Novel, potent, orally bioavailable and selective mycobacterial ATP synthase inhibitors that demonstrated activity against both replicating and non-replicating M. tuberculosis

https://doi.org/10.1016/j.bmc.2014.12.060Get rights and content

Abstract

The mycobacterial F0F1-ATP synthase (ATPase) is a validated target for the development of tuberculosis (TB) therapeutics. Therefore, a series of eighteen novel compounds has been designed, synthesized and evaluated against Mycobacterium smegmatis ATPase. The observed ATPase inhibitory activities (IC50) of these compounds range between 0.36 and 5.45 μM. The lead compound 9d [N-(7-chloro-2-methylquinolin-4-yl)-N-(3-((diethylamino)methyl)-4-hydroxyphenyl)-2,3-dichlorobenzenesulfonamide] with null cytotoxicity (CC50 >300 μg/mL) and excellent anti-mycobacterial activity and selectivity (mycobacterium ATPase IC50 = 0.51 μM, mammalian ATPase IC50 >100 μM, and selectivity >200) exhibited a complete growth inhibition of replicating Mycobacterium tuberculosis H37Rv at 3.12 μg/mL. In addition, it also exhibited bactericidal effect (approximately 2.4 log10 reductions in CFU) in the hypoxic culture of non-replicating M. tuberculosis at 100 μg/mL (32-fold of its MIC) as compared to positive control isoniazid [approximately 0.2 log10 reduction in CFU at 5 μg/mL (50-fold of its MIC)]. The pharmacokinetics of 9d after p.o. and IV administration in male Sprague–Dawley rats indicated its quick absorption, distribution and slow elimination. It exhibited a high volume of distribution (Vss, 0.41 L/kg), moderate clearance (0.06 L/h/kg), long half-life (4.2 h) and low absolute bioavailability (1.72%). In the murine model system of chronic TB, 9d showed 2.12 log10 reductions in CFU in both lung and spleen at 173 μmol/kg dose as compared to the growth of untreated control group of Balb/C male mice infected with replicating M. tuberculosis H37Rv. The in vivo efficacy of 9d is at least double of the control drug ethambutol. These results suggest 9d as a promising candidate molecule for further preclinical evaluation against resistant TB strains.

Introduction

Tuberculosis, primarily caused by Mycobacterium tuberculosis, is one of the globally leading infectious disease that continues as global epidemic with more than 9 million new cases and nearly 2 million deaths every year. 1 In addition, over 2 billion people harbor latent TB infection (LTBI), thus representing an enormous reservoir of M. tuberculosis that can subsequently progress to active TB.1 The main obstacles to the global TB control are: (i) the human immunodeficiency virus (HIV) epidemic proven to increase the risk of developing active TB dramatically, (ii) the increasing emergence of multi-drug resistant TB (MDR-TB), extensively drug-resistant TB (XDR-TB) and a recently identified TB state called total drug-resistant TB (TDR-TB).1, 2, 3, 4, 5 Current TB chemotherapy is mainly based on drugs that inhibit bacterial metabolism, and is characterized by efficient bactericidal and extremely weak sterilizing activities.6 The dormant or latent or metabolically inactive bacilli are tolerant to the anti-TB drugs that target cell division.7, 8 Although an extended chemotherapy may be partially successful in eradicating such dormant bacilli, drug(s) with efficient sterilizing activity are of current demand for properly tackling the dormant bacilli. In such scenario, targeting the particular enzyme that are essential for the survival of both actively growing and latent/dormant bacilli, is one of the most effective target-based drug discovery approach.2

The mycobacterial F0F1-ATP synthase (F0F1-ATPase) is a validated anti-TB target, which is responsible for the production of adenosine 5′-triphosphate (ATP) for energy homeostasis in different mycobacterial species.9 Bedaquiline (TMC207, Sirturo™) is a member of the diarylquinoline class of anti-TB drugs targeting this new enzyme and has been approved by the U.S. Food and Drug Administration (US-FDA) specifically for the treatment of MDR-TB. This drug has shown promising efficacy against both the drug-sensitive and drug-resistant TB, and has remarkable potential for restricting the duration of TB treatment due to the fact that this drug is bactericidal to both the replicating as well as non-replicating (dormant) bacilli.9, 10, 11, 12 Bedaquiline (Sirturo™) has been reported by the World Health Organization (WHO) to disturb the function of the heart and liver in particular. Common observed side effects include nausea, joint and chest pain, and headache.13 Bedaquiline is proven to be a weak hERG (human Ether-à-go-go) potassium (K+) channel blocker (in vitro IC50 = 0.2 μg/mL) that can cause prolongation of the heart QT interval (the time between the start of the Q wave and the end of the T wave in the heart’s electrical cycle), thus disturbing normal function of the heart.14 The observed efficacy of bedaquiline for the treatment of MDR-TB validates the target mycobacterium F0F1-ATPase, and the observed adverse events with this drug suggest further exploration of new chemical entities as selective F0F1-ATPase inhibitors lacking any potential adverse effects.

Structurally, the mycobacterial F0F1-ATPase is composed of a membrane-embedded F0 portion comprising a1b2c10–15 subunits and a hydrophilic F1 portion consisting of α3β3δε.15 Proton (H+) migration through F0 triggers the rotation of rotary ring formed by oligomeric subunits ‘c’ which is coupled to the rotation of the ‘γ’ subunit resided within the (αβ)3 hexamer of F1. This biochemical phenomenon thus drives the synthesis of ATP as a source of energy for mycobacterium species. A high-resolution crystal structure of mycobacterial ATP synthase or its subunits is lacking till date. Computational and biological studies have shown that TMC207 binds at the interface of subunit ‘a’, and rotary subunits ‘c’.15, 16, 17 This drug is biochemically shown to interfere with the rotary movement of subunit ‘c’ by mimicking a conserved basic residue Arg186 in the subunit ‘a’ needed for the proton transfer with Glu61 of the subunit ‘c’.16, 17 Through detailed computational studies, de Jonge and colleagues16 reported the chiral N,N-dimethylpropanol substructure of TMC207 to exhibit H-bond formation with the carboxylate side chain of Glu61, and a favorable location of quinoline substructure at the interface of subunits ‘a’ and ‘c’ that might lead to blockade of the rotation of rotary ring, and thus normal interfering with the proton transfer cascade process. Haagsma et al.15 have recently characterized the interaction between TMC207 and mycobacterial ATP synthase using biochemical assays and binding studies, where they have provided experimental support for ability of TMC207 to mimic a key residue Arg186 involved in the proton transfer cascade, and thus blocking rotary movement of subunits ‘c’ of the mycobacterial F0F1-ATP synthase.15, 16

In view of the success of mycobacterial F0F1-ATP synthase as validated target for TB chemotherapy, we utilized the state-of-the-art medicinal chemistry approach for the identification of novel inhibitors of mycobacterial F0F1-ATP synthase. This effort led to invention of quinoline class of aryl-sulfonamides as potent, orally bioavailable and selective mycobacterial ATP synthase inhibitors that we have patented recently.18 We have recently reported a short report on the biological screening of these compounds against Mycobacterium smegmatis ATP synthase along with in vitro testing against the dormant (non-replicating) M. tuberculosis H37Rv.12 Herein, we report a full-length description of the rational design, synthesis and structure-activity relationship (SAR) studies of these novel compounds proven as low micromolar inhibitors of the mycobacterial F0F1-ATP synthase. Among the synthesized series of 18 compounds, two lead compounds were biologically explored in greater detail where one representative compound was proven as an orally bioavailable and selective mycobacterium ATP synthase inhibitor that demonstrated approximately 2.12 log10 reductions in CFU in both lung and spleen at a dose of 173 μmol/kg as compared to the growth in the untreated control group of Balb/C male mice infected with M. tuberculosis H37Rv. Like the drug bedaquiline (Sirturo™, TMC207), this compound acts through a novel mechanism of action (ATP synthase), and is active against both the replicating as well as non-replicating M. tuberculosis H37Rv. This lead compound could be considered as candidate molecule for further preclinical evaluation that could lead to an effective TB therapeutics.

Section snippets

Chemicals, reagents, culture media and test compounds

All of the chemicals, culture media, and reagents were purchased from common commercial suppliers. Solvents were purified and dried by standard procedures, when required. Chromatographic separations of the synthesized intermediates and title compounds were performed on silica gel (Merck: 100–200 mesh). Thin layer chromatography was used to monitor the progress and/or completion of the reactions. Melting points (uncorrected) were determined with Büchi 510 apparatus. Characterization of the

Rational design and structure-based optimization

Through detailed computational studies, de Jonge and colleagues reported the important structural features in TMC207 governing its ATP synthase inhibitory activity.16 They reported the chiral N,N-dimethylpropanol substructure to exhibit H-bond formation with the carboxylate side chain of Glu61 residue, and the favorable binding of the quinoline substructure at the interface of subunits ‘a’ and ‘c’ of the mycobacterium F0F1-ATP synthase. In view of this, we carried out the preliminary database

Discussions

Despite the availability of highly efficacious treatment(s) for decades, TB is still a major global health problem mainly due to the emergence of MDR-, XDR- and TDR-TBs, latent/dormant infection of TB and its synergy with HIV/AIDS. The mycobacterial F0F1-ATP synthase is a well validated target for TB after the success launch of bedaquiline (Sirturo™, TMC207) into the market as an effective drug for MDR-TB. Bedaquiline has shown positive efficacy against both the resistant (MDR and XDR) and

Conclusion

Despite the availability of highly efficacious treatment(s) for decades, TB is still a major global health problem mainly due to the emergence of MDR-, XDR- and TDR-TBs, latent/ dormant infection of TB and its synergy with HIV/AIDS. The mycobacterial F0F1-ATP synthase is an anti-TB target for the bedaquiline (Sirturo™), a drug recently approved by the US-FDA specifically for the treatment of MDR-TB. In our efforts to identify novel compounds as ATP synthase inhibitors, a total of eighteen new

Acknowledgments

The authors (S.S. and K.K.R.) are thankful to Ministry of Health (MOH) and Council of Scientific and Industrial Research (CSIR), New Delhi, respectively for financial assistance in terms of fellowship. The authors thank Dr. P. K. Shukla and Ms. Pratiksha Singh of Fermentation Technology Division, CDRI for screening lead compounds against bacteria and fungi. The Sophisticated Analytical Instrument Facility (SAIF) department is acknowledged for the characterization of compounds. The technical

References and notes (26)

  • S.R. Khan et al.

    Int. J. Antimicrob. Agents

    (2013)
  • J.J. Garcia et al.

    J. Biol. Chem.

    (2000)
  • T. Mosmann

    J. Immunol. Methods

    (1983)
  • World Health Organization. Global Tuberculosis Report. WHO Report 2013; WHO Press: Geneva, Switzerland,...
  • A. Koul et al.

    Nature

    (2011)
  • G.B. Migliori et al.

    Clin. Infect. Dis.

    (2012)
  • Z.F. Udwadia

    Thorax

    (2012)
  • Z.F. Udwadia et al.

    Clin. Infect. Dis.

    (2012)
  • D.A. Mitchison

    Am. J. Respir. Crit. Care Med.

    (2005)
  • H.M. Vandiviere et al.

    Am. J. Med. Sci.

    (1956)
  • L.G. Wayne et al.

    Infect. Immun.

    (1996)
  • K. Andries et al.

    Science

    (2005)
  • A. Koul et al.

    Nat. Chem. Biol.

    (2007)
  • Cited by (53)

    View all citing articles on Scopus

    CDRI communication number allotted to this paper is 8882.

    View full text