1
|
Tangallapally R, Subramanian C, Yun MK, Edwards A, Sharma LK, Yang L, Creed K, Wang J, Jackowski S, Rock CO, White SW, Lee RE. Development of Brain Penetrant Pyridazine Pantothenate Kinase Activators. J Med Chem 2024; 67:14432-14442. [PMID: 39136313 PMCID: PMC11345825 DOI: 10.1021/acs.jmedchem.4c01211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024]
Abstract
Conversion of pantothenate to phosphopantothenate in humans is the first dedicated step in the coenzyme A (CoA) biosynthesis pathway and is mediated by four isoforms of pantothenate kinase. These enzymes are allosterically regulated by acyl-CoA levels, which control the rate of CoA biosynthesis. Small molecule activators of the PANK enzymes that overcome feedback suppression increase CoA levels in cultured cells and animals and have shown great potential for the treatment of pantothenate kinase-associated neurodegeneration and propionic acidemias. In this study, we detail the further optimization of PANK pyridazine activators using structure-guided design and focus on the cellular CoA activation potential, metabolic stability, and solubility as the primary drivers of the structure-activity relationship. These studies led to the prioritization of three late-stage preclinical lead PANK modulators with improved pharmacokinetic profiles and the ability to substantially increase brain CoA levels. Compound 22 (BBP-671) eventually advanced into clinical testing for the treatment of PKAN and propionic acidemia.
Collapse
Affiliation(s)
- Rajendra Tangallapally
- Department
of Chemical Biology and Therapeutics, St.
Jude Children’s Research Hospital, 262 Danny Thomas Place, MS1000, Memphis, Tennessee 38105, United States
| | - Chitra Subramanian
- Department
of Host Microbe Interactions, St. Jude Children’s
Research Hospital, 262
Danny Thomas Place, MS1000, Memphis, Tennessee 38105, United States
| | - Mi-Kyung Yun
- Department
of Structural Biology, St. Jude Children’s
Research Hospital, 262
Danny Thomas Place, MS1000, Memphis, Tennessee 38105, United States
| | - Anne Edwards
- Department
of Chemical Biology and Therapeutics, St.
Jude Children’s Research Hospital, 262 Danny Thomas Place, MS1000, Memphis, Tennessee 38105, United States
| | - Lalit Kumar Sharma
- Department
of Chemical Biology and Therapeutics, St.
Jude Children’s Research Hospital, 262 Danny Thomas Place, MS1000, Memphis, Tennessee 38105, United States
- Department
of Host Microbe Interactions, St. Jude Children’s
Research Hospital, 262
Danny Thomas Place, MS1000, Memphis, Tennessee 38105, United States
| | - Lei Yang
- Department
of Chemical Biology and Therapeutics, St.
Jude Children’s Research Hospital, 262 Danny Thomas Place, MS1000, Memphis, Tennessee 38105, United States
| | - Katie Creed
- Department
of Host Microbe Interactions, St. Jude Children’s
Research Hospital, 262
Danny Thomas Place, MS1000, Memphis, Tennessee 38105, United States
| | - Jina Wang
- Department
of Host Microbe Interactions, St. Jude Children’s
Research Hospital, 262
Danny Thomas Place, MS1000, Memphis, Tennessee 38105, United States
| | - Suzanne Jackowski
- Department
of Host Microbe Interactions, St. Jude Children’s
Research Hospital, 262
Danny Thomas Place, MS1000, Memphis, Tennessee 38105, United States
| | - Charles O. Rock
- Department
of Host Microbe Interactions, St. Jude Children’s
Research Hospital, 262
Danny Thomas Place, MS1000, Memphis, Tennessee 38105, United States
| | - Stephen W. White
- Department
of Structural Biology, St. Jude Children’s
Research Hospital, 262
Danny Thomas Place, MS1000, Memphis, Tennessee 38105, United States
| | - Richard E. Lee
- Department
of Chemical Biology and Therapeutics, St.
Jude Children’s Research Hospital, 262 Danny Thomas Place, MS1000, Memphis, Tennessee 38105, United States
| |
Collapse
|
2
|
Ma Y, Jiang M, Javeria H, Tian D, Du Z. Accurate prediction of K p,uu,brain based on experimental measurement of K p,brain and computed physicochemical properties of candidate compounds in CNS drug discovery. Heliyon 2024; 10:e24304. [PMID: 38298681 PMCID: PMC10828645 DOI: 10.1016/j.heliyon.2024.e24304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 02/02/2024] Open
Abstract
A mathematical equation model was developed by building the relationship between the fu,b/fu,p ratio and the computed physicochemical properties of candidate compounds, thereby predicting Kp,uu,brain based on a single experimentally measured Kp,brain value. A total of 256 compounds and 36 marketed published drugs including acidic, basic, neutral, zwitterionic, CNS-penetrant, and non-CNS penetrant compounds with diverse structures and physicochemical properties were involved in this study. A strong correlation was demonstrated between the fu,b/fu,p ratio and physicochemical parameters (CLogP and ionized fraction). The model showed good performance in both internal and external validations. The percentages of compounds with Kp,uu,brain predictions within 2-fold variability were 80.0 %-83.3 %, and more than 90 % were within a 3-fold variability. Meanwhile, "black box" QSAR models constructed by machine learning approaches for predicting fu,b/fu,p ratio based on the chemical descriptors are also presented, and the ANN model displayed the highest accuracy with an RMSE value of 0.27 and 86.7 % of the test set drugs fell within a 2-fold window of linear regression. These models demonstrated strong predictive power and could be helpful tools for evaluating the Kp,uu,brain by a single measurement parameter of Kp,brain during lead optimization for CNS penetration evaluation and ranking CNS drug candidate molecules in the early stages of CNS drug discovery.
Collapse
Affiliation(s)
- Yongfen Ma
- College of Chemistry, Beijing Key Laboratory of Environmentally Harmful Chemical Analysis, Beijing University of Chemical Technology, Beijing, 100029, China
- DMPK Department, Sironax (Beijing) Co., Ltd, Beijing, 102206, China
| | - Mengrong Jiang
- DMPK Department, Sironax (Beijing) Co., Ltd, Beijing, 102206, China
| | - Huma Javeria
- College of Chemistry, Beijing Key Laboratory of Environmentally Harmful Chemical Analysis, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Dingwei Tian
- College of Chemistry, Beijing Key Laboratory of Environmentally Harmful Chemical Analysis, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Zhenxia Du
- College of Chemistry, Beijing Key Laboratory of Environmentally Harmful Chemical Analysis, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
3
|
Jonchere B, Williams J, Zindy F, Liu J, Robinson S, Farmer DM, Min J, Yang L, Stripay JL, Wang Y, Freeman BB, Yu J, Shelat AA, Rankovic Z, Roussel MF. Combination of Ribociclib with BET-Bromodomain and PI3K/mTOR Inhibitors for Medulloblastoma Treatment In Vitro and In Vivo. Mol Cancer Ther 2023; 22:37-51. [PMID: 36318650 PMCID: PMC9808370 DOI: 10.1158/1535-7163.mct-21-0896] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 09/15/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
Despite improvement in the treatment of medulloblastoma over the last years, numerous patients with MYC- and MYCN-driven tumors still fail current therapies. Medulloblastomas have an intact retinoblastoma protein RB, suggesting that CDK4/6 inhibition might represent a therapeutic strategy for which drug combination remains understudied. We conducted high-throughput drug combination screens in a Group3 (G3) medulloblastoma line using the CDK4/6 inhibitor (CDK4/6i) ribociclib at IC20, referred to as an anchor, and 87 oncology drugs approved by FDA or in clinical trials. Bromodomain and extra terminal (BET) and PI3K/mTOR inhibitors potentiated ribociclib inhibition of proliferation in an established cell line and freshly dissociated tumor cells from intracranial xenografts of G3 and Sonic hedgehog (SHH) medulloblastomas in vitro. A reverse combination screen using the BET inhibitor JQ1 as anchor, revealed CDK4/6i as the most potentiating drugs. In vivo, ribociclib showed single-agent activity in medulloblastoma models whereas JQ1 failed to show efficacy due to high clearance and insufficient free brain concentration. Despite in vitro synergy, combination of ribociclib with the PI3K/mTOR inhibitor paxalisib did not significantly improve the survival of G3 and SHH medulloblastoma-bearing mice compared with ribociclib alone. Molecular analysis of ribociclib and paxalisib-treated tumors revealed that E2F targets and PI3K/AKT/MTORC1 signaling genes were depleted, as expected. Importantly, in one untreated G3MB model HD-MB03, the PI3K/AKT/MTORC1 gene set was enriched in vitro compared with in vivo suggesting that the pathway displayed increased activity in vitro. Our data illustrate the difficulty in translating in vitro findings in vivo. See related article in Mol Cancer Ther (2022) 21(8):1306-1317.
Collapse
Affiliation(s)
- Barbara Jonchere
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Justin Williams
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Frederique Zindy
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jingjing Liu
- Department of Tumor Cell Biology Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Sarah Robinson
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Dana M. Farmer
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jaeki Min
- Department of Tumor Cell Biology Chemical Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Lei Yang
- Department of Tumor Cell Biology Chemical Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jennifer L. Stripay
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Yingzhe Wang
- Department of Tumor Cell Biology Preclinical PK Shared Resource, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Burgess B. Freeman
- Department of Tumor Cell Biology Preclinical PK Shared Resource, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jiyang Yu
- Department of Tumor Cell Biology Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Anang A. Shelat
- Department of Tumor Cell Biology Chemical Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Zoran Rankovic
- Department of Tumor Cell Biology Chemical Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Martine F. Roussel
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
4
|
Macabuag N, Esmieu W, Breccia P, Jarvis R, Blackaby W, Lazari O, Urbonas L, Eznarriaga M, Williams R, Strijbosch A, Van de Bospoort R, Matthews K, Clissold C, Ladduwahetty T, Vater H, Heaphy P, Stafford DG, Wang HJ, Mangette JE, McAllister G, Beaumont V, Vogt TF, Wilkinson HA, Doherty EM, Dominguez C. Developing HDAC4-Selective Protein Degraders To Investigate the Role of HDAC4 in Huntington's Disease Pathology. J Med Chem 2022; 65:12445-12459. [PMID: 36098485 PMCID: PMC9512014 DOI: 10.1021/acs.jmedchem.2c01149] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Indexed: 11/30/2022]
Abstract
Huntington's disease (HD) is a lethal autosomal dominant neurodegenerative disorder resulting from a CAG repeat expansion in the huntingtin (HTT) gene. The product of translation of this gene is a highly aggregation-prone protein containing a polyglutamine tract >35 repeats (mHTT) that has been shown to colocalize with histone deacetylase 4 (HDAC4) in cytoplasmic inclusions in HD mouse models. Genetic reduction of HDAC4 in an HD mouse model resulted in delayed aggregation of mHTT, along with amelioration of neurological phenotypes and extended lifespan. To further investigate the role of HDAC4 in cellular models of HD, we have developed bifunctional degraders of the protein and report the first potent and selective degraders of HDAC4 that show an effect in multiple cell lines, including HD mouse model-derived cortical neurons. These degraders act via the ubiquitin-proteasomal pathway and selectively degrade HDAC4 over other class IIa HDAC isoforms (HDAC5, HDAC7, and HDAC9).
Collapse
Affiliation(s)
- Natsuko Macabuag
- Discovery
from Charles River, Charles River, Chesterford Research Park, Saffron Walden CB10 1XL, U.K.
| | - William Esmieu
- Discovery
from Charles River, Charles River, Chesterford Research Park, Saffron Walden CB10 1XL, U.K.
| | - Perla Breccia
- Discovery
from Charles River, Charles River, Chesterford Research Park, Saffron Walden CB10 1XL, U.K.
| | - Rebecca Jarvis
- Discovery
from Charles River, Charles River, Chesterford Research Park, Saffron Walden CB10 1XL, U.K.
| | - Wesley Blackaby
- Discovery
from Charles River, Charles River, Chesterford Research Park, Saffron Walden CB10 1XL, U.K.
| | - Ovadia Lazari
- Discovery
from Charles River, Charles River, Chesterford Research Park, Saffron Walden CB10 1XL, U.K.
| | - Liudvikas Urbonas
- Discovery
from Charles River, Charles River, Chesterford Research Park, Saffron Walden CB10 1XL, U.K.
| | - Maria Eznarriaga
- Discovery
from Charles River, Charles River, Chesterford Research Park, Saffron Walden CB10 1XL, U.K.
| | - Rachel Williams
- Discovery
from Charles River, Charles River, Chesterford Research Park, Saffron Walden CB10 1XL, U.K.
| | | | | | - Kim Matthews
- Discovery
from Charles River, Charles River, Chesterford Research Park, Saffron Walden CB10 1XL, U.K.
| | - Cole Clissold
- Discovery
from Charles River, Charles River, Chesterford Research Park, Saffron Walden CB10 1XL, U.K.
| | - Tammy Ladduwahetty
- Discovery
from Charles River, Charles River, Chesterford Research Park, Saffron Walden CB10 1XL, U.K.
| | - Huw Vater
- Discovery
from Charles River, Charles River, Chesterford Research Park, Saffron Walden CB10 1XL, U.K.
| | - Patrick Heaphy
- Curia, The Conventus Building, 1001 Main
Street, Buffalo, New York 14203, United States
| | - Douglas G. Stafford
- Curia, The Conventus Building, 1001 Main
Street, Buffalo, New York 14203, United States
| | - Hong-Jun Wang
- Curia, The Conventus Building, 1001 Main
Street, Buffalo, New York 14203, United States
| | - John E. Mangette
- Curia, The Conventus Building, 1001 Main
Street, Buffalo, New York 14203, United States
| | - George McAllister
- CHDI
Management/CHDI Foundation, 6080 Center Drive, Los Angeles, California 90045, United States
| | - Vahri Beaumont
- CHDI
Management/CHDI Foundation, 6080 Center Drive, Los Angeles, California 90045, United States
| | - Thomas F. Vogt
- CHDI
Management/CHDI Foundation, 6080 Center Drive, Los Angeles, California 90045, United States
| | - Hilary A. Wilkinson
- CHDI
Management/CHDI Foundation, 6080 Center Drive, Los Angeles, California 90045, United States
| | - Elizabeth M. Doherty
- CHDI
Management/CHDI Foundation, 6080 Center Drive, Los Angeles, California 90045, United States
| | - Celia Dominguez
- CHDI
Management/CHDI Foundation, 6080 Center Drive, Los Angeles, California 90045, United States
| |
Collapse
|
5
|
Fairhurst RA, Furet P, Imbach-Weese P, Stauffer F, Rueeger H, McCarthy C, Ripoche S, Oswald S, Arnaud B, Jary A, Maira M, Schnell C, Guthy DA, Wartmann M, Kiffe M, Desrayaud S, Blasco F, Widmer T, Seiler F, Gutmann S, Knapp M, Caravatti G. Identification of NVP-CLR457 as an Orally Bioavailable Non-CNS-Penetrant pan-Class IA Phosphoinositol-3-Kinase Inhibitor. J Med Chem 2022; 65:8345-8379. [PMID: 35500094 DOI: 10.1021/acs.jmedchem.2c00267] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Balanced pan-class I phosphoinositide 3-kinase inhibition as an approach to cancer treatment offers the prospect of treating a broad range of tumor types and/or a way to achieve greater efficacy with a single inhibitor. Taking buparlisib as the starting point, the balanced pan-class I PI3K inhibitor 40 (NVP-CLR457) was identified with what was considered to be a best-in-class profile. Key to the optimization to achieve this profile was eliminating a microtubule stabilizing off-target activity, balancing the pan-class I PI3K inhibition profile, minimizing CNS penetration, and developing an amorphous solid dispersion formulation. A rationale for the poor tolerability profile of 40 in a clinical study is discussed.
Collapse
Affiliation(s)
- Robin A Fairhurst
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Pascal Furet
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | | | - Frédéric Stauffer
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Heinrich Rueeger
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Clive McCarthy
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Sebastien Ripoche
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Susanne Oswald
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Bertrand Arnaud
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Aline Jary
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Michel Maira
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Christian Schnell
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Daniel A Guthy
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Markus Wartmann
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Michael Kiffe
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | | | - Francesca Blasco
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Toni Widmer
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Frank Seiler
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Sascha Gutmann
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Mark Knapp
- Novartis Institutes for BioMedical Research, Emeryville, California 94608, United States
| | - Giorgio Caravatti
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| |
Collapse
|
6
|
Jiang L, Kumar S, Nuechterlein M, Reyes M, Tran D, Cabebe C, Chiang P, Reynolds J, Carrier S, Sun Y, Eddershaw P, Hay T, Chen W, Feng B. Application of a high‐resolution in vitro human MDR1‐MDCK assay and in vivo studies in preclinical species to improve prediction of CNS drug penetration. Pharmacol Res Perspect 2022; 10:e00932. [PMID: 35156331 PMCID: PMC8929335 DOI: 10.1002/prp2.932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 11/23/2022] Open
Abstract
P‐glycoprotein (P‐gp, MDR1) is expressed at the blood–brain barrier (BBB) and restricts penetration of its substrates into the central nervous system (CNS). In vitro MDR1 assays are frequently used to predict the in vivo relevance of MDR1‐mediated efflux at the BBB. It has been well established that drug candidates with high MDR1 efflux ratios (ERs) display poor CNS penetration. Following a comparison of MDR1 transporter function between the MDR1‐MDCKI cell line from National Institutes of Health (NIH) and our internal MDR1‐MDCKII cell line, the former was found to provide better predictions of in vivo brain penetration than our in‐house MDR1‐MDCKII cell line. In particular, the NIH MDR1 assay has an improved sensitivity to differentiate the compounds with ERs of <3 in our internal cell line and is able to reduce the risk of false negatives. A better correlation between NIH MDR1 ERs and brain penetration in rat and non‐human primate (NHP) was demonstrated. Additionally, a comparison of brain penetration time course of MDR1 substrates and an MDR1 non‐substrate in NHP demonstrated that MDR1 interaction can delay the time to equilibrium of drug concentration in the brain with plasma. It is recommended to select highly permeable compounds without MDR1 interaction for rapid brain penetration to produce the maximal pharmacological effect in the CNS with a quicker onset.
Collapse
Affiliation(s)
| | | | | | | | - Dao Tran
- Vertex Pharmaceuticals Boston Massachusetts USA
| | | | | | | | | | - Yongkai Sun
- Vertex Pharmaceuticals Boston Massachusetts USA
| | | | - Tanya Hay
- Vertex Pharmaceuticals Boston Massachusetts USA
| | | | - Bo Feng
- Vertex Pharmaceuticals Boston Massachusetts USA
| |
Collapse
|
7
|
Gu X, Zhang H, Jiao M, Han B, Zhang Z, Li J, Zhang Q. Histone deacetylase 6 inhibitors with blood-brain barrier penetration as a potential strategy for CNS-Disorders therapy. Eur J Med Chem 2022; 229:114090. [PMID: 34992037 DOI: 10.1016/j.ejmech.2021.114090] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/20/2021] [Accepted: 12/26/2021] [Indexed: 11/27/2022]
Abstract
Histone deacetylase 6 inhibitors (HDAC6is) have been applied to certain cancer diseases and more recently to central nervous system (CNS) disorders including Rett syndrome, Alzheimer's and Parkinson's diseases, and major depressive disorder. Brain penetrance is the major challenge for the development of HDAC6is as potential therapeutics for CNS disorders due in part to the polarity of hydroxamate ZBG. Hence, only a handful of brain-penetrant HDAC6is have been reported and a few display appropriate in vitro and in vivo activities in models of neurological diseases in last decades. This review summarizes the contemporary research being done on HADC6is with brain penetration both the biological pathways involved and the structural modification attempts.
Collapse
Affiliation(s)
- Xiu Gu
- Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, 201203, China; School of Chemistry & Chemical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, China
| | - Hao Zhang
- Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, 201203, China; School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Minru Jiao
- Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, 201203, China
| | - Bo Han
- Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, 201203, China
| | - Zixue Zhang
- Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, 201203, China
| | - Jianqi Li
- Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, 201203, China
| | - Qingwei Zhang
- Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, 201203, China.
| |
Collapse
|
8
|
Haghighijoo Z, Zamani L, Moosavi F, Emami S. Therapeutic potential of quinazoline derivatives for Alzheimer's disease: A comprehensive review. Eur J Med Chem 2022; 227:113949. [PMID: 34742016 DOI: 10.1016/j.ejmech.2021.113949] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/02/2021] [Accepted: 10/22/2021] [Indexed: 12/24/2022]
Abstract
Quinazolines are considered as a promising class of bioactive heterocyclic compounds with broad properties. Particularly, the quinazoline scaffold has an impressive role in the design and synthesis of new CNS-active drugs. The drug-like properties and pharmacological characteristics of quinazoline could lead to different drugs with various targets. Among CNS disorders, Alzheimer's disease (AD) is a progressive neurodegenerative disorder with memory loss, cognitive decline and language dysfunction. AD is a complex and multifactorial disease therefore, the need for finding multi-target drugs against this devastative disease is urgent. A literature survey revealed that quinazoline derivatives have diverse therapeutic potential for AD as modulators/inhibitors of β-amyloid, tau protein, cholinesterases, monoamine oxidases, and phosphodiesterases as well as other protective effects. Thus, we describe here the most relevant and recent studies about anti-AD agents with quinazoline structure which can further aid the development and discovery of new anti-AD agents.
Collapse
Affiliation(s)
- Zahra Haghighijoo
- Department of Chemistry, University of Louisiana at Lafayette, Lafayette, LA, 70504, USA
| | - Leila Zamani
- Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
| | - Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Emami
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
9
|
Xiong B, Wang Y, Chen Y, Xing S, Liao Q, Chen Y, Li Q, Li W, Sun H. Strategies for Structural Modification of Small Molecules to Improve Blood-Brain Barrier Penetration: A Recent Perspective. J Med Chem 2021; 64:13152-13173. [PMID: 34505508 DOI: 10.1021/acs.jmedchem.1c00910] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In the development of central nervous system (CNS) drugs, the blood-brain barrier (BBB) restricts many drugs from entering the brain to exert therapeutic effects. Although many novel delivery methods of large molecule drugs have been designed to assist transport, small molecule drugs account for the vast majority of the CNS drugs used clinically. From this perspective, we review studies from the past five years that have sought to modify small molecules to increase brain exposure. Medicinal chemists make it easier for small molecules to cross the BBB by improving diffusion, reducing efflux, and activating carrier transporters. On the basis of their excellent work, we summarize strategies for structural modification of small molecules to improve BBB penetration. These strategies are expected to provide a reference for the future development of small molecule CNS drugs.
Collapse
Affiliation(s)
- Baichen Xiong
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Yuanyuan Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Ying Chen
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Shuaishuai Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Qinghong Liao
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Qi Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China.,School of Basic Medicine, Qingdao University, Qingdao 266071, People's Republic of China
| | - Wei Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| |
Collapse
|
10
|
Design, synthesis and biological evaluation of brain penetrant benzazepine-based histone deacetylase 6 inhibitors for alleviating stroke-induced brain infarction. Eur J Med Chem 2021; 218:113383. [PMID: 33799069 DOI: 10.1016/j.ejmech.2021.113383] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/13/2021] [Accepted: 03/08/2021] [Indexed: 12/20/2022]
Abstract
Histone deacetylase 6 (HDAC6) has become a promising therapeutic target for central nervous system diseases due to its more complex protein structure and biological functions. However, low brain penetration of reported HDAC6 inhibitors limits its clinical application in neurological disorders. Therefore, the benzazepine, a brain-penetrant rigid fragment, was utilized to design a series of selective HDAC6 inhibitors to improve brain bioavailability. Various synthetic strategies were applied to assemble the tetrahydro-benzazepine ring, and 22 compounds were synthesized. Among them, compound 5 showed low nanomolar potency and strong isozyme selectivity for the inhibition of HDAC6 (IC50 = 1.8 nM, 141-fold selectivity over HDAC1) with efficient binding patterns like coordination with the zinc ion and π-π stacking effect. Western blot results showed it could efficiently transport into SH-SY5Y cells and selectively enhance the acetylation level of α-tubulin with a moderate effect on Histone H3. Notably, pharmacokinetic studies demonstrated that compound 5 (brain/plasma ratio of 2.30) had an excellent ability to penetrate the blood-brain barrier of C57 mice. In male rats with transient middle cerebral artery occlusion (MCAO), compound 5 significantly reduced the cerebral infarction from 21.22% to 11.47% and alleviated neurobehavioral deficits in post-ischemic treatment, which provided a strong rationale for pursuing HDAC6-based therapies for ischemic stroke.
Collapse
|
11
|
Stott AJ, Maillard MC, Beaumont V, Allcock D, Aziz O, Borchers AH, Blackaby W, Breccia P, Creighton-Gutteridge G, Haughan AF, Jarvis RE, Luckhurst CA, Matthews KL, McAllister G, Pollack S, Saville-Stones E, Van de Poël AJ, Vater HD, Vann J, Williams R, Yates D, Muñoz-Sanjuán I, Dominguez C. Evaluation of 5-(Trifluoromethyl)-1,2,4-oxadiazole-Based Class IIa HDAC Inhibitors for Huntington's Disease. ACS Med Chem Lett 2021; 12:380-388. [PMID: 33738065 DOI: 10.1021/acsmedchemlett.0c00532] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/03/2021] [Indexed: 12/25/2022] Open
Abstract
Using an iterative structure-activity relationship driven approach, we identified a CNS-penetrant 5-(trifluoromethyl)-1,2,4-oxadiazole (TFMO, 12) with a pharmacokinetic profile suitable for probing class IIa histone deacetylase (HDAC) inhibition in vivo. Given the lack of understanding of endogenous class IIa HDAC substrates, we developed a surrogate readout to measure compound effects in vivo, by exploiting the >100-fold selectivity compound 12 exhibits over class I/IIb HDACs. We achieved adequate brain exposure with compound 12 in mice to estimate a class I/IIb deacetylation EC50, using class I substrate H4K12 acetylation and global acetylation levels as a pharmacodynamic readout. We observed excellent correlation between the compound 12 in vivo pharmacodynamic response and in vitro class I/IIb cellular activity. Applying the same relationship to class IIa HDAC inhibition, we estimated the compound 12 dose required to inhibit class IIa HDAC activity, for use in preclinical models of Huntington's disease.
Collapse
Affiliation(s)
- Andrew J. Stott
- Charles River Discovery, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, United Kingdom
| | - Michel C. Maillard
- CHDI Management/CHDI Foundation Inc., 6080 Center Drive, Suite 700, Los Angeles, California 90045, United States
| | - Vahri Beaumont
- CHDI Management/CHDI Foundation Inc., 6080 Center Drive, Suite 700, Los Angeles, California 90045, United States
| | - David Allcock
- Charles River Discovery, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, United Kingdom
| | - Omar Aziz
- Charles River Discovery, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, United Kingdom
| | - Alexander H. Borchers
- CHDI Management/CHDI Foundation Inc., 6080 Center Drive, Suite 700, Los Angeles, California 90045, United States
| | - Wesley Blackaby
- Charles River Discovery, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, United Kingdom
| | - Perla Breccia
- Charles River Discovery, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, United Kingdom
| | | | - Alan F. Haughan
- Charles River Discovery, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, United Kingdom
| | - Rebecca E. Jarvis
- Charles River Discovery, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, United Kingdom
| | - Christopher A. Luckhurst
- Charles River Discovery, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, United Kingdom
| | - Kim L. Matthews
- Charles River Discovery, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, United Kingdom
| | - George McAllister
- Charles River Discovery, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, United Kingdom
| | - Scott Pollack
- Charles River Discovery, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, United Kingdom
| | - Elizabeth Saville-Stones
- Charles River Discovery, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, United Kingdom
| | - Amanda J. Van de Poël
- Charles River Discovery, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, United Kingdom
| | - Huw D. Vater
- Charles River Discovery, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, United Kingdom
| | - Julie Vann
- Charles River Discovery, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, United Kingdom
| | - Rachel Williams
- Charles River Discovery, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, United Kingdom
| | - Dawn Yates
- Charles River Discovery, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, United Kingdom
| | - Ignacio Muñoz-Sanjuán
- CHDI Management/CHDI Foundation Inc., 6080 Center Drive, Suite 700, Los Angeles, California 90045, United States
| | - Celia Dominguez
- CHDI Management/CHDI Foundation Inc., 6080 Center Drive, Suite 700, Los Angeles, California 90045, United States
| |
Collapse
|
12
|
Saldaña-Shumaker SL, Grenning AJ, Cunningham CW. Modern approaches to the development of synthetic cannabinoid receptor probes. Pharmacol Biochem Behav 2021; 203:173119. [PMID: 33508249 DOI: 10.1016/j.pbb.2021.173119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 10/13/2020] [Accepted: 01/18/2021] [Indexed: 10/22/2022]
Abstract
The endocannabinoid system, which spans the central and peripheral nervous systems and regulates many biologic processes, is an important target for probe discovery and medications development. Whereas the earliest endocannabinoid receptor probes were derivatives of the non-selective phytocannabinoids isolated from Cannabis species, modern drug discovery techniques have expanded the definitions of what constitutes a CB1R or CB2R cannabinoid receptor ligand. This review highlights recent advances in synthetic cannabinoid receptor chemistry and pharmacology. We provide examples of new CB1R- and CB2R-selective probes, and discuss rational approaches to the design of peripherally-restricted agents. We also describe structural classes of positive- and negative allosteric modulators (PAMs and NAMs) of CB1R and CB2R. Finally, we introduce new opportunities for cannabinoid receptor probe development that have emerged in recent years, including biased agonists that may lead to medications lacking adverse effects.
Collapse
Affiliation(s)
- Savanah L Saldaña-Shumaker
- Department of Pharmaceutical Sciences, Concordia University Wisconsin, 12800 N. Lake Shore Drive, Mequon, WI 53097, USA
| | - Alexander J Grenning
- Department of Chemistry, University of Florida, PO Box 117200, Gainesville, FL 32611, USA
| | - Christopher W Cunningham
- Department of Pharmaceutical Sciences, Concordia University Wisconsin, 12800 N. Lake Shore Drive, Mequon, WI 53097, USA.
| |
Collapse
|
13
|
Noh K, Pietrasiewicz A, Liu X, Wei C. Use of Intravenous Infusion Study Design to Simultaneously Determine Brain Penetration and Systemic Pharmacokinetic Parameters in Rats. Drug Metab Dispos 2020; 49:142-151. [PMID: 33262223 DOI: 10.1124/dmd.120.000242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 11/10/2020] [Indexed: 11/22/2022] Open
Abstract
In drug discovery, the extent of brain penetration as measured by free brain/plasma concentration ratio (Kp,uu) is normally determined from one experiment after constant intravenous infusion, and pharmacokinetics (PK) parameters, including clearance (CL), volume of distribution at steady state (Vss), and effective half-life (t 1/2 ,eff) are determined from another experiment after a single intravenous bolus injection. The objective of the present study was to develop and verify a method to simultaneously determine Kp,uu and PK parameters from a single intravenous infusion experiment. In this study, nine compounds (atenolol, loperamide, minoxidil, N-[3-(4'-fluorophenyl)-3-(4'-phenylphenoxy)propyl]sarcosine, sulpiride, and four proprietary compounds) were intravenously infused for 4 hours at 1 mg/kg or 24 hours at 1 or 6 mg/kg or bolus injected at 1 mg/kg. Plasma samples were serially collected, and brain and cerebrospinal fluid samples were collected at the end of infusion. The PK parameters were obtained using noncompartmental analysis (NCA) and compartmental analysis. The Kp,uu,brain values of those compounds increased up to 2.86-fold from 4 to 24 hours. The CL calculated from infusion rate over steady-state concentration from the 24-hour infusion studies was more consistent with the CL from the intravenous bolus studies than that from 4-hour infusion studies (CL avg. fold of difference 1.19-1.44 vs. 2.10). The compartmental analysis using one- and two-compartment models demonstrated better performance than NCA regardless of study design. In addition, volume of distribution at steady state and t 1/2,eff could be accurately obtained by one-compartment analysis within 2-fold difference. In conclusion, both unbound brain-to-plasma ratio and PK parameters can be successfully estimated from a 24-hour intravenous infusion study design. SIGNIFICANCE STATEMENT: We demonstrated that the extent of brain penetration and pharmacokinetic parameters (such as clearance, Vss, and effective t 1/2) can be determined from a single constant intravenous infusion study in rats.
Collapse
Affiliation(s)
- Keumhan Noh
- Drug Metabolism and Pharmacokinetics, Biogen, Cambridge, Massachusetts
| | | | - Xingrong Liu
- Drug Metabolism and Pharmacokinetics, Biogen, Cambridge, Massachusetts
| | - Cong Wei
- Drug Metabolism and Pharmacokinetics, Biogen, Cambridge, Massachusetts
| |
Collapse
|
14
|
Ingersoll MA, Malloy EA, Caster LE, Holland EM, Xu Z, Zallocchi M, Currier D, Liu H, He DZZ, Min J, Chen T, Zuo J, Teitz T. BRAF inhibition protects against hearing loss in mice. SCIENCE ADVANCES 2020; 6:6/49/eabd0561. [PMID: 33268358 PMCID: PMC7821884 DOI: 10.1126/sciadv.abd0561] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 10/20/2020] [Indexed: 05/13/2023]
Abstract
Hearing loss caused by noise, aging, antibiotics, and chemotherapy affects 10% of the world population, yet there are no Food and Drug Administration (FDA)-approved drugs to prevent it. Here, we screened 162 small-molecule kinase-specific inhibitors for reduction of cisplatin toxicity in an inner ear cell line and identified dabrafenib (TAFINLAR), a BRAF kinase inhibitor FDA-approved for cancer treatment. Dabrafenib and six additional kinase inhibitors in the BRAF/MEK/ERK cellular pathway mitigated cisplatin-induced hair cell death in the cell line and mouse cochlear explants. In adult mice, oral delivery of dabrafenib repressed ERK phosphorylation in cochlear cells, and protected from cisplatin- and noise-induced hearing loss. Full protection was achieved in mice with co-treatment with oral AZD5438, a CDK2 kinase inhibitor. Our study explores a previously unidentified cellular pathway and molecular target BRAF kinase for otoprotection and may advance dabrafenib into clinics to benefit patients with cisplatin- and noise-induced ototoxicity.
Collapse
Affiliation(s)
- Matthew A Ingersoll
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Emma A Malloy
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Lauryn E Caster
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Eva M Holland
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Zhenhang Xu
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE 68178, USA
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Marisa Zallocchi
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Duane Currier
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Huizhan Liu
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - David Z Z He
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Jaeki Min
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jian Zuo
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Tal Teitz
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA.
| |
Collapse
|