Skip to main content
iScience logoLink to iScience
. 2025 Jan 30;28(3):111932. doi: 10.1016/j.isci.2025.111932

Predicting tuberculosis drug efficacy in preclinical and clinical models from in vitro data

Janice JN Goh 1,8, Anu Patel 1, Bernard Ngara 1, Rob C van Wijk 1, Natasha Strydom 1, Qianwen Wang 1, Nhi Van 2, Tracy M Washington 2,3, Eric L Nuermberger 4, Bree B Aldridge 2,3, Christine Roubert 5,6, Jansy Sarathy 7, Véronique Dartois 7, Rada M Savic 1,9,
PMCID: PMC11875147  PMID: 40034847

Summary

Multiple in vitro potency assays are used to evaluate compounds against Mycobacterium tuberculosis, but a consensus on clinically relevant assays is lacking. We aimed to identify an in vitro assay signature that predicts preclinical efficacy and early clinical outcome. Thirty-one unique in vitro assays were compiled for 10 TB drugs. In vitro EC50 values were compared to pharmacokinetic-pharmacodynamic (PK-PD)-model-derived EC50 values from mice evaluated via multinomial regression. External validation of best-performing in vitro assay combinations was performed using five new TB drugs. Best-performing assay signatures for acute and subacute infections were described by assays that reproduce conditions found in macrophages and foamy macrophages and chronic infection by the ex vivo caseum assay. Subsequent simulated mouse bacterial burden over time using predicted in vivo EC50 was within 2-fold of observations. This study helps us identify clinically relevant assays and prioritize successful drug candidates, saving resources and accelerating clinical success.

Subject areas: bioinformatics, biological sciences, natural sciences, pharmacoinformatics, pharmacology

Highlights

  • In TB, it is unknown which potency assays prioritize in vivo compound efficacy

  • We made a PK-PD platform using in vivo potency data to identify clinically relevant assays

  • This platform could also predict monotherapy outcomes in preclinical and clinical studies

  • This helps prioritize promising drug candidates and save resources in drug development


Bioinformatics; Biological sciences; Natural sciences; Pharmacoinformatics; Pharmacology

Introduction

Tuberculosis (TB) was the top killer among infectious diseases globally as of 2022.1 A standard therapy consisting of a cocktail of four drugs exists,2 but long treatment duration and strict adherence requirements make cure difficult to achieve.3 Thus, there is an urgent need for the development of new drugs that can shorten treatment duration. Many novel drug candidates have emerged in recent years4; however, the clinical trial process is often long and expensive. Therefore, it is essential to better prioritize which drugs are the most likely to succeed clinically.

In vitro assays are typically used to test a new compound for activity prior to animal studies. Traditionally, the minimum inhibitory concentration (MIC) is used to determine a drug’s potency and efficacy against Mycobacterium tuberculosis (Mtb).5 However, MIC assays are often carried out in a nutrient-rich media that promotes rapid bacterial growth and are not representative of the physiological conditions in which Mtb grows in human hosts. Furthermore, throughout treatment, Mtb growth slows, making it less susceptible to drug treatment.6,7 Therefore, novel in vitro assays have since been developed to better mimic this slow-growing, “persister” state, such as depriving Mtb of key nutrients or oxygen or adding immune cells to simulate what happens in an actual infection.8 There is not yet consensus on which of these assays are most predictive of in vivo drug efficacy, and thus most informative, for prioritizing preclinical and clinical candidates.

Previously, we developed an integrated pharmacokinetic-pharmacodynamic (PK-PD) model describing bacterial dynamics that allows us to estimate the in vivo potency (EC50) of a drug in mice after accounting for the adaptive immune effect.9 EC50 is defined as the drug concentration needed to achieve 50% of maximal response. In an in vivo model, drug concentration is dynamic and changes over time. However, by accounting for this using PK-PD modeling, we can integrate both drug concentration and pharmacodynamic response over time into a single model and estimate an in vivo EC50, similar to in vitro EC50, as a single concentration. We found that EC50 from mice was portable to humans and could be used to estimate clinical early bactericidal activity (EBA) outcomes in Phase 2a trials. Using in vivo EC50 from mice as a standard, we aimed to ask two questions: (1) Which in vitro assays, or combination of assays, are the most useful for predicting in vivo EC50? and (2) Are these in vitro correlates of in vivo EC50 useful for the prediction of both in vivo preclinical efficacy and clinical efficacy? Knowing which in vitro assays are most informative for the prediction of outcomes in preclinical in vivo models and clinical outcomes will help us to streamline compound progression and accelerate drug development.

Results

Compilation of a rich in vitro assay dataset

We compiled a rich dataset of 31 unique assays tested on 10 key first- and second-line drugs with corresponding PK-PD models and clinical data available (Figures 1 and 2A). These drugs were bedaquiline (BDQ), delamanid (DLM), ethambutol (EMB), isoniazid (INH), moxifloxacin (MFX), linezolid (LZD), pretomanid (PMD), pyrazinamide (PZA), rifampicin (RIF), and rifapentine (RPT). Of these 10 drugs, 7 had efficacy data in mouse acute models (EMB, INH, LZD, MXF, PMD, PZA, and RIF), 8 in subacute models (BDQ, DLM, INH, LZD, MXF, PMD, PZA, and RIF), and 7 in chronic models (INH, LZD, MXF, PMD, PZA, RIF, and RPT). Most of the assays only reported drug potencies rather than a full dose-response curve. Hence, we focused on EC50 rather than Emax to generate the exposure response.

Figure 1.

Figure 1

A three-step pipeline to translate in vitro potency (EC50) to in vivo mouse EC50 and in vivo efficacy when combined with a pharmacokinetic-pharmacodynamic (PK-PD) model

(A) In vitro assays were collected from literature and from collaborators for 10 drugs of interest: bedaquiline (BDQ), delamanid (DLM), ethambutol (EMB), isoniazid (INH), linezolid (LZD), moxifloxacin (MXF), pretomanid (PMD), pyrazinamide (PZA), rifampicin (RIF), and rifapentine (RPT). Mouse PK-PD models with a baseline describing bacterial dynamics were also previously built for these drugs.

(B) Univariate linear regression was first carried out to understand the individual relationships between in vitro EC50 and in vivo EC50 derived from mouse PK-PD models. A multinomial regression was then built to find the least number of in vitro assays with the best accuracy for predicting in vivo EC50.

(C) Predicted in vivo EC50 was used to make a new exposure-response relationship in the mouse PK-PD model to predict the bacterial colony-forming units (CFUs) over time profile in mouse.

Figure 2.

Figure 2

In vitro assay description and correlation

(A) Overview of all 31 in vitro assays across 10 drugs of interest, clustered by their in vitro potency (EC50). Missing data were denoted as an empty white square, whereas inactive drugs in an assay were arbitrarily assigned a value of 9999.

(B) Pairwise correlations between in vitro assays clustered by their Pearson correlation value (R) show many high positive correlations between in vitro assays.

(C) Network plot of high pairwise correlations between in vitro assays. In vitro assays with absolute pairwise correlations higher than 0.9 were joined by an edge in the network plot. Assays with six or more edges, or with the greatest number of edges in their cluster, filled in red, were chosen as representative of the many highly correlated assays. Table 2 also lists every significant pairwise correlation among the assays with high correlation. Assays that had no high pairwise correlations were also selected as features for further analysis. BDQ, bedaquiline, DLM, delamanid, EMB, ethambutol, INH, isoniazid, LZD, linezolid, MXF, moxifloxacin, PMD, pretomanid, PZA, pyrazinamide, RIF, rifampicin, and RPT, rifapentine.

Our assumption of a Hill coefficient of 1, which allowed us to easily convert all potencies to EC50, held true in our analysis. When comparing models with and without a Hill coefficient using a nested F test, the reported nested F value of 0.600 showed statistical insignificance.

To categorize the assays as a function of their biological relevance at the various sites of disease, we created four broad categories: (1) nutrient-rich media conditions representing extracellular rapidly replicating Mtb, (2) essential nutrient substitution or deletion slowing Mtb replication, (3) macrophage assays reflecting the involvement of the immune system slowing down Mtb growth to a more persistent state during acute infection, and (4) ex vivo assays representing the necrotic lesion core formed in severe cases of TB infection. Out of these 31 assays, 7 were under nutrient-rich conditions (e.g., MIC in Middlebrook 7H9 medium), 18 were a change in culture condition, via either a media formulation or oxygen deprivation, 5 were macrophage assays involving the uptake of Mtb into an immune cell, and 1 was an ex vivo assay using harvested caseum from New Zealand White rabbits (Figure 2A). Three assays—minimum bactericidal activity under non-replicating conditions (MBC NR) and two assays designed to quantify the impact of nonspecific drug binding to serum (rich medium plus 10% fetal bovine serum [FBS] or 4% bovine serum albumin [BSA])—did not have assay information for at least three drugs of interest and were discarded (Figure 2A). The full list of assays and data sources is displayed in Table 1.

Table 1.

In vitro assays compiled and their sources

Assay type Source References Assay classification/bacterial population
10% FBS MIC TBDA Upton et al. 201410 Nutrient-rich, nonspecific drug binding/fast-growing
3 Stress Assay TBDA Early et al. 201911 Nutrient substitution/slow-growing
4% BSA MIC TBDA Upton et al. 201410 Nutrient-rich, nonspecific drug binding/fast-growing
4 Stress NR CARA TBDA Gold et al. 201512 Nutrient substitution/slow-growing
4 Stress NR MIC TBDA Gold et al. 201512 Nutrient substitution/slow-growing
BCG Stationary TBDA Quezada 201913 Nutrient substitution/slow-growing
CARA MBC TBDA Gold et al. 201512 Nutrient-rich/fast-growing
Caseum Dartois lab, HMH Sarathy et al. 201814 Ex vivo caseum/slow-growing
Human Macrophage Sarathy lab, HMH Lanni et al. 202315 Macrophage/slow-growing
Acidic Aldridge lab, TU Larkins-Ford et al. 202116 Nutrient substitution/slow-growing
Butyrate Aldridge lab, TU Larkins-Ford et al. 202116 Nutrient substitution/slow-growing
Tufts Cholesterol Aldridge lab, TU Larkins-Ford et al. 202116 Nutrient substitution/slow-growing
Tufts High Cholesterol Aldridge lab, TU Larkins-Ford et al. 202116 Nutrient substitution/slow-growing
Dormancy Aldridge lab, TU Larkins-Ford et al. 202116 Nutrient substitution/slow-growing
J774 Aldridge lab, TU Larkins-Ford et al. 202116 Macrophage/slow-growing
Tufts Standard Aldridge lab, TU Larkins-Ford et al. 202116 Nutrient-rich/fast-growing
Valerate Aldridge lab, TU Larkins-Ford et al. 202116 Nutrient substitution/slow-growing
THP-1 Hypoxia Evotec S. Souriant et al. 202317 Macrophage/slow-growing
THP-1 Normoxia Evotec Badé et al. 202118 Macrophage/slow-growing
LCC Literature search Lakshminarayana et al. 2015,19 Sarathy et al. 201320 Nutrient substitution/slow-growing
LORA MBC TBDA Cho et al. 200721 Nutrient substitution/slow-growing
LORA MIC TBDA Cho et al. 200721 Nutrient substitution/slow-growing
RAW264.7 Literature search Lakshminarayana et al. 201519 Macrophage/fast-growing
MBC Literature search; Dartois Lab, HMH Lakshminarayana et al. 2015,19 Sarathy et al. 2013,20 Koul 200822 Nutrient-rich/fast-growing
MBC NR Literature search Xie et al. 2005,23 Upton et al. 2014,10 Sarathy et al. 201320 Nutrient substitution/slow-growing
MIC Literature search; Dartois Lab, HMH Lakshminarayana et al. 2015,19 Cho et al. 2007,21 Silva et al. 2018,24 Kim 2021,25 Drusano 2018,26 Upton et al. 201410 Nutrient-rich/fast-growing
MIC NR Gold Lab, WCMC Gold et al. 2015,12 Upton et al. 201410 Nutrient substitution/slow-growing
NARA MBC TBDA Gold et al. 201512 Nutrient-rich/fast-growing
Replicative Extracellular MIC Evotec Microplate Alamar Blue Assay in Zhang et al. 202327 Nutrient substitution/fast-growing
NIAID Cholesterol Boshoff lab, NIAID Nuermberger et al. 202228 Nutrient substitution/slow-growing
WCC Literature search Lakshminarayana et al. 2015,19 A. Koul 200822 Nutrient substitution/slow-growing

NR, non-replicative; MIC, minimum inhibitory concentration; MBC, minimum bactericidal concentration; FBS, fetal bovine serum; BSA, bovine serum albumin; BCG, Bacille Calmette-Guérin; CARA, Charcoal Agar Resazurin Assay; NARA, normal agar resazurin assay; NIAID, National Institute of Allergy and Infectious Diseases; NR, non-replicative; LCC, Loebel cidal concentration; LORA, Low-Oxygen Recovery Assay; WCC, Wayne cidal concentration; TBDA, Tuberculosis Drug Accelerator; TU, Tufts University; HMH, Hackensack Meridian Health; WCMC, Weill Cornell Medical College.

Feature selection of most informative in vitro assays

To reduce redundancy in the model, we carried out feature selection on all 31 assays, selecting those that had information on more than three drugs of interest, as well as those with a high number of pairwise correlations, which would be most representative of a large number of assays in the dataset (Figures 2B and 2C). A cutoff of >6 high pairwise correlations was selected, which gave us the upper 50% quantile of highly correlated assays. Table 2 lists each pairwise correlation for the assays with high numbers of pairwise correlations. Using pairwise correlation, we found 52 in vitro assay pairs with |R| >0.9, indicating a strong pairwise correlation. Four assays—caseum MBC, charcoal agar resazurin assay (CARA) MBC, low oxygen recovery assay (LORA) MIC, and intracellular potency assay in RAW264.7 macrophages—did not have high pairwise correlations with other assays and were therefore selected as informative features. Using network analysis, we found that a large cluster of highly correlated assays was formed with the remaining 25 assays. In this large cluster, eight assays with more than six pairwise correlations were chosen. The normal agar resazurin assay (NARA) MBC NR and the hypoxia-induced non-replication assay using the “Wayne cidal concentration” (WCC) as readout were also chosen, as they only had one high correlation with an assay in the large cluster (Figures 2B and 2C). In total, 14 in vitro assays were selected as features for further exploration.

Table 2.

In vitro assays with six or more high correlations (|R| > 0.9)

Assays with most high correlations Highly correlated assays Total number of assays
Butyrate Human Macrophage, Acidic, Tufts cholesterol, Tufts High cholesterol, Dormancy, Standard, NIAID cholesterol, 3 stress assay, Valerate 9
Tufts Cholesterol Human Macrophage, Acidic, Butyrate, Tufts High cholesterol, Dormancy, Standard, NIAID cholesterol, 3 stress assay, Valerate 9
Dormancy 4 stress NR MIC, Human Macrophage, Acidic, Butyrate, Tufts cholesterol, Standard, NIAID cholesterol, 3 stress assay, Valerate 9
NIAID Cholesterol 4 stress NR MIC, Human Macrophage, Butyrate, Tufts cholesterol, Dormancy, Standard, 3 stress assay, Valerate 8
Valerate Acidic, Butyrate, Tufts cholesterol, Tufts High cholesterol, Dormancy, J774, THP1 normoxia, NIAID cholesterol 8
Human Macrophage Butyrate, Tufts cholesterol, Dormancy, Standard, NARA MBC, NIAID cholesterol, 3 stress assay 7
Acidic Butyrate, Tufts cholesterol, Dormancy, J774, MBC, MIC, Valerate 7
3 Stress Assay 4 stress NR MIC, Human Macrophage, Butyrate, Tufts cholesterol, Dormancy, Standard, NIAID cholesterol 7
Standard Human Macrophage, Butyrate, Tufts cholesterol, Dormancy, NIAID cholesterol, 3 stress assay 6

Feature perturbation demonstrates that four or fewer in vitro assays can predict in vivo EC50 across mouse infection model types

Although data from three mouse models were available, we mainly discuss subacute mouse models in this text, as this was the mouse model previously used to validate our preclinical-to-clinical early bactericidal activity predictions.9 Acute and chronic mouse model results are described in Figures S1–S6.

Univariate analysis was first carried out to understand the individual relationships between in vitro EC50 and in vivo EC50 values (Figure S1). Likely due to the small size of the dataset, most univariate relationships were not statistically significant, leading us to perform feature perturbation instead.

Multivariate regression was unable to give good predictions using all 14 selected features due to overfitting of the training dataset. While the training dataset always gave an R2 of 0.99, the test drug EC50 was consistently predicted to be 100-fold or higher than its actual value. We therefore tried multinomial regression by classifying the in vivo EC50 values into four bins (very low, <0.1; low, 0.1–1; mid, 1–10; high, >10 mg/L). Although the test prediction improved slightly, it was still insufficient to get a reliable classification of the in vivo EC50. However, as we saw low-to-moderate correlations with in vivo mouse EC50 (Figure S1), there may have been multiple interacting features within the dataset, leading us to attempt feature perturbation (by creating all possible 1–5 combinations of in vitro assays based on the 14 assays from feature selection) to find the minimum number of features required to make a reliable prediction across two bins, low and high EC50. The model had a drop in accuracy from 100% to 62.5% when we further split EC50 predictions into all four bins, except for the NIAID cholesterol assay, which maintained its accuracy of 70% (Figure 3A). This allowed us to conclude that four or fewer assays were sufficient for the prediction of in vivo EC50.

Figure 3.

Figure 3

Feature perturbation identifies the best-performing combination of in vitro assays

(A) Feature perturbation was carried out to find the minimum number of features that could reliably classify in vivo mouse EC50 from in vitro potencies. All possible combinations of 1–5 in vitro assays were carried out.

(B) Test result of leave-one-out cross-validation performance in the training set with best-performing in vitro assay combinations. The lowest bin of <0.1 mg/L has an extended bin width to indicate that any drug with predicted EC50 < 0.1 mg/L will fall into that bin. BDQ, bedaquiline (BDQ), delamanid (DLM), ethambutol (EMB), isoniazid (INH), linezolid (LZD), moxifloxacin (MXF), pretomanid (PMD), pyrazinamide (PZA), rifampicin (RIF), and rifapentine (RPT).

We then compared the best-performing assay combinations against one another (Figure 3B). A four-assay combination of Human macrophage, Butyrate, Tufts cholesterol, and LORA MIC (62.5% accuracy), a three-assay combination of LORA MIC, RAW264.7, and valerate (62.5% accuracy), and single-assay NIAID high cholesterol (70% accuracy) were the top-performing assays. Across all the assays, the most difficult drug potencies to predict were for BDQ and LZD. LZD had a very different set of in vitro assays compared to other drugs in the same bin (DLM, PMD, and RIF), seen in Figure 2A. Here, LZD was on the extreme right of the clustered heatmap, whereas DLM, PMD, and RIF were clustered much more closely together, which could explain the difference in prediction. MFX, on the other hand, had an abnormally low in vivo EC50 of 0.0000586 mg/L (<0.1 bin), making it extremely difficult to predict compared to other drugs, as no other drugs had similar information. For BDQ, having an in vivo EC50 of 0.228 mg/L made it the only observed drug in the 0.1–1 bin. This led to all models having insufficient information for BDQ prediction in this analysis. In the next section’s external validation, however, we were able to predict new drugs in this 0.1–1 potency bin because the training set for that analysis included all eight drugs, including BDQ. Thus, we provide evidence that our top three models are reliable in making predictions for in vivo EC50. Our four-way in vitro assay combination of human macrophage, butyrate, BA cholesterol, and LORA MIC was preferentially chosen over top-performing single-assay combination NIAID high cholesterol based on the biological understanding of tuberculosis as a heterogeneous disease, making it crucial to incorporate multiple assays. Similar analyses for acute and chronic infection models are reported in Figure S3.

Selected in vitro assay combinations can reasonably predict in vivo EC50 of new drugs

As an external validation, we compiled a list of top-performing assays and reached out to collaborators to test new compounds with these assays (Figure 4A). The new compounds were sutezolid (SZD), TBAJ-587, and TBAJ-876 for the subacute mouse model, all of which are drugs currently being developed for the treatment of TB. Acute and chronic mouse models also had EC50 predictions for SZD, TBI223, and BTZ-043 and are reported in Figure S4. The top-performing acute model was able to predict TBI223, but not SZD, correctly, whereas the chronic model was able to predict BTZ-043 accurately. As all three top-performing combinations performed similarly, the four-way in vitro assay combination of foamy and hypoxic human macrophages (human macrophage), changing the main carbon source to butyrate (butyrate), changing the main carbon source to cholesterol (Tufts cholesterol), and LORA MIC was preferentially chosen over other top-performing assay combinations based on data availability for the new drugs. Using the model trained on all eight TB drugs with subacute infection models, we inputted newly generated in vitro data from three new drugs into their respective trained mouse infection models. Out of three new drugs used for validation, SZD, TBAJ-587, and TBAJ-876, only TBAJ-587 was predicted incorrectly (predicted in the 1–10 bin, whereas observed EC50 was in the 0.1–1 bin) (Figure 4B). The results for acute and chronic models are available in Figure S4, with similar results.

Figure 4.

Figure 4

External validation of the model with TB drug candidates

(A) Data availability of in vitro assays with five new TB drugs.

(B) Model performance across different in vitro assay combinations shows that some assay combinations are more generalizable to new drugs. Error bars represent the bin width of the predicted bin, and points align with the observed in vivo EC50 on the y axis and middle of the predicted bin on the x axis. The lowest bin of <0.1 mg/L has an extended bin width to indicate that any drug with predicted EC50 < 0.1 mg/L will fall into that bin. The training set consisted of the 10 drugs used to train the models, whereas the testing set consisted of new drugs used to validate the models. BDQ, bedaquiline, DLM, delamanid, EMB, ethambutol, INH, isoniazid, LZD, linezolid, MXF, moxifloxacin, PMD, pretomanid, PZA, pyrazinamide, RIF, rifampicin, RPT, rifapentine, and SZD, sutezolid.

Binned EC50 values integrated with mouse PK and the bacterial dynamics model gave good predictions of mouse CFU profiles

After predicting the in vivo EC50 from the best-performing combination of in vitro assays, we took the median of each bin containing the predicted in vivo EC50, along with the median Emax per mouse infection model as the exposure-response relationship in mice, in our integrated PK-PD model with bacterial dynamics. This allowed us to predict the colony-forming unit (CFU) drop in mice over time, over a range of drug doses for all eight training drugs in the subacute mouse model (Figure 5A). The 95% prediction intervals from the models overlapped with observed mouse CFU data, indicating that the predictions were accurate. The only exception was BDQ, whose efficacy was overpredicted in mice. Similarly, we were able to capture the mouse profiles for new drug candidates SZD, TBAJ-587, and TBAJ-876 as validation of the model (Figure 5B).

Figure 5.

Figure 5

Simulations versus observed mouse data using predicted mouse EC50

Five hundred simulations per drug per mouse infection model were run. Ribbons represent the 95% prediction interval and solid lines the median model prediction. Dotted lines are the median of observed values. Observed data are represented as points.

(A) Simulations with the initial eight drugs used for feature selection and model development.

(B) Simulations with three new drugs used as external validation demonstrate the extent to which our model was generalizable. The in vitro to in vivo EC50 prediction models selected for this simulation of mouse CFU are as listed in Table S1. Model performance across all mouse infection models is in Figure S5. BDQ, bedaquiline, DLM, delamanid, EMB, ethambutol, INH, isoniazid, LZD, linezolid, MXF, moxifloxacin, PMD, pretomanid, PZA, pyrazinamide, RIF, rifampicin, RPT, rifapentine, and SZD, sutezolid.

Predicted in vivo exposure-response informed the prediction of clinical EBA

Similarly, by combining clinical drug exposure with predicted exposure-response, we were able to get EBA predictions for all 10 drugs over 14 clinical trials (Figure 6). Our results were largely similar to preclinical mouse-to-human predictions, with most predictions overlapping well with the observed clinical data.9 Only BDQ had an overpredicted clinical EBA due to it being much more effective in a mouse model compared to clinical observations. However, our predicted in vivo exposure-response from in vitro data was able to capture the clinical observations more accurately, with higher overlap of the predictions and observations. This demonstrates that exposure-response relationships are translatable between systems.

Figure 6.

Figure 6

Prediction of early bactericidal activity outcomes in clinical populations

Simulations using clinical PK with the predicted exposure-response relationship show good fits with observed clinical trial data for the 10 training set drugs. Subacute model predictions were used for all models except for EMB, which used an acute model and RPT, which used a chronic model. BDQ, bedaquiline, DLM, delamanid, EMB, ethambutol, INH, isoniazid, LZD, linezolid, MXF, moxifloxacin, PMD, pretomanid, PZA, pyrazinamide, RIF, rifampicin, RPT, rifapentine. Observations are represented as mean +/− standard deviation, and simulations are represented as mean with 95% confidence intervals.

Discussion

We have systematically evaluated >30 in vitro assays used in TB drug development for their potential to predict in vivo outcome in mice and humans. Using in vivo EC50 as the primary outcome to identify which in vitro assays could predict mouse efficacy and best inform preclinical dosing, we were able to identify a set of four in vitro assays that could capture the in vivo potency well. This was then externally validated using three new drugs with good accuracy. When the predicted in vivo EC50 values were combined with our integrated bacterial dynamics PK-PD model, we were also able to capture the decrease in mouse CFU over time. This provides us with a framework for translating exposure-response relationships between different model systems, from in vitro to preclinical to clinical observations, which can guide dosing recommendations and prioritize drug candidates most likely to succeed in clinical trials.

In vitro assays are the least resource-intensive platform for gauging a compound’s potential in the discovery and development pipeline.29 However, in vitro drug potency should be coupled with drug exposure to fully capture the drivers of a drug’s in vivo efficacy. Thus, to translate these findings into interpretable biological outcomes, we combined predicted exposure-response relationships with either mouse or clinical PK data and identified a four-assay in vitro signature predictive of mouse bacterial burden upon treatment and short-term (Phase 2a) clinical outcomes. This is useful in prioritizing candidates for resource-intensive animal studies and suggesting reasonable first-in-human doses for clinical testing, provided they are safe.

We compiled data from a large panel of assays with differing complexities, from axenic culture in different types of culture media to macrophage infections and an ex vivo assay in rabbit caseum. Given that TB is a highly heterogeneous and dynamic disease over the course of infection,30,31,32 it is no surprise that multiple assays were required for the prediction of in vivo drug efficacy, especially with a mouse subacute model, where the adapted immune response has mounted, but bacteria are still actively replicating.33 Up to four in vitro assays describing key ecological niches were thus required.

Due to high correlation between in vitro assays, only a subset was required to describe the various pathophysiological states of Mtb. Both acute and subacute mouse infection models required more than one assay to have reliable prediction of in vivo EC50, suggesting that different assays describing different biological niches were required for in vivo EC50 prediction. Interestingly, the chronic mouse infection model was well described by the caseum assay alone, despite BALB/c mice not forming necrotic lesions.34,35 It is possible that after a long inoculation, the mouse lesions are still rich with foamy macrophages, whose lipid droplets are very similar in composition to caseum, found in the necrotic lesion core.36 This is consistent with ex vivo caseum as a single assay that reflects drug efficacy in vivo.14,36

We found that many in vitro assays where only the culture medium differed were highly correlated. However, none of these assays showed good correlation with the ex vivo caseum assay,14,36 which is currently understood as one of the most reflective of dormant Mtb bacilli found in hard-to-treat cavitary TB. This suggests that a single change in media formulation alone may not be sufficient to induce the same dormant state as seen within a necrotic TB lesion. Interestingly, the five macrophage assays were not strongly correlated to one another nor to the acidic in vitro assay meant to mimic the condition of Mtb surviving in the macrophage lysozyme, despite all macrophage assays expected to engulf Mtb into an acidic phagolysosome.37 This highlights that macrophage response in Mtb infection can be heterogeneous and/or differentially recapitulated in vitro, which has also been reported in other studies.38 We thus found that a multi-assay signature, rather than a single assay, could capture this physiological heterogeneity and predict in vivo EC50.

It was also interesting to note that despite both NIAID high cholesterol28 and Tufts high cholesterol16 having similar cholesterol concentrations (0.259 mM and 0.2 mM, respectively), the two assays had differences in media formulation and handling of the microbes after introduction to the culture prior to treatment initiation and reported different assay outcomes. This highlights that other biological factors might be at play in influencing Mtb and warrant further study.

To further test the utility of an in vitro to clinical EBA prediction, we similarly simulated clinical EBA predictions using the predicted in vivo EC50 alongside a clinical PK model and observed similarly good predictions to our translational mouse model predictions. Species-specific differences between mouse and human can exist, such as the case with BDQ, highlighting the potential utility of in vitro assays to overcome this limitation with those more tailored to human physiology.

Limitations of the study

Our initial dataset was limited in that we only had 8 drugs against a total of 14 features. We thus employed leave-one-out cross-validation, instead of the traditional 80:20 training-to-testing ratio for model validation.39 Trying to predict in vivo EC50 as a continuous variable using multiple linear regression led to substantial model overfitting in the training set and poor predictions on the left-out drug. Binning the in vivo EC50 into ranges rather than discrete values provided a good estimate of in vivo EC50 and allowed for reliable predictions to be made on the left-out testing drug. Other machine learning methods such as random forest were also tried with poor accuracy and robustness, leaving us to use simple multimodal regression. The median of the bin served as a sufficient estimate for mouse CFU prediction, with the prediction of mouse bacterial burden with drug treatment overlapping well with observed experimental data.

A few drugs did not have good predictions in mice due to their in vivo Emax deviating greatly from the median Emax of all other drugs. Due to multiple assays in our database reporting only EC50 and not Emax, we were unable to do a similar prediction for Emax, which is a limitation of this study. However, the sensitivity analysis showed that simulated mouse CFUs were mostly similar, whether median Emax or actual model-derived Emax was used (Figure S6).

Our dataset was biased toward drugs that had already been approved or are in clinical development for TB. This was a limitation due to the number of new compounds we were able to get data for. Access to a broader range of agents to validate the pipeline, including discovery compounds that failed preclinical development for lack of efficacy, would be useful to test the robustness of the model and its utility for decision-making and is being planned for future study. This study was limited to single drugs and short-term EBA. Due to the lack of information about the fraction unbound of the drugs in each assay, we were unable to correct for fraction unbound or dynamic free fraction, which may help to improve the predictions. Further work is ongoing to predict the long-term efficacy of drug combinations and investigate the role of the fraction unbound and dynamic free fraction in prioritized assays.

Furthermore, although we could have generated efficacy predictions for single drugs, we wanted to ensure that the most predictive assays across drug classes and mechanisms of action should be prioritized as most informative assays. This study has shown that such in vitro to in vivo translations are promising, and further efforts to expand this database and model validation would be pertinent as novel compounds are developed.

Resource availability

Lead contact

Further information and requests for resources and reagents should be directed to, and will be fulfilled by, the lead contact Rada Savic (Rada.savic@ucsf.edu).

Materials availability

This study did not generate new unique reagents.

Data and code availability

  • Data: the in vitro data used in this study are not made available due to confidentiality reasons but is available upon reasonable request from the authors with the approval of the individual asset holders.

  • Code: all equations used to build models are listed in the supplementary section of this paper. The pipeline to select clinically relevant assays from mouse EC50 is located under https://github.com/JaniceGoh93/MTB_IVIVC. Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

Acknowledgments

We wish to thank Carl Nathan, Benjamin Gold, and Helena Boshoff for sharing raw potency data. We would also like to thank Jacqueline Ernest, Belén Perez Solans, Annamarie Bustion, and Linda Chaba for their feedback and discussions on this manuscript.

This work was funded by award INV-002483 from the Bill and Melinda Gates Foundation to V.D. and R.S., NIH/NIAID UM1 AI179699 from the Preclinical Design and Clinical Translation of TB Regimens (PReDiCTR) Consortium, and INV-027276 from the Bill and Melinda Gates Foundation to B.B.A. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Author contributions

Conceptualization, J.J.N.G. and Q.W.W.; methodology and software, J.J.N.G.; formal analysis, J.J.N.G., A.P., B.N., R.C.W., N.S., and Q.W.W.; investigation and resources, J.J.N.G., N.V., T.M.W., V.D., J.S., B.B.A., and E.L.N.; data curation, J.J.N.G. and C.R.; writing—original draft, J.J.N.G. and V.D.; writing—review and editing, J.J.N.G., A.P., R.C.W., and V.D.; visualization, J.J.N.G.; supervision, R.M.S.; project administration, R.M.S.; funding acquisition, R.M.S., V.D., and B.B.A.

Declaration of interests

The authors declare no conflicts of interest.

STAR★Methods

Key resources table

REAGENT or RESOURCE SOURCE IDENTIFIER
Software and algorithms

NONMEM 7.5.1 ICON Plc. https://www.iconplc.com/solutions/technologies/nonmem
R 4.1.3. The Comprehensive R Archive Network https://cran.r-project.org/
Tidyverse posit https://www.tidyverse.org/
Pheatmap Raivo Kolde https://cran.r-project.org/web/packages/pheatmap/pheatmap.pdf
Nnet Brian Ripley https://cran.r-project.org/web/packages/nnet/nnet.pdf
Cor Guangbao Guo https://doi.org/10.32614/CRAN.package.COR

Deposited data

In vitro assays Multiple labs Details and references in Table 1
Mouse PK and PD data Multiple labs Details and references in Table S1

Experimental model and study participant details

Compiling a rich dataset of in vitro data

An initial set of drugs was selected based on the criteria that they had a corresponding integrated bacterial dynamics PK-PD model built across 3 different mouse infection models, acute, subacute, and chronic, and a minimum of 5 reported in vitro assay results. Detailed description of PK-PD model methods are described in Methods S1. For the sake of brevity, we focused on the subacute infection model, which was previously used to predict clinical Phase 2a outcomes. The same analyses were also carried out in acute and chronic mouse infection models, with their results reported in the supplemental information.

In vitro assays and their reported drug potencies (i.e., EC50) were compiled both from published literature and from collaborators (Table 1). To ensure fair comparison across drug potencies, all drug concentrations were standardized to mg/L and different measure outcomes (e.g., EC50, EC80, EC99) were converted to EC50 using the assumption that the Hill coefficient in a log-logistic dose-response curve was 1. After normalizing all values and doses across assays and drugs, curve fittings using both models with and without an estimated Hill coefficient were tested as a nested F test. When the percent activity was not stated for MIC, we assumed it to be EC90.

Building a translational mouse-to-human PK-PD dataset and model repository

Our model repository was built on rich longitudinal data pooled from available databases, including observed or published mouse PK data and mouse PD data (lung CFU counts) and human PD data (sputum CFU counts), as well as simulated human PK data using models as previously published.40 Multiple dose levels were investigated in both mouse PK and efficacy (PD) studies and human PK and EBA (PD) studies. All drugs were administered orally in mouse and human PK and PD studies. Mouse plasma samples were collected after either single or multiple doses of treatment, while human plasma samples were collected only after multiple doses.

Depending on the size of the infectious dose and the duration of incubation before treatment started, mouse efficacy studies were grouped as acute infection (inoculum size no less than 3.5 log10CFU/mL and incubation period no more than 8 days), subacute infection (inoculum size no less than 3.5 log10CFU/mL and incubation period between 10 and 17 days), and chronic infection studies (inoculum size less than 3.5 log10CFU/mL and incubation period no less than 21 days). Mice were dosed 5 days per week and CFU counts were collected 3 days after the last dose for any given mouse. In human EBA studies, the treatment duration ranged between 1 and 14 days. Integrated PK-PD models with bacterial dynamics were applied to the mouse data to elucidate the in vivo exposure-response using NONMEM 7.5.1 and Perl-Speaks-NONMEM (PsN) 5.3.0. Detailed methods on model building can be found in Ernest et al.9

Quantification and statistical analysis

Exploratory data analysis and determination of translatable assays

To first assess data availability and spread of EC50 values across different assays, initial exploratory analyses were done using tidyverse, pheatmap, drc, nnet, and cor R packages using R 4.1.3. Feature selection was carried out based on the following criteria (Figure 2): 1) the in vitro assay contained information for at least 3 drugs of interest, and 2) between highly correlated assays, the most representative assay with the most pairwise correlations was chosen.

While initial clustering and data exploration were done in μM, we decided to do the binning and predictions of EC50 in mg/L, as that is the concentration that is commonly reported for PK studies, and we wanted to be as consistent as possible with published models. Because the main driver of the drug’s effect is its exposure characterized as mg/L over time, rather than its potency directly, the predictions in mg/L are more translatable for directly adding into a PK-PD model without additional unit conversions.

In vitro assays were first evaluated individually using univariate linear regression. Models to predict in vivo mouse EC50 were built individually for each mouse infection type as the in vivo EC50 trends changed between infection models for the same drug. As the potency of inactive compounds was unknown, inactive compounds were excluded from the univariate analysis.

Feature selection was first carried out by performing pairwise correlations using Pearson’s correlation between all in vitro assays. Assays with no high pairwise correlations were selected as features, and from groups of assays with high pairwise correlations (R>0.9), one representative assay with the most correlations was chosen.

Feature perturbation to test which combination of in vitro assays was most informative using multinomial linear regression was then applied to the dataset to predict in vivo EC50 from in vitro EC50. All possible 1-5-way combinations of in vitro assays were generated for the evaluation of predictive in vitro assay combinations. As initial analyses found multiple linear regression to have large model overfitting, we applied multinomial regression instead, using bins of very low (<0.1 mg/L), low (0.1-1 mg/L), mid (1-10 mg/L), and high (>10 mg/L) concentration ranges to classify EC50 values. Due to the small size of the dataset, we applied leave-one-out cross-validation to evaluate model accuracy instead of K-fold validation. Accuracy was defined as the number of times the left-out drug was predicted in the correct bin for multinomial linear regression. For drugs with missing assay values, the assay values were imputed as the median potency of all other drugs within the same in vitro assay.41

Combining PK models from mice with in vitro-in vivo (IVIV)-predicted exposure-response

To find out how well IVIV-predicted exposure-response mapped to drug efficacy in mice, we substituted predicted EC50 values into previously validated mouse PK models. The Emax in each exposure-response curve was estimated as the median Emax of all 10 drugs for each mouse infection model. This was done as most in vitro assays reported only a single EC50 value rather than concentration-response curves. A sensitivity analysis comparing simulations done using either the actual PK-PD model’s Emax or median Emax values are detailed in Figure S6. Simulations of bacterial burden (CFU) in mice over time were done using both the raw EC50 values for selected in vitro assays as well as the median of the binned EC50 values as predicted by the multinomial regression algorithm. Typical bacterial dynamic parameters were used as detailed in Zhang et al.33 The PK-PD simulations were done using NONMEM 7.5.1. Variability was simulated using variance in CFU at day 1 post inoculation in mice for acute, subacute, and chronic mouse infection models respectively, before treatment. Model predictions were evaluated using visual predictive checks (VPCs), by overlaying the median and 95% prediction interval of the model over observed results from mouse studies. Models were considered appropriate when prediction intervals overlapped well with the observed data. Equations and a detailed methodology are described in supplementary methods.

Combining clinical PK models with IVIV-predicted exposure response

To simulate clinical EBA, the predicted in vivo exposure-response relationship was combined with previously validated population PK models from literature to build a PK-PD model that was then used for simulation.9 Again, our models were evaluated using VPCs against observed clinical EBA data from 14 different studies for all 10 drugs. The subacute model results were used for all drugs except for EMB, which used acute model results, and RPT, which used chronic model results, as these drugs did not have subacute mouse data. Details and references of the clinical EBA studies are in Table S2.

External model validation with a set of new TB drugs

To externally validate the model, we compiled a list of the best-performing combinations of in vitro assays from initial multinomial classification and reached out to the same labs that made these assays to test 5 new compounds. We then input the in vitro EC50 values into the multinomial classifier trained using the original model to predict mouse EC50 for these new compounds. Detailed information on the mouse PD data used for validation and the mouse PK models used in the prediction are listed in Table S1.

Published: January 30, 2025

Footnotes

Supplemental information can be found online at https://doi.org/10.1016/j.isci.2025.111932.

Supplemental information

Document S1. Figures S1–S6, Tables S1 and S2, and Methods S1
mmc1.pdf (2MB, pdf)

References

  • 1.Global Tuberculosis Report. 2022. https://www.who.int/teams/global-tuberculosis-programme/tb-reports/global-tuberculosis-report-2022
  • 2.Nahid P., Dorman S.E., Alipanah N., Barry P.M., Brozek J.L., Cattamanchi A., Chaisson L.H., Chaisson R.E., Daley C.L., Grzemska M., et al. Executive Summary: Official American Thoracic Society/Centers for Disease Control and Prevention/Infectious Diseases Society of America Clinical Practice Guidelines: Treatment of Drug-Susceptible Tuberculosis. Clin. Infect. Dis. 2016;63:853–867. doi: 10.1093/cid/ciw566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Vernon A., Fielding K., Savic R., Dodd L., Nahid P. The importance of adherence in tuberculosis treatment clinical trials and its relevance in explanatory and pragmatic trials. PLoS Med. 2019;16 doi: 10.1371/journal.pmed.1002884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Dartois V.A., Rubin E.J. Anti-tuberculosis treatment strategies and drug development: challenges and priorities. Nat. Rev. Microbiol. 2022;20:685–701. doi: 10.1038/s41579-022-00731-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Franzblau S.G., Witzig R.S., McLaughlin J.C., Torres P., Madico G., Hernandez A., Degnan M.T., Cook M.B., Quenzer V.K., Ferguson R.M., Gilman R.H. Rapid, low-technology MIC determination with clinical Mycobacterium tuberculosis isolates by using the microplate Alamar Blue assay. J. Clin. Microbiol. 1998;36:362–366. doi: 10.1128/jcm.36.2.362-366.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Connolly L.E., Edelstein P.H., Ramakrishnan L. Why is long-term therapy required to cure tuberculosis? PLoS Med. 2007;4:e120. doi: 10.1371/journal.pmed.0040120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ke H., Gui X., Sun W., Zhang S., Yang Y., Zhang Z., Fan L. The Safety and Efficacy of Prolonged Use of Bedaquiline for the Treatment of Patients with Pulmonary Multi-Drug Resistant/Rifampin-Resistant Tuberculosis: A Prospective, Cohort Study in China. Infect. Drug Resist. 2023;16:5055–5064. doi: 10.2147/IDR.S419996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Zhang Y., Yew W.W., Barer M.R. Targeting persisters for tuberculosis control. Antimicrob. Agents Chemother. 2012;56:2223–2230. doi: 10.1128/AAC.06288-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Ernest J.P., Goh J.J.N., Strydom N., Wang Q., van Wijk R.C., Zhang N., Deitchman A., Nuermberger E., Savic R.M. Translational predictions of phase 2a first-in-patient efficacy studies for antituberculosis drugs. Eur. Respir. J. 2023;62 doi: 10.1183/13993003.00165-2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Upton A.M., Cho S., Yang T.J., Kim Y., Wang Y., Lu Y., Wang B., Xu J., Mdluli K., Ma Z., Franzblau S.G. In vitro and in vivo activities of the nitroimidazole TBA-354 against Mycobacterium tuberculosis. Antimicrob. Agents Chemother. 2015;59:136–144. doi: 10.1128/AAC.03823-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Early J.V., Mullen S., Parish T. A rapid, low pH, nutrient stress, assay to determine the bactericidal activity of compounds against non-replicating Mycobacterium tuberculosis. PLoS One. 2019;14 doi: 10.1371/journal.pone.0222970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Gold B., Roberts J., Ling Y., Quezada L.L., Glasheen J., Ballinger E., Somersan-Karakaya S., Warrier T., Warren J.D., Nathan C. Rapid, Semiquantitative Assay To Discriminate among Compounds with Activity against Replicating or Nonreplicating Mycobacterium tuberculosis. Antimicrob. Agents Chemother. 2015;59:6521–6538. doi: 10.1128/AAC.00803-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Lopez Quezada L., Li K., McDonald S.L., Nguyen Q., Perkowski A.J., Pharr C.W., Gold B., Roberts J., McAulay K., Saito K., et al. Dual-pharmacophore pyrithione-containing cephalosporins kill both replicating and nonreplicating Mycobacterium tuberculosis. ACS Infect. Dis. 2019;5:1433–1445. doi: 10.1021/acsinfecdis.9b00112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Sarathy J.P., Via L.E., Weiner D., Blanc L., Boshoff H., Eugenin E.A., Barry C.E., 3rd, Dartois V.A. Extreme Drug Tolerance of Mycobacterium tuberculosis in Caseum. Antimicrob. Agents Chemother. 2018;62 doi: 10.1128/AAC.02266-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Lanni F., Wijnant G.J., Xie M., Osiecki P., Dartois V., Sarathy J.P. Adaptation to the intracellular environment of primary human macrophages influences drug susceptibility of Mycobacterium tuberculosis. Tuberculosis. 2023;139 doi: 10.1016/j.tube.2023.102318. [DOI] [PubMed] [Google Scholar]
  • 16.Larkins-Ford J., Greenstein T., Van N., Degefu Y.N., Olson M.C., Sokolov A., Aldridge B.B. Systematic measurement of combination-drug landscapes to predict in vivo treatment outcomes for tuberculosis. Cell Syst. 2021;12:1046–1063.e7. doi: 10.1016/j.cels.2021.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Souriant S. 2017. Role of macrophages during HIV-1 infection and in the context of co-infection with Mycobacterium tuberculosis.https://theses.fr/2017TOU30209?domaine=theses [Google Scholar]
  • 18.Bade P., Simonetti F., Sans S., Laboudie P., Kissane K., Chappat N., Lagrange S., Apparailly F., Roubert C., Duroux-Richard I. Integrative Analysis of Human Macrophage Inflammatory Response Related to Mycobacterium tuberculosis Virulence. Front. Immunol. 2021;12 doi: 10.3389/fimmu.2021.668060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Lakshminarayana S.B., Huat T.B., Ho P.C., Manjunatha U.H., Dartois V., Dick T., Rao S.P.S. Comprehensive physicochemical, pharmacokinetic and activity profiling of anti-TB agents. J. Antimicrob. Chemother. 2015;70:857–867. doi: 10.1093/jac/dku457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Sarathy J., Dartois V., Dick T., Gengenbacher M. Reduced drug uptake in phenotypically resistant nutrient-starved nonreplicating Mycobacterium tuberculosis. Antimicrob. Agents Chemother. 2013;57:1648–1653. doi: 10.1128/AAC.02202-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Cho S.H., Warit S., Wan B., Hwang C.H., Pauli G.F., Franzblau S.G. Low-oxygen-recovery assay for high-throughput screening of compounds against nonreplicating Mycobacterium tuberculosis. Antimicrob. Agents Chemother. 2007;51:1380–1385. doi: 10.1128/AAC.00055-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Koul A., Vranckx L., Dendouga N., Balemans W., Van den Wyngaert I., Vergauwen K., Göhlmann H.W.H., Willebrords R., Poncelet A., Guillemont J., et al. Diarylquinolines are bactericidal for dormant mycobacteria as a result of disturbed ATP homeostasis. J. Biol. Chem. 2008;283:25273–25280. doi: 10.1074/jbc.M803899200. [DOI] [PubMed] [Google Scholar]
  • 23.Xie Z., Siddiqi N., Rubin E.J. Differential antibiotic susceptibilities of starved Mycobacterium tuberculosis isolates. Antimicrob. Agents Chemother. 2005;49:4778–4780. doi: 10.1128/AAC.49.11.4778-4780.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.de Miranda Silva C., Hajihosseini A., Myrick J., Nole J., Louie A., Schmidt S., Drusano G.L. Effect of Linezolid plus Bedaquiline against Mycobacterium tuberculosis in Log Phase, Acid Phase, and Nonreplicating-Persister Phase in an In Vitro Assay. Antimicrob. Agents Chemother. 2018;62 doi: 10.1128/AAC.00856-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Kim S., Yamada W.M., Duncanson B., Nole J., Rogers S., Parker S., Bacci M., Mtchedlidze N., Peloquin C.A., Louie A., et al. Building Optimal Three-Drug Combination Chemotherapy Regimens To Eradicate Mycobacterium tuberculosis in Its Slow-Growth Acid Phase. Antimicrob. Agents Chemother. 2021;65 doi: 10.1128/AAC.00693-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Drusano G.L., Myrick J., Maynard M., Nole J., Duncanson B., Brown D., Schmidt S., Neely M., Scanga C.A., Peloquin C., et al. Linezolid Kills Acid-Phase and Nonreplicative-Persister-Phase Mycobacterium tuberculosis in a Hollow-Fiber Infection Model. Antimicrob. Agents Chemother. 2018;62 doi: 10.1128/AAC.00221-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Zhang J., Lair C., Roubert C., Amaning K., Barrio M.B., Benedetti Y., Cui Z., Xing Z., Li X., Franzblau S.G., et al. Discovery of natural-product-derived sequanamycins as potent oral anti-tuberculosis agents. Cell. 2023;186:1013–1025.e24. doi: 10.1016/j.cell.2023.01.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Nuermberger E.L., Martínez-Martínez M.S., Sanz O., Urones B., Esquivias J., Soni H., Tasneen R., Tyagi S., Li S.Y., Converse P.J., et al. GSK2556286 Is a Novel Antitubercular Drug Candidate Effective In Vivo with the Potential To Shorten Tuberculosis Treatment. Antimicrob. Agents Chemother. 2022;66 doi: 10.1128/aac.00132-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Shetye G.S., Choi K.B., Kim C.-Y., Franzblau S.G., Cho S. In Vitro Profiling of Antitubercular Compounds by Rapid, Efficient, and Nondestructive Assays Using Autoluminescent Mycobacterium tuberculosis. Antimicrob. Agents Chemother. 2021;65 doi: 10.1128/AAC.00282-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Lenaerts A., Barry C.E., 3rd, Dartois V. Heterogeneity in tuberculosis pathology, microenvironments and therapeutic responses. Immunol. Rev. 2015;264:288–307. doi: 10.1111/imr.12252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Cadena A.M., Fortune S.M., Flynn J.L. Heterogeneity in tuberculosis. Nat. Rev. Immunol. 2017;17:691–702. doi: 10.1038/nri.2017.69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Chung E.S., Johnson W.C., Aldridge B.B. Types and functions of heterogeneity in mycobacteria. Nat. Rev. Microbiol. 2022;20:529–541. doi: 10.1038/s41579-022-00721-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Zhang N., Strydom N., Tyagi S., Soni H., Tasneen R., Nuermberger E.L., Savic R.M. Mechanistic Modeling of Mycobacterium tuberculosis Infection in Murine Models for Drug and Vaccine Efficacy Studies. Antimicrob. Agents Chemother. 2020;64 doi: 10.1128/AAC.01727-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Keller C., Lauber J., Blumenthal A., Buer J., Ehlers S. Resistance and susceptibility to tuberculosis analysed at the transcriptome level: lessons from mouse macrophages. Tuberculosis. 2004;84:144–158. doi: 10.1016/j.tube.2003.12.003. [DOI] [PubMed] [Google Scholar]
  • 35.Kramnik I., Dietrich W.F., Demant P., Bloom B.R. Genetic control of resistance to experimental infection with virulent Mycobacterium tuberculosis. Proc. Natl. Acad. Sci. USA. 2000;97:8560–8565. doi: 10.1073/pnas.150227197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Sarathy J.P., Dartois V. Caseum: a Niche for Mycobacterium tuberculosis Drug-Tolerant Persisters. Clin. Microbiol. Rev. 2020;33 doi: 10.1128/CMR.00159-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Vandal O.H., Nathan C.F., Ehrt S. Acid resistance in Mycobacterium tuberculosis. J. Bacteriol. 2009;191:4714–4721. doi: 10.1128/JB.00305-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Marakalala M.J., Martinez F.O., Plüddemann A., Gordon S. Macrophage Heterogeneity in the Immunopathogenesis of Tuberculosis. Front. Microbiol. 2018;9:1028. doi: 10.3389/fmicb.2018.01028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sammut C., Webb G.I., editors. Encyclopedia of Machine Learning. Springer US; Boston, MA: 2010. Leave-One-Out Cross-Validation; pp. 600–601. [Google Scholar]
  • 40.Ernest J.P., Ni Goh J.J., Strydom N., Wang Q., van Wijk R.C., Zhang N., Deitchman A., Nuermberger E., Savic R.M. Translational predictions of phase 2a first-in-patient efficacy studies for antituberculosis drugs. bioRxiv. 2023 doi: 10.1101/2023.01.18.524608. Preprint at. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Donders A.R.T., van der Heijden G.J.M.G., Stijnen T., Moons K.G.M. Review: a gentle introduction to imputation of missing values. J. Clin. Epidemiol. 2006;59:1087–1091. doi: 10.1016/j.jclinepi.2006.01.014. [DOI] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

Document S1. Figures S1–S6, Tables S1 and S2, and Methods S1
mmc1.pdf (2MB, pdf)

Data Availability Statement

  • Data: the in vitro data used in this study are not made available due to confidentiality reasons but is available upon reasonable request from the authors with the approval of the individual asset holders.

  • Code: all equations used to build models are listed in the supplementary section of this paper. The pipeline to select clinically relevant assays from mouse EC50 is located under https://github.com/JaniceGoh93/MTB_IVIVC. Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.


Articles from iScience are provided here courtesy of Elsevier

RESOURCES