1
|
Liu S, Li X, Fan P, Gu Y, Yang A, Wang W, Zhou L, Chen H, Zheng F, Lin J, Xu Z, Zhao Q. The potential role of transcription factor sterol regulatory element binding proteins (SREBPs) in Alzheimer's disease. Biomed Pharmacother 2024; 180:117575. [PMID: 39442239 DOI: 10.1016/j.biopha.2024.117575] [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: 07/04/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
Sterol regulatory element binding proteins (SREBPs) are a series of cholesterol-related transcription factors. Their role in regulating brain cholesterol biosynthesis, amyloid accumulation, and tau tangles formation has been intensively studied in protein-protein interaction analysis based on genes in clinical databases. SREBPs play an important role in maintaining cholesterol homeostasis in the brain. There are three subtypes of SREBPs, SREBP-1a stimulates the expression of genes related to cholesterol and fatty acid synthesis, SREBP-1c stimulates adipogenesis, and SREBP-2 stimulates cholesterol synthase and LDL receptors. SREBP-2 is activated in response to cholesterol depletion and stimulates a compensatory upregulation of cholesterol uptake and synthesis. Previous studies have shown that inhibition of SREBP-2 reduces cholesterol and amyloid accumulation, and new research suggests that SREBPs play a multifaceted role in Alzheimer's disease. Here, we highlight the importance of SREBPs in AD, in terms of multiple pathways regulating cholesterol in the brain, and primarily demonstrate the potential of SREBP-2 inhibitors. There was a trend towards a significant increase in the expression levels of different SREBP isoforms in AD patients compared to healthy controls. Therefore, there is a close link between SREBPs and AD, and this review analyses the potential role of SREBPs in the treatment of AD. In addition, we systematically reviewed the research progress of SREBPs in AD, and this review will provide more innovative insights into the pathogenesis and treatment of AD and new strategies for drug development in AD.
Collapse
Affiliation(s)
- Siyuan Liu
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Xinzhu Li
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Panpan Fan
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Yujia Gu
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Aizhu Yang
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Weiyi Wang
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Lijun Zhou
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Huanhua Chen
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Fangyuan Zheng
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Junjie Lin
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Zihua Xu
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| | - Qingchun Zhao
- Teaching Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang 110016, PR China.
| |
Collapse
|
2
|
Peng Z, Chen L, Wang M, Yue X, Wei H, Xu F, Hou W, Li Y. SREBP inhibitors: an updated patent review for 2008-present. Expert Opin Ther Pat 2023; 33:669-680. [PMID: 38054657 DOI: 10.1080/13543776.2023.2291393] [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: 07/18/2023] [Accepted: 11/22/2023] [Indexed: 12/07/2023]
Abstract
INTRODUCTION Sterol regulatory element-binding proteins (SREBPs) are a family of membrane-binding transcription factors that activate genes encoding enzymes required for cholesterol and unsaturated fatty acid synthesis. Overactivation of SREBP is related to the occurrence and development of diabetes, nonalcoholic fatty liver, tumor, and other diseases. In the past period, many SREBP inhibitors have been found. AREAS COVERED This manuscript is a patent review of SREBP inhibitors. We searched 2008 to date for all data from the US patent database (https://www.uspto.gov/) and the European patent database (https://www.epo.org/) with 'SREBP' and 'inhibitor' as keywords and analyzed the search results. EXPERT OPINION Both synthetic and natural SREBP inhibitors have been reported. Despite the lack of cocrystal structure of SREBP inhibitor, the mechanisms of several compounds have been clarified. Importantly, some SREBP inhibitors have been proved to have good activity in preclinical studies. As the characteristics of lipid metabolism reprogramming in cardio-cerebrovascular diseases and tumors are gradually revealed, more and more attention will be focused on SREBP.
Collapse
Affiliation(s)
- Zhenyu Peng
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Leyuan Chen
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, China
| | - Manjiang Wang
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, China
| | - Xufan Yue
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huiqiang Wei
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, China
| | - Feifei Xu
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, China
| | - Wenbin Hou
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, China
| | - Yiliang Li
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, China
| |
Collapse
|
3
|
Guiselin T, Lecoutey C, Rochais C, Dallemagne P. Conceptual Framework of the Design of Pleiotropic Drugs against Alzheimer's Disease. Pharmaceutics 2023; 15:2382. [PMID: 37896142 PMCID: PMC10610275 DOI: 10.3390/pharmaceutics15102382] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/27/2023] [Accepted: 09/18/2023] [Indexed: 10/29/2023] Open
Abstract
The multifactorial nature of some diseases, particularly neurodegenerative diseases such as Alzheimer's disease, frequently requires the use of several drugs. These drug cocktails are not without drawbacks in terms of increased adverse effects, drug-drug interactions or low adherence to treatment. The use of pleiotropic drugs, which combine, within a single molecule, several activities directed against distinct therapeutic targets, makes it possible to overcome some of these problems. In addition, these pleiotropic drugs generally lead to the expression of a synergy of effects, sometimes greater than that observed with a combination of drugs. This article will review, through recent examples, the different kinds of pleiotropic drugs being studied or already present on the market of medicines, with a focus on the structural aspect of such drug design.
Collapse
Affiliation(s)
| | | | | | - Patrick Dallemagne
- Normandie University, Unicaen, Centre d’Etudes et de Recherche sur le Médicament de Normandie (CERMN), 14000 Caen, France; (T.G.); (C.L.); (C.R.)
| |
Collapse
|
4
|
Biocatalytic synthesis of lipophilic amides by the lipase of Candida antarctica type B. MOLECULAR CATALYSIS 2022. [DOI: 10.1016/j.mcat.2022.112635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
5
|
Deng J, Zhang J, Shi K, Liu Z. Drug development progress in duchenne muscular dystrophy. Front Pharmacol 2022; 13:950651. [PMID: 35935842 PMCID: PMC9353054 DOI: 10.3389/fphar.2022.950651] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/28/2022] [Indexed: 12/22/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe, progressive, and incurable X-linked disorder caused by mutations in the dystrophin gene. Patients with DMD have an absence of functional dystrophin protein, which results in chronic damage of muscle fibers during contraction, thus leading to deterioration of muscle quality and loss of muscle mass over time. Although there is currently no cure for DMD, improvements in treatment care and management could delay disease progression and improve quality of life, thereby prolonging life expectancy for these patients. Furthermore, active research efforts are ongoing to develop therapeutic strategies that target dystrophin deficiency, such as gene replacement therapies, exon skipping, and readthrough therapy, as well as strategies that target secondary pathology of DMD, such as novel anti-inflammatory compounds, myostatin inhibitors, and cardioprotective compounds. Furthermore, longitudinal modeling approaches have been used to characterize the progression of MRI and functional endpoints for predictive purposes to inform Go/No Go decisions in drug development. This review showcases approved drugs or drug candidates along their development paths and also provides information on primary endpoints and enrollment size of Ph2/3 and Ph3 trials in the DMD space.
Collapse
Affiliation(s)
- Jiexin Deng
- School of Nursing and Health, Henan University, Kaifeng, China
- *Correspondence: Jiexin Deng, ; Zhigang Liu,
| | - Junshi Zhang
- Department of Neurology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Keli Shi
- School of Medicine, Henan University, Kaifeng, China
| | - Zhigang Liu
- Department of Orthopedics, First Affiliated Hospital of Henan University, Kaifeng, China
- *Correspondence: Jiexin Deng, ; Zhigang Liu,
| |
Collapse
|
6
|
Finkel RS, McDonald CM, Lee Sweeney H, Finanger E, Neil Knierbein E, Wagner KR, Mathews KD, Marks W, Statland J, Nance J, McMillan HJ, McCullagh G, Tian C, Ryan MM, O'Rourke D, Müller-Felber W, Tulinius M, Bryan Burnette W, Nguyen CT, Vijayakumar K, Johannsen J, Phan HC, Eagle M, MacDougall J, Mancini M, Donovan JM. A Randomized, Double-Blind, Placebo-Controlled, Global Phase 3 Study of Edasalonexent in Pediatric Patients with Duchenne Muscular Dystrophy: Results of the PolarisDMD Trial. J Neuromuscul Dis 2021; 8:769-784. [PMID: 34120912 PMCID: PMC8543277 DOI: 10.3233/jnd-210689] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Background: Edasalonexent (CAT-1004) is an orally-administered novel small molecule drug designed to inhibit NF-κB and potentially reduce inflammation and fibrosis to improve muscle function and thereby slow disease progression and muscle decline in Duchenne muscular dystrophy (DMD). Objective: This international, randomized 2 : 1, placebo-controlled, phase 3 study in patients ≥4 – < 8 years old with DMD due to any dystrophin mutation examined the effect of edasalonexent (100 mg/kg/day) compared to placebo over 52 weeks. Methods: Endpoints were changes in the North Star Ambulatory Assessment (NSAA; primary) and timed function tests (TFTs; secondary). Assessment of health-related function used the Pediatric Outcomes Data Collection tool (PODCI). Results: One hundred thirty one patients received edasalonexent (n = 88) and placebo (n = 43). At week 52, differences between edasalonexent and placebo for NSAA total score and TFTs were not statistically significant, although there were consistently less functional declines in the edasalonexent group. A pre-specified analysis by age demonstrated that younger patients (≤6.0 years) showed more robust and statistically significant differences between edasalonexent and placebo for some assessments. Treatment was well-tolerated and the majority of adverse events were mild, and most commonly involved the gastrointestinal system (primarily diarrhea). Conclusions: Edasalonexent was generally well-tolerated with a manageable safety profile at the dose of 100 mg/kg/day. Although edasalonexent did not achieve statistical significance for improvement in primary and secondary functional endpoints for assessment of DMD, subgroup analysis suggested that edasalonexent may slow disease progression if initiated before 6 years of age. (NCT03703882)
Collapse
Affiliation(s)
- Richard S Finkel
- St. Jude Children's Research Hospital, Memphis, TN and Nemours Children's Hospital, Orlando, FL
| | | | - H Lee Sweeney
- University of Florida College of Medicine, Gainesville, FL
| | | | | | - Kathryn R Wagner
- Kennedy Krieger Institute, The Johns Hopkins School of Medicine, Baltimore, MD
| | | | | | | | | | | | | | - Cuixia Tian
- Cincinnati Children's Hospital & University of Cincinnati, Cincinnati, OH
| | | | | | | | - Mar Tulinius
- Queen Silvia Children's Hospital, Gothenburg, Sweden
| | | | | | | | | | - Han C Phan
- Rare Disease Research, LLC, Atlanta GA, Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
7
|
Finkel RS, Finanger E, Vandenborne K, Sweeney HL, Tennekoon G, Shieh PB, Willcocks R, Walter G, Rooney WD, Forbes SC, Triplett WT, Yum SW, Mancini M, MacDougall J, Fretzen A, Bista P, Nichols A, Donovan JM. Disease-modifying effects of edasalonexent, an NF-κB inhibitor, in young boys with Duchenne muscular dystrophy: Results of the MoveDMD phase 2 and open label extension trial. Neuromuscul Disord 2021; 31:385-396. [PMID: 33678513 DOI: 10.1016/j.nmd.2021.02.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/12/2020] [Accepted: 02/01/2021] [Indexed: 12/18/2022]
Abstract
Chronic activation of NF-κB is a key driver of muscle degeneration and suppression of muscle regeneration in Duchenne muscular dystrophy. Edasalonexent (CAT-1004) is an orally-administered novel small molecule that covalently links two bioactive compounds (salicylic acid and docosahexaenoic acid) that inhibit NF-κB. This placebo-controlled, proof-of-concept phase 2 study with open-label extension in boys ≥4-<8 years old with any dystrophin mutation examined the effect of edasalonexent (67 or 100 mg/kg/day) compared to placebo or off-treatment control. Endpoints were safety/tolerability, change from baseline in MRI T2 relaxation time of lower leg muscles and functional assessment, as well as pharmacodynamics and biomarkers. Treatment was well-tolerated and the majority of adverse events were mild, and most commonly of the gastrointestinal system (primarily diarrhea). There were no serious adverse events in the edasalonexent groups. Edasalonexent 100 mg/kg was associated with slowing of disease progression and preservation of muscle function compared to an off-treatment control period, with decrease in levels of NF-κB-regulated genes and improvements in biomarkers of muscle health and inflammation. These results support investigating edasalonexent in future trials and have informed the design of the edasalonexent phase 3 clinical trial in boys with Duchenne.
Collapse
Affiliation(s)
- Richard S Finkel
- St. Jude Children's Research Hospital, Memphis, TN and Nemours Children's Hospital, Orlando, FL, United States.
| | - Erika Finanger
- Oregon Health & Science University, Portland, OR, United States
| | | | - H Lee Sweeney
- University of Florida Health, Gainesville, FL, United States
| | - Gihan Tennekoon
- The Children's Hospital of Philadelphia, and the University of Pennsylvania, Philadelphia, PA, United States
| | - Perry B Shieh
- University of California, Los Angeles, Los Angeles, CA, United States
| | | | - Glenn Walter
- University of Florida Health, Gainesville, FL, United States
| | | | - Sean C Forbes
- University of Florida Health, Gainesville, FL, United States
| | | | - Sabrina W Yum
- The Children's Hospital of Philadelphia, and the University of Pennsylvania, Philadelphia, PA, United States
| | - Maria Mancini
- Catabasis Pharmaceuticals, Inc., Boston, MA, United States
| | | | | | - Pradeep Bista
- Catabasis Pharmaceuticals, Inc., Boston, MA, United States
| | - Andrew Nichols
- Catabasis Pharmaceuticals, Inc., Boston, MA, United States
| | | |
Collapse
|
8
|
Albertini C, Naldi M, Petralla S, Strocchi S, Grifoni D, Monti B, Bartolini M, Bolognesi ML. From Combinations to Single-Molecule Polypharmacology-Cromolyn-Ibuprofen Conjugates for Alzheimer's Disease. Molecules 2021; 26:1112. [PMID: 33669839 PMCID: PMC7923232 DOI: 10.3390/molecules26041112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 02/04/2023] Open
Abstract
Despite Alzheimer's disease (AD) incidence being projected to increase worldwide, the drugs currently on the market can only mitigate symptoms. Considering the failures of the classical paradigm "one target-one drug-one disease" in delivering effective medications for AD, polypharmacology appears to be a most viable therapeutic strategy. Polypharmacology can involve combinations of multiple drugs and/or single chemical entities modulating multiple targets. Taking inspiration from an ongoing clinical trial, this work aims to convert a promising cromolyn-ibuprofen drug combination into single-molecule "codrugs." Such codrugs should be able to similarly modulate neuroinflammatory and amyloid pathways, while showing peculiar pros and cons. By exploiting a linking strategy, we designed and synthesized a small set of cromolyn-ibuprofen conjugates (4-6). Preliminary plasma stability and neurotoxicity assays allowed us to select diamide 5 and ethanolamide 6 as promising compounds for further studies. We investigated their immunomodulatory profile in immortalized microglia cells, in vitro anti-aggregating activity towards Aβ42-amyloid self-aggregation, and their cellular neuroprotective effect against Aβ42-induced neurotoxicity. The fact that 6 effectively reduced Aβ-induced neuronal death, prompted its investigation into an in vivo model. Notably, 6 was demonstrated to significantly increase the longevity of Aβ42-expressing Drosophila and to improve fly locomotor performance.
Collapse
Affiliation(s)
- Claudia Albertini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6/Via Selmi 3, 40126 Bologna, Italy; (C.A.); (M.N.); (S.P.); (S.S.); (B.M.); (M.B.)
| | - Marina Naldi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6/Via Selmi 3, 40126 Bologna, Italy; (C.A.); (M.N.); (S.P.); (S.S.); (B.M.); (M.B.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, St. Orsola Hospital, Via Massarenti 9, 40138 Bologna, Italy
| | - Sabrina Petralla
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6/Via Selmi 3, 40126 Bologna, Italy; (C.A.); (M.N.); (S.P.); (S.S.); (B.M.); (M.B.)
| | - Silvia Strocchi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6/Via Selmi 3, 40126 Bologna, Italy; (C.A.); (M.N.); (S.P.); (S.S.); (B.M.); (M.B.)
| | - Daniela Grifoni
- Department of Life, Health and Environmental Sciences (MeSVA), University of L’Aquila, Via Vetoio, Coppito 2, 67100 L’Aquila, Italy;
| | - Barbara Monti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6/Via Selmi 3, 40126 Bologna, Italy; (C.A.); (M.N.); (S.P.); (S.S.); (B.M.); (M.B.)
| | - Manuela Bartolini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6/Via Selmi 3, 40126 Bologna, Italy; (C.A.); (M.N.); (S.P.); (S.S.); (B.M.); (M.B.)
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6/Via Selmi 3, 40126 Bologna, Italy; (C.A.); (M.N.); (S.P.); (S.S.); (B.M.); (M.B.)
| |
Collapse
|
9
|
Rossi M, Petralla S, Protti M, Baiula M, Kobrlova T, Soukup O, Spampinato SM, Mercolini L, Monti B, Bolognesi ML. α-Linolenic Acid-Valproic Acid Conjugates: Toward Single-Molecule Polypharmacology for Multiple Sclerosis. ACS Med Chem Lett 2020; 11:2406-2413. [PMID: 33329762 PMCID: PMC7734798 DOI: 10.1021/acsmedchemlett.0c00375] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022] Open
Abstract
![]()
Multiple
sclerosis (MS) is a complex inflammatory, degenerative,
and demyelinating disease of the central nervous system. Although
treatments exist, MS cannot be cured by available drugs, which primarily
target neuroinflammation. Thus, it is feasible that a well concerted
polypharmacological approach able to act at multiple points within
the intricate network of inflammation, neurodegeneration, and demyelination/remyelination
pathways would succeed where other drugs have failed. Starting from
reported beneficial effects of α-linolenic acid (ALA) and valproic
acid (VPA) in MS, and by applying a rational strategy, we developed
a small set of codrugs obtained by conjugating VPA and ALA through
proper linkers. A cellular profiling identified 1 as
a polypharmacological tool able not only to modulate microglia polarization,
but also to counteract neurodegeneration and demyelination and induce
oligodendrocyte precursor cell differentiation, by acting on multiple
biochemical and epigenetic pathways.
Collapse
Affiliation(s)
- Michele Rossi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Sabrina Petralla
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Michele Protti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Monica Baiula
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Tereza Kobrlova
- Biomedical Research Center, University Hospital, CZ-500 05 Hradec Kralove, Czech Republic
| | - Ondrej Soukup
- Biomedical Research Center, University Hospital, CZ-500 05 Hradec Kralove, Czech Republic
| | - Santi Mario Spampinato
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Laura Mercolini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Barbara Monti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| |
Collapse
|
10
|
Ferrara SJ, Scanlan TS. A CNS-Targeting Prodrug Strategy for Nuclear Receptor Modulators. J Med Chem 2020; 63:9742-9751. [DOI: 10.1021/acs.jmedchem.0c00868] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Skylar J. Ferrara
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Thomas S. Scanlan
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| |
Collapse
|
11
|
Albertini C, Salerno A, Sena Murteira Pinheiro P, Bolognesi ML. From combinations to multitarget‐directed ligands: A continuum in Alzheimer's disease polypharmacology. Med Res Rev 2020; 41:2606-2633. [DOI: 10.1002/med.21699] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/01/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Claudia Albertini
- Department of Pharmacy and Biotechnology Alma Mater Studiorum–University of Bologna Bologna Italy
| | - Alessandra Salerno
- Department of Pharmacy and Biotechnology Alma Mater Studiorum–University of Bologna Bologna Italy
| | - Pedro Sena Murteira Pinheiro
- Department of Pharmacy and Biotechnology Alma Mater Studiorum–University of Bologna Bologna Italy
- Programa de Pós‐Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas Universidade Federal do Rio de Janeiro Rio de Janeiro Rio de Janeiro Brazil
| | - Maria L. Bolognesi
- Department of Pharmacy and Biotechnology Alma Mater Studiorum–University of Bologna Bologna Italy
| |
Collapse
|
12
|
Wong T, Narayanan S, Brown DP, Chen ZS. Synthesis and Cytotoxicity Studies of Stilbene Long-Chain Fatty Acid Conjugates. JOURNAL OF NATURAL PRODUCTS 2020; 83:1563-1570. [PMID: 32243160 DOI: 10.1021/acs.jnatprod.0c00027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
A series of 16 conjugates of the tubulin polymerization inhibitor combretastatin A4 (CA-4) and other functionally related stilbene with four 18-carbon fatty acids, namely, stearic, oleic, linoleic, and linolenic acids, have been synthesized in good yields. These new derivatives have been evaluated against the KB-3-1 (human epidermoid carcinoma), NCI-H460 (human lung cancer), HEK293 (human embryonic kidney), and MCF-7 (human breast adenocarcinoma) cell lines for antiproliferative activity, with the exhibited cytotoxic activities comparable with those of CA-4 and colchicine. Compounds 22 and 23, CA-4 conjugates of linoleic and linolenic acids, respectively, were determined to have exhibited the most active in vitro assays, with compound 23 exhibiting very similar activity to the parent compound against the NCI-H460 cell line. Our studies further delineated the structurally required Z-geometry of the stilbene moiety and that conjugation of the less active E-stilbenes with the most active fatty acid had minimal or no improvement in their respective activities.
Collapse
Affiliation(s)
- Thomas Wong
- Department of Pharmaceutical Sciences, St. John's University, 8000 Utopia Parkway, Queens, New York 11439, United States
| | - Silpa Narayanan
- Department of Pharmaceutical Sciences, St. John's University, 8000 Utopia Parkway, Queens, New York 11439, United States
| | - David P Brown
- Department of Chemistry, St. John's University, 8000 Utopia Parkway, Queens, New York 11439, United States
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, St. John's University, 8000 Utopia Parkway, Queens, New York 11439, United States
| |
Collapse
|
13
|
Liu H, Bolleddula J, Nichols A, Tang L, Zhao Z, Prakash C. Metabolism of bioconjugate therapeutics: why, when, and how? Drug Metab Rev 2020; 52:66-124. [PMID: 32045530 DOI: 10.1080/03602532.2020.1716784] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Bioconjugation of therapeutic agents has been used as a selective drug delivery platform for many therapeutic areas. Bioconjugates are prepared by the covalent linkage of active compounds (small or large molecule) to a carrier molecule (lipids, proteins, peptides, carbohydrates, and polymers) through a chemical linker. The linkage of the active component to a carrier molecule enhances the therapeutic window through a targeted delivery and by reducing toxicity. Bioconjugates also possess improved pharmacokinetic properties such as a long half-life, increased stability, and cleavage by intracellular enzymes/environment. However, premature cleavage of the bioconjugates and the resulting metabolites/catabolites may produce undesirable toxic effects and, hence, it is critical to understand cleavage mechanisms, metabolism of bioconjugates, and translatability to human in the discovery stages. This article provides a comprehensive overview of linker cleavage pathways and catabolism/metabolism of antibody-drug conjugates, glycoconjugates, polymer-drug conjugates, lipid-drug conjugates, folate-targeted small molecule-drug conjugates, and drug-drug conjugates.
Collapse
Affiliation(s)
- Hanlan Liu
- KSQ Therapeutics Inc., Cambridge, MA, USA
| | | | | | | | | | | |
Collapse
|
14
|
Finanger E, Vandenborne K, Finkel RS, Lee Sweeney H, Tennekoon G, Yum S, Mancini M, Bista P, Nichols A, Liu H, Fretzen A, Donovan JM. Phase 1 Study of Edasalonexent (CAT-1004), an Oral NF-κB Inhibitor, in Pediatric Patients with Duchenne Muscular Dystrophy. J Neuromuscul Dis 2020; 6:43-54. [PMID: 30452422 PMCID: PMC6398836 DOI: 10.3233/jnd-180341] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background: Edasalonexent is an orally administered small molecule designed to inhibit NF-κB, which is activated from infancy in Duchenne muscular dystrophy and is central to causing muscle damage and preventing muscle regeneration. Objective: Evaluate the safety, tolerability, pharmacokinetics and exploratory pharmacodynamics of three doses of edasalonexent in ambulatory males ≥4 to <8 years of age with genetically confirmed Duchenne muscular dystrophy. Methods: This was a 1-week, open-label, multiple-dose study with 3 sequential ascending doses (33, 67 and 100 mg/kg/day) of edasalonexent administered under different dietary conditions to 17 males with a mean age of 5.5 years. Results: All doses of edasalonexent were well tolerated, with no serious adverse events, no drug discontinuations and no dose reductions. The majority of adverse events were mild, and the most common adverse events were gastrointestinal (primarily diarrhea). Edasalonexent was rapidly absorbed with peak levels observed 2–6 hours after dosing and exposures appeared to increase nearly proportionally to dose for the 2 lower and all 3 doses under low-fat and high-fat meal conditions, respectively. Only minor plasma accumulation of edasalonexent was observed with 7 days of dosing. After treatment with edasalonexent for 7 days, levels of NF-κB-regulated genes and serum proteins were decreased. Conclusions: This first report of edasalonexent oral administration for one week in male pediatric patients with Duchenne muscular dystrophy showed that treatment was well tolerated and inhibited NF-kB pathways.
Collapse
Affiliation(s)
- Erika Finanger
- Oregon Health Sciences University Pediatrics, Portland, OR, USA
| | | | - Richard S Finkel
- Nemours Children's Hospital, Division of Pediatric Neurology, Orlando, FL, USA
| | - H Lee Sweeney
- University of Florida Health Myology Institute, Gainesville, FL, USA
| | - Gihan Tennekoon
- Children's Hospital of Philadelphia Pediatric Neurology, Philadelphia, PA, USA
| | - Sabrina Yum
- Children's Hospital of Philadelphia Pediatric Neurology, Philadelphia, PA, USA
| | | | | | | | - Hanlan Liu
- Catabasis Pharmaceuticals, Inc., Cambridge, MA, USA
| | | | | |
Collapse
|
15
|
Winston-McPherson GN, Xie H, Yang K, Li X, Shu D, Tang W. Discovery of 2,3'-diindolylmethanes as a novel class of PCSK9 modulators. Bioorg Med Chem Lett 2019; 29:2345-2348. [PMID: 31227343 DOI: 10.1016/j.bmcl.2019.06.014] [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: 03/30/2019] [Revised: 05/20/2019] [Accepted: 06/11/2019] [Indexed: 12/12/2022]
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) promotes the degradation of low density lipoprotein receptor (LDLR). Anti-PCSK9 agents have been approved for the treatment of hypercholesterolemia. We recently discovered a series of small-molecule PCSK9 modulators that contains a relatively small pharmacophore of 2,3'-diindolylmethane with molecular weights around only 250. These molecules can significantly lower the amount of PCSK9 protein in a cell-based phenotypic assay. Our SAR studies yielded compound 16 with a IC50-value of 200 nM. No obvious cytotoxicity was observed at concentrations below 50 µM.
Collapse
Affiliation(s)
| | - Haibo Xie
- School of Pharmacy, University of Wisconsin, 777 Highland Avenue, Madison, WI 53705, United States
| | - Ka Yang
- School of Pharmacy, University of Wisconsin, 777 Highland Avenue, Madison, WI 53705, United States
| | - Xiaoxun Li
- School of Pharmacy, University of Wisconsin, 777 Highland Avenue, Madison, WI 53705, United States
| | - Dongxu Shu
- School of Pharmacy, University of Wisconsin, 777 Highland Avenue, Madison, WI 53705, United States; Department of Chemistry, University of Wisconsin, 1101 University Avenue, Madison, WI 53706, United States
| | - Weiping Tang
- School of Pharmacy, University of Wisconsin, 777 Highland Avenue, Madison, WI 53705, United States; Department of Chemistry, University of Wisconsin, 1101 University Avenue, Madison, WI 53706, United States.
| |
Collapse
|
16
|
Small molecules as inhibitors of PCSK9: Current status and future challenges. Eur J Med Chem 2018; 162:212-233. [PMID: 30448414 DOI: 10.1016/j.ejmech.2018.11.011] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/13/2018] [Accepted: 11/05/2018] [Indexed: 12/11/2022]
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) plays an important role in regulating lipoprotein metabolism by binding to low-density lipoprotein receptors (LDLRs), leading to their degradation. LDL cholesterol (LDL-C) lowering drugs that operate through the inhibition of PCSK9 are being pursued for the management of hypercholesterolemia and reducing its associated atherosclerotic cardiovascular disease (CVD) risk. Two PCSK9-blocking monoclonal antibodies (mAbs), alirocumab and evolocumab, were approved in 2015. However, the high costs of PCSK9 antibody drugs impede their prior authorization practices and reduce their long-term adherence. Given the potential of small-molecule drugs, the development of small-molecule PCSK9 inhibitors has attracted considerable attention. This article provides an overview of the recent development of small-molecule PCSK9 inhibitors disclosed in the literature and patent applications, and different approaches that have been pursued to modulate the functional activity of PCSK9 using small molecules are described. Challenges and potential strategies in developing small-molecule PCSK9 inhibitors are also discussed.
Collapse
|
17
|
Ward LM, Kinnett K, Bonewald L. Proceedings of a Parent Project Muscular Dystrophy Bone Health Workshop: Morbidity due to osteoporosis in DMD: The Path Forward May 12-13, 2016, Bethesda, Maryland, USA. Neuromuscul Disord 2017; 28:64-76. [PMID: 28756052 DOI: 10.1016/j.nmd.2017.05.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 05/04/2017] [Accepted: 05/11/2017] [Indexed: 10/19/2022]
Affiliation(s)
- Leanne M Ward
- Division of Endocrinology and Metabolism, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, ON, Canada.
| | - Kathi Kinnett
- Parent Project Muscular Dystrophy, Middletown, OH, USA
| | - Lynda Bonewald
- Indiana Center for Musculoskeletal Health, Departments of Anatomy and Cell Biology and Orthopaedic Surgery, Indiana University, Indianapolis, IN, USA
| | | |
Collapse
|
18
|
Zimmer M, Bista P, Benson EL, Lee DY, Liu F, Picarella D, Vega RB, Vu CB, Yeager M, Ding M, Liang G, Horton JD, Kleemann R, Kooistra T, Morrison MC, Wielinga PY, Milne JC, Jirousek MR, Nichols AJ. CAT-2003: A novel sterol regulatory element-binding protein inhibitor that reduces steatohepatitis, plasma lipids, and atherosclerosis in apolipoprotein E*3-Leiden mice. Hepatol Commun 2017; 1:311-325. [PMID: 29404461 PMCID: PMC5721391 DOI: 10.1002/hep4.1042] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 04/11/2017] [Indexed: 12/21/2022] Open
Abstract
CAT‐2003 is a novel conjugate of eicosapentaenoic acid (EPA) and niacin designed to be hydrolyzed by fatty acid amide hydrolase to release EPA inside cells at the endoplasmic reticulum. In cultured liver cells, CAT‐2003 blocked the maturation of sterol regulatory element‐binding protein (SREBP)‐1 and SREBP‐2 proteins and decreased the expression of multiple SREBP target genes, including HMGCR and PCSK9. Consistent with proprotein convertase subtilisin/kexin type 9 (PCSK9) reduction, both low‐density lipoprotein receptor protein at the cell surface and low‐density lipoprotein particle uptake were increased. In apolipoprotein E*3‐Leiden mice fed a cholesterol‐containing western diet, CAT‐2003 decreased hepatic inflammation and steatosis as evidenced by fewer inflammatory cell aggregates in histopathologic sections, decreased nuclear factor kappa B activity in liver lysates, reduced inflammatory gene expression, reduced intrahepatic cholesteryl ester and triglyceride levels, and decreased liver mass. Plasma PCSK9 was reduced and hepatic low‐density lipoprotein receptor protein expression was increased; plasma cholesterol and triglyceride levels were lowered. Aortic root segments showed reduction of several atherosclerotic markers, including lesion size, number, and severity. CAT‐2003, when dosed in combination with atorvastatin, further lowered plasma cholesterol levels and decreased hepatic expression of SREBP target genes. Conclusion: SREBP inhibition is a promising new strategy for the prevention and treatment of diseases associated with abnormal lipid metabolism, such as atherosclerosis and nonalcoholic steatohepatitis. (Hepatology Communications 2017;1:311–325)
Collapse
Affiliation(s)
| | | | | | | | - Feng Liu
- Catabasis Pharmaceuticals Cambridge MA
| | | | | | - Chi B Vu
- Catabasis Pharmaceuticals Cambridge MA
| | | | - Min Ding
- Department of Molecular Genetics University of Texas Southwestern Medical Center at Dallas Dallas TX
| | - Guosheng Liang
- Department of Molecular Genetics University of Texas Southwestern Medical Center at Dallas Dallas TX
| | - Jay D Horton
- Department of Molecular Genetics University of Texas Southwestern Medical Center at Dallas Dallas TX.,Department of Internal Medicine University of Texas Southwestern Medical Center at Dallas Dallas TX.,Center for Human Nutrition University of Texas Southwestern Medical Center at Dallas Dallas TX
| | - Robert Kleemann
- Netherlands Organization for Applied Scientific Research, Department of Metabolic Health Research Leiden the Netherlands
| | - Teake Kooistra
- Netherlands Organization for Applied Scientific Research, Department of Metabolic Health Research Leiden the Netherlands
| | - Martine C Morrison
- Netherlands Organization for Applied Scientific Research, Department of Metabolic Health Research Leiden the Netherlands
| | - Peter Y Wielinga
- Netherlands Organization for Applied Scientific Research, Department of Metabolic Health Research Leiden the Netherlands
| | | | | | | |
Collapse
|
19
|
Donovan JM, Zimmer M, Offman E, Grant T, Jirousek M. A Novel NF-κB Inhibitor, Edasalonexent (CAT-1004), in Development as a Disease-Modifying Treatment for Patients With Duchenne Muscular Dystrophy: Phase 1 Safety, Pharmacokinetics, and Pharmacodynamics in Adult Subjects. J Clin Pharmacol 2017; 57:627-639. [PMID: 28074489 PMCID: PMC5412838 DOI: 10.1002/jcph.842] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 10/17/2016] [Indexed: 12/11/2022]
Abstract
In Duchenne muscular dystrophy (DMD), NF‐κB is activated in skeletal muscle from infancy regardless of the underlying dystrophin mutation and drives inflammation and muscle degeneration while inhibiting muscle regeneration. Edasalonexent (CAT‐1004) is a bifunctional orally administered small molecule that covalently links 2 compounds known to inhibit NF‐κB, salicylic acid and docosahexaenoic acid (DHA). Edasalonexent is designed to inhibit activated NF‐κB upon intracellular cleavage to these bioactive components. Preclinical data demonstrate disease‐modifying activity in DMD animal models. Three placebo‐controlled studies in adult subjects assessed the safety, pharmacokinetics, and pharmacodynamics of single or multiple edasalonexent doses up to 6000 mg. Seventy‐nine adult subjects received edasalonexent, and 25 received placebo. Pharmacokinetic results were consistent with the intracellular cleavage of edasalonexent to its active components. Food increased plasma exposures of edasalonexent and salicyluric acid, an intracellularly formed metabolite of salicylic acid. The NF‐κB pathway and proteosome gene expression profiles in peripheral mononuclear cells were significantly decreased (P = .02 and P = .002, respectively) after 2 weeks of edasalonexent treatment. NF‐κB activity was inhibited following a single dose of edasalonexent but not by equimolar doses of salicylic acid and DHA. Edasalonexent was well tolerated, and the most common adverse events were mild diarrhea and headache. In first‐in‐human studies, edasalonexent was safe, well tolerated, and inhibited activated NF‐κB pathways, suggesting potential therapeutic utility in DMD regardless of the causative dystrophin mutation, as well as other NF‐κB–mediated diseases.
Collapse
|
20
|
Vu CB, Bridges RJ, Pena-Rasgado C, Lacerda AE, Bordwell C, Sewell A, Nichols AJ, Chandran S, Lonkar P, Picarella D, Ting A, Wensley A, Yeager M, Liu F. Fatty Acid Cysteamine Conjugates as Novel and Potent Autophagy Activators That Enhance the Correction of Misfolded F508del-Cystic Fibrosis Transmembrane Conductance Regulator (CFTR). J Med Chem 2016; 60:458-473. [PMID: 27976892 DOI: 10.1021/acs.jmedchem.6b01539] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
A depressed autophagy has previously been reported in cystic fibrosis patients with the common F508del-CFTR mutation. This report describes the synthesis and preliminary biological characterization of a novel series of autophagy activators involving fatty acid cysteamine conjugates. These molecular entities were synthesized by first covalently linking cysteamine to docosahexaenoic acid. The resulting conjugate 1 synergistically activated autophagy in primary homozygous F508del-CFTR human bronchial epithelial (hBE) cells at submicromolar concentrations. When conjugate 1 was used in combination with the corrector lumacaftor and the potentiator ivacaftor, it showed an additive effect, as measured by the increase in the chloride current in a functional assay. In order to obtain a more stable form for oral dosing, the sulfhydryl group in conjugate 1 was converted into a functionalized disulfide moiety. The resulting conjugate 5 is orally bioavailable in the mouse, rat, and dog and allows a sustained delivery of the biologically active conjugate 1.
Collapse
Affiliation(s)
- Chi B Vu
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Robert J Bridges
- Chicago Medical School, Rosalind Franklin University of Medicine and Science , 3333 Green Bay Road, North Chicago, Illinois 60064, United States
| | - Cecilia Pena-Rasgado
- Chicago Medical School, Rosalind Franklin University of Medicine and Science , 3333 Green Bay Road, North Chicago, Illinois 60064, United States
| | - Antonio E Lacerda
- Charles River Laboratories , 14656 Neo Parkway, Cleveland, Ohio 44128, United States
| | - Curtis Bordwell
- Charles River Laboratories , 14656 Neo Parkway, Cleveland, Ohio 44128, United States
| | - Abby Sewell
- Charles River Laboratories , 14656 Neo Parkway, Cleveland, Ohio 44128, United States
| | - Andrew J Nichols
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Sachin Chandran
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Pallavi Lonkar
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Dominic Picarella
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Amal Ting
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Allison Wensley
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Maisy Yeager
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Feng Liu
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
21
|
Hammers DW, Sleeper MM, Forbes SC, Coker CC, Jirousek MR, Zimmer M, Walter GA, Sweeney HL. Disease-modifying effects of orally bioavailable NF- κB inhibitors in dystrophin-deficient muscle. JCI Insight 2016; 1:e90341. [PMID: 28018975 DOI: 10.1172/jci.insight.90341] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a devastating muscle disease characterized by progressive muscle deterioration and replacement with an aberrant fatty, fibrous matrix. Chronic upregulation of nuclear factor κB (NF-κB) is implicated as a driver of the dystrophic pathogenesis. Herein, 2 members of a novel class of NF-κB inhibitors, edasalonexent (formerly CAT-1004) and CAT-1041, were evaluated in both mdx mouse and golden retriever muscular dystrophy (GRMD) dog models of DMD. These orally bioavailable compounds consist of a polyunsaturated fatty acid conjugated to salicylic acid and potently suppress the pathogenic NF-κB subunit p65/RelA in vitro. In vivo, CAT-1041 effectively improved the phenotype of mdx mice undergoing voluntary wheel running, in terms of activity, muscle mass and function, damage, inflammation, fibrosis, and cardiac pathology. We identified significant increases in dysferlin as a possible contributor to the protective effect of CAT-1041 to sarcolemmal damage. Furthermore, CAT-1041 improved the more severe GRMD phenotype in a canine case study, where muscle mass and diaphragm function were maintained in a treated GRMD dog. These results demonstrate that NF-κB modulation by edasalonexent and CAT-1041 is effective in ameliorating the dystrophic process and these compounds are candidates for new treatments for DMD patients.
Collapse
Affiliation(s)
- David W Hammers
- Department of Physiology and.,Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA.,Department of Pharmacology and Therapeutics.,Myology Institute and
| | - Margaret M Sleeper
- Myology Institute and.,Department of Clinical Studies, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA.,Department of Small Animal Clinical Sciences, University of Florida College of Veterinary Medicine
| | - Sean C Forbes
- Myology Institute and.,Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Cora C Coker
- Department of Pharmacology and Therapeutics.,Myology Institute and
| | | | - Michael Zimmer
- Catabasis Pharmaceuticals, Inc., Cambridge, Massachusetts, USA
| | - Glenn A Walter
- Myology Institute and.,Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida, USA
| | - H Lee Sweeney
- Department of Physiology and.,Department of Pharmacology and Therapeutics.,Myology Institute and
| |
Collapse
|