1
|
Stevens R, Shrives HJ, Cryan J, Klimaszewska D, Stacey P, Burley GA, Harling JD, Battersby DJ, Miah AH. Expanding the reaction toolbox for nanoscale direct-to-biology PROTAC synthesis and biological evaluation. RSC Med Chem 2024:d4md00760c. [PMID: 39720740 PMCID: PMC11664481 DOI: 10.1039/d4md00760c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/13/2024] [Indexed: 12/26/2024] Open
Abstract
High-throughput chemistry (HTC) and direct-to-biology (D2B) platforms allow for plate-based compound synthesis and biological evaluation of crude mixtures in cellular assays. The rise of these workflows has rapidly accelerated drug-discovery programs in the field of targeted protein degradation (TPD) in recent years by removing a key bottleneck of compound purification. However, the number of chemical transformations amenable to this methodology remain minimal, leading to limitations in the exploration of chemical space using existing library-based approaches. In this work, we expanded the toolbox by synthesising a library of degraders in D2B format. First, reaction conditions are established for performing key medicinal chemistry transformations, including reductive amination, SNAr, palladium-mediated cross-coupling and alkylation, in D2B format. Second, the utility of these alternative reactions is demonstrated by rapidly identifying developable PROTACs for a range of protein targets.
Collapse
Affiliation(s)
- Rebecca Stevens
- Modality Platform Technologies, GSK Stevenage SG1 2NY UK
- Department of Pure and Applied Chemistry, University of Strathclyde Glasgow G1 1BX UK
| | | | - Jenni Cryan
- Discovery Biology and Screening, GSK Stevenage SG1 2NY UK
| | | | - Peter Stacey
- Discovery Biology and Screening, GSK Stevenage SG1 2NY UK
| | - Glenn A Burley
- Department of Pure and Applied Chemistry, University of Strathclyde Glasgow G1 1BX UK
| | - John D Harling
- Modality Platform Technologies, GSK Stevenage SG1 2NY UK
| | | | - Afjal H Miah
- Modality Platform Technologies, GSK Stevenage SG1 2NY UK
| |
Collapse
|
2
|
Welsh A, Husbands D, Frei A. High-Throughput Combinatorial Metal Complex Synthesis. Angew Chem Int Ed Engl 2024:e202420204. [PMID: 39714355 DOI: 10.1002/anie.202420204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 12/24/2024]
Abstract
High-throughput combinatorial metal complex synthesis has emerged as a powerful tool for rapidly generating and screening diverse libraries of metal complexes, enabling accelerated discovery in fields such as catalysis, medicinal chemistry, and materials science. By systematically combining building blocks under mild and efficient conditions, researchers can explore broad chemical spaces, increasing the likelihood of identifying complexes with desired properties. This method streamlines hit identification and optimisation, especially when integrated with high-throughput screening and data-driven approaches like machine learning. Despite challenges such as scalability and purity control, recent advancements in automation and predictive modelling are enhancing the efficiency of combinatorial synthesis, opening new avenues for the development of metal-based catalysts, therapeutic agents, and functional materials.
Collapse
Affiliation(s)
- A Welsh
- Department of Chemistry, Biochemistry & Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012, Bern, Switzerland
- Department of Chemistry, University of York, York, YO10 5DD, U.K
| | - D Husbands
- Department of Chemistry, University of York, York, YO10 5DD, U.K
| | - A Frei
- Department of Chemistry, Biochemistry & Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012, Bern, Switzerland
- Department of Chemistry, University of York, York, YO10 5DD, U.K
| |
Collapse
|
3
|
Bogyo M, Upadhyay T, Woods E, Ahator S, Julin K, Faucher F, Hollander M, Pedowitz N, Abegg D, Hammond I, Eke I, Wang S, Chen S, Bennett J, Jo J, Lentz C, Adibekian A, Fellner M. Covalent-fragment screening identifies selective inhibitors of multiple Staphylococcus aureus serine hydrolases important for growth and biofilm formation. RESEARCH SQUARE 2024:rs.3.rs-5494070. [PMID: 39711551 PMCID: PMC11661381 DOI: 10.21203/rs.3.rs-5494070/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Staphylococcus aureus is a leading cause of bacteria-associated mortality worldwide. This is largely because infection sites are often difficult to localize and the bacteria forms biofilms which are not effectively cleared using classical antibiotics. Therefore, there is a need for new tools to both image and treat S. aureus infections. We previously identified a group of S. aureus serine hydrolases known as fluorophosphonate-binding hydrolases (Fphs), which regulate aspects of virulence and lipid metabolism. However, because their structures are similar and their functions overlap, it remains challenging to distinguish the specific roles of individual members of this family. In this study, we applied a high-throughput screening approach using a library of covalent electrophiles to identify inhibitors for FphB, FphE, and FphH. We identified inhibitors that irreversibly bind to the active-site serine residue of each enzyme with high potency and selectivity without requiring extensive medicinal chemistry optimization. Structural and biochemical analysis identified novel binding modes for several of the inhibitors. Selective inhibitors of FphH impaired both bacterial growth and biofilm formation while Inhibitors of FphB and FphE had no impact on cell growth and only limited impact on biofilm formation. These results suggest that all three hydrolases likely play functional, but non-equivalent roles in biofilm formation and FphH is a potential target for development of therapeutics that have both antibiotic and anti-biofilm activity. Overall, we demonstrate that focused covalent fragment screening can be used to rapidly identify highly potent and selective electrophiles targeting bacterial serine hydrolases. This approach could be applied to other classes of lipid hydrolases in diverse pathogens or higher eukaryotes.
Collapse
|
4
|
Tyagarajan S, Andrews CL, Beshore DC, Buevich AV, Curran PJ, Dandliker P, Greshock TJ, Hoar J, Kim A, Karnachi P, Knemeyer I, Kozlowski J, Liu J, Maletic M, Myers R, Rada V, Sha D, Sauvagnat B, Vachal P, Wolkenberg S, Yu W, Yu Y, Krska SW. Rapid Affinity and Microsomal Stability Ranking of Crude Mixture Libraries of Histone Deacetylase Inhibitors. ACS Med Chem Lett 2024; 15:1787-1794. [PMID: 39411537 PMCID: PMC11472384 DOI: 10.1021/acsmedchemlett.4c00345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 10/19/2024] Open
Abstract
The science of drug discovery involves multiparameter optimization of molecular structures through iterative design-make-test cycles. For medicinal chemistry library synthesis, traditional workflows involve the isolation of each individual compound, gravimetric quantitation, and preparation of a standard concentration solution for biological assays. In this work, we explore ways to expedite this process by testing unpurified library mixtures using a combination of mass spectrometry-based assays for affinity selection and microsomal metabolic stability. Utilizing this approach, microgram quantities of crude library mixtures can be used to identify high affinity, metabolically stable library members for isolation and full characterization. This streamlined approach was demonstrated for the synthesis and evaluation of two libraries of histone deacetylase inhibitors and was shown to generate decision-making data in line with traditional workflows. The advantages of this paradigm include greatly reduced cycle time, reduced material requirements, and concentration of resources on the most promising compounds.
Collapse
Affiliation(s)
- Sriram Tyagarajan
- Discovery
Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Christine L. Andrews
- Quantitative
Biosciences, Merck & Co., Inc., Boston, Massachusetts 02115, United States
| | - Douglas C. Beshore
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Alexei V. Buevich
- Analytical
Research & Development, Merck &
Co., Inc., Rahway, New Jersey 07065, United States
| | - Patrick J. Curran
- Quantitative
Biosciences, Merck & Co., Inc., Boston, Massachusetts 02115, United States
| | - Peter Dandliker
- Quantitative
Biosciences, Merck & Co., Inc., Boston, Massachusetts 02115, United States
| | - Thomas J. Greshock
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Jason Hoar
- Pharmacokinetics,
Pharmacodynamics and Drug Metabolism, Merck
& Co., Inc., Rahway, New Jersey 07065, United States
| | - Alex Kim
- Discovery
Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Prabha Karnachi
- Modeling
and Informatics, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Ian Knemeyer
- Pharmacokinetics,
Pharmacodynamics and Drug Metabolism, Merck
& Co., Inc., Boston, Massachusetts 02115, United States
| | - Joseph Kozlowski
- Discovery
Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Jian Liu
- Discovery
Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Milana Maletic
- Discovery
Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Robert Myers
- Department
of Pharmacology, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Vanessa Rada
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Deyou Sha
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Berengere Sauvagnat
- Quantitative
Biosciences, Merck & Co., Inc., Boston, Massachusetts 02115, United States
| | - Petr Vachal
- Discovery
Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Scott Wolkenberg
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Wensheng Yu
- Discovery
Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Younong Yu
- Discovery
Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Shane W. Krska
- Discovery
Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| |
Collapse
|
5
|
Wang SC, Zhou X, Li YX, Zhang CY, Zhang ZY, Xiong YS, Lu G, Dong J, Weng J. Enabling Modular Click Chemistry Library through Sequential Ligations of Carboxylic Acids and Amines. Angew Chem Int Ed Engl 2024; 63:e202410699. [PMID: 38943043 DOI: 10.1002/anie.202410699] [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: 06/06/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 06/30/2024]
Abstract
High-throughput synthesis and screening of chemical libraries play pivotal roles in drug discovery. Click chemistry has emerged as a powerful strategy for constructing highly modular chemical libraries. However, the development of new click reactions and unlocking new clickable building blocks remain exceedingly challenging. Herein, we describe a double-click strategy that enables the sequential ligations of widely available carboxylic acids and amines with fluorosulfuryl isocyanate (FSO2NCO) via a modular amidation/SuFEx (sulfur-fluoride exchange) process. This method provides facile access to chemical libraries of N-fluorosulfonyl amides (RCONHSO2F) and N-acylsulfamides (RCONHSO2NR'R'') in near-quantitative yields under simple and practical conditions. The robustness and efficiency of this double click strategy is showcased by the facile construction of chemical libraries in 96-well microtiter plates from a large number of carboxylic acids and amines. Preliminary biological activity screening reveals that some compounds exhibit high antimicrobial activities against Gram-positive bacterium S. aureus and drug-resistant MRSA (MIC up to 6.25 μg ⋅ mL-1). These results provide compelling evidence for the potential application of modular click chemistry library as an enabling technology in high-throughput medicinal chemistry.
Collapse
Affiliation(s)
- Sheng-Cai Wang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006, Guangzhou, China
| | - Xiang Zhou
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006, Guangzhou, China
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, P. R. China
| | - Ying-Xian Li
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006, Guangzhou, China
| | - Chun-Yan Zhang
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, P. R. China
| | - Zi-Yan Zhang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006, Guangzhou, China
| | - Yan-Shi Xiong
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, P. R. China
| | - Gui Lu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006, Guangzhou, China
| | - Jiajia Dong
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Jiang Weng
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006, Guangzhou, China
| |
Collapse
|
6
|
Li H, Peng M, Li J, Wang L, Do H, Ni K, Wang M, Yuan Z, Zhao T, Zhang X, Zhang X, Hu Z, Ren F, An J. SO 2F 2 mediated click chemistry enables modular disulfide formation in diverse reaction media. Nat Commun 2024; 15:8325. [PMID: 39333088 PMCID: PMC11436863 DOI: 10.1038/s41467-024-52606-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 09/12/2024] [Indexed: 09/29/2024] Open
Abstract
The dynamic disulfide linkage plays a vital role in various biological processes as well as drugs and biomaterials. The conversion of thiols to their corresponding disulfides is a hallmark of sulfur chemistry, but notoriously difficult to control. Achieving optimal reactivity and selectivity continues to pose significant challenges. Here, we describe a click chemistry for disulfide formation from thiols in both batch and flow-mode using SO2F2, which display exceptional selectivity toward disulfide formation through an effective nucleophilic substitution cascade. This reaction's unique characteristics satisfy the stringent click-criteria with its high thermodynamic driving force, straightforward conditions, wide scope, quantitative yields, exceptional chemoselectivity, and non-chromatographic purification process. The modular synthesis of symmetrical, unsymmetrical, cyclic and polydisulfides is demonstrated, along with the formation of disulfide cross-linked hydrogels.
Collapse
Affiliation(s)
- Hengzhao Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
- College of Plant Protection, Northwest A&F University, Yangling, 712100, China
- Food Laboratory of Zhongyuan, Luohe, 462000, China
| | - Mengqi Peng
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Junyu Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Lijun Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Hainam Do
- Department of Chemical and Environmental Engineering, University of Nottingham Ningbo China, Ningbo, 315100, China
| | - Ke Ni
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Minlong Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Zhankui Yuan
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Tianxiao Zhao
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Xiaohe Zhang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Xiaoxu Zhang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Zhaonong Hu
- College of Plant Protection, Northwest A&F University, Yangling, 712100, China
| | - Fazheng Ren
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Jie An
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China.
| |
Collapse
|
7
|
Zhao XZ, Barakat IA, Lountos GT, Wang W, Agama K, Mahmud MRA, Suazo KF, Andresson T, Pommier Y, Burke TR. Targeted sulfur(VI) fluoride exchange-mediated covalent modification of a tyrosine residue in the catalytic pocket of tyrosyl-DNA phosphodiesterase 1. Commun Chem 2024; 7:208. [PMID: 39284936 PMCID: PMC11405833 DOI: 10.1038/s42004-024-01298-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024] Open
Abstract
Developing effective inhibitors of the DNA repair enzyme tyrosyl-DNA phosphodiesterase 1 (TDP1) has been challenging because of the enzyme shallow catalytic pocket and non-specific substrate binding interactions. Recently, we discovered a quinolone-binding hot spot in TDP1's active site proximal to the evolutionary conserved Y204 and F259 residues that position DNA. Sulfur (VI) fluoride exchange (SuFEx) is a biocompatible click chemistry reaction that enables acylation of protein residues, including tyrosine. Selective protein modifications can provide insights into the biological roles of proteins and inform ligand design. As we report herein, we used SuFEx chemistries to prepare covalent TDP1-bound binders showing site-specific covalent bonds with Y204. Our work presents the first application of SuFEx chemistries to TDP1 ligands. It validates the ability to covalently modify specific TDP1 residues by designed targeting and adds to the chemical biology resource toolbox for studying TDP1.
Collapse
Affiliation(s)
- Xue Zhi Zhao
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA.
| | - Idris A Barakat
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - George T Lountos
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Wenjie Wang
- Developmental Therapeutics Branch & Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Keli Agama
- Developmental Therapeutics Branch & Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Md Rasel Al Mahmud
- Developmental Therapeutics Branch & Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kiall F Suazo
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
- Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Thorkell Andresson
- Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Yves Pommier
- Developmental Therapeutics Branch & Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Terrence R Burke
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| |
Collapse
|
8
|
Schnaider L, Tan S, Singh PR, Capuano F, Scott AJ, Hambley R, Lu L, Yang H, Wallace EJ, Jo H, DeGrado WF. SuFEx Chemistry Enables Covalent Assembly of a 280-kDa 18-Subunit Pore-Forming Complex. J Am Chem Soc 2024; 146:25047-25057. [PMID: 39190920 DOI: 10.1021/jacs.4c07920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Proximity-enhanced chemical cross-linking is an invaluable tool for probing protein-protein interactions and enhancing the potency of potential peptide and protein drugs. Here, we extend this approach to covalently stabilize large macromolecular assemblies. We used SuFEx chemistry to covalently stabilize an 18-subunit pore-forming complex, CsgG:CsgF, consisting of nine CsgG membrane protein subunits that noncovalently associate with nine CsgF peptides. Derivatives of the CsgG:CsgF pore have been used for DNA sequencing, which places high demands on the structural stability and homogeneity of the complex. To increase the robustness of the pore, we designed and synthesized derivatives of CsgF-bearing sulfonyl fluorides, which react with CsgG in very high yield to form a covalently stabilized CsgG:CsgF complex. The resulting pores formed highly homogeneous channels when added to artificial membranes. The high yield and rapid reaction rate of the SuFEx reaction prompted molecular dynamics simulations, which revealed that the SO2F groups in the initially formed complex are poised for nucleophilic reaction with a targeted Tyr. These results demonstrate the utility of SuFEx chemistry to structurally stabilize very large (here, 280 kDa) assemblies.
Collapse
Affiliation(s)
- Lee Schnaider
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94143, United States
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94143, United States
| | - Sophia Tan
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94143, United States
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94143, United States
| | | | | | | | | | - Lei Lu
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94143, United States
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94143, United States
| | - Hyunjun Yang
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94143, United States
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94143, United States
| | | | - Hyunil Jo
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94143, United States
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94143, United States
| | - William F DeGrado
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94143, United States
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94143, United States
| |
Collapse
|
9
|
Wang P, Lin L, Huang Y, Zhang H, Liao S. Radical Fluorosulfonamidation: A Facile Access to Sulfamoyl Fluorides. Angew Chem Int Ed Engl 2024; 63:e202405944. [PMID: 38837324 DOI: 10.1002/anie.202405944] [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: 03/27/2024] [Revised: 05/30/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024]
Abstract
Recently, the introduction of fluorosulfonyl (-SO2F) groups have attracted considerable research interests, as this moiety could often afford enhanced activities and new functions in the context of chemical biology and drug discovery. Herein, we report the design and synthesis of 1-fluorosulfamoyl-pyridinium (FSAP) salts, which could serve as an effective photoredox-active precursor to fluorosulfamoyl radicals and enable the direct radical C-H fluorosulfonamidation of a variety of (hetero)arenes. This method features mild conditions, visible light, broad substrate scope, good group tolerance, etc., and a metal-free protocol is also viable by using organic photocatalysts. Further, FSAP can also be applied to the radical functionalization of alkenes via 1,2-difunctionalization, radical distal migration, tandem radical-polar crossover reactions, etc. In addition, a formal C-H methylamination of (hetero)arenes by combining this radical C-H fluorosulfonamidation with subsequent hydrolysis as well as product derivatization are also demonstrated.
Collapse
Affiliation(s)
- Peng Wang
- Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou, 350108, China
- Key Laboratory of Green and Precise Synthetic Chemistry and Application, Ministry of Education, College of Chemistry and Materials Science, Huaibei Normal University, Huaibei, Anhui, 235000, China
| | - Lu Lin
- Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou, 350108, China
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Yao Huang
- Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou, 350108, China
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Honghai Zhang
- Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou, 350108, China
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Saihu Liao
- Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou, 350108, China
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| |
Collapse
|
10
|
Jaeger R, Rachor SG, Ahrens M, Braun T. Activation of SO 2F 2 at a Rhodium PNP Pincer Complex: Ligand Supported S-F Bond Cleavage to Generate NSO 2F Derivatives. Chemistry 2024; 30:e202401571. [PMID: 38757784 DOI: 10.1002/chem.202401571] [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: 04/23/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 05/18/2024]
Abstract
The κ2-(P,N)-phosphine ligand precursor NH(CH2CH2PCy2)2 can be used for the synthesis of the rhodium(I) complex [Rh(CO){ĸ3-(P,N,P)-Cy2PC2H4NHC2H4PCy2}][Cl] (1). The deprotonated complex [Rh(CO){ĸ3-(P,N,P)-Cy2PC2H4NC2H4PCy2}] (2) shows a cooperative reactivity of the PNP ligand in the activation reaction of SO2F2 to yield the rhodium fluorido complex trans-[Rh(F)(CO){ĸ2-(P,P)-Cy2PC2H4N(SO2F)C2H4PCy2}]2 (3) by S-F bond cleavage. It is remarkable that no reaction was observed when 3 was treated with hydrogen sources e. g. dihydrogen, organosilicon compounds such as triethylsilane or TMS-CF3 and different fluorine sources such as SF4 or Selectfluor®. However, the treatment of complex 3 with XeF2 in the presence of CsF resulted in the formation of the unique fluorido rhodium(III) complex cis,trans-[Rh(F)3(CO){ĸ2-(P,P)-Cy2PC2H4N(SO2F)C2H4PCy2}]2 (4). In the presence of pyridine(HF)X or BF3 the fluorido complex 3 converted into the dicationic complexes [Rh(CO){ĸ2-(P,P)-Cy2PC2H4N(SO2F)C2H4PCy2}]2[XF]2, X=HF (5) or BF3 (6), respectively.
Collapse
Affiliation(s)
- Ruben Jaeger
- Humboldt-Universität zu Berlin, Department of Chemistry, Brook-Taylor-Straße 2, 12489, Berlin, Germany
| | - Simon G Rachor
- Humboldt-Universität zu Berlin, Department of Chemistry, Brook-Taylor-Straße 2, 12489, Berlin, Germany
| | - Mike Ahrens
- Humboldt-Universität zu Berlin, Department of Chemistry, Brook-Taylor-Straße 2, 12489, Berlin, Germany
| | - Thomas Braun
- Humboldt-Universität zu Berlin, Department of Chemistry, Brook-Taylor-Straße 2, 12489, Berlin, Germany
| |
Collapse
|
11
|
Yan KN, Nie YQ, Wang JY, Yin GL, Liu Q, Hu H, Sun X, Chen XH. Accelerating PROTACs Discovery Through a Direct-to-Biology Platform Enabled by Modular Photoclick Chemistry. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400594. [PMID: 38689503 PMCID: PMC11234393 DOI: 10.1002/advs.202400594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/02/2024] [Indexed: 05/02/2024]
Abstract
Proteolysis targeting chimeras (PROTACs) have emerged as a promising strategy for drug discovery and exploring protein functions, offering a revolutionary therapeutic modality. Currently, the predominant approach to PROTACs discovery mainly relies on an empirical design-synthesis-evaluation process involving numerous cycles of labor-intensive synthesis-purification and bioassay data collection. Therefore, the development of innovative methods to expedite PROTAC synthesis and exploration of chemical space remains highly desired. Here, a direct-to-biology strategy is reported to streamline the synthesis of PROTAC libraries on plates, enabling the seamless transfer of reaction products to cell-based bioassays without the need for additional purification. By integrating amide coupling and light-induced primary amines and o-nitrobenzyl alcohols cyclization (PANAC) photoclick chemistry into a plate-based synthetic process, this strategy produces PROTAC libraries with high efficiency and structural diversity. Moreover, by employing this platform for PROTACs screening, we smoothly found potent PROTACs effectively inhibit triple-negative breast cancer (TNBC) cell growth and induce rapid, selective targeted degradation of cyclin-dependent kinase 9 (CDK9). The study introduces a versatile platform for assembling PROTACs on plates, followed by direct biological evaluation. This approach provides a promising opportunity for high-throughput synthesis of PROTAC libraries, thereby enhancing the efficiency of exploring chemical space and accelerating the discovery of PROTACs.
Collapse
Affiliation(s)
- Ke-Nian Yan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yong-Qiang Nie
- Jiangxi Key Laboratory of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Jia-Yu Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Guang-Liang Yin
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qia Liu
- University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Hao Hu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiaoxia Sun
- Jiangxi Key Laboratory of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Xiao-Hua Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| |
Collapse
|
12
|
Zogu A, Ullah K, Spanopoulos S, Ismalaj E, De Borggraeve WM, Demaerel J. Perfluorooxosulfate Salts as SOF 4-Gas-Free Precursors to Multidimensional SuFEx Electrophiles. Angew Chem Int Ed Engl 2024; 63:e202403797. [PMID: 38630865 DOI: 10.1002/anie.202403797] [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: 02/23/2024] [Revised: 04/03/2024] [Accepted: 04/11/2024] [Indexed: 04/19/2024]
Abstract
Sulfur(VI) Fluoride Exchange (SuFEx) chemistry stands as a well-established method for swiftly constructing complex molecules in a modular fashion. An especially promising segment of this toolbox is reserved for multidimensional SuFEx hubs: three or more substituents pluggable into a singular SVI centre to make 'beyond-linear' clicked constructions. Sulfurimidoyl difluorides (RNSOF2) stand out as the prime example of this, however their preparation from the scarcely available thionyl tetrafluoride (SOF4) limits this chemistry to only a few laboratories with access to this gas. In this work, we identify silver pentafluorooxosulfate (AgOSF5) as a viable SuFEx hub with reactivity equal to SOF4. The AgF2-mediated oxidation of SOCl2 gives rise to the hexacoordinate AgOSF5 adduct, which in contact with primary amines produces the sulfurimidoyl fluorides in high yields. In addition, we have found this workflow to be fully extendable to the trifluoromethyl homologue, AgOSF4CF3, and we propose the use of AgOSF4X salts as a general route to azasulfur SuFEx electrophiles from commercial starting materials.
Collapse
Affiliation(s)
- Armir Zogu
- Department of Chemistry, Sustainable Chemistry for Metals and Molecules (SCM2), KU Leuven Department of Chemistry, Celestijnenlaan 200F-box 2404, B-3001, Leuven, Belgium
| | - Karim Ullah
- Department of Chemistry, Sustainable Chemistry for Metals and Molecules (SCM2), KU Leuven Department of Chemistry, Celestijnenlaan 200F-box 2404, B-3001, Leuven, Belgium
- Department of Chemistry and Technologies of Drug, Sapienza, University of Rome, P.le A. Moro 5, 00185, Rome, Italy
| | - Stefanos Spanopoulos
- Department of Chemistry, Sustainable Chemistry for Metals and Molecules (SCM2), KU Leuven Department of Chemistry, Celestijnenlaan 200F-box 2404, B-3001, Leuven, Belgium
| | - Ermal Ismalaj
- Department of Chemistry, Sustainable Chemistry for Metals and Molecules (SCM2), KU Leuven Department of Chemistry, Celestijnenlaan 200F-box 2404, B-3001, Leuven, Belgium
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramon, 20014, San Sebastian, Guipuzcoa, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), 28029, Madrid, Spain
| | - Wim M De Borggraeve
- Department of Chemistry, Sustainable Chemistry for Metals and Molecules (SCM2), KU Leuven Department of Chemistry, Celestijnenlaan 200F-box 2404, B-3001, Leuven, Belgium
| | - Joachim Demaerel
- Department of Chemistry, Sustainable Chemistry for Metals and Molecules (SCM2), KU Leuven Department of Chemistry, Celestijnenlaan 200F-box 2404, B-3001, Leuven, Belgium
| |
Collapse
|
13
|
Kitamura S, Lin TH, Lee CCD, Takamura A, Kadam RU, Zhang D, Zhu X, Dada L, Nagai E, Yu W, Yao Y, Sharpless KB, Wilson IA, Wolan DW. Ultrapotent influenza hemagglutinin fusion inhibitors developed through SuFEx-enabled high-throughput medicinal chemistry. Proc Natl Acad Sci U S A 2024; 121:e2310677121. [PMID: 38753503 PMCID: PMC11145270 DOI: 10.1073/pnas.2310677121] [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: 06/24/2023] [Accepted: 04/19/2024] [Indexed: 05/18/2024] Open
Abstract
Seasonal and pandemic-associated influenza strains cause highly contagious viral respiratory infections that can lead to severe illness and excess mortality. Here, we report on the optimization of our small-molecule inhibitor F0045(S) targeting the influenza hemagglutinin (HA) stem with our Sulfur-Fluoride Exchange (SuFEx) click chemistry-based high-throughput medicinal chemistry (HTMC) strategy. A combination of SuFEx- and amide-based lead molecule diversification and structure-guided design led to identification and validation of ultrapotent influenza fusion inhibitors with subnanomolar EC50 cellular antiviral activity against several influenza A group 1 strains. X-ray structures of six of these compounds with HA indicate that the appended moieties occupy additional pockets on the HA surface and increase the binding interaction, where the accumulation of several polar interactions also contributes to the improved affinity. The compounds here represent the most potent HA small-molecule inhibitors to date. Our divergent HTMC platform is therefore a powerful, rapid, and cost-effective approach to develop bioactive chemical probes and drug-like candidates against viral targets.
Collapse
Affiliation(s)
- Seiya Kitamura
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA92037
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA92037
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY10461
| | - Ting-Hui Lin
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Chang-Chun David Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Akihiro Takamura
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA92037
| | - Rameshwar U. Kadam
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Ding Zhang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Xueyong Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Lucas Dada
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY10461
| | - Emiko Nagai
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY10461
| | - Wenli Yu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Yao Yao
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA92037
| | - K. Barry Sharpless
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
| | - Ian A. Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA92037
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Dennis W. Wolan
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA92037
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA92037
| |
Collapse
|
14
|
Hansen T, Danková D, Bæk M, Grlaš L, Olsen CA. Sulfur(VI) Fluoride Exchange Chemistry in Solid-Phase Synthesis of Compound Arrays: Discovery of Histone Deacetylase Inhibitors. JACS AU 2024; 4:1854-1862. [PMID: 38818074 PMCID: PMC11134391 DOI: 10.1021/jacsau.4c00042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 06/01/2024]
Abstract
Multistep synthesis performed on solid support is a powerful means to generate small-molecule libraries for the discovery of chemical probes to dissect biological mechanisms as well as for drug discovery. Therefore, expansion of the collection of robust chemical transformations amenable to solid-phase synthesis is desirable for achieving chemically diverse libraries for biological testing. Here, we show that sulfur(VI) fluoride exchange (SuFEx) chemistry, exemplified by pairing phenols with aryl fluorosulfates, can be used for the solid-phase synthesis of biologically active compounds. As a case study, we designed and synthesized a library of 84 hydroxamic acid-containing small molecules, providing a rich source of inhibitors with diverse selectivity profiles across the human histone deacetylase enzyme family. Among other discoveries, we identified a scaffold that furnished inhibitors of HDAC11 with exquisite selectivity in vitro and a selective inhibitor of HDAC6 that was shown to affect the acetylation of α-tubulin over histone sites H3K18, H3K27, as well as SMC3 in cultured cells. Our results encourage the further use of SuFEx chemistry for the synthesis of diverse small-molecule libraries and provide insight for future design of selective HDAC inhibitors.
Collapse
Affiliation(s)
| | | | | | - Linda Grlaš
- Center for Biopharmaceuticals
and Department of Drug Design and Pharmacology, Faculty of Health
and Medical Sciences, University of Copenhagen, Jagtvej 160, DK-2100 Copenhagen, Denmark
| | - Christian A. Olsen
- Center for Biopharmaceuticals
and Department of Drug Design and Pharmacology, Faculty of Health
and Medical Sciences, University of Copenhagen, Jagtvej 160, DK-2100 Copenhagen, Denmark
| |
Collapse
|
15
|
Stevens R, Thompson JDF, Fournier JCL, Burley GA, Battersby DJ, Miah AH. Innovative, combinatorial and high-throughput approaches to degrader synthesis. Chem Soc Rev 2024; 53:4838-4861. [PMID: 38596888 DOI: 10.1039/d3cs01127e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Targeted protein degraders such as PROTACs and molecular glues are a rapidly emerging therapeutic modality within industry and academia. Degraders possess unique mechanisms of action that lead to the removal of specific proteins by co-opting the cell's natural degradation mechanisms via induced proximity. Their optimisation thus far has often been largely empirical, requiring the synthesis and screening of a large number of analogues. In addition, the synthesis and development of degraders is often challenging, leading to lengthy optimisation campaigns to deliver candidate-quality compounds. This review highlights how the synthesis of degraders has evolved in recent years, in particular focusing on means of applying high-throughput chemistry and screening approaches to expedite these timelines, which we anticipate to be valuable in shaping the future of degrader optimisation campaigns.
Collapse
Affiliation(s)
- Rebecca Stevens
- Medicinal Chemistry, GSK, Stevenage, SG1 2NY, UK.
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, G1 1XQ, UK
| | | | | | - Glenn A Burley
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, G1 1XQ, UK
| | | | - Afjal H Miah
- Medicinal Chemistry, GSK, Stevenage, SG1 2NY, UK.
| |
Collapse
|
16
|
Gutiérrez-González A, Karlsson S, Leonori D, Plesniak MP. Mild Strategy for the Preparation of Alkyl Sulfonyl Fluorides from Alkyl Bromides and Alcohols Using Photoredox Catalysis and Flow Chemistry. Org Lett 2024; 26:3972-3976. [PMID: 38663015 DOI: 10.1021/acs.orglett.4c01216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
Facile access to sp3-rich scaffolds containing a sulfonyl fluoride group is still limited. Herein, we describe a mild and scalable strategy for the preparation of alkyl sulfonyl fluorides from readily available alkyl bromides and alcohols using photoredox catalysis. This approach is based on halogen atom transfer (XAT), followed by SO2 capture and fluorination. The method features mild conditions enabling fast access to high-value derivatives and has been scaled up to 5 g using a continuous stirred tank reactor cascade.
Collapse
Affiliation(s)
- Alejandro Gutiérrez-González
- Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca Gothenburg, 431 83 Mölndal, Sweden
| | - Staffan Karlsson
- Early Chemical Development, Pharmaceutical Sciences, Biopharmaceuticals R&D, AstraZeneca Gothenburg, 431 83 Mölndal, Sweden
| | - Daniele Leonori
- Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52056 Aachen, Germany
| | - Mateusz P Plesniak
- Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca Gothenburg, 431 83 Mölndal, Sweden
| |
Collapse
|
17
|
Carter TR, Milosevich N, Dada L, Shaum JB, Barry Sharpless K, Kitamura S, Erb MA. SuFEx-based chemical diversification for the systematic discovery of CRBN molecular glues. Bioorg Med Chem 2024; 104:117699. [PMID: 38608634 PMCID: PMC11195152 DOI: 10.1016/j.bmc.2024.117699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/18/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024]
Abstract
Molecular glues are small molecules that stabilize protein-protein interactions, enabling new molecular pharmacologies, such as targeted protein degradation. They offer advantages over proteolysis targeting chimeras (PROTACs), which present challenges associated with the size and properties of heterobifunctional constructions, but glues lack the rational design principles analogous to PROTACs. One notable exception is the ability to alter the structure of Cereblon (CRBN)-based molecular glues and redirect their activity toward new neo-substrate proteins. We took a focused approach toward modifying the CRBN ligand, 5'-amino lenalidomide, to alter its neo-substrate specificity using high-throughput chemical diversification by parallelized sulfur(VI)-fluoride exchange (SuFEx) transformations. We synthesized over 3,000 analogs of 5'-amino lenalidomide using this approach and screened the crude products using a phenotypic screen for cell viability, identifying dozens of analogs with differentiated activity. We characterized four compounds that degrade G-to-S phase transition 1 (GSPT1) protein, providing a proof-of-concept model for SuFEx-based discovery of CRBN molecular glues.
Collapse
Affiliation(s)
- Trever R Carter
- Department of Chemistry, The Scripps Research Institute, United States
| | | | - Lucas Dada
- Department of Biochemistry, Albert Einstein College of Medicine, United States
| | - James B Shaum
- Department of Chemistry, The Scripps Research Institute, United States
| | - K Barry Sharpless
- Department of Chemistry, The Scripps Research Institute, United States
| | - Seiya Kitamura
- Department of Chemistry, The Scripps Research Institute, United States; Department of Biochemistry, Albert Einstein College of Medicine, United States
| | - Michael A Erb
- Department of Chemistry, The Scripps Research Institute, United States
| |
Collapse
|
18
|
Hong J, Li C, Zhao K, Wang X, Feng R, Chen X, Wei C, Gong X, Zheng F, Zheng C. Stereoselective Fluorosulfonylation of Vinylboronic Acids for ( E)-Vinyl Sulfonyl Fluorides with Copper Participation. Org Lett 2024; 26:2332-2337. [PMID: 38478713 DOI: 10.1021/acs.orglett.4c00711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
A practical synthetic method for the synthesis of vinyl sulfonyl fluorides through copper-promoted direct fluorosulfonylation has been developed. The reaction of the vinylboronic acids with DABSO and then NFSI is performed under mild reaction conditions. This transformation efficiently affords aryl or alkyl vinyl sulfonyl fluorides with good reaction yields, exclusive E-configuration, broad substrate scope, excellent compatibility, and operational simplicity.
Collapse
Affiliation(s)
- Jianquan Hong
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu 214122, P. R. China
| | - Chunxiang Li
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu 214122, P. R. China
| | - Kui Zhao
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu 214122, P. R. China
| | - Xiaoyu Wang
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu 214122, P. R. China
| | - Ruilong Feng
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu 214122, P. R. China
| | - Xifei Chen
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu 214122, P. R. China
| | - Chongbin Wei
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu 214122, P. R. China
| | - Xinxin Gong
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu 214122, P. R. China
| | - Feng Zheng
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu 214122, P. R. China
| | - Changge Zheng
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu 214122, P. R. China
| |
Collapse
|
19
|
McFadden WM, Casey-Moore MC, Bare GAL, Kirby KA, Wen X, Li G, Wang H, Slack RL, Snyder AA, Lorson ZC, Kaufman IL, Cilento ME, Tedbury PR, Gembicky M, Olson AJ, Torbett BE, Sharpless KB, Sarafianos SG. Identification of clickable HIV-1 capsid-targeting probes for viral replication inhibition. Cell Chem Biol 2024; 31:477-486.e7. [PMID: 38518746 PMCID: PMC11257216 DOI: 10.1016/j.chembiol.2024.02.012] [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: 03/06/2023] [Revised: 12/15/2023] [Accepted: 02/27/2024] [Indexed: 03/24/2024]
Abstract
Of the targets for HIV-1 therapeutics, the capsid core is a relatively unexploited but alluring drug target due to its indispensable roles throughout virus replication. Because of this, we aimed to identify "clickable" covalent modifiers of the HIV-1 capsid protein (CA) for future functionalization. We screened a library of fluorosulfate compounds that can undergo sulfur(VI) fluoride exchange (SuFEx) reactions, and five compounds were identified as hits. These molecules were further characterized for antiviral effects. Several compounds impacted in vitro capsid assembly. One compound, BBS-103, covalently bound CA via a SuFEx reaction to Tyr145 and had antiviral activity in cell-based assays by perturbing virus production, but not uncoating. The covalent binding of compounds that target the HIV-1 capsid could aid in the future design of antiretroviral drugs or chemical probes that will help study aspects of HIV-1 replication.
Collapse
Affiliation(s)
- William M McFadden
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, 1760 Haygood Drive NE, Atlanta, GA 30322, USA; Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Mary C Casey-Moore
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA; Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Grant A L Bare
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Karen A Kirby
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, 1760 Haygood Drive NE, Atlanta, GA 30322, USA; Children's Healthcare of Atlanta, Atlanta, GA 30322, USA; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA; Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Xin Wen
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, 1760 Haygood Drive NE, Atlanta, GA 30322, USA; Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Gencheng Li
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Hua Wang
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ryan L Slack
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, 1760 Haygood Drive NE, Atlanta, GA 30322, USA; Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Alexa A Snyder
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, 1760 Haygood Drive NE, Atlanta, GA 30322, USA; Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Zachary C Lorson
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, 1760 Haygood Drive NE, Atlanta, GA 30322, USA; Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Isabella L Kaufman
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, 1760 Haygood Drive NE, Atlanta, GA 30322, USA; Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Maria E Cilento
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, 1760 Haygood Drive NE, Atlanta, GA 30322, USA; Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Philip R Tedbury
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, 1760 Haygood Drive NE, Atlanta, GA 30322, USA; Children's Healthcare of Atlanta, Atlanta, GA 30322, USA; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA; Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Milan Gembicky
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92521, United States
| | - Arthur J Olson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Bruce E Torbett
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98101, USA
| | - K Barry Sharpless
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Stefan G Sarafianos
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, 1760 Haygood Drive NE, Atlanta, GA 30322, USA; Children's Healthcare of Atlanta, Atlanta, GA 30322, USA; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA; Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA.
| |
Collapse
|
20
|
Homer JA, Koelln RA, Barrow AS, Gialelis TL, Boiarska Z, Steinohrt NS, Lee EF, Yang WH, Johnson RM, Chung T, Habowski AN, Vishwakarma DS, Bhunia D, Avanzi C, Moorhouse AD, Jackson M, Tuveson DA, Lyons SK, Lukey MJ, Fairlie WD, Haider SM, Steinmetz MO, Prota AE, Moses JE. Modular synthesis of functional libraries by accelerated SuFEx click chemistry. Chem Sci 2024; 15:3879-3892. [PMID: 38487227 PMCID: PMC10935723 DOI: 10.1039/d3sc05729a] [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: 10/26/2023] [Accepted: 02/09/2024] [Indexed: 03/17/2024] Open
Abstract
Accelerated SuFEx Click Chemistry (ASCC) is a powerful method for coupling aryl and alkyl alcohols with SuFEx-compatible functional groups. With its hallmark favorable kinetics and exceptional product yields, ASCC streamlines the synthetic workflow, simplifies the purification process, and is ideally suited for discovering functional molecules. We showcase the versatility and practicality of the ASCC reaction as a tool for the late-stage derivatization of bioactive molecules and in the array synthesis of sulfonate-linked, high-potency, microtubule targeting agents (MTAs) that exhibit nanomolar anticancer activity against multidrug-resistant cancer cell lines. These findings underscore ASCC's promise as a robust platform for drug discovery.
Collapse
Affiliation(s)
- Joshua A Homer
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | - Rebecca A Koelln
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | - Andrew S Barrow
- La Trobe Institute for Molecular Science, La Trobe University Melbourne VIC 3086 Australia
| | - Timothy L Gialelis
- La Trobe Institute for Molecular Science, La Trobe University Melbourne VIC 3086 Australia
| | - Zlata Boiarska
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut Villigen PSI 5232 Switzerland
- Department of Chemistry, Università degli Studi di Milano Via Golgi 19 20133 Milan Italy
| | - Nikita S Steinohrt
- Olivia Newton-John Cancer Research Institute Heidelberg Victoria 3084 Australia
- School of Cancer Medicine, La Trobe University Melbourne Victoria 3086 Australia
| | - Erinna F Lee
- Olivia Newton-John Cancer Research Institute Heidelberg Victoria 3084 Australia
- School of Cancer Medicine, La Trobe University Melbourne Victoria 3086 Australia
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University Melbourne Victoria 3086 Australia
| | - Wen-Hsuan Yang
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | - Robert M Johnson
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | - Taemoon Chung
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | - Amber N Habowski
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | | | - Debmalya Bhunia
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | - Charlotte Avanzi
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University Fort Collins CO 80523 USA
| | - Adam D Moorhouse
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University Fort Collins CO 80523 USA
| | - David A Tuveson
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | - Scott K Lyons
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | - Michael J Lukey
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| | - W Douglas Fairlie
- Olivia Newton-John Cancer Research Institute Heidelberg Victoria 3084 Australia
- School of Cancer Medicine, La Trobe University Melbourne Victoria 3086 Australia
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University Melbourne Victoria 3086 Australia
| | - Shozeb M Haider
- School of Pharmacy, University College London 29-39 Brunswick Square London WC1N 1AX UK
| | - Michel O Steinmetz
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut Villigen PSI 5232 Switzerland
- Biozentrum, University of Basel 4056 Basel Switzerland
| | - Andrea E Prota
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut Villigen PSI 5232 Switzerland
| | - John E Moses
- Cancer Center, Cold Spring Harbor Laboratory 1 Bungtown Rd Cold Spring Harbor NY 11724 USA
| |
Collapse
|
21
|
Yin CL, Qin RZ, Qin HL. One-Pot Three-Component Synthesis of Indolyl-4 H-chromene-3-sulfonyl Fluoride: A Class of Important Pharmacophore. J Org Chem 2024; 89:3618-3628. [PMID: 38358945 DOI: 10.1021/acs.joc.3c02706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
A one-pot, sequential three-component reaction between salicylaldehyde, indole, and 2-bromoprop-2-ene-1-sulfonyl fluoride (BPESF) has been demonstrated for the synthesis of sulfonyl fluoride substituted 4H-chromene derivatives in moderate to excellent yields (45%-94%). This one-pot sequential method features easily available starting materials, wide substrate scope, mild conditions, and great efficiency.
Collapse
Affiliation(s)
- Cheng-Lin Yin
- State Key Laboratory of Silicate Materials for Architectures; and School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, People's Republic of China
| | - Richard Zijian Qin
- State Key Laboratory of Silicate Materials for Architectures; and School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, People's Republic of China
| | - Hua-Li Qin
- State Key Laboratory of Silicate Materials for Architectures; and School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, People's Republic of China
| |
Collapse
|
22
|
Peng Z, Sun S, Zheng MM, Li Y, Li X, Li S, Xue XS, Dong J, Gao B. Enantioselective sulfur(VI) fluoride exchange reaction of iminosulfur oxydifluorides. Nat Chem 2024; 16:353-362. [PMID: 38355829 DOI: 10.1038/s41557-024-01452-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 01/17/2024] [Indexed: 02/16/2024]
Abstract
Linkage chemistry and functional molecules derived from the stereogenic sulfur(VI) centre have important applications in organic synthesis, bioconjugation, drug discovery, agrochemicals and polymeric materials. However, existing approaches for the preparation of optically active S(VI)-centred compounds heavily rely on synthetic chiral S(IV) pools, and the reported linkers of S(VI) lack stereocontrol. A modular assembly method, involving sequential ligand exchange at the S(VI) centre with precise control of enantioselectivity, is appealing but remains elusive. Here we report an asymmetric three-dimensional sulfur(VI) fluoride exchange (3D-SuFEx) reaction based on thionyl tetrafluoride gas (SOF4). A key step involves the chiral ligand-induced enantioselective defluorinative substitution of iminosulfur oxydifluorides using organolithium reagents. The resulting optically active sulfonimidoyl fluorides allow for further stereospecific fluoride-exchange by various nucleophiles, thereby establishing a modular platform for the asymmetric SuFEx ligation and the divergent synthesis of optically active S(VI) functional molecules.
Collapse
Affiliation(s)
- Zhiyuan Peng
- State Key Laboratory of Chemo/BioSensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha, China
| | - Shoujun Sun
- Institute of Translational Medicine, National Facility for Translational Medicine (Shanghai), Shanghai Jiao Tong University, Shanghai, China
| | - Meng-Meng Zheng
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Yangyang Li
- State Key Laboratory of Chemo/BioSensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha, China
| | - Xixi Li
- Institute of Translational Medicine, National Facility for Translational Medicine (Shanghai), Shanghai Jiao Tong University, Shanghai, China
| | - Suhua Li
- School of Chemistry, Sun Yat-Sen University, Guangzhou, China
| | - Xiao-Song Xue
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
- Key Laboratory of Fluorine and Nitrogen Chemistry and Advanced Materials, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Jiajia Dong
- Institute of Translational Medicine, National Facility for Translational Medicine (Shanghai), Shanghai Jiao Tong University, Shanghai, China.
| | - Bing Gao
- State Key Laboratory of Chemo/BioSensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha, China.
| |
Collapse
|
23
|
Zhao S, Zeng D, Wang M, Jiang X. C-SuFEx linkage of sulfonimidoyl fluorides and organotrifluoroborates. Nat Commun 2024; 15:727. [PMID: 38272934 PMCID: PMC10810801 DOI: 10.1038/s41467-024-44998-6] [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: 08/23/2023] [Accepted: 01/11/2024] [Indexed: 01/27/2024] Open
Abstract
Sulfur(VI) fluoride exchange, a new type of linkage reaction, has excellent potential for application in functional molecule linkage to prepare pharmaceuticals, biomolecules, and polymers. Herein, a C-SuFEx reaction is established to achieve fast (in minutes) linkage between sulfonimidoyl fluorides and aryl/alkyl organotrifluoroborates. Potassium organotrifluoroborates are instantaneously activated via a substoichiometric amount of trimethylsilyl triflate to afford organodifluoroboranes, releasing BF3 as an activating reagent in situ. This sulfur(VI) fluoride exchange technique is capable of forming S(VI)-C(alkyl), S(VI)-C(alkenyl) and S(VI)-C(aryl) bonds, demonstrating its broad scope. Natural products and pharmaceuticals with sensitive functional groups, such as valdecoxib, celecoxib and diacetonefructose, are compatible with this protocol, allowing the formation of diverse sulfoximines.
Collapse
Affiliation(s)
- Suqin Zhao
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, China
| | - Daming Zeng
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, China
| | - Ming Wang
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, China.
| | - Xuefeng Jiang
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, China.
- State Key Laboratory of Petroleum Molecular and Process engineering, SKLPMPE, Sinopec research institute of petroleum processing Co., LTD., Beijing 100083, China; East China Normal University, Shanghai, 200062, China.
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, 453007, China.
| |
Collapse
|
24
|
Shim SY. Late-Stage C-H Activation of Drug (Derivative) Molecules with Pd(ll) Catalysis. Chemistry 2023; 29:e202302620. [PMID: 37846586 DOI: 10.1002/chem.202302620] [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: 08/10/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/18/2023]
Abstract
This review comprehensively analyses representative examples of Pd(II)-catalyzed late-stage C-H activation reactions and demonstrates their efficacy in converting C-H bonds at multiple positions within drug (derivative) molecules into diverse functional groups. These transformative reactions hold immense potential in medicinal chemistry, enabling the efficient and selective functionalization of specific sites within drug molecules, thereby enhancing their pharmacological activity and expanding the scope of potential drug candidates. Although notable articles have focused on late-stage C-H functionalization reactions of drug-like molecules using transition-metal catalysts, reviews specifically focusing on late-stage C-H functionalization reactions of drug (derivative) molecules using Pd(II) catalysts are required owing to their prominence as the most widely utilized metal catalysts for C-H activation and their ability to introduce a myriad of functional groups at specific C-H bonds. The utilization of Pd-catalyzed C-H activation methodologies demonstrates impressive success in introducing various functional groups, such as cyano (CN), fluorine (F), chlorine (Cl), aromatic rings, olefin, alkyl, alkyne, and hydroxyl groups, to drug (derivative) molecules with high regioselectivity and functional-group tolerance. These breakthroughs in late-stage C-H activation reactions serve as invaluable tools for drug discovery and development, thereby offering strategic options to optimize drug candidates and drive the exploration of innovative therapeutic solutions.
Collapse
Affiliation(s)
- Su Yong Shim
- Infectious Diseases Therapeutic Research Center Division of Medicinal Chemistry and Pharmacology Korea Research Institute of Chemical Technology (KRICT) KRICT School, University of Science and Technology, Daejeon, 34114, Republic of Korea
| |
Collapse
|
25
|
Plesniak MP, Taylor EK, Eisele F, Kourra CMK, Michaelides IN, Oram A, Wernevik J, Valencia ZS, Rowbottom H, Mann N, Fredlund L, Pivnytska V, Novén A, Pirmoradian M, Lundbäck T, Storer RI, Pettersson M, De Donatis GM, Rehnström M. Rapid PROTAC Discovery Platform: Nanomole-Scale Array Synthesis and Direct Screening of Reaction Mixtures. ACS Med Chem Lett 2023; 14:1882-1890. [PMID: 38116431 PMCID: PMC10726452 DOI: 10.1021/acsmedchemlett.3c00314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/21/2023] [Accepted: 10/24/2023] [Indexed: 12/21/2023] Open
Abstract
Precise length, shape, and linker attachment points are all integral components to designing efficacious proteolysis targeting chimeras (PROTACs). Due to the synthetic complexity of these heterobifunctional degraders and the difficulty of computational modeling to aid PROTAC design, the exploration of structure-activity relationships remains mostly empirical, which requires a significant investment of time and resources. To facilitate rapid hit finding, we developed capabilities for PROTAC parallel synthesis and purification by harnessing an array of preformed E3-ligand-linker intermediates. In the next iteration of this approach, we developed a rapid, nanomole-scale PROTAC synthesis methodology using amide coupling that enables direct screening of nonpurified reaction mixtures in cell-based degradation assays, as well as logD and EPSA measurements. This approach greatly expands and accelerates PROTAC SAR exploration (5 days instead of several weeks) as well as avoids laborious and solvent-demanding purification of the reaction mixtures, thus making it an economical and more sustainable methodology for PROTAC hit finding.
Collapse
Affiliation(s)
- Mateusz P. Plesniak
- Medicinal
Chemistry, Research and Early Development, Cardiovascular, Renal and
Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | - Emilia K. Taylor
- Medicinal
Chemistry, Research and Early Development, Cardiovascular, Renal and
Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | - Frederik Eisele
- Mechanistic
& Structural Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | | | - Iacovos N. Michaelides
- Fragment
Based Lead Generation, Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 0WG, U.K.
| | - Alice Oram
- iLAB,
Compound Synthesis & Management, Discovery Sciences, R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | - Johan Wernevik
- Mechanistic
& Structural Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | | | - Hannah Rowbottom
- Mechanistic
& Structural Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | - Nadia Mann
- Mechanistic
& Structural Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | - Linda Fredlund
- Mechanistic
& Structural Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | - Valentyna Pivnytska
- Mechanistic
& Structural Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | - Anna Novén
- Mechanistic
& Structural Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | - Mohammad Pirmoradian
- Mechanistic
& Structural Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | - Thomas Lundbäck
- Mechanistic
& Structural Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | - R. Ian Storer
- Hit
Discovery, Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 0WG, U.K.
| | - Mariell Pettersson
- Medicinal
Chemistry, Research and Early Development, Cardiovascular, Renal and
Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | - Gian M. De Donatis
- Cellular
Assay Development, Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 0WG, U.K.
| | - Marie Rehnström
- Cell
Culture Sciences & Banking, Discovery Biology, Discovery Sciences,
R&D, AstraZeneca, Gothenburg 431 83, Sweden
| |
Collapse
|
26
|
Erb MA. Small-molecule tools for YEATS domain proteins. Curr Opin Chem Biol 2023; 77:102404. [PMID: 37924571 PMCID: PMC10842393 DOI: 10.1016/j.cbpa.2023.102404] [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: 08/09/2023] [Revised: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 11/06/2023]
Abstract
Chromatin reader domains are protein folds that bind to post-translational modifications of histones and other chromatin-associated proteins. Compared to other families of reader domains, the discovery that YEATS domains bind to acylated lysines is relatively recent. Four human proteins harbor a YEATS domain, and each is present in protein complexes that regulate chromatin and transcription (ENL, AF9, YEATS2, and YEATS4). Without chemical tools to enable temporally resolved perturbations, it is often unclear how reader domains contribute to protein function. Here, we will discuss recent progress in developing small-molecule tools for YEATS domains and highlight their usefulness for making biological discoveries.
Collapse
Affiliation(s)
- Michael A Erb
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
27
|
Julio AR, Shikwana F, Truong C, Burton NR, Dominguez E, Turmon AC, Cao J, Backus K. Pervasive aggregation and depletion of host and viral proteins in response to cysteine-reactive electrophilic compounds. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.30.564067. [PMID: 38014036 PMCID: PMC10680658 DOI: 10.1101/2023.10.30.564067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Protein homeostasis is tightly regulated, with damaged or misfolded proteins quickly eliminated by the proteasome and autophagosome pathways. By co-opting these processes, targeted protein degradation technologies enable pharmacological manipulation of protein abundance. Recently, cysteine-reactive molecules have been added to the degrader toolbox, which offer the benefit of unlocking the therapeutic potential of 'undruggable' protein targets. The proteome-wide impact of these molecules remains to be fully understood and given the general reactivity of many classes of cysteine-reactive electrophiles, on- and off-target effects are likely. Using chemical proteomics, we identified a cysteine-reactive small molecule degrader of the SARS-CoV-2 nonstructural protein 14 (nsp14), which effects degradation through direct modification of cysteines in both nsp14 and in host chaperones together with activation of global cell stress response pathways. We find that cysteine-reactive electrophiles increase global protein ubiquitylation, trigger proteasome activation, and result in widespread aggregation and depletion of host proteins, including components of the nuclear pore complex. Formation of stress granules was also found to be a remarkably ubiquitous cellular response to nearly all cysteine-reactive compounds and degraders. Collectively, our study sheds light on complexities of covalent target protein degradation and highlights untapped opportunities in manipulating and characterizing proteostasis processes via deciphering the cysteine-centric regulation of stress response pathways.
Collapse
Affiliation(s)
- Ashley R Julio
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095 (USA)
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA 90095 (USA)
| | - Flowreen Shikwana
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095 (USA)
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA 90095 (USA)
| | - Cindy Truong
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095 (USA)
| | - Nikolas R Burton
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095 (USA)
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA 90095 (USA)
| | - Emil Dominguez
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095 (USA)
| | - Alexandra C Turmon
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095 (USA)
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA 90095 (USA)
| | - Jian Cao
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA 90095 (USA)
| | - Keriann Backus
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095 (USA)
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA 90095 (USA)
- DOE Institute for Genomics and Proteomics, UCLA, Los Angeles, CA 90095 (USA)
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095 (USA)
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA 90095 (USA)
| |
Collapse
|
28
|
Hasan MN, Ray M, Saha A. Landscape of In Silico Tools for Modeling Covalent Modification of Proteins: A Review on Computational Covalent Drug Discovery. J Phys Chem B 2023; 127:9663-9684. [PMID: 37921534 DOI: 10.1021/acs.jpcb.3c04710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Covalent drug discovery has been a challenging research area given the struggle of finding a sweet balance between selectivity and reactivity for these drugs, the lack of which often leads to off-target activities and hence undesirable side effects. However, there has been a resurgence in covalent drug design following the success of several covalent drugs such as boceprevir (2011), ibrutinib (2013), neratinib (2017), dacomitinib (2018), zanubrutinib (2019), and many others. Design of covalent drugs includes many crucial factors, where "evaluation of the binding affinity" and "a detailed mechanistic understanding on covalent inhibition" are at the top of the list. Well-defined experimental techniques are available to elucidate these factors; however, often they are expensive and/or time-consuming and hence not suitable for high throughput screens. Recent developments in in silico methods provide promise in this direction. In this report, we review a set of recent publications that focused on developing and/or implementing novel in silico techniques in "Computational Covalent Drug Discovery (CCDD)". We also discuss the advantages and disadvantages of these approaches along with what improvements are required to make it a great tool in medicinal chemistry in the near future.
Collapse
Affiliation(s)
- Md Nazmul Hasan
- Department of Chemistry and Biochemistry, University of Wisconsin─Milwaukee, Milwaukee, Wisconsin 53211, United States
| | - Manisha Ray
- Department of Chemistry and Biochemistry, Loyola University Chicago, Chicago, Illinois 60660, United States
| | - Arjun Saha
- Department of Chemistry and Biochemistry, University of Wisconsin─Milwaukee, Milwaukee, Wisconsin 53211, United States
| |
Collapse
|
29
|
Pasieka A, Diamanti E, Uliassi E, Laura Bolognesi M. Click Chemistry and Targeted Degradation: A Winning Combination for Medicinal Chemists? ChemMedChem 2023; 18:e202300422. [PMID: 37706617 DOI: 10.1002/cmdc.202300422] [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] [Revised: 09/01/2023] [Indexed: 09/15/2023]
Abstract
Click chemistry is universally recognized as a powerful strategy for the fast and precise assembly of diverse building blocks. Targeted Protein Degradation (TPD) is a new therapeutic modality based on heterobifunctional small-molecule degraders that provides new opportunities to medicinal chemists dealing with undruggable targets and incurable diseases. Here, we highlight how very recently the TPD field and that of click chemistry have merged, opening up the possibility for fine-tuning the properties of a degrader, chemically assembled through a "click" synthesis. By reviewing concrete examples, we want to provide the reader with the insight that the application of click and bioorthogonal chemistry in the TDP field may be a winning combination.
Collapse
Affiliation(s)
- Anna Pasieka
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Eleonora Diamanti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Elisa Uliassi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| |
Collapse
|
30
|
Abstract
Ralstonia solanacearum species complex (RSSC) strains are devastating plant pathogens distributed worldwide. The primary cell density-dependent gene expression system in RSSC strains is phc quorum sensing (QS). It regulates the expression of about 30% of all genes, including those related to cellular activity, primary and secondary metabolism, pathogenicity, and more. The phc regulatory elements encoded by the phcBSRQ operon and phcA gene play vital roles. RSSC strains use methyl 3-hydroxymyristate (3-OH MAME) or methyl 3-hydroxypalmitate (3-OH PAME) as the QS signal. Each type of RSSC strain has specificity in generating and receiving its QS signal, but their signaling pathways might not differ significantly. In this review, I describe the genetic and biochemical factors involved in QS signal input and the regulatory network and summarize control of the phc QS system, new cell-cell communications, and QS-dependent interactions with soil fungi.
Collapse
Affiliation(s)
- Kenji Kai
- Graduate School of Agriculture, Osaka Metropolitan University, Osaka, Japan;
| |
Collapse
|
31
|
Audet F, Donnard M, Panossian A, Bernier D, Pazenok S, Leroux FR. New Chemical Transformations Involving SO 2 F 2 -Mediated Alcohol Activation. CHEM REC 2023; 23:e202300107. [PMID: 37236146 DOI: 10.1002/tcr.202300107] [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: 03/27/2023] [Revised: 05/02/2023] [Indexed: 05/28/2023]
Abstract
Sulfuryl fluoride is a gas produced on a multi-ton scale for its use as a fumigant. In the last decades, it has gained interest in organic synthesis as a reagent with unique properties in terms of stability and reactivity when compared to other sulfur-based reagents. Sulfuryl fluoride has not only been used for sulfur-fluoride exchange (SuFEx) chemistry but also encountered applications in classic organic synthesis as an efficient activator of both alcohols and phenols, forming a triflate surrogate, namely a fluorosulfonate. A long-standing industrial collaboration in our research group drove our work on the sulfuryl fluoride-mediated transformations that will be highlighted below. We will first describe recent works on metal-catalyzed transformations from aryl fluorosulfonates while emphasizing the one-pot processes from phenol derivatives. In a second section, nucleophilic substitution reactions on polyfluoroalkyl alcohols will be discussed and the value of polyfluoroalkyl fluorosulfonates in comparison to alternative triflate and halide reagents will be brought to light.
Collapse
Affiliation(s)
- Florian Audet
- Laboratoire d'Innovation Moléculaire et Applications (UMR7042), Université de Strasbourg, Université de Haute-Alsace, CNRS, 25 rue Becquerel, 67000, Strasbourg, France
| | - Morgan Donnard
- Laboratoire d'Innovation Moléculaire et Applications (UMR7042), Université de Strasbourg, Université de Haute-Alsace, CNRS, 25 rue Becquerel, 67000, Strasbourg, France
| | - Armen Panossian
- Laboratoire d'Innovation Moléculaire et Applications (UMR7042), Université de Strasbourg, Université de Haute-Alsace, CNRS, 25 rue Becquerel, 67000, Strasbourg, France
| | - David Bernier
- Bayer S.A.S., 14 impasse Pierre Baizet, 69263, Lyon, France
| | - Sergii Pazenok
- Bayer CropScience AG, Alfred Nobel Straße 50, 40789, Monheim, Germany
| | - Frédéric R Leroux
- Laboratoire d'Innovation Moléculaire et Applications (UMR7042), Université de Strasbourg, Université de Haute-Alsace, CNRS, 25 rue Becquerel, 67000, Strasbourg, France
| |
Collapse
|
32
|
Wang T, Xu L, Dong J. FSO 2N 3-Enabled Synthesis of Tetrazoles from Amidines and Guanidines. Org Lett 2023; 25:6222-6227. [PMID: 37581428 DOI: 10.1021/acs.orglett.3c02470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2023]
Abstract
Herein we report the facile syntheses of tetrazoles enabled by FSO2N3 under mild conditions. FSO2N3 has been shown as the most powerful diazotizing reagent, which converts thousands of primary amines to azides fast and orthogonally. As the follow-up studies of the diazo transfer reaction using FSO2N3, we discover that amidines and guanidines are rapidly transformed into tetrazole derivatives when reacting with FSO2N3 under an aqueous environment, which is unprecedented for tetrazole synthesis.
Collapse
Affiliation(s)
- Tianyu Wang
- Laboratory of Organofluorine Chemistry Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Long Xu
- Institute of Translational Medicine, Zhangjiang Institute for Advanced Study, National Facility for Translational Medicine (Shanghai), Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiajia Dong
- Institute of Translational Medicine, Zhangjiang Institute for Advanced Study, National Facility for Translational Medicine (Shanghai), Shanghai Jiao Tong University, Shanghai 200240, China
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Shanghai Artificial Intelligence Laboratory, Shanghai 200232, China
| |
Collapse
|
33
|
Abstract
The impact of click chemistry was recently recognized with the 2022 Nobel Prize in Chemistry. The breadth of areas where click chemistry has accelerated discovery is prodigal. In one of the most written about subjects in chemistry over recent years, this short perspective zones in on a small fragment of what we, the authors, consider are some of the most critical developments in synthetic chemistry, which have expanded access to the click chemistry toolbox. In addition, we touch upon areas within medicinal chemistry and novel approaches to drug discovery enabled by click chemistry, where we believe there is untapped potential for biological function to be found and exploited.
Collapse
Affiliation(s)
- Adam D Moorhouse
- Cancer Centre, Cold Spring Harbor Laboratory, 1 Bungtown Road, New York, NY 11724, USA
| | - Joshua A Homer
- Cancer Centre, Cold Spring Harbor Laboratory, 1 Bungtown Road, New York, NY 11724, USA
| | - John E Moses
- Cancer Centre, Cold Spring Harbor Laboratory, 1 Bungtown Road, New York, NY 11724, USA
- Lead Contact
| |
Collapse
|
34
|
Homer JA, Xu L, Kayambu N, Zheng Q, Choi EJ, Kim BM, Sharpless KB, Zuilhof H, Dong J, Moses JE. Sulfur fluoride exchange. NATURE REVIEWS. METHODS PRIMERS 2023; 3:58. [PMID: 38873592 PMCID: PMC11171465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Sulfur Fluoride Exchange (SuFEx) is a click reaction par excellence that has revolutionized multiple research fields. In this Primer, we delve into the essential elements of SuFEx operation, catalysis, and SuFExable connective hubs. We also explore the cutting-edge applications of SuFEx in drug development, polymer science, and biochemistry. Additionally, we examine the potential limitations and promising prospects for this versatile click reaction.
Collapse
Affiliation(s)
- Joshua A. Homer
- Cancer Center, Cold Spring Harbor Laboratory, 1 Bungtown Road, NY 11724, USA
| | - Long Xu
- Institute of Translational Medicine, Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Namitharan Kayambu
- Laboratory of Organic Chemistry, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Qinheng Zheng
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
- Current affiliation: Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA
| | - Eun Joung Choi
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, South Korea
| | - Byeong Moon Kim
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, South Korea
| | - K. Barry Sharpless
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Han Zuilhof
- Laboratory of Organic Chemistry, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
- School of Pharmaceutical Science & Technology, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Jiajia Dong
- Institute of Translational Medicine, Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Shanghai Artificial Intelligence Laboratory, Shanghai 200232, China
| | - John E. Moses
- Cancer Center, Cold Spring Harbor Laboratory, 1 Bungtown Road, NY 11724, USA
| |
Collapse
|
35
|
Yu B, Cao L, Li S, Klauser PC, Wang L. The proximity-enabled sulfur fluoride exchange reaction in the protein context. Chem Sci 2023; 14:7913-7921. [PMID: 37502323 PMCID: PMC10370592 DOI: 10.1039/d3sc01921g] [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: 04/12/2023] [Accepted: 06/20/2023] [Indexed: 07/29/2023] Open
Abstract
The proximity-enabled sulfur(vi) fluoride exchange (SuFEx) reaction generates specific covalent linkages between proteins in cells and in vivo, which opens innovative avenues for studying elusive protein-protein interactions and developing potent covalent protein drugs. To exploit the power and expand the applications of covalent proteins, covalent linkage formation between proteins is the critical step, for which fundamental kinetic and essential properties remain unexplored. Herein, we systematically studied SuFEx kinetics in different proteins and conditions. In contrast to in small molecules, SuFEx in interacting proteins conformed with a two-step mechanism involving noncovalent binding, followed by covalent bond formation, exhibiting nonlinear rate dependence on protein concentration. The protein SuFEx rate consistently changed with protein binding affinity as well as chemical reactivity of the functional group and was impacted by target residue identity and solution pH. In addition, kinetic analyses of nanobody SR4 binding with SARS-CoV-2 spike protein revealed that viral target mutations did not abolish covalent binding but decreased the SuFEx rate with affinity decrease. Moreover, off-target cross-linking of a SuFEx-capable nanobody in human serum was not detected, and the SuFEx-generated protein linkage was stable at cellular acidic pHs, suggesting SuFEx suitability for in vivo usage. These results advanced our understanding of SuFEx reactivity and kinetics in proteins, which is invaluable for ongoing exploration of SuFEx-enabled covalent proteins for basic biological research and creative biotherapeutics.
Collapse
Affiliation(s)
- Bingchen Yu
- Department of Pharmaceutical Chemistry, The Cardiovascular Research Institute, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco 555 Mission Bay Blvd. South San Francisco California 94158 USA
| | - Li Cao
- Department of Pharmaceutical Chemistry, The Cardiovascular Research Institute, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco 555 Mission Bay Blvd. South San Francisco California 94158 USA
| | - Shanshan Li
- Department of Pharmaceutical Chemistry, The Cardiovascular Research Institute, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco 555 Mission Bay Blvd. South San Francisco California 94158 USA
| | - Paul C Klauser
- Department of Pharmaceutical Chemistry, The Cardiovascular Research Institute, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco 555 Mission Bay Blvd. South San Francisco California 94158 USA
| | - Lei Wang
- Department of Pharmaceutical Chemistry, The Cardiovascular Research Institute, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco 555 Mission Bay Blvd. South San Francisco California 94158 USA
| |
Collapse
|
36
|
Smedley CJ, Giel MC, Fallon T, Moses JE. Ethene-1,1-disulfonyl Difluoride (EDSF) for SuFEx Click Chemistry: Synthesis of SuFExable 1,1-Bissulfonylfluoride Substituted Cyclobutene Hubs. Angew Chem Int Ed Engl 2023; 62:e202303916. [PMID: 37224463 PMCID: PMC10958772 DOI: 10.1002/anie.202303916] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/21/2023] [Accepted: 05/22/2023] [Indexed: 05/26/2023]
Abstract
We present the synthesis of 1,1-bis(fluorosulfonyl)-2-(pyridin-1-ium-1-yl)ethan-1-ide, a bench-stable precursor to ethene-1,1-disulfonyl difluoride (EDSF). The novel SuFEx reagent, EDSF, is demonstrated in the preparation of 26 unique 1,1-bissulfonylfluoride substituted cyclobutenes via a cycloaddition reaction. The regioselective click cycloaddition reaction is rapid, straightforward, and highly efficient, enabling the generation of highly functionalized 4-membered ring (4MR) carbocycles. These carbocycles are valuable structural motifs found in numerous bioactive natural products and pharmaceutically relevant small molecules. Additionally, we showcase diversification of the novel cyclobutene cores through selective Cs2 CO3 -activated SuFEx click chemistry between a single S-F group and an aryl alcohol, yielding the corresponding sulfonate ester products with high efficiency. Finally, density functional theory calculations offer mechanistic insights about the reaction pathway.
Collapse
Affiliation(s)
- Christopher J. Smedley
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
- La Trobe Institute for Molecular Science, Melbourne, VIC 3086, Australia
| | - Marie-Claire Giel
- La Trobe Institute for Molecular Science, Melbourne, VIC 3086, Australia
| | - Thomas Fallon
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - John E. Moses
- Cancer Center, Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724 (USA)
| |
Collapse
|
37
|
Abstract
A SuFEx linkage reaction between sulfonimidoyl fluoride and allyltrimethylsilane was achieved for the construction of N-modified allylsulfoximines in minutes with BF3 as a nonmetal difunctional activator enabling the activation of both S-F and C-Si bonds to forge the S-Callyl (sp3) bond swiftly. Mechanistic studies and DFT calculations indicated that the linkage was initiated with the activation of sulfonimidoyl fluoride and then followed with the transfer of the fluoride anion to the TMS group.
Collapse
Affiliation(s)
- Daming Zeng
- State Key Laboratory of Molecular and Process Engineering, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, P. R. China
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Suqin Zhao
- State Key Laboratory of Molecular and Process Engineering, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, P. R. China
| | - Wei-Ping Deng
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xuefeng Jiang
- State Key Laboratory of Molecular and Process Engineering, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, P. R. China
- State Key Laboratory of Elemento-organic Chemistry, Nankai University, Tianjin 300071, P. R. China
| |
Collapse
|
38
|
Vincent CA, Ripak A, Troian-Gautier L, Tambar UK. Photocatalytic conversion of aryl diazonium salts to sulfonyl fluorides. Tetrahedron 2023; 139:133364. [PMID: 38404686 PMCID: PMC10887421 DOI: 10.1016/j.tet.2023.133364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Sulfonyl fluorides have emerged as powerful tools in chemical biology for the selective labelling of proteins. A photocatalytic method is described for the conversion of aryl diazonium salts to aryl sulfonyl fluorides. The diazonium substrates are easily obtained in one step from functionalized anilines. We present the optimization of this mild method for the synthesis of sulfonyl fluorides, the scope of the transformation with a series of functionalized diazonium salts, and we discuss photophysical measurements that provide detailed information about the mechanism of the photochemical process.
Collapse
Affiliation(s)
- Cooper A. Vincent
- Department of Biochemistry, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9038, United States
| | - Alexia Ripak
- Université Catholique de Louvain (UCLouvain), Institut de la Matière Condensée et des Nanosciences (IMCN), Molecular Chemistry, Materials and Catalysis (MOST), Place Louis Pasteur 1, bte L4.01.02, 1348, Louvain-la-Neuve, Belgium
| | - Ludovic Troian-Gautier
- Université Catholique de Louvain (UCLouvain), Institut de la Matière Condensée et des Nanosciences (IMCN), Molecular Chemistry, Materials and Catalysis (MOST), Place Louis Pasteur 1, bte L4.01.02, 1348, Louvain-la-Neuve, Belgium
| | - Uttam K. Tambar
- Department of Biochemistry, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9038, United States
| |
Collapse
|
39
|
Zeng D, Deng WP, Jiang X. Advances in the construction of diverse SuFEx linkers. Natl Sci Rev 2023; 10:nwad123. [PMID: 37441224 PMCID: PMC10335383 DOI: 10.1093/nsr/nwad123] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 12/02/2022] [Accepted: 01/30/2023] [Indexed: 07/15/2023] Open
Abstract
Sulfur fluoride exchange (SuFEx), a new generation of click chemistry, was first presented by Sharpless, Dong and co-workers in 2014. Owing to the high stability and yet efficient reactivity of the SVI-F bond, SuFEx has found widespread applications in organic synthesis, materials science, chemical biology and drug discovery. A diverse collection of SuFEx linkers has emerged, involving gaseous SO2F2 and SOF4 hubs; SOF4-derived iminosulfur oxydifluorides; O-, N- and C-attached sulfonyl fluorides and sulfonimidoyl fluorides; and novel sulfondiimidoyl fluorides. This review summarizes the progress of these SuFEx connectors, with an emphasis on analysing the advantages and disadvantages of synthetic strategies of these connectors based on the SuFEx concept, and it is expected to be beneficial to researchers to rapidly and correctly understand this field, thus inspiring further development in SuFEx chemistry.
Collapse
Affiliation(s)
- Daming Zeng
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Shanghai Key Laboratory of Green Chemistry and Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | | | | |
Collapse
|
40
|
Petri L, Ábrányi-Balogh P, Csorba N, Keeley A, Simon J, Ranđelović I, Tóvári J, Schlosser G, Szabó D, Drahos L, Keserű GM. Activation-Free Sulfonyl Fluoride Probes for Fragment Screening. Molecules 2023; 28:molecules28073042. [PMID: 37049805 PMCID: PMC10096327 DOI: 10.3390/molecules28073042] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023] Open
Abstract
SuFEx chemistry is based on the unique reactivity of the sulfonyl fluoride group with a range of nucleophiles. Accordingly, sulfonyl fluorides label multiple nucleophilic amino acid residues, making these reagents popular in both chemical biology and medicinal chemistry applications. The reactivity of sulfonyl fluorides nominates this warhead chemotype as a candidate for an external, activation-free general labelling tag. Here, we report the synthesis and characterization of a small sulfonyl fluoride library that yielded the 3-carboxybenzenesulfonyl fluoride warhead for tagging tractable targets at nucleophilic residues. Based on these results, we propose that coupling diverse fragments to this warhead would result in a library of sulfonyl fluoride bits (SuFBits), available for screening against protein targets. SuFBits will label the target if it binds to the core fragment, which facilitates the identification of weak fragments by mass spectrometry.
Collapse
Affiliation(s)
- László Petri
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
- National Laboratory for Drug Research and Development, Research Centre for Natural Sciences, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
| | - Péter Ábrányi-Balogh
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
- National Laboratory for Drug Research and Development, Research Centre for Natural Sciences, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, Szent Gellért tér 4, 1111 Budapest, Hungary
| | - Noémi Csorba
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
- National Laboratory for Drug Research and Development, Research Centre for Natural Sciences, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, Szent Gellért tér 4, 1111 Budapest, Hungary
| | - Aaron Keeley
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
| | - József Simon
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
- Research Centre for Natural Sciences, MS Metabolomics Research Group, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
| | | | - József Tóvári
- Department of Experimental Pharmacology and National Tumor Biology Laboratory POB 21, National Institute of Oncology, 1525 Budapest, Hungary
| | - Gitta Schlosser
- MTA-ELTE Lendület Ion Mobility Mass Spectrometry Research Group, Institute of Chemistry, ELTE Eötvös Loránd University, Pázmány Péter Sétány 1/A, 1117 Budapest, Hungary
| | - Dániel Szabó
- MS Proteomics Research Group, Research Centre for Natural Sciences, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
| | - László Drahos
- MS Proteomics Research Group, Research Centre for Natural Sciences, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
| | - György M Keserű
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
- National Laboratory for Drug Research and Development, Research Centre for Natural Sciences, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, Szent Gellért tér 4, 1111 Budapest, Hungary
| |
Collapse
|
41
|
Zhang X, Wang F, Tan CH. Asymmetric Synthesis of S(IV) and S(VI) Stereogenic Centers. JACS AU 2023; 3:700-714. [PMID: 37006767 PMCID: PMC10052288 DOI: 10.1021/jacsau.2c00626] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/06/2022] [Accepted: 12/06/2022] [Indexed: 05/22/2023]
Abstract
Sulfur can form diverse S(IV) and S(VI) stereogenic centers, of which some have gained significant attention recently due to their increasing use as pharmacophores in drug discovery programs. The preparation of these sulfur stereogenic centers in their enantiopure form has been challenging, and progress made will be discussed in this Perspective. This Perspective summarizes different strategies, with selected works, for asymmetric synthesis of these moieties, including diastereoselective transformations using chiral auxiliaries, enantiospecific transformations of enantiopure sulfur compounds, and catalytic enantioselective synthesis. We will discuss the advantages and limitations of these strategies and will provide our views on how this field will develop.
Collapse
Affiliation(s)
- Xin Zhang
- West China
School of Public Health and West China Fourth Hospital, and State
Key Laboratory of Biotherapy, Sichuan University, 610041 Chengdu, China
| | - Fucheng Wang
- West China
School of Public Health and West China Fourth Hospital, and State
Key Laboratory of Biotherapy, Sichuan University, 610041 Chengdu, China
| | - Choon-Hong Tan
- School
of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, 637371 Singapore
| |
Collapse
|
42
|
Shreiber ST, Molander GA. Alkyl (Het)Arylsulfones from SuFEx Reagents via Photochemical S-F Bond Activation. Org Lett 2023; 25:2084-2087. [PMID: 36939137 DOI: 10.1021/acs.orglett.3c00447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
Abstract
A visible light-induced S-F bond activation of sulfur fluoride exchange (SuFEx) reagents to generate alkyl sulfones is described. The method is free of transition metals and relies on the use of commercially available commodity chemicals. In addition, this method demonstrates the first photoredox functionalization of SuFEx reagents. The reaction has a diverse substrate scope, with applications in various aryl sulfonyl fluorides, both activated and unactivated alkenes.
Collapse
Affiliation(s)
- Scott T Shreiber
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, United States
| | - Gary A Molander
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, United States
| |
Collapse
|
43
|
Vincent CA, Chiriac MI, Troian-Gautier L, Tambar UK. Photocatalytic Sulfonyl Fluorination of Alkyl Organoboron Substrates. ACS Catal 2023; 13:3668-3675. [PMID: 37124721 PMCID: PMC10135442 DOI: 10.1021/acscatal.3c00107] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Sulfonyl fluorides are highly versatile molecules for click chemistry that have found applications in many areas of chemistry and biology. Recent chemical approaches have focused on the synthesis of alkyl sulfonyl fluorides from readily available starting materials. Here, we report a photocatalytic synthesis of alkyl sulfonyl fluorides from organotrifluoroborates and boronic acid pinacol esters, which are building blocks commonly employed by medicinal chemists in the synthesis of bioactive molecules. Steady-state and time-resolved spectroscopy have confirmed that the absorption of photons by the acridinium catalysts leads to the oxidation of the organotrifluoroborate substrates. The reaction exhibits broad functional group tolerance, which can be attributed to the mild activation with visible light. Importantly, this general approach provides easy access to primary, secondary, and tertiary alkyl sulfonyl fluorides.
Collapse
Affiliation(s)
- Cooper A Vincent
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas 75390-9038, United States
| | - Maria Irina Chiriac
- Department of Discovery Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Ludovic Troian-Gautier
- Molecular Chemistry, Materials and Catalysis (MOST), Université catholique de Louvain (UCLouvain), Institut de la Matierè Condensée et des Nanosciences (IMCN), 1348 Louvain-la-Neuve, Belgium
| | - Uttam K Tambar
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas 75390-9038, United States
| |
Collapse
|
44
|
Lin L, Wang P, Dong T, Tsui GC, Liao S. Radical Fluorosulfonyl Heteroarylation of Unactivated Alkenes with Quinoxalin-2(1 H)-ones and Related N-Heterocycles. Org Lett 2023; 25:1088-1093. [PMID: 36775923 DOI: 10.1021/acs.orglett.2c04315] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
The incorporation of sulfonyl fluoride groups into molecules has been proved effective to enhance their biological activities or introduce new functions. Herein, we report a transition-metal-free and visible-light-mediated radical 1-fluorosulfonyl-2-heteroarylation of alkenes, which could allow access to a series of SO2F-containing quinoxalin-2(1H)-ones, which are a critical structural motif widely present in a number of biologically active molecules. Further application of the method to the modification of other heterocycles and drug molecules as well as ligation chemistry via SuFEx click reactions is also demonstrated.
Collapse
Affiliation(s)
- Lu Lin
- Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Peng Wang
- Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Tao Dong
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR 12333, China
| | - Gavin Chit Tsui
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR 12333, China
| | - Saihu Liao
- Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou 350108, China.,State Key Laboratory of Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361005, China
| |
Collapse
|
45
|
Carneiro SN, Khasnavis SR, Lee J, Butler TW, Majmudar JD, Am Ende CW, Ball ND. Sulfur(VI) fluorides as tools in biomolecular and medicinal chemistry. Org Biomol Chem 2023; 21:1356-1372. [PMID: 36662157 PMCID: PMC9929716 DOI: 10.1039/d2ob01891h] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 01/05/2023] [Indexed: 01/21/2023]
Abstract
Recent advances in the synthesis of sulfur(VI)-fluorides has enabled incredible growth in their application in biomolecular chemistry. This review aims to serve as a primer highlighting synthetic strategies toward a diversity of S(VI) fluorides and their application in chemical biology, bioconjugation, and medicinal chemistry.
Collapse
Affiliation(s)
- Sabrina N Carneiro
- Department of Chemistry, Pomona College, Claremont, California 91711, USA.
| | - Samuel R Khasnavis
- Department of Chemistry, Pomona College, Claremont, California 91711, USA.
| | - Jisun Lee
- Pfizer Worldwide Research, Development, Groton, Connecticut 06340, USA.
| | - Todd W Butler
- Pfizer Worldwide Research, Development, Groton, Connecticut 06340, USA.
| | - Jaimeen D Majmudar
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, USA
| | | | - Nicholas D Ball
- Department of Chemistry, Pomona College, Claremont, California 91711, USA.
| |
Collapse
|
46
|
Racioppo B, Qiu N, Adibekian A. Serine Hydrolase Activity‐Based Probes for use in Chemical Proteomics. Isr J Chem 2023. [DOI: 10.1002/ijch.202300016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Affiliation(s)
- Brittney Racioppo
- Department of Chemistry University of Illinois Chicago Chicago Illinois 60607 United States
- Skaggs Doctoral Program in the Chemical and Biological Sciences, Scripps Research La Jolla California 92037 United States
| | - Nan Qiu
- Department of Chemistry University of Illinois Chicago Chicago Illinois 60607 United States
- Skaggs Doctoral Program in the Chemical and Biological Sciences, Scripps Research La Jolla California 92037 United States
| | - Alexander Adibekian
- Department of Chemistry University of Illinois Chicago Chicago Illinois 60607 United States
| |
Collapse
|
47
|
Qin Y, Zhang Z, Ye X, Tan CH. Ion Pair Catalyst - Pentanidinium. CHEM REC 2023:e202200304. [PMID: 36762723 DOI: 10.1002/tcr.202200304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/27/2023] [Indexed: 02/11/2023]
Abstract
In this account, we further describe our already developed N-sp2 hybrid guanidinium as an efficient phase-transfer catalyst and ion pair catalysis based on N-sp2 hybrid pentanidinium and its application in some new reactions. The sp3 hybrid quaternary ammonium salt has a tetrahedral structure, which means that three sides of it can be effectively steric, allowing the remaining side to be close to the substrate. However, the sp2 hybrid ammonium salt allows the substrate to form ion pairs from both directions respectively, so it is a greater challenge to control the stereoselectivity of the reaction. Van der Waals forces, such as hydrogen bonds and π - π ${\pi -\pi }$ interactions, have been used to make electrophiles approach from a certain direction, leading to a higher enantioselectivity. Based on the above idea, we designed an N-sp2 hybrid phase-transfer catalyst, pentanidinium. Pentanidinium has five conjugated nitrogen atoms, one of which has a formal positive charge, which is necessary for it to become an ion pair catalyst. We have confirmed that pentanidinium can catalyze α-hydroxylation of 3-substituted-2-oxindoles, Michael addition of 3-alkyloxindoles with vinyl sulfone, and alkylation reactions of sulfenate anions and dihydrocoumarins, desymmetrization of pro-chiral sulfinate to afford enantioenriched sulfinate esters. Pentanidinium with side chain structure changes can also be catalyzed efficiently with enantioconvergent halogenophilic nucleophilic substitution, including azidation and thioesterification. In the reaction catalyzed by pentanidinium, it always attracts us with the advantages of low catalytic load and good enantioselectivity.
Collapse
Affiliation(s)
- Yimin Qin
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang Province, P.R. China
| | - Zhenqiang Zhang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang Province, P.R. China
| | - Xinyi Ye
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang Province, P.R. China
| | - Choon-Hong Tan
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| |
Collapse
|
48
|
Reineke TM. Bioconjugate Chemistry: Enabling Innovation and Fostering Community at the Nexus of Synthetic and Biological Research. Bioconjug Chem 2023; 34:1-2. [PMID: 36563340 DOI: 10.1021/acs.bioconjchem.2c00591] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
49
|
Zhang X, Qin HL. A General Procedure for the Construction of 2-Alkyl-Substituted Vinyl Sulfonyl Fluoride. Org Lett 2022; 24:9311-9315. [PMID: 36475782 DOI: 10.1021/acs.orglett.2c03936] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A series of compact and multifunctional 2-alkyl-substituted vinyl sulfonyl fluorides were efficiently prepared from the corresponding alkyl iodides and 2-chloroprop-2-ene-1-sulfonyl fluoride (CESF). This Giese-type radical approach provided new and general access to alkenyl sulfonyl fluorides, including structures that would otherwise be challenging to synthesize with previously established methods. A correspondingly large collection of derivatization reactions was also demonstrated on the alkenyl sulfonyl fluorides.
Collapse
Affiliation(s)
- Xu Zhang
- School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Hua-Li Qin
- School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, Hubei 430070, China
| |
Collapse
|
50
|
Bellia S, Teodoro LI, Barbosa AJ, Zeller M, Mirjafari A, Hillesheim PC. Contrasting the Noncovalent Interactions of Aromatic Sulfonyl Fluoride and Sulfonyl Chloride Motifs via Crystallography and Hirshfeld Surfaces. ChemistrySelect 2022; 7:e202203797. [PMID: 36643613 PMCID: PMC9835070 DOI: 10.1002/slct.202203797] [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: 09/28/2022] [Accepted: 11/18/2022] [Indexed: 12/13/2022]
Abstract
A heteroaryl sulfonyl(VI) fluoride, 4-chloro-7-fluorosulfonyl-2,1,3-benzoxadiazole, was synthesized from its chloride counterpart (4-chloro-7-chlorosulfonyl-2,1,3-benzoxadiazole) and the X-ray structure analysis of these compounds and the interactions in the solid-state were thoroughly examined. Hirshfeld surface analysis is used to provide a thorough and complete picture of the changes arising from the different halides in the functional groups. Surface analysis reveals that the fluoride does not participate in any hydrogen interactions as opposed to the chloride. However, the fluorine atom is observed to form close interactions with several π bonds. For both moieties, however, the sulfonyl oxygens show comparable interactions with respect to both magnitude and interatomic distances. The Hirshfeld surface analysis is coupled with computational studies to help elucidate the observed interactions that are found from the distinct nitrogen, chlorine, and oxygen atoms present in the molecules, providing new physical insights to the correlation between their structures and properties.
Collapse
Affiliation(s)
- Sophia Bellia
- Department of Chemistry and Physics, Ave Maria University, 34142 Ave Maria, FL, USA
| | - Lara I Teodoro
- Department of Chemistry and Physics, Ave Maria University, 34142 Ave Maria, FL, USA
| | - Antonio J Barbosa
- Department of Chemistry and Physics, Ave Maria University, 34142 Ave Maria, FL, USA
| | - Matthias Zeller
- Department of Chemistry, Purdue University, 47907 West Lafayette, Indiana, USA
| | - Arsalan Mirjafari
- Department of Chemistry, State University of New York at Oswego, 13126 Oswego, New York, USA
| | - Patrick C Hillesheim
- Department of Chemistry and Physics, Ave Maria University, 34142 Ave Maria, FL, USA
| |
Collapse
|