1
|
Lang S. Understanding the HIV-CA protein and the ligands that bind at the N-terminal domain (NTD) - C-terminal domain (CTD) interface. RSC Med Chem 2025:d5md00111k. [PMID: 40291137 PMCID: PMC12018806 DOI: 10.1039/d5md00111k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/12/2025] [Indexed: 04/30/2025] Open
Abstract
Treatment and prevention of HIV/AIDS infections represents a significant global challenge, with this being the cause of a substantial number of deaths each year. HIV-CA, the protein responsible for protecting the viral RNA and facilitating reverse transcription, has emerged as an important target in drug discovery. This review applies various computer drug discovery tools for the analysis and understanding of not only the HIV-CA protein, but also the ligands reported to bind to the site at the NTD-CTD interface between two capsid monomer units. Combining this evaluation with reported experimental data, highlights the effects that changes to the ligands make to the binding affinity. This analysis, including identifying areas of the ligand that have not been adequately explored, allows for the generation of guidelines that can be applied to the design of novel ligands that bind to HIV-CA.
Collapse
Affiliation(s)
- Stuart Lang
- New Cambridge House Bassingbourn Road, Litlington Cambridgeshire SG8 0SS UK
| |
Collapse
|
2
|
Yue D, Ng EWH, Hirao H. Hydrogen-Bond-Assisted Catalysis: Hydroxylation of Paclitaxel by Human CYP2C8. J Am Chem Soc 2024; 146:30117-30125. [PMID: 39441858 PMCID: PMC11544615 DOI: 10.1021/jacs.4c07937] [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: 06/12/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
Paclitaxel (PTX, or Taxol), a chemotherapeutic agent widely employed in the treatment of various cancers, undergoes metabolic transformations through the cytochrome P450 enzymes CYP3A4 and CYP2C8. CYP3A4 catalyzes the aromatic hydroxylation reaction of PTX, whereas CYP2C8 demonstrates a distinct reactivity pattern, producing 6α-hydroxypaclitaxel via alkane hydroxylation. Despite the significant impact of PTX metabolism on its anticancer efficacy, the detailed mechanisms underlying these transformations have remained largely unclear. In this study, we employed hybrid quantum mechanics and molecular mechanics (QM/MM) calculations to elucidate the mechanism of PTX metabolism by human CYP2C8. Our QM/MM results reveal that the hydroxylation of PTX by CYP2C8 follows an atypical rebound mechanism. Either of the two hydrogen atoms at the C6 position of PTX can be abstracted, leading to a common radical intermediate. Although the subsequent rebound barrier is unusually high, stereochemical scrambling is unlikely, as the rebound barrier for the formation of the 6α-hydroxylated PTX─the actual product─is significantly lower than that for the 6β-hydroxylated metabolite. Thus, product selectivity is determined by the non-rate-determining rebound step. Furthermore, the hydroxyl group at the C7 position of PTX plays a catalytic role by facilitating the hydrogen abstraction and rebound steps. Our study also confirms a pronounced stability of the transition state in the high-spin sextet spin state, enabled by the enzyme's specific substrate positioning.
Collapse
Affiliation(s)
- Dongxiao Yue
- Warshel
Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, P. R. China
| | - Elvis Wang Hei Ng
- Department
of Pharmacology and Pharmacy, The University
of Hong Kong, Pokfulam 999077, Hong Kong SAR, P. R. China
| | - Hajime Hirao
- Warshel
Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, P. R. China
| |
Collapse
|
3
|
Ali W, Oliver GA, Werz DB, Maiti D. Pd-catalyzed regioselective activation of C(sp 2)-H and C(sp 3)-H bonds. Chem Soc Rev 2024; 53:9904-9953. [PMID: 39212454 DOI: 10.1039/d4cs00408f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Differentiating between two highly similar C-H bonds in a given molecule remains a fundamental challenge in synthetic organic chemistry. Directing group assisted strategies for the functionalisation of proximal C-H bonds has been known for the last few decades. However, distal C-H bond functionalisation is strenuous and requires distinctly specialised techniques. In this review, we summarise the advancement in Pd-catalysed distal C(sp2)-H and C(sp3)-H bond activation through various redox manifolds including Pd(0)/Pd(II), Pd(II)/Pd(IV) and Pd(II)/Pd(0). Distal C-H functionalisation, where a Pd-catalyst is directly involved in the C-H activation step, either through assistance of an external directing group or directed by an inherent functionality or functional group incorporated at the site of the Pd-C bond is covered. The purpose of this review is to portray the current state of art in Pd-catalysed distal C(sp2)-H and C(sp3)-H functionalisation reactions, their mechanism and application in the late-stage functionalisation of medicinal compounds along with highlighting its limitations, thus leaving the field open for further synthetic adjustment.
Collapse
Affiliation(s)
- Wajid Ali
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, 400076, India.
| | - Gwyndaf A Oliver
- Albert-Ludwigs-Universität Freiburg, Institut für Organische Chemie, Albertstraße 21, D-79104 Freiburg, Germany.
| | - Daniel B Werz
- Albert-Ludwigs-Universität Freiburg, Institut für Organische Chemie, Albertstraße 21, D-79104 Freiburg, Germany.
- Freiburg Institute for Advanced Studies, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Debabrata Maiti
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, 400076, India.
- Freiburg Institute for Advanced Studies, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| |
Collapse
|
4
|
Zhang Y, Fan C, Zhang J, Tian X, Zuo W, He K. Lipid-conjugated nucleoside monophosphate and monophosphonate prodrugs: A versatile drug delivery paradigm. Eur J Med Chem 2024; 275:116614. [PMID: 38925014 DOI: 10.1016/j.ejmech.2024.116614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/16/2024] [Accepted: 06/20/2024] [Indexed: 06/28/2024]
Abstract
Integrating lipid conjugation strategies into the design of nucleoside monophosphate and monophosphonate prodrugs is a well-established approach for discovering potential therapeutics. The unique prodrug design endows nucleoside analogues with strong lipophilicity and structures resembling lysoglycerophospholipids, which improve cellular uptake, oral bioavailability and pharmacological activity. In addition, the metabolic stability, pharmacological activity, pharmacokinetic profiles and biodistribution of lipid prodrugs can be finely optimized by adding biostable caps, incorporating transporter-targeted groups, inserting stimulus-responsive bonds, adjusting chain lengths, and applying proper isosteric replacements. This review summarizes recent advances in the structural features and application fields of lipid-conjugated nucleoside monophosphate and monophosphonate prodrugs. This collection provides deep insights into the increasing repertoire of lipid prodrug development strategies and offers design inspirations for medicinal chemists for the development of novel chemotherapeutic agents.
Collapse
Affiliation(s)
- Yanhua Zhang
- College of Science, Xichang University, Sichuan, 615000, China.
| | - Conghua Fan
- Xichang People's Hospital, Xichang, Sichuan, 615000, China
| | - Junjie Zhang
- College of Science, Xichang University, Sichuan, 615000, China
| | - Xin Tian
- College of Science, Xichang University, Sichuan, 615000, China
| | - Wen Zuo
- Xichang People's Hospital, Xichang, Sichuan, 615000, China
| | - Kehan He
- College of Science, Xichang University, Sichuan, 615000, China
| |
Collapse
|
5
|
Wang S, Argikar UA, Chatzopoulou M, Cho S, Crouch RD, Dhaware D, Gu TJ, Heck CJS, Johnson KM, Kalgutkar AS, Liu J, Ma B, Miller GP, Rowley JA, Seneviratne HK, Zhang D, Khojasteh SC. Bioactivation and reactivity research advances - 2023 year in review. Drug Metab Rev 2024; 56:247-284. [PMID: 38963129 DOI: 10.1080/03602532.2024.2376023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/28/2024] [Indexed: 07/05/2024]
Abstract
Advances in the field of bioactivation have significantly contributed to our understanding and prediction of drug-induced liver injury (DILI). It has been established that many adverse drug reactions, including DILI, are associated with the formation and reactivity of metabolites. Modern methods allow us to detect and characterize these reactive metabolites in earlier stages of drug development, which helps anticipate and circumvent the potential for DILI. Improved in silico models and experimental techniques that better reflect in vivo environments are enhancing predictive capabilities for DILI risk. Further, studies on the mechanisms of bioactivation, including enzyme interactions and the role of individual genetic differences, have provided valuable insights for drug optimizations. Cumulatively, this progress is continually refining our approaches to drug safety evaluation and personalized medicine.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Upendra A Argikar
- Non-clinical Development, Bill and Melinda Gates Medical Research Institute, Cambridge, MA, USA
| | | | - Sungjoon Cho
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Rachel D Crouch
- Department of Pharmacy and Pharmaceutical Sciences, Lipscomb University College of Pharmacy, Nashville, TN, USA
| | | | - Ting-Jia Gu
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Carley J S Heck
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Groton, CT, USA
| | - Kevin M Johnson
- Drug Metabolism and Pharmacokinetics, Inotiv, Maryland Heights, MO, USA
| | - Amit S Kalgutkar
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| | - Joyce Liu
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Bin Ma
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Grover P Miller
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - Herana Kamal Seneviratne
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, MD, USA
| | - Donglu Zhang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - S Cyrus Khojasteh
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| |
Collapse
|
6
|
Richter JM, Gunaga P, Yadav N, Bora RO, Bhide R, Rajugowda N, Govindrajulu K, Godesi S, Akuthota N, Rao P, Sivaraman A, Panda M, Kaspady M, Gupta A, Mathur A, Levesque PC, Gulia J, Dokania M, Ramarao M, Kole P, Chacko S, Lentz KA, Sivaprasad Lvj S, Thatipamula RP, Sridhar S, Kamble S, Govindrajan A, Soleman SI, Gordon DA, Wexler RR, Priestley ES. Discovery of BMS-986308: A Renal Outer Medullary Potassium Channel Inhibitor for the Treatment of Heart Failure. J Med Chem 2024; 67:9731-9744. [PMID: 38807539 DOI: 10.1021/acs.jmedchem.4c00893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Recent literature reports highlight the importance of the renal outer medullary potassium (ROMK) channel in renal sodium and potassium homeostasis and emphasize the potential impact that ROMK inhibitors could have as a novel mechanism diuretic in heart failure patients. A series of piperazine-based ROMK inhibitors were designed and optimized to achieve excellent ROMK potency, hERG selectivity, and ADME properties, which led to the identification of compound 28 (BMS-986308). BMS-986308 demonstrated efficacy in the volume-loaded rat diuresis model as well as promising in vitro and in vivo profiles and was therefore advanced to clinical development.
Collapse
Affiliation(s)
- Jeremy M Richter
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Prashantha Gunaga
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Navnath Yadav
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Rajesh Onkardas Bora
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Rajeev Bhide
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Nagendra Rajugowda
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Kavitha Govindrajulu
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Sreenivasulu Godesi
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Nagarjuna Akuthota
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Prasanna Rao
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Aneesh Sivaraman
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Manoranjan Panda
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Mahammed Kaspady
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Anuradha Gupta
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Arvind Mathur
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Paul C Levesque
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Jyoti Gulia
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Manoj Dokania
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Manjunath Ramarao
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Prashant Kole
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Silvi Chacko
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Kimberley A Lentz
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Sankara Sivaprasad Lvj
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | | | - Srikanth Sridhar
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Shyam Kamble
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Arun Govindrajan
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Sharif I Soleman
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - David A Gordon
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Ruth R Wexler
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - E Scott Priestley
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| |
Collapse
|
7
|
Slafer B, Dessoy MA, de Oliveira RG, Mollo MC, Lee E, Matheeussen A, Maes L, Caljon G, Ferreira LLG, Krogh R, Andricopulo AD, Cruz LR, Mowbray CE, Kratz JM, Dias LC. Synthesis and Anti- Trypanosoma cruzi Activity of 3-Cyanopyridine Derivatives. ACS OMEGA 2024; 9:22360-22370. [PMID: 38799347 PMCID: PMC11112591 DOI: 10.1021/acsomega.4c01919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/14/2024] [Accepted: 04/17/2024] [Indexed: 05/29/2024]
Abstract
Chagas disease (CD) is a parasitic neglected tropical disease (NTD) caused by the protozoan Trypanosoma cruzi that affects 6 million people worldwide, often resulting in financial burden, morbidity, and mortality in endemic regions. Given a lack of highly efficient and safe treatments, new, affordable, and fit-for-purpose drugs for CD are urgently needed. In this work, we present a hit-to-lead campaign for novel cyanopyridine analogues as antichagasic agents. In a phenotypic screening against intracellular T. cruzi, hits 1 and 2 were identified and displayed promising potency combined with balanced physicochemical properties. As part of the Lead Optimization Latin America consortium, a set of 40 compounds was designed, synthesized, and tested against T. cruzi intracellular amastigotes and relevant human cell lines. The structural modifications were focused on three positions: cyanopyridine core, linker, and right-hand side. The ADME properties of selected compounds, lipophilicity, kinetic solubility, permeability, and liver microsomal stability, were evaluated. Compounds 1-9 displayed good potency (EC50T. cruzi amastigote <1 μM), and most compounds did not present significant cytotoxicity (CC50 MRC-5 = 32-64 μM). Despite the good balance between potency and selectivity, the antiparasitic activity of the series appeared to be driven by lipophilicity, making the progression of the series unfeasible due to poor ADME properties and potential promiscuity issues.
Collapse
Affiliation(s)
- Brian
W. Slafer
- Institute
of Chemistry, State University of Campinas, Campinas 13083-970, São Paulo, Brazil
| | - Marco A. Dessoy
- Institute
of Chemistry, State University of Campinas, Campinas 13083-970, São Paulo, Brazil
| | - Ramon G. de Oliveira
- Institute
of Chemistry, State University of Campinas, Campinas 13083-970, São Paulo, Brazil
| | - Maria C. Mollo
- Institute
of Chemistry, State University of Campinas, Campinas 13083-970, São Paulo, Brazil
| | - Eun Lee
- Institute
of Chemistry, State University of Campinas, Campinas 13083-970, São Paulo, Brazil
| | - An Matheeussen
- Laboratory
of Microbiology, Parasitology and Hygiene, University of Antwerp, Antwerp 2000, Belgium
| | - Louis Maes
- Laboratory
of Microbiology, Parasitology and Hygiene, University of Antwerp, Antwerp 2000, Belgium
| | - Guy Caljon
- Laboratory
of Microbiology, Parasitology and Hygiene, University of Antwerp, Antwerp 2000, Belgium
| | - Leonardo L. G. Ferreira
- Laboratory
of Medicinal and Computational Chemistry, Physics Institute of Sao Carlos, University of Sao Paulo, Sao Carlos 13566-590, São Paulo, Brazil
| | - Renata Krogh
- Laboratory
of Medicinal and Computational Chemistry, Physics Institute of Sao Carlos, University of Sao Paulo, Sao Carlos 13566-590, São Paulo, Brazil
| | - Adriano D. Andricopulo
- Laboratory
of Medicinal and Computational Chemistry, Physics Institute of Sao Carlos, University of Sao Paulo, Sao Carlos 13566-590, São Paulo, Brazil
| | - Luiza R. Cruz
- Drugs
for Neglected Diseases initiative, Geneva 1202, Switzerland
| | | | - Jadel M. Kratz
- Drugs
for Neglected Diseases initiative, Geneva 1202, Switzerland
| | - Luiz C. Dias
- Institute
of Chemistry, State University of Campinas, Campinas 13083-970, São Paulo, Brazil
| |
Collapse
|
8
|
Gu C, Huang J, Muste C, Zhong J, Walker GS, Obach RS, Shaffer CL. Radiolabel Uncovers Nonintuitive Metabolites of BIIB104: Novel Release of [ 14C]Cyanide from 2-Cyanothiophene and Subsequent Formation of [ 14C]Thiocyanate. Drug Metab Dispos 2024; 52:323-336. [PMID: 38360917 DOI: 10.1124/dmd.123.001462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 01/11/2024] [Accepted: 02/12/2024] [Indexed: 02/17/2024] Open
Abstract
BIIB104 (formerly PF-04958242), N-((3S,4S)-4-(4-(5-cyanothiophen-2-yl)phenoxy)tetrahydrofuran-3-yl)propane-2-sulfonamide, is an α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor potentiator investigated for the treatment of cognitive impairment associated with schizophrenia. Preliminary in vitro metabolism studies with non-radiolabeled BIIB104 in rat, dog, and human liver microsomes (RLM, DLM, and HLM) showed O-dealkylation in all three species, tetrahydrofuran hydroxylation dominating in DLM and HLM, and thiophene hydroxylation prevalent in RLM. However, a subsequent rat mass balance study with [nitrile-14C]BIIB104 showed incomplete recovery of administered radioactivity (∼80%) from urine and feces over 7 days following an oral dose, and an exceptionally long plasma total radioactivity half-life. Radiochromatographic metabolite profiling and identification, including chemical derivation, revealed that [14C]cyanide was a major metabolite of [nitrile-14C]BIIB104 in RLM, but a minor and trace metabolite in DLM and HLM, respectively. Correspondingly in bile duct-cannulated rats, [14C]thiocyanate accounted for ∼53% of total radioactivity excreted over 48 hours postdose and it, as an endogenous substance, explained the exceptionally long plasma radioactivity half-life. The release of [14C]cyanide from the 2-cyanothiophene moiety is postulated to follow an epoxidation-initiated thiophene-opening based on the detection of non-radiolabeled counterpart metabolites in RLM. This unusual biotransformation serves as a lesson regarding placement of the radioactive label on an aryl nitrile when material will be used for evaluating the metabolism of a new drug candidate. Additionally, the potential cyanide metabolite of nitrile-containing drug molecules may be detected in liver microsomes with liquid chromatography-mass spectrometry following a chemical derivatization. SIGNIFICANCE STATEMENT: Using [nitrile-14C]BIIB104, non-intuitive metabolites of BIIB104 were discovered involving a novel cyanide release from the 2-cyanothiophene motif via a postulated epoxidation-initiated thiophene-opening. This unusual biotransformation serves as a lesson regarding placement of the radioactive label on an aryl nitrile when material will be used for evaluating the metabolism of a new drug candidate.
Collapse
Affiliation(s)
- Chungang Gu
- Drug Metabolism and Pharmacokinetics (C.G., J.H., C.M.), External Innovation Unit (C.L.S.), and Physical Biochemistry (J.Z.), Biogen, Inc., Cambridge, Massachusetts and Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (G.S.W., R.S.O.)
| | - Jiansheng Huang
- Drug Metabolism and Pharmacokinetics (C.G., J.H., C.M.), External Innovation Unit (C.L.S.), and Physical Biochemistry (J.Z.), Biogen, Inc., Cambridge, Massachusetts and Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (G.S.W., R.S.O.)
| | - Cathy Muste
- Drug Metabolism and Pharmacokinetics (C.G., J.H., C.M.), External Innovation Unit (C.L.S.), and Physical Biochemistry (J.Z.), Biogen, Inc., Cambridge, Massachusetts and Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (G.S.W., R.S.O.)
| | - Jeremy Zhong
- Drug Metabolism and Pharmacokinetics (C.G., J.H., C.M.), External Innovation Unit (C.L.S.), and Physical Biochemistry (J.Z.), Biogen, Inc., Cambridge, Massachusetts and Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (G.S.W., R.S.O.)
| | - Gregory S Walker
- Drug Metabolism and Pharmacokinetics (C.G., J.H., C.M.), External Innovation Unit (C.L.S.), and Physical Biochemistry (J.Z.), Biogen, Inc., Cambridge, Massachusetts and Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (G.S.W., R.S.O.)
| | - R Scott Obach
- Drug Metabolism and Pharmacokinetics (C.G., J.H., C.M.), External Innovation Unit (C.L.S.), and Physical Biochemistry (J.Z.), Biogen, Inc., Cambridge, Massachusetts and Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (G.S.W., R.S.O.)
| | - Christopher L Shaffer
- Drug Metabolism and Pharmacokinetics (C.G., J.H., C.M.), External Innovation Unit (C.L.S.), and Physical Biochemistry (J.Z.), Biogen, Inc., Cambridge, Massachusetts and Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (G.S.W., R.S.O.)
| |
Collapse
|
9
|
Clune S, Awolade P, Zhou Q, Esquer H, Matter B, Kearns JT, Kellett T, Akintayo DC, Kompella UB, LaBarbera DV. The validation of new CHD1L inhibitors as a therapeutic strategy for cancer. Biomed Pharmacother 2024; 170:116037. [PMID: 38128184 PMCID: PMC10792906 DOI: 10.1016/j.biopha.2023.116037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/04/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Chromodomain helicase DNA-binding protein 1 like (CHD1L) is an oncogene that promotes tumor progression, metastasis, and multidrug resistance. CHD1L expression is indicative of poor outcomes and low survival in cancer patients with various cancer types. Herein, we report a set of CHD1L inhibitors (CHD1Li) discovered from high-throughput screening and evaluated using enzyme inhibition, 3D tumor organoid cytotoxicity and mechanistic assays. The structurally distinct compounds 8-11 emerged as hits with promising bioactivity by targeting CHD1L. CHD1Li were further examined for their stability in human and mouse liver microsomes, which showed compounds 9 and 11 to be the most metabolically stable. Additionally, molecular modeling studies of CHD1Li with the target protein shed light on key pharmacophore features driving CHD1L binding. Taken together, these results expand the chemical space of CHD1Li as a potential targeted therapy for colorectal cancer and other cancers.
Collapse
Affiliation(s)
- Sophia Clune
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Paul Awolade
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA; The CU Anschutz Center for Drug Discovery, USA
| | - Qiong Zhou
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA; The CU Anschutz Center for Drug Discovery, USA; The CU Cancer Center, USA
| | - Hector Esquer
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA; The CU Anschutz Center for Drug Discovery, USA; The CU Cancer Center, USA
| | - Brock Matter
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jeffrey T Kearns
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Timothy Kellett
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Damilola Caleb Akintayo
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Uday B Kompella
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA; The CU Anschutz Center for Drug Discovery, USA; The CU Cancer Center, USA
| | - Daniel V LaBarbera
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA; The CU Anschutz Center for Drug Discovery, USA; The CU Cancer Center, USA.
| |
Collapse
|
10
|
Choi K. Structure-property Relationships Reported for the New Drugs Approved in 2022. Mini Rev Med Chem 2024; 24:330-340. [PMID: 37211842 DOI: 10.2174/1389557523666230519162803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/20/2023] [Accepted: 03/15/2023] [Indexed: 05/23/2023]
Abstract
BACKGROUND The structure-property relationship illustrates how modifying the chemical structure of a pharmaceutical compound influences its absorption, distribution, metabolism, excretion, and other related properties. Understanding structure-property relationships of clinically approved drugs could provide useful information for drug design and optimization strategies. METHOD Among new drugs approved around the world in 2022, including 37 in the US, structure- property relationships of seven drugs were compiled from medicinal chemistry literature, in which detailed pharmacokinetic and/or physicochemical properties were disclosed not only for the final drug but also for its key analogues generated during drug development. RESULTS The discovery campaigns for these seven drugs demonstrate extensive design and optimization efforts to identify suitable candidates for clinical development. Several strategies have been successfully employed, such as attaching a solubilizing group, bioisosteric replacement, and deuterium incorporation, resulting in new compounds with enhanced physicochemical and pharmacokinetic properties. CONCLUSION The structure-property relationships hereby summarized illustrate how proper structural modifications could successfully improve the overall drug-like properties. The structure-property relationships of clinically approved drugs are expected to continue to provide valuable references and guides for the development of future drugs.
Collapse
Affiliation(s)
- Kihang Choi
- Department of Chemistry, Korea University, Seoul, 02841, Korea (ROK)
| |
Collapse
|
11
|
Wang S, Ballard TE, Christopher LJ, Foti RS, Gu C, Khojasteh SC, Liu J, Ma S, Ma B, Obach RS, Schadt S, Zhang Z, Zhang D. The Importance of Tracking "Missing" Metabolites: How and Why? J Med Chem 2023; 66:15586-15612. [PMID: 37769129 DOI: 10.1021/acs.jmedchem.3c01293] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
Technologies currently employed to find and identify drug metabolites in complex biological matrices generally yield results that offer a comprehensive picture of the drug metabolite profile. However, drug metabolites can be missed or are captured only late in the drug development process. This could be due to a variety of factors, such as metabolism that results in partial loss of the molecule, covalent bonding to macromolecules, the drug being metabolized in specific human tissues, or poor ionization in a mass spectrometer. These scenarios often draw a great deal of attention from chemistry, safety assessment, and pharmacology. This review will summarize scenarios of missing metabolites, why they are missing, and associated uncovering strategies from deeper investigations. Uncovering previously missed metabolites can have ramifications in drug development with toxicological and pharmacological consequences, and knowledge of these can help in the design of new drugs.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - T Eric Ballard
- Takeda Development Center Americas, Inc., 35 Landsdowne St, Cambridge, Massachusetts 02139, United States
| | - Lisa J Christopher
- Department of Clinical Pharmacology, Pharmacometrics, Disposition & Bioanalysis, Bristol-Myers Squibb, Route 206 & Province Line Road, Princeton, New Jersey 08543, United States
| | - Robert S Foti
- Preclinical Development, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Chungang Gu
- Drug Metabolism and Pharmacokinetics, Biogen Inc., 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - S Cyrus Khojasteh
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Joyce Liu
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Shuguang Ma
- Drug Metabolism and Pharmacokinetics, Pliant Therapeutics, 260 Littlefield Avenue, South San Francisco, California 94080, United States
| | - Bin Ma
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - R Scott Obach
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer, Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Simone Schadt
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacher Strasse 124, 4070 Basel, Switzerland
| | - Zhoupeng Zhang
- DMPK Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Donglu Zhang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
12
|
Chambers RK, Weaver JD, Kim J, Hoar JL, Krska SW, White MC. A preparative small-molecule mimic of liver CYP450 enzymes in the aliphatic C-H oxidation of carbocyclic N-heterocycles. Proc Natl Acad Sci U S A 2023; 120:e2300315120. [PMID: 37428920 PMCID: PMC10629554 DOI: 10.1073/pnas.2300315120] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 05/15/2023] [Indexed: 07/12/2023] Open
Abstract
An emerging trend in small-molecule pharmaceuticals, generally composed of nitrogen heterocycles (N-heterocycles), is the incorporation of aliphatic fragments. Derivatization of the aliphatic fragments to improve drug properties or identify metabolites often requires lengthy de novo syntheses. Cytochrome P450 (CYP450) enzymes are capable of direct site- and chemo-selective oxidation of a broad range of substrates but are not preparative. A chemoinformatic analysis underscored limited structural diversity of N-heterocyclic substrates oxidized using chemical methods relative to pharmaceutical chemical space. Here, we describe a preparative chemical method for direct aliphatic oxidation that tolerates a wide range of nitrogen functionality (chemoselective) and matches the site of oxidation (site-selective) of liver CYP450 enzymes. Commercial small-molecule catalyst Mn(CF3-PDP) selectively effects direct methylene oxidation in compounds bearing 25 distinct heterocycles including 14 out of 27 of the most frequent N-heterocycles found in U.S. Food and Drug Administration (FDA)-approved drugs. Mn(CF3-PDP) oxidations of carbocyclic bioisostere drug candidates (for example, HCV NS5B and COX-2 inhibitors including valdecoxib and celecoxib derivatives) and precursors of antipsychotic drugs blonanserin, buspirone, and tiospirone and the fungicide penconazole are demonstrated to match the major site of aliphatic metabolism obtained with liver microsomes. Oxidations are demonstrated at low Mn(CF3-PDP) loadings (2.5 to 5 mol%) on gram scales of substrate to furnish preparative amounts of oxidized products. A chemoinformatic analysis supports that Mn(CF3-PDP) significantly expands the pharmaceutical chemical space accessible to small-molecule C-H oxidation catalysis.
Collapse
Affiliation(s)
- Rachel K. Chambers
- Department of Chemistry, Roger Adams Laboratory, University of Illinois, Urbana, IL61801
| | - Jacob D. Weaver
- Department of Chemistry, Roger Adams Laboratory, University of Illinois, Urbana, IL61801
| | - Jinho Kim
- Department of Chemistry, Roger Adams Laboratory, University of Illinois, Urbana, IL61801
| | - Jason L. Hoar
- Department of Discovery Chemistry, Merck & Co., Inc., Rahway, NJ07065
| | - Shane W. Krska
- Department of Discovery Chemistry, Merck & Co., Inc., Rahway, NJ07065
| | - M. Christina White
- Department of Chemistry, Roger Adams Laboratory, University of Illinois, Urbana, IL61801
| |
Collapse
|
13
|
Sloane S, Vang ZP, Nelson G, Qi L, Sonstrom RE, Alansari IY, Behlow KT, Pate BH, Neufeldt SR, Clark JR. Precision Deuteration Using Cu-Catalyzed Transfer Hydrodeuteration to Access Small Molecules Deuterated at the Benzylic Position. JACS AU 2023; 3:1583-1589. [PMID: 37388686 PMCID: PMC10301681 DOI: 10.1021/jacsau.3c00053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 07/01/2023]
Abstract
A highly regio- and chemoselective Cu-catalyzed aryl alkyne transfer hydrodeuteration to access a diverse scope of aryl alkanes precisely deuterated at the benzylic position is described. The reaction benefits from a high degree of regiocontrol in the alkyne hydrocupration step, leading to the highest selectivities reported to date for an alkyne transfer hydrodeuteration reaction. Only trace isotopic impurities are formed under this protocol, and analysis of an isolated product by molecular rotational resonance spectroscopy confirms that high isotopic purity products can be generated from readily accessible aryl alkyne substrates.
Collapse
Affiliation(s)
- Samantha
E. Sloane
- Department
of Chemistry, Marquette University, Milwaukee, Wisconsin 53233-1881, United
States
| | - Zoua Pa Vang
- Department
of Chemistry, Marquette University, Milwaukee, Wisconsin 53233-1881, United
States
| | - Genevieve Nelson
- Department
of Chemistry & Biochemistry, Montana
State University, Bozeman, Montana 59717, United States
| | - Lihan Qi
- Department
of Chemistry, Marquette University, Milwaukee, Wisconsin 53233-1881, United
States
| | | | - Isabella Y. Alansari
- Department
of Chemistry, Marquette University, Milwaukee, Wisconsin 53233-1881, United
States
| | - Kiera T. Behlow
- Department
of Chemistry, Marquette University, Milwaukee, Wisconsin 53233-1881, United
States
| | - Brooks H. Pate
- Department
of Chemistry, University of Virginia, Charlottesville, Virginia 22904-4319, United
States
| | - Sharon R. Neufeldt
- Department
of Chemistry & Biochemistry, Montana
State University, Bozeman, Montana 59717, United States
| | - Joseph R. Clark
- Department
of Chemistry, Marquette University, Milwaukee, Wisconsin 53233-1881, United
States
| |
Collapse
|
14
|
Hwang DJ, He Y, Ponnusamy S, Thiyagarajan T, Mohler ML, Narayanan R, Miller DD. Metabolism-Guided Selective Androgen Receptor Antagonists: Design, Synthesis, and Biological Evaluation for Activity against Enzalutamide-Resistant Prostate Cancer. J Med Chem 2023; 66:3372-3392. [PMID: 36825758 PMCID: PMC10243532 DOI: 10.1021/acs.jmedchem.2c01858] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
A major challenge for new drug discovery in the area of androgen receptor (AR) antagonists lies in predicting the druggable properties that will enable small molecules to retain their potency and stability during further studies in vitro and in vivo. Indole (compound 8) is a first-in-class AR antagonist with very high potency (IC50 = 0.085 μM) but is metabolically unstable. During the metabolic studies described herein, we synthesized new small molecules that exhibit significantly improved stability while retaining potent antagonistic activity for an AR. This structure-activity relationship (SAR) study of more than 50 compounds classified with three classes (Class I, II, and III) and discovered two compounds (32c and 35i) that are potent AR antagonists (e.g., IC50 = 0.021 μM, T1/2 = 120 min for compound 35i). The new antagonists exhibited improved in vivo pharmacokinetics (PK) with high efficacy antiandrogen activity in Hershberger and antiandrogen Enz-Res tumor xenograft models that overexpress AR (LNCaP-AR).
Collapse
Affiliation(s)
- Dong-Jin Hwang
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Yali He
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Suriyan Ponnusamy
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Thirumagal Thiyagarajan
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Michael L Mohler
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Ramesh Narayanan
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Duane D Miller
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
15
|
Hintzsche SJ, Vang ZP, Rivera Torres E, Podoski M, Clark JR. Highly selective catalytic transfer hydrodeuteration of cyclic alkenes. J Labelled Comp Radiopharm 2023; 66:86-94. [PMID: 36772856 DOI: 10.1002/jlcr.4015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023]
Abstract
Selective deuterium installation into small molecules is becoming increasingly desirable not only for the elucidation of mechanistic pathways and studying biological processes but also because of deuterium's ability to favorably adjust the pharmacokinetic parameters of bioactive molecules. Fused bicyclic moieties, especially those containing heteroatoms, are prevalent in drug discovery and pharmaceuticals. Herein, we report a copper-catalyzed transfer hydrodeuteration of cyclic and heterocyclic alkenes, which enables the synthesis of chromans, quinolinones, and tetrahydronaphthalenes that are precisely deuterated at the benzylic position. We also demonstrate the ability to place one deuterium atom at the homobenzylic site of these scaffolds with high regioselectivity by swapping transfer reagents for their isotopic analogs. Furthermore, examples of chemoselective transfer hydrogenation and transfer deuteration are disclosed, allowing for the simultaneous incorporation of two vicinal hydrogen or deuterium atoms into a double bond.
Collapse
Affiliation(s)
- Samuel J Hintzsche
- Department of Chemistry, Marquette University, Milwaukee, Wisconsin, USA
| | - Zoua Pa Vang
- Department of Chemistry, Marquette University, Milwaukee, Wisconsin, USA
| | | | - Mykaela Podoski
- Department of Chemistry, Marquette University, Milwaukee, Wisconsin, USA
| | - Joseph R Clark
- Department of Chemistry, Marquette University, Milwaukee, Wisconsin, USA
| |
Collapse
|
16
|
Yao H, Sherer EC, Lu M, Small J, Martin GE, Lam YH, Chen Q, Helmy R, Liu Y, Chen H. One-Step Regio- and Stereoselective Electrochemical Synthesis of Orexin Receptor Antagonist Oxidative Metabolites. J Org Chem 2022; 87:15011-15021. [PMID: 36322780 PMCID: PMC10512451 DOI: 10.1021/acs.joc.2c01311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Synthesis of drug metabolites, which often have complex structures, is an integral step in the evaluation of drug candidate metabolism, pharmacokinetic (PK) properties, and safety profiles. Frequently, such synthetic endeavors entail arduous, multiple-step de novo synthetic routes. Herein, we present the one-step Shono-type electrochemical synthesis of milligrams of chiral α-hydroxyl amide metabolites of two orexin receptor antagonists, MK-8133 and MK-6096, as revealed by a small-scale (pico- to nano-mole level) reaction screening using a lab-built online electrochemistry (EC)/mass spectrometry (MS) (EC/MS) platform. The electrochemical oxidation of MK-8133 and MK-6096 was conducted in aqueous media and found to produce the corresponding α-piperidinols with exclusive regio- and stereoselectivity, as confirmed by high-resolution nuclear magnetic resonance (NMR) characterization of products. Based on density functional theory (DFT) calculations, the exceptional regio- and stereoselectivity for this electrochemical oxidation are governed by more favorable energetics of the transition state, leading to the preferred secondary carbon radical α to the amide group and subsequent steric hindrance associated with the U-shaped conformation of the cation derived from the secondary α-carbon radical, respectively.
Collapse
Affiliation(s)
- Huifang Yao
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, MRL, Merck & Co., Inc., PO Box 2000, Rahway, NJ 07065, USA
| | - Edward C. Sherer
- Analytical Research and Development, MRL, Merck & Co., Inc., PO Box 2000, Rahway, NJ 07065, USA
| | - Mei Lu
- Department of Chemistry & Biochemistry, Ohio University, Athens, OH 45701, USA
| | - James Small
- Analytical Research and Development, MRL, Merck & Co., Inc., PO Box 2000, Rahway, NJ 07065, USA
| | - Gary E. Martin
- Analytical Research and Development, MRL, Merck & Co., Inc., PO Box 2000, Rahway, NJ 07065, USA
| | - Yu-hong Lam
- Computational and Structural Chemistry, MRL, Merck & Co., Inc., PO Box 2000, Rahway, NJ 07065, USA
| | - Qinghao Chen
- Process Research and Development, MRL, Merck & Co., Inc., PO Box 2000, Rahway, NJ 07065, USA
| | - Roy Helmy
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, MRL, Merck & Co., Inc., PO Box 2000, Rahway, NJ 07065, USA
| | - Yong Liu
- Analytical Research and Development, MRL, Merck & Co., Inc., PO Box 2000, Rahway, NJ 07065, USA
| | - Hao Chen
- Department of Chemistry & Biochemistry, Ohio University, Athens, OH 45701, USA
- Department of Chemistry & Environmental Science, New Jersey Institute of Technology, Newark, NJ 07102, USA
| |
Collapse
|
17
|
Iwaki K, Maruno K, Nagata O, Shibata N. Ethynyl-SF 4-Pyridines: Reagents for SF 4-Alkynylation to Carbonyl Compounds. J Org Chem 2022; 87:6302-6311. [PMID: 35437010 DOI: 10.1021/acs.joc.2c00564] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The trans-tetrafluoro-λ6-sulfanyl (SF4) unit is medicinally attractive because of its high electronegativity, lipophilicity, and unique hypervalent structure. The trans-SF4 unit can characteristically connect two independent molecules linearly. However, there is no example of the use of this unit for medicinal chemistry due to difficulties in synthesis. We report the first synthesis of (ethynyl-trans-tetrafluoro-λ6-sulfanyl)pyridines (t-ethynyl-SF4-pyridines) and their use as versatile reagents for the first direct SF4-alkynylation to carbonyl compounds. The addition reaction of t-ethynyl-SF4-pyridines to the carbonyl group in the presence of MeLi smoothly afforded pyridine-SF4-propargylic tertiary and secondary alcohols in high yields.
Collapse
Affiliation(s)
- Kentaro Iwaki
- Department of Nanopharmaceutical Sciences & Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Gokiso, Showa-ku, Nagoya 466-8555, Japan
| | - Koki Maruno
- Department of Nanopharmaceutical Sciences & Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Gokiso, Showa-ku, Nagoya 466-8555, Japan
| | - Osamu Nagata
- Pharmaceutical Division, Ube Industries, Ltd., Seavans North Bldg, 1-2-1 Shibaura, Minato-ku, Tokyo 105-8449, Japan
| | - Norio Shibata
- Department of Nanopharmaceutical Sciences & Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Gokiso, Showa-ku, Nagoya 466-8555, Japan
| |
Collapse
|
18
|
Sorrentino JP, Altman RA. Fluoroalkylation of Dextromethorphan Improves CNS Exposure and Metabolic Stability. ACS Med Chem Lett 2022; 13:707-713. [PMID: 35450379 PMCID: PMC9014517 DOI: 10.1021/acsmedchemlett.2c00055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/11/2022] [Indexed: 11/28/2022] Open
Abstract
Aryl-methyl ethers, while present in many bioactive compounds, are subject to rapid O-dealkylation, which can generate bioinactive or toxic metabolites. Such is the case for dextromethorphan, which readily undergoes P450 mediated O-dealkylation to provide the psychoactive phenolic metabolite dextrorphan, an N-methyl-d-aspartate (NMDA) receptor antagonist that causes hallucinations and encourages recreational abuse. As a general strategy to minimize this undesired degradation, both deuteration and fluorination strategies might be exploited, though such strategies have rarely been compared in matched series. In this manuscript, we designed, synthesized, and evaluated in vitro and in vivo new fluoroalkyl analogs of dextromethorphan and D3-dextromethorphan that minimize metabolic degradation and increased CNS exposure relative to dextromethorphan and related deuterated analogs currently in clinical trials.
Collapse
Affiliation(s)
- Jacob P. Sorrentino
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Ryan A. Altman
- Department of Medicinal Chemistry and Molecular Pharmacology and Department of Chemistry, Purdue University, West Lafayette, Indiana 47906, United States
| |
Collapse
|
19
|
Reyes A, Torres ER, Vang ZP, Clark JR. Highly Regioselective Copper-Catalyzed Transfer Hydrodeuteration of Unactivated Terminal Alkenes. Chemistry 2021; 28:e202104340. [PMID: 34882859 DOI: 10.1002/chem.202104340] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Indexed: 12/21/2022]
Abstract
Catalytic transfer hydrodeuteration of unactivated alkenes is challenging because of the requirement that chemically similar hydrogen and deuterium undergo selective insertion across a π-bond. We now report a highly regioselective catalytic transfer hydrodeuteration of unactivated terminal alkenes across a variety of heteroatom- or heterocycle-containing substrates. The base-metal-catalyzed reaction is also demonstrated on two complex natural products. Reaction studies indicate modular conditions that can also be extended to perform either an alkene transfer hydrogenation or transfer deuteration.
Collapse
Affiliation(s)
- Albert Reyes
- Department of Chemistry, Marquette University, Milwaukee, WI 53233-1881, USA
| | | | - Zoua Pa Vang
- Department of Chemistry, Marquette University, Milwaukee, WI 53233-1881, USA
| | - Joseph R Clark
- Department of Chemistry, Marquette University, Milwaukee, WI 53233-1881, USA
| |
Collapse
|
20
|
Sinha SK, Guin S, Maiti S, Biswas JP, Porey S, Maiti D. Toolbox for Distal C-H Bond Functionalizations in Organic Molecules. Chem Rev 2021; 122:5682-5841. [PMID: 34662117 DOI: 10.1021/acs.chemrev.1c00220] [Citation(s) in RCA: 212] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Transition metal catalyzed C-H activation has developed a contemporary approach to the omnipresent area of retrosynthetic disconnection. Scientific researchers have been tempted to take the help of this methodology to plan their synthetic discourses. This paradigm shift has helped in the development of industrial units as well, making the synthesis of natural products and pharmaceutical drugs step-economical. In the vast zone of C-H bond activation, the functionalization of proximal C-H bonds has gained utmost popularity. Unlike the activation of proximal C-H bonds, the distal C-H functionalization is more strenuous and requires distinctly specialized techniques. In this review, we have compiled various methods adopted to functionalize distal C-H bonds, mechanistic insights within each of these procedures, and the scope of the methodology. With this review, we give a complete overview of the expeditious progress the distal C-H activation has made in the field of synthetic organic chemistry while also highlighting its pitfalls, thus leaving the field open for further synthetic modifications.
Collapse
Affiliation(s)
- Soumya Kumar Sinha
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Srimanta Guin
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Sudip Maiti
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Jyoti Prasad Biswas
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Sandip Porey
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Debabrata Maiti
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
21
|
Ripenko V, Vysochyn D, Klymov I, Zhersh S, Mykhailiuk PK. Large-Scale Synthesis and Modifications of Bicyclo[1.1.1]pentane-1,3-dicarboxylic Acid (BCP). J Org Chem 2021; 86:14061-14068. [PMID: 34166594 PMCID: PMC8524415 DOI: 10.1021/acs.joc.1c00977] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
![]()
In flow photochemical addition of propellane to
diacetyl allowed construction of the bicyclo[1.1.1]pentane (BCP) core
in a 1 kg scale within 1 day. Haloform reaction of the formed diketone
in batch afforded bicyclo[1.1.1]pentane-1,3-dicarboxylic acid in a
multigram amount. Representative gram scale transformations of the
diacid were also performed to obtain various BCP-containing building
blocks—alcohols, acids, amines, trifluoroborates, amino acids, etc.—for medicinal chemistry.
Collapse
Affiliation(s)
- Vasyl Ripenko
- Enamine Ltd., Chervonotkatska 78, 02094 Kyiv, Ukraine
| | | | - Ivan Klymov
- Enamine Ltd., Chervonotkatska 78, 02094 Kyiv, Ukraine
| | - Serhii Zhersh
- Enamine Ltd., Chervonotkatska 78, 02094 Kyiv, Ukraine
| | | |
Collapse
|
22
|
Candish L, Collins KD, Cook GC, Douglas JJ, Gómez-Suárez A, Jolit A, Keess S. Photocatalysis in the Life Science Industry. Chem Rev 2021; 122:2907-2980. [PMID: 34558888 DOI: 10.1021/acs.chemrev.1c00416] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the pursuit of new pharmaceuticals and agrochemicals, chemists in the life science industry require access to mild and robust synthetic methodologies to systematically modify chemical structures, explore novel chemical space, and enable efficient synthesis. In this context, photocatalysis has emerged as a powerful technology for the synthesis of complex and often highly functionalized molecules. This Review aims to summarize the published contributions to the field from the life science industry, including research from industrial-academic partnerships. An overview of the synthetic methodologies developed and strategic applications in chemical synthesis, including peptide functionalization, isotope labeling, and both DNA-encoded and traditional library synthesis, is provided, along with a summary of the state-of-the-art in photoreactor technology and the effective upscaling of photocatalytic reactions.
Collapse
Affiliation(s)
- Lisa Candish
- Drug Discovery Sciences, Pharmaceuticals, Bayer AG, 42113 Wuppertal, Germany
| | - Karl D Collins
- Bayer Foundation, Public Affairs, Science and Sustainability, Bayer AG, 51368 Leverkusen, Germany
| | - Gemma C Cook
- Discovery High-Throughput Chemistry, Medicinal Science and Technology, GlaxoSmithKline, Stevenage SG1 2NY, U.K
| | - James J Douglas
- Early Chemical Development, Pharmaceutical Sciences, R&D, AstraZeneca, Macclesfield SK10 2NA, U.K
| | - Adrián Gómez-Suárez
- Organic Chemistry, Bergische Universität Wuppertal, 42119 Wuppertal, Germany
| | - Anais Jolit
- Medicinal Chemistry Department, Neuroscience Discovery Research, AbbVie Deutschland GmbH & Co. KG, 67061 Ludwigshafen, Germany
| | - Sebastian Keess
- Medicinal Chemistry Department, Neuroscience Discovery Research, AbbVie Deutschland GmbH & Co. KG, 67061 Ludwigshafen, Germany
| |
Collapse
|
23
|
Sun S, Wesolowski SS. Biologically active metabolites in drug discovery. Bioorg Med Chem Lett 2021; 48:128255. [PMID: 34245850 DOI: 10.1016/j.bmcl.2021.128255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 12/30/2022]
Abstract
Biologically active metabolites are a valuable resource for development of drug candidates and lead structures for drug design. This digest highlights a selection of biologically active metabolites that have been used as new chemical entities for development or as lead structures for drug design.
Collapse
Affiliation(s)
- Shaoyi Sun
- Xenon Pharmaceuticals Inc, 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada.
| | - Steven S Wesolowski
- Xenon Pharmaceuticals Inc, 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| |
Collapse
|
24
|
Čikoš A, Dragojević S, Kubiček A. Degradation products of azetidine core G334089 - Isolation, structure elucidation and pathway. J Pharm Biomed Anal 2021; 203:114232. [PMID: 34246845 DOI: 10.1016/j.jpba.2021.114232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 10/21/2022]
Abstract
An extensive forced degradation study using hydrolytic degradation conditions was performed on G334089, the S-enantiomer of the free fatty acid receptor 2 (FFA2) antagonist GLPG0974, to identify the degradation product structures and discern degradation pathways. Not all degradation products generated ions in the MS spectra, while several others were isomers, so more rigorous degradation conditions were applied to increase the degradant yield. Esterification of the degradants facilitated isolation via preparative HPLC and subsequent NMR and MS characterisation. The determined structures, retention times and fragmentation patterns were used to identify the original degradation products and postulate a degradation pathway. In addition to the expected amide bond hydrolysis, a second degradation mechanism involving azetidine activation through formation of an azetidinium ion was demonstrated.
Collapse
Affiliation(s)
- Ana Čikoš
- Fidelta Ltd, Prilaz Baruna Filipovića 29, 10000, Zagreb, Croatia.
| | | | - Adrijana Kubiček
- Fidelta Ltd, Prilaz Baruna Filipovića 29, 10000, Zagreb, Croatia
| |
Collapse
|
25
|
Soltani S, Hallaj-Nezhadi S, Rashidi MR. A comprehensive review of in silico approaches for the prediction and modulation of aldehyde oxidase-mediated drug metabolism: The current features, challenges and future perspectives. Eur J Med Chem 2021; 222:113559. [PMID: 34119831 DOI: 10.1016/j.ejmech.2021.113559] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/10/2021] [Accepted: 05/13/2021] [Indexed: 01/09/2023]
Abstract
The importance of aldehyde oxidase (AOX) in drug metabolism necessitates the development and application of the in silico rational drug design methods as an integral part of drug discovery projects for the early prediction and modulation of AOX-mediated metabolism. The current study represents an up-to-date and thorough review of in silico studies of AOX-mediated metabolism and modulation methods. In addition, the challenges and the knowledge gap that should be covered have been discussed. The importance of aldehyde oxidase (AOX) in drug metabolism is a hot topic in drug discovery. Different strategies are available for the modulation of the AOX-mediated metabolism of drugs. Application of the rational drug design methods as an integral part of drug discovery projects is necessary for the early prediction of AOX-mediated metabolism. The current study represents a comprehensive review of AOX molecular structure, AOX-mediated reactions, AOX substrates, AOX inhibition, approaches to modify AOX-mediated metabolism, prediction of AOX metabolism/substrates/inhibitors, and the AOX related structure-activity relationship (SAR) studies. Furthermore, an up-to-date and thorough review of in silico studies of AOX metabolism has been carried out. In addition, the challenges and the knowledge gap that should be covered in the scientific literature have been discussed in the current review.
Collapse
Affiliation(s)
- Somaieh Soltani
- Pharmaceutical Analysis Research Center and Pharmacy Faculty, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Somayeh Hallaj-Nezhadi
- Drug Applied Research Center and Pharmacy Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Rashidi
- Stem Cell and Regenerative Medicine Institute and Pharmacy faculty, Tabriz University of Medical Sciences, Iran.
| |
Collapse
|
26
|
Vang ZP, Reyes A, Sonstrom RE, Holdren MS, Sloane SE, Alansari IY, Neill JL, Pate BH, Clark JR. Copper-Catalyzed Transfer Hydrodeuteration of Aryl Alkenes with Quantitative Isotopomer Purity Analysis by Molecular Rotational Resonance Spectroscopy. J Am Chem Soc 2021; 143:7707-7718. [PMID: 34000182 DOI: 10.1021/jacs.1c00884] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A copper-catalyzed alkene transfer hydrodeuteration reaction that selectively incorporates one hydrogen and one deuterium atom across an aryl alkene is described. The transfer hydrodeuteration protocol is selective across a variety of internal and terminal alkenes and is also demonstrated on an alkene-containing complex natural product analog. Beyond using 1H, 2H, and 13C NMR analysis to measure reaction selectivity, six transfer hydrodeuteration products were analyzed by molecular rotational resonance (MRR) spectroscopy. The application of MRR spectroscopy to the analysis of isotopic impurities in deuteration chemistry is further explored through a measurement methodology that is compatible with high-throughput sample analysis. In the first step, the MRR spectroscopy signatures of all isotopic variants accessible in the reaction chemistry are analyzed using a broadband chirped-pulse Fourier transform microwave spectrometer. With the signatures in hand, measurement scripts are created to quantitatively analyze the sample composition using a commercial cavity enhanced MRR spectrometer. The sample consumption is below 10 mg with analysis times on the order of 10 min using this instrument-both representing order-of-magnitude reduction compared to broadband MRR spectroscopy. To date, these measurements represent the most precise spectroscopic determination of selectivity in a transfer hydrodeuteration reaction and confirm that product regioselectivity ratios of >140:1 are achievable under this mild protocol.
Collapse
Affiliation(s)
- Zoua Pa Vang
- Department of Chemistry, Marquette University, Milwaukee, Wisconsin 53233-1881, United States
| | - Albert Reyes
- Department of Chemistry, Marquette University, Milwaukee, Wisconsin 53233-1881, United States
| | - Reilly E Sonstrom
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904-4319, United States
| | - Martin S Holdren
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904-4319, United States
| | - Samantha E Sloane
- Department of Chemistry, Marquette University, Milwaukee, Wisconsin 53233-1881, United States
| | - Isabella Y Alansari
- Department of Chemistry, Marquette University, Milwaukee, Wisconsin 53233-1881, United States
| | - Justin L Neill
- BrightSpec, Inc., Charlottesville, Virginia 22903, United States
| | - Brooks H Pate
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904-4319, United States
| | - Joseph R Clark
- Department of Chemistry, Marquette University, Milwaukee, Wisconsin 53233-1881, United States
| |
Collapse
|
27
|
Li D, Deng Y, Achab A, Bharathan I, Hopkins BA, Yu W, Zhang H, Sanyal S, Pu Q, Zhou H, Liu K, Lim J, Fradera X, Lesburg CA, Lammens A, Martinot TA, Cohen RD, Doty AC, Ferguson H, Nickbarg EB, Cheng M, Spacciapoli P, Geda P, Song X, Smotrov N, Abeywickrema P, Andrews C, Chamberlin C, Mabrouk O, Curran P, Richards M, Saradjian P, Miller JR, Knemeyer I, Otte KM, Vincent S, Sciammetta N, Pasternak A, Bennett DJ, Han Y. Carbamate and N-Pyrimidine Mitigate Amide Hydrolysis: Structure-Based Drug Design of Tetrahydroquinoline IDO1 Inhibitors. ACS Med Chem Lett 2021; 12:389-396. [PMID: 33738066 PMCID: PMC7957919 DOI: 10.1021/acsmedchemlett.0c00525] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/23/2021] [Indexed: 12/12/2022] Open
Abstract
Indoleamine-2,3-dioxygenase-1 (IDO1) has emerged as an attractive target for cancer immunotherapy. An automated ligand identification system screen afforded the tetrahydroquinoline class of novel IDO1 inhibitors. Potency and pharmacokinetic (PK) were key issues with this class of compounds. Structure-based drug design and strategic incorporation of polarity enabled the rapid improvement on potency, solubility, and oxidative metabolic stability. Metabolite identification studies revealed that amide hydrolysis in the D-pocket was the key clearance mechanism for this class. Strategic survey of amide isosteres revealed that carbamates and N-pyrimidines, which maintained exquisite potencies, mitigated the amide hydrolysis issue and led to an improved rat PK profile. The lead compound 28 is a potent IDO1 inhibitor, with clean off-target profiles and the potential for quaque die dosing in humans.
Collapse
Affiliation(s)
- Derun Li
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Yongqi Deng
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Abdelghani Achab
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Indu Bharathan
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Brett Andrew Hopkins
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Wensheng Yu
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Hongjun Zhang
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Sulagna Sanyal
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Qinglin Pu
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Hua Zhou
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Kun Liu
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Jongwon Lim
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Xavier Fradera
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Charles A. Lesburg
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Alfred Lammens
- Proteros
Biostructures GmbH, Bunsenstraße 7a, D-82152 Planegg-Martinsried, Germany
| | - Theodore A. Martinot
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Ryan D. Cohen
- Analytical
Research & Development, Merck &
Co., Inc., 126 East Lincoln Avenue, Rahway, New Jersey 07065 United States
| | - Amy C. Doty
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Heidi Ferguson
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Elliott B. Nickbarg
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Mangeng Cheng
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Peter Spacciapoli
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Prasanthi Geda
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Xuelei Song
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Nadya Smotrov
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Pravien Abeywickrema
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Christine Andrews
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Chad Chamberlin
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Omar Mabrouk
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Patrick Curran
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Matthew Richards
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Peter Saradjian
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - J. Richard Miller
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Ian Knemeyer
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Karin M. Otte
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Stella Vincent
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Nunzio Sciammetta
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Alexander Pasternak
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - David Jonathan Bennett
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| | - Yongxin Han
- Departments of Discovery Chemistry, Pharmacokinetics, Pharmacodynamics,
and Drug Metabolism, Computational and Structural Chemistry, Discovery Process Chemistry, Discovery Pharmaceutical
Science, and Quantitative Biosciences, Merck & Co.,
Inc., 33 Avenue Louis
Pasteur, Boston, Massachusetts 02115, United States
| |
Collapse
|
28
|
Veale CGL. Into the Fray! A Beginner's Guide to Medicinal Chemistry. ChemMedChem 2021; 16:1199-1225. [PMID: 33591595 DOI: 10.1002/cmdc.202000929] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Indexed: 12/31/2022]
Abstract
Modern medicinal chemistry is a complex, multidimensional discipline that operates at the interface of the chemical and biological sciences. The medicinal chemistry contribution to drug discovery is typically described in the context of the well-recited linear progression of the drug discovery pipeline. However, compound optimization is idiosyncratic to each project, and clear definitions of hit and lead molecules and the subsequent progress along the pipeline becomes easily blurred. In addition, this description lacks insight into the entangled relationship between chemical and pharmacological properties, and thus provides limited guidance on how innovative medicinal chemistry strategies can be applied to solve optimization problems, regardless of the stage in the pipeline. Through discussion and illustrative examples, this article seeks to provide insights into the finesse of medicinal chemistry and the subtlety of balancing chemical properties pharmacology. In so doing, it aims to serve as an accessible and simple-to-digest guide for anyone who wishes to learn about the underlying principles of medicinal chemistry, in a context that has been decoupled from the pipeline description.
Collapse
Affiliation(s)
- Clinton G L Veale
- School of Chemistry and Physics, Pietermaritzburg Campus, University of KwaZulu-Natal, Private Bag X01, Pietermaritzburg, Scottsville, 3209, South Africa
| |
Collapse
|
29
|
Chen X, Li Y. Identification of the stable and reactive metabolites of tetrahydropiperine using ultrahigh-performance liquid chromatography combined with diode-array detection and high-resolution mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2021; 35:e8975. [PMID: 33049799 DOI: 10.1002/rcm.8975] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/22/2020] [Accepted: 10/08/2020] [Indexed: 06/11/2023]
Abstract
RATIONALE Tetrahydropiperine is one of the natural arylpentanamide compounds isolated from Piper nigrum L., which has been demonstrated to have insecticidal activity. The aim of this study was to investigate the metabolic profiles of tetrahydropiperine in mouse, rat, dog, monkey and human hepatocytes. METHODS The in vitro metabolism of tetrahydropiperine was elucidated via incubation with hepatocytes for 2 h at 37°C. The samples were analyzed using ultrahigh-performance liquid chromatography combined with diode-array detection and high-resolution tandem mass spectrometry operated in positive electrospray ionization mode. The structures of the metabolites were characterized using their retention times and their tandem mass spectrometric product ions. RESULTS A total of 20 metabolites were detected and their structures were proposed. These metabolites were formed mainly through the following pathways: (1) 1,3-benzodioxole ring opening to form a catechol derivative (M12), which was prone to glucuronidation (M6 and M8), methylation (M17) and glutathione (GSH)-derived conjugation through an ortho-quinone intermediate (M4) or via an aldehyde intermediate (M7); (2) dehydrogenation to form a piperanine (M15), which was subsequently subject to hydroxylation (M2 and M5) and GSH conjugation (M10 and M11) via Michael addition; (3) hydroxylation (M13, M14, M16, M18 and M19); and (4) direct GSH conjugation through an aldehyde intermediate (M3). CONCLUSIONS The major metabolic pathways of tetrahydropiperine were hydroxylation, dehydrogenation, methylation, GSH conjugation and glucuronidation. Tetrahydropiperine was bioactivated through ortho-quinone, Michael receptor and aldehyde intermediates.
Collapse
Affiliation(s)
- Xiaoling Chen
- Department of Laboratory, Jinmen First People's Hospital, 67 Xiangshan Avenue, Jinmen, Hubei Province, 448000, China
| | - Yanghua Li
- Department of Pharmacy, Jinmen First People's Hospital, 67 Xiangshan Avenue, Jinmen, Hubei Province, 448000, China
| |
Collapse
|
30
|
Henry S, Anand JP, Brinkel AC, McMillan DM, Twarozynski JJ, Loo CE, Traynor JR, Mosberg HI. SAR Matrices Enable Discovery of Mixed Efficacy μ-Opioid Receptor Agonist Peptidomimetics with Simplified Structures through an Aromatic-Amine Pharmacophore. ACS Chem Neurosci 2021; 12:216-233. [PMID: 33346631 PMCID: PMC9923772 DOI: 10.1021/acschemneuro.0c00693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
We previously described the development of potent μ-opioid receptor (MOR)-agonist/δ-opioid receptor (DOR)-antagonist peptidomimetic ligands as an approach toward effective analgesics with reduced side effects. In this series, a tetrahydroquinoline (THQ) or substituted phenyl is employed to link two key pharmacophore elements, a dimethyltyrosine amino acid and typically an aromatic pendant. Using new and previously reported analogues, we constructed a structure-activity relationship (SAR) matrix that probes the utility of previously reported amine pendants. This matrix reveals that the MOR-agonist/DOR-antagonist properties of these ligands do not change when a tetrahydroisoquinoline (THIQ) pendant is used, despite removal of substituents on the core phenyl ring. Based on this observation, we retained the THIQ pendant and replaced the phenyl core with simpler aliphatic chain structures. These simpler analogues proved to be potent MOR-agonists with high variability in their effects at the DOR and the κ-opioid receptor (KOR). These data show that the amine of the THIQ pendant may be a novel pharmacophore element that favors high MOR-efficacy, whereas the aromatic ring of the THIQ pendant may produce high MOR-potency. Combined, the two pharmacophores within the THIQ pendant may be a structurally efficient means of converting opioid peptides and peptidomimetics into potent and efficacious MOR-agonists.
Collapse
Affiliation(s)
- Sean Henry
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jessica P. Anand
- Department of Pharmacology, Medical School and Edward F Domino Research Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ashley C. Brinkel
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Douglas M. McMillan
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jack. J. Twarozynski
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Christian E. Loo
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - John R. Traynor
- Department of Medicinal Chemistry, College of Pharmacy, Department of Pharmacology, Medical School, and Edward F Domino Research Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Henry I. Mosberg
- Department of Medicinal Chemistry, College of Pharmacy and Edward F Domino Research Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
31
|
Shahinozzaman M, Basak B, Emran R, Rozario P, Obanda DN. Artepillin C: A comprehensive review of its chemistry, bioavailability, and pharmacological properties. Fitoterapia 2020; 147:104775. [PMID: 33152464 DOI: 10.1016/j.fitote.2020.104775] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/14/2020] [Accepted: 10/31/2020] [Indexed: 02/06/2023]
Abstract
Artepillin C (ARC), a prenylated derivative of p-coumaric acid, is one of the major phenolic compounds found in Brazilian green propolis (BGP) and its botanical source Baccharis dracunculifolia. Numerous studies on ARC show that its beneficial health effects correlate with the health effects of both BGP and B. dracunculifolia. Its wide range of pharmacological benefits include antioxidant, antimicrobial, anti-inflammatory, anti-diabetic, neuroprotective, gastroprotective, immunomodulatory, and anti-cancer effects. Most studies have focused on anti-oxidation, inflammation, diabetic, and cancers using both in vitro and in vivo approaches. Mechanisms underlying anti-cancer properties of ARC are apoptosis induction, cell cycle arrest, and the inhibition of p21-activated kinase 1 (PAK1), a protein characterized in many human diseases/disorders including COVID-19 infection. Therefore, further pre-clinical and clinical studies with ARC are necessary to explore its potential as intervention for a wide variety of diseases including the recent pandemic coronaviral infection. This review summarizes the comprehensive data on the pharmacological effects of ARC and could be a guideline for its future study and therapeutic usage.
Collapse
Affiliation(s)
- Md Shahinozzaman
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA.
| | - Bristy Basak
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Rashiduzzaman Emran
- Department of Biochemistry, Sher-e-Bangla Agricultural University, Dhaka 1207, Bangladesh; Department of Agricultural Extension (DAE), Khamarbari, Farmgate, Dhaka 1215, Bangladesh
| | - Patricia Rozario
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka 1000, Bangladesh
| | - Diana N Obanda
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
32
|
Kumar R, Kenttämaa HI. Effects of Analyte Concentration on the Protonation Sites of 4-Aminobenzoic Acid upon Atmospheric Pressure Chemical Ionization As Revealed by Gas-Phase Ion-Molecule Reactions. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2020; 31:2210-2217. [PMID: 32852952 DOI: 10.1021/jasms.0c00285] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The most basic site of 4-aminobenzoic acid in aqueous solution is the amino nitrogen, while the carbonyl oxygen is calculated to be the most basic site in the gas phase. However, the preferred protonation site of 4-aminobenzoic acid upon electrospray ionization (ESI) and atmospheric pressure chemical ionization (APCI) depends upon the ionization solvent and ion source parameters. The influence of the concentration of the analyte on the manifested protonation sites upon APCI has not been investigated and is reported here. Gas-phase ion-molecule reactions of trimethoxymethylsilane were used to identify the protonation sites of 4-aminobenzoic acid ionized using APCI with methanol or acetonitrile-water as the solvent. The nitrogen-protomer was found to be about twice as abundant as the oxygen-protomer at low analyte concentrations (10-9-10-6 M) in methanol solvent. This finding was rationalized on the basis of a previous finding that when the O-protomer is surrounded by more than eight methanol molecules in the gas phase it starts behaving as if it were in an aqueous solution and converts to the N-protomer. At greater analyte concentrations (≥10-4 M), the amino group was predominantly protonated, which was rationalized based on the formation of a particularly stable proton-bound dimer of 4-aminobenzoic acid that preferentially dissociates to form the N-protomer. The above findings suggest that solution processes are much more important in APCI than commonly assumed, in agreement with recent literature. Indeed, when 1:1 (v/v) acetonitrile-water was used as the solvent system for 4-aminobenzoic acid, the N-protomer was predominantly generated at all analyte concentrations.
Collapse
Affiliation(s)
- Rashmi Kumar
- Chemistry Department, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| | - Hilkka I Kenttämaa
- Chemistry Department, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| |
Collapse
|
33
|
Agnew-Francis KA, Williams CM. Squaramides as Bioisosteres in Contemporary Drug Design. Chem Rev 2020; 120:11616-11650. [DOI: 10.1021/acs.chemrev.0c00416] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Kylie A. Agnew-Francis
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Craig M. Williams
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
34
|
Hu B, Zhou X, Mohutsky MA, Desai PV. Structure–Property Relationships and Machine Learning Models for Addressing CYP3A4-Mediated Victim Drug–Drug Interaction Risk in Drug Discovery. Mol Pharm 2020; 17:3600-3608. [DOI: 10.1021/acs.molpharmaceut.0c00637] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Bingjie Hu
- Computational ADME, ADME−Toxicology−PKPD, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Xin Zhou
- ADME−Toxicology−PKPD, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California 92121, United States
| | - Michael A. Mohutsky
- Investigational Drug Disposition, ADME−Toxicology−PKPD, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Prashant V. Desai
- Computational ADME, ADME−Toxicology−PKPD, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| |
Collapse
|
35
|
Miller RR, Madeira M, Wood HB, Geissler WM, Raab CE, Martin IJ. Integrating the Impact of Lipophilicity on Potency and Pharmacokinetic Parameters Enables the Use of Diverse Chemical Space during Small Molecule Drug Optimization. J Med Chem 2020; 63:12156-12170. [DOI: 10.1021/acs.jmedchem.9b01813] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Randy R. Miller
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Maria Madeira
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Harold B. Wood
- Chemistry, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Wayne M. Geissler
- Business Development & Licensing, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Conrad E. Raab
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Iain J. Martin
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| |
Collapse
|
36
|
Salamoun JM, Garcia CJ, Hargett SR, Murray JH, Chen SY, Beretta M, Alexopoulos SJ, Shah DP, Olzomer EM, Tucker SP, Hoehn KL, Santos WL. 6-Amino[1,2,5]oxadiazolo[3,4- b]pyrazin-5-ol Derivatives as Efficacious Mitochondrial Uncouplers in STAM Mouse Model of Nonalcoholic Steatohepatitis. J Med Chem 2020; 63:6203-6224. [PMID: 32392051 PMCID: PMC11042500 DOI: 10.1021/acs.jmedchem.0c00542] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Small molecule mitochondrial uncouplers have recently garnered great interest for their potential in treating nonalcoholic steatohepatitis (NASH). In this study, we report the structure-activity relationship profiling of a 6-amino[1,2,5]oxadiazolo[3,4-b]pyrazin-5-ol core, which utilizes the hydroxy moiety as the proton transporter across the mitochondrial inner membrane. We demonstrate that a wide array of substituents is tolerated with this novel scaffold that increased cellular metabolic rates in vitro using changes in oxygen consumption rate as a readout. In particular, compound SHS4121705 (12i) displayed an EC50 of 4.3 μM in L6 myoblast cells and excellent oral bioavailability and liver exposure in mice. In the STAM mouse model of NASH, administration of 12i at 25 mg kg-1 day-1 lowered liver triglyceride levels and improved liver markers such as alanine aminotransferase, NAFLD activity score, and fibrosis. Importantly, no changes in body temperature or food intake were observed. As potential treatment of NASH, mitochondrial uncouplers show promise for future development.
Collapse
Affiliation(s)
- Joseph M Salamoun
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Christopher J Garcia
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Stefan R Hargett
- Departments of Pharmacology and Medicine, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Jacob H Murray
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Sing-Young Chen
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2033, Australia
| | - Martina Beretta
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2033, Australia
| | - Stephanie J Alexopoulos
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2033, Australia
| | - Divya P Shah
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2033, Australia
| | - Ellen M Olzomer
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2033, Australia
| | - Simon P Tucker
- Continuum Biosciences, Pty Ltd., Sydney 2035, Australia
- Continuum Biosciences Inc., Boston, Massachusetts 02116, United States
| | - Kyle L Hoehn
- Departments of Pharmacology and Medicine, University of Virginia, Charlottesville, Virginia 22908, United States
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2033, Australia
| | - Webster L Santos
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
37
|
Fluorinated CRA13 analogues: Synthesis, in vitro evaluation, radiosynthesis, in silico and in vivo PET study. Bioorg Chem 2020; 99:103834. [PMID: 32334193 DOI: 10.1016/j.bioorg.2020.103834] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/21/2020] [Accepted: 04/06/2020] [Indexed: 12/19/2022]
Abstract
Fluorine is a unique atom that imparts distinct properties to bioactive molecules upon incorporation. Herein, we prepare and study fluorinated derivatives of the nanomolar affine peripherally restricted dual CB1R/CB2R agonist; CRA13 and its analogs. Binding affinity evaluation relative to CRA13 proved the stronger binding affinity of compound 7c to CB1R and CB2R by 6.95 and 5.64 folds. Physicochemical properties evaluation proved compound 7c improved lipophilicity profile suggesting some enhanced BBB penetration relative to CRA13. Radiosynthesis of 18F-labeled compound 7c was conducted conveniently affording pure hot ligand. In vivo PET study investigation demonstrated efficient distribution of 18F-labeled compound 7c in peripheral tissues visualizing peripheral CB1R/CB2R generating time-activity-curves showing good standard uptake values. Despite enhanced BBB penetration and increased cannabinoid receptors binding affinity, low brain uptake of 7c was observed. In silico docking study explained the measured binding affinities of compounds 7a-d to CB1R. While most of previous efforts aimed to develop central cannabinoid PET imaging agents, 18F-labeled compound 7c might be a promising agent serving as a universal CB1R/CB2R PET imaging agents for diagnosis and therapy of various diseases correlated with peripheral cannabinoid system. It might also serve as a lead compound for development of PET imaging of peripheral and central cannabinoid systems.
Collapse
|
38
|
Sorrentino JP, Ambler BR, Altman RA. Late-Stage Conversion of a Metabolically Labile Aryl Methyl Ether-Containing Natural Product to Fluoroalkyl Analogues. J Org Chem 2020; 85:5416-5427. [PMID: 32191836 DOI: 10.1021/acs.joc.0c00125] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We report the conversion of aryl methyl ethers and phenols into six fluoroalkyl analogues through late-stage functionalization of a natural product-derived FDA-approved therapeutic. This series of short synthetic sequences exploits a combination of both modern and traditional methods and demonstrates that some recently reported methods do not always work as well as desired on a natural product-like scaffold. Nonetheless, reaction optimization can deliver sufficient quantities of each target analogue for medicinal chemistry purposes. In some cases, classical reactions and synthetic sequences still outcompete modern organofluorine transformations, which should encourage the continued search for improved reactions. Overall, the project provides a valuable synthetic roadmap for medicinal chemists to access a range of fluorinated therapeutic candidates with distinct physicochemical properties relative to the original O-based analogue.
Collapse
Affiliation(s)
- Jacob P Sorrentino
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Brett R Ambler
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States.,Department of Discovery Chemistry MRL, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Ryan A Altman
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
| |
Collapse
|
39
|
Moreira da Silva R, Carrão DB, Habenschus MD, Jimenez PC, Lopes NP, Fenical W, Costa-Lotufo LV, de Oliveira ARM. Prediction of seriniquinone-drug interactions by in vitro inhibition of human cytochrome P450 enzymes. Toxicol In Vitro 2020; 65:104820. [PMID: 32142840 DOI: 10.1016/j.tiv.2020.104820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/18/2020] [Accepted: 03/02/2020] [Indexed: 10/24/2022]
Abstract
Seriniquinone is a secondary metabolite isolated from a rare marine bacterium of the genus Serinicoccus. This natural quinone is highlighted for its selective cytotoxic activity toward melanoma cancer cells, in which rapid metastatic properties are still a challenge for clinical treatment of malignant melanoma. The progress of seriniquinone as a promising bioactive molecule for drug development requires the assessment of its clinical interaction potential with other drugs. This study aimed to investigate the in vitro inhibitory effects of seriniquinone on the main human CYP450 isoforms involved in drug metabolism. The results showed strong inhibition of CYP1A2, CYP2E1 and CYP3A, with IC50 values up to 1.4 μM, and moderate inhibition of CYP2C19, with IC50 value >15 μM. Detailed experiments performed with human liver microsomes showed that the inhibition of CYP450 isoforms can be explained by competitive and non-competitive inhibition mechanisms. In addition, seriniquinone demonstrated to be an irreversible and time-dependent inhibitor of CYP1A2 and CYP3A. The low inhibition constants values obtained experimentally suggest that concomitant intake of seriniquinone with drug metabolized by these isoforms should be carefully monitored for adverse effects or therapeutic failure.
Collapse
Affiliation(s)
- Rodrigo Moreira da Silva
- Núcleo de Pesquisas de Produtos Naturais e Sintéticos, Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, 14090-903 Ribeirão Preto, SP, Brazil.
| | - Daniel Blascke Carrão
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901 Ribeirão Preto, SP, Brazil
| | - Maísa Daniela Habenschus
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901 Ribeirão Preto, SP, Brazil
| | - Paula Christine Jimenez
- Departamento de Ciências do Mar, Instituto do Mar, Universidade Federal de São Paulo, 11070-100 Santos, SP, Brazil
| | - Norberto Peporine Lopes
- Núcleo de Pesquisas de Produtos Naturais e Sintéticos, Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, 14090-903 Ribeirão Preto, SP, Brazil
| | - William Fenical
- CMBB, Scripps Institution of Oceanography, UC San Diego, 9500 Gilman Drive No. 0204, 92093-0204 La Jolla, CA, USA
| | - Letícia Vera Costa-Lotufo
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, 05508-900 São Paulo, SP, Brazil
| | - Anderson Rodrigo Moraes de Oliveira
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901 Ribeirão Preto, SP, Brazil; National Institute for Alternative Technologies of Detection, Toxicological Evaluation and Removal of Micropollutants and Radioactives (INCT-DATREM), Unesp, Institute of Chemistry, P.O. Box 355, 14800-900 Araraquara, SP, Brazil
| |
Collapse
|
40
|
Henry S, Anand JP, Twarozynski JJ, Brinkel AC, Pogozheva ID, Sears BF, Jutkiewicz EM, Traynor JR, Mosberg HI. Aromatic-Amine Pendants Produce Highly Potent and Efficacious Mixed Efficacy μ-Opioid Receptor (MOR)/δ-Opioid Receptor (DOR) Peptidomimetics with Enhanced Metabolic Stability. J Med Chem 2020; 63:1671-1683. [PMID: 31986033 DOI: 10.1021/acs.jmedchem.9b01818] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We previously reported a novel SAR campaign that converted a metabolically unstable series of μ-opioid receptor (MOR) agonist/δ-opioid receptor (DOR) antagonist bicyclic core peptidomimetics with promising analgesic activity and reduced abuse liabilities into a more stable series of benzylic core analogues. Herein, we expanded the SAR of that campaign and determined that the incorporation of amines into the benzylic pendant produces enhanced MOR-efficacy in this series, whereas the reincorporation of an aromatic ring into the pendant enhanced MOR-potency. Two compounds, which contain a piperidine (14) or an isoindoline (17) pendant, retained the desired opioid profile in vitro, possessed metabolic half-lives of greater than 1 h in mouse liver microsomes (MLMs), and were active antinociceptive agents in the acetic acid stretch assay (AASA) at subcutaneous doses of 1 mg/kg.
Collapse
Affiliation(s)
- Sean Henry
- Department of Medicinal Chemistry, College of Pharmacy , University of Michigan , 428 Church Street , Ann Arbor , Michigan 48109 , United States
| | - Jessica P Anand
- Department of Pharmacology, Medical School , University of Michigan , Ann Arbor , Michigan 48109 , United States.,Edward F. Domino Research Center , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Jack J Twarozynski
- Department of Pharmacology, Medical School , University of Michigan , Ann Arbor , Michigan 48109 , United States.,Edward F. Domino Research Center , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Ashley C Brinkel
- Department of Pharmacology, Medical School , University of Michigan , Ann Arbor , Michigan 48109 , United States.,Edward F. Domino Research Center , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Irina D Pogozheva
- Department of Medicinal Chemistry, College of Pharmacy , University of Michigan , 428 Church Street , Ann Arbor , Michigan 48109 , United States
| | - Bryan F Sears
- Department of Pharmacology, Medical School , University of Michigan , Ann Arbor , Michigan 48109 , United States.,Edward F. Domino Research Center , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Emily M Jutkiewicz
- Department of Pharmacology, Medical School , University of Michigan , Ann Arbor , Michigan 48109 , United States.,Edward F. Domino Research Center , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - John R Traynor
- Department of Medicinal Chemistry, College of Pharmacy , University of Michigan , 428 Church Street , Ann Arbor , Michigan 48109 , United States.,Department of Pharmacology, Medical School , University of Michigan , Ann Arbor , Michigan 48109 , United States.,Edward F. Domino Research Center , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Henry I Mosberg
- Department of Medicinal Chemistry, College of Pharmacy , University of Michigan , 428 Church Street , Ann Arbor , Michigan 48109 , United States.,Edward F. Domino Research Center , University of Michigan , Ann Arbor , Michigan 48109 , United States
| |
Collapse
|
41
|
Dalvie D, Di L. Aldehyde oxidase and its role as a drug metabolizing enzyme. Pharmacol Ther 2019; 201:137-180. [PMID: 31128989 DOI: 10.1016/j.pharmthera.2019.05.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 03/27/2019] [Indexed: 11/29/2022]
Abstract
Aldehyde oxidase (AO) is a cytosolic enzyme that belongs to the family of structurally related molybdoflavoproteins like xanthine oxidase (XO). The enzyme is characterized by broad substrate specificity and marked species differences. It catalyzes the oxidation of aromatic and aliphatic aldehydes and various heteroaromatic rings as well as reduction of several functional groups. The references to AO and its role in metabolism date back to the 1950s, but the importance of this enzyme in the metabolism of drugs has emerged in the past fifteen years. Several reviews on the role of AO in drug metabolism have been published in the past decade indicative of the growing interest in the enzyme and its influence in drug metabolism. Here, we present a comprehensive monograph of AO as a drug metabolizing enzyme with emphasis on marketed drugs as well as other xenobiotics, as substrates and inhibitors. Although the number of drugs that are primarily metabolized by AO are few, the impact of AO on drug development has been extensive. We also discuss the effect of AO on the systemic exposure and clearance these clinical candidates. The review provides a comprehensive analysis of drug discovery compounds involving AO with the focus on developmental candidates that were reported in the past five years with regards to pharmacokinetics and toxicity. While there is only one known report of AO-mediated clinically relevant drug-drug interaction (DDI), a detailed description of inhibitors and inducers of AO known to date has been presented here and the potential risks associated with DDI. The increasing recognition of the importance of AO has led to significant progress in predicting the site of AO-mediated metabolism using computational methods. Additionally, marked species difference in expression of AO makes it is difficult to predict human clearance with high confidence. The progress made towards developing in vivo, in vitro and in silico approaches for predicting AO metabolism and estimating human clearance of compounds that are metabolized by AO have also been discussed.
Collapse
Affiliation(s)
- Deepak Dalvie
- Drug Metabolism and Pharmacokinetics, Celgene Corporation, 10300, Campus Point Drive, San Diego, CA 92121, USA.
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT 06340, UK
| |
Collapse
|
42
|
Norcross NR, Wilson C, Baragaña B, Hallyburton I, Osuna‐Cabello M, Norval S, Riley J, Fletcher D, Sinden R, Delves M, Ruecker A, Duffy S, Meister S, Antonova‐Koch Y, Crespo B, de Cózar C, Sanz LM, Gamo FJ, Avery VM, Frearson JA, Gray DW, Fairlamb AH, Winzeler EA, Waterson D, Campbell SF, Willis PA, Read KD, Gilbert IH. Substituted Aminoacetamides as Novel Leads for Malaria Treatment. ChemMedChem 2019; 14:1329-1335. [PMID: 31188540 PMCID: PMC6899483 DOI: 10.1002/cmdc.201900329] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Indexed: 01/29/2023]
Abstract
Herein we describe the optimization of a phenotypic hit against Plasmodium falciparum based on an aminoacetamide scaffold. This led to N-(3-chloro-4-fluorophenyl)-2-methyl-2-{[4-methyl-3-(morpholinosulfonyl)phenyl]amino}propanamide (compound 28) with low-nanomolar activity against the intraerythrocytic stages of the malaria parasite, and which was found to be inactive in a mammalian cell counter-screen up to 25 μm. Inhibition of gametes in the dual gamete activation assay suggests that this family of compounds may also have transmission blocking capabilities. Whilst we were unable to optimize the aqueous solubility and microsomal stability to a point at which the aminoacetamides would be suitable for in vivo pharmacokinetic and efficacy studies, compound 28 displayed excellent antimalarial potency and selectivity; it could therefore serve as a suitable chemical tool for drug target identification.
Collapse
Affiliation(s)
- Neil R. Norcross
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Caroline Wilson
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Beatriz Baragaña
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Irene Hallyburton
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Maria Osuna‐Cabello
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Suzanne Norval
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Jennifer Riley
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Daniel Fletcher
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | | | | | | | - Sandra Duffy
- Discovery BiologyGriffith Institute for Drug DiscoveryGriffith UniversityNathanQueensland4111Australia
| | - Stephan Meister
- Department of PediatricsUniversity of California San Diego School of Medicine9500 Gilman Drive 0741La JollaCA92093USA
| | - Yevgeniya Antonova‐Koch
- Department of PediatricsUniversity of California San Diego School of Medicine9500 Gilman Drive 0741La JollaCA92093USA
| | - Benigno Crespo
- GlaxoSmithKline, Diseases of the Developing World – Tres Cantos Medicines Development Campusc/ Severo Ochoa 2, Tres Cantos28760MadridSpain
| | - Cristina de Cózar
- GlaxoSmithKline, Diseases of the Developing World – Tres Cantos Medicines Development Campusc/ Severo Ochoa 2, Tres Cantos28760MadridSpain
| | - Laura M. Sanz
- GlaxoSmithKline, Diseases of the Developing World – Tres Cantos Medicines Development Campusc/ Severo Ochoa 2, Tres Cantos28760MadridSpain
| | - Francisco Javier Gamo
- GlaxoSmithKline, Diseases of the Developing World – Tres Cantos Medicines Development Campusc/ Severo Ochoa 2, Tres Cantos28760MadridSpain
| | - Vicky M. Avery
- Discovery BiologyGriffith Institute for Drug DiscoveryGriffith UniversityNathanQueensland4111Australia
| | - Julie A. Frearson
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - David W. Gray
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Alan H. Fairlamb
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Elizabeth A. Winzeler
- Department of PediatricsUniversity of California San Diego School of Medicine9500 Gilman Drive 0741La JollaCA92093USA
| | - David Waterson
- Medicines for Malaria VentureInternational Centre, Cointrin, Entrance G, 3rd FloorRoute de Pré-Bois 20, PO Box 1826Geneva1215Switzerland
| | - Simon F. Campbell
- Medicines for Malaria VentureInternational Centre, Cointrin, Entrance G, 3rd FloorRoute de Pré-Bois 20, PO Box 1826Geneva1215Switzerland
| | - Paul A. Willis
- Medicines for Malaria VentureInternational Centre, Cointrin, Entrance G, 3rd FloorRoute de Pré-Bois 20, PO Box 1826Geneva1215Switzerland
| | - Kevin D. Read
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Ian H. Gilbert
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| |
Collapse
|
43
|
Henry SP, Fernandez TJ, Anand JP, Griggs NW, Traynor JR, Mosberg HI. Structural Simplification of a Tetrahydroquinoline-Core Peptidomimetic μ-Opioid Receptor (MOR) Agonist/δ-Opioid Receptor (DOR) Antagonist Produces Improved Metabolic Stability. J Med Chem 2019; 62:4142-4157. [PMID: 30924650 DOI: 10.1021/acs.jmedchem.9b00219] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We have previously reported a series of μ-opioid receptor (MOR) agonist/δ-opioid receptor (DOR) antagonist ligands to serve as potential nonaddictive opioid analgesics. These ligands have been shown to be active in vivo, do not manifest withdrawal syndromes or reward behavior in conditioned-place preference assays in mice, and do not produce dependence. Although these attributes are promising, these analogues exhibit poor metabolic stability in mouse liver microsomes, likely due to the central tetrahydroquinoline scaffold in this series. As such, a structure-activity relationship (SAR) campaign was pursued to improve their metabolic stability. This resulted in a shift from our original bicyclic tetrahydroquinoline core to a monocyclic benzylic-core system. By eliminating one of the rings in this scaffold and exploring the SAR of this new core, two promising analogues were discovered. These analogues (5l and 5m) had potency and efficacy values at MOR better or comparable to morphine, retained their DOR-antagonist properties, and showed a 10-fold improvement in metabolic stability.
Collapse
Affiliation(s)
- Sean P Henry
- Department of Medicinal Chemistry, College of Pharmacy , University of Michigan , 428 Church Street , Ann Arbor , Michigan 48109 , United States
| | | | | | | | - John R Traynor
- Department of Medicinal Chemistry, College of Pharmacy , University of Michigan , 428 Church Street , Ann Arbor , Michigan 48109 , United States
| | - Henry I Mosberg
- Department of Medicinal Chemistry, College of Pharmacy , University of Michigan , 428 Church Street , Ann Arbor , Michigan 48109 , United States
| |
Collapse
|
44
|
Gair JJ, Grey RL, Giroux S, Brodney MA. Palladium Catalyzed Hydrodefluorination of Fluoro-(hetero)arenes. Org Lett 2019; 21:2482-2487. [DOI: 10.1021/acs.orglett.9b00889] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Joseph J. Gair
- Vertex Pharmaceuticals Inc, 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Ronald L. Grey
- Vertex Pharmaceuticals Inc, 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Simon Giroux
- Vertex Pharmaceuticals Inc, 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Michael A. Brodney
- Vertex Pharmaceuticals Inc, 50 Northern Avenue, Boston, Massachusetts 02210, United States
| |
Collapse
|
45
|
Affiliation(s)
- Muhammad H. Rahman
- School of PharmacyUniversity of Birmingham Edgbaston B15 2TT United Kingdom
| | - Mandeep K. Bal
- Faculty of Science and EngineeringManchester Metropolitan University Chester Street Manchester M1 5GD United Kingdom
| | - Alan M. Jones
- School of PharmacyUniversity of Birmingham Edgbaston B15 2TT United Kingdom
| |
Collapse
|
46
|
Bal MK, Banks CE, Jones AM. Metabolism Mimicry: An Electrosynthetic Method for the Selective Deethylation of Tertiary Benzamides. ChemElectroChem 2019. [DOI: 10.1002/celc.201900028] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Mandeep K. Bal
- Faculty of Science and EngineeringManchester Metropolitan University Chester Street Manchester M1 5GD United Kingdom
| | - Craig E. Banks
- Faculty of Science and EngineeringManchester Metropolitan University Chester Street Manchester M1 5GD United Kingdom
| | - Alan M. Jones
- School of PharmacyUniversity of Birmingham Edgbaston B15 2TT United Kingdom
| |
Collapse
|
47
|
Kakutani N, Nanayama T, Nomura Y. Novel risk assessment of reactive metabolites from discovery to clinical stage. J Toxicol Sci 2019; 44:201-211. [DOI: 10.2131/jts.44.201] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Nobuyuki Kakutani
- Drug Metabolism & Pharmacokinetics Research Laboratories, Central Pharmaceutical Research Institute
| | - Toyomichi Nanayama
- Drug Metabolism & Pharmacokinetics Research Laboratories, Central Pharmaceutical Research Institute
| | - Yukihiro Nomura
- Drug Metabolism & Pharmacokinetics Research Laboratories, Central Pharmaceutical Research Institute
| |
Collapse
|
48
|
Ma B, Liu L, Zhang J. Gold-Catalyzed Site-Selective C−H Bond Functionalization with Diazo Compounds. ASIAN J ORG CHEM 2018. [DOI: 10.1002/ajoc.201800472] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Ben Ma
- School of Chemistry and Molecular Engineering; East China Normal University; 500 Dongchuan Road Shanghai 200241 China
| | - Lu Liu
- School of Chemistry and Molecular Engineering; East China Normal University; 500 Dongchuan Road Shanghai 200241 China
| | - Junliang Zhang
- School of Chemistry and Molecular Engineering; East China Normal University; 500 Dongchuan Road Shanghai 200241 China
| |
Collapse
|
49
|
He C, Wan H. Drug metabolism and metabolite safety assessment in drug discovery and development. Expert Opin Drug Metab Toxicol 2018; 14:1071-1085. [DOI: 10.1080/17425255.2018.1519546] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Chunyong He
- Department of DMPK/Tox, Shanghai Hengrui Pharmaceutical Co., Ltd., Shanghai, P. R. China
| | - Hong Wan
- Department of DMPK/Tox, Shanghai Hengrui Pharmaceutical Co., Ltd., Shanghai, P. R. China
| |
Collapse
|
50
|
Li Z, Zhang L, Yuan Y, Yang Z. Identification of metabolites of evobrutinib in rat and human hepatocytes by using ultra-high performance liquid chromatography coupled with diode array detector and Q Exactive Orbitrap tandem mass spectrometry. Drug Test Anal 2018; 11:129-139. [PMID: 30102849 DOI: 10.1002/dta.2477] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/20/2018] [Accepted: 08/02/2018] [Indexed: 01/19/2023]
Affiliation(s)
- Zeyun Li
- Department of Pharmacy; the First Affiliated Hospital of Zhengzhou University; Zhengzhou China
| | - Lizhen Zhang
- Department of Pharmacy; the First Affiliated Hospital of Zhengzhou University; Zhengzhou China
| | - Yongliang Yuan
- Department of Pharmacy; the First Affiliated Hospital of Zhengzhou University; Zhengzhou China
| | - Zhiheng Yang
- Department of Pharmacy; the First Affiliated Hospital of Zhengzhou University; Zhengzhou China
| |
Collapse
|