Inhibitors of FabI, an enzyme drug target in the bacterial fatty acid biosynthesis pathway.

The modern age of drug discovery, which had been slowly gathering momentum during the early part of the twentieth century, exploded into life in the 1940s with the isolation of penicillin and streptomycin. The immense success of these early drug discovery efforts prompted the general view that many infectious diseases would now be effectively controlled and even eradicated. However this initial optimism was misplaced, and pathogens such as multidrug-resistant Mycobacterium tuberculosis and methicillin-resistant Staphylococcus aureus present a major current threat to human health. Drug resistance arises through the unrelenting pressure of natural selection, and there is thus a continuing need to identify novel drug targets and develop chemotherapeutics that circumvent existing drug resistance mechanisms. In this Account, we summarize current progress in developing inhibitors of FabI, the NADH-dependent enoyl reductase from the type II bacterial fatty acid biosynthesis pathway (FAS-II), a validated but currently underexploited target for drug discovery. The FabI inhibitors have been divided into two groups, based on whether they form a covalent adduct with the NAD (+) cofactor. Inhibitors that form a covalent adduct include the diazaborines, as well as the front-line tuberculosis drug isoniazid. The NAD adducts formed with these compounds are formally bisubstrate enzyme inhibitors, and we summarize progress in developing novel leads based on these pharmacophores. Inhibitors that do not form covalent adducts form a much larger group, although generally these compounds also require the cofactor to be bound to the enzyme. Using structure-based approaches, we have developed a series of alkyl diphenyl ethers that are nanomolar inhibitors of InhA, the FabI from M. tuberculosis, and that are active against INH-resistant strains of M. tuberculosis. This rational approach to inhibitor development is based on the proposal that high-affinity inhibition of the FabI enzymes is coupled to the ordering of a loop of amino acids close to the active site. Compounds that promote loop ordering are slow onset FabI inhibitors with increased residence time on the enzyme. The diphenyl ether skeleton has also been used as a framework by us and others to develop potent inhibitors of the FabI enzymes from other pathogens such as Escherichia coli, S. aureus, and Plasmodium falciparum. Meanwhile chemical optimization of compounds identified in high-throughput screening programs has resulted in the identification of several classes of heteroaromatic FabI inhibitors with potent activity both in vitro and in vivo. Finally, screening of natural product libraries may provide useful chemical entities for the development of novel agents with low toxicity. While the discovery that not all pathogens contain FabI homologues has led to reduced industrial interest in FabI as a broad spectrum target, there is substantial optimism that FabI inhibitors can be developed for disease-specific applications. In addition, the availability of genome sequencing data, improved methods for target identification and validation, and the development of novel approaches for determining the mode of action of current drugs will all play critical roles in the road ahead and in exploiting other components of the FAS-II pathway.

[1]  P. Tonge,et al.  Structure-activity studies of the inhibition of FabI, the enoyl reductase from Escherichia coli, by triclosan: kinetic analysis of mutant FabIs. , 2003, Biochemistry.

[2]  D. Livermore The need for new antibiotics. , 2004, Clinical microbiology and infection : the official publication of the European Society of Clinical Microbiology and Infectious Diseases.

[3]  Charles O. Rock,et al.  erratum: A triclosan-resistant bacterial enzyme , 2000, Nature.

[4]  David Alland,et al.  Targeting Tuberculosis and Malaria through Inhibition of Enoyl Reductase , 2003, Journal of Biological Chemistry.

[5]  R. Bax,et al.  The future challenges facing the development of new antimicrobial drugs , 2002, Nature Reviews Drug Discovery.

[6]  R. Copeland,et al.  Drug–target residence time and its implications for lead optimization , 2007, Nature Reviews Drug Discovery.

[7]  P J Artymiuk,et al.  Common themes in redox chemistry emerge from the X-ray structure of oilseed rape (Brassica napus) enoyl acyl carrier protein reductase. , 1995, Structure.

[8]  W. Jacobs,et al.  inhA, a gene encoding a target for isoniazid and ethionamide in Mycobacterium tuberculosis. , 1994, Science.

[9]  R. Slayden,et al.  Isoniazid affects multiple components of the type II fatty acid synthase system of Mycobacterium tuberculosis , 2000, Molecular microbiology.

[10]  Xiayang Qiu,et al.  Discovery of aminopyridine-based inhibitors of bacterial enoyl-ACP reductase (FabI). , 2002, Journal of medicinal chemistry.

[11]  Robert Stroud,et al.  Pyrrolidine carboxamides as a novel class of inhibitors of enoyl acyl carrier protein reductase from Mycobacterium tuberculosis. , 2006, Journal of medicinal chemistry.

[12]  H. Bergler,et al.  The enoyl-[acyl-carrier-protein] reductase (FabI) of Escherichia coli, which catalyzes a key regulatory step in fatty acid biosynthesis, accepts NADH and NADPH as cofactors and is inhibited by palmitoyl-CoA. , 1996, European journal of biochemistry.

[13]  Jie J. Zheng,et al.  The structural biology of type II fatty acid biosynthesis. , 2005, Annual review of biochemistry.

[14]  S. Levy,et al.  Antibacterial resistance worldwide: causes, challenges and responses , 2004, Nature Medicine.

[15]  Antoni R. Slabas,et al.  A Mechanism of Drug Action Revealed by Structural Studies of Enoyl Reductase , 1996, Science.

[16]  P. Schultz,et al.  Studies on the Mechanism of Action of Isoniazid and Ethionamide in the Chemotherapy of Tuberculosis , 1995 .

[17]  M. Mann,et al.  Robust Salmonella metabolism limits possibilities for new antimicrobials , 2006, Nature.

[18]  W. Jacobs,et al.  Mechanisms of isoniazid resistance in Mycobacterium tuberculosis: enzymatic characterization of enoyl reductase mutants identified in isoniazid-resistant clinical isolates. , 1998, The Journal of infectious diseases.

[19]  Kun Song,et al.  Insight through molecular mechanics Poisson-Boltzmann surface area calculations into the binding affinity of triclosan and three analogues for FabI, the E. coli enoyl reductase. , 2006, Journal of medicinal chemistry.

[20]  M. Espinal,et al.  Tuberculosis as a major global health problem in the 21st century: a WHO perspective. , 2004, Seminars in respiratory and critical care medicine.

[21]  M. Head,et al.  Defining and Combating the Mechanisms of Triclosan Resistance in Clinical Isolates of Staphylococcus aureus , 2002, Antimicrobial Agents and Chemotherapy.

[22]  Jun Wang,et al.  Platensimycin is a selective FabF inhibitor with potent antibiotic properties , 2006, Nature.

[23]  D. Williams The glycopeptide story--how to kill the deadly 'superbugs'. , 1996, Natural product reports.

[24]  L. A. Basso,et al.  An inorganic iron complex that inhibits wild-type and an isoniazid-resistant mutant 2-trans-enoyl-ACP (CoA) reductase from Mycobacterium tuberculosis. , 2004, Chemical communications.

[25]  Deniz Tasdemir,et al.  Inhibition of Plasmodium falciparum fatty acid biosynthesis: evaluation of FabG, FabZ, and FabI as drug targets for flavonoids. , 2006, Journal of medicinal chemistry.

[26]  T. Scior,et al.  Isoniazid is not a lead compound for its pyridyl ring derivatives, isonicotinoyl amides, hydrazides, and hydrazones: a critical review. , 2006, Current medicinal chemistry.

[27]  J. Puglisi,et al.  Structure of the A Site of Escherichia coli 16S Ribosomal RNA Complexed with an Aminoglycoside Antibiotic , 1996, Science.

[28]  A. Maxwell DNA gyrase as a drug target. , 1997, Trends in microbiology.

[29]  Peter J. Tonge,et al.  The isoniazid-NAD adduct is a slow, tight-binding inhibitor of InhA, the Mycobacterium tuberculosis enoyl reductase: Adduct affinity and drug resistance , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[30]  S. Sharma,et al.  Green tea catechins potentiate triclosan binding to enoyl-ACP reductase from Plasmodium falciparum (PfENR). , 2007, Journal of medicinal chemistry.

[31]  Xiayang Qiu,et al.  Indole naphthyridinones as inhibitors of bacterial enoyl-ACP reductases FabI and FabK. , 2003, Journal of medicinal chemistry.

[32]  S. Parikh,et al.  Inhibition of InhA, the enoyl reductase from Mycobacterium tuberculosis, by triclosan and isoniazid. , 2000, Biochemistry.

[33]  K. Kumura,et al.  4-Pyridone derivatives as new inhibitors of bacterial enoyl-ACP reductase FabI. , 2007, Bioorganic & medicinal chemistry.

[34]  Tae-Gyu Lee,et al.  Fatty acid synthesis is a target for antibacterial activity of unsaturated fatty acids , 2005, FEBS letters.

[35]  R. Perozzo,et al.  Anti-protozoal and plasmodial FabI enzyme inhibiting metabolites of Scrophularia lepidota roots. , 2005, Phytochemistry.

[36]  Joel S. Freundlich,et al.  X-ray Structural Analysis of Plasmodium falciparum Enoyl Acyl Carrier Protein Reductase as a Pathway toward the Optimization of Triclosan Antimalarial Efficacy* , 2007, Journal of Biological Chemistry.

[37]  T. Schaeverbeke,et al.  In-vitro activity of grepafloxacin, a new fluoroquinolone, against mycoplasmas. , 1999, The Journal of antimicrobial chemotherapy.

[38]  C. Vilchèze,et al.  Crystal Structure of the Mycobacterium tuberculosis Enoyl-ACP Reductase, InhA, in Complex with NAD+ and a C16 Fatty Acyl Substrate* , 1999, The Journal of Biological Chemistry.

[39]  W. Delano The PyMOL Molecular Graphics System , 2002 .

[40]  J. Hildebrandt,et al.  Preparation and antibacterial activities of new 1,2,3-diazaborine derivatives and analogues. , 1984, Journal of medicinal chemistry.

[41]  P. Tonge,et al.  Inhibition of the bacterial enoyl reductase FabI by triclosan: a structure-reactivity analysis of FabI inhibition by triclosan analogues. , 2004, Journal of medicinal chemistry.

[42]  Matthew W Vetting,et al.  Mycobacterium tuberculosis dihydrofolate reductase is a target for isoniazid , 2006, Nature Structural &Molecular Biology.

[43]  L. L. Ling,et al.  Identification and Characterization of Inhibitors of Bacterial Enoyl-Acyl Carrier Protein Reductase , 2022 .

[44]  H. Schweizer,et al.  High-level triclosan resistance in Pseudomonas aeruginosa is solely a result of efflux. , 2003, American journal of infection control.

[45]  R. Bax,et al.  Antibiotic resistance - what can we do? , 1998, Nature Medicine.

[46]  P. McDermott,et al.  Genetic Evidence that InhA of Mycobacterium smegmatis Is a Target for Triclosan , 1999, Antimicrobial Agents and Chemotherapy.

[47]  V. Bernardes-Génisson,et al.  The first chemical synthesis of the core structure of the benzoylhydrazine-NAD adduct, a competitive inhibitor of the Mycobacterium tuberculosis enoyl reductase. , 2005, The Journal of organic chemistry.

[48]  M. Rosenberg,et al.  Exploiting genomics to discover new antibiotics. , 2001, Trends in microbiology.

[49]  D W Rice,et al.  Mechanism of action of diazaborines. , 1998, Biochemical pharmacology.

[50]  P. Warren,et al.  Bacterial fatty-acid biosynthesis: a genomics-driven target for antibacterial drug discovery. , 2001, Drug discovery today.

[51]  P. Tonge,et al.  Synthesis of 4-phenoxybenzamide adenine dinucleotide as NAD analogue with inhibitory activity against enoyl-ACP reductase (InhA) of Mycobacterium tuberculosis. , 2007, Bioorganic & medicinal chemistry letters.

[52]  S. Levy,et al.  Triclosan targets lipid synthesis , 1998, Nature.

[53]  J C Sacchettini,et al.  Enzymatic characterization of the target for isoniazid in Mycobacterium tuberculosis. , 1995, Biochemistry.

[54]  J. W. Campbell,et al.  Bacterial fatty acid biosynthesis: targets for antibacterial drug discovery. , 2001, Annual review of microbiology.

[55]  C. Rock,et al.  Evaluation of Epigallocatechin Gallate and Related Plant Polyphenols as Inhibitors of the FabG and FabI Reductases of Bacterial Type II Fatty-acid Synthase* , 2004, Journal of Biological Chemistry.

[56]  Christopher J. L. Murray,et al.  Tuberculosis: Commentary on a Reemergent Killer , 1992, Science.

[57]  T. Ramya,et al.  Novel diphenyl ethers: design, docking studies, synthesis and inhibition of enoyl ACP reductase of Plasmodium falciparum and Escherichia coli. , 2006, Bioorganic & medicinal chemistry.

[58]  A. Rattan,et al.  Multidrug-resistant Mycobacterium tuberculosis: molecular perspectives. , 1998, Emerging infectious diseases.

[59]  D. Pompliano,et al.  Drugs for bad bugs: confronting the challenges of antibacterial discovery , 2007, Nature Reviews Drug Discovery.

[60]  Teresa Quitugua,et al.  Single Nucleotide Polymorphisms in Genes Associated with Isoniazid Resistance in Mycobacterium tuberculosis , 2003, Antimicrobial Agents and Chemotherapy.

[61]  C. Walsh Molecular mechanisms that confer antibacterial drug resistance , 2000, Nature.

[62]  R. Heath,et al.  The Enoyl-[acyl-carrier-protein] Reductases FabI and FabL fromBacillus subtilis * , 2000, The Journal of Biological Chemistry.

[63]  S. Parikh,et al.  Structural basis and mechanism of enoyl reductase inhibition by triclosan. , 1999, Journal of molecular biology.

[64]  A. Kochi,et al.  The global tuberculosis situation and the new control strategy of the World Health Organization. , 1991, Tubercle.

[65]  S. Chirala,et al.  Animal fatty acid synthase: functional mapping and cloning and expression of the domain I constituent activities. , 1997, Proceedings of the National Academy of Sciences of the United States of America.

[66]  R. Dubos,et al.  THE ISOLATION OF BACTERICIDAL SUBSTANCES FROM CULTURES OF BACILLUS BREVIS , 1941 .

[67]  M. Blacow,et al.  INDUCTION OF LABOUR , 1975, The Lancet.

[68]  S. Parikh,et al.  Roles of tyrosine 158 and lysine 165 in the catalytic mechanism of InhA, the enoyl-ACP reductase from Mycobacterium tuberculosis. , 1999, Biochemistry.

[69]  W. Roper,et al.  The new tuberculosis. , 1992, The New England journal of medicine.

[70]  J. Mcgowan Resistance in nonfermenting gram-negative bacteria: multidrug resistance to the maximum. , 2006, American journal of infection control.

[71]  G. Domagk Ein Beitrag zur Chemotherapie der bakteriellen Infektionen , 1935 .

[72]  I. Taylor,et al.  Kinetic and structural characteristics of the inhibition of enoyl (acyl carrier protein) reductase by triclosan. , 1999, Biochemistry.

[73]  J. Sacchettini,et al.  Modification of the NADH of the isoniazid target (InhA) from Mycobacterium tuberculosis. , 1998, Science.

[74]  Peter J Tonge,et al.  High affinity InhA inhibitors with activity against drug-resistant strains of Mycobacterium tuberculosis. , 2006, ACS chemical biology.

[75]  O. N. de Souza,et al.  Slow-onset inhibition of 2-trans-enoyl-ACP (CoA) reductase from Mycobacterium tuberculosis by an inorganic complex. , 2006, Current pharmaceutical design.

[76]  Chang Ki Kim,et al.  In Vitro Activities of CG400549, a Novel FabI Inhibitor, against Recently Isolated Clinical Staphylococcal Strains in Korea , 2007, Antimicrobial Agents and Chemotherapy.

[77]  R. Angeletti,et al.  Proteome-wide profiling of isoniazid targets in Mycobacterium tuberculosis. , 2006, Biochemistry.

[78]  S. Waksman,et al.  Streptomycin, a Substance Exhibiting Antibiotic Activity Against Gram-Positive and Gram-Negative Bacteria.∗† , 1944, Clinical orthopaedics and related research.

[79]  N. Kaplan,et al.  In Vitro Activity of API-1252, a Novel FabI Inhibitor, against Clinical Isolates of Staphylococcus aureus and Staphylococcus epidermidis , 2007, Antimicrobial Agents and Chemotherapy.