Skip to main content
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Eukaryotic Cell
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems
  • Log in
  • My alerts
  • My Cart

Main menu

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • COVID-19 Special Collection
    • Archive
    • Minireviews
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About AAC
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • AAC Podcast
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Eukaryotic Cell
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems

User menu

  • Log in
  • My alerts
  • My Cart

Search

  • Advanced search
Antimicrobial Agents and Chemotherapy
publisher-logosite-logo

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • COVID-19 Special Collection
    • Archive
    • Minireviews
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About AAC
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • AAC Podcast
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
Pharmacology

Cumulative Fraction of Response for Once- and Twice-Daily Delamanid in Patients with Pulmonary Multidrug-Resistant Tuberculosis

Suresh Mallikaarjun, Moti L. Chapagain, Tomohiro Sasaki, Norimitsu Hariguchi, Devyani Deshpande, Shashikant Srivastava, Alexander Berg, Kuniko Hirota, Yusuke Inoue, Makoto Matsumoto, Jeffrey Hafkin, Lawrence Geiter, Xiaofeng Wang, Tawanda Gumbo, Yongge Liu
Suresh Mallikaarjun
aOtsuka Pharmaceutical Development & Commercialization, Inc., Rockville, Maryland, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Moti L. Chapagain
bBaylor Research Institute, Dallas, Texas, USA
cPraedicare, Dallas, Texas, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Tomohiro Sasaki
dOtsuka Pharmaceutical Co., Ltd., Tokushima, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Norimitsu Hariguchi
dOtsuka Pharmaceutical Co., Ltd., Tokushima, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Devyani Deshpande
bBaylor Research Institute, Dallas, Texas, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Shashikant Srivastava
bBaylor Research Institute, Dallas, Texas, USA
eUniversity of Texas Health Science Center, Tyler, Texas, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Shashikant Srivastava
Alexander Berg
fCritical Path Institute, Tucson, Arizona, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kuniko Hirota
dOtsuka Pharmaceutical Co., Ltd., Tokushima, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Yusuke Inoue
dOtsuka Pharmaceutical Co., Ltd., Tokushima, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Makoto Matsumoto
dOtsuka Pharmaceutical Co., Ltd., Tokushima, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jeffrey Hafkin
aOtsuka Pharmaceutical Development & Commercialization, Inc., Rockville, Maryland, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lawrence Geiter
aOtsuka Pharmaceutical Development & Commercialization, Inc., Rockville, Maryland, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Xiaofeng Wang
aOtsuka Pharmaceutical Development & Commercialization, Inc., Rockville, Maryland, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Xiaofeng Wang
Tawanda Gumbo
bBaylor Research Institute, Dallas, Texas, USA
cPraedicare, Dallas, Texas, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Tawanda Gumbo
Yongge Liu
aOtsuka Pharmaceutical Development & Commercialization, Inc., Rockville, Maryland, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Yongge Liu
DOI: 10.1128/AAC.01207-20
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

ABSTRACT

Pharmacokinetic (PK) and pharmacodynamic (PD) analyses were conducted to determine the cumulative fraction of response (CFR) for 100 mg twice-daily (BID) and 200 mg once-daily (QD) delamanid in patients with multidrug-resistant tuberculosis (MDR-TB), using a pharmacodynamic target (PDT) that achieves 80% of maximum efficacy. First, in the mouse model of chronic TB, the PK/PD index for delamanid efficacy was determined to be area under the drug concentration-time curve over 24 h divided by MIC (AUC0–24/MIC), with a PDT of 252. Second, in the hollow-fiber system model of tuberculosis, plasma-equivalent PDTs were identified as an AUC0–24/MIC of 195 in log-phase bacteria and 201 in pH 5.8 cultures. Third, delamanid plasma AUC0–24/MIC and sputum bacterial decline data from two early bactericidal activity trials identified a clinical PDT of AUC0–24/MIC of 171. Finally, the CFRs for the currently approved 100-mg BID dose were determined to be above 95% in two MDR-TB clinical trials. The CFR for the 200-mg QD dose, evaluated in a trial in which delamanid was administered as 100 mg BID for 8 weeks plus 200 mg QD for 18 weeks, was 89.3% based on the mouse PDT and >90% on the other PDTs. QTcF (QTc interval corrected for heart rate by Fridericia’s formula) prolongation was approximately 50% lower for the 200 mg QD dose than the 100 mg BID dose. In conclusion, while CFRs of 100 mg BID and 200 mg QD delamanid were close to or above 90% in patients with MDR-TB, more-convenient once-daily dosing of delamanid is feasible and likely to have less effect on QTcF prolongation.

TEXT

Worldwide, approximately 10 million people developed tuberculosis (TB) and 1.45 million (including those coinfected with HIV) died of the disease in 2018, making it the leading cause of mortality from a single infectious agent and one of the top 10 causes of death overall (1). While drug-susceptible TB has a high cure rate following a standard 6-month regimen of antibiotics in controlled settings, global treatment success rates are 85% in routine patient care and, thus, are still not optimal (1). Management of multidrug-resistant TB (MDR-TB) is even more challenging. Accounting for approximately 460,000 cases per year worldwide, MDR-TB requires longer therapy with agents that are less effective (current rate of treatment success, 56%) and substantially more toxic (1). Additionally, resistance to either second-line injectables or fluoroquinolones (pre-extensively drug-resistant TB [pre-XDR-TB]) or to both (XDR-TB) reduces the success rate to a mere 39% (1). Thus, new therapies for effective and safe treatment of MDR-TB and XDR-TB are urgently needed.

Delamanid (Deltyba; Otsuka Pharmaceutical Co., Ltd., Tokyo, Japan) is a bicyclic nitroimidazooxazole compound that inhibits the synthesis of mycolic acids (2), key components of the lipid-rich cell wall of Mycobacterium tuberculosis (3). In preclinical studies, delamanid exhibited the lowest MIC among approved TB drugs against both drug-susceptible and drug-resistant isolates of M. tuberculosis (4) and demonstrated dose-dependent bactericidal effects in mice (2). In humans, two randomized, placebo-controlled trials assessed the efficacy and safety of delamanid plus an optimized background regimen (OBR) in the treatment of MDR-TB. In one study (trial 204; Table S5), patients who received delamanid with OBR had significantly higher sputum culture conversion (SCC) proportions after 2 months of therapy than those receiving placebo with OBR (45.4% versus 29.6%; P = 0.0083) (5). In the second study (trial 213; Table S5), median time to SCC was numerically shorter in the delamanid/OBR arm (51 days) than in the placebo/OBR arm (57 days), although the difference did not reach the significance level of 0.05 (P = 0.0562 for the comparison) (6). As of August 2020, delamanid is approved in 15 countries, including the European Union and Japan, as well as several high-TB-burden countries, including China, India, Indonesia, Peru, the Philippines, the Russian Federation, South Africa, and Ukraine, as part of an appropriate combination regimen for treatment of pulmonary MDR-TB in adults, when an effective treatment regimen cannot otherwise be composed for reasons of resistance or tolerability (7). The World Health Organization (WHO) has also recommended that delamanid be added to the WHO-recommended longer regimen in children and adolescents (6 to 17 years of age) with multidrug- or rifampin-resistant TB who are not eligible for the shorter MDR-TB regimen, under specific conditions (8).

For the treatment of infectious diseases, including TB, antibiotics should be dosed at concentrations that balance efficacy and safety. Currently, delamanid is approved at a dose of 100 mg twice daily (BID) (7) based on data from a number of clinical trials (5, 6, 9–11). A treatment regimen consisting of once-daily delamanid plus OBR has not been examined in isolation in clinical trials, but patients in trial 213 did receive 18 weeks of 200-mg once-daily (QD) dosing after 8 weeks of prior treatment with delamanid 100 mg delamanid twice a day (BID) (Table S5) (6). The present study was designed to determine the cumulative fraction of responses (CFRs) for 100 mg delamanid BID and 200 mg delamanid QD to compare the optimality of these two dosing regimens, thereby addressing the feasibility of once-daily administration of delamanid. Given the complexity of treatment regimens for MDR-TB, reducing the frequency of dosing via a once-daily regimen could have significant benefits for patient compliance, which in turn would be expected to improve outcomes for this difficult-to-treat infection.

RESULTS

PK/PD index and PK/PD target for delamanid in mice.Pharmacokinetic (PK) parameters after the single administration of delamanid at doses of 0.625, 2.5, or 10 mg/kg in uninfected mice are shown in Table S1 for both plasma and lung tissue. Delamanid has a Kp (ratio of total concentration in tissue to that in plasma) value of 1.9 to 3 depending on the dose administered. Using a nonparametric superposition method, the single-dose plasma data were then used to estimate the plasma PK parameters after multiple-dose regimens, as shown in Table 1, that were used in an efficacy study (see below). The simulated PK profiles at the 4th week of a 4-week treatment for each individual regimen are shown in Fig. S2.

View this table:
  • View inline
  • View popup
  • Download powerpoint
TABLE 1

Simulated PK parameters and observed bactericidal effects of various delamanid dosing regimens in M. tuberculosis Kurono-infected micea

All tested delamanid dosing schedules significantly reduced bacterial burden as measured by log10 CFU/lung compared to untreated controls, with a minimum of 1.672 log10 CFU/lung reduction by the 2.5 mg/kg once-weekly treatment and a maximum of 2.994 log10 CFU/lung reduction by the 10-mg/kg once-daily treatment (Table 1). As shown in Table 2 and Fig. 1, the inhibitory sigmoid maximum-effect (Emax) model with a fixed Hill coefficient of 1.0 fitted the data well. The Emax of delamanid in mice was estimated as a 2.96 log10 CFU/lung reduction (95% confidence interval [CI], 2.63 to 3.29). The ratio of area under the concentration-time curve from 0 to 24 h to MIC (AUC0–24/MIC) was the pharmacokinetic-pharmacodynamic (PK/PD) parameter that best described delamanid efficacy (Pearson’s correlation coefficient = 0.97; P < 0.0001), while the percentage of time above the MIC (%T>MIC) exhibited moderate correlation (Spearman correlation coefficient = 0.53; P < 0.01). There was no significant correlation between efficacy and Cmax/MIC by Pearson’s correlation (P = 0.13). Additionally, inhibitory sigmoid Emax modeling of log10 CFU/lung reduction versus PK/PD exposure revealed corrected Akaike information criterion scores of 2.55 (r2 = 0.99), 21.03 (r2 = 0.86), and 22.07 (r2 = 0.84) for AUC0–24/MIC, %T>MIC, and Cmax/MIC, respectively. This confirmed that AUC0–24/MIC was the parameter that best explained the delamanid antimicrobial effect. Therefore, the key PK/PD index driving delamanid efficacy was determined to be AUC0–24/MIC.

View this table:
  • View inline
  • View popup
  • Download powerpoint
TABLE 2

Inhibitory sigmoid Emax parameters for delamanid AUC0–24/MIC from mouse and HFS studies and human EBA trialsa

FIG 1
  • Open in new tab
  • Download powerpoint
FIG 1

Log10 CFU/lung reduction by AUC0–24/MIC in mice. Each circle represents data from an individual mouse. The reduction was the difference between log10 CFU per lung in the treated mouse and the average value for the untreated mice. Multiple-dose AUC0–24 was simulated from single-dose PK data. EC50, EC80, and EC90 represent the AUC0–24/MIC to achieve 50%, 80%, and 90% of the maximum efficacy, respectively.

We then determined the pharmacodynamic target (PDT) for AUC0–24/MIC, using the AUC0–24 value that achieved 80% of the Emax, a cutoff that was used in previously published studies (12–16) and validated through the translation of HFS-TB data to clinical use for various TB drugs (as detailed in the Discussion). The PDT was an AUC0–24/MIC of 252 (95% CI, 139 to 649) (a plasma AUC0–24 of 3.021 mg · h/liter divided by an MIC of 0.012 mg/liter for the M. tuberculosis strain used in the study), which achieved a 2.36 log10 CFU/lung reduction from the untreated control.

PDT for delamanid from HFS-TB.The delamanid MIC of strain H37Rv, which was used in the study of the hollow-fiber system model for TB (HFS-TB), was determined to be 0.015 mg/liter. Maximal killing by delamanid of bacteria in log-phase growth and growth at pH 5.8 was recorded on day 7, after which all regimens failed as bacterial regrowth occurred. This regrowth in the HFS-TB has been observed for all TB drugs in monotherapy, including rifampin (17) and isoniazid (18), the two most important first-line TB drugs. The regrowth in the HFS-TB has been attributed to the emergence of resistance under monotherapy. Therefore, the killing (as calculated by the reduction of log10 CFU per milliliter in the delamanid treatment arms from the time-matched untreated control) at day 7 was used in the inhibitory sigmoid Emax model, as shown in Table 2 and Fig. 2. AUC0–24 and average log10 CFU-per-milliliter data for each regimen at day 7 can be found in Table S3 (for log-phase growth) and Table S4 (for growth at pH 5.8).

FIG 2
  • Open in new tab
  • Download powerpoint
FIG 2

Inhibitory sigmoid Emax model for log-phase growth (A) and growth at pH 5.8 (B). An inhibitory sigmoid Emax model was used to examine the log10 CFU-per-milliliter values for day 7 as the response variable versus AUC0–24/MIC from various regimens. (A) Each circle represents data from one HFS-TB with targeted AUC0–24/MIC. (B) Each circle represents data from one HFS-TB with observed AUC0–24/MIC. EC50 and EC80 represent the AUC0–24/MIC to achieve 50% and 80% of the maximum efficacy, respectively.

The EC80 (80% of the Emax) values derived on day 7 were AUC0–24/MIC ratios of 506 (95% CI, 360 to 777) for log-phase growth and 522 (95% CI, 210 to 910) for growth at pH 5.8 . PDTs obtained in the HFS-TB corresponded to the drug AUCs at the site of infection (i.e., lung), while the PK/PD analyses of animal and human data were based on the plasma levels. However, there are no human data on delamanid lung penetration and the relative ratio of AUC in lung versus plasma. Hence, the Kp value could only be inferred from animal data at this time. As shown Table S1, delamanid Kp values were 1.9, 2.9, and 3.0 when delamanid was administered as a single dose at 0.625, 2.5, and 10 mg/kg, respectively. Further, it was previously shown that after a single administration of delamanid at 3 mg/kg, the Kp was 2.4 (AUC0–24 = 13.768 mg · h/kg in lung versus 5.673 mg · h/liter in plasma) in mice (19). Using the average of 2.6 of the 4 mouse Kp values of 1.9, 2.4, 2.9, and 3, the plasma-equivalent PDTs for log-phase growth and growth at pH 5.8 were 195 (95% CI, 139 to 299) and 201 (95% CI, 81 to 350), respectively.

PDT for delamanid in humans.Individual delamanid AUC0–24/MIC and sputum culture bacterial burden reduction data from two early bactericidal activity (EBA) trials in drug-susceptible TB patients are shown in Table S6. The relationship between the reduction of log10 CFU per milliliter from baseline and AUC0–24/MIC was modeled using a nonlinear mixed-effect approach to assess the PDT of delamanid in humans. As shown in Fig. 3, an inhibitory sigmoid Emax model of bactericidal activity and plasma AUC0–24/MIC with random effect on maximum inhibition (Imax) provided the best fit and was selected as the final model. Parameter estimates are summarized in Table 2. Goodness of fit and visual predictive check plots are provided in Fig. S4 and S5, respectively. The plasma AUC0–24/MIC producing 80% inhibition (i.e., the PDT) was calculated to be 171 (95% CI, 79 to 292).

FIG 3
  • Open in new tab
  • Download powerpoint
FIG 3

Sputum log10 CFU reduction from baseline (prior to the start of treatment) to the end of treatment and AUC0–24/MIC relationship in trial 101 and trial 102. Each circle represents data from one patient. AUC0–24 is the daily area under the concentration-time curve on day 14 (trial 101) or day 7 (trial 102); EC50 and EC80 represent the AUC0–24/MIC to achieve 50% and 80% of the maximum efficacy, respectively.

CFR in patients with MDR-TB who were treated with 100 mg delamanid BID or 200 mg delamanid QD.As shown in Table 3, when patients were treated with 100 mg delamanid BID, the observed CFR was 100% in the trial 204 study population and ≥95% in the trial 213 study population, regardless of which PDT was used in the calculation. The CFR for 200 mg delamanid QD was 89.3% using the mouse PDT and above 90% for the other three PDTs in the trial 213 population.

View this table:
  • View inline
  • View popup
  • Download powerpoint
TABLE 3

Cumulative fraction of response following 100-mg BID or 200-mg QD delamanid dosinga

QTcF prolongation.In trial 213, patients with MDR-TB received 100 mg delamanid BID for 8 weeks, followed by 200 mg QD for 18 weeks (Table S5), thus providing an opportunity to compare the magnitude of corrected QT (QTc) interval prolongation under the two different dosing regimens. As shown previously, the mean differences from baseline in QTcF (QTc interval corrected for heart rate by Fridericia’s formula) between the delamanid-plus-OBR group (n = 341) and the placebo-plus-OBR group (n = 170) after 8 weeks of treatment with the 100-mg BID dose was 5.3 ms (90% CI, 2.8 to 7.9 ms) compared with 2.5 ms (90% CI, −0.3 to 5.3 ms) at the end of 18 weeks of treatment with the 200-mg QD dose (6). Thus, the QTc interval prolongation associated with 200-mg QD dosing was about 50% of that seen with 100-mg BID dosing.

DISCUSSION

The process to develop TB drugs is highly laborious due to the complexity of TB pathology, the spatial and temporal heterogeneity of TB lesions, which drives variable microbial killing and resistance emergence, and the lack of real-time biomarkers to measure treatment outcomes (20, 21). Delamanid was identified using in vitro and animal TB models that demonstrated bactericidal effects on replicating, dormant, and intracellular bacilli (22). Clinical development involved the traditional drug development pathway of testing on healthy subjects first, followed by proof-of-concept and dose selection phase II trials and a phase III trial in TB patients (22). In the EBA (phase IIa) trials, 200-mg QD and 300-mg QD doses of delamanid monotherapy appeared to produce similar bactericidal activity following 14 days of treatment in patients with drug-susceptible pulmonary TB (23). Nonetheless, due to observed delamanid dose-limiting absorption (23), the phase IIb trial used BID dosing. In the trial, 100 mg BID and 200 mg BID demonstrated similar proportions of sputum culture conversion after 8 weeks of treatment when added to an OBR in pulmonary MDR-TB patients (5). Therefore, the 100-mg BID dose regimen was subsequently approved by regulatory agencies, first in 2014 by the European Medicines Agency (EMA) (22). However, insufficient data were available for a thorough PK/PD analysis of delamanid at that time, including exploring alternate dosing regimens.

In the multipronged PK/PD analyses presented here, we attempted to understand the relationship between the PK and PD of delamanid using nonclinical and human PK/PD data, based on the principles outlined in a guideline from the EMA on the use of PK/PD in the development of antimicrobial medicinal products (24). Such principles are also consistent with the FDA guidance (25) for developing antibiotics and the methodology of establishing appropriate doses for TB drugs from the WHO (26). In addition to using data from the mouse model and human EBA trials, we utilized the data from the HFS-TB model, which was recently qualified by the EMA as a method for use in support of selection and development of antituberculosis drugs (27) and is supported by the FDA as a complementary tool for dose selection (28, 29). The HFS-TB is an in vitro system that can mimic human PK characteristics of antimycobacterial drugs at the site of infection and was built for the purpose of exploring the concentration-effect relationships potentially relevant to the treatment of TB. Another merit of the HFS-TB is that drug efficacy can be examined under different growth conditions of M. tuberculosis bacilli (log-phase growth and low pH to simulate a slowly replicating state) and, thus, may capture some of the microbial heterogenicity of TB lesions. In addition, repetitive sampling in the HFS-TB, as in patients’ sputum, allows determination of several quantitative PD measures, including kill slopes.

AUC0–24/MIC is the PK/PD index for delamanid, as determined in the mouse study. In the PDT and CFR calculations, AUC0–24 was obtained using noncompartmental methods for data from the mouse TB model, EBA trials (trials 101 and 102), and trial 204. For the data from the HFS and trial 213, model-based approaches were applied to obtain AUC0–24. As shown in Fig. S3 for the HFS and the companion article (30) for trial 213, the model predictions are reasonably consistent with observations. Thus, we determined that the calculated AUCs were accurate and could be used here for PK/PD analyses. The MICs of the infecting strains used in the mouse study (MIC of 0.012 mg/liter) and the HFS-TB study (MIC of 0.015 mg/liter) are in the range of the MICs in the EBA studies (0.006 mg/liter to 0.05 mg/liter) (Table S6 and Table S7) and trial 204 (0.001 to 0.05 mg/liter) (4). The individual MICs from trial 213 have not been published, but the distribution is similar to that in trial 204.

In our PK/PD analyses, two critical cutoff values were used: EC80 (80% of the Emax) and 90% for CFR. There is no regulatory or industry standard for a set percentage of the Emax to determine PDT. Since the Emax in the inhibitory sigmoid maximal microbial kill model is on an asymptote, a percentage of the Emax needs to be selected for the purpose of determining the PDT. Traditionally in the drug development industry, 80% or 90% of Emax has been selected (13, 31). We selected EC80 in our study based on 3 pieces of validation work that showed that at the level of 80%, PDTs selected from HFS-TB studies reliably predicted the drug exposures associated with clinical success. First, the accuracy to forecast optimal exposures and breakpoints in patients with TB using the EC80-based findings from the HFS-TB was validated by examining 20 clinical studies that were published after 26 HFS-TB experiments had been published (12). The HFS-TB EC80-based PDT predicted optimal drug exposures and doses that were identified in these 20 clinical studies based on agnostic machine learning analyses. The predictive accuracy of the HFS-TB in forecasting the clinical exposure values was 94.4% (95% CI, 84.3% to 99.9%). Second, a systematic analysis of HFS-TB and clinical findings indicated that EC80-based findings in tandem with Monte Carlo simulations for attainment of that target were similar between patients and the preclinical model (32). Third, a comprehensive analysis of all preclinical PK/PD studies in TB with clinical comparisons concluded that PDTs using 80% of Emax obtained in preclinical studies provided a reliable predictor of clinical success (31).

Importantly, increasing the efficacy level from 80% to 90% requires a large increase of AUC0–24, since the efficacy curve is flat from EC80 and above (i.e., on an asymptote). In our mouse model, the increase from EC80 to EC90 (only an additional 0.3 log10 CFU reduction) requires the increase of AUC0–24/MIC from 252 to 566 (AUC0–24 increases from 3.021 to 6.797 mg · h/liter) (Fig. 1), which represents very little gain in terms of microbial killing for a 2.2-fold jump in needed AUC. Doubling the AUC0–24 in humans from the 100-mg BID delamanid dose will likely increase side effects, such as the QTc interval prolongation. Therefore, taking into consideration the balance between efficacy and safety, we selected EC80 in this study. As for the cutoff of CFR, the EMA guidance provides 90% as an appropriate level for PK/PD analysis (24). FDA does not define a cutoff value for CFR as far as we are aware. However, it is generally accepted that the goal for PTA (probability of target attainment) or CFR should be at least 90% (33, 34). A white paper published in 2017 assessed the relationship between the PTA for antibacterial dosing regimens and FDA approval and concluded that, among the programs evaluated, FDA approval was granted for 88% of those achieving at least a 90% PTA versus much lower approval rates for development programs using lower percentages of PTA (35). Therefore, we felt that EC80 and CFR at 90% were reasonable selections for our PK/PD analyses.

We showed that the plasma or plasma-equivalent PDTs (AUC0–24/MIC) for delamanid ranged from 171, obtained from the human EBA trials, to 252, obtained in the mouse TB model. The PDTs from the HFS-TB assessments were 195 in log-phase growth and 201 in growth at pH 5.8. The values are within a narrow range, considering the very different models used to determine PDTs. Importantly, regardless of which PDT was used, the CFRs were >95% for the 100-mg BID dose, the dose currently approved for the treatment of pulmonary MDR-TB in adults, strongly indicating that this dose is appropriate to achieve the PDT. The similarity of the efficacy of the 200-mg BID dose evaluated in trial 204 to that of the 100-mg BID dose (5) is consistent with this conclusion.

While our analysis indicates that the 100-mg BID dose of delamanid can achieve EC80 in more than 90% of the patients, once-daily dosing of delamanid would be more convenient for patients and could lead to enhanced adherence. Our data show that the 200-mg QD dose could produce CFRs close to or above 90%, suggesting that 200 mg QD may be a feasible option for the MDR-TB population we evaluated. Furthermore, 200 mg QD likely causes less QTc interval prolongation than the 100-mg BID regimen, as shown in trial 213. To date, QTc interval prolongation is the major safety concern for delamanid.

The CFRs achieved with 200 mg delamanid QD are slightly lower than those from the 100-mg BID dose, and thus, once-daily doses higher than 200 mg could be explored. Data on once-daily delamanid doses higher than 200 mg, such as 300 mg, have been studied in healthy subjects, as well as in a 14-day EBA trial in pulmonary TB patients (23). In the EBA study, Cmax and AUC values for 300 mg QD were higher than those for 200 mg QD but still below those achieved with the 100-mg BID dose (5). Based on delamanid exposure, QTc interval prolongation from the 300 mg QD dose would be expected to be lower than that from the 100 mg BID dose, since the concentrations of both delamanid and DM-6705, a major metabolite causing QTc interval prolongation, are lower following the 300-mg QD dose than the 100-mg BID dose (7).

Our study has several limitations. First, the mouse multiple-dose PK profiles were simulated from single-dose data in healthy animals. However, PK parameters obtained using this method were similar to the actual measurements obtained with multiple doses of delamanid in M. tuberculosis Kurono-infected mice at the dose of 2.5 mg/kg and when using the one-compartment model method (see Table S2 and the other supplemental material). Thus, the nonparametric superposition method provided a reasonable estimation of the PK parameters following multiple doses of delamanid in this study. Further, delamanid PK parameters were similar between healthy subjects and patients with TB (7). Second, different phenotypes of TB bacilli (i.e., replicating, and dormant) respond differently to drug treatment, and animal models used to generate PK/PD measures of efficacy may not faithfully mimic TB disease in humans. It should be noted, however, that the HFS-TB examined low-pH conditions, which is thought to mimic certain caseous TB lesions (36–40). Third, the mouse and HFS-TB studies were designed to evaluate bactericidal activity and not sterilizing efficacy and relapse prevention of the drug. Fourth, as shown in Fig. 3, human EBA responses in trials 101 and 102 were variable. However, the model described well the central tendency values based on goodness of fit and visual predictive check (Fig. S4 and S5). Fifth, since no human lung tissue drug concentration data are available for delamanid, we used the lung tissue and plasma levels in a mouse study to calculate Kp and obtain the plasma-equivalent HFS-TB PDT. In a separate study using guinea pigs, the Kp was 18.4 (41), which would make the plasma equivalent PDT much lower. Since human TB lesions are heterogenous, TB drug penetrations are likely lesion type dependent (42). Future studies using animals and, ideally, human TB lungs are needed to investigate delamanid lesion penetration. Finally, the nonclinical PDTs were determined with monotherapy and the human PDT was determined from short-term monotherapy in EBA trials; therefore, we cannot exclude the possibility that PDTs for each drug component in a multidrug regimen when used for MDR-TB patients could be different.

In conclusion, in the present study, we identified AUC0–24/MIC as the index of delamanid efficacy. Using PDTs obtained from different models, we further showed that delamanid at 100 mg BID and 200 mg QD achieved cumulative fractions of response of ≥90% in two large trials of patients with pulmonary MDR-TB. Therefore, once-daily doses of delamanid, such as 200 mg or higher, may be possible options, likely with less QTc interval prolongation than with the 100-mg BID dose. Hence, QD doses of delamanid should be further evaluated in future clinical trials.

MATERIALS AND METHODS

General approach to the PK/PD analysis.To determine CFR for the 100-mg BID and 200-mg QD doses of delamanid, we used general principles for PK/PD analysis as outlined in several publications, including one from the EMA (24, 26, 33). We used data from a mouse and an HFS-TB study and several human trials for this effort (Table S5). The steps taken to obtain the CFRs are outlined in Fig. S1.

Materials.M. tuberculosis Kurono (ATCC 35812) was used in the mouse infection model, and H37Rv (ATCC 27294) was used in the HFS-TB studies. SLC:ICR mice were obtained from Japan SLC, Inc. (Hamamatsu, Shizuoka, Japan). The hollow-fiber cartridges with polysulfone hollow fibers were purchased from FiberCell System, Inc. (New Market, MD, USA). Delamanid was supplied by Otsuka Pharmaceutical Co., Ltd. (Tokushima, Japan).

PK analysis of single-dose delamanid in mice and estimation after multiple doses.All animal studies were carried out in accordance with the document “Guidelines for Animal Care and Use in Otsuka Pharmaceutical Co., Ltd.” To evaluate delamanid PK, uninfected SLC:ICR mice were administered a single dose of delamanid at 0.625, 2.5, or 10 mg/kg by oral gavage. Three to six mice in each dosage group were sacrificed at 1, 2, 4, 6, 8, 12, and 24 h to obtain blood and lung tissue. Plasma and lung concentrations of delamanid were determined by high-performance liquid chromatographic-tandem mass spectrometry (HPLC-MS/MS), according to the method described by Hirao et al. (19). Plasma AUC0–24 values were calculated by the linear trapezoidal method using Microsoft Excel.

Based on the data from the single-dose PK study, PK parameters from multidose delamanid at 0.625, 2.5, or 10 mg/kg were then simulated by the nonparametric superposition method using Phoenix WinNonlin software, version 6.3, in various 28-day treatment regimens—twice daily, once daily, three time per week, or once per week—the same regimens that were used in the efficacy experiments, as described below (Table 1). Results of these simulations, in combination with the delamanid MIC of 0.012 mg/liter for the M. tuberculosis Kurono strain (used in the efficacy study), were used to calculate the following PK/PD indices: weekly (504 to 672 h corresponds to the fourth week of treatment) AUC504–672, AUC504–672/MIC, daily AUC (AUC504–672/7), daily AUC/MIC, Cmax504–672, Cmax504–672/MIC, and %T>MIC, i.e., percentage of time above MIC during the fourth week of treatment.

PK/PD index and PDT assessments in mice.Mice were infected with M. tuberculosis Kurono by tail vein injection at 1.2 × 103 CFU/mouse. Starting from 4 weeks postinfection (average bacterial load at the time of treatment was 5.964 log10 CFU/lung for 5 mice), the mice were treated with delamanid by oral gavage for 28 days using eight different dosing regimens (Table 1), with five animals per treatment group, except for one group with only four mice due to a technical error (Table 1). One group of mice did not receive delamanid and served as untreated controls. At the end of the 28-day treatment period, mice were sacrificed by exsanguination through the abdominal inferior vena cava under ether anesthesia, and lung tissue was aseptically excised. Dilutions of lung homogenates were plated on 7H11 agar medium supplemented with 10% oleic acid-albumin-dextrose-catalase (OADC), and the plates were incubated at 37°C until colonies were sufficiently grown for visual counting (usually 3 weeks). The log10 CFU/lung reduction in each animal was calculated by subtracting the log10 CFU from the mean log10 CFU of untreated controls.

Correlations between the various PK/PD indices and log10 CFU/lung reduction were analyzed by Pearson’s or Spearman correlation using SAS software, releases 9.1 and 9.3 (SAS Institute Japan Ltd., Tokyo, Japan). A P value less than 0.05 was considered statistically significant. Additionally, corrected Akaike information criterion scores obtained from the inhibitory sigmoid Emax modeling of log10 CFU/lung reduction versus PK/PD indices were further used to determine the PK/PD index for delamanid.

After the PK/PD index was determined (which is AUC0–24/MIC for delamanid; see Results), the PDT of delamanid in mice (that is, the AUC0–24/MIC required for 80% of Emax [EC80]) was obtained using inhibitory sigmoid Emax modeling of reduction in log10 CFU per lung from the average log10 CFU per lung of untreated mice versus the AUC0–24/MIC ratios for each treatment group, as follows:log10 CFU/lung=Emax×(AUC0−24MIC)HillEC50Hill+(AUC0−24MIC)Hill(1)where Emax was the maximum log10 CFU/lung reduction from untreated controls, AUC0–24 was the average daily AUC from the last week of the 4-week treatment, MIC was 0.012 mg/liter for the infecting M. tuberculosis Kurono strain, and EC50 was the 50% AUC0–24/MIC ratio for maximum log10 CFU decline. The AUC0–24/MIC values corresponding to exposure required for 50% of Emax (EC50), EC80, and EC90 were calculated. These analyses were performed with SAS software, release 9.3 (SAS Institute Japan, Ltd.).

Pharmacodynamic target assessments in the HFS-TB.The design of the HFS-TB can be found in previous publications (43, 44) and is described briefly below. The HFS-TB was adapted for delamanid by addition of 0.1% Tween 80 and 10% bovine serum albumin to Middlebrook 7H9 broth. Delamanid concentration was measured by liquid chromatography with tandem mass spectrometry using a Waters (Milford, MA, USA) Acquity UPLC connected to a Waters Xevo TQ mass spectrometer. Data were collected using MassLynx version 4.1 SCN810 software.

Delamanid was infusion based on preliminary HFS PK experiments that identified the corresponding conditions to best mimic human PK parameters, with Cmax at 4 h and a terminal half-life of 30 h to match that in humans (30 to 38 h) (7). PK parameters were derived from a one-compartment model, which provided excellent estimation of actual measured concentrations, as shown in Fig. S3. The PK model-derived AUC0–24 at each efficacy sampling day was then obtained for the use in the inhibitory sigmoid Emax model, as described below. Bacterial killing was studied in two M. tuberculosis culture conditions: log-phase growth and culture at pH 5.8.

(i) Log-phase-growth study.To prepare the inoculum, M. tuberculosis H37Rv was grown to log phase in Middlebrook 7H9 broth with 10% OADC for 4 days under shaking conditions and 5% CO2. On day 0, 20 ml of 6 log10 CFU/ml culture was inoculated into each hollow-fiber system. Starting on day 1 (average bacterial load was 6.15 log10 CFU/ml), doses of delamanid were administered to achieve the targets described in Table S3. These dose regimens were designed to cover the EC80 identified in the mouse study with sufficient exposures below and above this level to allow the establishment of an exposure-response curve. Different concentrations of delamanid were prepared in a 2-ml volume and infused over 4 h using a syringe pump. Treatment was administered daily for 28 days. The peripheral compartment of each HFS-TB was sampled on study days 3, 7, 10, 14, 21, and 28 to determine the number of bacterial CFU per milliliter. Samples for PK analysis were collected from each HFS-TB central compartment starting 15 min prior to the day 28 dose (time zero), with time points at 2, 4, 12, 20, 24, 48, and 72 h after the start of the last infusion.

(ii) M. tuberculosis cultured at pH 5.8.Growth at pH 5.8 was carried out as described for log-phase growth with the following modifications: nonreplicating M. tuberculosis strain H37Rv was propagated under acidic conditions (pH 5.8) before being inoculated into the HFS at an inoculum of 1 × 105 (5 log10) CFU/ml. At day 1 of treatment initiation, the bacterial load was 5.98 log10 CFU/ml. The target exposures for the pH 5.8 study are shown in Table S4. Two additional exposures higher than those evaluated in the log-phase study were included in the anticipation that higher exposures were needed to kill bacilli cultured at pH 5.8.

(iii) Efficacy determination.Quantitative cultures were performed on bacterial samples collected from each HFS-TB peripheral compartment. The samples were obtained just before administration of the next scheduled dose of delamanid. To prevent drug carryover, each 1-ml sample was washed twice, resuspended, and serially diluted 10-fold in sterile saline for quantitative cultures. Each dilution was then cultured on antibiotic-free Middlebrook 7H10 agar plates supplemented with 10% OADC. All cultures were incubated under 5% CO2 at 37°C for 21 days for CFU counting.

As in the mouse study, the inhibitory sigmoid Emax model (equation 1) was used to estimate the EC50 and EC80. The MIC was 0.015 mg/liter for M. tuberculosis strain H37Rv, Emax was maximum bacterial killing from time-matched untreated controls, and the Hill coefficient was estimated for this analysis. S-ADAPT and ADAPT 5 (Biomedical Simulations Resources, University of Southern California) were used for data analysis.

PDT assessments in trials 101 and 102.Data from two EBA trials, trial 101 and trial 102 (Table S5), were used to assess the relationship between AUC0–24/MIC and bacterial kill trajectories in humans. AUC0–24/MIC was obtained using the individual daily AUC (trial 101, AUC on day 14; trial 102, AUC on day 7 [calculated by the linear trapezoidal rule with R version 3.2.2]) divided by the MIC for the clinical isolate from the same patient at baseline determined using the proportion method. Bacterial killing was calculated by the reduction of log10 CFU per milliliter from the baseline to the lowest sputum log10 CFU per milliliter for the 7-day treatment period in trial 101 and to the day 14 log10 CFU per milliliter in trial 101. The inhibitory sigmoidal Emax model (as shown in equation 1) and nonlinear mixed-effect analysis were used to model the data. The analysis was performed using NONMEM software, version 7.3.0 (ICON, Dublin, Ireland). Summary statistics and raw data used for the analysis are provided in Tables S6 and S7.

CFRs for the 100-mg BID and 200-mg QD delamanid doses.In trial 204, delamanid plasma concentrations were determined predose and 2, 3, 4, 10, 12, 13, 14, and 24 h postdose on days 1, 14, 28, and 56 following administration of delamanid at 100 mg BID in MDR-TB patients. Daily AUCs on days 1, 14, 28, and 56 were determined by noncompartmental analysis (NCA) methods. The average steady-state AUC0–24 was determined by averaging the AUC0–24 on days 14, 28, and 56. In trial 213, patients received 100 mg delamanid BID plus OBR for 2 months, followed by 200 mg delamanid QD plus OBR for 4 months. Due to sparse sampling of the PK in this study, the steady-state AUC0–24 values following the 100-mg BID and 200-mg QD dosing were estimated based on a population PK model (30). Baseline MICs for M. tuberculosis isolates from each patient were determined using the proportion method (4). Patients with M. tuberculosis isolates that had baseline MICs of ≤0.016 mg/liter were included in the analyses, as this MIC has been established as the critical concentration for delamanid (45). The calculated individual AUC0–24/MIC values from trial 204 and trial 213 were compared with the selected PDT values (mouse, HFS-TB plasma-equivalent, and human EBA PDTs) to determine the CFR.

QTc interval determination.In trial 213, 12-lead electrocardiograms (ECG) were recorded at screening baseline (day −1), at day 1, and at weekly visits from week 1 to week 12 and biweekly from week 14 to 26 during the 6-month treatment, with the subject supine and at rest for at least 10 min. During treatment, three ECGs were collected 5 to 10 min apart after the morning dose of IMP (investigational medicinal product). In addition to the initial clinical interpretation for ongoing safety evaluation by the investigator, digitally acquired ECGs were received by the central reader for processing and were analyzed by a central reader. QT intervals were corrected for heart rate using Fridericia’s formula (QTcF) and averaged from the 3 ECG readings. Delamanid’s QTc prolongation effect is mainly caused by one of its metabolites, DM-6705 (7), as DM-6705 concentrations were identified as a surrogate marker for QTc prolongation. The terminal half-life of DM-6705 is about 10 days; thus, plasma concentrations of this metabolite fluctuate very little at steady state, which is reached in about 7 weeks. Hence, the timing of performing ECGs, at or beyond 7 weeks, is not important to determine the effect of delamanid on QTc.

ACKNOWLEDGMENTS

We acknowledge and thank the patients and investigators who participated in the clinical trials discussed herein.

The HFS-TB study was supported by the Critical Path to TB Drug Regimens Initiative, a partnership between the Bill and Melinda Gates Foundation and the Critical Path Institute (Tucson, AZ, USA). The rest of the studies in this report were supported by Otsuka Pharmaceutical Development & Commercialization, Inc., Rockville, MD, USA (OPDC), and Otsuka Pharmaceutical Co., Ltd., Tokushima, Japan (OPC). David Norris (Ecosse Medical Communications, Falmouth, MA, USA) provided editorial services during the writing of the manuscript; these editorial services were supported by OPDC.

FOOTNOTES

    • Received 12 June 2020.
    • Returned for modification 15 July 2020.
    • Accepted 13 October 2020.
    • Accepted manuscript posted online 26 October 2020.
  • Supplemental material is available online only.

  • Copyright © 2020 Mallikaarjun et al.

This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license.

REFERENCES

  1. 1.↵
    World Health Organization. 2019. Global tuberculosis report 2019. https://apps.who.int/iris/bitstream/handle/10665/329368/9789241565714-eng.pdf?ua=1. Accessed January 2020.
  2. 2.↵
    1. Matsumoto M,
    2. Hashizume H,
    3. Tomishige T,
    4. Kawasaki M,
    5. Tsubouchi H,
    6. Sasaki H,
    7. Shimokawa Y,
    8. Komatsu M
    . 2006. OPC-67683, a nitro-dihydro-imidazooxazole derivative with promising action against tuberculosis in vitro and in mice. PLoS Med 3:e466. doi:10.1371/journal.pmed.0030466.
    OpenUrlCrossRefPubMed
  3. 3.↵
    1. Nataraj V,
    2. Varela C,
    3. Javid A,
    4. Singh A,
    5. Besra GS,
    6. Bhatt A
    . 2015. Mycolic acids: deciphering and targeting the Achilles' heel of the tubercle bacillus. Mol Microbiol 98:7–16. doi:10.1111/mmi.13101.
    OpenUrlCrossRefPubMed
  4. 4.↵
    1. Stinson K,
    2. Kurepina N,
    3. Venter A,
    4. Fujiwara M,
    5. Kawasaki M,
    6. Timm J,
    7. Shashkina E,
    8. Kreiswirth BN,
    9. Liu Y,
    10. Matsumoto M,
    11. Geiter L
    . 2016. MIC of delamanid (OPC-67683) against Mycobacterium tuberculosis clinical isolates and a proposed critical concentration. Antimicrob Agents Chemother 60:3316–3322. doi:10.1128/AAC.03014-15.
    OpenUrlAbstract/FREE Full Text
  5. 5.↵
    1. Gler MT,
    2. Skripconoka V,
    3. Sanchez-Garavito E,
    4. Xiao H,
    5. Cabrera-Rivero JL,
    6. Vargas-Vasquez DE,
    7. Gao M,
    8. Awad M,
    9. Park SK,
    10. Shim TS,
    11. Suh GY,
    12. Danilovits M,
    13. Ogata H,
    14. Kurve A,
    15. Chang J,
    16. Suzuki K,
    17. Tupasi T,
    18. Koh WJ,
    19. Seaworth B,
    20. Geiter LJ,
    21. Wells CD
    . 2012. Delamanid for multidrug-resistant pulmonary tuberculosis. N Engl J Med 366:2151–2160. doi:10.1056/NEJMoa1112433.
    OpenUrlCrossRefPubMedWeb of Science
  6. 6.↵
    1. von Groote-Bidlingmaier F,
    2. Patientia R,
    3. Sanchez E,
    4. Balanag V, Jr,
    5. Ticona E,
    6. Segura P,
    7. Cadena E,
    8. Yu C,
    9. Cirule A,
    10. Lizarbe V,
    11. Davidaviciene E,
    12. Domente L,
    13. Variava E,
    14. Caoili J,
    15. Danilovits M,
    16. Bielskiene V,
    17. Staples S,
    18. Hittel N,
    19. Petersen C,
    20. Wells C,
    21. Hafkin J,
    22. Geiter LJ,
    23. Gupta R
    . 2019. Efficacy and safety of delamanid in combination with an optimised background regimen for treatment of multidrug-resistant tuberculosis: a multicentre, randomised, double-blind, placebo-controlled, parallel group phase 3 trial. Lancet Respir Med 7:249–259. doi:10.1016/S2213-2600(18)30426-0.
    OpenUrlCrossRef
  7. 7.↵
    European Medicines Agency. 2020. Deltyba European public assessment report (EPAR). https://www.ema.europa.eu/en/documents/assessment-report/deltyba-epar-public-assessment-report_en.pdf. Accessed September 2020.
  8. 8.↵
    World Health Organization. 2016. The use of delamanid in the treatment of multidrug-resistant tuberculosis in children and adults: interim policy guidance. https://www.ncbi.nlm.nih.gov/books/NBK396145/. Accessed February 2020.
  9. 9.↵
    1. Gupta R,
    2. Geiter LJ,
    3. Wells CD,
    4. Gao M,
    5. Cirule A,
    6. Xiao H
    . 2015. Delamanid for extensively drug-resistant tuberculosis. N Engl J Med 373:291–292. doi:10.1056/NEJMc1415332.
    OpenUrlCrossRefPubMed
  10. 10.↵
    1. Skripconoka V,
    2. Danilovits M,
    3. Pehme L,
    4. Tomson T,
    5. Skenders G,
    6. Kummik T,
    7. Cirule A,
    8. Leimane V,
    9. Kurve A,
    10. Levina K,
    11. Geiter LJ,
    12. Manissero D,
    13. Wells CD
    . 2013. Delamanid improves outcomes and reduces mortality in multidrug-resistant tuberculosis. Eur Respir J 41:1393–1400. doi:10.1183/09031936.00125812.
    OpenUrlAbstract/FREE Full Text
  11. 11.↵
    1. Wells CD,
    2. Gupta R,
    3. Hittel N,
    4. Geiter LJ
    . 2015. Long-term mortality assessment of multidrug-resistant tuberculosis patients treated with delamanid. Eur Respir J 45:1498–1501. doi:10.1183/09031936.00176314.
    OpenUrlAbstract/FREE Full Text
  12. 12.↵
    1. Gumbo T,
    2. Pasipanodya JG,
    3. Romero K,
    4. Hanna D,
    5. Nuermberger E
    . 2015. Forecasting accuracy of the hollow fiber model of tuberculosis for clinical therapeutic outcomes. Clin Infect Dis 61(Suppl 1):S25–S31. doi:10.1093/cid/civ427.
    OpenUrlCrossRefPubMed
  13. 13.↵
    1. Tuntland T,
    2. Ethell B,
    3. Kosaka T,
    4. Blasco F,
    5. Zang RX,
    6. Jain M,
    7. Gould T,
    8. Hoffmaster K
    . 2014. Implementation of pharmacokinetic and pharmacodynamic strategies in early research phases of drug discovery and development at Novartis Institute of Biomedical Research. Front Pharmacol 5:174. doi:10.3389/fphar.2014.00174.
    OpenUrlCrossRefPubMed
  14. 14.↵
    1. Srivastava S,
    2. Magombedze G,
    3. Koeuth T,
    4. Sherman C,
    5. Pasipanodya JG,
    6. Raj P,
    7. Wakeland E,
    8. Deshpande D,
    9. Gumbo T
    . 2017. Linezolid dose that maximizes sterilizing effect while minimizing toxicity and resistance emergence for tuberculosis. Antimicrob Agents Chemother 61:e00751-17. doi:10.1128/AAC.00751-17.
    OpenUrlAbstract/FREE Full Text
  15. 15.↵
    1. Deshpande D,
    2. Srivastava S,
    3. Pasipanodya JG,
    4. Bush SJ,
    5. Nuermberger E,
    6. Swaminathan S,
    7. Gumbo T
    . 2016. Linezolid for infants and toddlers with disseminated tuberculosis: first steps. Clin Infect Dis 63:S80–S87. doi:10.1093/cid/ciw482.
    OpenUrlCrossRefPubMed
  16. 16.↵
    1. Modongo C,
    2. Pasipanodya JG,
    3. Magazi BT,
    4. Srivastava S,
    5. Zetola NM,
    6. Williams SM,
    7. Sirugo G,
    8. Gumbo T
    . 2016. Artificial intelligence and amikacin exposures predictive of outcomes in multidrug-resistant tuberculosis patients. Antimicrob Agents Chemother 60:5928–5932. doi:10.1128/AAC.00962-16.
    OpenUrlAbstract/FREE Full Text
  17. 17.↵
    1. Drusano GL,
    2. Sgambati N,
    3. Eichas A,
    4. Brown DL,
    5. Kulawy R,
    6. Louie A
    . 2010. The combination of rifampin plus moxifloxacin is synergistic for suppression of resistance but antagonistic for cell kill of Mycobacterium tuberculosis as determined in a hollow-fiber infection model. mBio 1:e00139-10. doi:10.1128/mBio.00139-10.
    OpenUrlAbstract/FREE Full Text
  18. 18.↵
    1. Gumbo T,
    2. Louie A,
    3. Liu W,
    4. Ambrose PG,
    5. Bhavnani SM,
    6. Brown D,
    7. Drusano GL
    . 2007. Isoniazid's bactericidal activity ceases because of the emergence of resistance, not depletion of Mycobacterium tuberculosis in the log phase of growth. J Infect Dis 195:194–201. doi:10.1086/510247.
    OpenUrlCrossRefPubMedWeb of Science
  19. 19.↵
    1. Hirao Y,
    2. Koga T,
    3. Koyama N,
    4. Shimokawa Y,
    5. Umehara K
    . 2015. Liquid chromatography-tandem mass spectrometry methods for determination of delamanid in mouse plasma and lung. Am J Anal Chem 6:98–105. doi:10.4236/ajac.2015.62009.
    OpenUrlCrossRef
  20. 20.↵
    1. Ordonez AA,
    2. Wang H,
    3. Magombedze G,
    4. Ruiz-Bedoya CA,
    5. Srivastava S,
    6. Chen A,
    7. Tucker EW,
    8. Urbanowski ME,
    9. Pieterse L,
    10. Fabian Cardozo E,
    11. Lodge MA,
    12. Shah MR,
    13. Holt DP,
    14. Mathews WB,
    15. Dannals RF,
    16. Gobburu JVS,
    17. Peloquin CA,
    18. Rowe SP,
    19. Gumbo T,
    20. Ivaturi VD,
    21. Jain SK
    . 2020. Dynamic imaging in patients with tuberculosis reveals heterogeneous drug exposures in pulmonary lesions. Nat Med 26:529–534. doi:10.1038/s41591-020-0770-2.
    OpenUrlCrossRef
  21. 21.↵
    1. Dheda K,
    2. Lenders L,
    3. Magombedze G,
    4. Srivastava S,
    5. Raj P,
    6. Arning E,
    7. Ashcraft P,
    8. Bottiglieri T,
    9. Wainwright H,
    10. Pennel T,
    11. Linegar A,
    12. Moodley L,
    13. Pooran A,
    14. Pasipanodya JG,
    15. Sirgel FA,
    16. van Helden PD,
    17. Wakeland E,
    18. Warren RM,
    19. Gumbo T
    . 2018. Drug-penetration gradients associated with acquired drug resistance in patients with tuberculosis. Am J Respir Crit Care Med 198:1208–1219. doi:10.1164/rccm.201711-2333OC.
    OpenUrlCrossRef
  22. 22.↵
    1. Liu Y,
    2. Matsumoto M,
    3. Ishida H,
    4. Ohguro K,
    5. Yoshitake M,
    6. Gupta R,
    7. Geiter L,
    8. Hafkin J
    . 2018. Delamanid: from discovery to its use for pulmonary multidrug-resistant tuberculosis (MDR-TB). Tuberculosis (Edinb) 111:20–30. doi:10.1016/j.tube.2018.04.008.
    OpenUrlCrossRef
  23. 23.↵
    1. Diacon AH,
    2. Dawson R,
    3. Hanekom M,
    4. Narunsky K,
    5. Venter A,
    6. Hittel N,
    7. Geiter LJ,
    8. Wells CD,
    9. Paccaly AJ,
    10. Donald PR
    . 2011. Early bactericidal activity of delamanid (OPC-67683) in smear-positive pulmonary tuberculosis patients. Int J Tuber Lung Dis 15:949–954. doi:10.5588/ijtld.10.0616.
    OpenUrlCrossRef
  24. 24.↵
    European Medicines Agency Committee for Medicinal Products for Human Use. 2016. Guideline on the use of pharmacokinetics and pharmacodynamics in the development of antimicrobial medicinal products. European Medicines Agency, London, United Kingdom.
  25. 25.↵
    Center for Drug Evaluation and Research. 2018. Microbiology data for systemic antibacterial drugs—development, analysis, and presentation. https://www.fda.gov/media/77442/download. Accessed September 2020.
  26. 26.↵
    World Health Organization. 2018. Technical report on the pharmacokinetics and pharmacodynamics (PK/PD) of medicines used in the treatment of drug-resistant tuberculosis. https://apps.who.int/iris/bitstream/handle/10665/260440/WHO-CDS-TB-2018.6-eng.pdf?sequence=1&isAllowed=y. Accessed September 2020.
  27. 27.↵
    1. Cavaleri M,
    2. Manolis E
    . 2015. Hollow fiber system model for tuberculosis: the European Medicines Agency experience. Clin Infect Dis 61(Suppl 1):S1–S4. doi:10.1093/cid/civ484.
    OpenUrlCrossRefPubMed
  28. 28.↵
    1. Romero K,
    2. Clay R,
    3. Hanna D
    . 2015. Strategic regulatory evaluation and endorsement of the hollow fiber tuberculosis system as a novel drug development tool. Clin Infect Dis 61(Suppl 1):S5–S9. doi:10.1093/cid/civ424.
    OpenUrlCrossRefPubMed
  29. 29.↵
    1. Chilukuri D,
    2. McMaster O,
    3. Bergman K,
    4. Colangelo P,
    5. Snow K,
    6. Toerner JG
    . 2015. The hollow fiber system model in the nonclinical evaluation of antituberculosis drug regimens. Clin Infect Dis 61(Suppl 1):S32–S33. doi:10.1093/cid/civ460.
    OpenUrlCrossRefPubMed
  30. 30.↵
    1. Wang X,
    2. Mallikaarjun S,
    3. Gibiansky E
    . 2021. Population pharmacokinetic analysis of delamanid in patients with pulmonary multidrug-resistant tuberculosis. Antimicrob Agents Chemother 65:e01202-20. doi:10.1128/AAC.01202-20.
    OpenUrlCrossRef
  31. 31.↵
    1. Gumbo T,
    2. Angulo-Barturen I,
    3. Ferrer-Bazaga S
    . 2015. Pharmacokinetic-pharmacodynamic and dose-response relationships of antituberculosis drugs: recommendations and standards for industry and academia. J Infect Dis 211(Suppl 3):S96–S106. doi:10.1093/infdis/jiu610.
    OpenUrlCrossRefPubMed
  32. 32.↵
    1. Pasipanodya J,
    2. Gumbo T
    . 2011. An oracle: antituberculosis pharmacokinetics-pharmacodynamics, clinical correlation, and clinical trial simulations to predict the future. Antimicrob Agents Chemother 55:24–34. doi:10.1128/AAC.00749-10.
    OpenUrlAbstract/FREE Full Text
  33. 33.↵
    1. Simner PJ,
    2. Miller L
    . 2018. Understanding pharmacokinetics (PK) and pharmacodynamics (PD). https://clsi.org/media/1990/ast_news_update_jan18-pkpd.pdf. Accessed September 2020.
  34. 34.↵
    National Institute of Allergy and Infectious Diseases. 2017. NIAID Workshop ‘Pharmacokinetics-Pharmacodynamics (PKPD) for Development of Therapeutics against Bacterial Pathogens’, June 14–15, 2017, Bethesda, MD. http://amr.solutions/wp-content/uploads/99/niaid_june2017_pkpdworkshopsummary.pdf. Accessed September 2020.
  35. 35.↵
    Institute for Clinical Pharmacodynamics. 2017. De-risking antibiotic drug development with PK-PD. http://icpd.com/downloads/ICPD-White-Paper-De-risking-Drug-Development-with-PK-PD.pdf. Accessed September 2020.
  36. 36.↵
    1. Srivastava S,
    2. Pasipanodya JG,
    3. Gumbo T
    . 2017. pH conditions under which pyrazinamide works in humans. Antimicrob Agents Chemother 61:e00854-17. doi:10.1128/AAC.00854-17.
    OpenUrlFREE Full Text
  37. 37.↵
    1. Kempker RR,
    2. Heinrichs MT,
    3. Nikolaishvili K,
    4. Sabulua I,
    5. Bablishvili N,
    6. Gogishvili S,
    7. Avaliani Z,
    8. Tukvadze N,
    9. Little B,
    10. Bernheim A,
    11. Read TD,
    12. Guarner J,
    13. Derendorf H,
    14. Peloquin CA,
    15. Blumberg HM,
    16. Vashakidze S
    . 2017. Lung tissue concentrations of pyrazinamide among patients with drug-resistant pulmonary tuberculosis. Antimicrob Agents Chemother 61:e00226-17. doi:10.1128/AAC.00226-17.
    OpenUrlAbstract/FREE Full Text
  38. 38.↵
    1. McDermott W,
    2. Tompsett R
    . 1954. Activation of pyrazinamide and nicotinamide in acidic environments in vitro. Am Rev Tuberc 70:748–754. doi:10.1164/art.1954.70.4.748.
    OpenUrlCrossRefPubMed
  39. 39.↵
    1. McCune RM,
    2. Feldmann FM,
    3. Lambert HP,
    4. McDermott W
    . 1966. Microbial persistence. I. The capacity of tubercle bacilli to survive sterilization in mouse tissues. J Exp Med 123:445–468. doi:10.1084/jem.123.3.445.
    OpenUrlAbstract
  40. 40.↵
    1. Mitchison DA
    . 1979. Basic mechanisms of chemotherapy. Chest 76:771–781. doi:10.1378/chest.76.6_supplement.771.
    OpenUrlCrossRefPubMedWeb of Science
  41. 41.↵
    1. Chen X,
    2. Hashizume H,
    3. Tomishige T,
    4. Nakamura I,
    5. Matsuba M,
    6. Fujiwara M,
    7. Kitamoto R,
    8. Hanaki E,
    9. Ohba Y,
    10. Matsumoto M
    . 2017. Delamanid kills dormant Mycobacteria in vitro and in a guinea pig model of tuberculosis. Antimicrob Agents Chemother 61:e02402-16. doi:10.1128/AAC.02402-16.
    OpenUrlAbstract/FREE Full Text
  42. 42.↵
    1. Strydom N,
    2. Gupta SV,
    3. Fox WS,
    4. Via LE,
    5. Bang H,
    6. Lee M,
    7. Eum S,
    8. Shim T,
    9. Barry CE, III,
    10. Zimmerman M,
    11. Dartois V,
    12. Savic RM
    . 2019. Tuberculosis drugs' distribution and emergence of resistance in patient's lung lesions: a mechanistic model and tool for regimen and dose optimization. PLoS Med 16:e1002773. doi:10.1371/journal.pmed.1002773.
    OpenUrlCrossRefPubMed
  43. 43.↵
    European Medicines Agency. 2015. In-vitro hollow fiber system model of tuberculosis (HSF-TB). https://www.ema.europa.eu/en/documents/regulatory-procedural-guideline/qualification-opinion-vitro-hollow-fibre-system-model-tuberculosis-hfs-tb_en.pdf. Accessed September 2019.
  44. 44.↵
    1. Srivastava S,
    2. Gumbo T
    . 2011. In vitro and in vivo modeling of tuberculosis drugs and its impact on optimization of doses and regimens. Curr Pharm Des 17:2881–2888. doi:10.2174/138161211797470192.
    OpenUrlCrossRefPubMed
  45. 45.↵
    World Health Organization. 2018. Technical report on critical concentrations for drug susceptibility testing of medicines used in the treatment of drug-resistant tuberculosis. https://www.who.int/tb/publications/2018/WHO_technical_report_concentrations_TB_drug_susceptibility/en/. Accessed September 2019.
PreviousNext
Back to top
Download PDF
Citation Tools
Cumulative Fraction of Response for Once- and Twice-Daily Delamanid in Patients with Pulmonary Multidrug-Resistant Tuberculosis
Suresh Mallikaarjun, Moti L. Chapagain, Tomohiro Sasaki, Norimitsu Hariguchi, Devyani Deshpande, Shashikant Srivastava, Alexander Berg, Kuniko Hirota, Yusuke Inoue, Makoto Matsumoto, Jeffrey Hafkin, Lawrence Geiter, Xiaofeng Wang, Tawanda Gumbo, Yongge Liu
Antimicrobial Agents and Chemotherapy Dec 2020, 65 (1) e01207-20; DOI: 10.1128/AAC.01207-20

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Print

Alerts
Sign In to Email Alerts with your Email Address
Email

Thank you for sharing this Antimicrobial Agents and Chemotherapy article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Cumulative Fraction of Response for Once- and Twice-Daily Delamanid in Patients with Pulmonary Multidrug-Resistant Tuberculosis
(Your Name) has forwarded a page to you from Antimicrobial Agents and Chemotherapy
(Your Name) thought you would be interested in this article in Antimicrobial Agents and Chemotherapy.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
Cumulative Fraction of Response for Once- and Twice-Daily Delamanid in Patients with Pulmonary Multidrug-Resistant Tuberculosis
Suresh Mallikaarjun, Moti L. Chapagain, Tomohiro Sasaki, Norimitsu Hariguchi, Devyani Deshpande, Shashikant Srivastava, Alexander Berg, Kuniko Hirota, Yusuke Inoue, Makoto Matsumoto, Jeffrey Hafkin, Lawrence Geiter, Xiaofeng Wang, Tawanda Gumbo, Yongge Liu
Antimicrobial Agents and Chemotherapy Dec 2020, 65 (1) e01207-20; DOI: 10.1128/AAC.01207-20
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Top
  • Article
    • ABSTRACT
    • TEXT
    • RESULTS
    • DISCUSSION
    • MATERIALS AND METHODS
    • ACKNOWLEDGMENTS
    • FOOTNOTES
    • REFERENCES
  • Figures & Data
  • Info & Metrics
  • PDF

KEYWORDS

Mycobacterium tuberculosis
PK-PD index
PK-PD target
cumulative fraction of response
delamanid
hollow-fiber system model of tuberculosis

Related Articles

Cited By...

About

  • About AAC
  • Editor in Chief
  • Editorial Board
  • Policies
  • For Reviewers
  • For the Media
  • For Librarians
  • For Advertisers
  • Alerts
  • AAC Podcast
  • RSS
  • FAQ
  • Permissions
  • Journal Announcements

Authors

  • ASM Author Center
  • Submit a Manuscript
  • Article Types
  • Ethics
  • Contact Us

Follow #AACJournal

@ASMicrobiology

       

ASM Journals

ASM journals are the most prominent publications in the field, delivering up-to-date and authoritative coverage of both basic and clinical microbiology.

About ASM | Contact Us | Press Room

 

ASM is a member of

Scientific Society Publisher Alliance

 

American Society for Microbiology
1752 N St. NW
Washington, DC 20036
Phone: (202) 737-3600

Copyright © 2021 American Society for Microbiology | Privacy Policy | Website feedback

Print ISSN: 0066-4804; Online ISSN: 1098-6596