Abstract

Background

Tuberculosis (TB) treatment is lengthy and complicated and patients often develop chronic lung disease. Recent attention has focused on host-directed therapies aimed at optimizing immune responses to Mycobacterium tuberculosis (Mtb), as adjunctive treatment given with antitubercular drugs. In addition to their cholesterol-lowering properties, 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) have broad anti-inflammatory and immunomodulatory activities.

Methods

In the current study, we screened 8 commercially available statins for cytotoxic effect, anti-TB activity, synergy with first-line drugs in macrophages, pharmacokinetics and adjunctive bactericidal activity, and, in 2 different mouse models, as adjunctive therapy to first-line TB drugs.

Results

Pravastatin showed the least toxicity in THP-1 and Vero cells. At nontoxic doses, atorvastatin and mevastatin were unable to inhibit Mtb growth in THP-1 cells. Simvastatin, fluvastatin, and pravastatin showed the most favorable therapeutic index and enhanced the antitubercular activity of the first-line drugs isoniazid, rifampin, and pyrazinamide in THP-1 cells. Pravastatin modulated phagosomal maturation characteristics in macrophages, phenocopying macrophage activation, and exhibited potent adjunctive activity in the standard mouse model of TB chemotherapy and in a mouse model of human-like necrotic TB lung granulomas.

Conclusions

These data provide compelling evidence for clinical evaluation of pravastatin as adjunctive, host-directed therapy for TB.

Despite more than a century of research, tuberculosis (TB) kills millions of people every year [1]. There is an imminent but unmet medical need for novel therapeutics to complement the current antimicrobial arsenal to combat drug-susceptible and drug-resistant TB [2–4]. In particular, attention has focused on the potential repurposing of existing US Food and Drug Administration-approved agents, many of which have broad immunomodulatory properties, and whose safety and tolerability profiles have been well characterized, in an effort to obtain accelerated approval for use as adjunctive host-directed therapy (HDT) for TB [5–8]. Host-directed therapy offers a novel strategy with mechanisms that include activating immune defense mechanisms or ameliorating tissue damage [9].

There has recently been considerable interest in repurposing 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (statins) as novel antimicrobials, including as antitubercular agents [10]. In addition to their cholesterol-lowering properties, statins have been shown to have broad anti-inflammatory and immunomodulatory properties [11]. More recently, Parihar et al [12] showed that peripheral blood mononuclear cells and monocyte-derived macrophages from patients with familial hypercholesterolemia receiving statin therapy were more effective in controlling Mycobacterium tuberculosis (Mtb) growth compared with those of healthy donors, and that statin therapy protected mice against TB-induced pathology. We found that simvastatin adjunctive therapy enhanced the first-line anti-TB regimen’s antimicrobial activity and shortened the time required to achieve cure in a standard mouse model of chronic TB infection [13, 14]. In addition, atorvastatin was found to be synergistic with the key sterilizing drug, rifampin [15]. Recent retrospective cohort studies found that statin use was associated with a reduced incidence of active TB disease [16, 17]. However, several important questions must be answered before statins can be used as adjunctive anti-TB agents in the clinical setting. For example, it remains to be determined whether the anti-TB activity of statins represents a class effect, or whether there is a hierarchy in antitubercular potency and toxicity among statins.

The objective of this study was to screen 8 commercially available statins (pravastatin, simvastatin, fluvastatin, atorvastatin, lovastatin, rosuvastatin, pitavastatin, and mevastatin) in various preclinical models to prioritize 1 or more statins for future prospective clinical studies.

MATERIALS AND METHODS

Assessment of Statin Cytotoxicity

To determine the 50% cytotoxic concentration (CC50) of each statin, exposures were performed with the human monocytic THP-1 cell line (TIB-202; ATCC) and Vero cells (CCL-81; ATCC). Drug cytotoxicity was determined by the MTS method (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt; Promega, Madison, WI). Cells were treated with a range of doses of each statin (with lowest dose corresponding to the inhibitory dose [IC50] reported in the literature), and toxicity was measured at 24 hours posttreatment. After treatment with statins, the MTS solution was added to each well and incubated at 37°C for 30 minutes. The absorbance at 490 nm was recorded, and the CC50 was determined using a nonlinear regression fitted with Prism 6 (GraphPad).

Assessment of Statin Efficacy in Macrophages

To determine the efficacy of statins against intracellular bacilli, a bioluminescent Mtb strain (H37Rv-lux), which stably expresses an integrated bioluminescent reporter (firefly luxABCDE full operon), including the luciferase enzyme and associated luciferin substrate, was used to infect THP-1 cells [14, 18]. Mycobacterial growth was monitored in real-time using a luminometer by measuring the relative light units (RLUs), which serve as a reliable surrogate for colony-forming units (CFUs) [19]. After differentiation, the cells were infected with a frozen stock of H37Rv-lux at a multiplicity of infection of 1:20 [14, 20]. Multiple concentrations of each statin (all causing <20% reduction of cell viability after 6 days of treatment) were tested to determine the concentration required to inhibit Mtb growth by 50% (half maximal effective concentration [EC50]) for each compound.

Evaluation of Synergy of Statins With Antituberculosis Drugs in THP-1 Cells

To determine whether statins can enhance the activity of anti-TB drugs against intracellular bacilli, we used the RLU assay described above. We first determined the EC50 of each of the 3 first-line drugs separately and then in combination. When each statin (simvastatin, pravastatin, or fluvastatin) was paired with the 3-drug combination of rifampin (0.0055 μM), isoniazid (0.006 μM), and pyrazinamide (81.23 μM) (collectively, RHZ) [14], each antibiotic in the combination was used at half the concentration required to reduce RLUs by 50% when dosed individually.

Phagosomal pH and Proteolysis Bead Assays

Carboxyfluorescein (pH readout) and DQ-BSA (proteolysis readout) beads were generated as previously described [21, 22]. Full details of the methods are provided in the Supplementary Materials.

Bone marrow-derived macrophages (BMDMs) were isolated from BALB/cJ mice (Jackson Laboratory, Bar Harbor, ME), and maintained in Dulbecco’s modified Eagle’s medium (Gibco, Grand Island, NY) containing 10% fetal bovine serum (FBS) (Gibco), 15% L-cell conditioned media, 2 mM L-glutamine (Sigma-Aldrich, St. Louis, MO), 1 mM sodium pyruvate (Gibco), and antibiotics (penicillin/streptomycin) (Gibco) at 37°C in a 5% CO2 atmosphere. For the plate reader assay, 2 × 105 macrophages/well were seeded into a 96-well clear bottom black plate (Corning Costar, Corning, NY). Where applicable, macrophages were activated by treatment with 100 U/mL interferon-γ (PeproTech, Rocky Hill, NJ) and 10 ng/mL lipopolysaccharide (List Biological Laboratories, Campbell, CA). Two days after seeding, the media were replaced with fresh media containing appropriate concentrations of pravastatin or water as a carrier control. Bead assays were carried out 2 days after treatment. Macrophages were washed 3 times with assay buffer (phosphate-buffered saline, pH 7.2, 5% FBS, 5 mM dextrose, 1 mM CaCl2, 2.7 mM KCl, 0.5 mM MgCl2), before addition of sensor beads at ~2–5 beads/macrophage in assay buffer [21, 22]. Data acquisition on a Biotek Synergy H1 microplate reader was started within 2–3 minutes of bead addition. Signal detection was from the bottom using the following excitation/emission wavelengths: carboxyfluorescein, 450 nm/520 nm and 490 nm/520 nm; DQ-BSA, 490 nm/520 nm; and Alexa Fluor (AF), 594–590 nm/617 nm. A total of 3–4 replicate wells/condition were used, with temperature maintained at 37°C throughout the assay. Reads were taken every 2 minutes for 2 hours for pH bead assays and every 2 minutes for 4 hours for proteolysis bead assays. Analyses of the results were performed as previously described [21, 22]. In brief, for the pH assay, the ratio of the carboxyfluorescein fluorescence signal at excitation 490 nm (pH-sensitive) versus 450 nm (pH-insensitive) provides a readout of relative pH. For the proteolysis assay, AF594 serves as an internal calibration control, allowing ratiometric measurement, with the DQ-BSA/AF594 ratio reflecting changes in proteolysis.

Ethics Statement

All procedures involving animals were performed according to protocols approved by the Institutional Animal Care and Use Committee at the Johns Hopkins University.

Tolerability and Pharmacokinetics of Statins in Mice

In steady-state studies, separate groups of female C3HeB/FeJ mice (aged 5 to 6 weeks; Jackson Laboratory) were given 1 of the 7 statins (mevastatin was not tested due to cytotoxicity) orally at a dose of 90 mg/kg together with isoniazid 10 mg/kg, rifampin 10 mg/kg, pyrazinamide 150 mg/kg, and ethambutol 100 mg/kg (collectively, RHZE) for 5 successive days. Rifampin doses were separated from the accompanying drugs by 1 hour to minimize drug-drug interactions during absorption [23, 24]. Whole lung tissue and serum at each time point were frozen at −20°C and later analyzed for concentrations of each drug by liquid chromatography-tandem mass spectrometry. Serum and lung drug concentration data were entered into a WinNonlin worksheet (WinNonlin version 4.0, 2002; Pharsight, Mountain View, CA) and analyzed using standard noncompartmental techniques [19, 25].

Antituberculosis Activity of Statins in C3HeB/FeJ Mice

A total of 110 female C3HeB/FeJ mice (aged 5 to 6 weeks; Jackson Laboratory) were aerosol-infected with Mtb H37Rv using an inhalation exposure system (Glas-Col, Terre Haute, IN) calibrated to deliver ~102 CFU/mouse lung. Six weeks after aerosol infection, mice were treated with human-equivalent doses of RHZE for 2 months and were randomized to receive one of the following adjuntive statin regimens designed to simulate human daily exposures (Supplementary Tables S2 and S3). The basic experimental scheme is shown in Supplementary Table S4. The treatment was administered once daily (5 days a week) for a total of 8 weeks. All statins were obtained from Toronto Research Chemicals (North York, ON, Canada), except pitavastatin, which was obtained from Kowa Company, Ltd. (Nagoya, Japan).

Dose-Ranging Activity of Pravastatin in the Standard Mouse Model of Tuberculosis Chemotherapy

Six-week-old female BALB/c mice (Charles River Laboratories, Wilmington, MA) were aerosol-infected with ~5 × 103 CFU of wild-type Mtb H37Rv, as described above. Treatment was initiated 4 weeks later with RHZE or RHZE + pravastatin at doses ranging from 30 to 180 mg/kg for a total of 8 weeks. Animal total body, lung, and spleen weights were recorded, and the lungs and spleen were examined grossly for visible lesions and were photographed at the time of sacrifice. Endpoints included the bactericidal activity of each regimen at defined time points.

Data Evaluation and Statistical Analysis

Log-transformed CFUs were used to calculate means and standard deviations (SDs). Comparisons of CFU data among experimental groups were performed by Student t test (2-tailed). Pairwise comparisons of group mean values for log10 counts were made by using one-way analysis of variance and Bonferroni’s multiple comparison test posttest with GraphPad Prism 7 (GraphPad, San Diego, CA), and P < .05 was considered significant.

RESULTS

Statins Show Differential Cytotoxicity

First, we tested the cytotoxicity of 8 commercially available statins in THP-1 cells and Vero cells. A range of concentrations of each statin was tested to determine the cytotoxic concentration leading to a 50% reduction in cell viability (CC50). An example is provided in Supplementary Figure S1 demonstrating how the CC50 of statin was determined in THP-1 cells. This approach was performed with the remaining 7 clinically available statins. Based on CC50, pravastatin was determined to be the least cytotoxic of the statins tested (Supplementary Table S1).

Simvastatin, Fluvastatin, and Pravastatin Exhibit the Most Potent Activity Against Intracellular Mycobacterium tuberculosis

At nontoxic doses, atorvastatin and mevastatin were unable to inhibit Mtb growth (Supplementary Table S1), whereas rosuvastatin, pitavastatin, and lovastatin gave <50% growth inhibition in our assay. The EC50 values (≥50% Mtb growth inhibition) could be obtained only for simvastatin, pravastatin, and fluvastatin (Figure 1A), and each of these drugs showed a favorable therapeutic index, as measured by CC50/EC50 (Supplementary Table S1). Thus, the majority of compounds inhibited intracellular Mtb growth, pointing to a class effect. Observed differences in antimicrobial activity may be due to differences in structure among the various statins.

Antitubercular activity of statins alone and in combination with the first-line drugs in THP-1 cells. (A) Mycobacterium tuberculosis H37Rv-lux survival in THP-1 cells after 6 days of exposure to statins at the EC50 (the lowest concentration of statin yielding 50% reduction in relative light units [RLU]). (B) In vitro anti-tuberculosis (TB) activity of simvastatin, pravastatin, and fluvastatin alone and in combination with first-line anti-TB drugs. Untreated (solvent control); SIMVA = simvastatin (0.2 μM); PRAVA = pravastatin (7.8 μM); FLUVA = fluvastatin (0.032 μM); PITA = pitavastatin (0.06 μM); ROSU = rosuvastatin (1.15 μM); LOVA = lovastatin (0.87 μM); RHZ = rifampin (0.0055 μM); isoniazid (0.006 μM); and pyrazinamide (81.23 μM). Significantly different values (*, P < .05; **, P < .01; ***, P < .001) between groups are indicated by “a” untreated vs treated; “b”, RHZE vs RHZE+ statin; “c”, statin vs RHZE+ statin. Student’s t test, 2-tailed, n = 3. Graph shows mean ± standard deviation.
Figure 1.

Antitubercular activity of statins alone and in combination with the first-line drugs in THP-1 cells. (A) Mycobacterium tuberculosis H37Rv-lux survival in THP-1 cells after 6 days of exposure to statins at the EC50 (the lowest concentration of statin yielding 50% reduction in relative light units [RLU]). (B) In vitro anti-tuberculosis (TB) activity of simvastatin, pravastatin, and fluvastatin alone and in combination with first-line anti-TB drugs. Untreated (solvent control); SIMVA = simvastatin (0.2 μM); PRAVA = pravastatin (7.8 μM); FLUVA = fluvastatin (0.032 μM); PITA = pitavastatin (0.06 μM); ROSU = rosuvastatin (1.15 μM); LOVA = lovastatin (0.87 μM); RHZ = rifampin (0.0055 μM); isoniazid (0.006 μM); and pyrazinamide (81.23 μM). Significantly different values (*, P < .05; **, P < .01; ***, P < .001) between groups are indicated by “a” untreated vs treated; “b”, RHZE vs RHZE+ statin; “c”, statin vs RHZE+ statin. Student’s t test, 2-tailed, n = 3. Graph shows mean ± standard deviation.

Statin Exposure Differentially Enhances the Activity of the First-Line Drugs Against Intracellular Mycobacterium tuberculosis

The EC50 values of isoniazid (H), rifampin (R), and pyrazinamide (Z) against Mtb in THP-1 cells were determined to be 0.006 μM, 0.0055 μM, and 81.23 μM, respectively. To test the synergistic activity of statins with standard antitubercular drugs, each statin was used at the respective EC50 in combination with the 3-drug combination HRZ, with each anti-TB drug dosed at half its EC50. We found that combination therapy with HRZ had more potent activity against intracellular Mtb than each statin alone (Figure 1B). Adjunctive therapy with pravastatin 7.8 μM significantly increased killing by HRZ (P  = 0.0024) (Figure 1B). A similar effect was observed with simvastatin 0.2 μM (P = .024) and fluvastatin 0.032 μM (P = .0004).

Pravastatin Treatment Phenocopies Phagosomal Acidification and Proteolytic Activity Profiles of Activated Macrophages

Previous reports have demonstrated that exposure of Mtb-infected macrophages to simvastatin promotes phagosome-lysosome fusion and autophagy [12]. To determine whether these activities also apply to pravastatin, we investigated the capacity of this drug to modulate phagosome acidification and proteolytic activity in mouse BMDMs. Treatment of resting BMDMs with pravastatin phenocopied the proteolytic activity and acidification profiles of phagosomes observed in interferon-γ- and lipopolysaccharide-activated BMDMs. This effect, which was concentration-dependent, suggests a contributing factor to intracellular Mtb killing by pravastatin (Figure 2).

Pravastatin modulates phagosome maturation in a dose-dependent manner. Murine bone marrow-derived macrophages were treated with the indicated pravastatin concentrations (or water as a vehicle control) for 2 days before assay start. Pravastatin treatment decreases phagosomal acidification (A) and proteolytic activity (B) in a concentration-dependent manner, phenocopying phagosome maturation profiles of macrophages activated by interferon-γ and lipopolysaccharide (C and D). Carboxyfluorescein (A and C) or DQ-BSA/AF594 (B and D) beads were added to the cells, or to wells containing only media, and fluorescence tracked with a microplate reader over time. Data are shown as means ± standard deviation from 3–4 wells.
Figure 2.

Pravastatin modulates phagosome maturation in a dose-dependent manner. Murine bone marrow-derived macrophages were treated with the indicated pravastatin concentrations (or water as a vehicle control) for 2 days before assay start. Pravastatin treatment decreases phagosomal acidification (A) and proteolytic activity (B) in a concentration-dependent manner, phenocopying phagosome maturation profiles of macrophages activated by interferon-γ and lipopolysaccharide (C and D). Carboxyfluorescein (A and C) or DQ-BSA/AF594 (B and D) beads were added to the cells, or to wells containing only media, and fluorescence tracked with a microplate reader over time. Data are shown as means ± standard deviation from 3–4 wells.

In Vivo Pharmacokinetics Studies and Selection of Statin Dose for Efficacy Studies in Mice

Before undertaking the in vivo efficacy studies, we studied the safety and pharmacokinetics (PK) of each statin dosed daily at 90 mg/kg by gavage in C3Heb/FeJ mice, a strain of mice that develops human-like, necrotic TB lung granulomas [26–28]. To select the dose of each statin for the efficacy studies, we aimed to reproduce the human area under the plasma drug concentration-time curve (AUC) at (1) the highest clinically licensed dose and (2) twice that dose for selected compounds exhibiting the most attractive cytotoxicity profile and anti-TB activity in macrophages (simvastatin and pravastatin). It is important to note that for most statins, matching the AUC resulted in a Cmax significantly higher than the corresponding value in humans (Supplementary Table S2).

Significant effects of rifampin coadministration on simvastatin or pravastatin exposures were not observed in mice (Supplementary Table S3), indicating that the standard mouse model does not recapitulate the known drug-drug interactions observed between rifampin and these statins in humans [29, 30]. No clinically apparent signs of toxicity were observed in any of the statin groups.

Statin Adjunctive Therapy Improves the Bactericidal Activity of the First-Line Antitubercular Regimen in C3HeB/FeJ Mice

After 8 weeks of treatment, Mtb-infected mice receiving statin adjunctive therapy had significantly reduced lung bacillary burdens relative to control mice receiving RHZE alone. Relative to the control regimen, adjunctive therapy with simvastatin 90 mg/kg, pravastatin 90 mg/kg, pravastatin 50 mg/kg, or fluvastatin 15 mg/kg reduced colony counts in the lungs by 1.28 log10 (P < .0001), 1.16 log10 (P < .01), 0.78 log10 (P < .05), and 0.90 log10 (P < .05), respectively (Figure 3A). There were no significant differences in total body weights, lung and spleen weights, gross lung pathology of mice lungs, or surface area of lung involved by inflammation in the treatment groups compared with control groups treated with RHZE alone (Figure 3B).

Adjunctive anti-tuberculosis (TB) activity of statins in C3HeB/FeJ mice. (A) C3HeBFeJ mice were infected with wild-type Mycobacterium tuberculosis H37Rv by the aerosol route. Four weeks after infection, the mice were left untreated (no drug) or treated with RHZE or RHZE plus statin daily (5 days/week) for 8 weeks. RHZE = rifampin (10 mg/kg), isoniazid (10 mg/kg), pyrazinamide (150 mg/kg), ethambutol (100 mg/kg). ATORVA = atorvastatin 40 mg/kg; FLUVA = fluvastatin 15 mg/kg; PITA = pitavastatin 3 mg/kg; PRAVA 50 = pravastatin 50 mg/kg; P/PRAVA 90 = pravastatin 90 mg/kg; ROSU 4 = rosuvastatin 4 mg/kg, ROSU 8 = rosuvastatin 8 mg/kg; SIMVA = simvastatin 90 mg/kg. One-way analysis of variance, Dunnett’s multiple comparison test, with a single pooled variance, n = 5. Significantly different values (****, P < .0001; **, P < .01) between groups are indicated by “a” untreated vs treated; “b”, RHZE vs RHZE+ statin. (B) Sections of C3HeB/FeJ mice lung were paraffin-embedded, sectioned and stained with hematoxylin and eosin. Treatment with RHZE showed substantial reduction in overall inflammation compared with no drug. However, the addition of pravastatin (P90) did not further reduce lung inflammation. Images were captured at ×2 magnification.
Figure 3.

Adjunctive anti-tuberculosis (TB) activity of statins in C3HeB/FeJ mice. (A) C3HeBFeJ mice were infected with wild-type Mycobacterium tuberculosis H37Rv by the aerosol route. Four weeks after infection, the mice were left untreated (no drug) or treated with RHZE or RHZE plus statin daily (5 days/week) for 8 weeks. RHZE = rifampin (10 mg/kg), isoniazid (10 mg/kg), pyrazinamide (150 mg/kg), ethambutol (100 mg/kg). ATORVA = atorvastatin 40 mg/kg; FLUVA = fluvastatin 15 mg/kg; PITA = pitavastatin 3 mg/kg; PRAVA 50 = pravastatin 50 mg/kg; P/PRAVA 90 = pravastatin 90 mg/kg; ROSU 4 = rosuvastatin 4 mg/kg, ROSU 8 = rosuvastatin 8 mg/kg; SIMVA = simvastatin 90 mg/kg. One-way analysis of variance, Dunnett’s multiple comparison test, with a single pooled variance, n = 5. Significantly different values (****, P < .0001; **, P < .01) between groups are indicated by “a” untreated vs treated; “b”, RHZE vs RHZE+ statin. (B) Sections of C3HeB/FeJ mice lung were paraffin-embedded, sectioned and stained with hematoxylin and eosin. Treatment with RHZE showed substantial reduction in overall inflammation compared with no drug. However, the addition of pravastatin (P90) did not further reduce lung inflammation. Images were captured at ×2 magnification.

Pravastatin Shows Adjunctive Activity in a Standard Mouse Model of Tuberculosis Chemotherapy

Given its favorable therapeutic index and reduced drug interactions with rifampin relative to simvastatin in humans [29, 30], we next performed a dose-ranging study of pravastatin adjunctive therapy in the mouse model of TB chemotherapy [25, 31]. The Mtb-infected BALB/c mice were treated with the standard TB regimen (RHZE) with or without increasing doses of pravastatin ranging from 30 to 180 mg/kg. Treatment was given orally 5 times weekly for 8 weeks (continuous phase) beginning 6 weeks after infection. Adjunctive treatment with pravastatin increased the bactericidal activity of the first-line regimen, reducing lung bacillary counts by 0.2–0.6 log10, 0.3–0.6 log10, and 0.3–0.8 log10 compared with RHZE alone after treatment for 2 weeks, 4 weeks, and 8 weeks, respectively (Figure 4).

Adjunctive treatment with pravastatin augments the bactericidal activity of the first-line antituberculosis regimen in a dose-dependent manner in BALB/c mice. RHZE = rifampicin (10 mg/kg), isoniazid (10 mg/kg), pyrazinamide (150 mg/kg), ethambutol (100 mg/kg). P = pravastatin. Numbers refer to the dose in mg/kg. One-way analysis of variance, Dunnett’s multiple comparison test, with a single pooled variance, n = 3. Significantly different values (*, P < .05; **, P < .01; ***, P < .001; ****, P < .0001) between groups are indicated by “a” untreated vs treated; “b”, RHZE vs RHZE+ statin. CFU, colony-forming units.
Figure 4.

Adjunctive treatment with pravastatin augments the bactericidal activity of the first-line antituberculosis regimen in a dose-dependent manner in BALB/c mice. RHZE = rifampicin (10 mg/kg), isoniazid (10 mg/kg), pyrazinamide (150 mg/kg), ethambutol (100 mg/kg). P = pravastatin. Numbers refer to the dose in mg/kg. One-way analysis of variance, Dunnett’s multiple comparison test, with a single pooled variance, n = 3. Significantly different values (*, P < .05; **, P < .01; ***, P < .001; ****, P < .0001) between groups are indicated by “a” untreated vs treated; “b”, RHZE vs RHZE+ statin. CFU, colony-forming units.

We also tested the effect of each drug regimen on lung inflammation by histological assessment at 8 weeks posttreatment. We observed that, in general, the percentage of lung surface area involved by inflammation correlated with the bactericidal activity of each drug regimen (Figure 5). Specifically, the percentage of lung surface area occupied by inflammatory cells was reduced by 61.51% in the RHZE-treated group (20.88% ± 7.72%) relative to the untreated control (82.40% ± 3.60%). Treatment with pravastatin adjunctive therapy showed a statistically nonsignificant further reduction in mean inflamed lung surface area relative to the RHZE control group (13.76% for RHZE + pravastatin 90 mg/kg, P = .3 and 9.61% for RHZE + pravastatin 180 mg/kg, P = .62).

Histological assessment of lung inflammation after 8 weeks of treatment. At least 1 entire hematoxylin and eosin-stained cross-section per animal BALB/c mouse lung (5 animals/group) was analyzed for degree of inflammation. (A) The surface area occupied by cellular inflammation was determined by ImageJ software-based morphometry of digitized images of lung sections, and results are represented as percentage of lung surface area involved. (B) Representative histopathology images of lungs from Mycobacterium tuberculosis-infected BALB/C mice treated with pravastatin adjunctive therapy (90 mg/kg) in combination with the first-line anti-tubercular regimen (RHZE). RHZE = rifampicin (10 mg/kg), isoniazid (10 mg/kg), pyrazinamide (150 mg/kg), ethambutol (100 mg/kg). One-way analysis of variance, Dunnett’s multiple comparison test, with a single pooled variance, n = 3. Significantly different values (****, P < .0001) between groups are indicated by “a” untreated vs treated. Images were at ×2 magnification. P, pravastatin.
Figure 5.

Histological assessment of lung inflammation after 8 weeks of treatment. At least 1 entire hematoxylin and eosin-stained cross-section per animal BALB/c mouse lung (5 animals/group) was analyzed for degree of inflammation. (A) The surface area occupied by cellular inflammation was determined by ImageJ software-based morphometry of digitized images of lung sections, and results are represented as percentage of lung surface area involved. (B) Representative histopathology images of lungs from Mycobacterium tuberculosis-infected BALB/C mice treated with pravastatin adjunctive therapy (90 mg/kg) in combination with the first-line anti-tubercular regimen (RHZE). RHZE = rifampicin (10 mg/kg), isoniazid (10 mg/kg), pyrazinamide (150 mg/kg), ethambutol (100 mg/kg). One-way analysis of variance, Dunnett’s multiple comparison test, with a single pooled variance, n = 3. Significantly different values (****, P < .0001) between groups are indicated by “a” untreated vs treated. Images were at ×2 magnification. P, pravastatin.

DISCUSSION

Because the TB drug development pipeline remains sparse, novel approaches are required to shorten the duration of treatment and improve microbiological and clinical outcomes. The following HDT agents are currently being evaluated in phase 2 clinical trials as adjuncts to rifabutin-modified standard therapy in adults with drug-sensitive, smear-positive pulmonary TB: (1) the mammalian target of rapamycin inhibitor, everolimus (0.5 mg), (2) auranofin (6 mg), (3) vitamin D3, and (4) the phosphodiesterase-4 (PDE4) inhibitor, CC-11050 (ClinicalTrials.gov Identifier: NCT02968927) [32–35]. In a recent study, an analysis of retrospective cohorts showed that metformin use was associated with improved control of Mtb infection and decreased disease severity in patients with pulmonary TB [36].

In addition to pravastatin’s antitubercular activity in mice described in the present study, additional considerations favor the clinical use of pravastatin over other members of the class, including simvastatin, which showed the most favorable CC50/EC50 in macrophages in vitro. First and foremost, unlike simvastatin, the metabolism of pravastatin is not significantly mediated by cytochrome P450 enzymes [37–39]. Thus, although the steady-state AUC of both simvastatin and its active metabolite, simvastatin acid, are reduced by ~90% during coadministration with rifampin in humans [30], pravastatin exposures are reduced only by 30%–50% by rifampin [29], and the latter drug interaction can be overcome by doubling the dose of pravastatin [30]. This is particularly important because rifampin is a key sterilizing drug, and its inclusion in the first-line regimen has contributed significantly to treatment shortening to the current 6-month regimen for drug-susceptible TB. In addition, whereas human immunodeficiency virus (HIV) protease inhibitors have been shown to increase the median AUC of simvastatin by as much as 30-fold [40], pravastatin is one of the preferred statins for use in individuals with HIV infection due to its favorable toxicity profile and relatively low potential for drug interactions with antiretroviral drugs, an important consideration in populations in which TB and HIV are coendemic. Our previous PK data indicate that simvastatin is almost entirely converted to its acid metabolite in mice [14], whereas in humans, metabolism to simvastatin acid is ~1:1 [41]. Because it is not known where the parent drug or its acid metabolite is responsible for the observed anti-TB activity, it is possible that the mouse data overestimate the adjunctive activity of simvastatin.

Statins inhibit HMG-CoA reductase, the enzyme that catalyzes the conversion of HMG-CoA to mevalonate. The mevalonate pathway is essential for cholesterol synthesis, but it also contributes to the production of isoprenoids such as farnesyl pyrophosphate and geranylgeranyl pyrophosphate that are important for cell signaling and structure. A recent study found that inhibiting cholesterol with statins decreased the burden of Mycobacterium leprae in macrophages, highlighting the importance of cholesterol in promoting bacterial growth [42] and enhancing macrophage killing ability [15]. The ability of statins to reduce cholesterol has also been linked to increased killing of Helicobacter pylori [43], Coxiella burnetii [44], and Rickettsia conorii [45]. However, the beneficial effects of statins are not limited to cholesterol reduction alone, and they may involve processes associated with isoprenylation. These molecules serve as lipid labels for the posttranslational modification of several proteins, including G protein gamma subunits and small GTP-binding proteins, such as Ras, Rho, Rab, Rac, Ral, or Rap [46, 47]. Given the central role of small GTP-binding proteins in controlling the immune response, it is not surprising that statins exerts immunomodulatory effects independent of cholesterol lowering.

Our data showing reduced phagosome acidification and proteolysis of macrophages after pravastatin exposure are consistent with the notion that pravastatin-treated macrophages display an activated phenotype. Decreased proteolysis can promote antigen presentation [48, 49], which is beneficial for bacterial clearance, whereas the observed delay in acidification reflects a balance in the production of reactive oxygen species, which consume protons, and phagosomal acidification upon immune cell activation [48, 50]. Our macrophage functional data add to the endosomal marker data of Parihar et al [12], and the 2 studies are consistent with the conclusion that statin exposure induces macrophage activation.

From our work and that of other groups, it appears that statins can exert their effect on various cell types, and their anti-TB activity is likely multifactorial. We acknowledge that the mechanistic studies in the current study showing altered phagosome acidification and proteolytic activity with statin exposure do not necessarily demonstrate causality; however, they pave the way for further mechanistic studies to determine the molecular basis of the anti-TB activity of statins.

CONCLUSIONS

The preponderance of evidence suggests that statins may be useful as adjunctive HDT for TB. However, a number of important questions remain to be addressed before introducing statins as adjunctive agents in TB therapy, including the precise dosing of pravastatin, taking into consideration known drug interactions with rifampin, and its potential utility in patients with HIV coinfection. A randomized clinical trial, Statins as Adjunctive Therapy for TB (StAT-TB), is currently underway to determine whether pravastatin adjunctive therapy shortens the median time to sputum-culture negativity and improves lung function outcomes among HIV-infected and uninfected patients with drug-susceptible pulmonary TB.

Supplementary Data

Supplementary materials are available at The Journal of Infectious Diseases online. Consisting of data provided by the authors to benefit the reader, the posted materials are not copyedited and are the sole responsibility of the authors, so questions or comments should be addressed to the corresponding author.

Notes

Acknowledgments. We thankfully acknowledge Michael L. Pinn for excellent technical assistance.

Disclaimer. The funding sources had no role in the study design, data collection, data analysis, data interpretation or writing of the report. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Financial support. This work was funded by Grant R00 AI114952 (to S. T.), BMGF Bill & Melinda Gates Foundation Grant OPP 1066499 (to V. D.), Grant R01 HL106788 (to M. L. G.), and ACTG AIDS Clinical Trials Group Grant 110007 and CFAR Centers for AIDS Research Supplement P30AI094189, R01 HL106786, and UH2/3 AI122309 (to P. C. K.).

Potential conflicts of interest. All authors: No reported conflicts of interest. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest.

References

1.

Velásquez
GE
,
Becerra
MC
,
Gelmanova
IY
, et al.
Improving outcomes for multidrug-resistant tuberculosis: aggressive regimens prevent treatment failure and death
.
Clin Infect Dis
2014
;
59
:
9
15
.

2.

Norrby
SR
,
Nord
CE
,
Finch
R
;
European Society of Clinical Microbiology and Infectious Diseases
.
Lack of development of new antimicrobial drugs: a potential serious threat to public health
.
Lancet Infect Dis
2005
;
5
:
115
9
.

3.

Lawn
SD
,
Zumla
AI
.
Tuberculosis
.
Lancet
2011
;
378
:
57
72
.

4.

Dutta
NK
,
Karakousis
PC
.
Tuberculosis chemotherapy: present situation, possible solutions, and progress towards a TB-free world
.
Indian J Med Microbiol
2012
;
30
:
261
3
.

5.

Hawn
TR
,
Matheson
AI
,
Maley
SN
,
Vandal
O
.
Host-directed therapeutics for tuberculosis: can we harness the host?
Microbiol Mol Biol Rev
2013
;
77
:
608
27
.

6.

Paige
C
,
Bishai
WR
.
Penitentiary or penthouse condo: the tuberculous granuloma from the microbe’s point of view
.
Cell Microbiol
2010
;
12
:
301
9
.

7.

Schwegmann
A
,
Brombacher
F
.
Host-directed drug targeting of factors hijacked by pathogens
.
Sci Signal
2008
;
1
:
re8
.

8.

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
.

9.

Frank
DJ
,
Horne
DJ
,
Dutta
NK
, et al.
Remembering the host in tuberculosis drug development
.
J Infect Dis
2019
;
219
:
1518
24
.

10.

Alffenaar
JC
,
Akkerman
OW
,
van Hest
R
.
Statin adjunctive therapy for tuberculosis treatment
.
Antimicrob Agents Chemother
2016
;
60
:
7004
.

11.

Hennessy
E
,
Adams
C
,
Reen
FJ
,
O’Gara
F
.
Is there potential for repurposing statins as novel antimicrobials?
Antimicrob Agents Chemother
2016
;
60
:
5111
21
.

12.

Parihar
SP
,
Guler
R
,
Khutlang
R
, et al.
Statin therapy reduces the mycobacterium tuberculosis burden in human macrophages and in mice by enhancing autophagy and phagosome maturation
.
J Infect Dis
2014
;
209
:
754
63
.

13.

Skerry
C
,
Pinn
ML
,
Bruiners
N
,
Pine
R
,
Gennaro
ML
,
Karakousis
PC
.
Simvastatin increases the in vivo activity of the first-line tuberculosis regimen
.
J Antimicrob Chemother
2014
;
69
:
2453
7
.

14.

Dutta
NK
,
Bruiners
N
,
Pinn
ML
, et al.
Statin adjunctive therapy shortens the duration of TB treatment in mice
.
J Antimicrob Chemother
2016
;
71
:
1570
7
.

15.

Lobato
LS
,
Rosa
PS
,
Ferreira
Jda S
, et al.
Statins increase rifampin mycobactericidal effect
.
Antimicrob Agents Chemother
2014
;
58
:
5766
74
.

16.

Lai
CC
,
Lee
MT
,
Lee
SH
, et al.
Statin treatment is associated with a decreased risk of active tuberculosis: an analysis of a nationally representative cohort
.
Thorax
2016
;
71
:
646
51
.

17.

Su
VY
,
Su
WJ
,
Yen
YF
, et al.
Statin use is associated with a lower risk of TB
.
Chest
2017
;
152
:
598
606
.

18.

Andreu
N
,
Fletcher
T
,
Krishnan
N
,
Wiles
S
,
Robertson
BD
.
Rapid measurement of antituberculosis drug activity in vitro and in macrophages using bioluminescence
.
J Antimicrob Chemother
2012
;
67
:
404
14
.

19.

Andreu N, Fletcher T, Krishnan N et al. Rapid measurement of antituberculosis drug activity in vitro and in macrophages using bioluminescence. J Antimicrob Chemother 2012; 67:404–14

20.

Salamon
H
,
Bruiners
N
,
Lakehal
K
, et al.
Cutting edge: vitamin D regulates lipid metabolism in Mycobacterium tuberculosis infection
.
J Immunol
2014
;
193
:
30
4
.

21.

Tan
S
,
Yates
RM
,
Russell
DG
.
Mycobacterium tuberculosis: readouts of bacterial fitness and the environment within the phagosome
.
Methods Mol Biol
2017
;
1519
:
333
47
.

22.

Podinovskaia
M
,
Lee
W
,
Caldwell
S
,
Russell
DG
.
Infection of macrophages with Mycobacterium tuberculosis induces global modifications to phagosomal function
.
Cell Microbiol
2013
;
15
:
843
59
.

23.

Grosset
J
,
Truffot-Pernot
C
,
Lacroix
C
,
Ji
B
.
Antagonism between isoniazid and the combination pyrazinamide-rifampin against tuberculosis infection in mice
.
Antimicrob Agents Chemother
1992
;
36
:
548
51
.

24.

Dhillon
J
,
Dickinson
JM
,
Sole
K
,
Mitchison
DA
.
Preventive chemotherapy of tuberculosis in Cornell model mice with combinations of rifampin, isoniazid, and pyrazinamide
.
Antimicrob Agents Chemother
1996
;
40
:
552
5
.

25.

Dutta
NK
,
Pinn
ML
,
Karakousis
PC
.
Reduced emergence of isoniazid resistance with concurrent use of thioridazine against acute murine tuberculosis
.
Antimicrob Agents Chemother
2014
;
58
:
4048
53
.

26.

Dutta
NK
,
Illei
PB
,
Jain
SK
,
Karakousis
PC
.
Characterization of a novel necrotic granuloma model of latent tuberculosis infection and reactivation in mice
.
Am J Pathol
2014
;
184
:
2045
55
.

27.

Dutta
NK
,
Karakousis
PC
.
PA-824 is as effective as isoniazid against latent tuberculosis infection in C3HeB/FeJ mice
.
Int J Antimicrob Agents
2014
;
44
:
564
6
.

28.

Harper
J
,
Skerry
C
,
Davis
SL
, et al.
Mouse model of necrotic tuberculosis granulomas develops hypoxic lesions
.
J Infect Dis
2012
;
205
:
595
602
.

29.

Kyrklund
C
,
Backman
JT
,
Kivistö
KT
,
Neuvonen
M
,
Laitila
J
,
Neuvonen
PJ
.
Rifampin greatly reduces plasma simvastatin and simvastatin acid concentrations
.
Clin Pharmacol Ther
2000
;
68
:
592
7
.

30.

Kyrklund
C
,
Backman
JT
,
Neuvonen
M
,
Neuvonen
PJ
.
Effect of rifampicin on pravastatin pharmacokinetics in healthy subjects
.
Br J Clin Pharmacol
2004
;
57
:
181
7
.

31.

Nuermberger
E
,
Tyagi
S
,
Tasneen
R
, et al.
Powerful bactericidal and sterilizing activity of a regimen containing PA-824, moxifloxacin, and pyrazinamide in a murine model of tuberculosis
.
Antimicrob Agents Chemother
2008
;
52
:
1522
4
.

32.

Lachmandas
E
,
Beigier-Bompadre
M
,
Cheng
SC
, et al.
Rewiring cellular metabolism via the AKT/mTOR pathway contributes to host defence against Mycobacterium tuberculosis in human and murine cells
.
Eur J Immunol
2016
;
46
:
2574
86
.

33.

Harbut
MB
,
Vilchèze
C
,
Luo
X
, et al.
Auranofin exerts broad-spectrum bactericidal activities by targeting thiol-redox homeostasis
.
Proc Natl Acad Sci U S A
2015
;
112
:
4453
8
.

34.

Rekha
RS
,
Mily
A
,
Sultana
T
, et al.
Immune responses in the treatment of drug-sensitive pulmonary tuberculosis with phenylbutyrate and vitamin D3 as host directed therapy
.
BMC Infect Dis
2018
;
18
:
303
.

35.

Subbian
S
,
Tsenova
L
,
Holloway
J
, et al.
Adjunctive phosphodiesterase-4 inhibitor therapy improves antibiotic response to pulmonary tuberculosis in a rabbit model
.
EBioMedicine
2016
;
4
:
104
14
.

36.

Singhal
A
,
Jie
L
,
Kumar
P
, et al.
Metformin as adjunct antituberculosis therapy
.
Sci Transl Med
2014
;
6
:
263ra159
.

37.

Jacobsen
W
,
Kirchner
G
,
Hallensleben
K
, et al.
Small intestinal metabolism of the 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitor lovastatin and comparison with pravastatin
.
J Pharmacol Exp Ther
1999
;
291
:
131
9
.

38.

Jacobsen
W
,
Kirchner
G
,
Hallensleben
K
, et al.
Comparison of cytochrome P-450-dependent metabolism and drug interactions of the 3-hydroxy-3-methylglutaryl-CoA reductase inhibitors lovastatin and pravastatin in the liver
.
Drug Metab Dispos
1999
;
27
:
173
9
.

39.

Everett
DW
,
Chando
TJ
,
Didonato
GC
,
Singhvi
SM
,
Pan
HY
,
Weinstein
SH
.
Biotransformation of pravastatin sodium in humans
.
Drug Metab Dispos
1991
;
19
:
740
8
.

40.

Fichtenbaum
CJ
,
Gerber
JG
,
Rosenkranz
SL
, et al. ;
NIAID AIDS Clinical Trials Group
.
Pharmacokinetic interactions between protease inhibitors and statins in HIV seronegative volunteers: ACTG Study A5047
.
AIDS
2002
;
16
:
569
77
.

41.

Jemal
M
,
Ouyang
Z
,
Powell
ML
.
Direct-injection LC-MS-MS method for high-throughput simultaneous quantitation of simvastatin and simvastatin acid in human plasma
.
J Pharm Biomed Anal
2000
;
23
:
323
40
.

42.

Mattos
KA
,
Oliveira
VC
,
Berrêdo-Pinho
M
, et al.
Mycobacterium leprae intracellular survival relies on cholesterol accumulation in infected macrophages: a potential target for new drugs for leprosy treatment
.
Cell Microbiol
2014
;
16
:
797
815
.

43.

Liao
WC
,
Huang
MZ
,
Wang
ML
, et al.
Statin decreases Helicobacter pylori burden in macrophages by promoting autophagy
.
Front Cell Infect Microbiol
2016
;
6
:
203
.

44.

Botelho-Nevers
E
,
Espinosa
L
,
Raoult
D
,
Rolain
JM
.
Lovastatin, but not pravastatin, limits in vitro infection due to Coxiella burnetii
.
J Antimicrob Chemother
2008
;
62
:
845
7
.

45.

Botelho-Nevers
E
,
Rolain
JM
,
Espinosa
L
,
Raoult
D
.
Statins limit Rickettsia conorii infection in cells
.
Int J Antimicrob Agents
2008
;
32
:
344
8
.

46.

Oesterle
A
,
Laufs
U
,
Liao
JK
.
Pleiotropic effects of statins on the cardiovascular system
.
Circ Res
2017
;
120
:
229
43
.

47.

Oesterle
A
,
Liao
JK
.
The pleiotropic effects of statins - from coronary artery disease and stroke to atrial fibrillation and ventricular tachyarrhythmia
.
Curr Vasc Pharmacol
2019
;
17
:
222
32
.

48.

Savina
A
,
Jancic
C
,
Hugues
S
, et al.
NOX2 controls phagosomal pH to regulate antigen processing during crosspresentation by dendritic cells
.
Cell
2006
;
126
:
205
18
.

49.

Delamarre
L
,
Pack
M
,
Chang
H
,
Mellman
I
,
Trombetta
ES
.
Differential lysosomal proteolysis in antigen-presenting cells determines antigen fate
.
Science
2005
;
307
:
1630
4
.

50.

Sokolovska
A
,
Becker
CE
,
Ip
WK
, et al.
Activation of caspase-1 by the NLRP3 inflammasome regulates the NADPH oxidase NOX2 to control phagosome function
.
Nat Immunol
2013
;
14
:
543
53
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)