1
|
Rice AJ, Sword TT, Chengan K, Mitchell DA, Mouncey NJ, Moore SJ, Bailey CB. Cell-free synthetic biology for natural product biosynthesis and discovery. Chem Soc Rev 2025; 54:4314-4352. [PMID: 40104998 PMCID: PMC11920963 DOI: 10.1039/d4cs01198h] [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: 11/28/2024] [Indexed: 03/20/2025]
Abstract
Natural products have applications as biopharmaceuticals, agrochemicals, and other high-value chemicals. However, there are challenges in isolating natural products from their native producers (e.g. bacteria, fungi, plants). In many cases, synthetic chemistry or heterologous expression must be used to access these important molecules. The biosynthetic machinery to generate these compounds is found within biosynthetic gene clusters, primarily consisting of the enzymes that biosynthesise a range of natural product classes (including, but not limited to ribosomal and nonribosomal peptides, polyketides, and terpenoids). Cell-free synthetic biology has emerged in recent years as a bottom-up technology applied towards both prototyping pathways and producing molecules. Recently, it has been applied to natural products, both to characterise biosynthetic pathways and produce new metabolites. This review discusses the core biochemistry of cell-free synthetic biology applied to metabolite production and critiques its advantages and disadvantages compared to whole cell and/or chemical production routes. Specifically, we review the advances in cell-free biosynthesis of ribosomal peptides, analyse the rapid prototyping of natural product biosynthetic enzymes and pathways, highlight advances in novel antimicrobial discovery, and discuss the rising use of cell-free technologies in industrial biotechnology and synthetic biology.
Collapse
Affiliation(s)
- Andrew J Rice
- Department of Biochemistry, School of Medicine - Basic Sciences, Vanderbilt University Medical Research Building-IV, Nashville, Tennessee, 37232, USA
| | - Tien T Sword
- Department of Chemistry, University of Tennessee-Knoxville, Knoxville, TN, USA
| | | | - Douglas A Mitchell
- Department of Biochemistry, School of Medicine - Basic Sciences, Vanderbilt University Medical Research Building-IV, Nashville, Tennessee, 37232, USA
- Department of Chemistry, Vanderbilt University, Medical Research Building-IV, Nashville, Tennessee, 37232, USA
| | - Nigel J Mouncey
- DOE Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, California, 94720, USA
| | - Simon J Moore
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK.
| | - Constance B Bailey
- School of Chemistry, University of Sydney, Camperdown, NSW, 2001, Australia.
| |
Collapse
|
2
|
Petchey MR, Schneider P, Harwood LA. Biocatalytic Amino Acid Functionalisation. ChemMedChem 2025; 20:e202400628. [PMID: 39540701 DOI: 10.1002/cmdc.202400628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/16/2024]
Abstract
The success of new therapeutic modalities relies on advancements in synthetic chemistry to produce compounds for evaluation throughout the drug discovery process. The use of non-canonical amino acids (ncAAs) allows the properties of peptide drugs to be modified and optimised beyond the defined characteristics of the 20 proteogenic amino acids. Synthesis of ncAAs can be either through a bespoke chemical synthesis, or directly from the parent compound - using either traditional chemical reagents or using enzymes - to achieve the desired modification. This review will highlight recent advancements in the enzymatic functionalisation of amino acids to produce a variety of ncAAs.
Collapse
Affiliation(s)
- Mark R Petchey
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Pascal Schneider
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Lucy A Harwood
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
3
|
He J, Yokoi K, Wixted B, Zhang B, Kawamata Y, Renata H, Baran PS. Biocatalytic C-H oxidation meets radical cross-coupling: Simplifying complex piperidine synthesis. Science 2024; 386:1421-1427. [PMID: 39700271 PMCID: PMC11760214 DOI: 10.1126/science.adr9368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/12/2024] [Indexed: 12/21/2024]
Abstract
Modern medicinal chemists are targeting more complex molecules to address challenging biological targets, which leads to synthesizing structures with higher sp3 character (Fsp3) to enhance specificity as well as physiochemical properties. Although traditional flat, high-fraction sp2 molecules, such as pyridine, can be decorated through electrophilic aromatic substitution and palladium (Pd)-based cross-couplings, general strategies to derivatize three-dimensional (3D) saturated molecules are far less developed. In this work, we present an approach for the rapid, modular, enantiospecific, and diastereoselective functionalization of piperidine (saturated analog of pyridine), combining robust biocatalytic carbon-hydrogen oxidation with radical cross-coupling. This combination is directly analogous to electrophilic aromatic substitution followed by Pd-couplings for flat molecules, streamlining synthesis of 3D molecules. This study offers a generalizable strategy for accessing complex architectures, appealing to both medicinal and process chemists.
Collapse
Affiliation(s)
- Jiayan He
- Department of Chemistry, Scripps Research, 10550 North Torrey Pines Road, La Jolla, CA, 92037, United States
| | - Kenta Yokoi
- Department of Chemistry, BioScience Research Collaborative, Rice University, Houston, TX, 77005, United States
| | - Breanna Wixted
- Department of Chemistry, BioScience Research Collaborative, Rice University, Houston, TX, 77005, United States
| | - Benxiang Zhang
- Department of Chemistry, Scripps Research, 10550 North Torrey Pines Road, La Jolla, CA, 92037, United States
| | - Yu Kawamata
- Department of Chemistry, Scripps Research, 10550 North Torrey Pines Road, La Jolla, CA, 92037, United States
| | - Hans Renata
- Department of Chemistry, BioScience Research Collaborative, Rice University, Houston, TX, 77005, United States
| | - Phil S. Baran
- Department of Chemistry, Scripps Research, 10550 North Torrey Pines Road, La Jolla, CA, 92037, United States
| |
Collapse
|
4
|
Ancajas CMF, Oyedele AS, Butt CM, Walker AS. Advances, opportunities, and challenges in methods for interrogating the structure activity relationships of natural products. Nat Prod Rep 2024; 41:1543-1578. [PMID: 38912779 PMCID: PMC11484176 DOI: 10.1039/d4np00009a] [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: 02/27/2024] [Indexed: 06/25/2024]
Abstract
Time span in literature: 1985-early 2024Natural products play a key role in drug discovery, both as a direct source of drugs and as a starting point for the development of synthetic compounds. Most natural products are not suitable to be used as drugs without further modification due to insufficient activity or poor pharmacokinetic properties. Choosing what modifications to make requires an understanding of the compound's structure-activity relationships. Use of structure-activity relationships is commonplace and essential in medicinal chemistry campaigns applied to human-designed synthetic compounds. Structure-activity relationships have also been used to improve the properties of natural products, but several challenges still limit these efforts. Here, we review methods for studying the structure-activity relationships of natural products and their limitations. Specifically, we will discuss how synthesis, including total synthesis, late-stage derivatization, chemoenzymatic synthetic pathways, and engineering and genome mining of biosynthetic pathways can be used to produce natural product analogs and discuss the challenges of each of these approaches. Finally, we will discuss computational methods including machine learning methods for analyzing the relationship between biosynthetic genes and product activity, computer aided drug design techniques, and interpretable artificial intelligence approaches towards elucidating structure-activity relationships from models trained to predict bioactivity from chemical structure. Our focus will be on these latter topics as their applications for natural products have not been extensively reviewed. We suggest that these methods are all complementary to each other, and that only collaborative efforts using a combination of these techniques will result in a full understanding of the structure-activity relationships of natural products.
Collapse
Affiliation(s)
| | | | - Caitlin M Butt
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA.
| | - Allison S Walker
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA.
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
5
|
Schedlbauer A, Han X, van Bakel W, Kaminishi T, Ochoa-Lizarralde B, Iturrioz I, Çapuni R, Parry R, Zegarra R, Gil-Carton D, López-Alonso JP, Barragan Sanz K, Brandi L, Gualerzi CO, Fucini P, Connell SR. A binding site for the antibiotic GE81112 in the ribosomal mRNA channel. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.614503. [PMID: 39386670 PMCID: PMC11463416 DOI: 10.1101/2024.09.26.614503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The initiation phase is the rate-limiting step of protein synthesis (translation) and is finely regulated, making it an important drug target. In bacteria, initiation is guided by three initiation factors and involves positioning the start site on the messenger RNA within the P-site on the small ribosomal subunit (30S), where it is decoded by the initiator tRNA. This process can be efficiently inhibited by GE81112, a natural hydrophilic, noncyclic, nonribosomal tetrapeptide. It is found in nature in three structural variants (A, B and B1 with molecular masses of 643-658 Da). Previous biochemical and structural characterisation of GE81112 indicates that the primary mechanism of action of this antibiotic is to (1) prevent the initiator tRNA from binding correctly to the P-site and (2) block conformational rearrangements in initiation factor IF3, resulting in an unlocked 30S pre/C state. In this study, using cryoEM, we have determined the binding site of GE81112 in initiation complexes (3.2-3.7Å) and on empty ribosomes (2.09 Å). This binding site is within the mRNA channel (E-site) but remote from the binding site of the initiation factors and initiator tRNA. This suggests that it acts allosterically to prevent the initiator tRNA from being locked into place. The binding mode is consistent with previous biochemical studies and recent work identifying the key pharmacophores of GE81112.
Collapse
Affiliation(s)
- Andreas Schedlbauer
- Center for Cooperative Research in Biosciences (CIC bioGUNE),
Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A,
48160 Derio, Spain
| | - Xu Han
- Center for Cooperative Research in Biosciences (CIC bioGUNE),
Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A,
48160 Derio, Spain
- Structural Biology of Cellular Machines Laboratory, Biobizkaia
Health Research Institute, Cruces University Hospital, 48903 Barakaldo, Bizkaia,
Spain
| | - Wouter van Bakel
- Structural Biology of Cellular Machines Laboratory, Biobizkaia
Health Research Institute, Cruces University Hospital, 48903 Barakaldo, Bizkaia,
Spain
| | - Tatsuya Kaminishi
- Center for Cooperative Research in Biosciences (CIC bioGUNE),
Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A,
48160 Derio, Spain
- Graduate School of Medicine, Osaka University, 2-2 Yamadaoka,
Suita, 565-0871 Osaka, Japan
| | - Borja Ochoa-Lizarralde
- Center for Cooperative Research in Biosciences (CIC bioGUNE),
Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A,
48160 Derio, Spain
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque
Country, 48940, Leioa, Spain
| | - Idoia Iturrioz
- Center for Cooperative Research in Biosciences (CIC bioGUNE),
Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A,
48160 Derio, Spain
| | - Retina Çapuni
- Center for Cooperative Research in Biosciences (CIC bioGUNE),
Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A,
48160 Derio, Spain
- Department of Technology and Bussiness, Western Balkans
University, Highway Tiranë-Durrës, KM 7, Kashar, 1001 Tirana, Albania
| | - Ransford Parry
- Research Centre for Experimental Marine Biology and
Biotechnology, Plentzia Marine Station of the University of the Basque Country
(PiE-UPV/EHU), Areatza Bidea, 48620 Plentzia, Basque Country, Spain
| | - Ronny Zegarra
- Research Centre for Experimental Marine Biology and
Biotechnology, Plentzia Marine Station of the University of the Basque Country
(PiE-UPV/EHU), Areatza Bidea, 48620 Plentzia, Basque Country, Spain
| | - David Gil-Carton
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque
Country, 48940, Leioa, Spain
- Basque Resource for Electron Microscopy, 48940, Leioa,
Spain
- IKERBASQUE, Basque Foundation for Science, 48011 Bilbao,
Spain
| | - Jorge P. López-Alonso
- Center for Cooperative Research in Biosciences (CIC bioGUNE),
Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A,
48160 Derio, Spain
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque
Country, 48940, Leioa, Spain
- Basque Resource for Electron Microscopy, 48940, Leioa,
Spain
| | - Kristina Barragan Sanz
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque
Country, 48940, Leioa, Spain
- Basque Resource for Electron Microscopy, 48940, Leioa,
Spain
| | - Letizia Brandi
- Laboratory of Genetics, Department of Biosciences and Veterinary
Medicine, University of Camerino, 62032 Camerino, Italy
| | - Claudio O. Gualerzi
- Laboratory of Genetics, Department of Biosciences and Veterinary
Medicine, University of Camerino, 62032 Camerino, Italy
| | - Paola Fucini
- Center for Cooperative Research in Biosciences (CIC bioGUNE),
Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A,
48160 Derio, Spain
- Research Centre for Experimental Marine Biology and
Biotechnology, Plentzia Marine Station of the University of the Basque Country
(PiE-UPV/EHU), Areatza Bidea, 48620 Plentzia, Basque Country, Spain
- IKERBASQUE, Basque Foundation for Science, 48011 Bilbao,
Spain
| | - Sean R. Connell
- Center for Cooperative Research in Biosciences (CIC bioGUNE),
Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A,
48160 Derio, Spain
- Structural Biology of Cellular Machines Laboratory, Biobizkaia
Health Research Institute, Cruces University Hospital, 48903 Barakaldo, Bizkaia,
Spain
- IKERBASQUE, Basque Foundation for Science, 48011 Bilbao,
Spain
| |
Collapse
|
6
|
Fernandes GFS, Kim SH, Castagnolo D. Harnessing biocatalysis as a green tool in antibiotic synthesis and discovery. RSC Adv 2024; 14:30396-30410. [PMID: 39318457 PMCID: PMC11420778 DOI: 10.1039/d4ra04824e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024] Open
Abstract
Biocatalysis offers a sustainable approach to drug synthesis, leveraging the high selectivity and efficiency of enzymes. This review explores the application of biocatalysis in the early-stage synthesis of antimicrobial compounds, emphasizing its advantages over traditional chemical methods. We discuss various biocatalysts, including enzymes and whole-cell systems, and their role in the selective functionalization and preparation of antimicrobials and antibacterial building blocks. The review underscores the potential of biocatalysis to advance the development of new antibiotics and suggests directions and potential applications of enzymes in drug development.
Collapse
Affiliation(s)
- Guilherme F S Fernandes
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
- School of Pharmacy, University College London 29-39 Brunswick Square London WC1N 1AX UK
| | - Seong-Heun Kim
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London 150 Stamford Street London SE1 9NH UK
| | - Daniele Castagnolo
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| |
Collapse
|
7
|
Chao TH, Renata H. Chemoenzymatic Synthesis of 4,5-Dihydroxyisoleucine Fragment of α-Amanitin. Org Lett 2024; 26:3263-3266. [PMID: 38598422 DOI: 10.1021/acs.orglett.4c00901] [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: 04/12/2024]
Abstract
The ability of α-amanitin to potently inhibit RNA polymerase II (RNAP II) has elicited further research into its use as a novel payload for antibody-drug conjugates. Despite this promise, the de novo synthesis of α-amanitin is still a major challenge as it possesses an unusual bicyclic octapeptide structure that contains several oxidized amino acids, most notably 4,5-dihydroxy-l-isoleucine. Here, we report a concise chemoenzymatic synthesis of this key amino acid residue, which features two regioselective and diastereoselective enzymatic C-H oxidations on l-isoleucine.
Collapse
Affiliation(s)
- Tsung-Han Chao
- Department of Chemistry, BioScience Research Collaborative, Rice University, Houston, Texas 77005, United States
| | - Hans Renata
- Department of Chemistry, BioScience Research Collaborative, Rice University, Houston, Texas 77005, United States
| |
Collapse
|
8
|
Bader CD, Nichols AL, Yang D, Shen B. Interplay of emerging and established technologies drives innovation in natural product antibiotic discovery. Curr Opin Microbiol 2023; 75:102359. [PMID: 37517368 PMCID: PMC11623919 DOI: 10.1016/j.mib.2023.102359] [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: 03/02/2023] [Revised: 05/04/2023] [Accepted: 06/21/2023] [Indexed: 08/01/2023]
Abstract
A continued rise of antibiotic resistance and shortages of effective antibiotics necessitate the discovery and development of new antibiotics with novel modes of action (MoAs) against resistant pathogens. While natural products remain the best resource for antibiotic discovery, their exploration faces many challenges, including (i) unknown MoAs, (ii) high rediscovery rates, (iii) tedious isolation and structure elucidation, and (iv) insufficient production for further development. We have identified recent innovations in screening methods, microbiology, bioinformatics, and metabolomics technologies, as well as natural product-inspired synthesis and synthetic biology, that have contributed to new natural product antibiotics in the past two years. We highlight their interplay as the key element for successful applications, driving future opportunities to increase the pool of natural product-based antibacterial antibiotics.
Collapse
Affiliation(s)
- Chantal D Bader
- Department of Chemistry, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, Florida 33458, United States
| | - Angela L Nichols
- Department of Chemistry, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, Florida 33458, United States; Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, Jupiter, Florida 33458, United States
| | - Dong Yang
- Department of Chemistry, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, Florida 33458, United States; Natural Products Discovery Center, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, Florida 33458, United States
| | - Ben Shen
- Department of Chemistry, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, Florida 33458, United States; Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, Florida 33458, United States; Natural Products Discovery Center, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, Florida 33458, United States; Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, Jupiter, Florida 33458, United States.
| |
Collapse
|
9
|
Abstract
The ability to site-selectively modify equivalent functional groups in a molecule has the potential to streamline syntheses and increase product yields by lowering step counts. Enzymes catalyze site-selective transformations throughout primary and secondary metabolism, but leveraging this capability for non-native substrates and reactions requires a detailed understanding of the potential and limitations of enzyme catalysis and how these bounds can be extended by protein engineering. In this review, we discuss representative examples of site-selective enzyme catalysis involving functional group manipulation and C-H bond functionalization. We include illustrative examples of native catalysis, but our focus is on cases involving non-native substrates and reactions often using engineered enzymes. We then discuss the use of these enzymes for chemoenzymatic transformations and target-oriented synthesis and conclude with a survey of tools and techniques that could expand the scope of non-native site-selective enzyme catalysis.
Collapse
Affiliation(s)
- Dibyendu Mondal
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Harrison M Snodgrass
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Christian A Gomez
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Jared C Lewis
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| |
Collapse
|
10
|
Wang J, Wang Y, Wu Q, Zhang Y. Multidimensional engineering of Escherichia coli for efficient biosynthesis of cis-3-hydroxypipecolic acid. BIORESOURCE TECHNOLOGY 2023; 382:129173. [PMID: 37187331 DOI: 10.1016/j.biortech.2023.129173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 05/17/2023]
Abstract
Cis-3-hydroxypipecolic acid (cis-3-HyPip) is the crucial part of many alkaloids and drugs. However, its bio-based industrial production remains challenging. Here, lysine cyclodeaminase from Streptomyces malaysiensis (SmLCD) and pipecolic acid hydroxylase from Streptomyces sp. L-49973 (StGetF) were screened to achieve the conversion of L-lysine to cis-3-HyPip. Considering the high-cost of cofactors, NAD(P)H oxidase from Lactobacillus sanfranciscensis (LsNox) was further overexpressed in chassis strain Escherichia coli W3110 ΔsucCD (α-ketoglutarate-producing strain) to construct the NAD+ regeneration system, thus realizing the bioconversion of cis-3-HyPip from low-cost substrate L-lysine without NAD+ and α-ketoglutarate addition. To further accelerate the transmission efficiency of cis-3-HyPip biosynthetic pathway, multiple-enzyme expression optimization and transporter dynamic regulation via promoter engineering were conducted. Through fermentation optimization, the final engineered strain HP-13 generated 78.4 g/L cis-3-HyPip with 78.9% conversion in a 5-L fermenter, representing the highest production level achieved so far. These strategies described herein show promising potentials for large-scale production of cis-3-HyPip.
Collapse
Affiliation(s)
- Jiaping Wang
- Hangzhou Wahaha Group Co. Ltd., Hangzhou 310018, China; Hangzhou Wahaha Technology Co. Ltd., Hangzhou 310018, China; Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou 310018, China
| | - Yaqiong Wang
- Hangzhou Wahaha Group Co. Ltd., Hangzhou 310018, China; Hangzhou Wahaha Technology Co. Ltd., Hangzhou 310018, China; Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou 310018, China
| | - Qin Wu
- Hangzhou Wahaha Group Co. Ltd., Hangzhou 310018, China; Hangzhou Wahaha Technology Co. Ltd., Hangzhou 310018, China; Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou 310018, China
| | - Yimin Zhang
- Hangzhou Wahaha Group Co. Ltd., Hangzhou 310018, China; Hangzhou Wahaha Technology Co. Ltd., Hangzhou 310018, China; Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou 310018, China.
| |
Collapse
|
11
|
Fayad S, Jafari A, Schuler SMM, Kurz M, Plettenburg O, Hammann PE, Bauer A, Jürjens G, Pöverlein C. Total Synthesis of GE81112A: An Orthoester-Based Approach. J Org Chem 2023; 88:5597-5608. [PMID: 37023463 PMCID: PMC10167690 DOI: 10.1021/acs.joc.3c00094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
The GE81112 series, consisting of three naturally occurring tetrapeptides and synthetic derivatives, is evaluated as a potential lead structure for the development of a new antibacterial drug. Although the first total synthesis of GE81112A reported by our group provided sufficient amounts of material for an initial in depth biological profiling of the compound, improvements of the routes toward the key building blocks were needed for further upscaling and structure-activity relationship studies. The major challenges identified were poor stereoselectivity in the synthesis of the C-terminal β-hydroxy histidine intermediate and a concise access to all four isomers of the 3-hydroxy pipecolic acid. Herein, we report a second-generation synthesis of GE81112A, which is also applicable to access further representatives of this series. Based on Lajoie's ortho-ester-protected serine aldehydes as key building blocks, the described route provides both a satisfactory improvement in stereoselectivity of the β-hydroxy histidine intermediate synthesis and a stereoselective approach toward both orthogonally protected cis and trans-3-hydroxy pipecolic acid.
Collapse
Affiliation(s)
- Scherin Fayad
- Sanofi-Aventis Deutschland GmbH, R&D, Integrated Drug Discovery, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
- Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Institut für Medizinalchemie, 30167 Hannover, Germany
| | - Ardalan Jafari
- Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Institut für Medizinalchemie, 30167 Hannover, Germany
| | - Sören M M Schuler
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Bioresources, 35392 Giessen, Germany
- Evotec International GmbH, 37079 Göttingen, Germany
| | - Michael Kurz
- Sanofi-Aventis Deutschland GmbH, R&D, Integrated Drug Discovery, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Oliver Plettenburg
- Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Institut für Medizinalchemie, 30167 Hannover, Germany
| | - Peter E Hammann
- Evotec International GmbH, 37079 Göttingen, Germany
- Sanofi-Aventis Deutschland GmbH, R&D, Infectious Diseases, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Armin Bauer
- Sanofi-Aventis Deutschland GmbH, R&D, Integrated Drug Discovery, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Gerrit Jürjens
- Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Institut für Medizinalchemie, 30167 Hannover, Germany
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Bioresources, 35392 Giessen, Germany
| | - Christoph Pöverlein
- Sanofi-Aventis Deutschland GmbH, R&D, Integrated Drug Discovery, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| |
Collapse
|
12
|
Zwick CR, Renata H. Overview of Amino Acid Modifications by Iron- and α-Ketoglutarate-Dependent Enzymes. ACS Catal 2023. [DOI: 10.1021/acscatal.3c00424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
13
|
Wu L, An J, Jing X, Chen CC, Dai L, Xu Y, Liu W, Guo RT, Nie Y. Molecular Insights into the Regioselectivity of the Fe(II)/2-Ketoglutarate-Dependent Dioxygenase-Catalyzed C–H Hydroxylation of Amino Acids. ACS Catal 2022. [DOI: 10.1021/acscatal.2c03106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Lunjie Wu
- Laboratory of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jianhong An
- Laboratory of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Ophthalmology and Optometry, and Eye Hospital, Wenzhou Medical University, State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang 325000, China
| | - Xiaoran Jing
- Laboratory of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Chun-Chi Chen
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Longhai Dai
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Yan Xu
- Laboratory of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Weidong Liu
- Industrial Enzymes National Engineering Laboratory, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Rey-Ting Guo
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Yao Nie
- Laboratory of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi, Jiangsu 214122, China
- Suqian Industrial Technology Research Institute of Jiangnan University, Suqian, Jiangsu 223814, China
| |
Collapse
|
14
|
Chemo-enzymatic synthesis of natural products and their analogs. Curr Opin Biotechnol 2022; 77:102759. [PMID: 35908314 DOI: 10.1016/j.copbio.2022.102759] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 11/20/2022]
Abstract
Enzymes continue to gain recognition as valuable tools in synthetic chemistry as they enable transformations, which elude conventional organochemical approaches. As such, the progressing expansion of the biocatalytic arsenal has introduced unprecedented opportunities for new synthetic strategies and retrosynthetic disconnections. As a result, enzymes have found a solid foothold in modern natural product synthesis for applications ranging from the generation of early chiral synthons to endgame transformations, convergent synthesis, and cascade reactions for the rapid construction of molecular complexity. As a primer to the state-of-the-art concerning strategic uses of enzymes in natural product synthesis and the underlying concepts, this review highlights selected recent literature examples, which make a strong case for the admission of enzymatic methodologies into the standard repertoire for complex small-molecule synthesis.
Collapse
|
15
|
Hu S, Li Y, Zhang A, Li H, Chen K, Ouyang P. Designing of an Efficient Whole-Cell Biocatalyst System for Converting L-Lysine Into Cis-3-Hydroxypipecolic Acid. Front Microbiol 2022; 13:945184. [PMID: 35832817 PMCID: PMC9271919 DOI: 10.3389/fmicb.2022.945184] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/03/2022] [Indexed: 11/30/2022] Open
Abstract
Cis-3-hydroxypipecolic acid (cis-3-HyPip), a key structural component of tetrapeptide antibiotic GE81112, which has attracted substantial attention for its broad antimicrobial properties and unique ability to inhibit bacterial translation initiation. In this study, a combined strategy to increase the productivity of cis-3-HyPip was investigated. First, combinatorial optimization of the ribosomal binding site (RBS) sequence was performed to tune the gene expression translation rates of the pathway enzymes. Next, in order to reduce the addition of the co-substrate α-ketoglutarate (2-OG), the major engineering strategy was to reconstitute the tricarboxylic acid (TCA) cycle of Escherichia coli to force the metabolic flux to go through GetF catalyzed reaction for 2-OG to succinate conversion, a series of engineered strains were constructed by the deletion of the relevant genes. In addition, the metabolic flux (gltA and icd) was improved and glucose concentrations were optimized to enhance the supply and catalytic efficiency of continuous 2-OG supply powered by glucose. Finally, under optimal conditions, the cis-3-HyPip titer of the best strain catalysis reached 33 mM, which was remarkably higher than previously reported.
Collapse
|
16
|
Charlton SN, Hayes MA. Oxygenating Biocatalysts for Hydroxyl Functionalisation in Drug Discovery and Development. ChemMedChem 2022; 17:e202200115. [PMID: 35385205 PMCID: PMC9323455 DOI: 10.1002/cmdc.202200115] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/05/2022] [Indexed: 11/12/2022]
Abstract
C-H oxyfunctionalisation remains a distinct challenge for synthetic organic chemists. Oxygenases and peroxygenases (grouped here as "oxygenating biocatalysts") catalyse the oxidation of a substrate with molecular oxygen or hydrogen peroxide as oxidant. The application of oxygenating biocatalysts in organic synthesis has dramatically increased over the last decade, producing complex compounds with potential uses in the pharmaceutical industry. This review will focus on hydroxyl functionalisation using oxygenating biocatalysts as a tool for drug discovery and development. Established oxygenating biocatalysts, such as cytochrome P450s and flavin-dependent monooxygenases, have widely been adopted for this purpose, but can suffer from low activity, instability or limited substrate scope. Therefore, emerging oxygenating biocatalysts which offer an alternative will also be covered, as well as considering the ways in which these hydroxylation biotransformations can be applied in drug discovery and development, such as late-stage functionalisation (LSF) and in biocatalytic cascades.
Collapse
Affiliation(s)
- Sacha N. Charlton
- School of ChemistryUniversity of Bristol, Cantock's CloseBristolBS8 1TSUK
| | - Martin A. Hayes
- Compound Synthesis and ManagementDiscovery SciencesBiopharmaceuticals R&DAstraZenecaGothenburgSweden
| |
Collapse
|
17
|
Neis N, Xie F, Krug D, Zhao H, Siebert A, Binz T, Fu C, Müller R, Kazmaier U. Stereoselective Syntheses of Deuterated Pipecolic Acids as Tools to Investigate the Stereoselectivity of the Hydroxylase GetF. European J Org Chem 2022. [DOI: 10.1002/ejoc.202200162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Nadine Neis
- Saarland University: Universitat des Saarlandes Organic Chemistry GERMANY
| | - Feng Xie
- Helmholtz Centre for Infection Research: Helmholtz-Zentrum fur Infektionsforschung GmbH Microbial Natural Products GERMANY
| | - Daniel Krug
- Helmholtz Centre for Infection Research: Helmholtz-Zentrum fur Infektionsforschung GmbH Microbial Natural Products GERMANY
| | - Haowen Zhao
- Helmholtz Centre for Infection Research: Helmholtz-Zentrum fur Infektionsforschung GmbH Microbial Natural Products GERMANY
| | - Andreas Siebert
- Saarland University: Universitat des Saarlandes Organic Chemistry GERMANY
| | - Tina Binz
- Helmholtz-Zentrum für Infektionsforschung GmbH: Helmholtz-Zentrum fur Infektionsforschung GmbH Microbial Natural Products GERMANY
| | - Chengzhang Fu
- Helmholtz-Zentrum für Infektionsforschung GmbH: Helmholtz-Zentrum fur Infektionsforschung GmbH Microbial Natural Products GERMANY
| | - Rolf Müller
- Helmholtz-Zentrum für Infektionsforschung GmbH: Helmholtz-Zentrum fur Infektionsforschung GmbH Microbial Natural Products GERMANY
| | - Uli Kazmaier
- Saarland University Organic Chemistry Campus, Bldg. C4.2 66123 Saarbrücken GERMANY
| |
Collapse
|
18
|
Doyon TJ, Kumar P, Thein S, Kim M, Stitgen A, Grieger AM, Madigan C, Willoughby PH, Buller AR. Scalable and Selective β-Hydroxy-α-Amino Acid Synthesis Catalyzed by Promiscuous l-Threonine Transaldolase ObiH. Chembiochem 2022; 23:e202100577. [PMID: 34699683 PMCID: PMC8796315 DOI: 10.1002/cbic.202100577] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Indexed: 01/21/2023]
Abstract
Enzymes from secondary metabolic pathways possess broad potential for the selective synthesis of complex bioactive molecules. However, the practical application of these enzymes for organic synthesis is dependent on the development of efficient, economical, operationally simple, and well-characterized systems for preparative scale reactions. We sought to bridge this knowledge gap for the selective biocatalytic synthesis of β-hydroxy-α-amino acids, which are important synthetic building blocks. To achieve this goal, we demonstrated the ability of ObiH, an l-threonine transaldolase, to achieve selective milligram-scale synthesis of a diverse array of non-standard amino acids (nsAAs) using a scalable whole cell platform. We show how the initial selectivity of the catalyst is high and how the diastereomeric ratio of products decreases at high conversion due to product re-entry into the catalytic cycle. ObiH-catalyzed reactions with a variety of aromatic, aliphatic and heterocyclic aldehydes selectively generated a panel of β-hydroxy-α-amino acids possessing broad functional-group diversity. Furthermore, we demonstrated that ObiH-generated β-hydroxy-α-amino acids could be modified through additional transformations to access important motifs, such as β-chloro-α-amino acids and substituted α-keto acids.
Collapse
Affiliation(s)
- Tyler J. Doyon
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, United States
| | - Prasanth Kumar
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, United States
| | - Sierra Thein
- Department of Chemistry, Ripon College, Ripon, WI 54971, United States
| | - Maeve Kim
- Department of Chemistry, Ripon College, Ripon, WI 54971, United States
| | - Abigail Stitgen
- Department of Chemistry, Ripon College, Ripon, WI 54971, United States
| | | | - Cormac Madigan
- Department of Chemistry, Ripon College, Ripon, WI 54971, United States
| | | | - Andrew R. Buller
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, United States
| |
Collapse
|
19
|
Hu S, Yang P, Li Y, Zhang A, Chen K, Ouyang P. Biosynthesis of cis-3-hydroxypipecolic acid from L-lysine using an in vivo dual-enzyme cascade. Enzyme Microb Technol 2021; 154:109958. [PMID: 34891103 DOI: 10.1016/j.enzmictec.2021.109958] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 11/13/2021] [Accepted: 11/30/2021] [Indexed: 11/03/2022]
Abstract
Cis-3-Hydroxypipecolic acid (cis-3-HyPip) is an important intermediate for the synthesis of GE81112 tetrapeptides, a small family of unusual nonribosomal peptide congeners with potent inhibitory activity against prokaryotic translation initiation. In this study, we constructed a microbial cell factory that can convert L-lysine into cis-3-hydroxypipecolic acid (cis-3-HyPip). Lysine cyclodeaminase SpLCD and Fe(II)/α-ketoglutarate (2-OG)-based oxygenase GetF were co-expressed in Escherichia coli. Plasmids with different copy numbers were used to balance the expression of these two enzymes, and the cell with the most appropriate balance of this kind for carrying plasmid pET-duet-getf-splcd was obtained. After determining the temperature (30 °C), pH (7.0), cell biomass, substrate concentration, Fe2+ concentration (10 mM), L-ascorbate concentration (10 mM), and TritonX-100 concentration (0.1% w/v) that were optimal for whole-cell catalysis, the yield of cis-3-HyPip reached as high as 25 mM (3.63 g/L).
Collapse
Affiliation(s)
- Shewei Hu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211800, PR China
| | - Pengfan Yang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211800, PR China
| | - Yangyang Li
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211800, PR China
| | - Alei Zhang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211800, PR China.
| | - Kequan Chen
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211800, PR China.
| | - Pingkai Ouyang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211800, PR China
| |
Collapse
|
20
|
Parker PD, Hou X, Dong VM. Reducing Challenges in Organic Synthesis with Stereoselective Hydrogenation and Tandem Catalysis. J Am Chem Soc 2021; 143:6724-6745. [PMID: 33891819 DOI: 10.1021/jacs.1c00750] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tandem catalysis enables the rapid construction of complex architectures from simple building blocks. This Perspective shares our interest in combining stereoselective hydrogenation with transformations such as isomerization, oxidation, and epimerization to solve diverse challenges. We highlight the use of tandem hydrogenation for preparing complex natural products from simple prochiral building blocks and present tandem catalysis involving transfer hydrogenation and dynamic kinetic resolution. Finally, we underline recent breakthroughs and opportunities for asymmetric hydrogenation.
Collapse
Affiliation(s)
- Patrick D Parker
- Department of Chemistry, University of California, Irvine, California 92697, United States
| | - Xintong Hou
- Department of Chemistry, University of California, Irvine, California 92697, United States
| | - Vy M Dong
- Department of Chemistry, University of California, Irvine, California 92697, United States
| |
Collapse
|
21
|
Stout CN, Renata H. Reinvigorating the Chiral Pool: Chemoenzymatic Approaches to Complex Peptides and Terpenoids. Acc Chem Res 2021; 54:1143-1156. [PMID: 33543931 DOI: 10.1021/acs.accounts.0c00823] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Biocatalytic transformations that leverage the selectivity and efficiency of enzymes represent powerful tools for the construction of complex natural products. Enabled by innovations in genome mining, bioinformatics, and enzyme engineering, synthetic chemists are now more than ever able to develop and employ enzymes to solve outstanding chemical problems, one of which is the reliable and facile generation of stereochemistry within natural product scaffolds. In recognition of this unmet need, our group has sought to advance novel chemoenzymatic strategies to both expand and reinvigorate the chiral pool. Broadly defined, the chiral pool comprises cheap, enantiopure feedstock chemicals that serve as popular foundations for asymmetric total synthesis. Among these building blocks, amino acids and enantiopure terpenes, whose core structures can be mapped onto several classes of structurally and pharmaceutically intriguing natural products, are of particular interest to the synthetic community.In this Account, we summarize recent efforts from our group in leveraging biocatalytic transformations to expand the chiral pool, as well as efforts toward the efficient application of these transformations in natural products total synthesis, the ultimate testing ground for any novel methodology. First, we describe several examples of enzymatic generation of noncanonical amino acids as means to simplify the synthesis of peptide natural products. By extracting amino acid hydroxylases from native biosynthetic pathways, we obtain efficient access to hydroxylated variants of proline, lysine, arginine, and their derivatives. The newly installed hydroxyl moiety then becomes a chemical handle that can facilitate additional complexity generation, thereby expanding the pool of amino acid-derived building blocks available for peptide synthesis. Next, we present our efforts in enzymatic C-H oxidations of diverse terpene scaffolds, in which traditional chemistry can be combined with strategic applications of biocatalysis to selectively and efficiently derivatize several commercial terpenoid skeletons. The synergistic logic of this approach enables a small handful of synthetic intermediates to provide access to a plethora of terpenoid natural product families. Taken together, these findings demonstrate the advantages of applying enzymes in total synthesis in conjunction with established methodologies, as well as toward the expansion of the chiral pool to enable facile incorporation of stereochemistry during synthetic campaigns.
Collapse
Affiliation(s)
- Carter N. Stout
- Department of Chemistry, Scripps Research, 110 Scripps Way, Jupiter, Florida 33458, United States
| | - Hans Renata
- Department of Chemistry, Scripps Research, 110 Scripps Way, Jupiter, Florida 33458, United States
| |
Collapse
|
22
|
Hill RA, Sutherland A. Hot off the press. Nat Prod Rep 2021. [DOI: 10.1039/d1np90013g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A personal selection of 32 recent papers is presented covering various aspects of current developments in bioorganic chemistry and novel natural products such as eurysoloid A from Eurysolen gracilis.
Collapse
Affiliation(s)
- Robert A. Hill
- School of Chemistry, Glasgow University, Glasgow, G12 8QQ, UK
| | | |
Collapse
|
23
|
Chakrabarty S, Wang Y, Perkins JC, Narayan ARH. Scalable biocatalytic C-H oxyfunctionalization reactions. Chem Soc Rev 2020; 49:8137-8155. [PMID: 32701110 PMCID: PMC8177087 DOI: 10.1039/d0cs00440e] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Catalytic C-H oxyfunctionalization reactions have garnered significant attention in recent years with their ability to streamline synthetic routes toward complex molecules. Consequently, there have been significant strides in the design and development of catalysts that enable diversification through C-H functionalization reactions. Enzymatic C-H oxygenation reactions are often complementary to small molecule based synthetic approaches, providing a powerful tool when deployable on preparative-scale. This review highlights key advances in scalable biocatalytic C-H oxyfunctionalization reactions developed within the past decade.
Collapse
Affiliation(s)
- Suman Chakrabarty
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| | | | | | | |
Collapse
|
24
|
Kanda Y, Nakamura H, Umemiya S, Puthukanoori RK, Murthy Appala VR, Gaddamanugu GK, Paraselli BR, Baran PS. Two-Phase Synthesis of Taxol. J Am Chem Soc 2020; 142:10526-10533. [PMID: 32406238 PMCID: PMC8349513 DOI: 10.1021/jacs.0c03592] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Taxol (a brand name for paclitaxel) is widely regarded as among the most famed natural isolates ever discovered, and has been the subject of innumerable studies in both basic and applied science. Its documented success as an anticancer agent, coupled with early concerns over supply, stimulated a furious worldwide effort from chemists to provide a solution for its preparation through total synthesis. Those pioneering studies proved the feasibility of retrosynthetically guided access to synthetic Taxol, albeit in minute quantities and with enormous effort. In practice, all medicinal chemistry efforts and eventual commercialization have relied upon natural (plant material) or biosynthetically derived (synthetic biology) supplies. Here we show how a complementary divergent synthetic approach that is holistically patterned off of biosynthetic machinery for terpene synthesis can be used to arrive at Taxol.
Collapse
Affiliation(s)
- Yuzuru Kanda
- Department of Chemistry, Scripps Research, 10550 North Torrey Pines Road, La Jolla, CA 92037, United States
| | - Hugh Nakamura
- Department of Chemistry, Scripps Research, 10550 North Torrey Pines Road, La Jolla, CA 92037, United States
| | - Shigenobu Umemiya
- Department of Chemistry, Scripps Research, 10550 North Torrey Pines Road, La Jolla, CA 92037, United States
| | - Ravi Kumar Puthukanoori
- Chemveda Life Sciences, Pvt. Ltd., Plot No. B – 11/1, IDA Uppal, Hyderabad, Telangana 500039, India
| | | | - Gopi Krishna Gaddamanugu
- Chemveda Life Sciences, Pvt. Ltd., Plot No. B – 11/1, IDA Uppal, Hyderabad, Telangana 500039, India
| | - Bheema Rao Paraselli
- Chemveda Life Sciences, Inc., 9920 Pacific Heights Blvd, Suite 150, San Diego, CA 92121, United States
| | - Phil S. Baran
- Department of Chemistry, Scripps Research, 10550 North Torrey Pines Road, La Jolla, CA 92037, United States
| |
Collapse
|