1
|
Ren J, Liu K, Wang N, Li J, Long X, Li C, Li K. Three-Component 1,2-Methylamidation of Alkynes via Coordinating Activation Strategy. ChemistryOpen 2025:e2500151. [PMID: 40244085 DOI: 10.1002/open.202500151] [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: 03/06/2025] [Revised: 04/04/2025] [Indexed: 04/18/2025] Open
Abstract
The selective functionalization of carbon-carbon triple bonds with methyl groups remains a challenging task. Herein, the successful development of a novel copper-catalyzed three-component 1,2-methylamidation of carbon-carbon triple bond is reported. The readily available coupling partners, picolinamides and alkynes with dicumyl peroxide, serve as both the methyl source and oxidant in this difunctional strategy to access methylated enamides; the substrate scope is broad, demonstrating good functional group compatibility. The synthetic utility of the reaction is also demonstrated through the 1,2-methylamidation of alkynes via late-stage functionalization of substrates bearing biologically relevant molecules.
Collapse
Affiliation(s)
- Jing Ren
- Institute of BiopharmaceuticalsWest China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, P. R. China
| | - Kaiyun Liu
- Institute of BiopharmaceuticalsWest China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, P. R. China
| | - Ning Wang
- Institute of BiopharmaceuticalsWest China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, P. R. China
| | - Jinlong Li
- Institute of BiopharmaceuticalsWest China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, P. R. China
| | - Xinyu Long
- Institute of BiopharmaceuticalsWest China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, P. R. China
| | - Chengming Li
- Institute of Organ Transplantation, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, P. R. China
| | - Kaizhi Li
- Institute of BiopharmaceuticalsWest China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, P. R. China
| |
Collapse
|
2
|
Lan JX, Huang LJ, Kang SS, Hao-Huang, Liu SL, Dai W, Xu XL, Wang JY, Shu GZ, Hou W. Design, synthesis, biological evaluation, and mechanism of action of new pyrazines as anticancer agents in vitro and in vivo. Bioorg Med Chem 2025; 121:118108. [PMID: 39955800 DOI: 10.1016/j.bmc.2025.118108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
Cancer is the second leading cause of mortality worldwide. The development of innovative antitumor pharmaceuticals is urgently needed to alter this circumstance. N-heterocycles, pyrazines for example are prevalent pharmacophores in the architecture of anticancer medicines. This research involved the design and synthesis of seventy-seven new pyrazine derivatives, followed by an evaluation of their anticancer activity in vitro and in vivo. Several new pyrazines exhibiting remarkable antiproliferative activity and selectivity were identified. The links between structure and function were analyzed, and the mechanisms of action were examined. Our mechanistic investigations indicated that these chemicals triggered mitochondria-associated apoptosis in cancer cells. Moreover, they suppressed the phosphorylation of STAT3, concomitant with the down-regulation of BcL-2, BcL-XL, c-Myc, XIAP, GLI1, TAZ, MCL1, JAK1, JAK2 and up-regulation of Bax, p21. Furthermore, the lead compounds B-11 and C-27 demonstrated significant anticancer activity in vivo in the SKOV3 xenograft nude mouse model. Our research establishes a basis for the identification of pyrazines as JAK/STAT3 inhibition based anticancer lead compounds.
Collapse
Affiliation(s)
- Jin-Xia Lan
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China; School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000 PR China
| | - Le-Jun Huang
- School of Rehabilitation, Gannan Medical University, Ganzhou 341000 PR China
| | - Si-Shuang Kang
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Hao-Huang
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Sheng-Lan Liu
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Wei Dai
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Xin-Liang Xu
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Jin-Yang Wang
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Guang-Zhao Shu
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Wen Hou
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China.
| |
Collapse
|
3
|
Abd El Hadi SR, Eldinary MA, Ghith A, Haffez H, Salman A, Sayed GA. Unravelling the potency of the 4-oxo-2-thioxo-1,2,3,4-tetrahydropyrimidine-5-carbonitrile scaffold with S-arylamide hybrids as PIM-1 kinase inhibitors: synthesis, biological activity and in silico studies. RSC Med Chem 2025:d5md00021a. [PMID: 40162200 PMCID: PMC11951167 DOI: 10.1039/d5md00021a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
PIM-1 is a type of serine/threonine kinase that plays a crucial role in controlling several vital processes, including proliferation and apoptosis. New synthetic S-amide tetrahydropyrimidinone derivatives were designed and synthesized as PIM-1 inhibitors with potential anticancer activity. Several biochemical assays were performed for anticancer assessment, including PIM-1 inhibitory assays, MTT, apoptosis and cell cycle, gene expression analysis, c-MYC analysis, and ATPase inhibitory assays. Compounds (8c, 8d, 8g, 8h, 8k, and 8l) exhibited strong in vitro broad antiproliferative activity against MCF-7, DU-145, and PC-3, with a relatively higher SI index suggesting minimal cytotoxicity to normal cells. Furthermore, these compounds induced mixed late apoptosis and necrosis with cell cycle arrest at the G2/M phase. Moreover, compounds 8b, 8f, 8g, 8k, and 8l showed potent inhibitory action against PIM-1 kinase, with corresponding IC50 values of 660, 909, 373, 518, and 501 nM. In silico prediction studies of physiochemical properties, molecular dynamics, and induced fit docking studies were performed for these compounds to explain their potent biological activity. In conclusion, new pyrimidinone compounds (8c, 8d, 8g, 8h, 8k, and 8l) exhibit potential PIM-1 inhibitory activity and can be used as promising scaffolds for further optimization of new leads with selective PIM-inhibitors and anticancer activity.
Collapse
Affiliation(s)
- Soha R Abd El Hadi
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian Russian University, Badr City Cairo 11829 Egypt
| | - Manar A Eldinary
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian Russian University, Badr City Cairo 11829 Egypt
| | - Amna Ghith
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital Woodville South SA 5011 Australia
- Robinson Research Institute, University of Adelaide Adelaide SA 5006 Australia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Future University in Egypt Cairo 11835 Egypt
| | - Hesham Haffez
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University P.O. Box 11795 Cairo Egypt
- Center of Scientific Excellence "Helwan Structural Biology Research (HSBR)", Helwan University Cairo 11795 Egypt
| | - Aya Salman
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City 11829 Cairo Egypt
| | - Ghadir A Sayed
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City 11829 Cairo Egypt
| |
Collapse
|
4
|
Borriello L, Jarray R, Rignault-Bricard R, Montes M, Lopez N, Maciel TT, Hermine O, Raynaud F, Demange L, Lepelletier Y. Neuropilin Antagonists (NRPas) Block the Phosphorylation of the Cancer Therapeutic Key Factor p38α Kinase Triggering Cell Death. Molecules 2025; 30:1494. [PMID: 40286084 DOI: 10.3390/molecules30071494] [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: 02/11/2025] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 04/29/2025] Open
Abstract
Neuropilin-1 is henceforth a relevant target in cancer treatment; however, its way of action remains partly elusive, and the development of small inhibitory molecules is therefore required for its study. Here, we report that two small-sized neuropilin antagonists (NRPa-47 and NRPa-48), VEGF-A165/NRP-1 binding inhibitors, are able to decrease VEGF-Rs phosphorylation and to modulate their downstream cascades in the triple-negative breast cancer cell line (MDA-MB-231). Nevertheless, NRPas exert a divergent pathway regulation of MAPK phosphorylation, such as JNK-1/-2/-3, ERK-1/-2, and p38β/γ/δ-kinases, as well as their respective downstream targets. However, NRPa-47 and NRPa-48 apply a common down-regulation of the p38α-kinase phosphorylation and their downstream targets, emphasising its central regulating role. More importantly, none of the 40 selected kinases, including SAPK2a/p38α, are affected in vitro by NRPas, strengthening their specificity. Taken together, NRPas induced cell death by the down-modulation of pro-apoptotic and anti-apoptotic proteins, cell death receptors and adaptors, heat shock proteins (HSP-27/-60/-70), cell cycle proteins (p21, p27, phospho-RAD17), and transcription factors (p53, HIF-1α). In conclusion, we showed for the first time how NRPas may alter tumour cell signalling and contribute to the down-modulation of the cancer therapeutic key factor p38α-kinase phosphorylation. Thus, the efficient association of NRPas and p38α-kinase inhibitor strengthened this hypothesis.
Collapse
Affiliation(s)
- Lucia Borriello
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Fox Chase Cancer Center, Cancer Signaling and Microenvironment Program, Philadelphia, PA 19140, USA
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques (LCBPT), UMR 8601 CNRS, Université Paris Descartes, Sorbonne Paris Cité, UFR Biomédicale des Saints Pères, 45 Rue des Saints Pères, 75270 Paris, CEDEX 06, France
| | - Rafika Jarray
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques (LCBPT), UMR 8601 CNRS, Université Paris Descartes, Sorbonne Paris Cité, UFR Biomédicale des Saints Pères, 45 Rue des Saints Pères, 75270 Paris, CEDEX 06, France
- Division of Prions and Related Diseases (SEPIA), CEA (Commissariat à l'Énergie Atomique), Institute of Emerging Diseases and Innovative Therapies (iMETI), 92265 Fontenay-aux-Roses, France
| | - Rachel Rignault-Bricard
- INSERM UMR 1163, Laboratory of Cellular and Molecular Basis of Normal Hematopoiesis and Hematological Disorders: Therapeutical Implications, 24 Boulevard Montparnasse, 75015 Paris, France
- Imagine Institute, Université Paris Cité, 24 Boulevard Montparnasse, 75015 Paris, France
| | - Matthieu Montes
- Laboratoire Génomique, Bioinformatique et Chimie Moléculaire, EA7528, Conservatoire National des Arts et Métiers, 292 Rue Saint Martin, 75003 Paris, France
| | - Nicolas Lopez
- W-MedPhys, 128 Rue la Boétie, 75008 Paris, France
- ENOES/ENESIA, 62 Rue de Miromesnil, 75008 Paris, France
- Unité Mixte de Recherche "Institut de Physique Théorique (IPhT)" CEA-CNRS, UMR 3681, Route de l'Orme des Merisiers, 91191 St Aubin-Gif-sur-Yvette, France
| | - Thiago Trovati Maciel
- INSERM UMR 1163, Laboratory of Cellular and Molecular Basis of Normal Hematopoiesis and Hematological Disorders: Therapeutical Implications, 24 Boulevard Montparnasse, 75015 Paris, France
- Imagine Institute, Université Paris Cité, 24 Boulevard Montparnasse, 75015 Paris, France
| | - Olivier Hermine
- INSERM UMR 1163, Laboratory of Cellular and Molecular Basis of Normal Hematopoiesis and Hematological Disorders: Therapeutical Implications, 24 Boulevard Montparnasse, 75015 Paris, France
- Imagine Institute, Université Paris Cité, 24 Boulevard Montparnasse, 75015 Paris, France
| | - Françoise Raynaud
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques (LCBPT), UMR 8601 CNRS, Université Paris Descartes, Sorbonne Paris Cité, UFR Biomédicale des Saints Pères, 45 Rue des Saints Pères, 75270 Paris, CEDEX 06, France
| | - Luc Demange
- UMR 8038 CNRS CiTCoM, Team PNAS, Faculté de Pharmacie, Université Paris Cité, 4 Avenue de l'Observatoire, 75006 Paris, France
| | - Yves Lepelletier
- INSERM UMR 1163, Laboratory of Cellular and Molecular Basis of Normal Hematopoiesis and Hematological Disorders: Therapeutical Implications, 24 Boulevard Montparnasse, 75015 Paris, France
- Imagine Institute, Université Paris Cité, 24 Boulevard Montparnasse, 75015 Paris, France
- W-MedPhys, 128 Rue la Boétie, 75008 Paris, France
| |
Collapse
|
5
|
Zhang Z, Zhu BK, Yi ZY, Fang T, Jin Z, He L, Chen BB, Qi X, Wang CJ. Catalytic Asymmetric Synthesis and Applications of Stereogenic β'-Methyl Enones and β,β'-Dimethyl Ketones. Angew Chem Int Ed Engl 2025; 64:e202414449. [PMID: 39658841 DOI: 10.1002/anie.202414449] [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: 07/30/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/12/2024]
Abstract
The "Magic Methyl" effect has received tremendous interest in medicinal chemistry due to the significant pharmacological and physical modification of properties that have been observed upon introducing a methyl group, especially, a stereogenic methyl group into potential chiral drug candidates. The prevalence of stereogenic β-methyl ketone structural motifs in bioactive compounds and natural products has long motivated the development of enantioselective strategies toward their synthesis. Herein, we have rationally designed a Rh-catalyzed asymmetric monohydrogenation of readily-available β'-methylene conjugated enones with high efficiency and remarkable site-selectivity and enantioselectivity control for the practical construction of enantioenriched β'-methyl unsaturated enones that are difficult to access by other methods. Control experiments revealed that the conjugated C=C bond in β'-methylene conjugated enones plays a significant role in enhancing the reactivity of monohydrogenation. This methodology is applicable for the preparation of chiral β,β'-dimethyl ketones through consecutive double asymmetric hydrogenation of β,β'-dimethylene ketones. Detailed mechanistic investigation and DFT studies further provided strong support for a unique processive catalysis pathway for double asymmetric hydrogenation. The synthetic utilities have been demonstrated in the concise synthesis of several key intermediates for bioactive molecules, asymmetric total synthesis of natural products (S)-(+)-ar-Turmerone and (S)-(+)-dihydro-ar-Turmerone, and two C2-symmetric chiral spirocyclic diol frameworks.
Collapse
Affiliation(s)
- Zongpeng Zhang
- Hubei Research Center of Fundamental Science-Chemistry, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Bing-Ke Zhu
- Hubei Research Center of Fundamental Science-Chemistry, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Zhi-Yuan Yi
- Hubei Research Center of Fundamental Science-Chemistry, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Ting Fang
- Hubei Research Center of Fundamental Science-Chemistry, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Zhuan Jin
- Hubei Research Center of Fundamental Science-Chemistry, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Ling He
- Hubei Research Center of Fundamental Science-Chemistry, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Bo-Bin Chen
- Hubei Research Center of Fundamental Science-Chemistry, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Xiaotian Qi
- Hubei Research Center of Fundamental Science-Chemistry, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Chun-Jiang Wang
- Hubei Research Center of Fundamental Science-Chemistry, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
- State Key Laboratory of Elemento-organic Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
6
|
Farajat D, Zhang Y, Li CJ. Magic methylation with methyl-containing peroxides. Chem Sci 2025; 16:507-529. [PMID: 39640027 PMCID: PMC11615666 DOI: 10.1039/d4sc05620e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
Methyl groups rank among the most abundant carbon fragments found in natural products and small-molecule pharmaceuticals. The late-stage and environmentally friendly installation of these groups onto biologically active molecules has attracted widespread attention in both industry and academia. In 2008, we published the first use of a methyl radical derived from a peroxide toward a directed transition-metal catalysed C-H methylation. In the past sixteen years, methyl-containing peroxides have proven themselves as robust reagents for introducing methyl groups onto organic molecules. In this review, our goal is to provide a thorough summary of the research advancements achieved in this field thus far.
Collapse
Affiliation(s)
- Daliah Farajat
- Department of Chemistry, McGill University 801 Sherbrooke Street West Montreal Quebec H3A 2K6 Canada
| | - Yuhua Zhang
- Accustandard Inc. 125 Market Street New Haven Connecticut 06513 USA
| | - Chao-Jun Li
- Department of Chemistry, McGill University 801 Sherbrooke Street West Montreal Quebec H3A 2K6 Canada
- FRQNT Centre for Green Chemistry and Catalysis Canada
| |
Collapse
|
7
|
Wu P, Goujon G, Pan S, Tuccio B, Pégot B, Dagousset G, Anselmi E, Magnier E, Bolm C. Cyclic Sulfoximines as Methyl and Perdeuteromethyl Transfer Agents and Their Applications in Photoredox Catalysis. Angew Chem Int Ed Engl 2024; 63:e202412418. [PMID: 39234959 DOI: 10.1002/anie.202412418] [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: 07/02/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/06/2024]
Abstract
Benzo[1,3,2]dithiazole-1,1,3-trioxides are bench-stable and easy-to-use reagents. In photoredox catalysis, they generate methyl and perdeuteromethyl radicals which can add to a variety of radical acceptors, including olefins, acrylamides, quinoxalinones, isocyanides, enol silanes, and N-Ts acrylamide. As byproduct, a salt is formed which can be regenerated to the original methylating agent. Flow chemistry provides an option for reaction scale-up further underscoring the synthetic usefulness of these methylation reagents. Mechanistic investigations suggest a single-electron transfer (SET) pathway induced by photoredox catalysis.
Collapse
Affiliation(s)
- Peng Wu
- Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52074, Aachen, Germany
| | - Gabriel Goujon
- Institut Lavoisier de Versailles, Université Paris-Saclay, 78000, Versailles, France
| | - Shulei Pan
- Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52074, Aachen, Germany
| | - Béatrice Tuccio
- Aix-Marseille Université, CNRS, Institut de Chimie Radicalaire, UMR 7273, F-13397, Marseille Cedex 20, France
| | - Bruce Pégot
- Institut Lavoisier de Versailles, Université Paris-Saclay, 78000, Versailles, France
| | - Guillaume Dagousset
- Institut Lavoisier de Versailles, Université Paris-Saclay, 78000, Versailles, France
| | - Elsa Anselmi
- Institut Lavoisier de Versailles, Université Paris-Saclay, 78000, Versailles, France
- Université de Tours, Faculté des Sciences et Techniques, 37200, Tours, France
| | - Emmanuel Magnier
- Institut Lavoisier de Versailles, Université Paris-Saclay, 78000, Versailles, France
| | - Carsten Bolm
- Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52074, Aachen, Germany
| |
Collapse
|
8
|
Sabt A, Tawfik HO, Khaleel EF, Badi RM, Ibrahim HAA, Elkaeed EB, Eldehna WM. An overview of recent advancements in small molecules suppression of oncogenic signaling of K-RAS: an updated review. Mol Divers 2024; 28:4581-4608. [PMID: 38289431 DOI: 10.1007/s11030-023-10777-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/17/2023] [Indexed: 12/21/2024]
Abstract
RAS (rat sarcoma) oncoproteins are crucial for the growth of some human cancers, including lung, colorectal, and pancreatic adenocarcinomas. The RAS family contains three known human isoforms H(Harvey)-RAS, N(Neuroblastoma)-RAS, and K(Kirsten)-RAS. Mutations in RAS proteins cause up to ~ 30% of cancer cases. For almost 30 years, mutant proteins druggable pockets remained undiscovered, they are nearly identical to their essential, wild-type counterparts and cause cancer. Recent research has increased our knowledge of RAS's structure, processing, and signaling pathways and revealed novel insights into how it works in cancer cells. We highlight several approaches that inhibit RAS activity with small compounds in this review: substances that blocked farnesyltransferase (FTase), isoprenylcysteine carboxyl methyltransferase (Icmt), and RAS-converting enzyme 1 (Rce1) three important enzymes required for RAS localization. Inhibitors block the son of sevenless (SOS) protein's role in nucleotide exchange activity, small molecules that interfered with the phosphodiesterase (PDEδ)-mediated intracellular RAS transport processes, substances that focused on inhibiting RAS-effector interactions. Inhibitors are made to suppress the oncogenic K-RAS G12C mutant only when the nucleophilic cysteine residue at codon 12 is present and many inhibitors with various mechanisms like breaking the organization membrane of K-RAS nano-clustering. So, this is a thorough analysis of the most recent advancements in K-RAS-targeted anticancer techniques, hopefully offering insight into the field's future.
Collapse
Affiliation(s)
- Ahmed Sabt
- Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, Cairo, Egypt.
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| | - Eman F Khaleel
- Department of Medical Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Rehab Mustafa Badi
- Department of Medical Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | | | - Eslam B Elkaeed
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, 13713, Riyadh, Saudi Arabia
| | - Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, P.O. Box 33516, Egypt.
| |
Collapse
|
9
|
Nkoana JK, More GK, Mphahlele MJ, Elhenawy AA. Synthesis and in vitro exploration of the 8-carbo substituted 5-methoxyflavones as anti-breast and anti-lung cancer agents targeting protein kinases (VEGFR-2 & EGFR). Bioorg Chem 2024; 153:107875. [PMID: 39396454 DOI: 10.1016/j.bioorg.2024.107875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/23/2024] [Accepted: 10/04/2024] [Indexed: 10/15/2024]
Abstract
The 8-aryl-, 8-styryl- and 8-arylethynyl substituted 5-methoxyflavones were synthesized and characterized using a combination of spectroscopic techniques. Single crystal X-ray diffraction (XRD) study on a representative compound 3h shows an inverted dimer linked by fused ten and six-membered ring motifs involving intermolecular CO⋯HC and CH⋯OC hydrogen bonds. Compounds 3b, 3c, 3d, 4a and 4b exhibited strong activity against the human breast (MCF-7) cancer cell line (IC50 = 13.68 ± 0.72, 16.91 ± 0.40, 13.63 ± 0.36, 14.66 ± 0.47 and 12.26 ± 0.45 μM, respectively) and lung (A549) cancer cell line (IC50 = 15.38 ± 0.33, 10.00 ± 0.28, 12.38 ± 0.30, 12.84 ± 0.33 and 8.47 ± 0.30 μM, respectively) compared to quercetin (IC50 = 40.61 ± 1.07 and 58.17 ± 0.50 μM, respectively). Compounds 3b, 3c and 4b exhibited dual inhibitory effect against the vascular endothelial growth factor receptor-2 (VEGFR-2) and the epidermal growth factor receptor (EGFR) tyrosine kinase phosphorylation. Molecular docking revealed that strong alignment with the enzyme backbone is achieved mostly by hydrophobic (π-π, and π-H) contacts and by hydrogen bonding interaction with the residues in the active sites of VEGFR-2 and EGFR. The test compounds possess favorable drug-likeness properties, supporting their potential as promising therapeutic candidates.
Collapse
Affiliation(s)
- Jackson K Nkoana
- Department of Chemistry, College of Science, Engineering and Technology, University of South Africa, Private Bag X06, Florida 1710, South Africa
| | - Garland K More
- College of Agriculture and Environmental Sciences, University of South Africa, Private Bag X06, Florida 1710, South Africa
| | - Malose J Mphahlele
- Department of Chemistry, College of Science, Engineering and Technology, University of South Africa, Private Bag X06, Florida 1710, South Africa.
| | - Ahmed A Elhenawy
- Chemistry Department, Faculty of Science, Al-Azhar University, Nasr City, 11884 Cairo, Egypt; Chemistry Department, Faculty of Science, Al-Baha University, Al-Baha 1988, Saudi Arabia
| |
Collapse
|
10
|
Olomola TO, Nkoana JK, More GK, Gildenhuys S, Mphahlele MJ. Enzyme (α-Glucosidase, α-Amylase, PTP1B & VEGFR-2) Inhibition and Cytotoxicity of Fluorinated Benzenesulfonic Ester Derivatives of the 5-Substituted 2-Hydroxy-3-nitroacetophenones. Int J Mol Sci 2024; 25:11862. [PMID: 39595931 PMCID: PMC11594133 DOI: 10.3390/ijms252211862] [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: 10/15/2024] [Revised: 11/01/2024] [Accepted: 11/02/2024] [Indexed: 11/28/2024] Open
Abstract
The prevalence of small multi-target drugs containing a fluorinated aromatic moiety among approved drugs in the market is due to the unique properties of this halogen atom. With the aim to develop potent antidiabetic agents, a series of phenylsulfonic esters based on the conjugation of the 5-substituted 2-hydroxy-3-nitroacetophenones 1a-d with phenylsulfonyl chloride derivatives substituted with a fluorine atom or fluorine-containing (-CF3 or -OCF3) group were prepared. Their structures were characterized using a combination of spectroscopic techniques complemented with a single-crystal X-ray diffraction (XRD) analysis on a representative example. The compounds were, in turn, assayed for inhibitory effect against α-glucosidase, α-amylase, protein tyrosine phosphatase 1 B (PTP1B) and the vascular endothelial growth factor receptor-2 (VEGFR-2) all of which are associated with the pathogenesis and progression of type 2 diabetes mellitus (T2DM). The antigrowth effect of selected compounds was evaluated on the human breast (MCF-7) and lung (A549) cancer cell lines. The compounds were also evaluated for cytotoxicity against the African Green Monkey kidney (Vero) cell line. The results of an in vitro enzymatic study were augmented by molecular docking (in silico) analysis. Their ADME (absorption, distribution, metabolism and excretion) properties have been evaluated on the most active compounds against α-glucosidase and/or α-amylase to predict their drug likeness.
Collapse
Affiliation(s)
- Temitope O. Olomola
- Department of Chemistry, College of Science, Engineering and Technology, University of South Africa, Private Bag X06, Florida 1710, South Africa; (T.O.O.); (J.K.N.)
- Department of Chemistry, Faculty of Science, Obafemi Awolowo University, Ile-Ife 220005, Nigeria
| | - Jackson K. Nkoana
- Department of Chemistry, College of Science, Engineering and Technology, University of South Africa, Private Bag X06, Florida 1710, South Africa; (T.O.O.); (J.K.N.)
| | - Garland K. More
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Private Bag X06, Florida 1710, South Africa
| | - Samantha Gildenhuys
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Private Bag X06, Florida 1710, South Africa
| | - Malose J. Mphahlele
- Department of Chemistry, College of Science, Engineering and Technology, University of South Africa, Private Bag X06, Florida 1710, South Africa; (T.O.O.); (J.K.N.)
| |
Collapse
|
11
|
Wei F, Zhang Y. Palladium-Catalyzed Cascade Distal C-H Methylation and Cyclization for the Construction of Spirooxindole Skeletons. Org Lett 2024; 26:9221-9226. [PMID: 39423361 DOI: 10.1021/acs.orglett.4c03315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
Transition metal-catalyzed C-H methylation represents a straightforward approach for introducing methyl groups into organic molecules. Herein, we report a palladium-catalyzed alkene-relayed remote C-H methylation reaction that utilizes dimethyl carbonate as the methylation reagent. The aryl groups distal to a bromo group were dimethylated via C-H activation, leading to the formation of spirooxindoles as the final products through C(sp3)-H activation and C(sp3)-C(sp3) coupling. This cascade process involves the formation of four C-C bonds and the activation of three C-H bonds. The reaction not only provides a new approach to C-H methylation but also offers a novel method for constructing spirooxindole skeletons by merging skeleton construction and methylation into a single step.
Collapse
Affiliation(s)
- Feng Wei
- School of Chemical Science and Engineering, Shanghai Key Laboratory of Chemical Assessment and Sustainability, Tongji University, Shanghai 200092, China
| | - Yanghui Zhang
- School of Chemical Science and Engineering, Shanghai Key Laboratory of Chemical Assessment and Sustainability, Tongji University, Shanghai 200092, China
| |
Collapse
|
12
|
Konowalchuk DJ, Clement HA, Lofstrand VA, Kim JY, Burford KN. Diastereoselective Hydrodifluoromethylation of Alkenyl N-Heterocycles via Photocatalytic Radical-Polar Crossover. J Org Chem 2024; 89:16060-16064. [PMID: 39397589 DOI: 10.1021/acs.joc.4c02169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
A diastereoselective hydrodifluoromethylation of N-heteroaryl alkenes was successfully established. This method was applicable to an array of N-heteroaryl substrates with both cyclic and acyclic alkenes while displaying tolerance to a variety of functional groups. The conditions were also expanded to obtain hydrotrifluoromethylated products with similar results. Initial mechanistic studies suggest that the final protonation step is accessed through a radical-polar crossover process.
Collapse
Affiliation(s)
- Dawson J Konowalchuk
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, British Columbia, Canada, V5G 4W8
| | - Helen A Clement
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, British Columbia, Canada, V5G 4W8
| | - Verner A Lofstrand
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, British Columbia, Canada, V5G 4W8
| | - Jung Y Kim
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, British Columbia, Canada, V5G 4W8
| | - Kristen N Burford
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, British Columbia, Canada, V5G 4W8
| |
Collapse
|
13
|
Sumit, Sachin, Chandra D, Sharma U. Ru(II)-Catalyzed Sustainable C-H Methylation of Indolines with Organoboranes in Ethanol. J Org Chem 2024; 89:14880-14886. [PMID: 39240126 DOI: 10.1021/acs.joc.4c01650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
A sustainable protocol for Ru(II)-catalyzed regioselective C(sp2)-H methylation of indolines in the presence of ethanol has been explored. A wide array of substituted indolines were successfully methylated via the developed protocol with good to excellent yields. Deuterium labeling experiments suggested the reversible nature of the C-H activation step. Kinetic isotope effect studies revealed that C-H activation might be the rate-determining step. Gram scale reaction and post-transformation reactions of the methylated product demonstrated the potential of the developed protocol.
Collapse
Affiliation(s)
- Sumit
- C-H Activation & Phytochemistry Lab, Chemical Technology Division, CSIR-IHBT, Palampur 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sachin
- C-H Activation & Phytochemistry Lab, Chemical Technology Division, CSIR-IHBT, Palampur 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Devesh Chandra
- C-H Activation & Phytochemistry Lab, Chemical Technology Division, CSIR-IHBT, Palampur 176061, India
| | - Upendra Sharma
- C-H Activation & Phytochemistry Lab, Chemical Technology Division, CSIR-IHBT, Palampur 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
14
|
Tan JF, Kang YC, Hartwig JF. Catalytic undirected methylation of unactivated C(sp 3)-H bonds suitable for complex molecules. Nat Commun 2024; 15:8307. [PMID: 39333063 PMCID: PMC11437150 DOI: 10.1038/s41467-024-52245-1] [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: 04/23/2024] [Accepted: 08/28/2024] [Indexed: 09/29/2024] Open
Abstract
In pharmaceutical discovery, the "magic methyl" effect describes a substantial improvement in the pharmacological properties of a drug candidate with the incorporation of methyl groups. Therefore, to expedite the synthesis of methylated drug analogs, late-stage, undirected methylations of C(sp3)-H bonds in complex molecules would be valuable. However, current methods for site-selective methylations are limited to activated C(sp3)-H bonds. Here we describe a site-selective, undirected methylation of unactivated C(sp3)-H bonds, enabled by photochemically activated peroxides and a nickel(II) complex whose turnover is enhanced by an ancillary ligand. The methodology displays compatibility with a wide range of functional groups and a high selectivity for tertiary C-H bonds, making it suitable for the late-stage methylation of complex organic compounds that contain multiple alkyl C-H bonds, such as terpene natural products, peptides, and active pharmaceutical ingredients. Overall, this method provides a synthetic tool to explore the "magic methyl" effect in drug discovery.
Collapse
Affiliation(s)
- Jin-Fay Tan
- Department of Chemistry, University of California, Berkeley, CA, USA
| | - Yi Cheng Kang
- Department of Chemistry, University of California, Berkeley, CA, USA
| | - John F Hartwig
- Department of Chemistry, University of California, Berkeley, CA, USA.
| |
Collapse
|
15
|
Yao H, Ren Y, Wu F, Liu J, Li J, Cao L, Yan M, Li X. Discovery of new cyclopropane sulfonamide derivatives as EGFR inhibitors to overcome C797S-mediated resistance and EGFR double mutation. Eur J Med Chem 2024; 275:116590. [PMID: 38908104 DOI: 10.1016/j.ejmech.2024.116590] [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: 02/02/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/24/2024]
Abstract
The C797S mutation of EGFR leads to Osimertinib resistance by blocking the covalent binding of Cys797. To develop new agents that can overcome EGFR mutation resistance, thirty seven new cyclopropane sulfonamide derivatives were synthesized and evaluated as EGFRL858R/T790M/C797S or EGFRDel19/T790M/C797S inhibitors by structure-based screening. Most of the synthesized compounds exhibit good to excellent anti proliferation activity against to BaF3-EGFR L858R/T790M/C797S and BaF3-C797S/Del19/T790M cancer cell lines. Representative compounds 8l showed inhibitory activity against the two cancer cell lines with the IC50 values of 0.0012 and 0.0013 μM, respectively. Another compound 8h, exhibited slightly lower activity (0.0042 and 0.0034 μM of the IC50 values) to both of the two tri-mutation cell lines, but excellent activities against H1975 and PC9 cells with IC50 values of 13 and 19 nM, respectively. Considering the acquired drug resistance of tumors is a gradual process, we chose 8h for further in vivo and mechanism study. 8h was demonstrated significantly inhibited tumor growth with 72.1 % of the TGI in the BaF3/EGFR-TM xenograft tumor model and 83.5 % in the H1975-DM xenograft tumor model. Compound 8h was confirmed to be safe with no significant side effects as showed by the results of in vitro assay of human normal cells and the sections of animals major organs. Mechanism studies showed that in addition to inhibiting EGFR mutations, 8h can also target the tumor microenvironment and induce tumor cell apoptosis. All these results indicate that 8h deserves further investigation as an EGFR inhibitor to overcome C797S-mediated resistance.
Collapse
Affiliation(s)
- Han Yao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yuanyuan Ren
- Guangdong Provincial Key Laboratory of Drug Non-Clinical Evaluation and Research, Guangzhou, 510990, PR China
| | - Feng Wu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jiadai Liu
- Guangdong Provincial Key Laboratory of Drug Non-Clinical Evaluation and Research, Guangzhou, 510990, PR China
| | - Jianheng Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Longcai Cao
- Guangdong Provincial Key Laboratory of Drug Non-Clinical Evaluation and Research, Guangzhou, 510990, PR China
| | - Ming Yan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Xingshu Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Drug Non-Clinical Evaluation and Research, Guangzhou, 510990, PR China.
| |
Collapse
|
16
|
Chiodi D, Ishihara Y. The role of the methoxy group in approved drugs. Eur J Med Chem 2024; 273:116364. [PMID: 38781921 DOI: 10.1016/j.ejmech.2024.116364] [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: 01/04/2024] [Revised: 03/12/2024] [Accepted: 03/23/2024] [Indexed: 05/25/2024]
Abstract
The methoxy substituent is prevalent in natural products and, consequently, is present in many natural product-derived drugs. It has also been installed in modern drug molecules with no remnant of natural product features because medicinal chemists have been taking advantage of the benefits that this small functional group can bestow on ligand-target binding, physicochemical properties, and ADME parameters. Herein, over 230 methoxy-containing small-molecule drugs, as well as several fluoromethoxy-containing drugs, are presented from the vantage point of the methoxy group. Biochemical mechanisms of action, medicinal chemistry SAR studies, and numerous X-ray cocrystal structures are analyzed to identify the precise role of the methoxy group for many of the drugs and drug classes. Although the methoxy substituent can be considered as the hybridization of a hydroxy and a methyl group, the combination of these functionalities often results in unique effects that can amount to more than the sum of the individual parts.
Collapse
Affiliation(s)
- Debora Chiodi
- Department of Chemistry, Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, CA, 92121, USA
| | - Yoshihiro Ishihara
- Department of Chemistry, Vividion Therapeutics, 5820 Nancy Ridge Drive, San Diego, CA, 92121, USA.
| |
Collapse
|
17
|
Lin PC, Wong CD, Jarvo ER. Cross-selective Deoxygenative Coupling of Aliphatic Alcohols: Installation of Methyl Groups including Isotopic Labels by Nickel Catalysis. Angew Chem Int Ed Engl 2024; 63:e202403119. [PMID: 38604974 DOI: 10.1002/anie.202403119] [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: 02/13/2024] [Revised: 03/28/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
Nickel-catalyzed cross-electrophile coupling reactions of two aliphatic alcohol derivatives remain a challenge. Herein, we report a nickel-catalyzed reductive methylation reaction of aliphatic mesylates with methyl tosylate. This reaction provides straightforward access to compounds bearing aliphatic methyl groups from alkyl alcohol derivatives. Isotopically labelled substrates and reagents can be employed in the reaction to provide perdeuterated and 13C-labelled products. This transformation can be achieved by employing stoichiometric Mn reductant or electrochemically. Additionally, mechanistic experiments show that alkyl iodides are key intermediates in the transformation which undergo a stereoablative reaction via radical intermediates.
Collapse
Affiliation(s)
- Patricia C Lin
- Department of Chemistry, University of California, Irvine, California, 92617, United States
| | - Chloe D Wong
- Department of Chemistry, University of California, Irvine, California, 92617, United States
| | - Elizabeth R Jarvo
- Department of Chemistry, University of California, Irvine, California, 92617, United States
| |
Collapse
|
18
|
Zhao WW, Tian MY, Zhou YL, Liu LJ, Tian SF, He CY, Yang XZ, Chen YZ, Han WY. Trifluoromethyl Rhodium-Carbynoid in [2+1+2] Cycloadditions. Angew Chem Int Ed Engl 2024; 63:e202318887. [PMID: 38237082 DOI: 10.1002/anie.202318887] [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: 12/10/2023] [Indexed: 02/24/2024]
Abstract
Trifluoromethyl cationic carbyne (CF3 C+ :) possessing dual carbene-carbocation behavior emulated as trifluoromethyl metal-carbynoid (CF3 C+ =M) has not been explored yet, and its reaction characteristics are unknown. Herein, a novel α-diazotrifluoroethyl sulfonium salt was prepared and used in Rh-catalyzed three-component [2+1+2] cycloadditions for the first time with commercially available N-fused heteroarenes and nitriles, yielding a series of imidazo[1,5-a] N-heterocycles that are of interest in medicinal chemistry, in which the insertion of trifluoromethyl Rh-carbynoid (CF3 C+ =Rh) into C=N bonds of N-fused heteroarenes was involved. This strategy demonstrates synthetic applications in late-stage modification of pharmaceuticals, construction of CD3 -containing N-heterocycles, gram-scale experiments, and synthesis of phosphodiesterase 10A inhibitor analog. These highly valuable and modifiable imidazo[1,5-a] N-heterocycles exhibit good antitumor activity in vitro, thus demonstrating their potential applications in medicinal chemistry.
Collapse
Affiliation(s)
- Wen-Wen Zhao
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, No. 6 West Xuefu Rd., 563006, Zunyi, China
| | - Meng-Yang Tian
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, No. 6 West Xuefu Rd., 563006, Zunyi, China
| | - Yi-Lin Zhou
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, No. 6 West Xuefu Rd., 563006, Zunyi, China
| | - Lu-Jie Liu
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, No. 6 West Xuefu Rd., 563006, Zunyi, China
| | - Shao-Fang Tian
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, No. 6 West Xuefu Rd., 563006, Zunyi, China
| | - Chun-Yang He
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, No. 6 West Xuefu Rd., 563006, Zunyi, China
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, No. 6 West Xuefu Rd., 563006, Zunyi, China
| | - Xing-Zhi Yang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, 132 Lanhei Road, 650201, Kunming, China
| | - Yong-Zheng Chen
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, No. 6 West Xuefu Rd., 563006, Zunyi, China
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, No. 6 West Xuefu Rd., 563006, Zunyi, China
| | - Wen-Yong Han
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, No. 6 West Xuefu Rd., 563006, Zunyi, China
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, No. 6 West Xuefu Rd., 563006, Zunyi, China
| |
Collapse
|
19
|
Li QY, He Y, Lin YM, Gong L. Photo-Induced C-H Methylation Reactions. Chemistry 2023; 29:e202302542. [PMID: 37800464 DOI: 10.1002/chem.202302542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/21/2023] [Accepted: 10/04/2023] [Indexed: 10/07/2023]
Abstract
Direct C-H methylation is a highly valuable approach for introducing methyl groups into organic molecules, particularly in pharmaceutical chemistry. Among the various methodologies available, photo-induced methylation stands out as an exceptional choice due to its mild reaction conditions, energy efficiency, and compatibility with functional groups. This article offers a comprehensive review of photochemical strategies employed for the direct and selective methylation of C(sp3 )-H, C(sp2 )-H, and C(sp)-H bonds in various organic molecules. The discussed methodologies encompass transition-metal-based photocatalysis, organophotocatalysis, as well as other metal-free approaches, including electron donor-acceptor (EDA)-enabled transformations. Importantly, a wide range of easily accessible agents such as tert-butyl peroxide, methanol, DMSO, methyl tert-butyl ether, TsOMe, N-(acetoxy)phthalimide, acetic acid, methyl halides, and even methane can serve as effective methylating reagents for modifying diverse targets. These advancements in photochemical C-H methylation are anticipated to drive further progress in the fields of organic synthesis, photocatalysis, and pharmaceutical development, opening up exciting avenues for creating novel organic molecules and discovering new drug compounds.
Collapse
Affiliation(s)
- Qian-Yu Li
- Key Laboratory of Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, 361005, China
| | - Yuhang He
- Key Laboratory of Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, 361005, China
| | - Yu-Mei Lin
- Key Laboratory of Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, 361005, China
| | - Lei Gong
- Key Laboratory of Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, 361005, China
- Innovation Laboratory for Sciences and, Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen, Fujian, 361005, China
| |
Collapse
|
20
|
Menke MJ, Schneider P, Badenhorst CPS, Kunzendorf A, Heinz F, Dörr M, Hayes MA, Bornscheuer UT. A Universal, Continuous Assay for SAM-dependent Methyltransferases. Angew Chem Int Ed Engl 2023; 62:e202313912. [PMID: 37917964 DOI: 10.1002/anie.202313912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/04/2023]
Abstract
Enzyme-catalyzed late-stage functionalization (LSF), such as methylation of drug molecules and lead structures, enables direct access to more potent active pharmaceutical ingredients (API). S-adenosyl-l-methionine-dependent methyltransferases (MTs) can play a key role in the development of new APIs, as they catalyze the chemo- and regioselective methylation of O-, N-, S- and C-atoms, being superior to traditional chemical routes. To identify suitable MTs, we developed a continuous fluorescence-based, high-throughput assay for SAM-dependent methyltransferases, which facilitates screening using E. coli cell lysates. This assay involves two enzymatic steps for the conversion of S-adenosyl-l-homocysteine into H2 S to result in a selective fluorescence readout via reduction of an azidocoumarin sulfide probe. Investigation of two O-MTs and an N-MT confirmed that this assay is suitable for the determination of methyltransferase activity in E. coli cell lysates.
Collapse
Affiliation(s)
- Marian J Menke
- Department of Biotechnology and Enzyme Catalysis, Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, 17487, Greifswald, Germany
| | - Pascal Schneider
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, 43183, Mölndal, Sweden
| | - Christoffel P S Badenhorst
- Department of Biotechnology and Enzyme Catalysis, Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, 17487, Greifswald, Germany
| | - Andreas Kunzendorf
- Department of Biotechnology and Enzyme Catalysis, Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, 17487, Greifswald, Germany
| | - Florian Heinz
- Department of Biotechnology and Enzyme Catalysis, Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, 17487, Greifswald, Germany
| | - Mark Dörr
- Department of Biotechnology and Enzyme Catalysis, Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, 17487, Greifswald, Germany
| | - Martin A Hayes
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, 43183, Mölndal, Sweden
| | - Uwe T Bornscheuer
- Department of Biotechnology and Enzyme Catalysis, Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, 17487, Greifswald, Germany
| |
Collapse
|
21
|
Kanjariya DC, Naik HN, Sherashiya MJ, Naliapara YT, Ahmad I, Patel H, Rajani D, Jauhari S. α-Amylase and mycobacterium-TB H37Rv antagonistic efficacy of novel pyrazole-coumarin hybrids: an in vitro and in silico investigation. J Biomol Struct Dyn 2023; 42:12788-12805. [PMID: 37904535 DOI: 10.1080/07391102.2023.2273436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/15/2023] [Indexed: 11/01/2023]
Abstract
The present investigation of minutiae to acquire structural information of the novel pyrazole-coumarin hybrids (PC1-PC10) synthesized using ultrasound methods and characterized using different spectroscopic techniques: mass, 1H-NMR, 13 C-NMR and IR spectroscopy, and theoretically explored using the DFT approach with a B3LYP/6-311G (d, p) basis set, and there in vitro, antagonistic efficacy against α-amylase and mycobacterium-TB H37Rv are described in this article. Pyrazole-coumarin hybrids (PC1-PC10) showed α-amylase inhibition ranging from IC50 (0.32-0.58 mM) when compared with acarbose (IC50 = 0.34 mM). Similarly, Mycobacterium-TB H37Rv strain inhibition screening showed MIC values ranging from 62.5 to 1000 µg/mL when compared with rifampicin and isoniazid MIC = 0.25 and 0.20 µg/mL, respectively. Molecular docking and MD simulation studies were performed to determine the active sites and rationalize the activities of the active compounds. To investigate the binding conformation and dynamics responsible for their activity, the three most active compounds (PC1, PC3 and PC6) were docked into the porcine pancreatic α-amylase active site (PDB ID:1OSE), and mycobacterium-TB H37Rv active site (PDB ID: 4TZK). The binding interactions between PC1, PC3, and PC6 with α-amylase were like those responsible for inhibiting α-amylase by acarbose. Also, the mycobacterium-TB H37Rv inhibiting responsible residues were compared with standard isoniazid and rifampicin.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Dilip C Kanjariya
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat, India
| | - Hem N Naik
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat, India
| | | | | | - Iqrar Ahmad
- Department of Pharmaceutical Chemistry, Prof. Ravindra Nikam College of Pharmacy, Dhule, India
| | - Harun Patel
- Department of Pharmaceutical Chemistry, Division of Computer Aided Drug Design, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| | - Dhanji Rajani
- Microcare Laboratory and Tuberculosis Research Center, Surat, India
| | - Smita Jauhari
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat, India
| |
Collapse
|
22
|
Meger FS, Murphy JA. Recent Advances in C-H Functionalisation through Indirect Hydrogen Atom Transfer. Molecules 2023; 28:6127. [PMID: 37630379 PMCID: PMC10459052 DOI: 10.3390/molecules28166127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/09/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
The functionalisation of C-H bonds has been an enormous achievement in synthetic methodology, enabling new retrosynthetic disconnections and affording simple synthetic equivalents for synthons. Hydrogen atom transfer (HAT) is a key method for forming alkyl radicals from C-H substrates. Classic reactions, including the Barton nitrite ester reaction and Hofmann-Löffler-Freytag reaction, among others, provided early examples of HAT. However, recent developments in photoredox catalysis and electrochemistry have made HAT a powerful synthetic tool capable of introducing a wide range of functional groups into C-H bonds. Moreover, greater mechanistic insights into HAT have stimulated the development of increasingly site-selective protocols. Site-selectivity can be achieved through the tuning of electron density at certain C-H bonds using additives, a judicious choice of HAT reagent, and a solvent system. Herein, we describe the latest methods for functionalizing C-H/Si-H/Ge-H bonds using indirect HAT between 2018-2023, as well as a critical discussion of new HAT reagents, mechanistic aspects, substrate scopes, and background contexts of the protocols.
Collapse
Affiliation(s)
- Filip S. Meger
- Institute of Chemical Research of Catalonia (ICIQ), The Barcelona Institute of Science and Technology, 16 Avinguda dels Països Catalans, 43007 Tarragona, Catalonia, Spain
| | - John A. Murphy
- Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, UK
| |
Collapse
|
23
|
Guo F, Shan S, Gong X, Dai C, Quan Z, Cheng X, Fan X. Deuteration Degree-Controllable Methylation via a Cascade Assembly Strategy using Methylamine-Water as Methyl Source. Chemistry 2023; 29:e202301458. [PMID: 37222652 DOI: 10.1002/chem.202301458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 05/24/2023] [Accepted: 05/24/2023] [Indexed: 05/25/2023]
Abstract
We present a novel and effective photocatalytic method for the methylation of β-diketones with controllable degrees of deuterium incorporation via development of new methyl sources. By utilizing a methylamine-water system as the methyl precursor and a cascade assembly strategy for deuteration degree control, we synthesized methylated compounds with varying degrees of deuterium incorporation, showcasing the versatility of this approach. We examined a range of β-diketone substrates and synthesized key intermediates for drug and bioactive compounds with varying degrees of deuterium incorporation, ranging from 0 to 3. We also investigated and discussed the postulated reaction pathway. This work demonstrates the utility of readily available reagents, methylamines and water, as a new methyl source, and provides a simple and efficient strategy for the synthesis of degree-controllable deuterium-labelled compounds.
Collapse
Affiliation(s)
- Fuhu Guo
- College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, Gansu, 730070, China
- Synthetic and Functional Biomolecules Center, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Shiquan Shan
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Xu Gong
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Cancan Dai
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Zhengjun Quan
- College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, Gansu, 730070, China
| | - Xiamin Cheng
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Xinyuan Fan
- Synthetic and Functional Biomolecules Center, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| |
Collapse
|
24
|
Kathiravan S, Zhang T, Nicholls IA. Iridium catalysed C2 site-selective methylation of indoles using a pivaloyl directing group through weak chelation-assistance. RSC Adv 2023; 13:11291-11295. [PMID: 37057266 PMCID: PMC10088075 DOI: 10.1039/d3ra02031b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 04/04/2023] [Indexed: 04/15/2023] Open
Abstract
Here we present an iridium catalysed C2-selective methylation of indoles using methyltrifluoroborate as a source of methyl group. The iridium catalyst selectively discriminates the indole C2 and C4 C-H bonds by coordination with a pivaloyl directing group.
Collapse
Affiliation(s)
| | - Tianshu Zhang
- Bioorganic & Biophysical Chemistry Laboratory, Linnaeus University Centre for Biomaterials Chemistry, Department of Chemistry & Biomedical Sciences, Linnaeus University Kalmar SE-39182 Sweden
| | - Ian A Nicholls
- Bioorganic & Biophysical Chemistry Laboratory, Linnaeus University Centre for Biomaterials Chemistry, Department of Chemistry & Biomedical Sciences, Linnaeus University Kalmar SE-39182 Sweden
| |
Collapse
|
25
|
Song S, Cheng X, Cheng S, Lin YM, Gong L. Fe-Catalyzed Aliphatic C-H Methylation of Glycine Derivatives and Peptides. Chemistry 2023; 29:e202203404. [PMID: 36545842 DOI: 10.1002/chem.202203404] [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: 11/02/2022] [Revised: 12/14/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
Direct and selective C-H methylation is a powerful tool with which to install methyl groups into organic molecules, and is particularly useful in pharmaceutical chemistry. However, practical methods for such modification of biologically interesting targets have been rarely developed. We here report an iron-catalyzed C(sp3 )-H methylation reaction of glycine derivatives, peptides and drug-like molecules in an alcohol in the presence of di-tert-butyl peroxide. A readily available iron catalyst plays multiple roles in the transformation, which accelerates oxidation of C-N bonds to C=N double bonds, activates imine intermediates as Lewis acids by bidentate chelation, and at the same time facilitates cleavage of the peroxide to generate methyl radicals. A variety of methylated N-aryl glycine derivatives and peptides were obtained in good yield and with excellent chemo- and site-selectivity. This reaction is scalable, easily managed, and can be completed within 1-2 h. It features an economic, bio-friendly catalyst, a green solvent and low toxic reagents, and will provide effective access to precise C-H modification of biomolecules and natural products.
Collapse
Affiliation(s)
- Silin Song
- Key Laboratory of Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, 361005, Xiamen, China
| | - Xiuliang Cheng
- Key Laboratory of Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, 361005, Xiamen, China
| | - Shiyan Cheng
- Key Laboratory of Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, 361005, Xiamen, China
| | - Yu-Mei Lin
- Key Laboratory of Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, 361005, Xiamen, China
| | - Lei Gong
- Key Laboratory of Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, 361005, Xiamen, China
- Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), 361005, Xiamen, China
| |
Collapse
|
26
|
Dutta S, Sahoo AK. Three Component syn-1,2-Arylmethylation of Internal Alkynes. Angew Chem Int Ed Engl 2023; 62:e202300610. [PMID: 36701082 DOI: 10.1002/anie.202300610] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 01/26/2023] [Accepted: 01/26/2023] [Indexed: 01/27/2023]
Abstract
A Pd-catalyzed three-component syn-1,2-arylmethylation of internal alkynes (ynamides/yne-acetates/alkynes) is described. The readily available and bench stable coupling partners iodo-arenes, and methyl boronic acid are successfully used in this coupling strategy to access the methyl-containing tetra-substituted olefins; the scope is broad showing excellent functional-group tolerance. Notably, the transformation is regio- as well as stereoselective. The biologically relevant motifs (BRM) bearing iodo-arenes and ynamides are also used for the late-stage syn-1,2-arylmethylation of alkynes. Aryl-alkylation, aryl-trideuteriomethylation, alkynyl-methylation, and alkenyl-methylation of ynamides are also presented. The Me-substituted alkenes are further transformed into synthetically important β-amino-indenones and α-fluoro-α'-methyl ketones.
Collapse
Affiliation(s)
- Shubham Dutta
- School of Chemistry, University of Hyderabad, Hyderabad, 500046, India
| | - Akhila K Sahoo
- School of Chemistry, University of Hyderabad, Hyderabad, 500046, India
| |
Collapse
|
27
|
Wang FY, Li YX, Jiao L. Functionalized Cycloolefin Ligand as a Solution to Ortho-Constraint in the Catellani-Type Reaction. J Am Chem Soc 2023; 145:4871-4881. [PMID: 36795897 DOI: 10.1021/jacs.3c00329] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
The Catellani reaction, i.e., the Pd/norbornene (NBE) catalysis, has been evolved into a versatile approach to multisubstituted arenes via the ortho-functionalization/ipso-termination process of a haloarene. Despite significant advances over the past 25 years, this reaction still suffered from an intrinsic limitation in the substitution pattern of haloarene, referred to as "ortho-constraint". When an ortho substituent is absent, the substrate often fails to undergo an effective mono ortho-functionalization process, and either ortho-difunctionalization products or NBE-embedded byproducts predominate. To tackle this challenge, structurally modified NBEs (smNBEs) have been developed, which were proved effective for the mono ortho-aminative, -acylative, and -arylative Catellani reactions of ortho-unsubstituted haloarenes. However, this strategy is incompetent for solving the ortho-constraint in Catellani reactions with ortho-alkylation, and to date there lacks a general solution to this challenging but synthetically useful transformation. Recently, our group developed the Pd/olefin catalysis, in which an unstrained cycloolefin ligand served as a covalent catalytic module to enable the ortho-alkylative Catellani reaction without NBE. In this work, we show that this chemistry could afford a new solution to ortho-constraint in the Catellani reaction. A functionalized cycloolefin ligand bearing an amide group as the internal base was designed, which allowed for mono ortho-alkylative Catellani reaction of iodoarenes suffering from ortho-constraint before. Mechanistic study revealed that this ligand is capable of both accelerating the C-H activation and inhibiting side reactions, which accounts for its superior performance. The present work showcased the uniqueness of the Pd/olefin catalysis as well as the power of rational ligand design in metal catalysis.
Collapse
Affiliation(s)
- Feng-Yuan Wang
- Center of Basic Molecular Science (CBMS), Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Yu-Xiu Li
- Center of Basic Molecular Science (CBMS), Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Lei Jiao
- Center of Basic Molecular Science (CBMS), Department of Chemistry, Tsinghua University, Beijing 100084, China
| |
Collapse
|
28
|
Tang J, He J, Zhao SY, Liu W. Manganese-Catalyzed Chemoselective Coupling of Secondary Alcohols, Primary Alcohols and Methanol. Angew Chem Int Ed Engl 2023; 62:e202215882. [PMID: 36847452 DOI: 10.1002/anie.202215882] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 03/01/2023]
Abstract
Herein, we report a manganese-catalyzed three-component coupling of secondary alcohols, primary alcohols and methanol for the synthesis of β,β-methylated/alkylated secondary alcohols. Using our method, a series of 1-arylethanol, benzyl alcohol derivatives, and methanol undergo sequential coupling efficiently to construct assembled alcohols with high chemoselectivity in moderate to good yields. Mechanistic studies suggest that the reaction proceeds via methylation of a benzylated secondary alcohol intermediate to generate the final product.
Collapse
Affiliation(s)
- Jun Tang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Key Lab of Science and Technology of Eco-Textile, Ministry of Education, College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China
| | - Jingxi He
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Key Lab of Science and Technology of Eco-Textile, Ministry of Education, College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China
| | - Sheng-Yin Zhao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Key Lab of Science and Technology of Eco-Textile, Ministry of Education, College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China
| | - Weiping Liu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Key Lab of Science and Technology of Eco-Textile, Ministry of Education, College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China
| |
Collapse
|
29
|
da Rocha MJ, Pires CS, Presa MH, Besckow EM, Nunes GD, Gomes CS, Penteado F, Lenardão EJ, Bortolatto CF, Brüning CA. Involvement of the serotonergic system in the antidepressant-like effect of 1-(phenylselanyl)-2-(p-tolyl)indolizine in mice. Psychopharmacology (Berl) 2023; 240:373-389. [PMID: 36645465 DOI: 10.1007/s00213-023-06313-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 01/05/2023] [Indexed: 01/17/2023]
Abstract
RATIONALE Depression is a mental disorder that affects approximately 280 million people worldwide. In the search for new treatments for mood disorders, compounds containing selenium and indolizine derivatives show promising results. OBJECTIVES AND METHODS To evaluate the antidepressant-like effect of 1-(phenylselanyl)-2-(p-tolyl)indolizine (MeSeI) (0.5-50 mg/kg, intragastric-i.g.) on the tail suspension test (TST) and the forced swim test (FST) in adult male Swiss mice and to elucidate the role of the serotonergic system in this effect through pharmacological and in silico approaches, as well to evaluate acute oral toxicity at a high dose (300 mg/kg). RESULTS MeSeI administered 30 min before the FST and the TST reduced immobility time at doses from 1 mg/kg and at 50 mg/kg and increased the latency time for the first episode of immobility, demonstrating an antidepressant-like effect. In the open field test (OFT), MeSeI did not change the locomotor activity. The antidepressant-like effect of MeSeI (50 mg/kg, i.g.) was prevented by the pre-treatment with p-chlorophenylalanine (p-CPA), a selective tryptophan hydroxylase inhibitor (100 mg/kg, intraperitoneally-i.p. for 4 days), with ketanserin, a 5-HT2A/2C receptor antagonist (1 mg/kg, i.p.), and with GR113808, a 5-HT4 receptor antagonist (0.1 mg/kg, i.p.), but not with WAY100635, a selective 5-HT1A receptor antagonist (0.1 mg/kg, subcutaneous-s.c.) and ondansetron, a 5-HT3 receptor antagonist (1 mg/kg, i.p.). MeSeI showed a binding affinity with 5-HT2A, 5 -HT2C, and 5-HT4 receptors by molecular docking. MeSeI (300 mg/kg, i.g.) demonstrated low potential to cause acute toxicity in adult female Swiss mice. CONCLUSION In summary, MeSeI exhibits an antidepressant-like effect mediated by the serotonergic system and could be considered for the development of new treatment strategies for depression.
Collapse
Affiliation(s)
- Marcia Juciele da Rocha
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão Do Leão Campus, Pelotas, RS, 96010-900, Brazil
| | - Camila Simões Pires
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão Do Leão Campus, Pelotas, RS, 96010-900, Brazil
| | - Marcelo Heinemann Presa
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão Do Leão Campus, Pelotas, RS, 96010-900, Brazil
| | - Evelyn Mianes Besckow
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão Do Leão Campus, Pelotas, RS, 96010-900, Brazil
| | - Gustavo D'Avila Nunes
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão Do Leão Campus, Pelotas, RS, 96010-900, Brazil
| | - Caroline Signorini Gomes
- Clean Organic Synthesis Laboratory (LASOL), Postgraduate Program in Chemistry (PPGQ), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão Do Leão Campus, Pelotas, RS, 96010-900, Brazil
| | - Filipe Penteado
- Clean Organic Synthesis Laboratory (LASOL), Postgraduate Program in Chemistry (PPGQ), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão Do Leão Campus, Pelotas, RS, 96010-900, Brazil
| | - Eder João Lenardão
- Clean Organic Synthesis Laboratory (LASOL), Postgraduate Program in Chemistry (PPGQ), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão Do Leão Campus, Pelotas, RS, 96010-900, Brazil
| | - Cristiani Folharini Bortolatto
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão Do Leão Campus, Pelotas, RS, 96010-900, Brazil.
| | - César Augusto Brüning
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão Do Leão Campus, Pelotas, RS, 96010-900, Brazil.
| |
Collapse
|
30
|
Diversification of pharmaceutical molecules via late-stage C(sp2)–H functionalization. GREEN SYNTHESIS AND CATALYSIS 2023. [DOI: 10.1016/j.gresc.2022.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
31
|
Sheetal, Mehara P, Das P. Methanol as a greener C1 synthon under non-noble transition metal-catalyzed conditions. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
32
|
Zhao H, Zeng Q, Yang J, Xu B, Lei H, Xu L, Walsh PJ. Rhodium(I)-catalyzed directed trideuteromethylation of (hetero)arene C-H bonds with CD 3CO 2D. Org Biomol Chem 2022; 20:7645-7649. [PMID: 36125438 DOI: 10.1039/d2ob01581a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A Rh(I)-catalyzed trideuteromethylation of heteroarenes with inexpensive and readily available deuterated acetic acid (CD3CO2D) with the aid of a N-containing directing groups is developed. The oxidant-free reaction is applicable to a wide range of heteroarene substrates, including 2-pyridones, indoles, aryl rings, pyrroles and carbazoles. It allows installation of CD3 groups under straightforward reaction conditions. It is expected that the salient and practical features of this trideuteromethylation protocol will be of use to academic and industrial researchers.
Collapse
Affiliation(s)
- Haoqiang Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China. .,Department of Chemistry, Renmin University of China, Beijing 100872, China. .,Roy and Diana Vagelos Laboratories, Penn/Merck Laboratory for High-Throughput Experimentation, Department of Chemistry, University of Pennsylvania, 231 South 34th Street, Philadelphia, Pennsylvania 19104-6323, USA.
| | - Qi Zeng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Ji Yang
- Department of Chemistry, Renmin University of China, Beijing 100872, China.
| | - Bing Xu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Haimin Lei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Lijin Xu
- Department of Chemistry, Renmin University of China, Beijing 100872, China.
| | - Patrick J Walsh
- Roy and Diana Vagelos Laboratories, Penn/Merck Laboratory for High-Throughput Experimentation, Department of Chemistry, University of Pennsylvania, 231 South 34th Street, Philadelphia, Pennsylvania 19104-6323, USA.
| |
Collapse
|
33
|
SAR study of oxidative DIMs analogs targeting the Nur77-mediated apoptotic pathway of cancer cells. Bioorg Chem 2022; 129:106156. [PMID: 36179441 DOI: 10.1016/j.bioorg.2022.106156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/21/2022] [Accepted: 09/12/2022] [Indexed: 11/21/2022]
Abstract
Nur77, an orphan nuclear receptor, is implicated in regulating diverse cellular biological processes including apoptosis and inflammation. We previously identified BI1071 (DIM-C-pPhCF3+MeSO3-), an oxidized methanesulfonate salt of (4-CF3-Ph-C-DIM), was a direct ligand of Nur77, which could activate the Nur77-Bcl-2 apoptotic pathway. To obtain more effective compounds targeting the Nur77-mediated apoptotic pathway, we designed and synthesized a series of BI1071 analogs by introducing various substituent groups in the indolyl-rings of BI1071. Structure-activity relationship study identified A11, B5 and B15 as improved analogs with stronger binding affinity to Nur77 and enhanced apoptotic activity compared to BI1071. Nur77-binding studies demonstrated that A11, B5 and B15 bind to Nur77 with a Kd of 34 nM, 19 nM and 16 nM, respectively. Furthermore, mechanism studies showed that A11, B5 and B15 induced apoptosis through utilizing the Nur77-Bcl-2 pathway.
Collapse
|
34
|
Abdelraheem E, Thair B, Varela RF, Jockmann E, Popadić D, Hailes HC, Ward JM, Iribarren AM, Lewkowicz ES, Andexer JN, Hagedoorn P, Hanefeld U. Methyltransferases: Functions and Applications. Chembiochem 2022; 23:e202200212. [PMID: 35691829 PMCID: PMC9539859 DOI: 10.1002/cbic.202200212] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/10/2022] [Indexed: 11/25/2022]
Abstract
In this review the current state-of-the-art of S-adenosylmethionine (SAM)-dependent methyltransferases and SAM are evaluated. Their structural classification and diversity is introduced and key mechanistic aspects presented which are then detailed further. Then, catalytic SAM as a target for drugs, and approaches to utilise SAM as a cofactor in synthesis are introduced with different supply and regeneration approaches evaluated. The use of SAM analogues are also described. Finally O-, N-, C- and S-MTs, their synthetic applications and potential for compound diversification is given.
Collapse
Affiliation(s)
- Eman Abdelraheem
- BiocatalysisDepartment of BiotechnologyDelft University of TechnologyVan der Maasweg 92629 HZDelft (TheNetherlands
| | - Benjamin Thair
- Department of ChemistryUniversity College London20 Gordon StreetLondonWC1H 0AJUK
| | - Romina Fernández Varela
- Laboratorio de Biotransformaciones y Química de Ácidos NucleicosUniversidad Nacional de QuilmesRoque S. Peña 352B1876BXDBernalArgentina
| | - Emely Jockmann
- Institute of Pharmaceutical SciencesUniversity of FreiburgAlbertstr. 2579104FreiburgGermany
| | - Désirée Popadić
- Institute of Pharmaceutical SciencesUniversity of FreiburgAlbertstr. 2579104FreiburgGermany
| | - Helen C. Hailes
- Department of ChemistryUniversity College London20 Gordon StreetLondonWC1H 0AJUK
| | - John M. Ward
- Department of Biochemical EngineeringBernard Katz BuildingUniversity College LondonLondonWC1E 6BTUK
| | - Adolfo M. Iribarren
- Laboratorio de Biotransformaciones y Química de Ácidos NucleicosUniversidad Nacional de QuilmesRoque S. Peña 352B1876BXDBernalArgentina
| | - Elizabeth S. Lewkowicz
- Laboratorio de Biotransformaciones y Química de Ácidos NucleicosUniversidad Nacional de QuilmesRoque S. Peña 352B1876BXDBernalArgentina
| | - Jennifer N. Andexer
- Institute of Pharmaceutical SciencesUniversity of FreiburgAlbertstr. 2579104FreiburgGermany
| | - Peter‐Leon Hagedoorn
- BiocatalysisDepartment of BiotechnologyDelft University of TechnologyVan der Maasweg 92629 HZDelft (TheNetherlands
| | - Ulf Hanefeld
- BiocatalysisDepartment of BiotechnologyDelft University of TechnologyVan der Maasweg 92629 HZDelft (TheNetherlands
| |
Collapse
|
35
|
Dong Y, Shin K, Mai BK, Liu P, Buchwald SL. Copper Hydride-Catalyzed Enantioselective Olefin Hydromethylation. J Am Chem Soc 2022; 144:16303-16309. [PMID: 36044255 PMCID: PMC9994624 DOI: 10.1021/jacs.2c07489] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The enantioselective installation of a methyl group onto a small molecule can result in the significant modification of its biological properties. While hydroalkylation of olefins represents an attractive approach to introduce alkyl substituents, asymmetric hydromethylation protocols are often hampered by the incompatibility of highly reactive methylating reagents and a lack of general applicability. Herein, we report an asymmetric olefin hydromethylation protocol enabled by CuH catalysis. This approach leverages methyl tosylate as a methyl source compatible with the reducing base-containing reaction environment, while a catalytic amount of iodide ion transforms the methyl tosylate in situ into the active reactant, methyl iodide, to promote the hydromethylation. This method tolerates a wide range of functional groups, heterocycles, and pharmaceutically relevant frameworks. Density functional theory studies suggest that after the stereoselective hydrocupration, the methylation step is stereoretentive, taking place through an SN2-type oxidative addition mechanism with methyl iodide followed by a reductive elimination.
Collapse
Affiliation(s)
- Yuyang Dong
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Kwangmin Shin
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Binh Khanh Mai
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Peng Liu
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Stephen L Buchwald
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
36
|
Di Mola A, Nicastro G, Serusi L, Filosa R, Waser M, Massa A. Scalable (Enantioselective) Syntheses of Novel 3-Methylated Analogs of Pazinaclone, (S)-PD172938 and Related Biologically Relevant Isoindolinones. Molecules 2022; 27:molecules27175647. [PMID: 36080411 PMCID: PMC9458024 DOI: 10.3390/molecules27175647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Herein, we report the application of an efficient and practical K2CO3 promoted cascade reaction of 2-acetylbenzonitrile in the synthesis of novel 3-methylated analogs of Pazinaclone and PD172938, belonging to isoindolinones heterocyclic class bearing a tetrasubstituted stereocenter. Organocatalytic asymmetric synthesis of the key intermediate and its transformation into highly enantioenriched 3-methylated analog of (S)-PD172938 was also developed. These achievements can be of particular interest also for medicinal chemistry, since the methyl group is a very useful structural modification in the rational design of new and more effective bioactive compounds.
Collapse
Affiliation(s)
- Antonia Di Mola
- Dipartimento di Chimica e Biologia “A. Zambelli”, Università degli Studi di Salerno, Via Giovanni Paolo II, 84084 Fisciano, Italy
- Correspondence: (A.D.M.); (A.M.)
| | - Giorgia Nicastro
- Dipartimento di Chimica e Biologia “A. Zambelli”, Università degli Studi di Salerno, Via Giovanni Paolo II, 84084 Fisciano, Italy
| | - Lorenzo Serusi
- Dipartimento di Chimica e Biologia “A. Zambelli”, Università degli Studi di Salerno, Via Giovanni Paolo II, 84084 Fisciano, Italy
| | - Rosanna Filosa
- Dipartimento di Scienze e Tecnologia, Università degli Studi del Sannio, Via De Sanctis, 82100 Benevento, Italy
| | - Mario Waser
- Institute of Organic Chemistry, Johannes Kepler University Linz, Altenbergerstr. 69, 4040 Linz, Austria
| | - Antonio Massa
- Dipartimento di Chimica e Biologia “A. Zambelli”, Università degli Studi di Salerno, Via Giovanni Paolo II, 84084 Fisciano, Italy
- Correspondence: (A.D.M.); (A.M.)
| |
Collapse
|
37
|
Improved flotation separation of sulfide minerals by synthesized surfactant based on para-position methyl effect. Sep Purif Technol 2022. [DOI: 10.1016/j.seppur.2022.121550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
38
|
Lee BJ, Ickes AR, Gupta AK, Ensign SC, Ho TD, Tarasewicz A, Vanable EP, Kortman GD, Hull KL. Synthesis of Unsymmetrical Vicinal Diamines via Directed Hydroamination. Org Lett 2022; 24:5513-5518. [PMID: 35862860 PMCID: PMC9757009 DOI: 10.1021/acs.orglett.2c01911] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Vicinal diamines are a common motif found in biologically active molecules. The hydroamination of allyl amine derivatives is a powerful approach for the synthesis of substituted 1,2-diamines. Herein, the rhodium-catalyzed hydroamination of primary and secondary allylic amines using diverse amine nucleophiles, including primary, secondary, acyclic, and cyclic aliphatic amines to access a wide range of unsymmetrical vicinal diamines, is presented. The utility of this methodology is further demonstrated through the rapid synthesis of several bioactive molecules and analogs.
Collapse
Affiliation(s)
- Byung Joo Lee
- Department of Chemistry, University of Texas at Austin, 105 E 24th St. Austin, TX 78712, United States
| | - Andrew R. Ickes
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 S. Mathews, Urbana, IL 61812, United States
| | - Anil K. Gupta
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 S. Mathews, Urbana, IL 61812, United States
| | - Seth C. Ensign
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 S. Mathews, Urbana, IL 61812, United States
| | - Tam D. Ho
- Department of Chemistry, University of Texas at Austin, 105 E 24th St. Austin, TX 78712, United States
| | - Anika Tarasewicz
- Department of Chemistry, University of Texas at Austin, 105 E 24th St. Austin, TX 78712, United States
| | - Evan P. Vanable
- Department of Chemistry and Biochemistry, Elmhust University, 190 Prospect Avenue, Elmhurst, IL 60126, United States
| | - Gregory D. Kortman
- Department of Chemistry, University of Texas at Austin, 105 E 24th St. Austin, TX 78712, United States
| | - Kami L. Hull
- Department of Chemistry, University of Texas at Austin, 105 E 24th St. Austin, TX 78712, United States
- Department of Chemistry, University of Illinois Urbana-Champaign, 600 S. Mathews, Urbana, IL 61812, United States
| |
Collapse
|
39
|
Parcella K, Wang T, Eastman K, Zhang Z, Yin Z, Patel M, Tu Y, Zheng BZ, Walker MA, Saulnier MG, Frennesson D, Bowsher M, Gillis E, Peese K, Belema M, Cianci C, Dicker IB, McAuliffe B, Ding B, Falk P, Simmermacher J, Parker DD, Sivaprakasam P, Kish K, Lewis H, Hanumegowda U, Jenkins S, Kadow JF, Krystal M, Meanwell NA, Naidu BN. Discovery and Preclinical Profiling of GSK3839919, a Potent HIV-1 Allosteric Integrase Inhibitor. ACS Med Chem Lett 2022; 13:972-980. [DOI: 10.1021/acsmedchemlett.2c00115] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/04/2022] [Indexed: 11/28/2022] Open
Affiliation(s)
- Kyle Parcella
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Tao Wang
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Kyle Eastman
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Zhongxing Zhang
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Zhiwei Yin
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Manoj Patel
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Yong Tu
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Barbara Zhizhen Zheng
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Michael A. Walker
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Mark G. Saulnier
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - David Frennesson
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Michael Bowsher
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Eric Gillis
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Kevin Peese
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Makonen Belema
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Christopher Cianci
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Ira B. Dicker
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Brian McAuliffe
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Bo Ding
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Paul Falk
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Jean Simmermacher
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Dawn D. Parker
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Prasanna Sivaprakasam
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Kevin Kish
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Hal Lewis
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Umesh Hanumegowda
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Susan Jenkins
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - John F. Kadow
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Mark Krystal
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Nicholas A. Meanwell
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - B. Narasimhulu Naidu
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| |
Collapse
|
40
|
Yang QL, Liu Y, Liang L, Li ZH, Qu GR, Guo HM. Facilitating Rh-Catalyzed C-H Alkylation of (Hetero)arenes and 6-Arylpurine Nucleosides (Nucleotides) with Electrochemistry. J Org Chem 2022; 87:6161-6178. [PMID: 35438486 DOI: 10.1021/acs.joc.2c00391] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
An electrochemical approach to promote the ortho-C-H alkylation of (hetero)arenes via rhodium catalysis under mild conditions is described. This approach features mild conditions with high levels of regio- and monoselectivity that tolerate a variety of aromatic and heteroaromatic groups and offers a widely applicable method for late-stage diversification of complex molecular architectures including tryptophan, estrone, diazepam, nucleosides, and nucleotides. Alkyl boronic acids and esters and alkyl trifluoroborates are demonstrated as suitable coupling partners. The isolation of key rhodium intermediates and mechanistic studies provided strong support for a rhodium(III/IV or V) regime.
Collapse
Affiliation(s)
- Qi-Liang Yang
- NMPA Key Laboratory for Research and Evaluation of Innovative Drug, Henan Key Laboratory of Organic Functional Molecules and Drug Innovation, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Ying Liu
- NMPA Key Laboratory for Research and Evaluation of Innovative Drug, Henan Key Laboratory of Organic Functional Molecules and Drug Innovation, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Lei Liang
- School of Chemistry and Chemical Engineering, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Zhi-Hao Li
- NMPA Key Laboratory for Research and Evaluation of Innovative Drug, Henan Key Laboratory of Organic Functional Molecules and Drug Innovation, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Gui-Rong Qu
- NMPA Key Laboratory for Research and Evaluation of Innovative Drug, Henan Key Laboratory of Organic Functional Molecules and Drug Innovation, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Hai-Ming Guo
- NMPA Key Laboratory for Research and Evaluation of Innovative Drug, Henan Key Laboratory of Organic Functional Molecules and Drug Innovation, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| |
Collapse
|
41
|
Templ J, Schnürch M. Selective α-Methylation of Aryl Ketones Using Quaternary Ammonium Salts as Solid Methylating Agents. J Org Chem 2022; 87:4305-4315. [PMID: 35253422 PMCID: PMC8938946 DOI: 10.1021/acs.joc.1c03158] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Indexed: 11/28/2022]
Abstract
We describe the use of phenyl trimethylammonium iodide (PhMe3NI) as an alternative methylating agent for introducing a CH3 group in α-position to a carbonyl group. Compared to conventional methylating agents, quaternary ammonium salts have the advantages of being nonvolatile, noncancerogenic, and easy-to-handle solids. This regioselective method is characterized by ease of operational setup, use of anisole as green solvent, and yields up to 85%.
Collapse
Affiliation(s)
- Johanna Templ
- Institute of Applied Synthetic
Chemistry, TU Wien, Getreidemarkt 9/163, 1060 Wien, Austria
| | - Michael Schnürch
- Institute of Applied Synthetic
Chemistry, TU Wien, Getreidemarkt 9/163, 1060 Wien, Austria
| |
Collapse
|
42
|
Ramesh D, Sarkar D, Joji A, Singh M, Mohanty AK, G Vijayakumar B, Chatterjee M, Sriram D, Muthuvel SK, Kannan T. First-in-class pyrido[2,3-d]pyrimidine-2,4(1H,3H)-diones against leishmaniasis and tuberculosis: Rationale, in vitro, ex vivo studies and mechanistic insights. Arch Pharm (Weinheim) 2022; 355:e2100440. [PMID: 35106845 DOI: 10.1002/ardp.202100440] [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: 11/08/2021] [Revised: 12/22/2021] [Accepted: 01/07/2022] [Indexed: 11/06/2022]
Abstract
Pyrido[2,3-d]pyrimidine-2,4(1H,3H)-diones were synthesized, for the first time, from indole chalcones and 6-aminouracil, and their ability to inhibit leishmaniasis and tuberculosis (Tb) infections was evaluated. The in vitro antileishmanial activity against promastigotes of Leishmania donovani revealed exceptional activities of compounds 3, 12 and 13, with IC50 values ranging from 10.23 ± 1.50 to 15.58 ± 1.67 µg/ml, which is better than the IC50 value of the standard drug pentostam of 500 μg/ml. The selectivity of the compounds towards Leishmania parasites was evaluated via ex vivo studies in Swiss albino mice. The efficiency of these compounds against Tb infection was then evaluated using the in vitro anti-Tb microplate Alamar Blue assay. Five compounds, 3, 7, 8, 9 and 12, showed MIC100 values against the Mycobacterium tuberculosis H37 Rv strain at 25 µg/ml, and compound 20 yielded an MIC100 value of 50 µg/ml. Molecular modelling of these compounds highlighted interactions with binding sites of dihydrofolate reductase, pteridine reductase and thymidylate kinase, thus establishing the rationale of their pharmacological activity against both pathogens, which is consistent with the in vitro results. From the above results, it is clear that compounds 3 and 12 are promising lead candidates for Leishmania and Mycobacterium infections and may be promising for coinfections.
Collapse
Affiliation(s)
- Deepthi Ramesh
- Department of Chemistry, Pondicherry University, Kalapet, Puducherry, India
| | - Deblina Sarkar
- Department of Pharmacology, Institute of Post Graduate Medical Education & Research (IPGME&R), Kolkata, West Bengal, India
| | - Annu Joji
- Department of Chemistry, Pondicherry University, Kalapet, Puducherry, India
| | - Monica Singh
- Department of Pharmacy, Birla Institute of Technology & Science Pilani, Hyderabad, India
| | - Amaresh K Mohanty
- Department of Bioinformatics, Pondicherry University, Kalapet, Puducherry, India
| | | | - Mitali Chatterjee
- Department of Pharmacology, Institute of Post Graduate Medical Education & Research (IPGME&R), Kolkata, West Bengal, India
| | - Dharmarajan Sriram
- Department of Pharmacy, Birla Institute of Technology & Science Pilani, Hyderabad, India
| | - Suresh K Muthuvel
- Department of Bioinformatics, Pondicherry University, Kalapet, Puducherry, India
| | | |
Collapse
|
43
|
Kariofillis SK, Jiang S, Żurański AM, Gandhi SS, Martinez Alvarado JI, Doyle AG. Using Data Science To Guide Aryl Bromide Substrate Scope Analysis in a Ni/Photoredox-Catalyzed Cross-Coupling with Acetals as Alcohol-Derived Radical Sources. J Am Chem Soc 2022; 144:1045-1055. [PMID: 34985904 PMCID: PMC8810294 DOI: 10.1021/jacs.1c12203] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ni/photoredox catalysis has emerged as a powerful platform for C(sp2)-C(sp3) bond formation. While many of these methods typically employ aryl bromides as the C(sp2) coupling partner, a variety of aliphatic radical sources have been investigated. In principle, these reactions enable access to the same product scaffolds, but it can be hard to discern which method to employ because nonstandardized sets of aryl bromides are used in scope evaluation. Herein, we report a Ni/photoredox-catalyzed (deutero)methylation and alkylation of aryl halides where benzaldehyde di(alkyl) acetals serve as alcohol-derived radical sources. Reaction development, mechanistic studies, and late-stage derivatization of a biologically relevant aryl chloride, fenofibrate, are presented. Then, we describe the integration of data science techniques, including DFT featurization, dimensionality reduction, and hierarchical clustering, to delineate a diverse and succinct collection of aryl bromides that is representative of the chemical space of the substrate class. By superimposing scope examples from published Ni/photoredox methods on this same chemical space, we identify areas of sparse coverage and high versus low average yields, enabling comparisons between prior art and this new method. Additionally, we demonstrate that the systematically selected scope of aryl bromides can be used to quantify population-wide reactivity trends and reveal sources of possible functional group incompatibility with supervised machine learning.
Collapse
Affiliation(s)
- Stavros K. Kariofillis
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
- Department of Chemistry & Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Shutian Jiang
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| | - Andrzej M. Żurański
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| | - Shivaani S. Gandhi
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
- Department of Chemistry & Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States
| | | | - Abigail G. Doyle
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
- Department of Chemistry & Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States
| |
Collapse
|
44
|
Azeem Z, Mandal PK. Recent advances in palladium-catalyzed C(sp 3)/C(sp 2)-H bond functionalizations: access to C-branched glycosides. Org Biomol Chem 2022; 20:264-281. [PMID: 34904995 DOI: 10.1039/d1ob02142g] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Over the recent decades, tremendous interest has developed in the transformation of complex substrates by C-H activation and functionalization. In particular, palladium-catalyzed directing and non-directing group-assisted C-H functionalization has emerged as a powerful avenue to access C-branched glycosides. Due to the extreme complexity, delicate functionalities, and high stability of C-H bonds, site-selective functionalization of carbohydrate under mild conditions is highly desirable. The purpose of this review is to cover most of the recent advances in palladium-catalyzed C(sp3) and C(sp2)-H bond functionalizations for the synthesis of C-branched glycosides along with future directions.
Collapse
Affiliation(s)
- Zanjila Azeem
- Medicinal & Process Chemistry Division, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram extn, Sitapur Road, P.O. Box 173, Lucknow 226031, India. .,Academy of Scientific and Innovative Research, Ghaziabad-201002, India
| | - Pintu Kumar Mandal
- Medicinal & Process Chemistry Division, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram extn, Sitapur Road, P.O. Box 173, Lucknow 226031, India. .,Academy of Scientific and Innovative Research, Ghaziabad-201002, India
| |
Collapse
|
45
|
Feng B, Zhang G, Feng X, Chen Y. Palladium-catalyzed decarbonylative methylation of aryl carboxylic acids. Org Chem Front 2022. [DOI: 10.1039/d1qo01756j] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Described herein is a palladium-catalyzed decarbonylative methylation of general carboxylic acids, providing an efficient method for Ar–Me bond formation.
Collapse
Affiliation(s)
- Boya Feng
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, The Jiangsu Provincial Platform for Conservation and Utilization of Agricultural Germplasm, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing, 210014, China
| | - Guodong Zhang
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, The Jiangsu Provincial Platform for Conservation and Utilization of Agricultural Germplasm, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing, 210014, China
| | - Xu Feng
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, The Jiangsu Provincial Platform for Conservation and Utilization of Agricultural Germplasm, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing, 210014, China
| | - Yu Chen
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, The Jiangsu Provincial Platform for Conservation and Utilization of Agricultural Germplasm, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing, 210014, China
- Jiangsu Province Engineering Research Center of Eco-cultivation and High-value Utilization of Chinese Medicinal Materials, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing, 210014, China
| |
Collapse
|
46
|
Yang X, Wang G, Ye ZS. Palladium-catalyzed nucleomethylation of alkynes for synthesis of methylated heteroaromatic compounds. Chem Sci 2022; 13:10095-10102. [PMID: 36128232 PMCID: PMC9430495 DOI: 10.1039/d2sc03294e] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022] Open
Abstract
Herein, we disclosed a novel and efficient palladium-catalyzed nucleomethylation of alkynes for the simultaneous construction of the heteroaromatic ring and methyl group. The 3-methylindoles, 3-methylbenzofurans and 4-methylisoquinolines were obtained in moderate to excellent yields. Notably, this methodology was employed as a key step for synthesis of a pregnane X receptor antagonist, zindoxifene, bazedoxifene and AFN-1252. The kinetic studies revealed that reductive elimination might be the rate-determining step. A novel palladium-catalyzed nucleomethylation of alkynes is developed, affording 3-methylindoles, 3-methylbenzofurans and 4-methylisoquinolines in moderate to excellent yields.![]()
Collapse
Affiliation(s)
- Xi Yang
- Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian 116024, P. R. China
| | - Gang Wang
- Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian 116024, P. R. China
| | - Zhi-Shi Ye
- Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian 116024, P. R. China
| |
Collapse
|
47
|
Liu K, Song YF, Gao Y, Luo JQ, Jia YX. NiH-catalyzed dearomative hydroalkylation of indoles. Chem Commun (Camb) 2022; 58:5893-5896. [DOI: 10.1039/d2cc01650h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A NiH-catalyzed intermolecular dearomative hydroalkylation of indoles has been developed, which assembles an array of structurally diverse C2-alkylated indolines with moderate to excellent yields. This protocol exhibits good functional group...
Collapse
|
48
|
Ding C, Ren Y, Sun C, Long J, Yin G. Regio- and Stereoselective Alkylboration of Endocyclic Olefins Enabled by Nickel Catalysis. J Am Chem Soc 2021; 143:20027-20034. [PMID: 34734714 DOI: 10.1021/jacs.1c09214] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Whereas there is a significant interest in the rapid construction of diversely substituted saturated heterocycles, direct and modular access is currently limited to the mono-, 2,3-, or 3,4-substitution pattern. This Communication describes the straightforward and modular construction of 2,4-substituted saturated heterocycles from readily available materials in a highly stereo- and regioselective manner, which sets the stage for numerous readily accessible drug motifs. The strategy relies on chain walking catalysis.
Collapse
Affiliation(s)
- Chao Ding
- The Institute for Advanced Studies, Wuhan University, Wuhan, Hubei 430072, P. R. China
| | - Yaoyu Ren
- The Institute for Advanced Studies, Wuhan University, Wuhan, Hubei 430072, P. R. China
| | - Caocao Sun
- The Institute for Advanced Studies, Wuhan University, Wuhan, Hubei 430072, P. R. China
| | - Jiao Long
- The Institute for Advanced Studies, Wuhan University, Wuhan, Hubei 430072, P. R. China
| | - Guoyin Yin
- The Institute for Advanced Studies, Wuhan University, Wuhan, Hubei 430072, P. R. China
| |
Collapse
|
49
|
Jin T, Xu L, Wang P, Hu X, Zhang R, Wu Z, Du W, Kan W, Li K, Wang C, Zhou Y, Li J, Liu T. Discovery and Development of a Potent, Selective, and Orally Bioavailable CHK1 Inhibitor Candidate: 5-((4-((3-Amino-3-methylbutyl)amino)-5-(trifluoromethyl)pyrimidin-2-yl)amino)picolinonitrile. J Med Chem 2021; 64:15069-15090. [PMID: 34665631 DOI: 10.1021/acs.jmedchem.1c00994] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Checkpoint kinase 1 (CHK1) plays an important role in the DNA damage response pathway, being a potential anti-cancer drug target. In this study, we used a strategy for trifluoromethyl substitution to obtain orally bioavailable CHK1 inhibitors to overcome the limitations of lead compound 1, which can only be administered intravenously. After detailed investigation, we identified compound 6c as an oral CHK1 inhibitor, which demonstrated a considerably higher plasma exposure in mice. Compound 6c also showed good kinase selectivity. Moreover, it exhibited a significant antiproliferative effect in MV-4-11 cells singly and a synergistic effect in combination with gemcitabine in HT-29, A549, and RPMI-8226 cells. Additionally, compound 6c could inhibit tumor growth in the MV-4-11 xenograft mouse model. The combination of 6c and gemcitabine exhibited synergistic effect in the HT-29 xenograft mouse model. Thus, compound 6c was found to be a selective and oral potential anticancer CHK1 inhibitor.
Collapse
Affiliation(s)
- Tingting Jin
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Lei Xu
- State key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Zhongshan, Guangdong 528400, China
| | - Peipei Wang
- State key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiaobei Hu
- State key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Zhongshan, Guangdong 528400, China
| | - Runyuan Zhang
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Zhiqi Wu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Zhongshan, Guangdong 528400, China
| | - Wenxin Du
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Weijuan Kan
- State key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Kun Li
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Chang Wang
- State key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yubo Zhou
- State key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Zhongshan, Guangdong 528400, China
| | - Jia Li
- State key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Zhongshan, Guangdong 528400, China
| | - Tao Liu
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| |
Collapse
|
50
|
Jana R, Begam HM, Dinda E. The emergence of the C-H functionalization strategy in medicinal chemistry and drug discovery. Chem Commun (Camb) 2021; 57:10842-10866. [PMID: 34596175 DOI: 10.1039/d1cc04083a] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Owing to the market competitiveness and urgent societal need, an optimum speed of drug discovery is an important criterion for successful implementation. Despite the rapid ascent of artificial intelligence and computational and bioanalytical techniques to accelerate drug discovery in big pharma, organic synthesis of privileged scaffolds predicted in silico for in vitro and in vivo studies is still considered as the rate-limiting step. C-H activation is the latest technology added into an organic chemist's toolbox for the rapid construction and late-stage modification of functional molecules to achieve the desired chemical and physical properties. Particularly, elimination of prefunctionalization steps, exceptional functional group tolerance, complexity-to-diversity oriented synthesis, and late-stage functionalization of privileged medicinal scaffolds expand the chemical space. It has immense potential for the rapid synthesis of a library of molecules, structural modification to achieve the required pharmacological properties such as absorption, distribution, metabolism, excretion, toxicology (ADMET) and attachment of chemical reporters for proteome profiling, metabolite synthesis, etc. for preclinical studies. Although heterocycle synthesis, late-stage drug modification, 18F labelling, methylation, etc. via C-H functionalization have been reviewed from the synthetic standpoint, a general overview of these protocols from medicinal and drug discovery aspects has not been reviewed. In this feature article, we will discuss the recent trends of C-H activation methodologies such as synthesis of medicinal scaffolds through C-H activation/annulation cascade; C-H arylation for sp2-sp2 and sp2-sp3 cross-coupling; C-H borylation/silylation to introduce a functional linchpin for further manipulation; C-H amination for N-heterocycles and hydrogen bond acceptors; C-H fluorination/fluoroalkylation to tune polarity and lipophilicity; C-H methylation: methyl magic in drug discovery; peptide modification and macrocyclization for therapeutics and biologics; fluorescent labelling and radiolabelling for bioimaging; bioconjugation for chemical biology studies; drug-metabolite synthesis for biodistribution and excretion studies; late-stage diversification of drug-molecules to increase efficacy and safety; cutting-edge DNA encoded library synthesis and improved synthesis of drug molecules via C-H activation in medicinal chemistry and drug discovery.
Collapse
Affiliation(s)
- Ranjan Jana
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata-700032, India.
| | - Hasina Mamataj Begam
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata-700032, India.
| | - Enakshi Dinda
- Department of Chemistry and Environment, Heritage Institute of Technology, Kolkata-700107, India
| |
Collapse
|