1
|
Zahara AJ, Haines BE, Wilkerson-Hill SM. Programmed Heterocycle Synthesis Using Halomucononitriles as Pyridinimine Precursors. Org Lett 2024; 26:2976-2981. [PMID: 38557087 DOI: 10.1021/acs.orglett.4c00547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Herein we report a method to convert primary amines, ubiquitous motifs found in pharmaceutical libraries, to either imidazo[1,2-a]pyridines or 7-alkyl azaindoles in two steps from known compounds. Using halomucononitrile reagents, we can directly access 5-bromo-6-imino-1-alkyl-1,6-dihydropyridine-2-carbonitriles (pyridinimines) in a single step from primary amines (25-93% yield) through the cyclization of transient aminomucononitrile intermediates. We then demonstrate that these compounds can be readily converted to 7-alkylazaindoles using Sonogashira cross-coupling conditions (13 examples, up to 91% yield). Under oxidative conditions, the pyridinimines serve as directing groups for C-H functionalization reactions to afford imidazo[1,2-a]pyridines. We also studied the mechanism of the cyclization event using DFT calculations and propose that this takes place via sequential base-mediated E/Z isomerization and cyclization steps.
Collapse
Affiliation(s)
- Adam J Zahara
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Brandon E Haines
- Department of Chemistry, Westmont College, Santa Barbara, California 93108, United States
| | - Sidney M Wilkerson-Hill
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
2
|
Samanta S, Kumar S, Aratikatla EK, Ghorpade SR, Singh V. Recent developments of imidazo[1,2- a]pyridine analogues as antituberculosis agents. RSC Med Chem 2023; 14:644-657. [PMID: 37122538 PMCID: PMC10131611 DOI: 10.1039/d3md00019b] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Over the past 2000 years, tuberculosis (TB) has killed more people than any other infectious disease. In 2021, TB claimed 1.6 million lives worldwide, making it the second leading cause of death from an infectious disease after COVID-19. Unfortunately, TB drug discovery research was neglected in the last few decades of the twentieth century. Recently, the World Health Organization has taken the initiative to develop new TB drugs. Imidazopyridine, an important fused bicyclic 5,6 heterocycle has been recognized as a "drug prejudice" scaffold for its wide range of applications in medicinal chemistry. A few examples of imidazo[1,2-a]pyridine exhibit significant activity against multidrug-resistant TB (MDR-TB) and extensively drug-resistant TB (XDR-TB). Here, we critically review anti-TB compounds of the imidazo[1,2-a]pyridine class by discussing their development based on the structure-activity relationship, mode-of-action, and various scaffold hopping strategies over the last decade, which is identified as a renaissance era of TB drug discovery research.
Collapse
Affiliation(s)
- Sauvik Samanta
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town Rondebosch 7701 South Africa
| | - Sumit Kumar
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town Rondebosch 7701 South Africa
| | - Eswar K Aratikatla
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town Rondebosch 7701 South Africa
| | - Sandeep R Ghorpade
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town Rondebosch 7701 South Africa
| | - Vinayak Singh
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town Rondebosch 7701 South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Institute of Infectious Disease and Molecular Medicine, University of Cape Town Rondebosch 7701 South Africa
| |
Collapse
|
3
|
Dasmahapatra U, Chanda K. Synthetic approaches to potent heterocyclic inhibitors of tuberculosis: A decade review. Front Pharmacol 2022; 13:1021216. [PMID: 36386156 PMCID: PMC9661889 DOI: 10.3389/fphar.2022.1021216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/03/2022] [Indexed: 09/08/2024] Open
Abstract
Tuberculosis (TB) continues to be a significant global health concern with about 1.5 million deaths annually. Despite efforts to develop more efficient vaccines, reliable diagnostics, and chemotherapeutics, tuberculosis has become a concern to world health due to HIV, the rapid growth of bacteria that are resistant to treatment, and the recently introduced COVID-19 pandemic. As is well known, advances in synthetic organic chemistry have historically enabled the production of important life-saving medications that have had a tremendous impact on patients' lives and health all over the world. Small-molecule research as a novel chemical entity for a specific disease target offers in-depth knowledge and potential therapeutic targets. In this viewpoint, we concentrated on the synthesis of a number of heterocycles reported in the previous decade and the screening of their inhibitory action against diverse strains of Mycobacterium tuberculosis. These findings offer specific details on the structure-based activity of several heterocyclic scaffolds backed by their in vitro tests as a promising class of antitubercular medicines, which will be further useful to build effective treatments to prevent this terrible illness.
Collapse
Affiliation(s)
| | - Kaushik Chanda
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
4
|
Khan A, Poojary SS, Bhave KK, Nandan SR, Iyer KR, Coutinho EC. Prediction of QcrB Inhibition as a Measure of Antitubercular Activity with Machine Learning Protocols. ACS OMEGA 2022; 7:18094-18102. [PMID: 35664614 PMCID: PMC9161412 DOI: 10.1021/acsomega.2c01613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/06/2022] [Indexed: 05/07/2023]
Abstract
It has always been a challenge to develop interventional therapies for Mycobacterium tuberculosis. Over the years, several attempts at developing such therapies have hit a dead-end owing to rapid mutation rates of the tubercular bacilli and their ability to lay dormant for years. Recently, cytochrome bcc complex (QcrB) has shown some promise as a novel target against the tubercular bacilli, with Q203 being the first molecule acting on this target. In this paper, we report the deployment of several ML-based approaches to design molecules against QcrB. Machine learning (ML) models were developed based on a data set of 350 molecules using three different sets of molecular features, i.e., MACCS keys, ECFP6 fingerprints, and Mordred descriptors. Each feature set was trained on eight ML classifier algorithms and optimized to classify molecules accurately. The support vector machine-based classifier using the ECFP6 feature set was found to be the best classifier in this study. Further, screening of the known imidazopyridine amide inhibitors demonstrated that the model correctly classified the most potent molecules as actives, hence validating the model for future applications.
Collapse
Affiliation(s)
- Afreen
A. Khan
- Department
of Pharmaceutical Chemistry, Vasvik Research Centre, Bombay College of Pharmacy, Kalina, Santacruz (E), Mumbai 400 098, India
| | - Sannidhi S. Poojary
- Department
of Pharmaceutical Chemistry, Vasvik Research Centre, Bombay College of Pharmacy, Kalina, Santacruz (E), Mumbai 400 098, India
| | - Ketki K. Bhave
- Department
of Pharmaceutical Chemistry, Vasvik Research Centre, Bombay College of Pharmacy, Kalina, Santacruz (E), Mumbai 400 098, India
| | - Santosh R. Nandan
- Ambernath
Organics Pvt. Ltd., 222,
The Summit Business Bay, Andheri (E), Mumbai 400 093, India
| | - Krishna R. Iyer
- Department
of Pharmaceutical Chemistry, Vasvik Research Centre, Bombay College of Pharmacy, Kalina, Santacruz (E), Mumbai 400 098, India
| | - Evans C. Coutinho
- Department
of Pharmaceutical Chemistry, Vasvik Research Centre, Bombay College of Pharmacy, Kalina, Santacruz (E), Mumbai 400 098, India
| |
Collapse
|
5
|
Emerging impact of triazoles as anti-tubercular agent. Eur J Med Chem 2022; 238:114454. [PMID: 35597009 DOI: 10.1016/j.ejmech.2022.114454] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 05/04/2022] [Accepted: 05/08/2022] [Indexed: 01/08/2023]
Abstract
Tuberculosis, a disease of poverty is a communicable infection with a reasonably high mortality rate worldwide. 10 Million new cases of TB were reported with approx 1.4 million deaths in the year 2019. Due to the growing number of drug-sensitive and drug-resistant tuberculosis cases, there is a vital need to develop new and effective candidates useful to combat this deadly disease. Despite tremendous efforts to identify a mechanism-based novel antitubercular agent, only a few have entered into clinical trials in the last six decades. In recent years, triazoles have been well explored as the most valuable scaffolds in drug discovery and development. Triazole framework possesses favorable properties like hydrogen bonding, moderate dipole moment, enhanced water solubility, and also the ability to bind effectively with biomolecular targets of M. tuberculosis and therefore this scaffold displayed excellent potency against TB. This review is an endeavor to summarize an up-to-date innovation of triazole-appended hybrids during the last 10 years having potential in vitro and in vivo antitubercular activity with structure activity relationship analysis. This review may help medicinal chemists to explore the triazole scaffolds for the rational design of potent drug candidates having better efficacy, improved selectivity and minimal toxicity so that these hybrid NCEs can effectively be explored as potential lead to fight against M. tuberculosis.
Collapse
|
6
|
Jampilek J. Drug repurposing to overcome microbial resistance. Drug Discov Today 2022; 27:2028-2041. [PMID: 35561965 DOI: 10.1016/j.drudis.2022.05.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/02/2022] [Accepted: 05/06/2022] [Indexed: 12/15/2022]
Abstract
Infections are a growing global threat, and the number of resistant species of microbial pathogens is alarming. However, the rapid development of cross-resistant or multidrug-resistant strains and the development of so-called 'superbugs' are in stark contrast to the number of newly launched anti-infectives on the market. In this review, I summarize the causes of antimicrobial resistance, briefly discuss different approaches to the discovery and development of new anti-infective drugs, and focus on drug repurposing strategy, which is discussed from all possible perspectives. A comprehensive overview of drugs of other indications tested for their in vitro antimicrobial activity to support existing anti-infective therapeutics is provided, including several critical remarks on this strategy of repurposing non-antibiotics to antibacterial drugs.
Collapse
Affiliation(s)
- Josef Jampilek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia; Department of Chemical Biology, Faculty of Science, Palacky University, Slechtitelu 27, 783 71 Olomouc, Czech Republic.
| |
Collapse
|
7
|
Wani MA, Dhaked DK. Targeting the cytochrome bc 1 complex for drug development in M. tuberculosis: review. Mol Divers 2021; 26:2949-2965. [PMID: 34762234 DOI: 10.1007/s11030-021-10335-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/04/2021] [Indexed: 11/26/2022]
Abstract
The terminal oxidases of the oxidative phosphorylation pathway play a significant role in the survival and growth of M. tuberculosis, targeting these components lead to inhibition of M. tuberculosis. Many drug candidates targeting various components of the electron transport chain in M. tuberculosis have recently been discovered. The cytochrome bc1-aa3 supercomplex is one of the most important components of the electron transport chain in M. tuberculosis, and it has emerged as the novel target for several promising candidates. There are two cryo-electron microscopy structures (PDB IDs: 6ADQ and 6HWH) of the cytochrome bc1-aa3 supercomplex that aid in the development of effective and potent inhibitors for M. tuberculosis. In recent years, a number of potential candidates targeting the QcrB subunit of the cytochrome bc1 complex have been developed. In this review, we describe the recently identified inhibitors that target the electron transport chain's terminal oxidase enzyme in M. tuberculosis, specifically the QcrB subunit of the cytochrome bc1 complex.
Collapse
Affiliation(s)
- Mushtaq Ahmad Wani
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER)-Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, West Bengal, 700054, India
| | - Devendra Kumar Dhaked
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER)-Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, West Bengal, 700054, India.
| |
Collapse
|
8
|
Alsayed SSR, Lun S, Bailey AW, Suri A, Huang CC, Mocerino M, Payne A, Sredni ST, Bishai WR, Gunosewoyo H. Design, synthesis and evaluation of novel indole-2-carboxamides for growth inhibition of Mycobacterium tuberculosis and paediatric brain tumour cells. RSC Adv 2021; 11:15497-15511. [PMID: 35481189 PMCID: PMC9029315 DOI: 10.1039/d0ra10728j] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/10/2021] [Indexed: 12/17/2022] Open
Abstract
The omnipresent threat of tuberculosis (TB) and the scant treatment options thereof necessitate the development of new antitubercular agents, preferably working via a novel mechanism of action distinct from the current drugs. Various studies identified the mycobacterial membrane protein large 3 transporter (MmpL3) as the target of several classes of compounds, including the indole-2-caboxamides. Herein, several indoleamide analogues were rationally designed, synthesised, and evaluated for their antitubercular and antitumour activities. Compound 8g displayed the highest activity (MIC = 0.32 μM) against the drug-sensitive (DS) Mycobacterium tuberculosis (M. tb) H37Rv strain. This compound also exhibited high selective activity towards M. tb over mammalian cells [IC50 (Vero cells) = 40.9 μM, SI = 128], suggesting its minimal cytotoxicity. In addition, when docked into the MmpL3 active site, 8g adopted a binding profile similar to the indoleamide ligand ICA38. A related compound 8f showed dual antitubercular (MIC = 0.62 μM) and cytotoxic activities against paediatric glioblastoma multiforme (GBM) cell line KNS42 [IC50 (viability) = 0.84 μM]. Compound 8f also showed poor cytotoxic activity against healthy Vero cells (IC50 = 39.9 μM). Compounds 9a and 15, which were inactive against M. tb, showed potent cytotoxic (IC50 = 8.25 and 5.04 μM, respectively) and antiproliferative activities (IC50 = 9.85 and 6.62 μM, respectively) against KNS42 cells. Transcriptional analysis of KNS42 cells treated with compound 15 revealed a significant downregulation in the expression of the carbonic anhydrase 9 (CA9) and the spleen tyrosine kinase (SYK) genes. The expression levels of these genes in GBM tumours were previously shown to contribute to tumour progression, suggesting their involvement in our observed antitumour activities. Compounds 9a and 15 were selected for further evaluations against three different paediatric brain tumour cell lines (BT12, BT16 and DAOY) and non-neoplastic human fibroblast cells HFF1. Compound 9a showed remarkable cytotoxic (IC50 = 0.89 and 1.81 μM, respectively) and antiproliferative activities (IC50 = 7.44 and 6.06 μM, respectively) against the two tested atypical teratoid/rhabdoid tumour (AT/RT) cells BT12 and BT16. Interestingly, compound 9a was not cytotoxic when tested against non-neoplastic HFF1 cells [IC50 (viability) = 119 μM]. This suggests that an indoleamide scaffold can be fine-tuned to confer a set of derivatives with selective antitubercular and/or antitumour activities. In this study, we demonstrated that an indoleamide scaffold can be fine-tuned to confer a set of derivatives with selective antitubercular and/or antitumour activities.![]()
Collapse
Affiliation(s)
- Shahinda S R Alsayed
- Curtin Medical School, Faculty of Health Sciences, Curtin University Bentley Perth WA 6102 Australia
| | - Shichun Lun
- Center for Tuberculosis Research, Department of Medicine, Division of Infectious Disease, Johns Hopkins School of Medicine 1550, Orleans Street Baltimore Maryland 21231-1044 USA
| | - Anders W Bailey
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital of Chicago Chicago IL 60611 USA
| | - Amreena Suri
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital of Chicago Chicago IL 60611 USA
| | - Chiang-Ching Huang
- Department of Biostatistics, Zilber School of Public Health, University of Wisconsin Milwaukee WI 53205 USA
| | - Mauro Mocerino
- School of Molecular and Life Sciences, Curtin University Perth WA 6102 Australia
| | - Alan Payne
- School of Molecular and Life Sciences, Curtin University Perth WA 6102 Australia
| | - Simone Treiger Sredni
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital of Chicago Chicago IL 60611 USA.,Department of Surgery, Northwestern University, Feinberg School of Medicine Chicago IL 60611 USA
| | - William R Bishai
- Center for Tuberculosis Research, Department of Medicine, Division of Infectious Disease, Johns Hopkins School of Medicine 1550, Orleans Street Baltimore Maryland 21231-1044 USA .,Howard Hughes Medical Institute 4000 Jones Bridge Road Chevy Chase Maryland 20815-6789 USA
| | - Hendra Gunosewoyo
- Curtin Medical School, Faculty of Health Sciences, Curtin University Bentley Perth WA 6102 Australia
| |
Collapse
|
9
|
Nandikolla A, Srinivasarao S, Khetmalis YM, Kumar BK, Murugesan S, Shetye G, Ma R, Franzblau SG, Sekhar KVGC. Design, synthesis and biological evaluation of novel 1,2,3-triazole analogues of Imidazo-[1,2-a]-pyridine-3-carboxamide against Mycobacterium tuberculosis. Toxicol In Vitro 2021; 74:105137. [PMID: 33684466 DOI: 10.1016/j.tiv.2021.105137] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/28/2021] [Accepted: 03/04/2021] [Indexed: 10/22/2022]
Abstract
Twenty-eight novel 1,2,3-triazole analogues of imidazo-[1,2-a]-pyridine-3-carboxamide were designed and synthesized based on hybridization approach. The structure of the final compounds are characterized using 1HNMR, 13CNMR, LCMS and elemental analyses and are screened in vitro for anti-tubercular activity using low-oxygen recovery assay (LORA) non-replicating and using microplate alamar blue assay (MABA) against replicating M. tuberculosis. MIC was determined. From the obtained results, it was observed that, among (2,7-dimethylimidazo[1,2-a]pyridin-3-yl)(4-((1-subtituted phenyl-1H-1,2,3-triazol-4-yl)methyl)piperazin-1-yl)methanones and (6-chloro-2-methylimidazo[1,2-a]pyridin-3-yl)(4-((1-substituted phenyl-1H-1,2,3-triazol-4-yl)methyl)piperazin-1-yl)methanones, compounds with substitution at para position with electron electron releasing groups exhibited the best activity (< 34 μg/mL). Amidst, (2,7-dimethylimidazo[1,2-a]pyridin-3-yl)(4-(2-(4-alkyl/substituted aryl-1H-1,2,3-triazol-1-yl)ethyl)piperazin-1-yl)methanones and (6-chloro-2-methylimidazo[1,2-a]pyridin-3-yl)(4-(2-(4- alkyl/substituted aryl -1H-1,2,3-triazol-1-yl)ethyl)piperazin-1-yl)methanones, compounds with long alkyl chain or cyclo propyl group were most active (< 21 μg/mL) in MABA method against the tested strain of MTB. Compound 10b emerged to be the most active compound in MABA and LORA with MIC values 13.74 and 24.63 μg/mL respectively. In-silico ADMET parameters were also predicted for the significantly active compound. Finally, molecular docking study was carried out to predict the feasible binding pattern of the most active compound at the active site of enoyl acyl carrier protein reductase from Mycobacterium tuberculosis (PDB-4TZK) using Glide module of Schrodinger software.
Collapse
Affiliation(s)
- Adinarayana Nandikolla
- Department of Chemistry, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Hyderabad 500078, Telangana, India
| | - Singireddi Srinivasarao
- Department of Chemistry, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Hyderabad 500078, Telangana, India
| | - Yogesh Mahadu Khetmalis
- Department of Chemistry, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Hyderabad 500078, Telangana, India
| | - Banoth Karan Kumar
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani 333031, Rajasthan. India
| | - Sankaranarayanan Murugesan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani 333031, Rajasthan. India
| | - Gauri Shetye
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, IL 60612. USA
| | - Rui Ma
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, IL 60612. USA
| | - Scott G Franzblau
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, IL 60612. USA
| | - Kondapalli Venkata Gowri Chandra Sekhar
- Department of Chemistry, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Hyderabad 500078, Telangana, India.
| |
Collapse
|
10
|
Bahuguna A, Rawat S, Rawat DS. QcrB in Mycobacterium tuberculosis: The new drug target of antitubercular agents. Med Res Rev 2021; 41:2565-2581. [PMID: 33400275 DOI: 10.1002/med.21779] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/23/2020] [Accepted: 12/12/2020] [Indexed: 11/08/2022]
Abstract
Drug-resistance in mycobacterial infections is a major global health problem that leads to high mortality and socioeconomic pressure in developing countries around the world. From finding new targets to discovering novel chemical scaffolds, there is an urgent need for the development of better approaches for the cure of tuberculosis. Recently, energy metabolism in mycobacteria, particularly the oxidative phosphorylation pathway of cellular respiration, has emerged as a novel target pathway in drug discovery. New classes of antibacterials which target oxidative phosphorylation pathway either by interacting with a protein or any step in the pathway of oxidative phosphorylation can combat dormant mycobacterial infections leading to shortening of tuberculosis chemotherapy. Adenosine triphosphate synthase is one such recently discovered target of the newly approved antitubercular drug bedaquiline. Cytochrome bcc is another new target of the antitubercular drug candidate Q203, currently in phase II clinical trial. Research suggests that b subunit of cytochrome bcc, QcrB, is the target of Q203. The review article describes the structure, function, and importance of targeting QcrB throwing light on all chemical classes of QcrB inhibitors discovered to date. An understanding of the structure and function of validated targets and their inhibitors would enable the development of new chemical entities.
Collapse
Affiliation(s)
| | - Srishti Rawat
- Department of Chemistry, University of Delhi, Delhi, India
| | - Diwan S Rawat
- Department of Chemistry, University of Delhi, Delhi, India
| |
Collapse
|
11
|
Small organic molecules targeting the energy metabolism of Mycobacterium tuberculosis. Eur J Med Chem 2020; 212:113139. [PMID: 33422979 DOI: 10.1016/j.ejmech.2020.113139] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 11/21/2022]
Abstract
Causing approximately 10 million incident cases and 1.3-1.5 million deaths every year, Mycobacterium tuberculosis remains a global health problem. The risk is further exacerbated with latent tuberculosis (TB) infection, the HIV pandemic, and increasing anti-TB drug resistance. Therefore, unexplored chemical scaffolds directed towards new molecular targets are increasingly desired. In this context, mycobacterial energy metabolism, particularly the oxidative phosphorylation (OP) pathway, is gaining importance. Mycobacteria possess primary dehydrogenases to fuel electron transport; aa3-type cytochrome c oxidase and bd-type menaquinol oxidase to generate a protonmotive force; and ATP synthase, which is essential for both growing mycobacteria as well as dormant mycobacteria because ATP is produced under both aerobic and hypoxic conditions. Small organic molecules targeting OP are active against latent TB as well as resistant TB strains. FDA approval of the ATP synthase inhibitor bedaquiline and the discovery of clinical candidate Q203, which both interfere with the cytochrome bc1 complex, have already confirmed mycobacterial energy metabolism to be a valuable anti-TB drug target. This review highlights both preferable molecular targets within mycobacterial OP and promising small organic molecules targeting OP. Progressive research in the area of mycobacterial OP revealed several highly potent anti-TB compounds with nanomolar-range MICs as low as 0.004 μM against Mtb H37Rv. Therefore, we are convinced that targeting the OP pathway can combat resistant TB and latent TB, leading to more efficient anti-TB chemotherapy.
Collapse
|
12
|
Bajeli S, Baid N, Kaur M, Pawar GP, Chaudhari VD, Kumar A. Terminal Respiratory Oxidases: A Targetables Vulnerability of Mycobacterial Bioenergetics? Front Cell Infect Microbiol 2020; 10:589318. [PMID: 33330134 PMCID: PMC7719681 DOI: 10.3389/fcimb.2020.589318] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022] Open
Abstract
Recently, ATP synthase inhibitor Bedaquiline was approved for the treatment of multi-drug resistant tuberculosis emphasizing the importance of oxidative phosphorylation for the survival of mycobacteria. ATP synthesis is primarily dependent on the generation of proton motive force through the electron transport chain in mycobacteria. The mycobacterial electron transport chain utilizes two terminal oxidases for the reduction of oxygen, namely the bc1-aa3 supercomplex and the cytochrome bd oxidase. The bc1-aa3 supercomplex is an energy-efficient terminal oxidase that pumps out four vectoral protons, besides consuming four scalar protons during the transfer of electrons from menaquinone to molecular oxygen. In the past few years, several inhibitors of bc1-aa3 supercomplex have been developed, out of which, Q203 belonging to the class of imidazopyridine, has moved to clinical trials. Recently, the crystal structure of the mycobacterial cytochrome bc1-aa3 supercomplex was solved, providing details of the route of transfer of electrons from menaquinone to molecular oxygen. Besides providing insights into the molecular functioning, crystal structure is aiding in the targeted drug development. On the other hand, the second respiratory terminal oxidase of the mycobacterial respiratory chain, cytochrome bd oxidase, does not pump out the vectoral protons and is energetically less efficient. However, it can detoxify the reactive oxygen species and facilitate mycobacterial survival during a multitude of stresses. Quinolone derivatives (CK-2-63) and quinone derivative (Aurachin D) inhibit cytochrome bd oxidase. Notably, ablation of both the two terminal oxidases simultaneously through genetic methods or pharmacological inhibition leads to the rapid death of the mycobacterial cells. Thus, terminal oxidases have emerged as important drug targets. In this review, we have described the current understanding of the functioning of these two oxidases, their physiological relevance to mycobacteria, and their inhibitors. Besides these, we also describe the alternative terminal complexes that are used by mycobacteria to maintain energized membrane during hypoxia and anaerobic conditions.
Collapse
Affiliation(s)
- Sapna Bajeli
- Molecular Mycobacteriology, Council of Scientific and Industrial Research, Institute of Microbial Technology, Chandigarh, India
| | - Navin Baid
- Molecular Mycobacteriology, Council of Scientific and Industrial Research, Institute of Microbial Technology, Chandigarh, India
| | - Manjot Kaur
- Division of Medicinal Chemistry, Council of Scientific and Industrial Research, Institute of Microbial Technology, Chandigarh, India
| | - Ganesh P Pawar
- Division of Medicinal Chemistry, Council of Scientific and Industrial Research, Institute of Microbial Technology, Chandigarh, India
| | - Vinod D Chaudhari
- Division of Medicinal Chemistry, Council of Scientific and Industrial Research, Institute of Microbial Technology, Chandigarh, India
| | - Ashwani Kumar
- Molecular Mycobacteriology, Council of Scientific and Industrial Research, Institute of Microbial Technology, Chandigarh, India
| |
Collapse
|
13
|
Appetecchia F, Consalvi S, Scarpecci C, Biava M, Poce G. SAR Analysis of Small Molecules Interfering with Energy-Metabolism in Mycobacterium tuberculosis. Pharmaceuticals (Basel) 2020; 13:E227. [PMID: 32878317 PMCID: PMC7557483 DOI: 10.3390/ph13090227] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/28/2020] [Accepted: 08/28/2020] [Indexed: 12/13/2022] Open
Abstract
Tuberculosis remains the world's top infectious killer: it caused a total of 1.5 million deaths and 10 million people fell ill with TB in 2018. Thanks to TB diagnosis and treatment, mortality has been falling in recent years, with an estimated 58 million saved lives between 2000 and 2018. However, the emergence of multidrug-resistant (MDR) and extensively drug-resistant (XDR) Mtb strains is a major concern that might reverse this progress. Therefore, the development of new drugs acting upon novel mechanisms of action is a high priority in the global health agenda. With the approval of bedaquiline, which targets mycobacterial energy production, and delamanid, which targets cell wall synthesis and energy production, the energy-metabolism in Mtb has received much attention in the last decade as a potential target to investigate and develop new antimycobacterial drugs. In this review, we describe potent anti-mycobacterial agents targeting the energy-metabolism at different steps with a special focus on structure-activity relationship (SAR) studies of the most advanced compound classes.
Collapse
Affiliation(s)
| | | | | | | | - Giovanna Poce
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, piazzale A. Moro 5, 00185 Rome, Italy; (F.A.); (S.C.); (C.S.); (M.B.)
| |
Collapse
|
14
|
Onajole OK, Lun S, Yun YJ, Langue DY, Jaskula-Dybka M, Flores A, Frazier E, Scurry AC, Zavala A, Arreola KR, Pierzchalski B, Ayitou AJL, Bishai WR. Design, synthesis, and biological evaluation of novel imidazo[1,2-a]pyridinecarboxamides as potent anti-tuberculosis agents. Chem Biol Drug Des 2020; 96:1362-1371. [PMID: 32515129 DOI: 10.1111/cbdd.13739] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/13/2020] [Accepted: 05/31/2020] [Indexed: 12/14/2022]
Abstract
Tuberculosis (TB) is a highly infectious disease that has been plaguing the human race for centuries. The emergence of multidrug-resistant strains of TB has been detrimental to the fight against tuberculosis with very few safe therapeutic options available. As part of an ongoing effort to identify potent anti-tuberculosis agents, we synthesized and screened a series of novel imidazo[1,2-a]pyridinecarboxamide derivatives for their anti-tuberculosis properties. These compounds were designed based on reported anti-tuberculosis properties of the indolecarboxamides (I2Cs) and imidazo[1,2-a]pyridinecarboxamides (IPAs). In this series, we identified compounds 15 and 16 with excellent anti-TB activity against H37Rv strain of tuberculosis (MIC = 0.10-0.19 μM); these compounds were further screened against selected clinical isolates of Mtb. Compounds 15 and 16 showed excellent activities against multidrug-resistant (MDR) and extensively drug-resistant (XDR) strains of TB (MIC range: 0.05-1.5 μM) with excellent selectivity indices. In addition, preliminary ADME studies on compound 16 showed favorable pharmacokinetic properties.
Collapse
Affiliation(s)
- Oluseye K Onajole
- Department of Biological, Physical and Health Sciences, Roosevelt University, Chicago, IL, USA
| | - Shichun Lun
- Division of Infectious Disease, Department of Medicine, Center for Tuberculosis Research, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Young Ju Yun
- Department of Chemistry, Illinois Institute of Technology, Chicago, IL, USA
| | - Damkam Y Langue
- Department of Biological, Physical and Health Sciences, Roosevelt University, Chicago, IL, USA
| | - Michelle Jaskula-Dybka
- Department of Biological, Physical and Health Sciences, Roosevelt University, Chicago, IL, USA
| | - Adrian Flores
- Department of Biological, Physical and Health Sciences, Roosevelt University, Chicago, IL, USA
| | - Eriel Frazier
- Department of Biological, Physical and Health Sciences, Roosevelt University, Chicago, IL, USA
| | - Ashle C Scurry
- Department of Biological, Physical and Health Sciences, Roosevelt University, Chicago, IL, USA
| | - Ambernice Zavala
- Department of Biological, Physical and Health Sciences, Roosevelt University, Chicago, IL, USA
| | - Karen R Arreola
- Department of Biological, Physical and Health Sciences, Roosevelt University, Chicago, IL, USA
| | - Bryce Pierzchalski
- Department of Biological, Physical and Health Sciences, Roosevelt University, Chicago, IL, USA
| | - A Jean-Luc Ayitou
- Department of Chemistry, Illinois Institute of Technology, Chicago, IL, USA
| | - William R Bishai
- Division of Infectious Disease, Department of Medicine, Center for Tuberculosis Research, Johns Hopkins School of Medicine, Baltimore, MD, USA.,Howard Hughes Medical Institute, Chevy Chase, MD, USA
| |
Collapse
|
15
|
|
16
|
Sofi FA, Dubey G, Sharma R, Das P, Bharatam PV. Palladium-catalyzed aminocarbonylation of 2-phenyimidazo[1,2-a] pyridines using chloroform as carbon monoxide source and their mechanistic studies. Tetrahedron 2020. [DOI: 10.1016/j.tet.2020.131060] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
17
|
Lee BS, Sviriaeva E, Pethe K. Targeting the cytochrome oxidases for drug development in mycobacteria. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2020; 152:45-54. [PMID: 32081616 DOI: 10.1016/j.pbiomolbio.2020.02.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 01/15/2020] [Accepted: 02/06/2020] [Indexed: 11/19/2022]
Abstract
Mycobacterium tuberculosis strictly depends on oxygen to multiply, and the terminal oxidases are a vital part of the oxidative phosphorylation pathway. The bacterium possesses two aerobic respiratory branches: a cytochrome bcc-aa3 and a bacteria-specific cytochrome bd oxidase. The identification of small-molecule inhibitors of the cytochrome bcc-aa3 under numerous experimental conditions reflects the essentiality of the pathway for the optimum growth of M. tuberculosis. Recent findings on the biology of the cytochrome bcc-aa3 as well as the report of the first high-resolution structure of a mycobacterial cytochrome bcc-aa3 complex will help in the characterization and further development of potent inhibitors. Although the aerobic cytochrome bd respiratory branch is not strictly essential for growth, the discovery of a strong synthetic lethal interaction with the cytochrome bcc-aa3 placed the cytochrome bd oxidase under the spotlight as an attractive drug target for its synergistic role in potentiating the efficacy of cytochrome bcc-aa3 inhibitors and other drugs targeting oxidative phosphorylation. In this review, we are discussing current knowledge about the two mycobacterial aerobic respiratory branches, their potential as drug targets, as well as potential drawbacks.
Collapse
Affiliation(s)
- Bei Shi Lee
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Ekaterina Sviriaeva
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Kevin Pethe
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, 636921, Singapore.
| |
Collapse
|
18
|
Moraski GC, Deboosère N, Marshall KL, Weaver HA, Vandeputte A, Hastings C, Woolhiser L, Lenaerts AJ, Brodin P, Miller MJ. Intracellular and in vivo evaluation of imidazo[2,1-b]thiazole-5-carboxamide anti-tuberculosis compounds. PLoS One 2020; 15:e0227224. [PMID: 31905374 PMCID: PMC6944458 DOI: 10.1371/journal.pone.0227224] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/13/2019] [Indexed: 01/02/2023] Open
Abstract
The imidazo[2,1-b]thiazole-5-carboxamides (ITAs) are a promising class of anti-tuberculosis agents shown to have potent activity in vitro and to target QcrB, a key component of the mycobacterial cytochrome bcc-aa3 super complex critical for the electron transport chain. Herein we report the intracellular macrophage potency of nine diverse ITA analogs with MIC values ranging from 0.0625-2.5 μM and mono-drug resistant potency ranging from 0.0017 to 7 μM. The in vitro ADME properties (protein binding, CaCo-2, human microsomal stability and CYP450 inhibition) were determined for an outstanding compound of the series, ND-11543. ND-11543 was tolerable at >500 mg/kg in mice and at a dose of 200 mg/kg displayed good drug exposure in mice with an AUC(0-24h) >11,700 ng·hr/mL and a >24 hr half-life. Consistent with the phenotype observed with other QcrB inhibitors, compound ND-11543 showed efficacy in a chronic murine TB infection model when dosed at 200 mg/kg for 4 weeks. The efficacy was not dependent upon exposure, as pre-treatment with a known CYP450-inhibitor did not substantially improve efficacy. The ITAs are an interesting scaffold for the development of new anti-TB drugs especially in combination therapy based on their favorable properties and novel mechanism of action.
Collapse
Affiliation(s)
- Garrett C. Moraski
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, United States of America
| | - Nathalie Deboosère
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 –UMR 8204 –CIIL–Center for Infection and Immunity of Lille, Lille, France
| | - Kate L. Marshall
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, United States of America
| | - Heath A. Weaver
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, United States of America
| | - Alexandre Vandeputte
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 –UMR 8204 –CIIL–Center for Infection and Immunity of Lille, Lille, France
| | - Courtney Hastings
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Lisa Woolhiser
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Anne J. Lenaerts
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Priscille Brodin
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 –UMR 8204 –CIIL–Center for Infection and Immunity of Lille, Lille, France
| | - Marvin J. Miller
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, United States of America
| |
Collapse
|
19
|
Wang J, Zhu J, Zhou A. One-pot synthesis of imidazo[1,2-α]pyridine thioethers using imidazo[1,2-α]pyridines, arylsulfonyl chlorides and hydrazine. PHOSPHORUS SULFUR 2019. [DOI: 10.1080/10426507.2019.1686376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Jin Wang
- Department of Pharmaceutical Engineering, Pharmacy School, Jiangsu University, Zhenjiang City, Jiangsu, China
| | - Jie Zhu
- Department of Pharmaceutical Engineering, Pharmacy School, Jiangsu University, Zhenjiang City, Jiangsu, China
| | - Aihua Zhou
- Department of Pharmaceutical Engineering, Pharmacy School, Jiangsu University, Zhenjiang City, Jiangsu, China
| |
Collapse
|
20
|
Harrison GA, Mayer Bridwell AE, Singh M, Jayaraman K, Weiss LA, Kinsella RL, Aneke JS, Flentie K, Schene ME, Gaggioli M, Solomon SD, Wildman SA, Meyers MJ, Stallings CL. Identification of 4-Amino-Thieno[2,3- d]Pyrimidines as QcrB Inhibitors in Mycobacterium tuberculosis. mSphere 2019; 4:e00606-19. [PMID: 31511370 PMCID: PMC6739496 DOI: 10.1128/msphere.00606-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 12/24/2022] Open
Abstract
Antibiotic resistance is a global crisis that threatens our ability to treat bacterial infections, such as tuberculosis, caused by Mycobacterium tuberculosis Of the 10 million cases of tuberculosis in 2017, approximately 19% of new cases and 43% of previously treated cases were caused by strains of M. tuberculosis resistant to at least one frontline antibiotic. There is a clear need for new therapies that target these genetically resistant strains. Here, we report the discovery of a new series of antimycobacterial compounds, 4-amino-thieno[2,3-d]pyrimidines, that potently inhibit the growth of M. tuberculosis To elucidate the mechanism by which these compounds inhibit M. tuberculosis, we selected for mutants resistant to a representative 4-amino-thieno[2,3-d]pyrimidine and sequenced these strains to identify the mutations that confer resistance. We isolated a total of 12 resistant mutants, each of which harbored a nonsynonymous mutation in the gene qcrB, which encodes a subunit of the electron transport chain (ETC) enzyme cytochrome bc1 oxidoreductase, leading us to hypothesize that 4-amino-thieno[2,3-d]pyrimidines target this enzyme complex. We found that addition of 4-amino-thieno[2,3-d]pyrimidines to M. tuberculosis cultures resulted in a decrease in ATP levels, supporting our model that these compounds inhibit the M. tuberculosis ETC. Furthermore, 4-amino-thieno[2,3-d]pyrimidines had enhanced activity against a mutant of M. tuberculosis deficient in cytochrome bd oxidase, which is a hallmark of cytochrome bc1 inhibitors. Therefore, 4-amino-thieno[2,3-d]pyrimidines represent a novel series of QcrB inhibitors that build on the growing number of chemical scaffolds that are able to inhibit the mycobacterial cytochrome bc1 complex.IMPORTANCE The global tuberculosis (TB) epidemic has been exacerbated by the rise in drug-resistant TB cases worldwide. To tackle this crisis, it is necessary to identify new vulnerable drug targets in Mycobacterium tuberculosis, the causative agent of TB, and develop compounds that can inhibit the bacterium through novel mechanisms of action. The QcrB subunit of the electron transport chain enzyme cytochrome bc1 has recently been validated to be a potential drug target. In the current work, we report the discovery of a new class of QcrB inhibitors, 4-amino-thieno[2,3-d]pyrimidines, that potently inhibit M. tuberculosis growth in vitro These compounds are chemically distinct from previously reported QcrB inhibitors, and therefore, 4-amino-thieno[2,3-d]pyrimidines represent a new scaffold that can be exploited to inhibit this drug target.
Collapse
Affiliation(s)
- Gregory A Harrison
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Anne E Mayer Bridwell
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Megh Singh
- Center for World Health and Medicine, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Keshav Jayaraman
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Leslie A Weiss
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Rachel L Kinsella
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Janessa S Aneke
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Kelly Flentie
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Miranda E Schene
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Margaret Gaggioli
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Samantha D Solomon
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Scott A Wildman
- University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Marvin J Meyers
- Center for World Health and Medicine, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
- Department of Chemistry, Saint Louis University, Saint Louis, Missouri, USA
| | - Christina L Stallings
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
21
|
Reddyrajula R, Dalimba UK. Structural modification of zolpidem led to potent antimicrobial activity in imidazo[1,2-a]pyridine/pyrimidine-1,2,3-triazoles. NEW J CHEM 2019. [DOI: 10.1039/c9nj03462e] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
New imidazo[1,2-a]pyridine/pyrimidine-1,2,3-triazoles (IPTs) designed by specific structural modifications of zolpidem exhibited superior antitubercular activity than the parent compound.
Collapse
Affiliation(s)
- Rajkumar Reddyrajula
- Organic Chemistry Laboratory
- Department of Chemistry
- National Institute of Technology Karnataka
- Mangalore-575025
- India
| | - Udaya Kumar Dalimba
- Organic Chemistry Laboratory
- Department of Chemistry
- National Institute of Technology Karnataka
- Mangalore-575025
- India
| |
Collapse
|
22
|
Nitha PR, Joseph MM, Gopalan G, Maiti KK, Radhakrishnan KV, Das P. Chloroform as a carbon monoxide source in palladium-catalyzed synthesis of 2-amidoimidazo[1,2-a]pyridines. Org Biomol Chem 2018; 16:6430-6437. [PMID: 30132779 DOI: 10.1039/c8ob01486h] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A palladium-catalyzed aminocarbonylation strategy exploiting chloroform as a CO source has been developed for the synthesis of biologically potent 2-amidoimidazopyridine scaffolds. The aminocarbonylation reaction was found to be general with a range of amines and substituted imidazopyridines. Preliminary biological evaluation of cytotoxicity on selected examples provides scope for future investigations.
Collapse
Affiliation(s)
- P R Nitha
- National Institute for Interdisciplinary Science and Technology (CSIR), Thiruvananthapuram-19, India.
| | | | | | | | | | | |
Collapse
|
23
|
Lee BS, Pethe K. Therapeutic potential of promiscuous targets in Mycobacterium tuberculosis. Curr Opin Pharmacol 2018; 42:22-26. [PMID: 30015177 DOI: 10.1016/j.coph.2018.06.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/31/2018] [Accepted: 06/20/2018] [Indexed: 11/16/2022]
Abstract
In the field of tuberculosis drug development, the term 'promiscuous' was coined to collectively describe targets that repeatedly show up in whole-cell screenings. With the current climate leaning towards the exclusion of these targets in future drug screens, this review discusses and clarifies misconceptions surrounding this classification, the prospects of developing compounds targeting promiscuous targets, and their potential impact on tuberculosis drug development. The dominance of these targets in cell-based screens reflect not only bias introduced by experimental setup, but also some of the pathogen's greatest vulnerabilities. Coupled with favourable predictions of their in vivo efficacies and synergism with other TB drugs, these targets open opportunities to be explored for the development of rational drug combination for tuberculosis.
Collapse
Affiliation(s)
- Bei Shi Lee
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Kevin Pethe
- School of Biological Sciences, Nanyang Technological University, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Experimental Medicine Building, Singapore.
| |
Collapse
|
24
|
Imidazopyridine Compounds Inhibit Mycobacterial Growth by Depleting ATP Levels. Antimicrob Agents Chemother 2018; 62:AAC.02439-17. [PMID: 29632008 PMCID: PMC5971599 DOI: 10.1128/aac.02439-17] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 03/27/2018] [Indexed: 01/13/2023] Open
Abstract
The imidazopyridines are a promising new class of antitubercular agents with potent activity in vitro and in vivo. We isolated mutants of Mycobacterium tuberculosis resistant to a representative imidazopyridine; the mutants had large shifts (>20-fold) in MIC. Whole-genome sequencing revealed mutations in Rv1339, a hypothetical protein of unknown function. We isolated mutants resistant to three further compounds from the series; resistant mutants isolated from two of the compounds had single nucleotide polymorphisms in Rv1339 and resistant mutants isolated from the third compound had single nucleotide polymorphisms in QcrB, the proposed target for the series. All the strains were resistant to two compounds, regardless of the mutation, and a strain carrying the QcrB T313I mutation was resistant to all of the imidazopyridine derivatives tested, confirming cross-resistance. By monitoring pH homeostasis and ATP generation, we confirmed that compounds from the series were targeting QcrB; imidazopyridines disrupted pH homeostasis and depleted ATP, providing further evidence of an effect on the electron transport chain. A representative compound was bacteriostatic against replicating bacteria, consistent with a mode of action against QcrB. The series had a narrow inhibitory spectrum, with no activity against other bacterial species. No synergy or antagonism was seen with other antituberculosis drugs under development. In conclusion, our data support the hypothesis that the imidazopyridine series functions by reducing ATP generation via inhibition of QcrB.
Collapse
|
25
|
Campaniço A, Moreira R, Lopes F. Drug discovery in tuberculosis. New drug targets and antimycobacterial agents. Eur J Med Chem 2018; 150:525-545. [DOI: 10.1016/j.ejmech.2018.03.020] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/05/2018] [Accepted: 03/06/2018] [Indexed: 01/24/2023]
|
26
|
Bown L, Srivastava SK, Piercey BM, McIsaac CK, Tahlan K. Mycobacterial Membrane Proteins QcrB and AtpE: Roles in Energetics, Antibiotic Targets, and Associated Mechanisms of Resistance. J Membr Biol 2017; 251:105-117. [DOI: 10.1007/s00232-017-9997-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/20/2017] [Indexed: 02/08/2023]
|
27
|
Moraski GC, Bristol R, Seeger N, Boshoff HI, Tsang PSY, Miller MJ. Preparation and Evaluation of Potent Pentafluorosulfanyl-Substituted Anti-Tuberculosis Compounds. ChemMedChem 2017; 12:1108-1115. [PMID: 28654200 PMCID: PMC5603227 DOI: 10.1002/cmdc.201700170] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/13/2017] [Indexed: 01/10/2023]
Abstract
The global fight to stop tuberculosis (TB) remains a great challenge, particularly with the increase in drug-resistant strains and a lack of funding to support the development of new treatments. To bolster a precarious drug pipeline, we prepared a focused panel of eight pentafluorosulfanyl (SF5 ) compounds which were screened for their activity against Mycobacterium tuberculosis (Mtb) H37Rv in three different assay conditions and media. All eight compounds had sub-micromolar potency, and four displayed MICs <100 nm. Seven compounds were evaluated against non-replicating and mono-drug-resistant Mtb, and for their ability to inhibit Mtb within the macrophage. The greatest potency was observed against intracellular Mtb (MIC <10 nm for three compounds), which is often the most challenging to target. In general, the SF5 -bearing compounds were very similar to their CF3 counterparts, with the major differences observed being their in vitro ADME properties. Two SF5 -bearing compounds were found to have greater protein binding than their corresponding CF3 counterparts, but were also less metabolized in human microsomes, resulting in longer half-lives.
Collapse
Affiliation(s)
- Garrett C Moraski
- Department of Chemistry and Biochemistry, 103 Chemistry and Biochemistry, Montana State University, Bozeman, MT, 59717, USA
| | - Ryan Bristol
- Department of Chemistry and Biochemistry, 103 Chemistry and Biochemistry, Montana State University, Bozeman, MT, 59717, USA
| | - Natalie Seeger
- Department of Chemistry and Biochemistry, 103 Chemistry and Biochemistry, Montana State University, Bozeman, MT, 59717, USA
| | - Helena I Boshoff
- Tuberculosis Research Section, Laboratory of Clinical Infectious Diseases (LCID), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), 33 North Drive, Bethesda, MD, 20892, USA
| | - Patricia Siu-Yee Tsang
- Tuberculosis Research Section, Laboratory of Clinical Infectious Diseases (LCID), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), 33 North Drive, Bethesda, MD, 20892, USA
| | - Marvin J Miller
- Department of Chemistry and Biochemistry, University of Notre Dame, 347 Nieuwland Hall of Science, Notre Dame, IN, 46556, USA
| |
Collapse
|
28
|
Kumar V, Patel S, Jain R. New structural classes of antituberculosis agents. Med Res Rev 2017; 38:684-740. [DOI: 10.1002/med.21454] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 04/03/2017] [Accepted: 05/02/2017] [Indexed: 01/02/2023]
Affiliation(s)
- Vajinder Kumar
- Department of Medicinal Chemistry; National Institute of Pharmaceutical Education and Research; S.A.S. Nagar Punjab India
- Present address: Department of Chemistry; Akal University; Talwandi Sabo Punjab 151 302 India
| | - Sanjay Patel
- Department of Medicinal Chemistry; National Institute of Pharmaceutical Education and Research; S.A.S. Nagar Punjab India
| | - Rahul Jain
- Department of Medicinal Chemistry; National Institute of Pharmaceutical Education and Research; S.A.S. Nagar Punjab India
| |
Collapse
|
29
|
Anti-tubercular drug discovery: in silico implications and challenges. Eur J Pharm Sci 2017; 104:1-15. [PMID: 28341614 DOI: 10.1016/j.ejps.2017.03.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/08/2017] [Accepted: 03/19/2017] [Indexed: 12/18/2022]
Abstract
Tuberculosis (TB) has been reported as a major public health concern, especially in the developing countries. WHO report on tuberculosis 2016 shows a high mortality rate caused by TB leading to 1.8 million deaths worldwide (including deaths due to TB in HIV positive individuals), which is one of the top 10 causes of mortality in 2015. However, the main therapy used for the treatment of TB is still the Direct Observed Therapy Short-course (DOTS) that consists of four main first-line drugs. Due to the prolonged and unorganized use of these drugs, Mycobacterium tuberculosis (Mtb) has developed drug-resistance against them. To overcome this drug-resistance, efforts are continuously being made to develop new therapeutics. New drug-targets of Mtb are pursued by the researchers to develop their inhibitors. For this, new methodologies that comprise of the computational drug designing techniques are vigorously applied. A major limitation that is found with these techniques is the inability of the newly identified target-based inhibitors to inhibit the whole cell bacteria. A foremost factor for this limitation is the inability of these inhibitors to penetrate the bacterial cell wall. In this regard, various strategies to overcome this limitation have been discussed in detail in this review, along with new targets and new methodologies. A bunch of in silico tools available for the prediction of physicochemical properties that need to be explored to deal with the permeability issue of the Mtb inhibitors has also been discussed.
Collapse
|
30
|
Devi N, Singh D, Kaur G, Mor S, Putta VPRK, Polina S, Malakar CC, Singh V. In(OTf)3 assisted synthesis of β-carboline C-3 tethered imidazo[1,2-a]azine derivatives. NEW J CHEM 2017. [DOI: 10.1039/c6nj03210a] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Synthesis of β-carboline based natural products and synthetic derivatives is one of the frontier areas of research owing to their medicinal properties.
Collapse
Affiliation(s)
- Nisha Devi
- Department of Chemistry
- National Institute of Technology (NIT) Jalandhar
- India
| | - Dharmender Singh
- Department of Chemistry
- National Institute of Technology (NIT) Jalandhar
- India
| | - Gurpreet Kaur
- Department of Chemistry
- National Institute of Technology (NIT) Jalandhar
- India
| | - Satbir Mor
- Department of Chemistry
- GJ University of Science and Technology Hisar
- India
| | | | - Saibabu Polina
- Department of Medicinal Chemistry
- Jubilant Biosys
- Bangalore
- India
| | - Chandi C. Malakar
- Department of Chemistry
- National Institute of Technology (NIT) Manipur
- Imphal
- India
| | - Virender Singh
- Department of Chemistry
- National Institute of Technology (NIT) Jalandhar
- India
| |
Collapse
|
31
|
In(OTf)3-HBF4Assisted Multicomponent Approach for One-Pot Synthesis of Pyrazolopyridinone Fused Imidazopyridines. ChemistrySelect 2016. [DOI: 10.1002/slct.201601133] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
32
|
Phummarin N, Boshoff HI, Tsang PS, Dalton J, Wiles S, Barry Rd CE, Copp BR. SAR and identification of 2-(quinolin-4-yloxy)acetamides as Mycobacterium tuberculosis cytochrome bc1 inhibitors. MEDCHEMCOMM 2016; 7:2122-2127. [PMID: 28337336 PMCID: PMC5292992 DOI: 10.1039/c6md00236f] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 08/17/2016] [Indexed: 12/13/2022]
Abstract
New antimycobacterial 2-(quinoline-4-yloxy)acetamides were prepared, and using gene deletion and resistant mutants, we conclude that the compound class inhibits the mycobacterial cytochrome bc1 complex.
A previous phenotypic screen by GSK identified 2-(quinolin-4-yloxy)acetamides as potent growth inhibitors of Mycobacterium tuberculosis (Mtb). We report the results of a preliminary structure–activity relationship (SAR) study of the compound class which has yielded more potent inhibitors. An Mtb cytochrome bd oxidase deletion mutant (cydKO) was found to be hypersensitive to most members of the compound library, while strains carrying single-nucleotide polymorphisms of the qcrB gene, which encodes a subunit of the menaquinol cytochrome c oxidoreductase (bc1) complex, were resistant to the library. These results identify that the 2-(quinolin-4-yloxy)acetamide class of Mtb growth inhibitors can be added to the growing number of scaffolds that target the M. tuberculosis bc1 complex.
Collapse
Affiliation(s)
- Narisa Phummarin
- School of Chemical Sciences , University of Auckland , Private Bag 92019 , Auckland 1142 , New Zealand . ; ; Tel: +64 9 3737599
| | - Helena I Boshoff
- Tuberculosis Research Section , Laboratory of Clinical Infectious Diseases , National Institute of Allergy and Infectious Disease , National Institutes of Health , Bethesda , Maryland , USA
| | - Patricia S Tsang
- Tuberculosis Research Section , Laboratory of Clinical Infectious Diseases , National Institute of Allergy and Infectious Disease , National Institutes of Health , Bethesda , Maryland , USA
| | - James Dalton
- Bioluminescent Superbugs Lab , Department of Molecular Medicine and Pathology , University of Auckland , Private Bag 92019 , Auckland 1142 , New Zealand
| | - Siouxsie Wiles
- Bioluminescent Superbugs Lab , Department of Molecular Medicine and Pathology , University of Auckland , Private Bag 92019 , Auckland 1142 , New Zealand ; Te Pūnaha Matatini , c/o University of Auckland , Private Bag 92019 , Auckland 1142 , New Zealand
| | - Clifton E Barry Rd
- Tuberculosis Research Section , Laboratory of Clinical Infectious Diseases , National Institute of Allergy and Infectious Disease , National Institutes of Health , Bethesda , Maryland , USA
| | - Brent R Copp
- School of Chemical Sciences , University of Auckland , Private Bag 92019 , Auckland 1142 , New Zealand . ; ; Tel: +64 9 3737599
| |
Collapse
|
33
|
Imidazo[1,2-a]Pyridine-3-Carboxamides Are Active Antimicrobial Agents against Mycobacterium avium Infection In Vivo. Antimicrob Agents Chemother 2016; 60:5018-22. [PMID: 27216051 DOI: 10.1128/aac.00618-16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/13/2016] [Indexed: 11/20/2022] Open
Abstract
A panel of six imidazo[1,2-a]pyridine-3-carboxamides (IAPs) were shown to have low-micromolar activity against Mycobacterium avium strains. Compound ND-10885 (compound 2) showed significant activity in the lung, spleen, and liver in a mouse M. avium infection model. A combined regimen consisting of ND-10885 (compound 2) and rifampin was additive in its anti-M. avium activity in the lung. Our data indicate that IAPs represent a new class of antibiotics that are active against M. avium and could potentially serve as an effective addition to a combined treatment regimen.
Collapse
|
34
|
Pulipati L, Sridevi JP, Yogeeswari P, Sriram D, Kantevari S. Synthesis and antitubercular evaluation of novel dibenzo[ b , d ]thiophene tethered imidazo[1,2- a ]pyridine-3-carboxamides. Bioorg Med Chem Lett 2016; 26:3135-3140. [DOI: 10.1016/j.bmcl.2016.04.088] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 04/27/2016] [Accepted: 04/29/2016] [Indexed: 10/21/2022]
|
35
|
Hoagland DT, Liu J, Lee RB, Lee RE. New agents for the treatment of drug-resistant Mycobacterium tuberculosis. Adv Drug Deliv Rev 2016; 102:55-72. [PMID: 27151308 PMCID: PMC4903924 DOI: 10.1016/j.addr.2016.04.026] [Citation(s) in RCA: 236] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 04/20/2016] [Accepted: 04/22/2016] [Indexed: 02/06/2023]
Abstract
Inadequate dosing and incomplete treatment regimens, coupled with the ability of the tuberculosis bacilli to cause latent infections that are tolerant of currently used drugs, have fueled the rise of multidrug-resistant tuberculosis (MDR-TB). Treatment of MDR-TB infections is a major clinical challenge that has few viable or effective solutions; therefore patients face a poor prognosis and years of treatment. This review focuses on emerging drug classes that have the potential for treating MDR-TB and highlights their particular strengths as leads including their mode of action, in vivo efficacy, and key medicinal chemistry properties. Examples include the newly approved drugs bedaquiline and delaminid, and other agents in clinical and late preclinical development pipeline for the treatment of MDR-TB. Herein, we discuss the challenges to developing drugs to treat tuberculosis and how the field has adapted to these difficulties, with an emphasis on drug discovery approaches that might produce more effective agents and treatment regimens.
Collapse
Affiliation(s)
- Daniel T Hoagland
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Pharmaceutical Sciences Graduate Program, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jiuyu Liu
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Robin B Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Richard E Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
36
|
Moraski GC, Seeger N, Miller PA, Oliver AG, Boshoff HI, Cho S, Mulugeta S, Anderson JR, Franzblau SG, Miller MJ. Arrival of Imidazo[2,1-b]thiazole-5-carboxamides: Potent Anti-tuberculosis Agents That Target QcrB. ACS Infect Dis 2016; 2:393-8. [PMID: 27627627 DOI: 10.1021/acsinfecdis.5b00154] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Increasing interest in the potent anti-tuberculosis activity and the novel target (QcrB) of imidazo[1,2-a]pyridine-3-carboxamides encouraged extended structure-activity relationship studies of additional scaffolds. This study reports on the in vitro profiling of the imidazo[2,1-b]thiazole-5-carboxamides as a new promising class of anti-tuberculosis compounds endowed with nanomolar potency against replicating and drug-resistant Mycobacterium tuberculosis (Mtb) as well as low toxicity to VERO cells. Compounds 6, 16, and 17 had MIC values <10 nM and toxicity >100 μM. On-target selectivity of this series was confirmed by cross-resistance of specific QcrB mutants as well as the hypersusceptibility of a mutant with a functional gene deletion of the alternative cytochrome bd oxidase. Additionally, to demonstrate selectivity, three analogues (6, 15, 17) were broadly screened against a diverse set of eight strains of bacteria, including both Gram-positive and Gram-negative as well as six disease-causing non-tuberculosis mycobacteria. Finally, compounds 16 and 17 were found to be active in macrophages infected with Mtb.
Collapse
Affiliation(s)
- Garrett C. Moraski
- Department of Chemistry and Biochemistry, Montana State University, 103 Chemistry and Biochemistry Building, Bozeman, Montana 59717, United States
| | - Natalie Seeger
- Department of Chemistry and Biochemistry, Montana State University, 103 Chemistry and Biochemistry Building, Bozeman, Montana 59717, United States
| | - Patricia A. Miller
- Department of Chemistry and Biochemistry, University of Notre Dame, 251 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| | - Allen G. Oliver
- Department of Chemistry and Biochemistry, University of Notre Dame, 251 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| | - Helena I. Boshoff
- Tuberculosis Research Section, Laboratory of Clinical
Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Sanghyun Cho
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612, United States
| | - Surafel Mulugeta
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612, United States
| | - Jeffery R. Anderson
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612, United States
| | - Scott G. Franzblau
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612, United States
| | - Marvin J. Miller
- Department of Chemistry and Biochemistry, University of Notre Dame, 251 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| |
Collapse
|
37
|
Madaiah M, Prashanth MK, Revanasiddappa HD, Veeresh B. Synthesis and evaluation of novel imidazo[4,5-c]pyridine derivatives as antimycobacterial agents against Mycobacterium tuberculosis. NEW J CHEM 2016. [DOI: 10.1039/c6nj02069k] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Novel imidazo[4,5-c]pyridine derivatives showed cytotoxicity and decreased the bacterial load in lung and spleen tissues in the in vivo animal model.
Collapse
Affiliation(s)
| | | | | | - Bantal Veeresh
- Department of Pharmacology
- G. Pullareddy College of Pharmacy
- Mehdipatnam
- India
| |
Collapse
|
38
|
Abstract
The practice of medicine was profoundly transformed by the introduction of the antibiotics (compounds isolated from Nature) and the antibacterials (compounds prepared by synthesis) for the control of bacterial infection. As a result of the extraordinary success of these compounds over decades of time, a timeless biological activity for these compounds has been presumed. This presumption is no longer. The inexorable acquisition of resistance mechanisms by bacteria is retransforming medical practice. Credible answers to this dilemma are far better recognized than they are being implemented. In this perspective we examine (and in key respects, reiterate) the chemical and biological strategies being used to address the challenge of bacterial resistance.
Collapse
Affiliation(s)
- Jed F. Fisher
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame IN 46556–5670, USA
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame IN 46556–5670, USA
| |
Collapse
|