Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Sep 10.
Published in final edited form as: J Med Chem. 2020 Apr 20;63(17):8917–8955. doi: 10.1021/acs.jmedchem.9b02075

Molecule Property Analyses of Active Compounds for Mycobacterium Tuberculosis

Vadim Makarov 1, Elena Salina 1, Robert C Reynolds 2, Phyo Phyo Kyaw Zin 3,4, Sean Ekins 5
PMCID: PMC7702311  NIHMSID: NIHMS1619908  PMID: 32259446

Abstract

Tuberculosis (TB) continues to claim the lives of around 1.7 million people per year. Most concerning are the reports of multidrug drug resistance. Paradoxically, this global health pandemic is demanding new therapies when resources and interest are waning. However, continued tuberculosis drug discovery is critical to address the global health need and burgeoning multidrug resistance. Many diverse classes of antitubercular compounds have been identified with activity in vitro and in vivo. Our analyses of over 100 active leads are representative of thousands of active compounds generated over the past decade suggesting they come from few chemical classes or natural product sources. We are therefore repeatedly identifying compounds that are similar to those that preceded them. Our molecule-centred cheminformatics analyses points to the need to dramatically increase the diversity of chemical libraries tested and get outside of the historic Mtb property space if we are to generate novel improved antitubercular leads.

Keywords: Antituberculars, Drug Discovery, Tuberculosis

Graphical Abstract

graphic file with name nihms-1619908-f0001.jpg

INTRODUCTION

It is widely known that Mycobacterium tuberculosis (Mtb) infection is causative of tuberculosis (TB), and claims the lives of around 1.7 million people with 10.4 million infections reported per year.3 Hence TB continues to be the focus of many international efforts to develop new therapeutics to address resistance to the available first- and second-line drugs.4 Due to increasing TB morbidity in developed countries in the late 1990’s and the global expansion of drug-resistant strains, a high priority has since been given to the development of new TB drugs by the World Health Organization, Bill & Melinda Gates Foundation, National Institutes of Health as well as other international institutions and funds. While the widely used TB drugs were developed in the late 1950’s-1960’s (Table 1, compounds 1–4), it is only recently that the first new drugs (bedaquiline, delamanid and pretomanid) came on the market in over 40 years5, 6 (Table 1, compounds 5–7); these latter molecules are currently not widely used because of their cost (Table 1) and their use is limited to multidrug resistant Mtb (MDR-TB).7, 8 Interestingly, these three molecules also have the highest hydrophobicity (ALogP) out of the 7 approved TB drugs and no hydrogen bond donors which is unusual (Table S1).

Table 1.

First line and recently approved antituberculars.

Drug Year approved Pros (and target where known) Cons References
Isoniazid 1
graphic file with name nihms-1619908-t0002.jpg
1952 A prodrug activated by KatG. Inhibits synthesis of mycolic acids by targeting InhA.
In the United States a month of treatment costs less than $25.
Extensive side effects including neuropathy and liver enzyme effects. 9
Pyrazinamide 2
graphic file with name nihms-1619908-t0003.jpg
1954 Can penetrate granulomas. Kills non-replicating Mtb. A prodrug that is converted by pyrazinamidase (PncA) to the active pyrazinoic acid. PZA resistance exists. 10, 11
Ethambutol 3
graphic file with name nihms-1619908-t0004.jpg
1962 Inhibits the enzyme arabinosyl transferase. In the United States it costs $100 to 200 per month. Ocular side effects. 12
Rifampicin 4
graphic file with name nihms-1619908-t0005.jpg
1967 Targets RNA polymerase. In the United States costs about $120 per month. PXR inducer and leads to hepatotoxicity 13
Bedaquiline 5
graphic file with name nihms-1619908-t0006.jpg
2012 Blocks the proton pump for ATP synthase.1418 Discovered by M. smegmatis whole-cell screening19, MIC99 − 0.03 μg/ml Used for multidrug resistant TB. induce phospholipidosis at high doses20 potent inhibition of hERG increasing clinical QTc interval prolongation21. Costs $30,000 in high income countries. Mutations in atpE can lead to resistance 22, 23
Delamanid 6
graphic file with name nihms-1619908-t0007.jpg
2014 (EU) Blocks manufacture of mycolic acids in cell wall. Increased sputum culture conversion. Used for multidrug resistant TB. QT prolongation. Costs $30,000 in high income countries 23
Pretomanid 7
graphic file with name nihms-1619908-t0008.jpg
2019 (USA) Blocks manufacture of mycolic acids in cell wall.
Costs $364 for a six-month treatment.
Used in combination regimen with bedaquiline and linezolid for people with XDR-TB or treatment-intolerant/non-responsive MDR-TB 24, 25

The appearance and increasing number of Mtb strains with multiple drug resistance (i.e, MDR is defined as resistance to at least rifampicin (RPN) and isoniazid (INH)) and extensive drug resistance (i.e., XDR means MDR and additional resistance to at least one fluoroquinolone and one injectable second-line drug) represent a potential serious public health crisis. These intractable TB strains are associated with low cure rate and high mortality3, 26 as well as significant added treatment costs. Even more concerning, totally drug-resistant strains (TDR-TB) have been reported in clinical practice.27, 28

Widespread latent tuberculosis29 and the low potency of current TB drugs against latent disease represent one more weakness with current TB therapy.30, 31 According to the WHO, Mtb now exists as latent disease in approximately one quarter of the global population. Latent infection remains asymptomatic for a long time. Over a lifetime, it becomes active in about 5% of infected persons3, and reactivation is associated with a weakened immune system (including pediatric and geriatric cases) and in patients with compromised immune status, such as individuals undergoing immunosuppressive therapy or infected with HIV. HIV-positive patients carrying latent tuberculosis also have a several-fold higher risk of developing active tuberculosis (8–10 % annually). Therefore, although millions of cases of reactivated tuberculosis are reported worldwide, reliable methods of timely diagnosis and treatment of latent TB have not been developed. Thus, latent TB and reactivation risk represent a significant health issue. There is no doubt that the development of advanced TB drugs will play a key role in the reduction of morbidity and mortality levels required for achievement of WHO’s global international goals.32 This state of affairs and the general importance of this issue were also emphasized during a recent WHO Ministerial Conference held in Russia33, where TB represents a major healthcare challenge.

There are several other difficulties involved with Mtb drug discovery, namely the bacteria is slow growing, a BSL3 laboratory is required to work with the bacteria and studies are complicated by the need to assess MIC and intracellular killing in infected macrophages.34, 35 Ultimately in vivo assessment is also slow because of the length of infections required as well as the need for acute and chronic animal models. Subsequently, the pipeline for antituberculars, which is mostly driven by the TB Alliance, is by any standards very thin with approximately a dozen new tuberculosis drugs undergoing Phase 1–3 clinical studies currently as summarized by the working group on new TB drugs (Figure 1).

Figure 1.

Figure 1.

Current TB drug pipeline1, 2, * denotes new chemical class. Molecules in phase 3 have all been FDA (bedaquiline and pretomanid) or EU (delamanid) approved but also components of ongoing trials. Image courtesy of Dr. Barbara Laughon (NIAID) www.newtbdrugs.org.

Some of these drugs are expected to become viable products. In particular, six compounds to combat MDR-TB are being investigated within the framework of Phase II and III clinical trials. Bedaquiline (5, Sirturo®, Janssen Therapeutics, Titusville, NJ, USA), delamanid (6, Deltyba®, Otsuka Pharmaceuticals, Tokyo, Japan) and pretomanid (7, TB Alliance, USA) have been approved for MDR-TB, respectively and are still undergoing additional clinical trials as combinations with other drugs. Repurposing of the known drugs clofazimine and levofloxacin are in Phase II and III trials, as tuberculosis therapy.36, 37 Mtb strains resistant to some of these molecules have been already reported3840, which suggests the limited time window with a new drug before losing sensitivity in the field. This issue also points to the continued urgent need for rapid development of the most advanced TB drugs and continual feeding of the research pipeline. For all the decades and billions of dollars spent on tuberculosis drug discovery, this begs the question why is there not more to show for it? Let us begin by looking at the two of the most recent drugs to be approved for Mtb.

Nitroimidazoles as ‘new’ drugs for Mtb

Initially, nitroimidazoles were produced as bicyclic nitroimidazofuran derivatives developed for cancer chemotherapy as radiosensitizers.41 Once they were identified in 1989 to be potent against actively growing non-fissile tuberculosis, interest in this class of compounds took off and this work has been extensively reviewed.41 Delamanid (OPC-67683 (6)) and Pretomanid (PA-824 (7)) are examples of this class of compounds (Table 2) and there is continued interest in developing additional compounds in this series.42 This class of compounds is therefore far from new dating back 30 years. When you compare the two compounds it is apparent that they are very similar in terms of activity against drug sensitive and resistant Mtb.

Table 2.

Comparison of Delamanid and Pretomanid

Delamanid 6 Pretomanid 7
MIC (μg/ml) 0.012 μg/ml43 (MIC90) of 0.039 μg/ml44
Drug resistant clinical isolates MIC (μg/ml) ~0.01 μg/ml45 0.031–0.531 μg/ml44
Compound class nitro-dihydro-imidazooxazole nitroimidazopyran
Mechanism Non-mutagenic. Delamanid inhibited the synthesis of methoxy- and keto-mycolic acid.45 Protein synthesis and lipid synthesis are substantially inhibited.
Pretomanid is activated by a process that requires the unique deazaflavin F420 and depends on electron transport. Active against latent TB.
Human dosing Delamanid was well tolerated in initial studies.46 Delamanid was associated with an increase in sputum-culture conversion at 2 months among patients with multidrug-resistant tuberculosis.47 It was also shown to improve outcomes and survival in MDR TB.48 Maximum plasma levels in 4–5h, steady state after 5–6 days daily dosing, elimination half life 16–20 h49, causes creatinine to rise by inhibiting renal tubular creatinine secretion50, bactericidal from 200–1200 mg daily over 14 days51, a dose of 100 mg – 200 mg appears safe and effective52, pretomanid-moxifloxacin-pyrazinamide is potentially suitable for drug sensitive and MDR TB53, pretomanid does not inhibit CYP3A4 in a clinically relevant manner54, increase in exposure with high fat meal55, combination with bedaquiline and pyrazinamide indicates a new treatment approach56, moxifloxacin, pretomanid and pyrazinamide was safe and well tolerated and showed superior efficacy.57

Compound 6 is a nitroimidazoxazole that was developed45 for the treatment of MDR-TB. Compound 6 was developed from a smaller nitroimidazole58 and 6 inhibits biosynthesis of methoxy-mycolic and keto-mycolic acids (Table 2) which are essential components of the TB mycobacterial cellular wall. It is notable that, both 6 and 7 require metabolic activation by deazaflavin-dependent nitroreductase (Ddn) to provide anti-tubercular activity.59 Compound 6-resistant cells were characterized by Rv3547 (ddn) showing the gene encoding Ddn mutations and lacking the ability to provide metabolic activation of this compound.45 Mtb produces the inactive desnitro-imidazoxazole as the major metabolite of 6. We can infer that Rv3547 reduces the delamanid nitro group forming an intermediate between the 6 prodrug and the inactive terminal metabolite (i.e., desnitro-imidazoxazole).45 Compound 6 has potent anti-tubercular activity including against drug-resistant strains and no cross resistance has been reported with any current tuberculosis drug.60 Compound 7 is a bicyclic nitroimidazole, nitroimidazooxazine or nitroimidazopyran derivative. Studies have shown both bactericidal and sterilizing effects on drug-resistant, drug-susceptible Mtb strains and resting mycobacterial cells.61 In vitro studies using Mtb clinical multidrug resistant isolates and in vivo studies using mouse and guinea pig models demonstrated anti-tubercular activity.44, 62 According to mouse preclinical studies, threshold toxicity is more than 1000 mg/kg after single oral administration.62 Compound 7 does not demonstrate cross resistance with conventional tuberculosis drugs.44 Compound 7 is transformed into three major metabolites causing a release of nitric oxide (NO, a natural antibiotic released by macrophages63). Similar to isoniazid, 7 inhibits cell wall growth by affecting the biosynthesis of mycolic acids under aerobic conditions. Under anaerobic conditions it causes a cyanide-like action, such that pretomanid acts as a respiratory poison leading to NO production and respiratory inhibition in Mtb by binding to the heme/ cytochrome iron orbital that normally binds oxygen and acts like cyanide would.64 NO is produced by imidazole ring two-electron reduction rather than extraction from the nitro group.61 Compound 7 effect on the mycobacterial respiratory chain is manifested at lower ATP intracellular levels.64 According to a recent study, intracellular accumulation of methyl glyoxal (i.e., a toxic metabolite) may be another 7-induced mechanism of mycobacterial killing.65 Curiously, 7 also “imitates” the natural immune response of the host organism such that only penetration into Mtb can induce NO production.64 If we are to greatly improve upon these compounds and leverage the same approach of NO production, we would need to address the high frequency of resistance and overcome drug resistance seen in the clinic.66

ANALYSIS OF ACTIVE ANTITUBERCULAR COMPOUNDS

In recent years, there has been an abundance of reviews written on the topic of tuberculosis drug discovery as well as advances in treatment regimens and newer approaches to therapy. While we do not intend to cover the same ground, it is worth noting that most of these prior publications tend to focus on the underlying biology and targets used.67 They also hone in on a relatively small number of targets such as InhA, DNA gyrase, RNA polymerase combined with a few newer common targets like MmpL3, DprE1, CTP synthetase and QcrB.68, 69 Others have described emerging targets with some of the hits from high throughput screens that appear structurally similar across different groups.70 A recent review71 covered more current research but makes no attempt to organize the compounds by structure class. Other reviews have also covered a larger period of time but without a distinction of any kind of bioactivity threshold.72, 73 While such reviews have tackled why TB drug discovery is so challenging, they focused heavily on the TB Alliance pipeline with little if any discussion of the chemistry.74 More recent reviews have provided small selections of lead compounds for TB grouped by pathways targeted, while also commenting on the difficulty in developing fast acting drugs.75 In contrast to these many reports, the objective of the current review is to provide a new perspective on the design and development of TB drugs. Perhaps importantly none of these prior reviews has attempted to analyze or understand recent antitubercular chemistry in the context of the large numbers of active compounds described over the past few decades.We will therefore focus our attention on issues with these compounds and specifically the gains made in the development of these molecules (including against drug-resistant strains and MDR-TB). In order to dramatically limit the number of compounds addressed over the last decade, we will focus on those affecting drug-susceptible or drug-resistant Mtb strains that have demonstrated predominantly minimum inhibitory concentrations (MICs) < 10 μM, low toxicity and in many cases have shown promising in vivo efficacy. While we have also assessed those publications that have taken a target-based approach and identified potent molecules, in many cases they do not have an acceptable MIC. For example, a fragment-based screening approach took a weak 4-phenyl-1H-imidazole compound hit for inosine-5’-monophosphate dehydrogenase (IMPDH) and improved it to IC50 of 0.27 μM, but did not show inhibition of bacterial growth up to 50 μM.76 This molecule therefore does not fit our selection criteria.

Most of the compounds assessed are from relatively recent years with a few older compounds for comparison. Many of the scaffolds described have significant liabilities in selectivity or drug disposition/ toxicity which we will highlight. While several of the papers highlighted have been published in this journal, we are not limiting our scope to this alone. It is also unlikely we have captured every TB drug discovery paper to all readers satisfaction, but our intent is to provide a sampling, a “taste of TB drug discovery” if you will, and then place them in the bigger context. We have therefore divided the 118 molecules into separate tables (Tables 312). The selection of what are the actual cores of these molecules may vary by the differing perspectives of readers. We will demonstrate how the fruitful search for these new highly active anti-tuberculosis compounds performed by many groups globally has led to some chemical diversification of antitubercular drugs, but we question whether it is enough. Our goal is to structurally categorize the most promising leads rather than focus on biology, allowing an analysis from a molecular property perspective, which is fundamentally different compared with any of the preceding reviews. Thus, we can attempt to more clearly understand the chemical as well as mechanistic diversity of these potential innovative drugs that have been identified to date.

Table 3.

Pyrrole, indole-2-carboxamide, Pyrazole [1,5-a] pyridine-3 carboxamide, Pyrazolepyridone, imidazole [1,2-a] pyridine and oxazolidinone chemical classes. MmpL3 = Mycobacterial membrane protein Large 3, DprE1 = decaprenylphosphoryl-β-d-ribose oxidase. PAINS were calculated as described by Lagorce et al.105 Compound assay promiscuity was also predicted with BadApple.106

Molecule ID Chemical class Mtb Activity Target Notes References
graphic file with name nihms-1619908-t0009.jpg 8 Pyrrole MICs 0.7–6.2 μg/ml also active against drug-resistant bacilli (including ones with multiple resistance) and infected macrophages MmpL3 PAINS pyrrole_B(29)
BadApple: 2 Alerts
Also known as BM212.
107, 108
graphic file with name nihms-1619908-t0010.jpg 9 Pyrrole MIC: 0.5 MmpL3 PAINS pyrrole_B(29)
BadApple: 2 Alerts
Thiomorpholine fragment improves antimycobacterial activity
40,
graphic file with name nihms-1619908-t0011.jpg 10 Pyrrole MIC90 – 0.5 μg/ml PAINS pyrrole_B(29)
BadApple: 2 Alerts
Hybrid molecule.
109
graphic file with name nihms-1619908-t0012.jpg 11 Pyrrole MIC 0.025–0.12 μg/ml also activity against isoniazid- and rifampicin-resistant Mtb strains BadApple: 2 Alerts
Sudoterb, in vivo activity in infected mice as 12-week therapy (12.5 mg/kg) led to complete lung and spleen sterilization. Has undergone phase I clinical studies.
110, 111
graphic file with name nihms-1619908-t0013.jpg 12 Indole-2-carboxamide MIC90 was 0.006–0.047 μM against Mtb including MDR- and XDR-TB strains MmpL3 BadApple: 1 Alert
in vivo efficiency assessment using an infected mouse model demonstrated that the compound can reduce pulmonary bacterial load by 2.12 orders of magnitude in 4 weeks (dose - 100 mg/kg)
112
graphic file with name nihms-1619908-t0014.jpg 13 Pyrazole [1,5-a] pyridine-3 carboxamide MIC90: 11.1–223 nM PAINS
anil_di_alk_D(198)
anil_di_alk_D(198)
In vivo efficacy led to lower visceral bacterial load
113
graphic file with name nihms-1619908-t0015.jpg 14 Pyrazolepyridone MIC 1.6 μg/mL DprE1 Suboptimal water solubility (1 μM, logD 3.9), free plasma protein binding (1 %) and clearance (27 ml/min/kg) 81, 114116
graphic file with name nihms-1619908-t0016.jpg 15 Imidazole [1,2-a] pyridine MIC90: 0.09–0.13 μM active against two INH- and rifampicin resistant MDR clinical isolates BadApple: 1 Alert
In vivo studies in mice and rats showed satisfactory pharmacokinetic properties (peak serum concentration (Cmax) was 225 ng/ml, half-life (T1/2) was 1.5 h and clearance was 86.284 ml/h/kg)
117
graphic file with name nihms-1619908-t0017.jpg 16 Oxazolidinone MIC50 1 μg/ml and MIC90 0.5 μg/ml Binding with mycobacterial 50S ribosomal subunit PAINS
anil_di_alk_A(478)
Associated with peripheral and optic neuropathies which are the most common side effects. Toxicity detected during clinical trials resulted in the withdrawal
118123
graphic file with name nihms-1619908-t0018.jpg 17 Oxazolidinone MIC: 0.03–0.50 μg/ml Binding with mycobacterial 50S ribosomal subunit PAINS
anil_di_alk_A(478)
The therapeutic regimen involves combination with moxifloxacin and pyrazinamide which is more potent than rifampicin + isoniazid + pyrazinamide
124126
graphic file with name nihms-1619908-t0019.jpg 18 Oxazolidinone MIC 0.25 to 1 μg/ml against a panel of clinical isolates of M. tuberculosis, including single-drug-resistant strains Binding with mycobacterial 50S ribosomal subunit Multiple use resulted in several non-life-threatening side effects, the most common being gastrointestinal disorders and a slightly increased reticulocyte level observed during the course of drug treatment 127129

Table 12.

More recent lead compounds of different structural classes. Ddn = Deazaflavin-dependent nitroreductase, InhA = enoyl-ACP reductase, PyrG = CTP synthetase, QcrB = The b subunit of cytochrome bcc complex, IMPDH = inosine-5’-monophosphate dehydrogenase, DprE1 = decaprenylphosphoryl-β-d-ribose oxidase, Pks13 = polyketide synthase 13, Mmpl3 = Mycobacterial membrane protein Large 3, Mptpb = protein tyrosine phosphatase B. PAINS were calculated as described by Lagorce et al.105 Compound assay promiscuity was also predicted with BadApple.106

Structure ID Chemical class Activity Target Notes references
graphic file with name nihms-1619908-t0105.jpg 104 Triazine MIC 0.078 μM Ddn and InhA BadApple: 1 Alert
Original hit from a Bayesian model screen
66
graphic file with name nihms-1619908-t0106.jpg 105 Triazine MIC 0.63 μM Ddn and InhA BadApple: 1 Alert
Optimised lead, did not show efficacy in mouse
66
graphic file with name nihms-1619908-t0107.jpg 106 Thiophenecarboxamide MIC 0.5 μg/ml PyrG BadApple: 1 Alert
Compound activated by EthA
86
graphic file with name nihms-1619908-t0108.jpg 107 Quinoxoline di-N-oxide MIC 1.2 μg/ml Lead optimization of hit discovered using a pharmacophore search. 87
graphic file with name nihms-1619908-t0109.jpg 108 Pyrazolo[1,5-a]pyrimidine MIC 1.1 μg/ml BadApple: 1 Alert
Compound identified by a Bayesian model screen
85
graphic file with name nihms-1619908-t0110.jpg 109 Imidazole MIC 4.8 μg/ml Compound identified by a Bayesian model screen 225
graphic file with name nihms-1619908-t0111.jpg 110 Thymidine MIC 7.8 – 15.6 μM thymidylate kinase BadApple: 1 Alert
IC50 of 0.95 μM versus Mtb TMPK
88
graphic file with name nihms-1619908-t0112.jpg 111 Indolyl Azaspiroketal MIC90 versus Mtb BCG of 0.8 μM Selective membrane permeabilization of Mtb membranes BadApple: 1 Alert
Modest reduction in CFU counts in the lungs (−0.75 log10) and spleens (−0.84 log10) of Mtb H37Rv infected mice when dosed at 100 mg/kg daily over 4 weeks (6 days per week, total of 24 doses)
89
graphic file with name nihms-1619908-t0113.jpg 112 Morpholino-Thiophenes MIC of 0.24 μM QcrB BadApple: 1 Alert
Activity in a murine intratracheal infection model at 100 mg/kg for four days led to a modest reduction of CFU in the lung of 0.8 log compared to 2.8 log for moxifloxacin
graphic file with name nihms-1619908-t0114.jpg 113 Benzoxazole MIC = 1.0 μM IMPDH BadApple: 2 Alerts
Issues with mouse microsomal stability and high serum binding
91
graphic file with name nihms-1619908-t0115.jpg 114 Bis-Substituted Cyclam 6.25 μg/mL BadApple: 1 Alert
Relatively large (MW = 967.31 amu), charged species which might impact oral bioavailability
92
graphic file with name nihms-1619908-t0116.jpg 115 Phenyl Aminothiazole MIC90 against several strains of Mtb in the range of 0.060 to 0.125 μg/mL BadApple: 2 Alerts
Compound is metabolically stable and apparently is not susceptible to drug efflux pumps
93
graphic file with name nihms-1619908-t0117.jpg 116 6-Dialkylaminopyrimidine Carboxamide MIC99 1.3 μM BCG_3193 (Rv3169) and BCG_3827 (Rv3768) BadApple: 2 Alerts
The compound has a relatively high cLogP (5.0) and was tested in various assays for PK and demonstrated moderate uptake, stability, and showed low clearance
95
graphic file with name nihms-1619908-t0118.jpg 117 Heterocyclic N-oxide Growing Mtb H37Rv (MIC90 = 1.10 μM) and dormant (hypoxic) Mtb H37Rv (MIC90 = 6.62 μM) BadApple: 2 Alerts
Complete sterilization in the lungs of female BALB/c mice at 200 mg/kg given daily for 5/7 days over a three week period.
96
graphic file with name nihms-1619908-t0119.jpg 118 Hydantoin MIC 8.3 μM DprE1 pIC50 = 7.0, no toxicity to HepG2 cells up to 100 μM, and good kinetic aqueous solubility (202 μM) 97
graphic file with name nihms-1619908-t0120.jpg 119 Coumestan MIC90 = 0.125−0.25 μg/mL Pks13 PAINS
mannich_A(296)
(SIT) assay by dosing BALB/c mice at 100 mg/kg via oral gavage and collecting serum at 30, 60, and 120 minutes. After processing the serum, the 30 minute cohort gave good inhibition of Mtb H37Rv in the in vitro MABA assay suggesting that the lead is orally available and that metabolism likely intervenes past the 30 min time point.
99
graphic file with name nihms-1619908-t0121.jpg 120 Coumestan 0.0039 to 0.0156 μg/mL Pks13 PAINS
mannich_A(296)
Significantly reduced toxicity against the four human-derived cell lines
98
graphic file with name nihms-1619908-t0122.jpg 121 Riminophenazine MIC of 0.016 μg/mL PAINS
quinone_A_ter(370)
BadApple: 1 Alert
3–5 logs of CFU reduction in the lungs after 20 days in comparison to untreated control animals
100
graphic file with name nihms-1619908-t0123.jpg 122 Spirocyclic MIC90 = 0.06 μM MmpL3 BadApple: 1 Alert
At a 50 mg/kg dosing regimen, 123 gave a cidal response and a 4.2 log reduction in CFU in the lungs compared to untreated controls
101
graphic file with name nihms-1619908-t0124.jpg 123 Biarylpyrazole imidazole MIC of 6.25 μg/mL CYP121A1 BadApple: 1 Alert
The MIC was found to correlate with calculated logP values
102
graphic file with name nihms-1619908-t0125.jpg 124 Hydroxypyrimidinone MIC 4.7 μM DprE1 A single dose of 10 mg/kg indicated poor absorption 103
graphic file with name nihms-1619908-t0126.jpg 125 Phenyl-isoxazole Not reported Mptpb BadApple: 1 Alert
IC50 Mptpb = 2.98 μM
at least 1 log reduction in bacterial burden in lungs and spleens of guinea pig.
104

Our combined decades of experience in drug discovery applied to tuberculosis spans several areas such as high throughput screening (HTS) of hundreds of thousands of compounds7779, medicinal chemistry that has led to two compounds currently in clinical trials for TB80, 81 and machine learning computational approaches to categorize and identify new antitubercular leads as well as developing a structural understanding of crucial features for antituberculosis activity.8284 Therefore, we will focus on some of these key areas to highlight molecules of interest. Writing this perspective from our different unique viewpoints has indicated that there is scope for more diversity in chemistry and approaches to attack Mtb.

It is apparent from the recent antitubercular leads that there is a very limited palate of chemical classes. Some of the structural classes shown in this perspective represent common chemistries that have been applied elsewhere for anti-infectives or other types of therapeutics. For example, we have included pyrrole, indole-2-carboxamide, pyrazole [1,5-a] pyridine-3 carboxamide, pyrazolepyridone, imidazole [1,2-a] pyridine and oxazolidinone (Table 3), ethylenediamine and dipiperidine, hydrazone (Table 4), indole, 1,3,4-oxadiazole, 1,3,4-thiadiazole, oxadiazole, tetrazole-5-thiol, thiazole, benzothiazole, thiadiazole (Table 5), riminophenazine, pyridinone, phthalimide, quinoline, 4-aminoquinolone (Table 6), formamidopyrimidine, pyrimidine, diarylpyrazole, tetrahydropyrazolopyrimidine, pyrimidine-azaindole, indole (Table 7), quinazolinone, oxadiazole-pyranopyridine, chromeno[3,2-с]pyridine, methylcoumarin, triazoloquinolone and benzothiazinone (Table 8), dispiropyrrolothiazole, spiro-pyrrolothiazoles, octylberberine, capuramycins, caprazamycins, spectinomycins and galactose linked nitroimidazoles (Table 9), isooxazoline, oxazole, Benzo[d]oxazole and triazole (Table 10), benzimidazole, indazole, phthalazine, nitrofuran and 3-aracylphthalide and oxoborol (Table 11).

Table 4.

Ethylenediamine, dipiperidine, hydrazone chemical classes. MmpL3 = Mycobacterial membrane protein Large 3. Compound assay promiscuity was also predicted with BadApple106.

Molecule ID Chemical class Activity Target Notes References
graphic file with name nihms-1619908-t0020.jpg 19 Ethylenediamine MIC 0.16–0.64 μg/ml MmpL3 Low oral bioavailability, efficacy in mouse model when combined with isoniazid and rifampicin. has been in Phase I, IIA and IIB clinical studies 130137
graphic file with name nihms-1619908-t0021.jpg 20 Dipiperidine MIC: 4 μg/ml identified in a high throughput screen of over 10,000 compounds. in vivo Mtb models and favourable pharmacological safety 138, 139
graphic file with name nihms-1619908-t0022.jpg 21 Hydrazone MIC − 0.1 μg/ml inhibition of mycolic acid cyclopropanation Thioacetazone is used in combination with isoniazid 140
graphic file with name nihms-1619908-t0023.jpg 22 Hydrazone MIC
3.1 μg/ml
inhibition of mycolic acid cyclopropanation BadApple: 1 Alert
Greater potency in a mouse model
141
graphic file with name nihms-1619908-t0024.jpg 23 Hydrazone MIC < 0.05 μg/ml inhibition of mycolic acid cyclopropanation greater potency in a mouse model 141

Table 5.

Indole, 1,3,4-oxadiazole, 1,3,4-thiadiazole, oxadiazole, tetrazole-5-thiol, thiazole, benzothiazole, thiadiazole chemical classes. PanC = pantothenate synthetase, PrrB and PrrA are a two-component regulatory system, DprE1 = decaprenylphosphoryl-β-d-ribose oxidase. PAINS were calculated as described by Lagorce et al.105. Compound assay promiscuity was also predicted with BadApple.106

Molecule ID Chemical class Activity Target Notes References
graphic file with name nihms-1619908-t0025.jpg 24 Indole MIC90 is 6.16 μM against MDR Mtb clinical isolates PanC Docking in PanC 142, 143
graphic file with name nihms-1619908-t0026.jpg 25 1,3,4-oxadiazole MIC90 < 0.25 μM also active against non-replicating Mtb strains (streptomycin-starved Mtb18b) Nucleic acid biosynthesis BadApple: 1 Alert
Low cytotoxicity and non mutagenic
144
graphic file with name nihms-1619908-t0027.jpg 26 1,3,4-thiadiazole MIC90 < 0.25 μM also active against non-replicating Mtb strains (streptomycin-starved Mtb18b) Nucleic acid biosynthesis Low cytotoxicity and non mutagenic 144
graphic file with name nihms-1619908-t0028.jpg 27 Oxadiazole Addition of 25 μM BDM31343 to 0.1 μg ml−1 ethionamide abolished the growth of Mtb whereas 1 μg ml−1 ethionamide was necessary to obtain a similar growth inhibition in the absence of BDM31343 Allosteric inactivation of DNA-binding EthR activity BadApple: 1 Alert
A booster for ethionamide co-administration of ethionamide and BDM31343 to M. tuberculosis–infected mice for 3 weeks reduced the bacterial load throughout the entire treatment as efficiently as a three times–higher dose of ethionamide administered alone
145147
graphic file with name nihms-1619908-t0029.jpg 28 Tetrazole-5-thiol MIC90 < 0.5 μM active and selective agents to combat MDR and XDR Mtb strains Low cytotoxicity, might utilize the FAD systems 147
graphic file with name nihms-1619908-t0030.jpg 29 1,3,4-oxadiazole MIC90 < 0.5 μM active and selective agents to combat MDR and XDR Mtb strains BadApple: 1 Alert
Low cytotoxicity, might utilize the FAD systems
147
graphic file with name nihms-1619908-t0031.jpg 30 Thiazole MIC90 < 1.68 μM PrrB and PrrA BadApple: 2 Alerts
solubility is 31 μM, mouse plasma protein binding is 17 % and mouse clearance is 170.4 ul/min/mg
148150
graphic file with name nihms-1619908-t0032.jpg 31 Thiazole MIC90 of 7.1–12.0 μM PrrB and PrrA BadApple: 4 Alerts
PAINS
thiaz_ene_D_bis(8)
151
graphic file with name nihms-1619908-t0033.jpg 32 Benzothiazole MIC90 < 4.62 μM DprE1 BadApple: 1 Alert
Low-toxicity on the A549 human cell line (IC50 > 100 μM), negative Ames test and moderate CYP inhibition
152
graphic file with name nihms-1619908-t0034.jpg 33 Thiadiazole MIC90: 0.08–0.66 μM Isocitrate lyase BadApple: 1 Alert
Target plays a key role in Mtb persistence (chronic and latent infection)
153, 154

Table 6.

Riminophenazine, pyridinone, phthalimide, quinoline, 4-aminoquinolone chemical classes. QcrB = The b subunit of cytochrome bcc complex, DprE1 = decaprenylphosphoryl-β-d-ribose oxidase. PAINS were calculated as described by Lagorce et al.105 Compound assay promiscuity was also predicted with BadApple.106

Molecule ID Chemical class Activity Target Notes References
graphic file with name nihms-1619908-t0035.jpg 34 Riminophenazine MIC 1 μg/ml DNA intercalation PAINS
quinone_A_ter(370)
The riminophenazines lack interactions with CYPs, are generally active against various mycobacterial infections (such as leprosy), but they often have poor solubility and drug-induced side effects. Cross resistance with other classes of tuberculosis drugs has not been observed so they have the potential to treat MDR-TB
155157
graphic file with name nihms-1619908-t0036.jpg 35 Riminophenazine MIC 0.25 μg/ml DNA intercalation PAINS
quinone_A_ter(370)
Milder skin pigmentation side effects
155157
graphic file with name nihms-1619908-t0037.jpg 36 Riminophenazine MIC 0.12 μg/ml DNA intercalation PAINS
quinone_A_ter(370)
Milder skin pigmentation side effects
155157
graphic file with name nihms-1619908-t0038.jpg 37 Pyridinone MIC90 1.56 μg/mL
is active against the MDR-TB strain
RNA polymerase PAINS
anil_di_alk_A(478)
BadApple: 2 Alerts
Half-life in human hepatic microsomes (14.4 h) and a slight inhibition of CYP3A4 and CYP2D6
158
graphic file with name nihms-1619908-t0039.jpg 38 Phthalimide MIC90 0.132 μM QcrB Identified by phenotypic screening of 6000 molecules 159
graphic file with name nihms-1619908-t0040.jpg 39 Phthalimide MIC90 0.065 μM QcrB Mouse pharmacokinetic studies showed high clearance and low plasma levels 159
graphic file with name nihms-1619908-t0041.jpg 40 Quinoline MIC90 0.05 μM cytochrome bc1 BadApple: 1 Alert
In vitro intrinsic clearance (Clint) was 14.3 ml/min/kg and T1/2 was 21.8 min
160
graphic file with name nihms-1619908-t0042.jpg 41 Quinoline MIC90 1 nM against MDR-TB clinical isolate cytochrome bc1 BadApple: 1 Alert
Clint (14.8 ml/min/kg and T1/2 19.4 min)
161, 162
graphic file with name nihms-1619908-t0043.jpg 42 4-aminoquinolone MIC90 of 0.2–3.12 μM DprE1 A non-covalent inhibitor. Cmax of 4.9 μM, plasma clearance of 34.4 ml/min/kg and a T1/2 of 0.5 h 163

Table 7.

Formamidopyrimidine, pyrimidine, diarylpyrazole, tetrahydropyrazolopyrimidine, pyrimidine-azaindole, indole chemical classes. DprE1= decaprenylphosphoryl-β-d-ribose oxidase. Compound assay promiscuity was also predicted with BadApple,106

Molecule ID Chemical class Activity Target Notes References
graphic file with name nihms-1619908-t0044.jpg 43 Formamidopyrimidine MIC90 ≤ 0.2 μg/ml BadApple: 2 Alerts
No toxicity in Vero cells
164, 165
graphic file with name nihms-1619908-t0045.jpg 44 Quinolinyl pyrimidine MIC 0.87 μg/ml type II NADH-menaquinone oxidoreductase (Ndh) BadApple: 4 Alerts
Analogs with improved ADME properties were generated
166, 167
graphic file with name nihms-1619908-t0046.jpg 45 Pyrimidine MIC90 − 0.7 μg/ml and an identical activity versus an MDR strain BadApple: 2 Alerts
LD50 in mouse of 315 mg/kg
168
graphic file with name nihms-1619908-t0047.jpg 46 Pyrimidine MIC90 – 0.7 μg/ml and an identical activity versus an MDR strain BadApple: 2 Alerts
LD50 600 mg/kg
169
graphic file with name nihms-1619908-t0048.jpg 47 Pyrimidine MIC90 of 0.37 μM and activity against isoniazid- and rifampicin-resistant Mtb strains BadApple: 2 Alerts
LD50 45 mg/kg
170
graphic file with name nihms-1619908-t0049.jpg 48 Diarylpyrazole MIC90 0.125 μg/ml BadApple: 3 Alerts 171
graphic file with name nihms-1619908-t0050.jpg 49 Diarylpyrazole MIC90 0.25 μg/ml BadApple: 3 Alerts 171
graphic file with name nihms-1619908-t0051.jpg 50 Tetrahydropyrazolopyrimidine MIC 0.15 μM High potency in animal studies after oral administration (100 mg/kg; once a day; therapeutic course - 28 days) leading to a decrease in lung CFU by 3.5 orders of magnitude 172
graphic file with name nihms-1619908-t0052.jpg 51 Pyrimidine-azaindole MIC90: 0.78–1.56 μM DprE1 BadApple: 1 Alert
(dose − 300 mg/kg) was also tested in vivo and found to reduce lung bacterial load by 1 log
173
graphic file with name nihms-1619908-t0053.jpg 52 Indole MIC90 14.3 μM And inhibited INH-resistant strains DprE1 BadApple: 2 Alerts 174

Table 8.

quinazolinone, oxadiazole-pyranopyridine, chromeno[3,2-с]pyridine, methylcoumarin, triazoloquinolone, benzothiazinone chemical classes. AHAS = Acetohydroxyacid Synthases, DprE1 = decaprenylphosphoryl-β-d-ribose oxidase. Compound assay promiscuity was also predicted with BadApple.106

Molecule ID Chemical class Activity Target Notes References
graphic file with name nihms-1619908-t0054.jpg 53 Quinazolinone MIC 2.5–10 mg/L Also had intracellular activity against XDR clinical isolates. AHAS Compound Identified by a virtual screen 175
graphic file with name nihms-1619908-t0055.jpg 54 Oxadiazole-pyranopyridine MIC 0.31 μg/ml 176
graphic file with name nihms-1619908-t0056.jpg 55 Oxadiazole-pyranopyridine MIC 0.73 μg/ml 176
graphic file with name nihms-1619908-t0057.jpg 56 Oxadiazole-pyranopyridine MIC 0.07 and 0.14 μg/ml against drug-susceptible M. tb and MDR-TB 177
graphic file with name nihms-1619908-t0058.jpg 57 Chromeno[3,2-с]pyridine MICs 0.22 and 0.07 μg/ml against drug-resistant Mtb and MDR-TB BadApple: 1 Alert
Reduced lung and spleen bacterial load by 1.11 and 2.94 orders of magnitude, respectively in vivo (dose − 25 mg/kg)
178
graphic file with name nihms-1619908-t0059.jpg 58 Methylcoumarin MIC 0.78 μg/ml BadApple: 1 Alert 179
graphic file with name nihms-1619908-t0060.jpg 59 Methylcoumarin MIC 1.56 μg/ml 179
graphic file with name nihms-1619908-t0061.jpg 60 Triazoloquinolone MIC 0.5 μg/ml Also active in macrophages 180
graphic file with name nihms-1619908-t0062.jpg 61 Benzothiazinone MIC 1ng/ml DprE1 High activity against all the clinical isolates of Mtb (including MDR and XDR strains). Efficacy in the chronic mouse model at 37.5mg/kg 81, 181
graphic file with name nihms-1619908-t0063.jpg 62 Benzothiazinone MIC 1 ng/ml (2.3 nM) and 4 ng/ml (9.2 nM), respectively against Mtb H37Rv and M. smegmatis DprE1 Has completed an open-labelled, dose-escalation phase I study in healthy male volunteers followed by a multiple ascending dose trial in 2016 (in Russia) and in 2018 (in Switzerland). This most recent study investigated the safety, tolerability and pharmacokinetics at doses up to 640 mg once daily for 14 days and revealed a good safety profile. In 2017, a phase IIa EBA study (monotherapy during 14 days) was initiated in drug sensitive-TB patients in Russia and Belarus. It also confirmed safety in drug sensitive-TB patients and statistically significant EBA after 14 days monotherapy in the group of patients treated with 640 mg of 182184

Table 9.

Dispiropyrrolothiazole, spiro-pyrrolothiazoles, octylberberine, capuramycins, caprazamycins, spectinomycins, galactose linked nitroimidazoles chemical classes. MraY = phospho-MurNAc-pentapeptide translocase, WecA = phosphoglycosyltransferase, InhA – enoyl-ACP reductase. Compound assay promiscuity was also predicted with BadApple.106

Molecule ID Chemical class Activity Target Notes References
graphic file with name nihms-1619908-t0064.jpg 63 Dispiropyrrolothiazole MIC 0.21 μM Also active against the isoniazid-resistant Mtb strain MIC 8.31 μM Compound was not cytotoxic in Vero cells 185
graphic file with name nihms-1619908-t0065.jpg 64 Spiro-pyrrolothiazoles MIC − 0.07 μM Reduced bacterial count in lungs and spleen by 1.30–3.73 orders of magnitude 186
graphic file with name nihms-1619908-t0066.jpg 65 Octylberberine MIC 0.125 μg/ml Significant activity against RIF- and isoniazid-resistant Mtb strains. These quaternary lipophilic agents may not be ideal and show FtsZ activity which may be related to membrane disruption 187, 188
graphic file with name nihms-1619908-t0067.jpg 66 Capuramycins MIC 1.0 μg/ml MraY Selected in a library including more than 7000 analogs -natural nucleoside compound extracted from Streptomyces griseus - poor water solubility and fast elimination as well as P-glycoprotein-mediated efflux are limiting factors, poor in vivo activity in the mouse 189193
graphic file with name nihms-1619908-t0068.jpg 67 Caprazamycins MIC 1.56 μg/ml Also active agains an MDR strain MIC 6.25 μg/ml WecA Extracted from Streptomyces sp. affects both non-fissile and fissile tuberculosis in vitro, so, it may be an efficient drug to treat latent TB 194197
graphic file with name nihms-1619908-t0069.jpg 68 Pyridomycin MIC 0.4 μg/L InhA BadApple: 1 Alert
From Dactylosporangium fulvum or Streptomyces pyridomyceticus
enables killing of isoniazid-resistant Mtb clinical isolates that have katG mutations
198200
graphic file with name nihms-1619908-t0070.jpg 69 Spectinomycins MIC 0.8 μg/L Ribosome and ability to overcome intrinsic efflux mediated by the Mtb Rv1258c efflux pump BadApple: 1 Alert
Synergized with 11 drugs from six antibiotic classes (including clarithromycin, doxycycline and clindamycin)
clarithromycin provided additional bacterial killing in a mouse model of acute tuberculosis infection, but not in a chronic infection model due to mismatched drug exposure
201203
graphic file with name nihms-1619908-t0071.jpg 70 Spectinomycins MIC 1.6 μg/L ribosome and ability to overcome intrinsic efflux mediated by the Mtb Rv1258c efflux pump BadApple: 1 Alert
Synergized with 11 drugs from six antibiotic classes (including clarithromycin, doxycycline and clindamycin)
clarithromycin provided additional bacterial killing in a mouse model of acute tuberculosis infection, but not in a chronic infection model due to mismatched drug exposure
201, 202
graphic file with name nihms-1619908-t0072.jpg 71 Glycosylated beta-amino acids MIC 3.12 μg/ml inhibit biosynthesis of mycobacterial cellular wall peptidoglycan and arabinogalactan In vivo mouse studies showed toxicity at 50 mg/kg/day 204
graphic file with name nihms-1619908-t0073.jpg 72 Galactose linked nitroimidazoles MIC 1.56 μg/ml Active under aerobic conditions 205
graphic file with name nihms-1619908-t0074.jpg 73 Galactose linked nitroimidazoles MIC 1.56 μg/ml Active under aerobic conditions 205

Table 10.

Oxadiazole, isooxazoline, oxazole, Benzo[d]oxazole, triazole chemical classes. InhA = enoyl-ACP reductase. PAINS were calculated as described by Lagorce et al.105 Compound assay promiscuity was also predicted with BadApple.106

Molecule ID Chemical class Activity Target Notes References
graphic file with name nihms-1619908-t0075.jpg 74 Oxadiazole MIC 1.6 μg/ml BadApple: 2 Alerts 206
graphic file with name nihms-1619908-t0076.jpg 75 Oxadiazole MIC 1.5 μg/ml BadApple: 1 Alert 206
graphic file with name nihms-1619908-t0077.jpg 76 Isoxazoline MIC 0.0001 μg/ml PAINS
anil_di_alk_E(186)
BadApple: 1 Alert
207
graphic file with name nihms-1619908-t0078.jpg 77 Isoxazoline MIC 0.00156 μg/ml PAINS
anil_di_alk_E(186)
BadApple: 1 Alert
207
graphic file with name nihms-1619908-t0079.jpg 78 Isoxazoline MIC 0.0002 μg/ml PAINS
anil_di_alk_E(186)
BadApple: 1 Alert
0.83 Log10 reduction in Mtb CFU in lung versus untreated controls
207
graphic file with name nihms-1619908-t0080.jpg 79 Isoxazoline MIC90 1.56 μg/ml PAINS
anil_di_alk_E(186)
207
graphic file with name nihms-1619908-t0081.jpg 80 Oxazole MIC90 − 8 μg/ml 1 μg/mL against XDR-TB and 2 μg/mL MDR-TB BadApple: 1 Alert
Original hit from a screen of 45,000 compounds tested against M. smegmatis
208
graphic file with name nihms-1619908-t0082.jpg 81 Benzo[d]oxazole MIC90 − 11.47 μM InhA BadApple: 1 Alert
Active against a XDR-TB clinical isolate
209
graphic file with name nihms-1619908-t0083.jpg 82 Triazole MIC 2.50 μg/ml InhA 210
graphic file with name nihms-1619908-t0084.jpg 83 Triazole MIC 2.50 μg/ml InhA 210
graphic file with name nihms-1619908-t0085.jpg 84 Triazole MIC 0.50 μg/ml 211
graphic file with name nihms-1619908-t0086.jpg 85 Triazole MIC 0.25 μg/ml 211
graphic file with name nihms-1619908-t0087.jpg 86 Triazole MIC 1 μg/ml 212
graphic file with name nihms-1619908-t0088.jpg 87 Triazole MIC 1 μg/ml BadApple: 1 Alert 212
graphic file with name nihms-1619908-t0089.jpg 88 Triazole MIC 2.5 μg/ml
Similarly active against clinical isolates
PAINS
imine_one_A(321)
No cytotoxicity to human HCT116 and GM637 cells
212, 213
graphic file with name nihms-1619908-t0090.jpg 89 Triazole MIC 2.5 μg/ml
Similarly active against clinical isolates
PAINS
imine_one_A(321)
No cytotoxicity to human HCT116 and GM637 cells
213

Table 11.

Benzimidazole, indazole, phthalazine, nitrofuran, 3-aracylphthalide, oxoborol chemical classes. FtsZ = Filamenting temperature-sensitive mutant Z, KasA = β-Ketoacyl ACP synthase I, LeuRS = leucyl-tRNA synthetase. PAINS were calculated as described by Lagorce et al.105 Compound assay promiscuity was also predicted with BadApple.106

Molecule ID Chemical class Activity Target Notes References
graphic file with name nihms-1619908-t0091.jpg 90 Benzimidazole MIC 4.3 μM FtsZ BadApple: 1 Alert 214
graphic file with name nihms-1619908-t0092.jpg 91 Benzimidazole MIC99 0.39 μg/mL FtsZ BadApple: 1 Alert
in vivo equivalent to isoniazid in a murine tuberculosis model with a reported log10 reduction of 2.10 (i.p.) and 1.72 (p.o.) in the lungs and log10 reduction of 3.36 (i.p.) and 2.49 (p.o.) in the spleen. Computational modelling and QSAR optimization led to other analogs in this class and the suggestion that the trisubstituted benzimidazoles bind in the GTP site of FtsZ
215217
graphic file with name nihms-1619908-t0093.jpg 92 Benzimidazole MIC 1.5 μg/ml 218
graphic file with name nihms-1619908-t0094.jpg 93 Benzimidazole MIC 3.1 μg/ml PAINS
quinone_A_ter(370)
218
graphic file with name nihms-1619908-t0095.jpg 94 Benzimidazole MIC90 − 0.75 μM BadApple: 1 Alert 118
graphic file with name nihms-1619908-t0096.jpg 95 Benzimidazole MIC90 5 μM against MDR Mtb BadApple: 1 Alert
IC50 of 58 and 7.8 μM (newborn skin fibroblast cell lines (HDFn) and human epidermal keratinocyte precursors (HEK), respectively)
124
graphic file with name nihms-1619908-t0097.jpg 96 Indazole MIC 0.8 μM KasA Active in vivo in mice showing 2.4 −3.5 log reduction in chronic and acute mouse models 219, 220
graphic file with name nihms-1619908-t0098.jpg 97 Phthalazine MIC 1.4 μM 100–1800 times higher activity against isoniazid-resistant Mtb strains than isoniazid - mechanism of action related to mycolic acid biosynthesis inhibition 221
graphic file with name nihms-1619908-t0099.jpg 98 Nitrofuran MIC 0.19 μg/ml BadApple: 2 Alerts
Excellent selectivity index > 1800
222
graphic file with name nihms-1619908-t0100.jpg 99 3-aracylphthalide MIC 0.97 μg/ml Lactones and ketones represent chemical alerts 223
graphic file with name nihms-1619908-t0101.jpg 100 3-aracylphthalide MIC 0.93 μg/ml Lactones and ketones represent chemical alerts 223
graphic file with name nihms-1619908-t0102.jpg 101 3-aracylphthalide MIC 0.81 μg/ml BadApple: 1 Alert
Lactones and ketones represent chemical alerts
223
graphic file with name nihms-1619908-t0103.jpg 102 3-aracylphthalide MIC 1.24 μg/ml BadApple: 1 Alert
Lactones and ketones represent chemical alerts
223
graphic file with name nihms-1619908-t0104.jpg 103 Oxoborol MIC 0.08 μM LeuRS Highly specific (IC50 = 0.216 μM), as the IC50 for human cytoplasmic LeuRS is 140 μM 224

In terms of multiple structural classes inhibiting specific targets, this outcome is clearly revealed for pyroles, indole-2-carboxamide (Table 3), ethylenediamine (Table 4), which have been shown to inhibit MmpL3, pyrazolepyridone (Table 3) benzothiazole (Table 5), 4-aminoquinlone (Table 6), pyrimidine-azaindole, indole (Table 7), benzothiazinone (Table 8) which inhibit DprE1. While several other structural classes have evidence on their targets, such information is typically absent (Table 3Table 11, compounds 8–125).

As examples using this perspective, we will specifically highlight some relatively recent publications or those that take different approaches to identify lead compounds (Table 12). For instance, a study described the triazine antitubercular 10466 which was initially discovered using a machine learning model.85 This compound was recently found to be activated by F420H2 and one or more nitroreductases in addition to Ddn. Formation of NO and a des-nitro metabolite were identified.66 An analog 105 was generated in an attempt to improve the in vivo pharmacokinetics (PK). Both 104 and 105 achieved their activity via release of intrabacterial NO along with inhibition of InhA.66 The thiophenecarboxamide 106 was found to be activated by EthA and then inhibited PyrG identifying the importance of metabolic activation.86 A computational pharmacophore identified quinoxoline di N-oxides and a lead optimisation program produced several compounds such as 107.87 An additional study used a Bayesian model to identify compounds, one of which 108 was pyrazolo[1,5-a]pyrimidine85, a class of compounds that had also been identified by several others through a concurrent high throughput screening program. A third compound identified in a separate Bayesian model screen was an imidazole 109 which demonstrated reasonable in vitro activity. These computationally-derived hits are vastly outnumbered by those discovered by other approaches such as HTS.

For example, Song et al. have reported the structure activity relationship (SAR) of about 30 new analogs of an earlier thymidine-based hit against the Mtb thymidylate kinase (TMPK) ostensibly using ligand efficiency (LE) metrics.88 LE ranged from 0.18 – 0.29 for all compounds. Compound 110 emerged as the lead active giving an IC50 of 0.95 μM versus Mtb TMPK and activity against whole cell Mtb H37Rv (MIC = 7.8 – 15.6 μM). Screening against Mtb H37Rv in the presence of verapamil, an efflux pump inhibitor, modestly improved activity (MIC with verapamil (40 μg/mL) = 3.9 – 7.8 μM) suggesting that efflux may play a role in the differential activity against Mtb TMPK and the whole cell MIC. A crystal structure of the lead compound with Mtb TMPK was also reported.

A series of indolyl azaspiroketal Mannich bases has been reported to show selective membrane permeabilization of Mtb membranes.89 The lead agent 111 gave an MIC90 versus Mtb BCG of 0.8 μM and an IC50 against VERO cells of 29 μM for a modest selectivity index (SI) of 36 (SI commonly presented as IC50 VERO/MIC90 Mtb). Not surprisingly, the compound has a relatively high logP by medicinal chemistry standards (5.12) and demonstrates 50% hemolysis of human red blood cells at 64 μM. The lead demonstrated acceptable PK in mice and gave a modest reduction in CFU counts in the lungs (−0.75 log10) and spleens (−0.84 log10) of Mtb H37Rv infected mice when dosed at 100 mg/kg daily over 4 weeks (6 days per week, total of 24 doses).

Whole cell aerobic Mtb screening of the corporate Lilly screening deck produced a sub micromolar active in the morpholino-thiophene class.90 Further assessment validated the active hit leading to structure-activity relationship (SAR) optimization and advanced screening. The minimal pharmacophore was the morpholino-thiophene and produced a new lead compound 112 with an MIC of 0.24 μM against Mtb H37Rv, VERO cell toxicity of 37 μM, and a resulting Selectivity Index (SI = VERO IC50/Mtb MIC) of 154. Microsomal stability was improved over the initial hit but intrinsic clearance in mice using mouse liver microsomes remained significant at 8 mL/min/gram liver. Activity in a murine intratracheal infection model at 100 mg/kg for four days led to a modest reduction of CFU in the lung of 0.8 log compared to 2.8 log for moxifloxacin. Initial mechanism of action (MOA) studies suggested that QcrB, a subunit of the menaquinol cytochrome c oxidoreductase and part of the Mtb bc1-aa3-type cytochrome c oxidase complex was a potential target.

Mtb IMPDH has been the target of numerous drug discovery programs. Hedstrom et al. have pursued the SAR of the benzoxazole scaffold to produce a new lead inhibitor 113.91 This compound had showed potent inhibition of Mtb IMPDH (Ki,app = 18 nM) and good activity versus Mtb H37Rv (MIC = 1.0 μM). Additionally, the compound is active against conditional and knockdown strains easing current concerns that guanine salvage can protect Mtb bacilli in the host. The compound showed reasonable pharmacokinetics in mice, but there are issues with mouse microsomal stability and high serum binding suggesting that optimization of the class for targeting Mtb in vivo will be required.

The group of Spain et al. recently reported continuing studies developing the SAR of bis-substituted cyclams as antimycobacterial agents.92 One compound in the series 114 demonstrated modest activity against Mtb H37Rv (6.25 μg/mL) and showed improved thermodynamic solubility as the ZnCl2 salt relative to other metal salt forms. The compound showed approximately a 3-fold reduction in M. marinum fluorescence/infection in the zebrafish embryo model using tdTomato-fluorescent M. marinum at a dose of 10 μM, refreshed every second day of infection. It remains unclear how effective the molecule is as the metal salt or whether it is just a delivery mechanism for Zn2+, a metal known to have antibacterial activity. Beyond the issue of stability of the metal salt to gut pH, these are relatively large (MW = 967.31 amu), charged species which might impact oral bioavailability and utility as drugs.

Several substituted phenyl-aminothiazoles were reported through the TAACF screens in 2009 as potent and relatively selective inhibitors versus M. tuberculosis. Other groups have explored the SAR of this class since that time, but no definitive target(s) have emerged. Azzali et al. recently published over 30 analogs designed to further develop SAR of the class leading to compound 115.93 This lead had similar activity to highly related structures reported out of the TAACF screens, and the reported compound showed MIC90 values against several strains of Mtb in the range of 0.060 to 0.125 μg/mL with retention of activity against several established drug resistant Mtb strains. Furthermore, 115 was relatively selective for Mtb versus a mammalian cell line, HMDM, (IC50 = 53 μg/mL) verifying earlier toxicity reports for the class by the TAACF (VERO cells). The paper also includes metabolism (T1/2 and intrinsic clearance) from human liver microsomes (T1/2 = 16.1 ± 0.2 min and CL’int = 38.8 mL/min/kg) and information relating to efflux in drug resistant Mtb strains suggesting that the compound is metabolically stable and apparently is not susceptible to drug efflux pumps. The paper does not address concerns regarding chelating ability of the class. This group has also described a similar compound that is antitubercular and inhibits efflux activity.94

Wilson et al. report the SAR development of an HTS hit from the Biofocus DPI Softfocus Library that is in the 6‑dialkylaminopyrimidine carboxamide class.95 A lead compound 116 was identified from some 50 analogs that demonstrated high activity against Mtb H37Rv (MIC99 = 1.3 μM) and comparable activity against several clinical strains. The compound has a relatively high cLogP (5.0) and was tested in various assays for PK and demonstrated moderate uptake, stability, and showed low clearance via intravenous administration despite poor microsomal stability, most likely resulting from extensive tissue distribution as indicated by a high volume of distribution (Vd = 21.8 L/kg). Early MOA studies using an affinity anchored analog and sepharose beads in a chemoproteomic approach suggested two potential novel targets of unknown function, BCG_3193 (Rv3169) and BCG_3827 (Rv3768).

A SAR was recently reported for approximately 20 heterocyclic N-oxides leading to advanced screening of a novel hybrid of INH and a benxo-oxadiazole-N-oxide, (E)-6-((2-isonicotinoylhydrazono)methyl) benzo[c][1,2,5]-oxadiazole 1-oxide.96 Compound 117 shows good antitubercular activity against actively growing Mtb H37Rv (MIC90 = 1.1 μM) and dormant (hypoxic) Mtb H37Rv (MIC90 = 6.6 μM) including a small number of drug resistant strains (e.g. INH-R MIC90 = 8.6 μM, RMP-R MIC90 = 3.8 μM, BDQ-R MIC90 = 1.2 μM). While the agent was relatively nontoxic against MRC-5 cells with IC50 of 519.2 μM, it showed some toxicity versus HepG2 (IC50 = 16.0 μM). The active lead was further evaluated for mutagenicity using the micronucleus test in peripheral mouse blood reticulocytes, and there was no indication of genotoxicity up to a single dose of 500 mg/kg after 30 hours. Compound 117 was bactericidal in culture against Mtb H37Rv infection in the J774A.1, a macrophage cell line, at 23.86 μg/mL (4× MIC), 5.84 μg/mL (MIC), and 1.46 μg/mL (MIC/4), essentially sterilizing the culture at all doses. Extensive absorption, distribution, metabolism, excretion and toxicity (ADMET) properties including liver toxicity were determined for the lead resulting in screening in vivo against Mtb Erdman in an acute murine model of tuberculosis. Compound 117 also showed complete sterilization in the lungs of female BALB/c mice at 200 mg/kg given daily for 5/7 days over a three-week period.

Interest in the target DprE1 has led to a HTS screening campaign at GSK and the discovery of a novel hit in the hydantoin class 118.97 In a recent report by Rogacki et al, the activity of the initial hit and an extensive SAR program are described.97 The initial hit 118 showed potent inhibition of DprE1 (pIC50 = 7.0), good activity against Mtb H37Rv (8.3 μM) using the resazurin reduction assay with fluorescent readout, no toxicity to HepG2 cells up to 100 μM, and good kinetic aqueous solubility (202 μM) relative to the MIC. A relatively thorough SAR exploration of the scaffold resulting in over 100 analogs gave only modest improvements in activity and other medicinal chemistry parameters. Mechanistically, the compound is a reversible inhibitor of DprE1. The authors concluded that the compound class has few or no major liabilities, is generally characterized by good metabolic stability, has no appreciable cytotoxicity, and possesses a physicochemical profile acceptable for further development. Further optimization was reported as underway including in vivo experiments in a respiratory rodent model of Mtb.97

Two recent publications present the discovery and further natural product-inspired SAR optimization of the coumestan scaffold. The first publication reports nearly 50 analogs resulting in the lead 119, demonstrating significant activity against Mtb H37Rv in the MABA assay (MIC90 = 0.125−0.25 μg/mL).98 The compound showed relatively significant toxicity to VERO cells (IC50 = 4 μg/mL.), yielding a modest selectivity index - SI (IC50/MIC90) = 16–32. Interestingly, four other human derived cell lines [MRC-5 and HFL1 (human lung fibroblast cells), QSG-7701 (human liver cell), and HEK-293 (human embryonic kidney cell)] showed no toxicity up to 50 μg/mL. The lead was further evaluated in the Serum Inhibition Titration (SIT) assay by dosing BALB/c mice at 100 mg/kg via oral gavage and collecting serum at 30, 60, and 120 minutes. After processing the serum, the 30 min cohort gave good inhibition of Mtb H37Rv in the in vitro MABA assay suggesting that the lead is orally available, and that metabolism likely intervenes past the 30 min time point. Selection of resistant Mtb mutants and sequencing identified Polyketide Synthase 13 as a putative target. In a second paper98, Zhang et al. continued optimization of the scaffold identifying 120 as a second generation lead that shows improved activity (MIC90 = 0.0039 μg/mL) against Mtb H37Rv and MIC90s in the range of 0.0039 to 0.0156 μg/mL against several drug resistant strains.99 Compound 120 also demonstrated toxicity against VERO cells (IC50 = 4 μg/mL.), but, had significantly reduced toxicity against the four human-derived cell lines discussed previously. Compound 120 gave a good half-life (T1/2 = 120.1 min) in human liver microsomes and showed 8-fold higher activity than INH in the SIT assay dosed at 100 mg/kg with serum taken at 30 min.99

Clofazimine (CFZ) 34 is a highly active antitubercular agent, although the drug suffers from liabilities including high lipophilicity and fat solubility causing discoloration of fatty tissues in the patient. Zhang et al. describe optimization of the CFZ riminophenazine scaffold with the goal of reducing lipohilicity to manage these drug liabilities.100 Several CFZ analogs gave comparable or better activity as compared to CFZ with significantly reduced lipophilicity (ClogP). Six compounds with potent activity against M. tuberculosis in vitro, low acute toxicity in mice, and excellent PK profiles were further screened in an acute mouse model of MDR-TB infection. All compounds were highly efficacious, yielding 3–5 logs of CFU reduction in the lungs after 20 days in comparison to untreated control animals.100 Two compounds exhibited equal or better in vivo efficacy against MDR-TB compared to CFZ with the potential for reduced coloration in fatty tissues, and these analogs are being further evaluated for advanced development. For comparison, 121 gave an MIC of 0.016 μg/mL (CFZ 0.12 μg/mL), VERO cell toxicity of >64 μg/mL (CFZ 68.6 μg/mL), and a measured logP of 3.74 (CFZ 5.34).100

Screening of the GSK compound library yielded a novel spirocyclic hit that was further optimized in a recent SAR program and made available through their open source program.101 This effort yielded an improved compound 122 with potent antitubercular activity (Mtb H37Rv MIC90 = 0.06 μM). Cell health for the SAR set was measured in HepG2 cells and is reported as a triple-readout (membrane permeability, mitochondrial potential, and nuclear morphology) phenotypic assay measuring changes in cell structure as a preliminary consequence of cytotoxic injury. The assay used automated imaging to measure IC50 values for the effect of compounds on human liver-derived, HepG2 cells (48 h incubation) as a surrogate of hepatotoxicity. For high doses (>100 mg/kg/day), numbers <80 μM raises alarms for hepatotoxicity. Compound 122 gave results 12.6 μM, 10.7 μM, and 11.0 μM respectively. Compound 122 also had high lipophilicity and poor solubility as well as activity in the hERG assay leading to cardiotoxicity concerns. PK results for 122 showed acceptable bioavailability with lower intrinsic clearance, a longer half-life, and oral AUC values suitable for progression into efficacy studies relative to other analogs. Compound 122 was further assayed in an acute murine model of Mtb – female CB57BL/6J mice infected with Mtb H37Rv intratracheally at ~105 CFU. In a quick assay where the compound is administered for 8 days, the ED99 to give a 2-log reduction in lung CFU was reported to be 11.6 mg/kg. At a 50 mg/kg dosing regimen, 122 gave a cidal response and a 4.2 log reduction in CFU in the lungs compared to untreated controls.101 Further analoging showed modest effects on some of the liabilities but confirmed potential cardiotoxicity and tolerability issues leading to sidelining of this compound series. Mtb MmpL3 was proposed as a possible target either directly or through a related perturbation in the transmembrane proton gradient.101

Mtb cytochrome P450 CYP121A1 is essential for bacterial viability, and a recent paper by Taban et al. describes a small SAR study of three series of biarylpyrazole imidazoles and triazoles including antitubercular and Mtb CYP121A1 inhibition data.102 About 20 compounds were reported in the three series with MIC values ranging from 6.25 to >100 μg/mL. The −CH2-imidazole series was the most active in terms of KD for CYP121A1 and inhibition of Mtb H7Rv in the Resazurin Microtiter Assay. For example, 123 gave a KD of 13.6 μM against CYP121A1 and a MIC of 6.25 μg/mL.102 In general, the MIC was found to correlate with calculated logP values. It is not clear regarding whether logP or logD is more relevant as the imidazole moiety is protonatable at physiological pH. Furthermore, there was no discussion relating to effects on human CYP enzymes or selectivity versus mammalian cells (e.g. VERO, HepG2 etc.). Co-crystal structures are reported for certain analogs with CYP121A1.102

Oh et al. have reported a novel hydroxypyrimidinone hit 124 from the chemical library of the St. Jude Children’s Research Hospital that appears to target decaprenylphosphoryl-β-D-ribose 2-oxidase (DprE1, Rv3790).103 The compound shows good activity against Mtb H37Rv in two media 7H9/ADC (MIC 4.7 μM) and GAST (MIC 0.4 μM) and no toxicity to HepG2 cells up to 100 μM. The hit was bactericidal to actively replicating Mtb in vitro, with a 1–2 log reduction in CFU units observed over 7 days with weak dose dependence over a wide range of concentrations.103 Compound 124 did not show bactericidal activity against anaerobic, nonreplicating Mtb cells generated in the Wayne hypoxic Mtb model. PK profiling of 124 in C57BL/6 mice administered an oral suspension at a single dose of 10 mg/kg indicated poor absorption and in vivo efficacy against Mtb was not undertaken. Ex vivo activity against Mtb H37Rv in J774 murine macrophages gave apparent bactericidal activity. Nearly 20 analogs of the initial hit were reported with very little improvement in activity results. Furthermore, 124 is considerably less active than other reported DprE1 inhibitors that are progressing towards clinical use.103

A structure-based program targeting Mycobacterium tuberculosis protein-tyrosine-phosphatase B (MptpB) was recently reported by Vickers et al. that resulted in a lead compound 125 (IC50 Mptpb = 2.98 μM).104 Certain analogs showed high selectivity for the mycobacterial phosphatase relative to human counterparts and low toxicity up to 150 μM in the mouse J774 macrophage cell line. Compound 125 showed reasonable kinetic solubility (200 μM) and acceptable results in the PAMPA screen (78.1 nM/s), suggesting the potential for good cell penetration. Compound 125 also showed improved pharmacological properties relative to other analogs with reasonable orally bioavailability and a good PK profile. Since the target is only essential in vivo, it is not surprising that it has a very modest impact on the growth of H37Rv in vitro. However, at both 20 and 100 μM, the compound significantly impacts bacterial burden in the human THP-1 macrophage model against both MDR strain (Beijing-W) and the drug-susceptible M. tuberculosis H37Rv strains.104 Furthermore, the compound at the low dose of 5 μM demonstrates compatibility in combination with rifampin and isoniazid (0.1 μg/mL for INH and 0.3 μg/mL for RIF) reducing bacterial burden by 93% in BCG infected J774 mouse macrophages. In vivo profiling of 125 in guinea pigs showed high exposure (Cmax 112 μg/mL, AUC 230 μg·h/mL), long half-life (t1/2 5 h), good oral availability, and appropriate tissue distribution in guinea pigs under either parental or oral dosing. Compound 125 was further tested for efficacy as monotherapy in the acute and chronic guinea pig models of TB (female Hartley Duncan). In the acute infection, animals were infected with approximately 96 CFU and, after 24 h, orally dosed once daily at 100 mg/kg for 4 weeks. Treatment resulted in a 0.9 log reduction of bacterial burden in the lungs relative to vehicle.104 For the chronic infection, guinea pigs were infected with ca. 63 CFU, and 125 was orally administered daily at 100 mg/kg for 4 weeks starting at 28 days post infection. In this model, treatment with 125 resulted in at least 1 log reduction in bacterial burden in lungs and spleens.104

MOLECULAR DIVERSITY OF RECENT COMPOUNDS FOR TB

Interestingly, only a relatively small number (17) of the 118 molecules in Tables 312 (representing 14%) are Pan Assay Interference compounds (PAINS) as assessed using the FAF-3 online software105. However more compounds (62) are flagged with the BadApple software (representing 52% in Tables 312), which attempts to identify compounds with assay promiscuity.106 There is generally good agreement with these two approaches. Previous comparisons of TB in vitro and in vivo datasets have determined the mean of molecular descriptors/properties across actives and inactives.226, 227 We have determined 8 simple molecular properties for the molecules in Table 2 (Table S1) and Tables 312 (Table S2). These data are remarkably similar to our previous analyses suggesting a mean molecular weight of 390.96 and ALogP for 2.84 for approved TB drugs and mean molecular weight 399.57 and ALogP of 3.66 for TB leads, which are comparable to in vivo actives described earlier with a mean molecular weight of 411 and ALogP of 3.11.226 These may represent property ranges to target for TB activity. A more extensive cheminformatics analysis is now described in order to assess the chemical property space coverage of these Mtb leads.

To assess the molecular diversity of the compounds active against Mtb described in this perspective, we have generated a principal component analysis (PCA) plot based on eight 2D descriptors which included molecular weight, topological polar surface area, number of aromatic rings, number of hydrogen bond acceptors, number of hydrogen bond donors, number of rotatable bonds, ring count and MolLogP. All these descriptor calculations were generated with manual python scripts using RDKit228, Pandas229, NumPy230 libraries and Jupyter Notebook.231 Before conducting PCA, the explicit descriptors were imputed “NAN” and ensured there were no missing values; and scaled from zero to one for each feature using MinMaxScaler from scikit-learn.232 We then conducted PCA using three sets of descriptors on (1) Mtb approved drugs (Table 1) and 118 Mtb leads (Tables 312), (2) 7762 active compounds (MIC < 10 μM) from a set of 18,886 compounds with Mtb data233 (Mtb actives) and 118 Mtb leads datasets, and (3) Mtb actives, 118 Mtb compound dataset, approved drugs and Microsource spectrum datasets. The PCA figures were generated using Matplotlib234 library and Jupyter Notebook.

PCA analysis of Mtb approved drugs (Table 1) and 118 Mtb leads (Tables 312) indicates coverage of the same property space (Figure 2A). When visualized using the chemical network visualization this may represent a more useful approach to grouping the molecules than the Tables 312. We have summarized several “outlier” molecules away from the main cluster shown on Figure 2A. These molecules are also shown in Table S2 and S3. What is apparent is that many of these “natural product like” such as 66, 67, 69, 70, 71 or very large 114 with generally low ALogP (Table S2). Many of these molecules cluster together (Figure 2B). Several of outliers are very lipophilic 19, 41, 84, 85, 94. Interestingly, the high ALogP outliers are to the left and the low ALogP molecules are to the right. As one would expect similar molecules group together such as 1 and 2. (Figure 2B).

Figure 2.

Figure 2.

Data visualization of Mtb approved drugs and Mtb lead compounds in this review. A. The PCA generated with eight descriptors showed that Mtb approved drugs are overlapped with the 118 Mtb leads dataset; with the three components of PCA representing 82.8% of the variance (x = 41.7%, y = 28.5%, z = 12.6%). Molecule numbers represent those outside the main cluster with structures shown (see also Table S3). B. Chemical network visualization containing compounds from Table 1 (green) and Tables 312 (red). This was generated using the same eight descriptors, with unsupervised hierarchical clustering using the Euclidean distance method, Wards linkage. Numbering corresponds to molecules in Tables 1, 312.

We have compared these TB lead molecules to a recently curated set of 7762 active compounds (MIC < 10 μM) from a set of 18,886 compounds with Mtb data233 (Figure 3). The only TB lead that appears separate from the main cluster of TB Actives is 114 which is a Bis-Substituted Cyclam (Table S2 and S3) with a high molecular weight and low ALogP. This analysis shows that the compounds in this review are representative of the chemical property space of the thousands of Mtb “active” compounds published by many labs to date and when compared to approved drugs (Figure 4). Digging deeper into the outlier molecules as described herein suggests that the extremes of ALogP and molecular weight (and perhaps other simple descriptors) need to be further explored in detail, as most of the TB leads seem to be in the “sweet spot” of mean molecule properties that we have seen covered over the years with our various analyses.226, 227

Figure 3.

Figure 3.

Principal Component Analysis of compounds in this review (blue) alongside a curated set of Mtb actives228 (red, MIC < 10 μM threshold). The PCA generated with eight descriptors showed that Mtb actives are overlapped with the 118 Mtb compound dataset; with the three components of PCA representing 85.78% of the variance (x = 55.82%, y = 18.65%, z = 11.31%). Lead molecule 114 is clearly outside of the main cluster of TB actives (for structure see Table S3).

Figure 4.

Figure 4.

Principal Component Analysis of 118 lead compounds in this review (blue) alongside a curated set of Mtb actives228 (red, MIC < 10 μM threshold), approved drugs (green) and Microsource spectrum (yellow) and we then generated a PCA fitted with eight descriptors of the merged frame including all four datasets. The three components explained a variation of 89.57% (x = 52.89%, y = 24.39%, z = 12.29%). A majority of the four datasets were found overlapped in the same PCA space.

While we can attempt to classify the lead compounds structurally, we can also organize them by each property (Figure 5). This type of circular dendrogram approach while showing a relatively close proximity in log P property space alone, clearly indicates very few low logP molecules amongst these Mtb leads, for example structurally similar natural products or analogs 66 and 67, 69 and 70. This would suggest the need to further explore natural product or natural product derivative property space. These various molecule visualization methods are far more realistic in grouping the molecules than by classes based on structure types.

Figure 5.

Figure 5.

Circular dendrogram obtained from the hierarchical clustering of the 118 lead compounds for Mtb using eight descriptors, Ward linkage, and Euclidian distance. Compound nodes and names are colored according to their MolLogP.

MACHINE LEARNING FOR TB DRUG DISCOVERY

Alternative approaches to identify new leads for TB can learn from past data. We and others235 have published widely using the naïve Bayesian approach for tuberculosis drug discovery.227, 236. Models that combined bioactivity and cytotoxicity data were used to rank compounds such as the GSK antimalarial dataset.237 From the top 46 molecules, seven were chosen and five had MIC ≤ 2 μg/mL, the most active 104 being 0.0625 μg/mL.238 A second example used two different Mtb whole cell models to score three vendor libraries from which 550 compounds were tested and 124 actives identified.239 A third example filtered a library of >150,000 molecules and tested 48 compounds of which 11 were active225 including 109. The models achieved screening hit rates of 15–71% for suggested compounds, far higher than the 0.6 – 1.5% typical for random library HTS screening.85, 239, 240 Fusion of three dual activity models gave an excellent ROC value with a fourth external dataset from the same laboratory.241 These models have also been used individually with a test set of 1,924 molecules for which cytotoxicity was determined in three cell lines and enrichments of 11.8 fold were observed in the best case.242 Fusing single point data (bioactivity only) with dual activity data ultimately led to models with 345,011 molecules in them but these were no more predictive that the smaller dual activity datasets when tested with external data.243 We have also used Bayesian and additional machine learning approaches to model data from the mouse Mtb efficacy model that have been published over the last 70 years. Models were initially constructed with 773 compounds and used to predict 11 molecules from the literature (eight were correctly predicted).244 More recently these models have been updated and used with a test set of 60 molecules. The best Mtb in vivo models in this case now possess 5-fold ROC values > 0.7, sensitivity > 80% and concordance > 60%. These results indicated Bayesian models using literature in vivo Mtb data generated by different laboratories in many different mouse models can be predictive and also be used alongside other models to select antitubercular compounds with in vivo efficacy.245 We have also recently curated the Mtb data and developed newer machine learning models with 18,886 molecules (with activity cut offs of 10 μM, 1 μM and 100 nM).233 These datasets were used to evaluate many different machine learning methods (including deep learning and support vector machines) and were assessed with additional molecules published in 2017. The 100 nM cutoff model was tested with an evaluation set (n = 153 compounds) and showed comparable statistics to those seen with 5-fold cross validation (Accuracy = 0.83, Precision = 0.27, Recall = 1.00, Specificity = 0.81, Kappa = 0.36, and MCC = 0.47). Bayesian machine learning models constructed with literature Mtb data generated by different laboratories generally were equivalent to or outperformed Deep Neural Networks with external test sets.233 There are also individual efforts that have developed large numbers of promising leads such as the HTS of two million compounds that resulted in the GSK set of 177 lead compounds.246 We have previously shown that our Bayesian models identified some of these compounds before they were even published when they were a subset of an antimalarial library (described earlier237) providing an excellent example of prospective prediction.247 This raises the larger question of whether these models are able to identify the GSK leads because the models generated with large numbers of compounds from the SRI/TAACF screens were of a broad diversity that they essentially cover Mtb property space. Alternatively, we may be interpolating from these compounds and did the GSK library just sample close analogs of the SRI/TAACF library? Based on analysis of the literature it is clear from this work that we are quite adept at rediscovering compounds from the past, finding close analogs or structural scaffolds from years previously. Finding truly novel original molecules that are active against Mtb (and for that matter any bacteria) from machine learning models is a challenge we need to explore further in future because it is still a work in progress.

CONCLUSION

The recent development of tuberculosis research and development programs has been fruitful in terms of generating thousands of preclinical leads. Over the past decade alone, TB drug discovery has advanced with the screening of large drug-like and pharmaceutical company libraries starting with the TAACF in 2009 screening over 300,000 compounds.7779 Several companies such as Novartis and GSK have screened even more compounds, in the many millions.172, 246 The GSK library phenotypic screens alone covered 2 million molecules.246 It is likely that in total well over five million compounds have been tested against Mtb by academia and big pharmaceutical companies. These chemical libraries likely have some overlap but nevertheless many chemical classes have been examined, and more unique lead compounds were discovered in this way. Therefore, there is now a very low chance of finding new compounds and we will have to switch to looking for compounds against known targets or explore other sources of molecules. It may also be a challenge to come up with designs of new compounds which can address drug resistance. An alternative approach is the design and development of compounds which do not kill bacteria but instead block their virulence.248, 249 These compounds likely will not have an MIC in in vitro testing but will have in vivo activity. This points to further testing of compounds in in vivo animal models which is certainly expensive or optimizing combinations of drugs using other biochemical approaches. These alternative approaches will also have issues in how one goes after them and the need for expensive downstream animal/combination studies.

It should be noted that academics and industry are not always truly using and acknowledging available public resources such as the earlier NIH screens, but are often times rediscovering known actives and only poorly improving on them. That includes the data on their toxicity, the drug-like properties of the scaffolds and general oral availability of these Mtb active molecules. Our earlier efforts with the Collaborative Drug Discovery database to make these Mtb data and machine learning models available represent some of the earliest attempts to help other research programs learn from this accumulated data and support open source drug discovery.83, 84 Mtb drug discovery needs a truly field-wide effort to succeed (it takes a village rather than isolated academics), and more needs to be done faster even still with all the issues of cost, compliance, drug resistance and many thousands of people still dying annually. The Global Alliance’s recent advances (their approval of pretomanid and general ownership of the TB pipeline) are worthy of highlighting especially as it has likely involved a massive investment in collaborative drug discovery and development. The GSK Open Source efforts (at a fraction of the Global Alliance’s budget) are also notable and should be lauded for their making hit and lead compounds accessible to the public as well as highlighting true modern pharmaceutical drug discovery research, particularly paradigms for advancement of compounds for in vivo and preclinical toxicity and pharmacokinetics assessment. Unfortunately, these many efforts have not yet led to new drugs and most of these resulting publications (some of which are included herein) represent molecules that did not perform well when they reached in vivo. One of our observations that derived from the efforts at collating leads for this publication was that many papers still appear on the same poor, toxic insoluble scaffolds without all the necessary minimal information to truly evaluate candidates (i.e. solubility, toxicity in mammalian cells etc.) that are now standards in antibacterial drug discovery. Still, the increasing academic and industry collaborations has tended to at least anecdotally improve the drug-likeness of the compounds that resulted. Unfortunately, our cheminformatics analysis suggests that still, the recent lead compounds occupy the same property space as known drugs (Figure 2) as well as the larger set of TB in vitro or in vivo actives (Figure 3 and 4). There are however, several “natural product like” or large molecules 66, 67, 69, 70, 71, 114 or others 19, 41, 84, 85, 94 that are at the extremes of the ALogP scale (Table S2) compared to the majority of the TB leads (Figure 2A) and they stand out from the rest. It may be worth exploring these outlier areas of property space further. Some of the TB lead molecules also appear to contain PAINS105 and/or structural features that represent increased compound assay promiscuity.106 These molecules may need to be avoided or deprioritized. It is likely that limited structural diversity within screening libraries is also limiting the target diversity covered, which might explain why we see so many compounds for the promiscuous targets such as Mmpl3 and DprE1. Alternatively, and a common perception, it is possible that Mtb drug discovery has unique challenges due to the cellular barriers and druggability of targets requiring extremely small molecules or larger lipophilic compounds to access and bind targets within the cell. While there are many issues with Mtb drug discovery still to be answered from the chemistry/target perspective, the question of why lipophilic sulfur-containing compounds are hits in many Mtb screens is also intriguing. Is this just a case of metabolic activation or is there something else involved?

In this perspective, we have reviewed many different classes of compounds representing respectable coverage of Mtb chemical property space that might suggest reasonable target diversity and a promising foundation for the development of new drugs that will address burgeoning Mtb drug-resistance in the clinic. This abundance of preclinical leads, many of which are active in mice and / or active versus MDR strains of Mtb provides some hope, although the current clinical trial pipeline remains limited (Figure 1). Other reviews have suggested there is a need for novel targets.75 While the current perspective certainly confirms this finding, it is notable that there are also many compounds where the MOAs are unknown, and it is possible that unique and new targets may be hiding in plain sight. Perhaps a more critical observation is the generally limited variety of structural classes utilized and the general homogeneity of chemistry space. These current successes in generating new leads for Mtb have been driven primarily by funding from NIH/NIAID and BMGF as well as to some extent EU funding and to a lesser-extent, investments from pharmaceutical companies.5 While results from these programs are promising, there is still a dire need to develop more new leads in the hope that some of them will advance to the clinic for TB. More commitment to developing shorter treatment regimens are dependent on strong synergistic interactions between drugs and more screens for synergy are also needed. However, rarely does a compound with in vitro synergy translate to in vivo synergy250, more often than not the differences in ADME and PK properties of the two or more combined drugs will result in antagonism or no benefit250. Additionally, efforts to identify compounds affecting latent TB are of crucial importance to better target an essentially untreated population of bacilli in both actively diseased as well as latently infected patients251. Certainly, the compatibility of new TB agents and antiretroviral drugs to treat HIV-infected TB patients is another key issue that has to be addressed.252, 253 The complexity of developing treatments for TB has led to the exploration of various chemical cores described herein that are outside traditional classes of antibacterials or antituberculars as shown in some of the nearly 118 lead compounds selected (Table 312). Furthermore, many perfectly good drugs are available that could be revisited for delivery via different routes. For example, a recent review has summarized prospects for using inhaled drug delivery to reinvigorate TB treatment.254 In this vein, there are a large number of published examples of leads that possess poor PK and efficacy when dosed orally (including those described in this review) and this set might serve as an excellent reservoir of new drugs using an alternative delivery route such as inhalation. After decades of relatively modest outcomes in Mtb drug discovery and development, we think it is time for a ‘breath of fresh air’ in this area.

While there have been some moderate successes in the field over the past decade, as researchers we are having to run faster just to stay in place. Clearly the investments to date have under-delivered but perhaps our expectations may be set unrealistically high. For example: should we move beyond relying on oral drug delivery, and do we always need to know the target for a new hit or lead? Hundreds of high-quality papers over the past decade that delve into the detailed mechanistic underpinnings of molecules for Mtb have not delivered more than a small fraction that are viable clinical candidates. How can we shift from having generations of TB scientists producing the same types of closely related molecules? We hope that this goes some way towards recognizing the need for fresh perspectives on Mtb drug discovery in order to answer some of these questions and lead us to another golden era in antibacterial drug discovery. The current pipeline is dominated by one organization, the TB Alliance. Maybe it is time for them and the community in general to rethink expectations of Mtb preclinical candidates and how we could perform drug discovery differently. We have presented evidence for how we can learn from the published Mtb data on lead compounds using computational models such as machine learning and cheminformatics analyses which may help us to explore new structural classes and property space. These types of approaches have been under-utilized in Mtb drug discovery likely due to a lack of funding and also acceptance, this may change with the many recent articles on machine learning. It is abundantly clear there is still a bias towards small molecules when there may be very large natural products (e.g. macrolactones etc.) that could have promising antitubercular activity. We certainly have not explored all of these large molecules to date. It is well overdue to do this and, as importantly, we need to better learn from the molecules and data that have accumulated over the years. It is not in our best interests to ignore the collective preceding chemistry for antituberculars, but we do need to shift to new areas of chemistry because we are continuously covering the same ground. This perspective represents our latest efforts in nearly a decade of analysis of small molecules for Mtb in order to provide fresh insights to improve our productivity and likelihood of success in future.

Supplementary Material

supplemental material

ACKNOWLEDGMENTS

We kindly acknowledge NIH NINDS: 1R01NS102164-01, RFBR 17-54-30007, SC RF AAAA-A19-119010590004-2 and NIAID R41AI13456. SE kindly acknowledges Dr. Miriam Braunstein (UNC-Chapel Hill), Dr. Anthony Hickey (RTI), Dr. Joel Freundlich (Rutgers University), Dr. Ana Puhl (Collaborations Pharmaceuticals, Inc.) and Dr. Thomas Lane (Collaborations Pharmaceuticals, Inc.) for collaborations on tuberculosis and Dr. Barbara Laughon (NIAID) for permission to use the TB pipeline figure. SE kindly thanks BIOVIA for providing Discovery Studio.

ABBREVIATIONS USED

ADMET

absorption, distribution, metabolism, excretion and toxicity

AHAS

Acetohydroxyacid Synthases

Ddn

deazaflavin-dependent nitroreductase

DprE1

decaprenylphosphoryl-β-d-ribose oxidase

FtsZ

Filamenting temperature-sensitive mutant Z

GSK

GlaxoSmithKline

hERG

human ether-a-gogo-related gene

HIV

human immunodeficiency virus

HTS

high throughput screening

IMPDH

inosine-5’-monophosphate dehydrogenase

InhA

enoyl-ACP reductase

KasA

β-Ketoacyl ACP synthase I

LeuRS

leucyl-tRNA synthetase

LE

Ligand Efficiency

MDR

multidrug resistant

MmpL3

Mycobacterial membrane protein Large 3

MOA

mechanism of action

Mptpb

protein tyrosine phosphatase

MraY

phospho-MurNAc-pentapeptide translocase

Mtb

Mycobacterium tuberculosis

NIAID

National Institute of Allergy and Infectious Diseases

NO

nitric oxide

PAINS

Pan Assay Interference compounds

PanC

pantothenate synthetase

PCA

principal component analysis

PK

pharmacokinetics

Pks13

polyketide synthase 13

PyrG

CTP synthetase

QcrB

The b subunit of cytochrome bcc complex

ROC

receiver operator characteristic

SAR

Structure Activity Relationship

SI

Selectivity Index, SRI/TAACF Southern Research International/Tuberculosis Antimicrobial Acquisition and Coordinating Facility

TB

Tuberculosis

TDR

totally drug resistant

WecA

phosphoglycosyltransferase

WHO

World Health Organisation

XDR

extensively drug resistant

Biographies

Biographies

Vadim Makarov is the Chief of Lab in the Dept. of Stresses of Microorganisms, A.N. Bach Institute of Biochemistry, Moscow, Russian Federation. He is an expert in medicinal chemistry and rational drug design with special interest in developing antimicrobial and antiviral agents. His recent work in drug development has focused on antituberculosis drugs and as result of his efforts in cooperation with several European scientific groups (NM4TB; LSHP-CT-2005–018923) a new class of synthetic organic compounds having high antituberculosis activity in vivo and in vitro was discovered (1,3-benzothiazinones). Two compounds from this group are under in-depth preclinical investigation, with PBTZ169 being the lead candidate. Dr Makarov’s team makes extensive use of advanced methods of medicinal chemistry, rational drug design and study of metabolic transformations.

Elena G. Salina received a M.Sc. degree from the Mendeleev University of Chemical Technology of Russia in 2002. Then she began graduate studies at the Bach Institute of Biochemistry of the Russian Academy of Sciences where she received a Ph.D. in Biochemistry under the direction of Professor A. Kaprelyants in 2006. She conducted postdoctoral studies at the same Institute working on M. tuberculosis persistence and modelling mycobacterial dormancy. Later she worked in the field of finding new drugs for latent tuberculosis at the Federal Research Center of Biotechnology of the Russian Academy of Sciences where she is currently a Senior Scientist. Dr. Elena G. Salina’s research interests concern the discovery of new drugs for the treatment of latent tuberculosis infection and multi-targeted non-selective inhibition.

Robert C. Reynolds received a B.S. in Chemistry from the University of Virginia in 1977 and a PhD. from Duke University in 1985. While a student at Duke University under Dr. Barbara Shaw in Chemistry and Dr. David Sedwick in the Department of Medicine. After postdoctoral fellowship at NIEHS in the Research Triangle Park with Dr. Robert London in the area of probe development for in vivo NMR metabolism, Dr. Reynolds began a 25-year career in drug design and synthesis at Southern Research Institute in Birmingham, Alabama reaching the position of Director of Drug Discovery Technology. His major areas of emphasis were new target and drug discovery against cancer and infectious diseases, particularly tuberculosis. In 2012, Dr. Reynolds joined the University of Alabama at Birmingham.

Phyo Phyo Kyaw Zin received her B.A. in Chemistry and Computer Science from Berea College. She is now a Ph.D. candidate in the Fourches lab at North Carolina State University and a recipient of the AAUW International Doctoral Fellowship and Olive Ruth Russell Scholarships. Her research areas involve molecular informatics, computer-aided drug design, development of cheminformatics approaches/software to generate virtual chemical libraries, analysis and visualization of big chemical databases, and implementation of AI-powered (ML such as DLNN) cheminformatics methods to build QSAR models.

Sean Ekins graduated from the University of Aberdeen; receiving his M.Sc., Ph.D. and D.Sc in Clinical Pharmacology. He was a postdoctoral fellow at Lilly Research Laboratories then worked as a senior scientist at Pfizer and Lilly Research Laboratories, Associate Director of Computational Drug Discovery at Concurrent Pharmaceuticals, Inc., Vice President of Computational Biology at GeneGo, Then worked for a number of companies, as CSO at Collaborative Drug Discovery and is currently CEO and Founder at Collaborations Pharmaceuticals, Inc. He is an Entrepreneur-in-residence at the Eshelman School of Pharmacy, UNC Chapel Hill. Sean has co-authored >300 peer reviewed scientific papers, book chapters and patents as well as edited/ co-edited five books. He has worked for over a decade on tuberculosis drug discovery using machine learning.

Footnotes

COMPETING INTERESTS

S.E. is CEO and owner, of Collaborations Pharmaceuticals, Inc.

References

  • 1.Anon. The Stop TB Partnership’s Working Group On New TB Drugs www.newtbdrugs.org (accessed Mar 3, 2020).
  • 2.Low M The Tuberculosis Treatment Pipeline: A Breakthrough Year For The Treatment Of XDR-TB. https://www.treatmentactiongroup.org/resources/pipeline-report/2017-pipeline-report/the-tuberculosis-treatment-pipeline-a-breakthrough-year-for-the-treatment-of-xdr-tb/ (accessed Mar 3, 2020,
  • 3.WHO. Global Tuberculosis Report 2018. https://www.who.int/tb/publications/global_report/en/ (accessed Mar 3, 2020).
  • 4.Jakab Z; Acosta CD; Kluge HH; Dara M Consolidated action plan to prevent and combat multidrug- and extensively drug-resistant tuberculosis in the WHO european region 2011–2015: Cost-effectiveness analysis. Tuberculosis (Edinb) 2015, 95 Suppl 1, S212–S216. [DOI] [PubMed] [Google Scholar]
  • 5.Mikusova K; Ekins S Learning from the past for tb drug discovery in the future. Drug Discov Today 2017, 22, 534–545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Cole ST Tuberculosis drug discovery needs public-private consortia. Drug Discov Today 2016, 22, 477–478. [DOI] [PubMed] [Google Scholar]
  • 7.Conradie F; Diacon AH; Ngubane N; Howell P; Everitt D; Crook AM; Mendel CM; Egizi E; Moreira J; Timm J; McHugh TD; Wills GH; Bateson A; Hunt R; Van Niekerk C; Li M; Olugbosi M; Spigelman M; Nix TBTT Treatment of highly drug-resistant pulmonary tuberculosis. N Engl J Med 2020, 382, 893–902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Thwaites G; Nahid P Triumph and tragedy of 21st century tuberculosis drug development. N Engl J Med 2020, 382, 959–960. [DOI] [PubMed] [Google Scholar]
  • 9.Vilcheze C; Jacobs WR Jr. The mechanism of isoniazid killing: Clarity through the scope of genetics. Annu Rev Microbiol 2007, 61, 35–50. [DOI] [PubMed] [Google Scholar]
  • 10.Scorpio A; Zhang Y Mutations in pncA, a gene encoding pyrazinamidase/nicotinamidase, cause resistance to the antituberculous drug pyrazinamide in tubercle bacillus. Nat Med 1996, 2, 662–667. [DOI] [PubMed] [Google Scholar]
  • 11.Heifets LB; Flory MA; Lindholm-Levy PJ Does pyrazinoic acid as an active moiety of pyrazinamide have specific activity against mycobacterium tuberculosis? Antimicrob Agents Chemother 1989, 33, 1252–1254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Botha FJ; Sirgel FA; Parkin DP; van de Wal BW; Donald PR; Mitchison DA Early bactericidal activity of ethambutol, pyrazinamide and the fixed combination of isoniazid, rifampicin and pyrazinamide (Rifater) in patients with pulmonary tuberculosis. S Afr Med J 1996, 86, 155–158. [PubMed] [Google Scholar]
  • 13.Nusrath Unissa A; Hanna LE Molecular mechanisms of action, resistance, detection to the first-line anti tuberculosis drugs: Rifampicin and pyrazinamide in the post whole genome sequencing era. Tuberculosis (Edinb) 2017, 105, 96–107. [DOI] [PubMed] [Google Scholar]
  • 14.Andries K; Verhasselt P; Guillemont J; Gohlmann HW; Neefs JM; Winkler H; Van Gestel J; Timmerman P; Zhu M; Lee E; Williams P; de Chaffoy D; Huitric E; Hoffner S; Cambau E; Truffot-Pernot C; Lounis N; Jarlier V A diarylquinoline drug active on the ATP synthase of mycobacterium tuberculosis. Science 2005, 307, 223–227. [DOI] [PubMed] [Google Scholar]
  • 15.Diacon AH; Pym A; Grobusch M; Patientia R; Rustomjee R; Page-Shipp L; Pistorius C; Krause R; Bogoshi M; Churchyard G; Venter A; Allen J; Palomino JC; De Marez T; van Heeswijk RP; Lounis N; Meyvisch P; Verbeeck J; Parys W; de Beule K; Andries K; Mc Neeley DF The diarylquinoline TMC207 for multidrug-resistant tuberculosis. N Engl J Med 2009, 360, 2397–2405. [DOI] [PubMed] [Google Scholar]
  • 16.Koul A; Dendouga N; Vergauwen K; Molenberghs B; Vranckx L; Willebrords R; Ristic Z; Lill H; Dorange I; Guillemont J; Bald D; Andries K Diarylquinolines target subunit c of mycobacterial ATP synthase. Nat Chem Biol 2007, 3, 323–324. [DOI] [PubMed] [Google Scholar]
  • 17.Koul A; Vranckx L; Dendouga N; Balemans W; Van den Wyngaert I; Vergauwen K; Göhlmann HW; Willebrords R; Poncelet A; Guillemont J; Bald D; Andries K Diarylquinolines are bactericidal for dormant mycobacteria as a result of disturbed ATP homeostasis. J Biol Chem 2008, 283, 25273–25280. [DOI] [PubMed] [Google Scholar]
  • 18.Haagsma AC; Abdillahi-Ibrahim R; Wagner MJ; Krab K; Vergauwen K; Guillemont J; Andries K; Lill H; Koul A; Bald D Selectivity of TMC207 towards mycobacterial atp synthase compared with that towards the eukaryotic homologue. Antimicrob Agents Chemother 2009, 53, 1290–1292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.de Jonge MR; Koymans LH; Guillemont JE; Koul A; Andries K A computational model of the inhibition of mycobacterium tuberculosis ATPase by a new drug candidate r207910. Proteins 2007, 67, 971–980. [DOI] [PubMed] [Google Scholar]
  • 20.Smyej I; De Jonghe S; Looszova A; Mannens G; Verhaeghe T; Thijssen S; Starckx S; Lampo A; Rouan MC Dose- and time-dependency of the toxicity and pharmacokinetic profiles of bedaquiline and its n-desmethyl metabolite in dogs. Toxicol Pathol 2017, 45, 663–675. [DOI] [PubMed] [Google Scholar]
  • 21.Tong AST; Choi PJ; Blaser A; Sutherland HS; Tsang SKY; Guillemont J; Motte M; Cooper CB; Andries K; Van den Broeck W; Franzblau SG; Upton AM; Denny WA; Palmer BD; Conole D 6-cyano analogues of bedaquiline as less lipophilic and potentially safer diarylquinolines for tuberculosis. ACS Med Chem Lett 2017, 8, 1019–1024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Guglielmetti L; Le Du D; Jachym M; Henry B; Martin D; Caumes E; Veziris N; Metivier N; Robert J; Mycobacteria M-T M. G. o. t. F. N. R. C. f.; the Physicians of the French, M. D. R. T. B. C. Compassionate use of bedaquiline for the treatment of multidrug-resistant and extensively drug-resistant tuberculosis: Interim analysis of a french cohort. Clin Infect Dis 2015, 60, 188–194. [DOI] [PubMed] [Google Scholar]
  • 23.Esposito S; Bianchini S; Blasi F Bedaquiline and delamanid in tuberculosis. Expert Opin Pharmacother 2015, 16, 2319–2330. [DOI] [PubMed] [Google Scholar]
  • 24.Stover CK; Warrener P; VanDevanter DR; Sherman DR; Arain TM; Langhorne MH; Anderson SW; Towell JA; Yuan Y; McMurray DN; Kreiswirth BN; Barry CE; Baker WR A small-molecule nitroimidazopyran drug candidate for the treatment of tuberculosis. Nature 2000, 405, 962–966. [DOI] [PubMed] [Google Scholar]
  • 25.Anon. FDA Approves New Treatment For Highly Drug-Resistant Forms Of Tuberculosis. https://www.tballiance.org/news/fda-approves-new-treatment-highly-drug-resistant-forms-tuberculosis (accessed Mar 3, 2020).
  • 26.Zumla A; Nahid P; Cole ST Advances in the development of new tuberculosis drugs and treatment regimens. Nat Rev Drug Discov 2013, 12, 388–404. [DOI] [PubMed] [Google Scholar]
  • 27.Klopper M; Warren RM; Hayes C; Gey van Pittius NC; Streicher EM; Müller B; Sirgel FA; Chabula-Nxiweni M; Hoosain E; Coetzee G; David van Helden P; Victor TC; Trollip AP Emergence and spread of extensively and totally drug-resistant tuberculosis, South Africa. Emerg Infect Dis 2013, 19, 449–455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Slomski A South Africa warns of emergence of “totally” drug-resistant tuberculosis. JAMA 2013, 309, 1097–1098. [DOI] [PubMed] [Google Scholar]
  • 29.WHO. Latent Tuberculosis Infection. apps.who.int/iris/bitstream/10665/260233/1/9789241550239-eng.pdf (accessed Mar 3, 2020).
  • 30.Zhang Y; Yew WW; Barer MR Targeting persisters for tuberculosis control. Antimicrob Agents Chemother 2012, 56, 2223–2230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Iacobino A; Piccaro G; Giannoni F; Mustazzolu A; Fattorini L Fighting tuberculosis by drugs targeting nonreplicating mycobacterium tuberculosis bacilli. Int J Mycobacteriol 2017, 6, 213–221. [DOI] [PubMed] [Google Scholar]
  • 32.WHO. Global Tuberculosis Report 2017; World Health Organisation: 2017. https://www.who.int/tb/publications/global_report/gtbr2017_main_text.pdf (accessed Mar 3, 2020).
  • 33.WHO. Global Ministerial Conference On Ending TB In The Sustainable Development Era. http://www.who.int/mediacentre/events/2017/ministerial-conference-tb/en/ (accessed Mar 3, 2020).
  • 34.Dooley KE; Phillips PP; Nahid P; Hoelscher M Challenges in the clinical assessment of novel tuberculosis drugs. Adv Drug Deliv Rev 2016, 102, 116–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Manjunatha UH; Smith PW Perspective: Challenges and opportunities in TB drug discovery from phenotypic screening. Bioorg Med Chem 2015, 23, 5087–5097. [DOI] [PubMed] [Google Scholar]
  • 36.Pai M; Behr MA; Dowdy D; Dheda K; Divangahi M; Boehme CC; Ginsberg A; Swaminathan S; Spigelman M; Getahun H; Menzies D; Raviglione M Tuberculosis. Nat Rev Dis Primers 2016, 2, 16076. [DOI] [PubMed] [Google Scholar]
  • 37.Wallis RS; Maeurer M; Mwaba P; Chakaya J; Rustomjee R; Migliori GB; Marais B; Schito M; Churchyard G; Swaminathan S; Hoelscher M; Zumla A Tuberculosis--advances in development of new drugs, treatment regimens, host-directed therapies, and biomarkers. Lancet Infect Dis 2016, 16, e34–46. [DOI] [PubMed] [Google Scholar]
  • 38.Bloemberg GV; Keller PM; Stucki D; Stuckia D; Trauner A; Borrell S; Latshang T; Coscolla M; Rothe T; Hömke R; Ritter C; Feldmann J; Schulthess B; Gagneux S; Böttger EC Acquired resistance to bedaquiline and delamanid in therapy for tuberculosis. N Engl J Med 2015, 373, 1986–1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Segala E; Sougakoff W; Nevejans-Chauffour A; Jarlier V; Petrella S New mutations in the mycobacterial atp synthase: New insights into the binding of the diarylquinoline TMC207 to the atp synthase c-ring structure. Antimicrob Agents Chemother 2012, 56, 2326–2334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Zhang S; Chen J; Cui P; Shi W; Shi X; Niu H; Chan D; Yew WW; Zhang W; Zhang Y Mycobacterium tuberculosis mutations associated with reduced susceptibility to linezolid. Antimicrob Agents Chemother 2016, 60, 2542–2544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Mukherjee T; Boshoff H Nitroimidazoles for the treatment of TB: Past, present and future. Future Med Chem 2011, 3, 1427–1454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Palmer BD; Sutherland HS; Blaser A; Kmentova I; Franzblau SG; Wan B; Wang Y; Ma Z; Denny WA; Thompson AM Synthesis and structure-activity relationships for extended side chain analogues of the antitubercular drug (6s)-2-nitro-6-{[4-(trifluoromethoxy)benzyl]oxy}−6,7-dihydro-5h-imidazo[2,1-b][1, 3]oxazine (pa-824). J Med Chem 2015, 58, 3036–3059. [DOI] [PubMed] [Google Scholar]
  • 43.Stinson K; Kurepina N; Venter A; Fujiwara M; Kawasaki M; Timm J; Shashkina E; Kreiswirth BN; Liu Y; Matsumoto M; Geiter L MIC of delamanid (OPC-67683) against mycobacterium tuberculosis clinical isolates and a proposed critical concentration. Antimicrob Agents Chemother 2016, 60, 3316–3322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Lenaerts AJ; Gruppo V; Marietta KS; Johnson CM; Driscoll DK; Tompkins NM; Rose JD; Reynolds RC; Orme IM Preclinical testing of the nitroimidazopyran PA-824 for activity against mycobacterium tuberculosis in a series of in vitro and in vivo models. Antimicrob Agents Chemother 2005, 49, 2294–2301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Matsumoto M; Hashizume H; Tomishige T; Kawasaki M; Tsubouchi H; Sasaki H; Shimokawa Y; Komatsu M OPC-67683, a nitro-dihydro-imidazooxazole derivative with promising action against tuberculosis in vitro and in mice. PLoS Med 2006, 3, e466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Diacon AH; Dawson R; Hanekom M; Narunsky K; Venter A; Hittel N; Geiter LJ; Wells CD; Paccaly AJ; Donald PR Early bactericidal activity of delamanid (OPC-67683) in smear-positive pulmonary tuberculosis patients. Int J Tuberc Lung Dis 2011, 15, 949–954. [DOI] [PubMed] [Google Scholar]
  • 47.Gler MT; Skripconoka V; Sanchez-Garavito E; Xiao H; Cabrera-Rivero JL; Vargas-Vasquez DE; Gao M; Awad M; Park SK; Shim TS; Suh GY; Danilovits M; Ogata H; Kurve A; Chang J; Suzuki K; Tupasi T; Koh WJ; Seaworth B; Geiter LJ; Wells CD Delamanid for multidrug-resistant pulmonary tuberculosis. N Engl J Med 2012, 366, 2151–2160. [DOI] [PubMed] [Google Scholar]
  • 48.Skripconoka V; Danilovits M; Pehme L; Tomson T; Skenders G; Kummik T; Cirule A; Leimane V; Kurve A; Levina K; Geiter LJ; Manissero D; Wells CD Delamanid improves outcomes and reduces mortality in multidrug-resistant tuberculosis. Eur Respir J 2013, 41, 1393–1400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Ginsberg AM; Laurenzi MW; Rouse DJ; Whitney KD; Spigelman MK Safety, tolerability, and pharmacokinetics of PA-824 in healthy subjects. Antimicrob Agents Chemother 2009, 53, 3720–3725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ginsberg AM; Laurenzi MW; Rouse DJ; Whitney KD; Spigelman MK Assessment of the effects of the nitroimidazo-oxazine PA-824 on renal function in healthy subjects. Antimicrob Agents Chemother 2009, 53, 3726–3733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Diacon AH; Dawson R; Hanekom M; Narunsky K; Maritz SJ; Venter A; Donald PR; van Niekerk C; Whitney K; Rouse DJ; Laurenzi MW; Ginsberg AM; Spigelman MK Early bactericidal activity and pharmacokinetics of PA-824 in smear-positive tuberculosis patients. Antimicrob Agents Chemother 2010, 54, 3402–3407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Diacon AH; Dawson R; du Bois J; Narunsky K; Venter A; Donald PR; van Niekerk C; Erondu N; Ginsberg AM; Becker P; Spigelman MK Phase ii dose-ranging trial of the early bactericidal activity of PA-824. Antimicrob Agents Chemother 2012, 56, 3027–3031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Diacon AH; Dawson R; von Groote-Bidlingmaier F; Symons G; Venter A; Donald PR; van Niekerk C; Everitt D; Winter H; Becker P; Mendel CM; Spigelman MK 14-day bactericidal activity of PA-824, bedaquiline, pyrazinamide, and moxifloxacin combinations: A randomised trial. Lancet 2012, 380, 986–993. [DOI] [PubMed] [Google Scholar]
  • 54.Winter H; Egizi E; Erondu N; Ginsberg A; Rouse DJ; Severynse-Stevens D; Pauli E; Everitt D Evaluation of pharmacokinetic interaction between PA-824 and midazolam in healthy adult subjects. Antimicrob Agents Chemother 2013, 57, 3699–3703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Winter H; Ginsberg A; Egizi E; Erondu N; Whitney K; Pauli E; Everitt D Effect of a high-calorie, high-fat meal on the bioavailability and pharmacokinetics of PA-824 in healthy adult subjects. Antimicrob Agents Chemother 2013, 57, 5516–5520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Diacon AH; Dawson R; von Groote-Bidlingmaier F; Symons G; Venter A; Donald PR; van Niekerk C; Everitt D; Hutchings J; Burger DA; Schall R; Mendel CM Bactericidal activity of pyrazinamide and clofazimine alone and in combinations with pretomanid and bedaquiline. Am J Respir Crit Care Med 2015, 191, 943–953. [DOI] [PubMed] [Google Scholar]
  • 57.Dawson R; Diacon AH; Everitt D; van Niekerk C; Donald PR; Burger DA; Schall R; Spigelman M; Conradie A; Eisenach K; Venter A; Ive P; Page-Shipp L; Variava E; Reither K; Ntinginya NE; Pym A; von Groote-Bidlingmaier F; Mendel CM Efficiency and safety of the combination of moxifloxacin, pretomanid (PA-824), and pyrazinamide during the first 8 weeks of antituberculosis treatment: A phase 2b, open-label, partly randomised trial in patients with drug-susceptible or drug-resistant pulmonary tuberculosis. Lancet 2015, 385, 1738–1747. [DOI] [PubMed] [Google Scholar]
  • 58.Ashtekar DR; Costa-Perira R; Nagrajan K; Vishvanathan N; Bhatt AD; Rittel W In vitro and in vivo activities of the nitroimidazole CGI 17341 against mycobacterium tuberculosis. Antimicrob Agents Chemother 1993, 37, 183–186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Manjunatha UH; Boshoff H; Dowd CS; Zhang L; Albert TJ; Norton JE; Daniels L; Dick T; Pang SS; Barry CE 3rd. Identification of a nitroimidazo-oxazine-specific protein involved in PA-824 resistance in mycobacterium tuberculosis. Proc Natl Acad Sci U S A 2006, 103, 431–436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Zhang Q; Liu Y; Tang S; Sha W; Xiao H Clinical benefit of delamanid (opc-67683) in the treatment of multidrug-resistant tuberculosis patients in china. Cell Biochem Biophys 2013, 67, 957–963. [DOI] [PubMed] [Google Scholar]
  • 61.Singh R; Manjunatha U; Boshoff HI; Ha YH; Niyomrattanakit P; Ledwidge R; Dowd CS; Lee IY; Kim P; Zhang L; Kang S; Keller TH; Jiricek J; Barry CE PA-824 kills nonreplicating mycobacterium tuberculosis by intracellular no release. Science 2008, 322, 1392–1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Stover CK; Warrener P; VanDevanter DR; Sherman DR; Arain TM; Langhorne MH; Anderson SW; Towell JA; Yuan Y; McMurray DN; Kreiswirth BN; Barry CE; Baker WR A small-molecule nitroimidazopyran drug candidate for the treatment of tuberculosis. Nature 2000, 405, 962–966. [DOI] [PubMed] [Google Scholar]
  • 63.Parikh R; Dalwadi S; Aboti P; Patel L Inhaled microparticles of antitubercular antibiotic for in vitro and in vivo alveolar macrophage targeting and activation of phagocytosis. J Antibiot (Tokyo) 2014, 67, 387–394. [DOI] [PubMed] [Google Scholar]
  • 64.Manjunatha U; Boshoff HI; Barry CE The mechanism of action of PA-824: Novel insights from transcriptional profiling. Commun Integr Biol 2009, 2, 215–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Baptista R; Fazakerley DM; Beckmann M; Baillie L; Mur LAJ Untargeted metabolomics reveals a new mode of action of pretomanid (PA-824). Sci Rep 2018, 8, 5084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Wang X; Inoyama D; Russo R; Li SG; Jadhav R; Stratton TP; Mittal N; Bilotta JA; Singleton E; Kim T; Paget SD; Pottorf RS; Ahn YM; Davila-Pagan A; Kandasamy S; Grady C; Hussain S; Soteropoulos P; Zimmerman MD; Ho HP; Park S; Dartois V; Ekins S; Connell N; Kumar P; Freundlich JS Antitubercular triazines: Optimization and intrabacterial metabolism. Cell Chem Biol 2019, 7 (2), 172–185. e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Swaminathan S; Sundaramurthi JC; Palaniappan AN; Narayanan S Recent developments in genomics, bioinformatics and drug discovery to combat emerging drug-resistant tuberculosis. Tuberculosis (Edinb) 2016, 101, 31–40. [DOI] [PubMed] [Google Scholar]
  • 68.Campanico A; Moreira R; Lopes F Drug discovery in tuberculosis. New drug targets and antimycobacterial agents. Eur J Med Chem 2018, 150, 525–545. [DOI] [PubMed] [Google Scholar]
  • 69.Chiarelli LR; Mori G; Esposito M; Orena BS; Pasca MR New and old hot drug targets in tuberculosis. Curr Med Chem 2016, 23, 3813–3846. [DOI] [PubMed] [Google Scholar]
  • 70.Mdluli K; Kaneko T; Upton A The tuberculosis drug discovery and development pipeline and emerging drug targets. Cold Spring Harb Perspect Med 2015, 5. doi: 10.1101/cshperspect.a021154 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Fernandes G; Man Chin C; Dos Santos JL Advances in drug discovery of new antitubercular multidrug-resistant compounds. Pharmaceuticals (Basel) 2017, 10. doi: 10.3390/ph10020051 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Kumar V; Patel S; Jain R New structural classes of antituberculosis agents. Med Res Rev 2018, 38, 684–740. [DOI] [PubMed] [Google Scholar]
  • 73.da Silva PB; Campos DL; Ribeiro CM; da Silva IC; Pavan FR New antimycobacterial agents in the pre-clinical phase or beyond: Recent advances in patent literature (2001–2016). Expert Opin Ther Pat 2017, 27, 269–282. [DOI] [PubMed] [Google Scholar]
  • 74.Evangelopoulos D; McHugh TD Improving the tuberculosis drug development pipeline. Chem Biol Drug Des 2015, 86, 951–960. [DOI] [PubMed] [Google Scholar]
  • 75.Libardo MDJ; Boshoff HI; Barry CE 3rd. The present state of the tuberculosis drug development pipeline. Curr Opin Pharmacol 2018, 42, 81–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Trapero A; Pacitto A; Singh V; Sabbah M; Coyne AG; Mizrahi V; Blundell TL; Ascher DB; Abell C Fragment-based approach to targeting inosine-5’-monophosphate dehydrogenase (IMPDH) from mycobacterium tuberculosis. J Med Chem 2018, 61, 2806–2822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Ananthan S; Faaleolea ER; Goldman RC; Hobrath JV; Kwong CD; Laughon BE; Maddry JA; Mehta A; Rasmussen L; Reynolds RC; Secrist JA 3rd; Shindo N; Showe DN; Sosa MI; Suling WJ; White EL High-throughput screening for inhibitors of mycobacterium tuberculosis h37rv. Tuberculosis (Edinb) 2009, 89, 334–353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Maddry JA; Ananthan S; Goldman RC; Hobrath JV; Kwong CD; Maddox C; Rasmussen L; Reynolds RC; Secrist JA 3rd; Sosa MI; White EL; Zhang W Antituberculosis activity of the molecular libraries screening center network library. Tuberculosis (Edinb) 2009, 89, 354–363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Reynolds RC; Ananthan S; Faaleolea E; Hobrath JV; Kwong CD; Maddox C; Rasmussen L; Sosa MI; Thammasuvimol E; White EL; Zhang W; Secrist JA 3rd. High throughput screening of a library based on kinase inhibitor scaffolds against mycobacterium tuberculosis h37rv. Tuberculosis (Edinb) 2012, 92, 72–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Makarov V; Lechartier B; Zhang M; Neres J; van der Sar AM; Raadsen SA; Hartkoorn RC; Ryabova OB; Vocat A; Decosterd LA; Widmer N; Buclin T; Bitter W; Andries K; Pojer F; Dyson PJ; Cole ST Towards a new combination therapy for tuberculosis with next generation benzothiazinones. EMBO Mol Med 2014, 6, 372–383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Makarov V; Manina G; Mikusova K; Mollmann U; Ryabova O; Saint-Joanis B; Dhar N; Pasca MR; Buroni S; Lucarelli AP; Milano A; De Rossi E; Belanova M; Bobovska A; Dianiskova P; Kordulakova J; Sala C; Fullam E; Schneider P; McKinney JD; Brodin P; Christophe T; Waddell S; Butcher P; Albrethsen J; Rosenkrands I; Brosch R; Nandi V; Bharath S; Gaonkar S; Shandil RK; Balasubramanian V; Balganesh T; Tyagi S; Grosset J; Riccardi G; Cole ST Benzothiazinones kill mycobacterium tuberculosis by blocking arabinan synthesis. Science 2009, 324, 801–804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Ekins S; Freundlich JS; Choi I; Sarker M; Talcott C Computational databases, pathway and cheminformatics tools for tuberculosis drug discovery. Trends Microbiol 2011, 19, 65–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Ekins S; Kaneko T; Lipinski CA; Bradford J; Dole K; Spektor A; Gregory K; Blondeau D; Ernst S; Yang J; Goncharoff N; Hohman MM; Bunin BA Analysis and hit filtering of a very large library of compounds screened against mycobacterium tuberculosis. Mol Biosyst 2010, 6, 2316–2324. [DOI] [PubMed] [Google Scholar]
  • 84.Ekins S; Bradford J; Dole K; Spektor A; Gregory K; Blondeau D; Hohman M; Bunin BA A collaborative database and computational models for tuberculosis drug discovery. Mol Biosyst 2010, 6, 840–851. [DOI] [PubMed] [Google Scholar]
  • 85.Ekins S; Reynolds RC; Kim H; Koo MS; Ekonomidis M; Talaue M; Paget SD; Woolhiser LK; Lenaerts AJ; Bunin BA; Connell N; Freundlich JS Bayesian models leveraging bioactivity and cytotoxicity information for drug discovery. Chem Biol 2013, 20, 370–378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Mori G; Chiarelli LR; Esposito M; Makarov V; Bellinzoni M; Hartkoorn RC; Degiacomi G; Boldrin F; Ekins S; de Jesus Lopes Ribeiro AL; Marino LB; Centarova I; Svetlikova Z; Blasko J; Kazakova E; Lepioshkin A; Barilone N; Zanoni G; Porta A; Fondi M; Fani R; Baulard AR; Mikusova K; Alzari PM; Manganelli R; de Carvalho LP; Riccardi G; Cole ST; Pasca MR Thiophenecarboxamide derivatives activated by etha kill mycobacterium tuberculosis by inhibiting the CTP synthetase PyrG. Chem Biol 2015, 22, 917–927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ekins S; Madrid PB; Sarker M; Li SG; Mittal N; Kumar P; Wang X; Stratton TP; Zimmerman M; Talcott C; Bourbon P; Travers M; Yadav M; Freundlich JS Combining metabolite-based pharmacophores with bayesian machine learning models for mycobacterium tuberculosis drug discovery. PLoS One 2015, 10, e0141076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Song L; Merceron R; Gracia B; Quintana AL; Risseeuw MDP; Hulpia F; Cos P; Ainsa JA; Munier-Lehmann H; Savvides SN; Van Calenbergh S Structure guided lead generation toward nonchiral m. Tuberculosis thymidylate kinase inhibitors. J Med Chem 2018, 61, 2753–2775. [DOI] [PubMed] [Google Scholar]
  • 89.Nyantakyi SA; Li M; Gopal P; Zimmerman M; Dartois V; Gengenbacher M; Dick T; Go ML Indolyl azaspiroketal mannich bases are potent antimycobacterial agents with selective membrane permeabilizing effects and in vivo activity. J Med Chem 2018, 61, 5733–5750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Cleghorn LAT; Ray PC; Odingo J; Kumar A; Wescott H; Korkegian A; Masquelin T; Lopez Moure A; Wilson C; Davis S; Huggett M; Turner P; Smith A; Epemolu O; Zuccotto F; Riley J; Scullion P; Shishikura Y; Ferguson L; Rullas J; Guijarro L; Read KD; Green SR; Hipskind P; Parish T; Wyatt PG Identification of morpholino thiophenes as novel mycobacterium tuberculosis inhibitors, targeting QcrB. J Med Chem 2018, 61, 6592–6608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Chacko S; Boshoff HIM; Singh V; Ferraris DM; Gollapalli DR; Zhang M; Lawson AP; Pepi MJ; Joachimiak A; Rizzi M; Mizrahi V; Cuny GD; Hedstrom L Expanding benzoxazole-based inosine 5’-monophosphate dehydrogenase (IMPDH) inhibitor structure-activity as potential antituberculosis agents. J Med Chem 2018, 61, 4739–4756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Spain M; Wong JK; Nagalingam G; Batten JM; Hortle E; Oehlers SH; Jiang XF; Murage HE; Orford JT; Crisologo P; Triccas JA; Rutledge PJ; Todd MH Antitubercular bis-substituted cyclam derivatives: Structure-activity relationships and in vivo studies. J Med Chem 2018, 61, 3595–3608. [DOI] [PubMed] [Google Scholar]
  • 93.Azzali E; Machado D; Kaushik A; Vacondio F; Flisi S; Cabassi CS; Lamichhane G; Viveiros M; Costantino G; Pieroni M Substituted n-phenyl-5-(2-(phenylamino)thiazol-4-yl)isoxazole-3-carboxamides are valuable antitubercular candidates that evade innate efflux machinery. J Med Chem 2017, 60, 7108–7122. [DOI] [PubMed] [Google Scholar]
  • 94.Machado D; Azzali E; Couto I; Costantino G; Pieroni M; Viveiros M Adjuvant therapies against tuberculosis: Discovery of a 2-aminothiazole targeting mycobacterium tuberculosis energetics. Future Microbiol 2018, 13, 1383–1402. [DOI] [PubMed] [Google Scholar]
  • 95.Wilson CR; Gessner RK; Moosa A; Seldon R; Warner DF; Mizrahi V; Soares de Melo C; Simelane SB; Nchinda A; Abay E; Taylor D; Njoroge M; Brunschwig C; Lawrence N; Boshoff HIM; Barry CE 3rd; Sirgel FA; van Helden P; Harris CJ; Gordon R; Ghidelli-Disse S; Pflaumer H; Boesche M; Drewes G; Sanz O; Santos G; Rebollo-Lopez MJ; Urones B; Selenski C; Lafuente-Monasterio MJ; Axtman M; Lelievre J; Ballell L; Mueller R; Street LJ; Ghorpade SR; Chibale K Novel antitubercular 6-dialkylaminopyrimidine carboxamides from phenotypic whole-cell high throughput screening of a softfocus library: Structure-activity relationship and target identification studies. J Med Chem 2017, 60, 10118–10134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Dos Santos Fernandes GF; de Souza PC; Moreno-Viguri E; Santivanez-Veliz M; Paucar R; Perez-Silanes S; Chegaev K; Guglielmo S; Lazzarato L; Fruttero R; Man Chin C; da Silva PB; Chorilli M; Solcia MC; Ribeiro CM; Silva CSP; Marino LB; Bosquesi PL; Hunt DM; de Carvalho LPS; de Souza Costa CA; Cho SH; Wang Y; Franzblau SG; Pavan FR; Dos Santos JL Design, synthesis, and characterization of N-oxide-containing heterocycles with in vivo sterilizing antitubercular activity. J Med Chem 2017, 60, 8647–8660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Rogacki MK; Pitta E; Balabon O; Huss S; Lopez-Roman EM; Argyrou A; Blanco-Ruano D; Cacho M; Vande Velde CML; Augustyns K; Ballell L; Barros D; Bates RH; Cunningham F; Van der Veken P Identification and profiling of hydantoins-a novel class of potent antimycobacterial dpre1 inhibitors. J Med Chem 2018, 61, 11221–11249. [DOI] [PubMed] [Google Scholar]
  • 98.Zhang W; Lun S; Wang SH; Jiang XW; Yang F; Tang J; Manson AL; Earl AM; Gunosewoyo H; Bishai WR; Yu LF Identification of novel coumestan derivatives as polyketide synthase 13 inhibitors against mycobacterium tuberculosis. J Med Chem 2018, 61, 791–803. [DOI] [PubMed] [Google Scholar]
  • 99.Zhang W; Lun S; Liu LL; Xiao S; Duan G; Gunosewoyo H; Yang F; Tang J; Bishai WR; Yu LF Identification of novel coumestan derivatives as polyketide synthase 13 inhibitors against mycobacterium tuberculosis. Part ii. J Med Chem 2019, 62, 3575–3589. [DOI] [PubMed] [Google Scholar]
  • 100.Zhang D; Lu Y; Liu K; Liu B; Wang J; Zhang G; Zhang H; Liu Y; Wang B; Zheng M; Fu L; Hou Y; Gong N; Lv Y; Li C; Cooper CB; Upton AM; Yin D; Ma Z; Huang H Identification of less lipophilic riminophenazine derivatives for the treatment of drug-resistant tuberculosis. J Med Chem 2012, 55, 8409–8417. [DOI] [PubMed] [Google Scholar]
  • 101.Guardia A; Baiget J; Cacho M; Perez A; Ortega-Guerra M; Nxumalo W; Khanye SD; Rullas J; Ortega F; Jimenez E; Perez-Herran E; Fraile-Gabaldon MT; Esquivias J; Fernandez R; Porras-De Francisco E; Encinas L; Alonso M; Giordano I; Rivero C; Miguel-Siles J; Osende JG; Badiola KA; Rutledge PJ; Todd MH; Remuinan M; Alemparte C Easy-to-synthesize spirocyclic compounds possess remarkable in vivo activity against mycobacterium tuberculosis. J Med Chem 2018, 61, 11327–11340. [DOI] [PubMed] [Google Scholar]
  • 102.Taban IM; Elshihawy H; Torun B; Zucchini B; Williamson CJ; Altuwairigi D; Ngu AST; McLean KJ; Levy CW; Sood S; Marino LB; Munro AW; de Carvalho LPS; Simons C Novel aryl substituted pyrazoles as small molecule inhibitors of cytochrome P450 CYP121a1: Synthesis and antimycobacterial evaluation. J Med Chem 2017, 60, 10257–10267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Oh S; Park Y; Engelhart CA; Wallach JB; Schnappinger D; Arora K; Manikkam M; Gac B; Wang H; Murgolo N; Olsen DB; Goodwin M; Sutphin M; Weiner DM; Via LE; Boshoff HIM; Barry CE 3rd. Discovery and structure-activity-relationship study of n-alkyl-5-hydroxypyrimidinone carboxamides as novel antitubercular agents targeting decaprenylphosphoryl-beta-d-ribose 2’-oxidase. J Med Chem 2018, 61, 9952–9965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Vickers CF; Silva APG; Chakraborty A; Fernandez P; Kurepina N; Saville C; Naranjo Y; Pons M; Schnettger LS; Gutierrez MG; Park S; Kreiswith BN; Perlin DS; Thomas EJ; Cavet JS; Tabernero L Structure-based design of mptpb inhibitors that reduce multidrug-resistant mycobacterium tuberculosis survival and infection burden in vivo. J Med Chem 2018, 61, 8337–8352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Lagorce D; Sperandio O; Baell JB; Miteva MA; Villoutreix BO FAF-drugs3: A web server for compound property calculation and chemical library design. Nucleic Acids Res 2015, 43, W200–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Yang JJ; Ursu O; Lipinski CA; Sklar LA; Oprea TI; Bologa CG Badapple: Promiscuity patterns from noisy evidence. J Cheminform 2016, 8, 29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Deidda D; Lampis G; Fioravanti R; Biava M; Porretta GC; Zanetti S; Pompei R Bactericidal activities of the pyrrole derivative BM212 against multidrug-resistant and intramacrophagic mycobacterium tuberculosis strains. Antimicrob Agents Chemother 1998, 42, 3035–3037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.La Rosa V; Poce G; Canseco JO; Buroni S; Pasca MR; Biava M; Raju RM; Porretta GC; Alfonso S; Battilocchio C; Javid B; Sorrentino F; Ioerger TR; Sacchettini JC; Manetti F; Botta M; De Logu A; Rubin EJ; De Rossi E Mmpl3 is the cellular target of the antitubercular pyrrole derivative BM212. Antimicrob Agents Chemother 2012, 56, 324–331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Bhakta S; Scalacci N; Maitra A; Brown AK; Dasugari S; Evangelopoulos D; McHugh TD; Mortazavi PN; Twist A; Petricci E; Manetti F; Castagnolo D Design and synthesis of 1-((1,5-bis(4-chlorophenyl)-2-methyl-1h-pyrrol-3-yl)methyl)-4-methylpiperazine (bm212) and n-adamantan-2-yl-n’-((e)-3,7-dimethylocta-2,6-dienyl)ethane-1,2-diamine (SQ109) pyrrole hybrid derivatives: Discovery of potent antitubercular agents effective against multidrug-resistant mycobacteria. J Med Chem 2016, 59, 2780–2793. [DOI] [PubMed] [Google Scholar]
  • 110.Anon. Ll-3858. Tuberculosis (Edinb) 2008, 88, 126. [DOI] [PubMed] [Google Scholar]
  • 111.Cardona P-J. Understanding tuberculosis. New Approaches to Fighting Against Drug Resistance. IntechOpen, London, 2012. [Google Scholar]
  • 112.Stec J; Onajole OK; Lun S; Guo H; Merenbloom B; Vistoli G; Bishai WR; Kozikowski AP Indole-2-carboxamide-based MmpL3 inhibitors show exceptional antitubercular activity in an animal model of tuberculosis infection. J Med Chem 2016, 59, 6232–6247. [DOI] [PubMed] [Google Scholar]
  • 113.Tang J; Wang B; Wu T; Wan J; Tu Z; Njire M; Wan B; Franzblauc SG; Zhang T; Lu X; Ding K Design, synthesis, and biological evaluation of pyrazolo[1,5-a]pyridine-3-carboxamides as novel antitubercular agents. ACS Med Chem Lett 2015, 6, 814–818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Panda M; Ramachandran S; Ramachandran V; Shirude PS; Humnabadkar V; Nagalapur K; Sharma S; Kaur P; Guptha S; Narayan A; Mahadevaswamy J; Ambady A; Hegde N; Rudrapatna SS; Hosagrahara VP; Sambandamurthy VK; Raichurkar A Discovery of pyrazolopyridones as a novel class of noncovalent DprE1 inhibitor with potent anti-mycobacterial activity. J Med Chem 2014, 57, 4761–4771. [DOI] [PubMed] [Google Scholar]
  • 115.Riccardi G; Pasca MR; Chiarelli LR; Manina G; Mattevi A; Binda C The dpre1 enzyme, one of the most vulnerable targets of mycobacterium tuberculosis. Appl Microbiol Biotechnol 2013, 97, 8841–8848. [DOI] [PubMed] [Google Scholar]
  • 116.Brecik M; Centárová I; Mukherjee R; Kolly GS; Huszár S; Bobovská A; Kilacsková E; Mokošová V; Svetlíková Z; Šarkan M; Neres J; Korduláková J; Cole ST; Mikušová K DprE1 is a vulnerable tuberculosis drug target due to its cell wall localization. ACS Chem Biol 2015, 10, 1631–1636. [DOI] [PubMed] [Google Scholar]
  • 117.Wu Z; Lu Y; Li L; Zhao R; Wang B; Lv K; Liu M; You X Identification of n‐(2-phenoxyethyl)imidazo[1,2‐a]pyridine-3- carboxamides as new antituberculosis agents. ACS Med Chem Lett 2016, 7, 1130–1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Zhang Y; Post-Martens K; Denkin S New drug candidates and therapeutic targets for tuberculosis therapy. Drug Discov Today 2006, 11, 21–27. [DOI] [PubMed] [Google Scholar]
  • 119.Leach KL; Brickner SJ; Noe MC; Miller PF Linezolid, the first oxazolidinone antibacterial agent. Ann N Y Acad Sci 2011, 1222, 49–54. [DOI] [PubMed] [Google Scholar]
  • 120.Falagas ME; Siempos II; Vardakas KZ Linezolid versus glycopeptide or beta-lactam for treatment of gram-positive bacterial infections: Meta-analysis of randomised controlled trials. Lancet Infect Dis 2008, 8, 53–66. [DOI] [PubMed] [Google Scholar]
  • 121.Narita M; Tsuji BT; Yu VL Linezolid-associated peripheral and optic neuropathy, lactic acidosis, and serotonin syndrome. Pharmacotherapy 2007, 27, 1189–1197. [DOI] [PubMed] [Google Scholar]
  • 122.Bressler AM; Zimmer SM; Gilmore JL; Somani J Peripheral neuropathy associated with prolonged use of linezolid. Lancet Infect Dis 2004, 4, 528–531. [DOI] [PubMed] [Google Scholar]
  • 123.Chao CC; Sun HY; Chang YC; Hsieh ST Painful neuropathy with skin denervation after prolonged use of linezolid. J Neurol Neurosurg Psychiatry 2008, 79, 97–99. [DOI] [PubMed] [Google Scholar]
  • 124.Williams KN; Stover CK; Zhu T; Tasneen R; Tyagi S; Grosset JH; Nuermberger E Promising antituberculosis activity of the oxazolidinone PNU-100480 relative to that of linezolid in a murine model. Antimicrob Agents Chemother 2009, 53, 1314–1319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Alffenaar JW; van der Laan T; Simons S; van der Werf TS; van de Kasteele PJ; de Neeling H; van Soolingen D Susceptibility of clinical mycobacterium tuberculosis isolates to a potentially less toxic derivate of linezolid, PNU-100480. Antimicrob Agents Chemother 2011, 55, 1287–1289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Williams KN; Brickner SJ; Stover CK; Zhu T; Ogden A; Tasneen R; Tyagi S; Grosset JH; Nuermberger EL Addition of PNU-100480 to first-line drugs shortens the time needed to cure murine tuberculosis. Am J Respir Crit Care Med 2009, 180, 371–376. [DOI] [PubMed] [Google Scholar]
  • 127.Balasubramanian V; Solapure S; Iyer H; Ghosh A; Sharma S; Kaur P; Deepthi R; Subbulakshmi V; Ramya V; Ramachandran V; Balganesh M; Wright L; Melnick D; Butler SL; Sambandamurthy VK Bactericidal activity and mechanism of action of AZD5847, a novel oxazolidinone for treatment of tuberculosis. Antimicrob Agents Chemother 2014, 58, 495–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Jadhavar PS; Vaja MD; Dhameliya TM; Chakraborti AK Oxazolidinones as anti-tubercular agents: Discovery, development and future perspectives. Curr Med Chem 2015, 22, 4379–4397. [DOI] [PubMed] [Google Scholar]
  • 129.Furin JJ; Du Bois J; van Brakel E; Chheng P; Venter A; Peloquin CA; Alsultan A; Thiel BA; Debanne SM; Boom WH; Diacon AH; Johnson JL Early bactericidal activity of AZD5847 in patients with pulmonary tuberculosis. Antimicrob Agents Chemother 2016, 60, 6591–6599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Lee RE; Protopopova M; Crooks E; Slayden RA; Terrot M; Barry CE 3rd. Combinatorial lead optimization of [1,2]-diamines based on ethambutol as potential antituberculosis preclinical candidates. J Comb Chem 2003, 5, 172–187. [DOI] [PubMed] [Google Scholar]
  • 131.Protopopova M; Hanrahan C; Nikonenko B; Samala R; Chen P; Gearhart J; Einck L; Nacy CA Identification of a new antitubercular drug candidate, SQ109, from a combinatorial library of 1,2-ethylenediamines. J Antimicrob Chemother 2005, 56, 968–974. [DOI] [PubMed] [Google Scholar]
  • 132.Chen P; Gearhart J; Protopopova M; Einck L; Nacy CA Synergistic interactions of SQ109, a new ethylene diamine, with front-line antitubercular drugs in vitro. J Antimicrob Chemother 2006, 58, 332–337. [DOI] [PubMed] [Google Scholar]
  • 133.Jia L; Noker PE; Coward L; Gorman GS; Protopopova M; Tomaszewski JE Interspecies pharmacokinetics and in vitro metabolism of SQ109. Br J Pharmacol 2006, 147, 476–485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Tahlan K; Wilson R; Kastrinsky DB; Arora K; Nair V; Fischer E; Barnes SW; Walker JR; Alland D; Barry CE; Boshoff HI SQ109 targets mmpl3, a membrane transporter of trehalose monomycolate involved in mycolic acid donation to the cell wall core of mycobacterium tuberculosis. Antimicrob Agents Chemother 2012, 56, 1797–1809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Grzegorzewicz AE; Pham H; Gundi VA; Scherman MS; North EJ; Hess T; Jones V; Gruppo V; Born SE; Korduláková J; Chavadi SS; Morisseau C; Lenaerts AJ; Lee RE; McNeil MR; Jackson M Inhibition of mycolic acid transport across the mycobacterium tuberculosis plasma membrane. Nat Chem Biol 2012, 8, 334–341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Nikonenko BV; Protopopova M; Samala R; Einck L; Nacy CA Drug therapy of experimental tuberculosis (tb): Improved outcome by combining SQ109, a new diamine antibiotic, with existing tb drugs. Antimicrob Agents Chemother 2007, 51, 1563–1565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Li K; Wang Y; Yang G; Byun S; Rao G; Shoen C; Yang H; Gulati A; Crick DC; Cynamon M; Huang G; Docampo R; No JH; Oldfield E Oxa, thia, heterocycle, and carborane analogues of SQ109: Bacterial and protozoal cell growth inhibitors. ACS Infect Dis 2015, 1, 215–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Bogatcheva E; Hanrahan C; Chen P; Gearhart J; Sacksteder K; Einck L; Nacy C; Protopopova M Discovery of dipiperidines as new antitubercular agents. Bioorg Med Chem Lett 2010, 20, 201–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Bogatcheva E; Hanrahan C; Nikonenko B; de los Santos G; Reddy V; Chen P; Barbosa F; Einck L; Nacy C; Protopopova M Identification of SQ609 as a lead compound from a library of dipiperidines. Bioorg Med Chem Lett 2011, 21, 5353–5357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Bermudez LE; Reynolds R; Kolonoski P; Aralar P; Inderlied CB; Young LS Thiosemicarbazole (thiacetazone-like) compound with activity against mycobacterium avium in mice. Antimicrob Agents Chemother 2003, 47, 2685–2687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Alahari A; Trivelli X; Guérardel Y; Dover LG; Besra GS; Sacchettini JC; Reynolds RC; Coxon GD; Kremer L Thiacetazone, an antitubercular drug that inhibits cyclopropanation of cell wall mycolic acids in mycobacteria. PLoS One 2007, 2, e1343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Naidu KM; Srinivasarao S; Agnieszka N; Ewa AK; Kumar MM; Chandra Sekhar KV Seeking potent anti-tubercular agents: Design, synthesis, anti-tubercular activity and docking study of various ((triazoles/indole)-piperazin-1-yl/1,4-diazepan-1-yl)benzo[d]isoxazole derivatives. Bioorg Med Chem Lett 2016, 26, 2245–2250. [DOI] [PubMed] [Google Scholar]
  • 143.Zheng R; Blanchard JS Steady-state and pre-steady-state kinetic analysis of mycobacterium tuberculosis pantothenate synthetase. Biochemistry 2001, 40, 12904–12912. [DOI] [PubMed] [Google Scholar]
  • 144.Karabanovich G; Zemanová J; Smutný T; Székely R; Šarkan M; Centárová I; Vocat A; Pávková I; Čonka P; Němeček J; Stolaříková J; Vejsová M; Vávrová K; Klimešová V; Hrabálek A; Pávek P; Cole ST; Mikušová K; Roh J Development of 3,5-dinitrobenzylsulfanyl-1,3,4-oxadiazoles and thiadiazoles as selective antitubercular agents active against replicating and nonreplicating mycobacterium tuberculosis. J Med Chem 2016, 59, 2362–2380. [DOI] [PubMed] [Google Scholar]
  • 145.Carette X; Blondiaux N; Willery E; Hoos S; Lecat-Guillet N; Lens Z; Wohlkönig A; Wintjens R; Soror SH; Frénois F; Dirié B; Villeret V; England P; Lippens G; Deprez B; Locht C; Willand N; Baulard AR Structural activation of the transcriptional repressor EthR from mycobacterium tuberculosis by single amino acid change mimicking natural and synthetic ligands. Nucleic Acids Res 2012, 40, 3018–3030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Willand N; Dirie B; Carette X; Bifani P; Singhal A; Desroses M; Leroux F; Willery E; Mathys V; Deprez-Poulain R; Delcroix G; Frenois F; Aumercier M; Locht C; Villeret V; Deprez B; Baulard AR Synthetic EthR inhibitors boost antituberculous activity of ethionamide. Nat Med 2009, 15, 537–544. [DOI] [PubMed] [Google Scholar]
  • 147.Karabanovich G; Němeček J; Valášková L; Carazo A; Konečná K; Stolaříková J; Hrabálek A; Pavliš O; Pávek P; Vávrová K; Roh J; Klimešová V S-substituted 3,5-dinitrophenyl 1,3,4-oxadiazole-2-thiols and tetrazole-5-thiols as highly efficient antitubercular agents. Eur J Med Chem 2017, 126, 369–383. [DOI] [PubMed] [Google Scholar]
  • 148.Bellale E; Naik M; V B V; Ambady A; Narayan A; Ravishankar S; Ramachandran V; Kaur P; McLaughlin R; Whiteaker J; Morayya S; Guptha S; Sharma S; Raichurkar A; Awasthy D; Achar V; Vachaspati P; Bandodkar B; Panda M; Chatterji M Diarylthiazole: An antimycobacterial scaffold potentially targeting PrrB-PrrA two-component system. J Med Chem 2014, 57, 6572–6582. [DOI] [PubMed] [Google Scholar]
  • 149.Haydel SE; Malhotra V; Cornelison GL; Clark-Curtiss JE The prrab two-component system is essential for mycobacterium tuberculosis viability and is induced under nitrogen-limiting conditions. J Bacteriol 2012, 194, 354–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Ewann F; Jackson M; Pethe K; Cooper A; Mielcarek N; Ensergueix D; Gicquel B; Locht C; Supply P Transient requirement of the prra-prrb two-component system for early intracellular multiplication of mycobacterium tuberculosis. Infect Immun 2002, 70, 2256–2263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Pieroni M; Wan B; Cho S; Franzblau SG; Costantino G Design, synthesis and investigation on the structure-activity relationships of n-substituted 2-aminothiazole derivatives as antitubercular agents. Eur J Med Chem 2014, 72, 26–34. [DOI] [PubMed] [Google Scholar]
  • 152.Landge S; Mullick AB; Nagalapur K; Neres J; Subbulakshmi V; Murugan K; Ghosh A; Sadler C; Fellows MD; Humnabadkar V; Mahadevaswamy J; Vachaspati P; Sharma S; Kaur P; Mallya M; Rudrapatna S; Awasthy D; Sambandamurthy VK; Pojer F; Cole ST; Balganesh TS; Ugarkar BG; Balasubramanian V; Bandodkar BS; Panda M; Ramachandran V Discovery of benzothiazoles as antimycobacterial agents: Synthesis, structure-activity relationships and binding studies with mycobacterium tuberculosis decaprenylphosphoryl-β-d-ribose 2’-oxidase. Bioorg Med Chem 2015, 23, 7694–7710. [DOI] [PubMed] [Google Scholar]
  • 153.Mahajan NS; Dhawale SC Linked pyridinyl-thiadiazoles: Design and synthesis as potential candidate for treatment of xdr and mdr tuberculosis. Eur J Med Chem 2015, 102, 243–248. [DOI] [PubMed] [Google Scholar]
  • 154.Sharma V; Sharma S; Hoener zu Bentrup K; McKinney JD; Russell DG; Jacobs WR; Sacchettini JC Structure of isocitrate lyase, a persistence factor of mycobacterium tuberculosis. Nat Struct Biol 2000, 7, 663–668. [DOI] [PubMed] [Google Scholar]
  • 155.Reddy VM; O’Sullivan JF; Gangadharam PR Antimycobacterial activities of riminophenazines. J Antimicrob Chemother 1999, 43, 615–623. [DOI] [PubMed] [Google Scholar]
  • 156.Arbiser JL; Moschella SL Clofazimine: A review of its medical uses and mechanisms of action. J Am Acad Dermatol 1995, 32, 241–247. [DOI] [PubMed] [Google Scholar]
  • 157.Morrison NE; Marley GM The mode of action of clofazimine DNA binding studies. Int J Lepr Other Mycobact Dis 1976, 44, 133–134. [PubMed] [Google Scholar]
  • 158.Nair V; Okello MO; Mangu NK; Seo BI; Gund MG A novel molecule with notable activity against multi-drug resistant tuberculosis. Bioorg Med Chem Lett 2015, 25, 1269–1273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.van der Westhuyzen R; Winks S; Wilson CR; Boyle GA; Gessner RK; Soares de Melo C; Taylor D; de Kock C; Njoroge M; Brunschwig C; Lawrence N; Rao SP; Sirgel F; van Helden P; Seldon R; Moosa A; Warner DF; Arista L; Manjunatha UH; Smith PW; Street LJ; Chibale K Pyrrolo[3,4-c]pyridine-1,3(2h)-diones: A novel antimycobacterial class targeting mycobacterial respiration. J Med Chem 2015, 58, 9371–9381. [DOI] [PubMed] [Google Scholar]
  • 160.Pissinate K; Villela AD; Rodrigues-Junior V; Giacobbo BC; Grams ES; Abbadi BL; Trindade RV; Roesler Nery L; Bonan CD; Back DF; Campos MM; Basso LA; Santos DS; Machado P 2-(quinolin-4-yloxy)acetamides are active against drug-susceptible and drug-resistant mycobacterium tuberculosis strains. ACS Med Chem Lett 2016, 7, 235–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Giacobbo BC; Pissinate K; Rodrigues-Junior V; Villela AD; Grams ES; Abbadi BL; Subtil FT; Sperotto N; Trindade RV; Back DF; Campos MM; Basso LA; Machado P; Santos DS New insights into the sar and drug combination synergy of 2-(quinolin-4-yloxy)acetamides against mycobacterium tuberculosis. Eur J Med Chem 2017, 126, 491–501. [DOI] [PubMed] [Google Scholar]
  • 162.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. Medchemcomm 2016, 7, 2122–2127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Naik M; Humnabadkar V; Tantry SJ; Panda M; Narayan A; Guptha S; Panduga V; Manjrekar P; Jena LK; Koushik K; Shanbhag G; Jatheendranath S; Manjunatha MR; Gorai G; Bathula C; Rudrapatna S; Achar V; Sharma S; Ambady A; Hegde N; Mahadevaswamy J; Kaur P; Sambandamurthy VK; Awasthy D; Narayan C; Ravishankar S; Madhavapeddi P; Reddy J; Prabhakar K; Saralaya R; Chatterji M; Whiteaker J; McLaughlin B; Chiarelli LR; Riccardi G; Pasca MR; Binda C; Neres J; Dhar N; Signorino-Gelo F; McKinney JD; Ramachandran V; Shandil R; Tommasi R; Iyer PS; Narayanan S; Hosagrahara V; Kavanagh S; Dinesh N; Ghorpade SR 4-aminoquinolone piperidine amides: Noncovalent inhibitors of DprE1 with long residence time and potent antimycobacterial activity. J Med Chem 2014, 57, 5419–5434. [DOI] [PubMed] [Google Scholar]
  • 164.Read ML; Braendvang M; Miranda PO; Gundersen LL Synthesis and biological evaluation of pyrimidine analogs of antimycobacterial purines. Bioorg Med Chem 2010, 18, 3885–3897. [DOI] [PubMed] [Google Scholar]
  • 165.Anand N; Singh P; Sharma A; Tiwari S; Singh V; Singh DK; Srivastava KK; Singh BN; Tripathi RP Synthesis and evaluation of small libraries of triazolylmethoxy chalcones, flavanones and 2-aminopyrimidines as inhibitors of mycobacterial FAS-II and PknG. Bioorg Med Chem 2012, 20, 5150–5163. [DOI] [PubMed] [Google Scholar]
  • 166.Shirude PS; Paul B; Roy Choudhury N; Kedari C; Bandodkar B; Ugarkar BG Quinolinyl pyrimidines: Potent inhibitors of NDH-2 as a novel class of anti-TB agents. ACS Med Chem Lett 2012, 3, 736–740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Yano T; Rahimian M; Aneja KK; Schechter NM; Rubin H; Scott CP Mycobacterium tuberculosis type ii nadh-menaquinone oxidoreductase catalyzes electron transfer through a two-site ping-pong mechanism and has two quinone-binding sites. Biochemistry 2014, 53, 1179–1190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Kravchenko MA; Verbitskiy EV; Medvinskiy ID; Rusinov GL; Charushin VN Synthesis and antituberculosis activity of novel 5-styryl-4-(hetero)aryl-pyrimidines via combination of the pd-catalyzed suzuki cross-coupling and s(n)(h) reactions. Bioorg Med Chem Lett 2014, 24, 3118–3120. [DOI] [PubMed] [Google Scholar]
  • 169.Verbitskiy EV; Baskakova SA; Kravchenko MA; Skornyakov SN; Rusinov GL; Chupakhin ON; Charushin VN Synthesis and evaluation of antitubercular activity of fluorinated 5-aryl-4-(hetero)aryl substituted pyrimidines. Bioorg Med Chem 2016, 24, 3771–3780. [DOI] [PubMed] [Google Scholar]
  • 170.Verbitskiy EV; Cheprakova EM; Slepukhin PA; Kravchenko MA; Skornyakov SN; Rusinov GL; Chupakhin ON; Charushin VN Synthesis, and structure-activity relationship for c(4) and/or c(5) thienyl substituted pyrimidines, as a new family of antimycobacterial compounds. Eur J Med Chem 2015, 97, 225–234. [DOI] [PubMed] [Google Scholar]
  • 171.Yadlapalli RK; Chourasia OP; Vemuri K; Sritharan M; Perali RS Synthesis and in vitro anticancer and antitubercular activity of diarylpyrazole ligated dihydropyrimidines possessing lipophilic carbamoyl group. Bioorg Med Chem Lett 2012, 22, 2708–2711. [DOI] [PubMed] [Google Scholar]
  • 172.Yokokawa F; Wang G; Chan WL; Ang SH; Wong J; Ma I; Rao SP; Manjunatha U; Lakshminarayana SB; Herve M; Kounde C; Tan BH; Thayalan P; Ng SH; Nanjundappa M; Ravindran S; Gee P; Tan M; Wei L; Goh A; Chen PY; Lee KS; Zhong C; Wagner T; Dix I; Chatterjee AK; Pethe K; Kuhen K; Glynne R; Smith P; Bifani P; Jiricek J Discovery of tetrahydropyrazolopyrimidine carboxamide derivatives as potent and orally active antitubercular agents. ACS Med Chem Lett 2013, 4, 451–455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Shirude PS; Shandil R; Sadler C; Naik M; Hosagrahara V; Hameed S; Shinde V; Bathula C; Humnabadkar V; Kumar N; Reddy J; Panduga V; Sharma S; Ambady A; Hegde N; Whiteaker J; McLaughlin RE; Gardner H; Madhavapeddi P; Ramachandran V; Kaur P; Narayan A; Guptha S; Awasthy D; Narayan C; Mahadevaswamy J; Vishwas KG; Ahuja V; Srivastava A; Prabhakar KR; Bharath S; Kale R; Ramaiah M; Choudhury NR; Sambandamurthy VK; Solapure S; Iyer PS; Narayanan S; Chatterji M Azaindoles: Noncovalent dpre1 inhibitors from scaffold morphing efforts, kill mycobacterium tuberculosis and are efficacious in vivo. J Med Chem 2013, 56, 9701–9708. [DOI] [PubMed] [Google Scholar]
  • 174.Velezheva V; Brennan P; Ivanov P; Kornienko A; Lyubimov S; Kazarian K; Nikonenko B; Majorov K; Apt A Synthesis and antituberculosis activity of indole-pyridine derived hydrazides, hydrazide-hydrazones, and thiosemicarbazones. Bioorg Med Chem Lett 2016, 26, 978–985. [DOI] [PubMed] [Google Scholar]
  • 175.Lu W; Baig IA; Sun HJ; Cui CJ; Guo R; Jung IP; Wang D; Dong M; Yoon MY; Wang JG Synthesis, crystal structure and biological evaluation of substituted quinazolinone benzoates as novel antituberculosis agents targeting acetohydroxyacid synthase. Eur J Med Chem 2015, 94, 298–305. [DOI] [PubMed] [Google Scholar]
  • 176.Ranjith Kumar R; Perumal S; Menéndez JC; Yogeeswari P; Sriram D Antimycobacterial activity of novel 1,2,4-oxadiazole-pyranopyridine/chromene hybrids generated by chemoselective 1,3-dipolar cycloadditions of nitrile oxides. Bioorg Med Chem 2011, 19, 3444–3450. [DOI] [PubMed] [Google Scholar]
  • 177.Almansour AI; Suresh Kumar R; Arumugam N; Sriram D A solvent free, four-component synthesis and 1,3-dipolar cycloaddition of 4(h)-pyrans with nitrile oxides: Synthesis and discovery of antimycobacterial activity of enantiomerically pure 1,2,4-oxadiazoles. Eur J Med Chem 2012, 53, 416–423. [DOI] [PubMed] [Google Scholar]
  • 178.Sriram D; Yogeeswari P; Dinakaran M; Banerjee D; Bhat P; Gadhwal S Discovery of novel antitubercular 2,10-dihydro-4ah-chromeno[3,2-c]pyridin-3-yl derivatives. Eur J Med Chem 2010, 45, 120–123. [DOI] [PubMed] [Google Scholar]
  • 179.Jeyachandran M; Ramesh P; Sriram D; Senthilkumar P; Yogeeswari P Synthesis and in vitro antitubercular activity of 4-aryl/alkylsulfonylmethylcoumarins as inhibitors of mycobacterium tuberculosis. Bioorg Med Chem Lett 2012, 22, 4807–4809. [DOI] [PubMed] [Google Scholar]
  • 180.Carta A; Palomba M; Briguglio I; Corona P; Piras S; Jabes D; Guglierame P; Molicotti P; Zanetti S Synthesis and anti-mycobacterial activities of triazoloquinolones. Eur J Med Chem 2011, 46, 320–326. [DOI] [PubMed] [Google Scholar]
  • 181.Makarov V; Neres J; Hartkoorn RC; Ryabova OB; Kazakova E; Sarkan M; Huszar S; Piton J; Kolly GS; Vocat A; Conroy TM; Mikusova K; Cole ST The 8-pyrrole-benzothiazinones are noncovalent inhibitors of DprE1 from mycobacterium tuberculosis. Antimicrob Agents Chemother 2015, 59, 4446–4452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Lechartier B; Hartkoorn RC; Cole ST In vitro combination studies of benzothiazinone lead compound BTZ043 against mycobacterium tuberculosis. Antimicrob Agents Chemother 2012, 56, 5790–5793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Karoli T; Becker B; Zuegg J; Möllmann U; Ramu S; Huang JX; Cooper MA Identification of antitubercular benzothiazinone compounds by ligand-based design. J Med Chem 2012, 55, 7940–7944. [DOI] [PubMed] [Google Scholar]
  • 184.Anon. Macozinone (MCZ, PBTZ-169). https://www.newtbdrugs.org/pipeline/compound/macozinone-mcz-pbtz-169 (accessed Mar 3, 2020).
  • 185.Almansour AI; Ali S; Ali MA; Ismail R; Choon TS; Sellappan V; Elumalai KK; Pandian S A regio- and stereoselective 1,3-dipolar cycloaddition for the synthesis of new-fangled dispiropyrrolothiazoles as antimycobacterial agents. Bioorg Med Chem Lett 2012, 22, 7418–7421. [DOI] [PubMed] [Google Scholar]
  • 186.Karthikeyan SV; Bala BD; Raja VP; Perumal S; Yogeeswari P; Sriram D A highly atom economic, chemo-, regio- and stereoselective synthesis and evaluation of spiro-pyrrolothiazoles as antitubercular agents. Bioorg Med Chem Lett 2010, 20, 350–353. [DOI] [PubMed] [Google Scholar]
  • 187.Liu YX; Xiao CL; Wang YX; Li YH; Yang YH; Li YB; Bi CW; Gao LM; Jiang JD; Song DQ Synthesis, structure-activity relationship and in vitro anti-mycobacterial evaluation of 13-n-octylberberine derivatives. Eur J Med Chem 2012, 52, 151–158. [DOI] [PubMed] [Google Scholar]
  • 188.Keffer JL; Huecas S; Hammill JT; Wipf P; Andreu JM; Bewley CA Chrysophaentins are competitive inhibitors of FtsZ and inhibit z-ring formation in live bacteria. Bioorg Med Chem 2013, 21, 5673–5678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Hotoda H; Furukawa M; Daigo M; Murayama K; Kaneko M; Muramatsu Y; Ishii MM; Miyakoshi S; Takatsu T; Inukai M; Kakuta M; Abe T; Harasaki T; Fukuoka T; Utsui Y; Ohya S Synthesis and antimycobacterial activity of capuramycin analogues. Part 1: Substitution of the azepan-2-one moiety of capuramycin. Bioorg Med Chem Lett 2003, 13, 2829–2832. [DOI] [PubMed] [Google Scholar]
  • 190.Kimura K; Bugg TD Recent advances in antimicrobial nucleoside antibiotics targeting cell wall biosynthesis. Nat Prod Rep 2003, 20, 252–273. [DOI] [PubMed] [Google Scholar]
  • 191.Bugg TD; Lloyd AJ; Roper DI Phospho-MurNAc-pentapeptide translocase (MraY) as a target for antibacterial agents and antibacterial proteins. Infect Disord Drug Targets 2006, 6, 85–106. [DOI] [PubMed] [Google Scholar]
  • 192.Nikonenko BV; Reddy VM; Protopopova M; Bogatcheva E; Einck L; Nacy CA Activity of SQ641, a capuramycin analog, in a murine model of tuberculosis. Antimicrob Agents Chemother 2009, 53, 3138–3139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Koga T; Fukuoka T; Doi N; Harasaki T; Inoue H; Hotoda H; Kakuta M; Muramatsu Y; Yamamura N; Hoshi M; Hirota T Activity of capuramycin analogues against mycobacterium tuberculosis, mycobacterium avium and mycobacterium intracellulare in vitro and in vivo. J Antimicrob Chemother 2004, 54, 755–760. [DOI] [PubMed] [Google Scholar]
  • 194.Nakamura H; Yoshida T; Tsukano C; Takemoto Y Synthesis of cpzen-45: Construction of the 1,4-diazepin-2-one core by the cu-catalyzed intramolecular amidation of a vinyl iodide. Org Lett 2016, 18, 2300–2303. [DOI] [PubMed] [Google Scholar]
  • 195.Igarashi M; Takahashi Y; Shitara T; Nakamura H; Naganawa H; Miyake T; Akamatsu Y Caprazamycins, novel lipo-nucleoside antibiotics, from streptomyces sp. Ii. Structure elucidation of caprazamycins. J Antibiot (Tokyo) 2005, 58, 327–337. [DOI] [PubMed] [Google Scholar]
  • 196.Igarashi M; Nakagawa N; Doi N; Hattori S; Naganawa H; Hamada M Caprazamycin b, a novel anti-tuberculosis antibiotic, from streptomyces sp. J Antibiot (Tokyo) 2003, 56, 580–583. [DOI] [PubMed] [Google Scholar]
  • 197.Ishizaki Y; Hayashi C; Inoue K; Igarashi M; Takahashi Y; Pujari V; Crick DC; Brennan PJ; Nomoto A Inhibition of the first step in synthesis of the mycobacterial cell wall core, catalyzed by the glcnac-1-phosphate transferase weca, by the novel caprazamycin derivative cpzen-45. J Biol Chem 2013, 288, 30309–30319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Horlacher OP; Hartkoorn RC; Cole ST; Altmann KH Synthesis and antimycobacterial activity of 2,1’-dihydropyridomycins. ACS Med Chem Lett 2013, 4, 264–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Hartkoorn RC; Pojer F; Read JA; Gingell H; Neres J; Horlacher OP; Altmann KH; Cole ST Pyridomycin bridges the NADH- and substrate-binding pockets of the enoyl reductase InhA. Nat Chem Biol 2014, 10, 96–98. [DOI] [PubMed] [Google Scholar]
  • 200.Dong M; Pfeiffer B; Altmann KH Recent developments in natural product-based drug discovery for tuberculosis. Drug Discov Today 2016, 22, 585–591. [DOI] [PubMed] [Google Scholar]
  • 201.Liu J; Bruhn DF; Lee RB; Zheng Z; Janusic T; Scherbakov D; Scherman MS; Boshoff HI; Das S; Rakesh; Waidyarachchi SL; Brewer TA; Gracia B; Yang L; Bollinger J; Robertson GT; Meibohm B; Lenaerts AJ; Ainsa J; Bottger EC; Lee RE Structure-activity relationships of spectinamide antituberculosis agents: A dissection of ribosomal inhibition and native efflux avoidance contributions. ACS Infect Dis 2017, 3, 72–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Vaddady PK; Trivedi A; Rathi C; Madhura DB; Liu J; Lee RE; Meibohm B Dynamic time-kill curve characterization of spectinamide antibiotics 1445 and 1599 for the treatment of tuberculosis. Eur J Pharm Sci 2019, 127, 233–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Anon. Spectinamide 1810. http://www.newtbdrugs.org/pipeline/compound/spectinamide-1810 (accessed Mar 3, 2020).
  • 204.Tripathi RP; Tripathi R; Tiwari VK; Bala L; Sinha S; Srivastava A; Srivastava R; Srivastava BS Synthesis of glycosylated beta-amino acids as new class of antitubercular agents. Eur J Med Chem 2002, 37, 773–781. [DOI] [PubMed] [Google Scholar]
  • 205.Mugunthan G; Ramakrishna K; Sriram D; Yogeeswari P; Ravindranathan Kartha KP Synthesis and screening of galactose-linked nitroimidazoles and triazoles against mycobacterium tuberculosis. Eur J Med Chem 2011, 46, 4725–4732. [DOI] [PubMed] [Google Scholar]
  • 206.Rane RA; Gutte SD; Sahu NU Synthesis and evaluation of novel 1,3,4-oxadiazole derivatives of marine bromopyrrole alkaloids as antimicrobial agent. Bioorg Med Chem Lett 2012, 22, 6429–6432. [DOI] [PubMed] [Google Scholar]
  • 207.Tangallapally RP; Sun D; Rakesh; Budha N; Lee RE; Lenaerts AJ; Meibohm B; Lee RE Discovery of novel isoxazolines as anti-tuberculosis agents. Bioorg Med Chem Lett 2007, 17, 6638–6642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Li D; Gao N; Zhu N; Lin Y; Li Y; Chen M; You X; Lu Y; Wan K; Jiang JD; Jiang W; Si S Discovery of the disubstituted oxazole analogues as a novel class anti-tuberculotic agents against mdr- and xdr-mtb. Bioorg Med Chem Lett 2015, 25, 5178–5181. [DOI] [PubMed] [Google Scholar]
  • 209.Pedgaonkar GS; Sridevi JP; Jeankumar VU; Saxena S; Devi PB; Renuka J; Yogeeswari P; Sriram D Development of benzo[d]oxazol-2(3h)-ones derivatives as novel inhibitors of mycobacterium tuberculosis inha. Bioorg Med Chem 2014, 22, 6134–6145. [DOI] [PubMed] [Google Scholar]
  • 210.Costa MS; Boechat N; Rangel EA; da Silva F. e. C.; de Souza AM; Rodrigues CR; Castro HC; Junior IN; Lourenço MC; Wardell SM; Ferreira VF Synthesis, tuberculosis inhibitory activity, and sar study of n-substituted-phenyl-1,2,3-triazole derivatives. Bioorg Med Chem 2006, 14, 8644–8653. [DOI] [PubMed] [Google Scholar]
  • 211.Menendez C; Chollet A; Rodriguez F; Inard C; Pasca MR; Lherbet C; Baltas M Chemical synthesis and biological evaluation of triazole derivatives as inhibitors of InhA and antituberculosis agents. Eur J Med Chem 2012, 52, 275–283. [DOI] [PubMed] [Google Scholar]
  • 212.Thomas KD; Adhikari AV; Chowdhury IH; Sumesh E; Pal NK New quinolin-4-yl-1,2,3-triazoles carrying amides, sulphonamides and amidopiperazines as potential antitubercular agents. Eur J Med Chem 2011, 46, 2503–2512. [DOI] [PubMed] [Google Scholar]
  • 213.Menendez C; Rodriguez F; Ribeiro AL; Zara F; Frongia C; Lobjois V; Saffon N; Pasca MR; Lherbet C; Baltas M Synthesis and evaluation of α-ketotriazoles and α,β-diketotriazoles as inhibitors of mycobacterium tuberculosis. Eur J Med Chem 2013, 69, 167–173. [DOI] [PubMed] [Google Scholar]
  • 214.Kumar K; Awasthi D; Lee SY; Zanardi I; Ruzsicska B; Knudson S; Tonge PJ; Slayden RA; Ojima I Novel trisubstituted benzimidazoles, targeting Mtb FtsZ, as a new class of antitubercular agents. J Med Chem 2011, 54, 374–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Haranahalli K; Tong S; Ojima I Recent advances in the discovery and development of antibacterial agents targeting the cell-division protein ftsz. Bioorg Med Chem 2016, 24, 6354–6369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Knudson SE; Awasthi D; Kumar K; Carreau A; Goullieux L; Lagrange S; Vermet H; Ojima I; Slayden RA Cell division inhibitors with efficacy equivalent to isoniazid in the acute murine mycobacterium tuberculosis infection model. J Antimicrob Chemother 2015, 70, 3070–3073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Ahamad S; Islam A; Ahmad F; Dwivedi N; Hassan MI 2/3D-QSAR, molecular docking and md simulation studies of ftsz protein targeting benzimidazoles derivatives. Comput Biol Chem 2019, 78, 398–413. [DOI] [PubMed] [Google Scholar]
  • 218.Gobis K; Foks H; Bojanowski K; Augustynowicz-Kopeć E; Napiórkowska A Synthesis of novel 3-cyclohexylpropanoic acid-derived nitrogen heterocyclic compounds and their evaluation for tuberculostatic activity. Bioorg Med Chem 2012, 20, 137–144. [DOI] [PubMed] [Google Scholar]
  • 219.Abrahams KA; Chung CW; Ghidelli-Disse S; Rullas J; Rebollo-Lopez MJ; Gurcha SS; Cox JA; Mendoza A; Jimenez-Navarro E; Martinez-Martinez MS; Neu M; Shillings A; Homes P; Argyrou A; Casanueva R; Loman NJ; Moynihan PJ; Lelievre J; Selenski C; Axtman M; Kremer L; Bantscheff M; Angulo-Barturen I; Izquierdo MC; Cammack NC; Drewes G; Ballell L; Barros D; Besra GS; Bates RH Identification of KasA as the cellular target of an anti-tubercular scaffold. Nat Commun 2016, 7, 12581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Kumar P; Capodagli GC; Awasthi D; Shrestha R; Maharaja K; Sukheja P; Li SG; Inoyama D; Zimmerman M; Ho Liang HP; Sarathy J; Mina M; Rasic G; Russo R; Perryman AL; Richmann T; Gupta A; Singleton E; Verma S; Husain S; Soteropoulos P; Wang Z; Morris R; Porter G; Agnihotri G; Salgame P; Ekins S; Rhee KY; Connell N; Dartois V; Neiditch MB; Freundlich JS; Alland D Synergistic lethality of a binary inhibitor of mycobacterium tuberculosis KasA. MBio 2018, 9, e02101–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.De P; Koumba Yoya G; Constant P; Bedos-Belval F; Duran H; Saffon N; Daffé M; Baltas M Design, synthesis, and biological evaluation of new cinnamic derivatives as antituberculosis agents. J Med Chem 2011, 54, 1449–1461. [DOI] [PubMed] [Google Scholar]
  • 222.Tawari NR; Bairwa R; Ray MK; Rajan MG; Degani MS Design, synthesis, and biological evaluation of 4-(5-nitrofuran-2-yl)prop-2-en-1-one derivatives as potent antitubercular agents. Bioorg Med Chem Lett 2010, 20, 6175–6178. [DOI] [PubMed] [Google Scholar]
  • 223.Limaye RA; Kumbhar VB; Natu AD; Paradkar MV; Honmore VS; Chauhan RR; Gample SP; Sarkar D One pot solvent free synthesis and in vitro antitubercular screening of 3-aracylphthalides against mycobacterium tuberculosis. Bioorg Med Chem Lett 2013, 23, 711–714. [DOI] [PubMed] [Google Scholar]
  • 224.Anon. GSK 070, GSK 3036656. https://www.newtbdrugs.org/pipeline/compound/gsk-070-gsk-3036656 (accessed Mar 3, 2020).
  • 225.Ekins S; Casey AC; Roberts D; Parish T; Bunin BA Bayesian models for screening and TB mobile for target inference with mycobacterium tuberculosis. Tuberculosis (Edinb) 2014, 94, 162–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Ekins S; Pottorf R; Reynolds RC; Williams AJ; Clark AM; Freundlich JS Looking back to the future: Predicting in vivo efficacy of small molecules versus mycobacterium tuberculosis. J Chem Inf Model 2014, 54, 1070–1082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Ekins S; Kaneko T; Lipinksi CA; Bradford J; Dole K; Spektor A; Gregory K; Blondeau D; Ernst S; Yang J; Goncharoff N; Hohman M; Bunin B Analysis and hit filtering of a very large library of compounds screened against mycobacterium tuberculosis Mol BioSyst 2010, 6, 2316–2324. [DOI] [PubMed] [Google Scholar]
  • 228.Landrum G RDkit Documentation Release 2017.09.1 10.5281/zenodo.60510 (accessed Mar 3, 2020). [DOI]
  • 229.McKinney W; Team PD Pandas - Powerful Python Data Analysis Toolkit. https://pandas.pydata.org/pandas-docs/stable/ (accessed March 3, 2020).
  • 230.Community N Numpy Reference. 2016, 1, 1–1146. [Google Scholar]
  • 231.DataCamp. Jupyter Notebook. 10.1023/a:1009769707641 (accessed Mar 3, 2020). [DOI]
  • 232.Anon. Scikit-learn User Guide. https://scikit-learn.org/stable/_downloads/scikit-learn-docs.pdf (accessed Mar 3, 2020).
  • 233.Lane T; Russo DP; Zorn KM; Clark AM; Korotcov A; Tkachenko V; Reynolds RC; Perryman AL; Freundlich JS; Ekins S Comparing and validating machine learning models for mycobacterium tuberculosis drug discovery. Mol Pharm 2018, 15, 4346–4360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Hunter J; Dale D; Firing E; Droettboom M Matplotlib release 2.2.2. Release June 28, 2018.
  • 235.Prathipati P; Ma NL; Keller TH Global bayesian models for the prioritization of antitubercular agents. J Chem Inf Model 2008, 48, 2362–2370. [DOI] [PubMed] [Google Scholar]
  • 236.Ekins S; Bradford J; Dole K; Spektor A; Gregory K; Blondeau D; Hohman M; Bunin B A collaborative database and computational models for tuberculosis drug discovery. Mol BioSystems 2010, 6, 840–851. [DOI] [PubMed] [Google Scholar]
  • 237.Gamo F-J; Sanz LM; Vidal J; de Cozar C; Alvarez E; Lavandera J-L; Vanderwall DE; Green DVS; Kumar V; Hasan S; Brown JR; Peishoff CE; Cardon LR; Garcia-Bustos JF Thousands of chemical starting points for antimalarial lead identification. Nature 2010, 465, 305–310. [DOI] [PubMed] [Google Scholar]
  • 238.Ekins S; Reynolds R; Kim H; Koo M-S; Ekonomidis M; Talaue M; Paget SD; Woolhiser LK; Lenaerts AJ; Bunin BA; Connell N; Freundlich JS Bayesian models leveraging bioactivity and cytotoxicity information for drug discovery. Chem Biol 2013, 20, 370–378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Ekins S; Reynolds RC; Franzblau SG; Wan B; Freundlich JS; Bunin BA Enhancing hit identification in mycobacterium tuberculosis drug discovery using validated dual-event bayesian models PLOSONE 2013, 8, e63240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Ekins S; Casey AC; Roberts D; Parish T; Bunin BA Bayesian models for screening and tb mobile for target inference with mycobacterium tuberculosis Tuberculosis (Edinb) 2014, 94, 162–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Ekins S; Freundlich JS; Reynolds RC Fusing dual-event datasets for mycobacterium tuberculosis machine learning models and their evaluation. J Chem Inf Model 2013, 53, 3054–3063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Ekins S; Freundlich JS; Hobrath JV; Lucile White E; Reynolds RC Combining computational methods for hit to lead optimization in mycobacterium tuberculosis drug discovery. Pharm Res 2014, 31, 414–435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Ekins S; Freundlich JS; Reynolds RC Are bigger data sets better for machine learning? Fusing single-point and dual-event dose response data for mycobacterium tuberculosis. J Chem Inf Model 2014, 54, 2157–2165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Ekins S; Pottorf R; Reynolds RC; Williams AJ; Clark AM; Freundlich JS Looking back to the future: Predicting in vivo efficacy of small molecules versus mycobacterium tuberculosis. J Chem Inf Model 2014, 54, 1070–1082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Ekins S; Perryman AL; Clark AM; Reynolds RC; Freundlich JS Machine learning model analysis and data visualization with small molecules tested in a mouse model of mycobacterium tuberculosis infection (2014–2015). J Chem Inf Model 2016, 56 (7), 1332–1343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Ballell L; Bates RH; Young RJ; Alvarez-Gomez D; Alvarez-Ruiz E; Barroso V; Blanco D; Crespo B; Escribano J; Gonzalez R; Lozano S; Huss S; Santos-Villarejo A; Martin-Plaza JJ; Mendoza A; Rebollo-Lopez MJ; Remuinan-Blanco M; Lavandera JL; Perez-Herran E; Gamo-Benito FJ; Garcia-Bustos JF; Barros D; Castro JP; Cammack N Fueling open-source drug discovery: 177 small-molecule leads against tuberculosis. ChemMedChem 2013, 8, 313–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Mikusova K; Ekins S Learning from the past for tb drug discovery in the future. Drug Discov Today 2017, 22, 534–545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Paolino M; Brindisi M; Vallone A; Butini S; Campiani G; Nannicini C; Giuliani G; Anzini M; Lamponi S; Giorgi G; Sbardella D; Ferraris DM; Marini S; Coletta M; Palucci I; Minerva M; Delogu G; Pepponi I; Goletti D; Cappelli A; Gemma S; Brogi S Development of potent inhibitors of the mycobacterium tuberculosis virulence factor Zmp1 and evaluation of their effect on mycobacterial survival inside macrophages. ChemMedChem 2018, 13, 422–430. [DOI] [PubMed] [Google Scholar]
  • 249.Zhang D; Lin Y; Chen X; Zhao W; Chen D; Gao M; Wang Q; Wang B; Huang H; Lu Y; Lu Y Docking- and pharmacophore-based virtual screening for the identification of novel mycobacterium tuberculosis protein tyrosine phosphatase b (MptpB) inhibitor with a thiobarbiturate scaffold. Bioorg Chem 2019, 85, 229–239. [DOI] [PubMed] [Google Scholar]
  • 250.Bruhn DF; Scherman MS; Liu J; Scherbakov D; Meibohm B; Bottger EC; Lenaerts AJ; Lee RE In vitro and in vivo evaluation of synergism between anti-tubercular spectinamides and non-classical tuberculosis antibiotics. Sci Rep 2015, 5, 13985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Nathan C; Barry CE 3rd. Tb drug development: Immunology at the table. Immunol Rev 2015, 264, 308–318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Tornheim JA; Dooley KE Challenges of TB and HIV co-treatment: Updates and insights. Curr Opin HIV AIDS 2018, 13, 486–491. [DOI] [PubMed] [Google Scholar]
  • 253.Sadiq S; Khajuria V; Tandon VR; Mahajan A; Singh JB Adverse drug reaction profile in patients on anti-tubercular treatment alone and in combination with highly active antiretroviral therapy. J Clin Diagn Res 2015, 9, FC01–04. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Braunstein M; Hickey AJ; Ekins S Why wait? The case for treating tuberculosis with inhaled drugs. Pharm Res 2019, 36, 166. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

supplemental material

RESOURCES