesity, insulin resistance, hyperglycemia, dyslipidemia and hypertension, is the major risk factor predisposing the NAFLD and NASH. There are multiple potential pharmacologic strategies to rebalance aspects of disordered metabolism in NAFLD. These include therapies aimed at reducing hepatic steatosis by directly modulating lipid metabolism within the liver, inhibiting fructose metabolism, altering delivery of free fatty acids from the adipose to the liver by targeting insulin resistance and/or adipose metabolism, modulating glycemia, and altering pleiotropic metabolic pathways simultaneously. Emerging data from human genetics also supports a role for metabolic drivers in NAFLD and risk for progression to NASH. In this review, we highlight the prominent metabolic drivers of NAFLD pathogenesis and discuss the major metabolic targets of NASH pharmacotherapy.
Collapse
Key Words
- acc, acetyl-coa carboxylase
- alt, alanine aminotransferase
- aso, anti-sense oligonucleotide
- ast, aspartate aminotransferase
- chrebp, carbohydrate response element binding protein
- ci, confidence interval
- dgat, diacylglycerol o-acyltransferase
- dnl, de novo lipogenesis
- fas, fatty acid synthase
- ffa, free fatty acid
- fgf, fibroblast growth factor
- fxr, farnesoid x receptor
- glp-1, glucagon-like peptide-1
- hdl, high-density lipoprotein
- homa-ir, homeostatic model assessment of insulin resistance
- ldl, low-density lipoprotein
- nafld, nonalcoholic fatty liver disease
- nas, nonalcoholic fatty liver disease activity score
- nash, nonalcoholic steatohepatitis
- or, odds ratio
- pdff, proton density fat fraction
- ppar, peroxisome proliferator-activated receptor
- sglt2, sodium glucose co-transporter 2
- srebp-1c, sterol regulatory element binding protein-1c
- t2d, type 2 diabetes
- t2dm, type 2 diabetes mellitus
- tg, triglyceride
- th, thyroid hormone
- thr, thyroid hormone receptor
- treg, regulatory t cells
- tzd, thiazolidinedione
- vldl, very low-density lipoprotein
Collapse
Affiliation(s)
- William P Esler
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development, and Medical, Cambridge, Massachusetts
| | - Kendra K Bence
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development, and Medical, Cambridge, Massachusetts.
| |
Collapse
|
30
|
Mortenson PN, Erlanson DA, de Esch IJP, Jahnke W, Johnson CN. Fragment-to-Lead Medicinal Chemistry Publications in 2017. J Med Chem 2018; 62:3857-3872. [PMID: 30462504 DOI: 10.1021/acs.jmedchem.8b01472] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This Miniperspective is the third in a series reviewing fragment-to-lead publications from a given year. Following our reviews for 2015 and 2016, this Miniperspective provides tabulated summaries of relevant articles published in 2017 along with some general observations. In addition, we discuss insights obtained from analysis of the combined data set of 85 examples from all three years of publications.
Collapse
Affiliation(s)
- Paul N Mortenson
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road , Cambridge CB4 0QA , United Kingdom
| | - Daniel A Erlanson
- Carmot Therapeutics Inc. , 740 Heinz Avenue , Berkeley , California 94710 , United States
| | - Iwan J P de Esch
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS) , Vrije Universiteit Amsterdam , De Boelelaan 1108 , 1081 HZ , Amsterdam , The Netherlands
| | - Wolfgang Jahnke
- Chemical Biology and Therapeutics , Novartis Institutes for Biomedical Research , 4002 Basel , Switzerland
| | - Christopher N Johnson
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road , Cambridge CB4 0QA , United Kingdom
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW To review recent evidence for the role of dietary carbohydrate in de novo lipogenesis (DNL) and nonalcoholic fatty liver disease (NAFLD). RECENT FINDINGS A large body of evidence suggests that increased hepatic DNL is a significant pathway contributing to the development of NAFLD. Dietary carbohydrates, in particular, fructose, have been shown to stimulate DNL and increase liver fat, although it is debated whether this is due to excess energy or fructose per se. Recent dietary intervention studies conducted in energy balance show that high-fructose diets increase DNL and liver fat, whereas fructose restriction decreases DNL and liver fat. SUMMARY The association of high-carbohydrate and high-sugar diets with NAFLD may in part be explained by the effect of sugar on increasing hepatic DNL.
Collapse
Affiliation(s)
- Sally Chiu
- College of Osteopathic Medicine, Touro University California, Vallejo
- Children's Hospital Oakland Research Institute, Oakland
| | - Kathleen Mulligan
- College of Osteopathic Medicine, Touro University California, Vallejo
- Department of Medicine, University of California, San Francisco
- Division of Endocrinology, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Jean-Marc Schwarz
- College of Osteopathic Medicine, Touro University California, Vallejo
- Department of Medicine, University of California, San Francisco
- Division of Endocrinology, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| |
Collapse
|
32
|
Filipski KJ, Sammons MF, Bhattacharya SK, Panteleev J, Brown JA, Loria PM, Boehm M, Smith AC, Shavnya A, Conn EL, Song K, Weng Y, Facemire C, Jüppner H, Clerin V. Discovery of Orally Bioavailable Selective Inhibitors of the Sodium-Phosphate Cotransporter NaPi2a (SLC34A1). ACS Med Chem Lett 2018; 9:440-445. [PMID: 29795756 DOI: 10.1021/acsmedchemlett.8b00013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 04/04/2018] [Indexed: 11/30/2022] Open
Abstract
Sodium-phosphate cotransporter 2a, or NaPi2a (SLC34A1), is a solute-carrier (SLC) transporter located in the kidney proximal tubule that reabsorbs glomerular-filtered phosphate. Inhibition of NaPi2a may enhance urinary phosphate excretion and correct maladaptive mineral and hormonal derangements associated with increased cardiovascular risk in chronic kidney disease-mineral and bone disorder (CKD-MBD). To date, only nonselective NaPi inhibitors have been described. Herein, we detail the discovery of the first series of selective NaPi2a inhibitors, resulting from optimization of a high-throughput screening hit. The oral PK profile of inhibitor PF-06869206 (6f) in rodents allows for the exploration of the pharmacology of selective NaPi2a inhibition.
Collapse
Affiliation(s)
- Kevin J. Filipski
- Pfizer Worldwide Research & Development, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Matthew F. Sammons
- Pfizer Worldwide Research & Development, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Samit K. Bhattacharya
- Pfizer Worldwide Research & Development, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Jane Panteleev
- Pfizer Worldwide Research & Development, 558 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Janice A. Brown
- Pfizer Worldwide Research & Development, 558 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Paula M. Loria
- Pfizer Worldwide Research & Development, 558 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Markus Boehm
- Pfizer Worldwide Research & Development, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Aaron C. Smith
- Pfizer Worldwide Research & Development, 558 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Andre Shavnya
- Pfizer Worldwide Research & Development, 558 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Edward L. Conn
- Pfizer Worldwide Research & Development, 558 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kun Song
- Pfizer Worldwide Research & Development, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Yan Weng
- Pfizer Worldwide Research & Development, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Carie Facemire
- Pfizer Worldwide Research & Development, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Harald Jüppner
- Endocrine Unit and Pediatric Nephrology Unit, Thier 10, Massachusetts General Hospital and Harvard Medical School, 50 Blossom Street, Boston, Massachusetts 02114, United States
| | - Valerie Clerin
- Pfizer Worldwide Research & Development, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| |
Collapse
|