Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Acc Chem Res ; 54(8): 1891-1908, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33792282

ABSTRACT

Continued, rapid development of antimicrobial resistance has become worldwide health crisis and a burden on the global economy. Decisive and comprehensive action is required to slow down the spread of antibiotic resistance, including increased investment in antibiotic discovery, sustainable policies that provide returns on investment for newly launched antibiotics, and public education to reduce the overusage of antibiotics, especially in livestock and agriculture. Without significant changes in the current antibiotic pipeline, we are in danger of entering a post-antibiotic era.In this Account, we summarize our recent efforts to develop next-generation streptogramin and lankacidin antibiotics that overcome bacterial resistance by means of modular chemical synthesis. First, we describe our highly modular, scalable route to four natural group A streptogramins antibiotics in 6-8 steps from seven simple chemical building blocks. We next describe the application of this route to the synthesis of a novel library of streptogramin antibiotics informed by in vitro and in vivo biological evaluation and high-resolution cryo-electron microscopy. One lead compound showed excellent inhibitory activity in vitro and in vivo against a longstanding streptogramin-resistance mechanism, virginiamycin acetyltransferase. Our results demonstrate that the combination of rational design and modular chemical synthesis can revitalize classes of antibiotics that are limited by naturally arising resistance mechanisms.Second, we recount our modular approaches toward lankacidin antibiotics. Lankacidins are a group of polyketide natural products with activity against several strains of Gram-positive bacteria but have not been deployed as therapeutics due to their chemical instability. We describe a route to several diastereomers of 2,18-seco-lankacidinol B in a linear sequence of ≤8 steps from simple building blocks, resulting in a revision of the C4 stereochemistry. We next detail our modular synthesis of several diastereoisomers of iso-lankacidinol that resulted in the structural reassignment of this natural product. These structural revisions raise interesting questions about the biosynthetic origin of lankacidins, all of which possessed uniform stereochemistry prior to these findings. Finally, we summarize the ability of several iso- and seco-lankacidins to inhibit the growth of bacteria and to inhibit translation in vitro, providing important insights into structure-function relationships for the class.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Macrolides/chemical synthesis , Streptogramins/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Escherichia coli/metabolism , Gram-Negative Bacteria , Gram-Positive Bacteria/drug effects , Macrolides/chemistry , Macrolides/pharmacology , Microbial Sensitivity Tests , Molecular Conformation , Molecular Dynamics Simulation , Ribosomes/chemistry , Ribosomes/metabolism , Streptogramins/chemistry , Streptogramins/pharmacology , Virginiamycin/analogs & derivatives , Virginiamycin/chemical synthesis , Virginiamycin/metabolism , Virginiamycin/pharmacology
2.
J Am Chem Soc ; 139(38): 13304-13307, 2017 09 27.
Article in English | MEDLINE | ID: mdl-28902996

ABSTRACT

Streptogramin antibiotics are used clinically to treat multidrug-resistant bacterial infections, but their poor physicochemical properties and narrow spectra of activity have limited their utility. New methods to chemically modify streptogramins would enable structural optimization to overcome these limitations as well as to combat growing resistance to the class. Here we report a modular, scalable synthesis of group A streptogramin antibiotics that proceeds in 6-8 linear steps from simple chemical building blocks. We have applied our route to the synthesis of four natural products in this class including two that have never before been accessed by fully synthetic routes. We anticipate that this work will lead to the discovery of new streptogramin antibiotics that overcome previous limitations of the class.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Streptogramin Group A/chemical synthesis , Anti-Bacterial Agents/chemistry , Biological Products/chemical synthesis , Biological Products/chemistry , Molecular Structure , Streptogramin Group A/chemistry , Virginiamycin/chemical synthesis , Virginiamycin/chemistry
3.
J Org Chem ; 76(24): 9900-18, 2011 Dec 16.
Article in English | MEDLINE | ID: mdl-22070230

ABSTRACT

A stereoselective synthesis of the antibiotic (-)-virginiamycin M(2) is detailed. A convergent strategy was utilized that proceeded in 10 steps (longest linear sequence) from enantioenriched silane (S)-15. This reagent, which was prepared via a Rh(II)- or Cu(I)-catalyzed carbenoid Si-H insertion, was used to introduce the desired olefin geometry and stereocenters of the C1-C5 propionate subunit. A modified Negishi cross-coupling or an efficient alkoxide-directed titanium-mediated alkyne-alkyne reductive coupling strategy was utilized to assemble the trisubstituted (E,E)-diene. An underutilized late-stage SmI(2)-mediated macrocyclization was employed to construct the 23-membered macrocycle scaffold of the natural product.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Copper/chemistry , Rhodium/chemistry , Silanes/chemistry , Silicon/chemistry , Virginiamycin/analogs & derivatives , Alkenes/chemistry , Alkynes/chemistry , Catalysis , Chromatography, High Pressure Liquid , Cyclization , Humans , Hydrogen/chemistry , Magnetic Resonance Spectroscopy , Molecular Structure , Oxidation-Reduction , Stereoisomerism , Virginiamycin/chemical synthesis
4.
Org Lett ; 9(16): 3105-8, 2007 Aug 02.
Article in English | MEDLINE | ID: mdl-17608433

ABSTRACT

A de novo approach to the formal total synthesis of the macrolide natural product (-)-virginiamycin M2 has been achieved via a convergent approach. The absolute and relative stereochemistry of the nonpeptide portion of (-)-virginiamycin M2 was introduced by two Sharpless asymmetric dihydroxylation reactions.


Subject(s)
Macrolides/chemical synthesis , Virginiamycin/analogs & derivatives , Virginiamycin/chemical synthesis , Catalysis , Macrolides/chemistry , Models, Molecular , Molecular Structure , Stereoisomerism , Virginiamycin/chemistry
6.
Curr Pharm Des ; 4(2): 155-80, 1998 Apr.
Article in English | MEDLINE | ID: mdl-10197038

ABSTRACT

The streptogramins are a class of antibiotics remarkable for their antibacterial activity and their unique mechanism of action. These antibiotics are produced naturally, but the therapeutic use of the natural compounds is limited because they do not dissolve in water. New semisynthetic derivatives, in particular the injectable streptogramin quinupristin/dalfopristin, offer promise for treating the rising number of infections that are caused by multiply resistant bacteria. The streptogramins consist of two structurally unrelated compounds, group A and group B. The group A compounds are polyunsaturated macrolactones: the group B compounds are cyclic hexadepsipeptides. Modifications of the group B components have been mainly performed on the 3-hydroxypicolinoyl, the 4-dimethylaminophenylalanine and the 4-oxo pipecolinic residues. Semi-synthesis on this third residue led to the water-soluble derivative quinupristin. Water-soluble group A derivatives were obtained by Michael addition of aminothiols to the dehydroproline ring of pristinamycin IIA. Followed by oxidation of the intermediate sulfide into the sulfone derivatives (i.e., dalfopristin). Water-soluble derivatives (both group A and group B) can now be obtained at the industrial scale. Modified group B compounds are now also being produced by mutasynthesis, via disruption of the papA gene. Mutasynthesis has proved particularly useful for producing PIB, the group B component of the oral streptogramin RPR 106972. The streptogramins inhibit bacterial growth by disrupting the translation of mRNA into protein. Both the group A and group B compounds bind to the peptidyltransferase domain of the bacterial ribosome. The group A compounds interfere with the elongation of the polypeptide chain by preventing the binding of aa-tRNA to the ribosome and the formation of peptide bonds, while the B compounds stimulate the dissociation of the peptidyl-tRNA and may also interfere with the release of the completed polypeptide by blocking its access to the channel through which it normally leaves the ribosome. The synergy between the group A and group B compounds appears to result from an enhanced affinity of the group B compounds for the ribosome. Apparently, the group A compound induces a conformational change such that B compound binds with greater affinity. The natural streptogramins are produced as mixtures of the group A and B compounds, the combination of which is a more potent antibacterial agent than either type of compound alone. Whereas the type A or type B compound alone has, in vitro and in animal models of infection, a moderate bacteriostatic activity, the combination of the two has strong bacteriostatic activity and often bactericidal activity. Minimal inhibitory concentrations of quinupristin/dalfopristin range from 0.20 to 1 mg/l for Streptococcus pneumonae, from 0.25 to 2 mg/l for Staphylococcus aureus and from 0.50 to 4 for Enterococcus faecium, the principal target organisms of this drug. Quinupristin/dalfopristin also has activity against mycoplasmas, Neisseria gonorrhoeae, Haemophilus influenz, Legionella spp. and Moraxella catarrhalis. Bacteria develop resistance to the streptogramms by ribosomal modification, by producing inactivating enzymes, or by causing an efflux of the antibiotic. Dimethylation of an adenine residue in rRNA, a reaction that is catalyzed by a methylase encoded by the erm gene class, affects the binding of group B compounds (as well as the macrolides and lincosamides; hence, MLSB resistance), but group A and B compounds usually maintain their synergy and their bactericidal effect against MLSB-resistant strains. erm genes are widespread both geographically and throughout numerous bacterial genera. Several types of enzymes (acetyltransferases, hydrolases) have been identified that inactivate the group A or the group B compounds. Genes involved in streptogramin efflux have so far been found only in staphylococci, particularly in coagulase-negative species


Subject(s)
Anti-Bacterial Agents/pharmacology , Virginiamycin/pharmacology , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Bacteria/drug effects , Bacterial Infections/drug therapy , Drug Resistance, Microbial , Drug Resistance, Multiple , Drug Synergism , Humans , Microbial Sensitivity Tests , Virginiamycin/chemical synthesis , Virginiamycin/chemistry
7.
Curr Opin Investig Drugs ; 1(2): 173-80, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11249570

ABSTRACT

RhƓne-Poulenc Rorer (RPR) developed Synercid (RP-59500), an injectable synergistic combination of quinupristin and dalfopristin as a treatment for a variety of infections caused by Gram-positive anaerobic bacteria. The treatment was approved in the UK in July 1999, for use in patients with nosocomial pneumonia, skin and soft tissue infections and clinically significant infections due to Enterococcus faecium when there is no other active antibacterial agent [337556,335257]. It was launched in the UK and the US in September 1999 [342899]. In December 1999, Synercid successfully completed the Mutual Recognition Procedure in the EU under Aventis Pharma for use in patients with these infections [351525]. In September 2000, Merrill Lynch predicted first-year sales in 1999 of Euro 15 million, rising to Euro 171 million in 2004 [384874]. In January 1999, BT Alex Brown predicted sales of US $88 million in 1999 rising to US $450 million in 2002 [318220]. In April 1999, ABN Amro predicted annual sales of DM 30 million in 1999, rising to DM 150 million in 2002 [328676].


Subject(s)
Bacterial Infections/drug therapy , Drug Therapy, Combination/therapeutic use , Virginiamycin/therapeutic use , Animals , Bacterial Infections/microbiology , Clinical Trials as Topic , Contraindications , Drug Therapy, Combination/adverse effects , Drug Therapy, Combination/chemical synthesis , Drug Therapy, Combination/metabolism , Drug Therapy, Combination/pharmacology , Drug Therapy, Combination/toxicity , Humans , Structure-Activity Relationship , Virginiamycin/adverse effects , Virginiamycin/chemical synthesis , Virginiamycin/metabolism , Virginiamycin/pharmacology , Virginiamycin/toxicity
8.
Ann Pharm Fr ; 57(3): 255-65, 1999 May.
Article in French | MEDLINE | ID: mdl-10427862

ABSTRACT

To gain a better understanding of the effect exerted by the 3-hydroxypicolinoyl residue on the antibiotic activity of Pristinamycin IA, the C-N bond of picolinamide was cleaved electrochemically. A mechanistic study demonstrated that the presence of the peptidic macrolactone M markedly modified the expected cathodic behavior of pyridylcarboxamides. In order to assess the influence of steric crowding exerted by M on this original behavior, we look for models using two different approachs. First, tertiary pyridylcarboxamides were used to increase steric hindrance at the amide nitrogen position; second, M was opened by ammonolysis to decrease steric crowding at the amide nitrogen position. The electrochemical behavior of the selected compounds is presented in the first and the second parts of this study. Determination of pyridine nitrogen basicity in an N-substituted-3-methoxypicolinamide series is treated in the third part as a useful probe to evaluate the intensity of steric crowding at the amide nitrogen position. Finally, in the last part of this work, we propose the use of the picolinoyl residue (C6H4N-CO-ou Pic) as a protecting group for amines in peptide synthesis.


Subject(s)
Amides/chemistry , Peptides/chemical synthesis , Pyridines/chemistry , Anti-Bacterial Agents/chemical synthesis , Electrochemistry , Molecular Conformation , Oxidation-Reduction , Peptides/chemistry , Virginiamycin/chemical synthesis , Virginiamycin/chemistry
10.
Int J Pept Protein Res ; 41(2): 102-17, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8458684

ABSTRACT

We achieved the reconstruction of VS1-analogues containing a substitute for the fifth residue, gamma-oxo-Pip (Pip = pipecolic acid), starting from VS1-pentapeptide (VS5P;3) the latter being prepared by a two-step degradation process of the native antibiotic VS1 (1a). Protecting groups during the procedure were chosen in order to realize a minimal number of steps. Most of these gave excellent yields, including final cyclization between the fourth and fifth residue. In total, four analogues were synthesized with Ala, Asp, Asn and Lys (1b) replacing gamma-oxo-Pip. Among these, [Lys5(Tfa salt)]-VS1 is water-soluble, which is an important characteristic for eventual application of VS1 as a pharmaceutical agent. In the proposed reaction sequence, we made sure that residues 4 (MePhe) and 6 (Phg) became partially epimerised. We therefore obtained each time after cyclization a total of four epimers that have been separated by preparative TLC. The chiral identity of the final residues was realized by GC (Chirasil Val-III) on the total hydrolysates.


Subject(s)
Virginiamycin/analogs & derivatives , Amino Acid Sequence , Magnetic Resonance Spectroscopy , Methods , Molecular Sequence Data , Molecular Structure , Stereoisomerism , Virginiamycin/chemical synthesis , Virginiamycin/chemistry
11.
J Antimicrob Chemother ; 16 Suppl A: 13-21, 1985 Jul.
Article in English | MEDLINE | ID: mdl-3932299

ABSTRACT

Lincomycin is a 6-amino, 6-deoxy-octopyraninose with strong antibiotic activity. To maintain or enhance this activity, one must have the configuration of the first five asymmetric carbons of the sugar residue with the thioglycolic moiety in the alpha-position. The nitrogen on carbon 6 with the R configuration is also a prerequisite for the activity. Substitutions at the level of C-7 as well as the pyrolidine moiety can dramatically enhance this activity. The streptogramin group is composed of a wide variety of structures which can be classified into two subgroups. Group A or M is composed of polyunsaturated cyclic peptolides, e.g. pristinamycin IIA; group B or S is composed of cyclic hexadepsipeptides. Separately, these molecules have a bacteriostatic activity on Gram-positive organisms, whereas in association they exhibit a strong and synergistic bactericidal effect. The 13-OH function of PIIA is essential for antibiotic activity to occur whereas the 15-carbonyl function can be reduced with the retention of the biological properties of the drug. The carbonyl function of PIA can be reduced and the pipecolic moiety can be replaced by other groups without a dramatic influence on the antibiotic activity. The macrocyclic lactone ring is necessary for antibiotic activity.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Macrolides , Virginiamycin/chemical synthesis , Acetylation , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Lincosamides , Oxidation-Reduction , Structure-Activity Relationship , Virginiamycin/pharmacology
12.
J Org Chem ; 65(15): 4553-9, 2000 Jul 28.
Article in English | MEDLINE | ID: mdl-10959858

ABSTRACT

The synthetic potentialities of cerium(III) chloride are demonstrated by the synthesis of a nine-membered ring heterocycle component of Griseoviridin (3) in optically active form. The key step involves the stereospecific formation of the alpha-carbalkoxy alkenyl sulfide moiety using a combination system of cerium(III) chloride heptahydrate and sodium iodide.


Subject(s)
Anti-Bacterial Agents/chemistry , Heterocyclic Compounds/chemical synthesis , Peptides , Streptomyces/chemistry , Sulfides/chemical synthesis , Virginiamycin/chemical synthesis , Cerium/chemistry , Heterocyclic Compounds/chemistry , Magnetic Resonance Spectroscopy , Sodium Iodide/chemistry , Stereoisomerism , Sulfides/chemistry , Virginiamycin/chemistry
13.
Chemistry ; 10(17): 4334-40, 2004 Sep 06.
Article in English | MEDLINE | ID: mdl-15352116

ABSTRACT

We describe the first solid-phase synthesis of dihydrovirginiamycin S(1), a member of the streptogramin B family of antibiotics, which are nonribosomal-peptide natural products produced by Streptomyces. These compounds, along with the synergistic group A components, are "last line of defense" antimicrobial agents for the treatment of life-threatening infections such as vancomycin-resistant enterococci. The synthesis features an on-resin cyclization and is designed to allow production of streptogramin B analogues with diversification at positions 1', 1, 2, 3, 4, and 6. Several synthetic challenges known to hinder the synthesis of this class of compounds were solved, including sensitivity to acids and bases, and epimerization and rearrangements, through the judicious choice of deprotection conditions, coupling conditions, and synthetic strategy. This work should enable a better understanding of structure-activity relationships in the streptogramin B compounds, possible identification of analogues that bypass known resistance mechanisms, and perhaps the identification of analogues with novel biological activities.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Streptogramin B/chemical synthesis , Streptogramin Group B/chemical synthesis , Virginiamycin/chemical synthesis , Anti-Bacterial Agents/pharmacology , Bacillus subtilis/drug effects , Cyclization , Indicators and Reagents , Microbial Sensitivity Tests , Models, Molecular , Molecular Conformation , Streptogramin B/analogs & derivatives , Streptogramin B/pharmacology , Streptogramin Group B/pharmacology , Structure-Activity Relationship , Virginiamycin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL