• No results found

University of Groningen Therapeutic drug monitoring Pranger, Anna Diewerke

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Therapeutic drug monitoring Pranger, Anna Diewerke"

Copied!
19
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Therapeutic drug monitoring

Pranger, Anna Diewerke

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from

it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date:

2018

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Pranger, A. D. (2018). Therapeutic drug monitoring: How to improve moxifloxacin exposure in tuberculosis

patients. Rijksuniversiteit Groningen.

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

4B

Chapter

Low moxifloxacin exposure

in male tuberculosis patients

and the need for monitoring

in early stages of treatment

A.D. Pranger, R. van Altena, O.W. Akkerman,W.C.M. de Lange, D. van Soolingen, D.J. Touw, D.R.A. Uges, T.S. van der Werf, J.G.W. Kosterink, and J.W.C. Alffenaar Submitted

(3)

Abstract Background

Moxifloxacin (MFX) is an important component of the current core rifampicin and multidrug-resistant tuberculosis treatment-regimen. However, low MFX exposure is frequently observed and is associated with poor response to treatment and acquired drug-resistance. We hypothesized that stage of disease and clinical condition during tuberculosis (TB) treatment influences MFX pharmacokinetics (PK).

Methods

We retrospectively reviewed the medical charts of adult TB patients treated with MFX 400 mg/day at our TB Centre from January 2006 to January 2013. Demographic, clinical, treatment and PK data were collected. We determined the correlation of time passed since start of TB treatment, and other possible explanatory variables, with MFX exposure. In patients with PK data on more than one day, we analyzed the impact of elapsed time since start of TB treatment on MFX exposure.

Results

Of 39 patients, a MFX AUC0-24h was available for analysis. The MFX exposure on 400mg QD

varied substantially from 10 to 73 mg*h*L-1. Median (IQR) time of PK sampling was after 50

(19-81) days of treatment. A disproportional increase of AUC0-24h over time was seen in

patients with PK data on more than one day. In multivariable analysis, rifampicin drug-drug interaction (P=0.004) and male gender (P=0.019) were independent predictors of low MFX exposure, but not duration of TB treatment.

Conclusions

In this TB cohort, time elapsed since start of treatment, as surrogate parameter of stage of disease, and other clinical variables, were not independent predictors of MFX exposure, but MFX AUC0-24h values changed over time for individual TB patients. Furthermore, our findings

confirm the well-known rifampicin drug-drug interaction, but also suggest that, particularly, male TB patients may benefit from MFX concentration monitoring.

Introduction

The global elimination of tuberculosis (TB) is seriously threatened by drug-resistant TB, with an estimated incidence of 600,000 cases resistant against the most powerful anti-TB agent (rifampicin) in 2016 (1). Also, patients with “extensively” and “totally” drug-resistant TB have been reported (2-4). To promote patients’ treatment compliance and thus to prevent spread of resistance against critical anti-TB agents, today’s TB drug pipeline is focused on shorter regimens for drug-susceptible and drug-resistant TB (5). However, to end the global TB epidemic, intensified efforts are needed as some of the patients acquire drug-resistance due to low drug-exposure while compliant to therapy (6-8).

Moxifloxacin (MFX) has a high bactericidal activity against M. tuberculosis and is therefore an established important component of the current core rifampicin and multidrug-resistant (MDR) treatment-regimen. Also, MFX is a useful fluoroquinolone if patients cannot tolerate one of the first-line anti-TB agents (9,10). A protein-unbound area under the concentration-time curve up to 24 hours post dosage (AUC0-24h) / minimal inhibitory concentration (MIC) of

53 is associated with optimal kill and suppression of drug-resistant mutant selection of M.

tuberculosis in log-phase growth, and 800mg/day instead of the registered daily dose of

400mg MFX is probably needed to attain this target in most of the TB patients (11). Furthermore, in our TB patients treated under direct observation (DOT), we reported a large variation in pharmacokinetics (PK) on 400mg MFX/day (12). Drug-drug interaction with rifampicin is one possible explanation for a low MFX exposure (13,14). However, a substantial MFX PK variability was also observed in a MDR-TB cohort (15). Other factors like co-morbid conditions (HIV, diabetes) or TB disease activity with wasting and loss of fat, lean body mass and reduced serum proteins, and reduced drug absorption resulting from intestinal dysfunction, could affect PK parameters of MFX (16,17). Data of the impact of malnutrition on MFX PK are lacking. However, a low geometric mean MFX exposure (25 mg*h*L-1) and a short, variable half-life (geometric mean: 8h, CV: 49%) on 400mg,

intravenously administered, was observed in patients with severe sepsis, compared to healthy volunteers (34 mg*h*L-1 and 15 h) (18,19).

We hypothesize that the stage of disease and clinical condition of TB patients influences MFX PK. Therefore, the objective of this study was to explore MFX PK variability over time during the course of TB treatment and its possible relation with different (clinical) variables.

(4)

Chapter

4

b

Abstract Background

Moxifloxacin (MFX) is an important component of the current core rifampicin and multidrug-resistant tuberculosis treatment-regimen. However, low MFX exposure is frequently observed and is associated with poor response to treatment and acquired drug-resistance. We hypothesized that stage of disease and clinical condition during tuberculosis (TB) treatment influences MFX pharmacokinetics (PK).

Methods

We retrospectively reviewed the medical charts of adult TB patients treated with MFX 400 mg/day at our TB Centre from January 2006 to January 2013. Demographic, clinical, treatment and PK data were collected. We determined the correlation of time passed since start of TB treatment, and other possible explanatory variables, with MFX exposure. In patients with PK data on more than one day, we analyzed the impact of elapsed time since start of TB treatment on MFX exposure.

Results

Of 39 patients, a MFX AUC0-24h was available for analysis. The MFX exposure on 400mg QD

varied substantially from 10 to 73 mg*h*L-1. Median (IQR) time of PK sampling was after 50

(19-81) days of treatment. A disproportional increase of AUC0-24h over time was seen in

patients with PK data on more than one day. In multivariable analysis, rifampicin drug-drug interaction (P=0.004) and male gender (P=0.019) were independent predictors of low MFX exposure, but not duration of TB treatment.

Conclusions

In this TB cohort, time elapsed since start of treatment, as surrogate parameter of stage of disease, and other clinical variables, were not independent predictors of MFX exposure, but MFX AUC0-24h values changed over time for individual TB patients. Furthermore, our findings

confirm the well-known rifampicin drug-drug interaction, but also suggest that, particularly, male TB patients may benefit from MFX concentration monitoring.

Introduction

The global elimination of tuberculosis (TB) is seriously threatened by drug-resistant TB, with an estimated incidence of 600,000 cases resistant against the most powerful anti-TB agent (rifampicin) in 2016 (1). Also, patients with “extensively” and “totally” drug-resistant TB have been reported (2-4). To promote patients’ treatment compliance and thus to prevent spread of resistance against critical anti-TB agents, today’s TB drug pipeline is focused on shorter regimens for drug-susceptible and drug-resistant TB (5). However, to end the global TB epidemic, intensified efforts are needed as some of the patients acquire drug-resistance due to low drug-exposure while compliant to therapy (6-8).

Moxifloxacin (MFX) has a high bactericidal activity against M. tuberculosis and is therefore an established important component of the current core rifampicin and multidrug-resistant (MDR) treatment-regimen. Also, MFX is a useful fluoroquinolone if patients cannot tolerate one of the first-line anti-TB agents (9,10). A protein-unbound area under the concentration-time curve up to 24 hours post dosage (AUC0-24h) / minimal inhibitory concentration (MIC) of

53 is associated with optimal kill and suppression of drug-resistant mutant selection of M.

tuberculosis in log-phase growth, and 800mg/day instead of the registered daily dose of

400mg MFX is probably needed to attain this target in most of the TB patients (11). Furthermore, in our TB patients treated under direct observation (DOT), we reported a large variation in pharmacokinetics (PK) on 400mg MFX/day (12). Drug-drug interaction with rifampicin is one possible explanation for a low MFX exposure (13,14). However, a substantial MFX PK variability was also observed in a MDR-TB cohort (15). Other factors like co-morbid conditions (HIV, diabetes) or TB disease activity with wasting and loss of fat, lean body mass and reduced serum proteins, and reduced drug absorption resulting from intestinal dysfunction, could affect PK parameters of MFX (16,17). Data of the impact of malnutrition on MFX PK are lacking. However, a low geometric mean MFX exposure (25 mg*h*L-1) and a short, variable half-life (geometric mean: 8h, CV: 49%) on 400mg,

intravenously administered, was observed in patients with severe sepsis, compared to healthy volunteers (34 mg*h*L-1 and 15 h) (18,19).

We hypothesize that the stage of disease and clinical condition of TB patients influences MFX PK. Therefore, the objective of this study was to explore MFX PK variability over time during the course of TB treatment and its possible relation with different (clinical) variables.

(5)

Patients and methods

Study design and data collection

We retrospectively studied TB patients, aged ≥ 18 years, receiving a once-daily dose of 400mg of MFX orally for at least five days (steady state) as part of their directly observed TB treatment at Tuberculosis Centre Beatrixoord, University Medical Center Groningen (Groningen, the Netherlands), between January 1st 2006 and January 1st 2013. Part of the

data (2006-2009) was collected earlier (12). In our Center, the attending chest physician prescribes a TB drug regimen individually tailored to the patients’ drug-tolerance and the drug susceptibility tests (DST) of the M. tuberculosis complex isolate. MFX PK curves were ordered based on clinical considerations, suspected drug-drug interactions, or part of study procedures of ongoing clinical trials (20-23).

Time points of start of clinical supervision, start of TB treatment, commencement of MFX treatment and of PK measurements were recorded and intervals were computed. At each time point, patients’ weight and length were collected. C-reactive protein (CRP) and Erythrocyte Sedimentation Rate (ESR) were collected at the moment of PK sampling. The most important drug-drug interaction (DDI) of MFX is with rifampicin (RIF), with estimated maximum liver enzyme induction after ten days of RIF-treatment and liver enzyme induction subsided two or more weeks after discontinuation of RIF-treatment (24). A RIF DDI thus was defined as ≥ 10 days of RIF-treatment or < 3 weeks washout after at least 10 days of RIF. Also, data on TB treatment, and demographic and other relevant data were collected, including geographic region of origin, co-infections (HIV, hepatitis B and hepatitis C) and diabetes mellitus.

As retrospective data were collected anonymously the Institutional Ethical Review Board of the University Medical Center Groningen waived the requirement for research subjects to give informed consent (METc 2013-492).

Pharmacokinetic parameters

MFX concentrations were determined using a validated liquid-chromatography tandem mass-spectrometry method (25). A plasma PK curve based on at least two concentration-measurements, one between 0.5 and 4 hours (absorption phase) and one more than 4 hours (elimination phase) post dosage was set as a criterion to have sufficient information about MFX PK. The AUC0-24h was estimated using an open one compartment population PK model

with Bayesian regression (26).

Correlations of variables with moxifloxacin exposure

To explore the stage of TB disease as a predictor of MFX AUC0-24h, we assessed the

correlation between the patients’ first MFX AUC0-24h (400 mg) and time since start of

treatment. Also, explanatory variables of disease severity were taken into account, such as BMI < 18.5, CRP and ESR. We compared the MFX AUC0-24h between groups, divided by

gender, RIF DDI, HIV co-infection, diabetes mellitus and geographical region of origin. Subsequently, all significant correlations, and/or factors suspected to influence PK, were eligible for multivariable analysis. Also because of the number of patients, we aimed to construct an optimal model reducing the redundant variables. If more than one PK curve was initiated, regardless of dosage, descriptive statistics were used to observe the dose-adjusted AUC0-24h in relation to time-span for each individual patient. MFX PK was expected to be

linear over the dose range 400 - 800mg QD (18,27). The AUC0-24h values on 600mg or

800mg QD were therefore considered to be dose-proportional compared to the AUC0-24h

values on 400mg QD.

Statistical analysis

Continuous variables were expressed as median plus interquartile range (IQR). For categorical variables, percentages of the category relative to the total group were described. Second, a Spearman correlation coefficient, or standardized β, using a simple linear regression, was calculated to determine correlations between continuous variables and MFX AUC0-24h or between categorical variables and MFX AUC0-24h, respectively. Finally, a

multivariable linear regression analysis was performed to determine any association between the outcome measure in the research question (i.e. AUC0-24h) and multiple variables. By

removing and hence correcting for non-significant variables, using a P-value of 0.100 or higher as criterion-to-remove, in descending P-value order, significant values, independently associated with outcome, will remain. In general, a two-sided P-value < 0.05 was considered statistically significant. The AUC0-24h value was log10-transformed for univariable and

multivariable analyses to meet the test assumptions. All statistical evaluations were performed using SPSS version 23.0 (IBM SPSS, Chicago, IL, USA).

(6)

Chapter

4

b

Patients and methods

Study design and data collection

We retrospectively studied TB patients, aged ≥ 18 years, receiving a once-daily dose of 400mg of MFX orally for at least five days (steady state) as part of their directly observed TB treatment at Tuberculosis Centre Beatrixoord, University Medical Center Groningen (Groningen, the Netherlands), between January 1st 2006 and January 1st 2013. Part of the

data (2006-2009) was collected earlier (12). In our Center, the attending chest physician prescribes a TB drug regimen individually tailored to the patients’ drug-tolerance and the drug susceptibility tests (DST) of the M. tuberculosis complex isolate. MFX PK curves were ordered based on clinical considerations, suspected drug-drug interactions, or part of study procedures of ongoing clinical trials (20-23).

Time points of start of clinical supervision, start of TB treatment, commencement of MFX treatment and of PK measurements were recorded and intervals were computed. At each time point, patients’ weight and length were collected. C-reactive protein (CRP) and Erythrocyte Sedimentation Rate (ESR) were collected at the moment of PK sampling. The most important drug-drug interaction (DDI) of MFX is with rifampicin (RIF), with estimated maximum liver enzyme induction after ten days of RIF-treatment and liver enzyme induction subsided two or more weeks after discontinuation of RIF-treatment (24). A RIF DDI thus was defined as ≥ 10 days of RIF-treatment or < 3 weeks washout after at least 10 days of RIF. Also, data on TB treatment, and demographic and other relevant data were collected, including geographic region of origin, co-infections (HIV, hepatitis B and hepatitis C) and diabetes mellitus.

As retrospective data were collected anonymously the Institutional Ethical Review Board of the University Medical Center Groningen waived the requirement for research subjects to give informed consent (METc 2013-492).

Pharmacokinetic parameters

MFX concentrations were determined using a validated liquid-chromatography tandem mass-spectrometry method (25). A plasma PK curve based on at least two concentration-measurements, one between 0.5 and 4 hours (absorption phase) and one more than 4 hours (elimination phase) post dosage was set as a criterion to have sufficient information about MFX PK. The AUC0-24h was estimated using an open one compartment population PK model

with Bayesian regression (26).

Correlations of variables with moxifloxacin exposure

To explore the stage of TB disease as a predictor of MFX AUC0-24h, we assessed the

correlation between the patients’ first MFX AUC0-24h (400 mg) and time since start of

treatment. Also, explanatory variables of disease severity were taken into account, such as BMI < 18.5, CRP and ESR. We compared the MFX AUC0-24h between groups, divided by

gender, RIF DDI, HIV co-infection, diabetes mellitus and geographical region of origin. Subsequently, all significant correlations, and/or factors suspected to influence PK, were eligible for multivariable analysis. Also because of the number of patients, we aimed to construct an optimal model reducing the redundant variables. If more than one PK curve was initiated, regardless of dosage, descriptive statistics were used to observe the dose-adjusted AUC0-24h in relation to time-span for each individual patient. MFX PK was expected to be

linear over the dose range 400 - 800mg QD (18,27). The AUC0-24h values on 600mg or

800mg QD were therefore considered to be dose-proportional compared to the AUC0-24h

values on 400mg QD.

Statistical analysis

Continuous variables were expressed as median plus interquartile range (IQR). For categorical variables, percentages of the category relative to the total group were described. Second, a Spearman correlation coefficient, or standardized β, using a simple linear regression, was calculated to determine correlations between continuous variables and MFX AUC0-24h or between categorical variables and MFX AUC0-24h, respectively. Finally, a

multivariable linear regression analysis was performed to determine any association between the outcome measure in the research question (i.e. AUC0-24h) and multiple variables. By

removing and hence correcting for non-significant variables, using a P-value of 0.100 or higher as criterion-to-remove, in descending P-value order, significant values, independently associated with outcome, will remain. In general, a two-sided P-value < 0.05 was considered statistically significant. The AUC0-24h value was log10-transformed for univariable and

multivariable analyses to meet the test assumptions. All statistical evaluations were performed using SPSS version 23.0 (IBM SPSS, Chicago, IL, USA).

(7)

Results Patients

From January 1st 2006 to January 1st 2013, 211 patients were treated with MFX for TB; 149

patients (70.6%) were classified as pulmonary TB. DST results (n = 179, 84.8%) revealed susceptibility for 1st-line drugs (n = 108, 60.3%) or multidrug-resistance (MDR) (n = 50, 28%)

in most patients. MFX exposure data were available in 39 patients. In 23/39 cases, patients had MDR-TB (Supplemental table 1). Patients’ baseline demographics are presented in

Table 1. Data on TB treatment are summarized in Supplemental table 1. Table 1: Baseline demographics of 39 patients with a known MFX exposure.

Parameter Value

Female 15 (38)

Age (years) 30 (23-42)

Weight (kg) 57.4 (49.4-62.1)

Body Mass Index (kg*m-2) 19.4 (17.9-20.3)

Geographic sub-region of origin1

East Africa 10 (26) West Asia 5 (13) West Africa 4 (10) East Asia 3 (8) South-East Asia 3 (8) East Europe 3 (8) South America 3 (8)

Rest of the world (n < 3 per sub-region) 8 (21) Comorbidities

Human immunodeficiency virus (HIV) 3 (8)

Hepatitis B 3 (8)

Hepatitis C 4 (10)

Diabetes Mellitus 3 (8)

Data are presented as n (%) or median (interquartile range).1United Nations classification.

Moxifloxacin exposure and correlations with variables

All parameters collected at the first moment of PK sampling are presented in Table 2. The

distribution of patients’ weight and BMI was not significantly different during PK sampling compared to baseline (Sign Test, P = 0.100). Median (IQR) MFX AUC0-24h (400mg QD) was

22 (16-31) mg*h*L-1 (Table 2). MFX AUC

0-24h displayed a large variation (range: 10-73

mg*h*L-1).

Table 2: Parameters at the moment of PK sampling (n=39).

Parameter Value

Dose (mg/kg) 6.66 (6.16-7.78)

Rifampicin drug-drug interaction 13 (33)‡

CRP (mg*L-1) 11 (<5 – 28)#

Time periods

Time-to-clinical supervision days 41 (22-76) Time-to-start of TB treatment days 50 (19-81) Time-to-start of Moxifloxacin days 26 (9-76) Pharmacokinetics

AUC0-24h (mg*h*L-1) 22 (16-31)

Data are presented as n (%) or median (interquartile range). ‡9/39 patients: unknown. #24/39 patients:

unknown; CRP (or ESR) is no routine marker of TB treatment response at Beatrixoord. In case of an unknown CRP, ESR was unknown as well.

Univariable analysis identified gender, a RIF DDI, an HIV co-infection and time-to-start of TB treatment as strong correlates of log10 MFX AUC0-24h (Table 3). Also, the MFX AUC0-24h of the

TB patients with Diabetes Mellitus was relatively low; 11, 13 and 24 mg*h*L-1.

Model 1 (Table 4) was the optimal multivariable linear regression model. RIF drug-drug

interaction and gender were identified as significant independent predictors of MFX AUC0-24h.

The model did not improve by including the time of PK measurement since start of TB treatment (Model 2, Table 4). RIF continued to be the strongest correlate (standardized β =

-0.46, p = 0.004). In model 1, the effect size B (95% CI) of RIF on log10AUC0-24h was estimated

as -0.18 (-0.29 - -0.062), i.e. MFX exposure was reduced from 25 (95% CI, 20-31) to 17 mg*h*L-1. Similarly, an effect size B of 0.14 (95% CI: 0.025-0.25) resulted in an estimated

MFX exposure for female patients of 100.14 x 100% = 138% compared to men (100%). The

third determinant of our model, HIV co-infection, was associated with a low MFX exposure (B = -0.18, 95% CI -0.38 – 0.030).

(8)

Chapter

4

b

Results Patients

From January 1st 2006 to January 1st 2013, 211 patients were treated with MFX for TB; 149

patients (70.6%) were classified as pulmonary TB. DST results (n = 179, 84.8%) revealed susceptibility for 1st-line drugs (n = 108, 60.3%) or multidrug-resistance (MDR) (n = 50, 28%)

in most patients. MFX exposure data were available in 39 patients. In 23/39 cases, patients had MDR-TB (Supplemental table 1). Patients’ baseline demographics are presented in

Table 1. Data on TB treatment are summarized in Supplemental table 1. Table 1: Baseline demographics of 39 patients with a known MFX exposure.

Parameter Value

Female 15 (38)

Age (years) 30 (23-42)

Weight (kg) 57.4 (49.4-62.1)

Body Mass Index (kg*m-2) 19.4 (17.9-20.3)

Geographic sub-region of origin1

East Africa 10 (26) West Asia 5 (13) West Africa 4 (10) East Asia 3 (8) South-East Asia 3 (8) East Europe 3 (8) South America 3 (8)

Rest of the world (n < 3 per sub-region) 8 (21) Comorbidities

Human immunodeficiency virus (HIV) 3 (8)

Hepatitis B 3 (8)

Hepatitis C 4 (10)

Diabetes Mellitus 3 (8)

Data are presented as n (%) or median (interquartile range).1United Nations classification.

Moxifloxacin exposure and correlations with variables

All parameters collected at the first moment of PK sampling are presented in Table 2. The

distribution of patients’ weight and BMI was not significantly different during PK sampling compared to baseline (Sign Test, P = 0.100). Median (IQR) MFX AUC0-24h (400mg QD) was

22 (16-31) mg*h*L-1 (Table 2). MFX AUC

0-24h displayed a large variation (range: 10-73

mg*h*L-1).

Table 2: Parameters at the moment of PK sampling (n=39).

Parameter Value

Dose (mg/kg) 6.66 (6.16-7.78)

Rifampicin drug-drug interaction 13 (33)‡

CRP (mg*L-1) 11 (<5 – 28)#

Time periods

Time-to-clinical supervision days 41 (22-76) Time-to-start of TB treatment days 50 (19-81) Time-to-start of Moxifloxacin days 26 (9-76) Pharmacokinetics

AUC0-24h (mg*h*L-1) 22 (16-31)

Data are presented as n (%) or median (interquartile range). ‡9/39 patients: unknown. #24/39 patients:

unknown; CRP (or ESR) is no routine marker of TB treatment response at Beatrixoord. In case of an unknown CRP, ESR was unknown as well.

Univariable analysis identified gender, a RIF DDI, an HIV co-infection and time-to-start of TB treatment as strong correlates of log10 MFX AUC0-24h (Table 3). Also, the MFX AUC0-24h of the

TB patients with Diabetes Mellitus was relatively low; 11, 13 and 24 mg*h*L-1.

Model 1 (Table 4) was the optimal multivariable linear regression model. RIF drug-drug

interaction and gender were identified as significant independent predictors of MFX AUC0-24h.

The model did not improve by including the time of PK measurement since start of TB treatment (Model 2, Table 4). RIF continued to be the strongest correlate (standardized β =

-0.46, p = 0.004). In model 1, the effect size B (95% CI) of RIF on log10AUC0-24h was estimated

as -0.18 (-0.29 - -0.062), i.e. MFX exposure was reduced from 25 (95% CI, 20-31) to 17 mg*h*L-1. Similarly, an effect size B of 0.14 (95% CI: 0.025-0.25) resulted in an estimated

MFX exposure for female patients of 100.14 x 100% = 138% compared to men (100%). The

third determinant of our model, HIV co-infection, was associated with a low MFX exposure (B = -0.18, 95% CI -0.38 – 0.030).

(9)

Table 3: Univariable correlates of Moxifloxacin AUC0-24h.

Determinant Spearman’s rho Standardized β P-value

Female 0.45 0.004

Asian1,2 0.16$ 0.363

European1,2 0.037$ 0.833

American1,2 0.27$ 0.118

Human immunodeficiency virus (HIV)3 -0.36 0.027

Diabetes Mellitus3 -0.28 0.081

Rifampicin drug-drug interaction -0.58 0.001

BMI < 18.5 -0.18 0.313

Time-to-start of TB treatment ρ= 0.36 0.025

AUC0-24h was log10 transformed. 1regrouped into United Nations’ macro geographical regions: Africa,

Asia, Europe, America to create valid dummy variables. African patients originated mainly from Sub-Saharan Africa (n=15/17). Three out of four American patients originated from Suriname. 2compared

to African. 3n patients with determinant small (Table 1). $By exclusion of a visible outlier (American)

standardized β (p) for determinants Asian, European and American were 0.18 (0.334), 0.040 (0.823), 0.076 (0.670), respectively.

Table 4: Multivariable linear regression analysis: determinants of MFX AUC0-24h.

Model Determinant Standardized β P-value R2

1 Rifampicin drug-drug interaction -0.46 0.004 0.51

Female 0.35 0.019

Human immunodeficiency virus (HIV) -0.25 0.091

2 Rifampicin drug-drug interaction -0.44 0.005 0.52

Female 0.36 0.019

Human immunodeficiency virus (HIV) -0.23 0.117

Time-to-start of TB treatment 0.10 0.476

AUC0-24h was log10 transformed. Model 1 is the optimal regression model. Model 2 assessed the effect

of all significant variables from the univariable analysis.

Of 9 patients multiple MFX AUC0-24h values were available during their TB treatment (Table 5). In some patients the MFX exposure was disproportionally higher after more treatment

days. For example, on 800mg/day the AUC0-24h increased from 33 to 59 mg*h*L-1 (179%) in

63 days, but almost the same MFX exposure (57 mg*h*L-1) was measured after 28 more

treatment days for patient 7, without an HIV co-infection or RIF DDI. Also, an equal MFX exposure was observed for patient 5 on 600 and 800mg/day with 47 days in between, without an HIV co-infection and RIF DDI.

Table 5: Patients with more than one PK curve (n=9).

Patient Moment 1 Moment 2 Moment 3 Moment 4 Determinants1

Days AUC Days AUC Days AUC Days AUC

1 414 21 448 22 - - - 2 7 15(X) 24 22* - - HIV 3 6 15(X) 48 37* 89 22 - Female, HIV 4 20 25(X) 40 36 - - - 5 6 13(R) 161 46** 208 47* - - 6 8 22(X) 36 53* - - Female 7 19 20(X) 49 33** 112 59** 140 57** Female 8 82 31 172 42* 208 55* - - 9 41 62* 103 48 - - Female

MFX AUC0-24h on 400 mg/day unless indicated otherwise. *600mg/day. **800mg/day. AUC, indication

of disproportional PK compared to the previous AUC. (R), rifampicin drug-drug interaction (RIF DDI). (X), RIF DDI due to any pre-clinical RIF treatment unknown. 1female, HIV and/or Diabetes Mellitus.

(10)

Chapter

4

b

Table 3: Univariable correlates of Moxifloxacin AUC0-24h.

Determinant Spearman’s rho Standardized β P-value

Female 0.45 0.004

Asian1,2 0.16$ 0.363

European1,2 0.037$ 0.833

American1,2 0.27$ 0.118

Human immunodeficiency virus (HIV)3 -0.36 0.027

Diabetes Mellitus3 -0.28 0.081

Rifampicin drug-drug interaction -0.58 0.001

BMI < 18.5 -0.18 0.313

Time-to-start of TB treatment ρ= 0.36 0.025

AUC0-24h was log10 transformed. 1regrouped into United Nations’ macro geographical regions: Africa,

Asia, Europe, America to create valid dummy variables. African patients originated mainly from Sub-Saharan Africa (n=15/17). Three out of four American patients originated from Suriname. 2compared

to African. 3n patients with determinant small (Table 1). $By exclusion of a visible outlier (American)

standardized β (p) for determinants Asian, European and American were 0.18 (0.334), 0.040 (0.823), 0.076 (0.670), respectively.

Table 4: Multivariable linear regression analysis: determinants of MFX AUC0-24h.

Model Determinant Standardized β P-value R2

1 Rifampicin drug-drug interaction -0.46 0.004 0.51

Female 0.35 0.019

Human immunodeficiency virus (HIV) -0.25 0.091

2 Rifampicin drug-drug interaction -0.44 0.005 0.52

Female 0.36 0.019

Human immunodeficiency virus (HIV) -0.23 0.117

Time-to-start of TB treatment 0.10 0.476

AUC0-24h was log10 transformed. Model 1 is the optimal regression model. Model 2 assessed the effect

of all significant variables from the univariable analysis.

Of 9 patients multiple MFX AUC0-24h values were available during their TB treatment (Table 5). In some patients the MFX exposure was disproportionally higher after more treatment

days. For example, on 800mg/day the AUC0-24h increased from 33 to 59 mg*h*L-1 (179%) in

63 days, but almost the same MFX exposure (57 mg*h*L-1) was measured after 28 more

treatment days for patient 7, without an HIV co-infection or RIF DDI. Also, an equal MFX exposure was observed for patient 5 on 600 and 800mg/day with 47 days in between, without an HIV co-infection and RIF DDI.

Table 5: Patients with more than one PK curve (n=9).

Patient Moment 1 Moment 2 Moment 3 Moment 4 Determinants1

Days AUC Days AUC Days AUC Days AUC

1 414 21 448 22 - - - 2 7 15(X) 24 22* - - HIV 3 6 15(X) 48 37* 89 22 - Female, HIV 4 20 25(X) 40 36 - - - 5 6 13(R) 161 46** 208 47* - - 6 8 22(X) 36 53* - - Female 7 19 20(X) 49 33** 112 59** 140 57** Female 8 82 31 172 42* 208 55* - - 9 41 62* 103 48 - - Female

MFX AUC0-24h on 400 mg/day unless indicated otherwise. *600mg/day. **800mg/day. AUC, indication

of disproportional PK compared to the previous AUC. (R), rifampicin drug-drug interaction (RIF DDI). (X), RIF DDI due to any pre-clinical RIF treatment unknown. 1female, HIV and/or Diabetes Mellitus.

(11)

Discussion

The main finding of our multivariable analysis is that not time elapsed since start of TB treatment, as surrogate parameter of stage of disease, but rifampicin drug-drug interaction (P=0.004) and gender (P=0.019) were independent predictors of MFX exposure. An HIV co-infection (P=0.091) was associated with a lower MFX AUC0-24h. Second, in some individual

patients with multiple PK curves, without RIF DDI, we clearly observed a disproportional increase of the dose-adjusted MFX AUC0-24h over time.

To our knowledge a gender-related difference in MFX concentration has not been previously described, but relatively high concentrations of other anti-TB drugs have been found in female TB patients (28,29). The lower MFX AUC0-24h in our male TB patients tends to be

(partly) driven by a lower peak-level (Figure 1), indicating a reduced MFX bioavailability. A

gender-difference in intestinal P-glycoprotein expression due to a worse clinical condition (17), could be a possible mechanism for a reduced MFX absorption in male patients. A prospective crossover PK study of oral and intravenous administration of MFX in TB patients, investigating a gender-difference in absolute bioavailability, is needed to test this hypothesis.

Figure 1: Moxifloxacin peak-level in male (n=24) and female (n=15) TB patients.

Low concentrations of first-line anti-TB drugs are not uncommon in HIV co-infected patients, but the underlying cause is not entirely unraveled (30). Drug-absorption might be influenced by acquired immune deficiency syndrome (AIDS)-related intestinal malfunction, which often presents as diarrhea (16,31). Data on anti-retroviral drugs influencing MFX PK is limited to one recent population PK study in TB patients estimating a -30% change in MFX exposure and a +42% change in hepatic clearance in HIV co-infected patients treated with efavirenz, possibly due to induction of UDP-glucuronosyltransferase (32). Two-out-of-three HIV co-infected patients in our cohort were on efavirenz-based antiretroviral treatment during the period of PK sampling, and for one of them defecation problems, for which no specific cause could be found, were also reported. The pharmacological challenge of treating both HIV and TB with an effective and less-toxic drug-regimen for a longer period warrants for more knowledge on potential PK DDI’s.

In our TB center, a TB drug regimen is individually tailored to the patients’ drug-tolerance and the drug susceptibility tests. Therefore, by using the surrogate parameter ‘time of PK measurement since start of TB treatment’, the assumption was made that every patient was treated with a regimen of initially effective anti-TB drugs with equal gradual improvement of their clinical condition. Although this surrogate parameter did not emerge in our cohort by correcting for a RIF DDI, gender and an HIV co-infection (n=39, Table 4), a disproportional

increase of MFX AUC0-24h was observed over time for individual patients with multiple PK

curves (n=9, Table 5). A longitudinal analysis of the effect of inflammation and nutrition on

MFX PK in a larger cohort might therefore be more informative.

Our study has several limitations. Out of 211 patients treated with MFX, a MFX AUC0-24h

value was available in only 39 patients. First, the small sample size is a limitation of the intensive PK evaluation needed to assess MFX AUC0-24h (see method section). Also, PK

sampling was typically driven by suspected problems with drug exposure, e.g. a RIF DDI or a high MIC value, or by the study procedure of clinical trials, investigating safety and efficacy of an escalated MFX dose (20,22), evaluating dried-blood spot sampling (21), or describing the population PK of anti-TB drugs (23). The retrospective nature of our study makes these results thus sensitive for bias incurred by selection by indication. However, the PK variability observed in this broad selection of patients, only partly explained by three variables, justifies therapeutic drug monitoring (TDM) in patients at risk for a mismatch between exposure and antimicrobial activity. The significant impact of the well-known RIF DDI (13,14) supports the

(12)

Chapter

4

b

Discussion

The main finding of our multivariable analysis is that not time elapsed since start of TB treatment, as surrogate parameter of stage of disease, but rifampicin drug-drug interaction (P=0.004) and gender (P=0.019) were independent predictors of MFX exposure. An HIV co-infection (P=0.091) was associated with a lower MFX AUC0-24h. Second, in some individual

patients with multiple PK curves, without RIF DDI, we clearly observed a disproportional increase of the dose-adjusted MFX AUC0-24h over time.

To our knowledge a gender-related difference in MFX concentration has not been previously described, but relatively high concentrations of other anti-TB drugs have been found in female TB patients (28,29). The lower MFX AUC0-24h in our male TB patients tends to be

(partly) driven by a lower peak-level (Figure 1), indicating a reduced MFX bioavailability. A

gender-difference in intestinal P-glycoprotein expression due to a worse clinical condition (17), could be a possible mechanism for a reduced MFX absorption in male patients. A prospective crossover PK study of oral and intravenous administration of MFX in TB patients, investigating a gender-difference in absolute bioavailability, is needed to test this hypothesis.

Figure 1: Moxifloxacin peak-level in male (n=24) and female (n=15) TB patients.

Low concentrations of first-line anti-TB drugs are not uncommon in HIV co-infected patients, but the underlying cause is not entirely unraveled (30). Drug-absorption might be influenced by acquired immune deficiency syndrome (AIDS)-related intestinal malfunction, which often presents as diarrhea (16,31). Data on anti-retroviral drugs influencing MFX PK is limited to one recent population PK study in TB patients estimating a -30% change in MFX exposure and a +42% change in hepatic clearance in HIV co-infected patients treated with efavirenz, possibly due to induction of UDP-glucuronosyltransferase (32). Two-out-of-three HIV co-infected patients in our cohort were on efavirenz-based antiretroviral treatment during the period of PK sampling, and for one of them defecation problems, for which no specific cause could be found, were also reported. The pharmacological challenge of treating both HIV and TB with an effective and less-toxic drug-regimen for a longer period warrants for more knowledge on potential PK DDI’s.

In our TB center, a TB drug regimen is individually tailored to the patients’ drug-tolerance and the drug susceptibility tests. Therefore, by using the surrogate parameter ‘time of PK measurement since start of TB treatment’, the assumption was made that every patient was treated with a regimen of initially effective anti-TB drugs with equal gradual improvement of their clinical condition. Although this surrogate parameter did not emerge in our cohort by correcting for a RIF DDI, gender and an HIV co-infection (n=39, Table 4), a disproportional

increase of MFX AUC0-24h was observed over time for individual patients with multiple PK

curves (n=9, Table 5). A longitudinal analysis of the effect of inflammation and nutrition on

MFX PK in a larger cohort might therefore be more informative.

Our study has several limitations. Out of 211 patients treated with MFX, a MFX AUC0-24h

value was available in only 39 patients. First, the small sample size is a limitation of the intensive PK evaluation needed to assess MFX AUC0-24h (see method section). Also, PK

sampling was typically driven by suspected problems with drug exposure, e.g. a RIF DDI or a high MIC value, or by the study procedure of clinical trials, investigating safety and efficacy of an escalated MFX dose (20,22), evaluating dried-blood spot sampling (21), or describing the population PK of anti-TB drugs (23). The retrospective nature of our study makes these results thus sensitive for bias incurred by selection by indication. However, the PK variability observed in this broad selection of patients, only partly explained by three variables, justifies therapeutic drug monitoring (TDM) in patients at risk for a mismatch between exposure and antimicrobial activity. The significant impact of the well-known RIF DDI (13,14) supports the validity of our multivariable linear regression model.

(13)

Conclusions

In conclusion, the MFX AUC0-24h on 400mg/day orally is highly variable. Low MFX exposure

was found in male TB patients and in patients with a RIF-based TB regimen. The AUC0-24h

tended to be lower in HIV-TB patients and at the early stage of TB disease.

References

1. World Health Organization (WHO) (ed.). 2017. Global tuberculosis report 2017. World Health Organization, Geneva, Switzerland.

2. Migliori G.B., De laco G., Besozzi G., Centis R., and Cirillo D.M. 2007. First tuberculosis cases in Italy resistant to all tested drugs. Eurosurveillance. 12(5):E070517.1.

3. Velyati A.A., Masjedi M.R., Farnia P., Tabarsi P., Ghanavi J., Ziazarifi A.H., and Hoffner S.E. 2009. Emergence of new forms of totally drug-resistant tuberculosis bacilli: super extensively drug-resistant tuberculosis or totally drug-resistant strains in iran. Chest. 136(2):420-5. 4. Udwadia Z.F., Amale R.A., Ajbani K.K., and Rodrigues C. 2012. Totally drug-resistant

tuberculosis in India. Clin Infect Dis. 54(4):579-81.

5. Stop TB Partnership (ed.). Drug pipeline. Stop TB Partnership. Accessed: March 18th 2018

via www.newtbdrugs.org.

6. Srivastava S., Pasipanodya J.G., Meek C., Leff R., and Gumbo T. 2011.Multidrug-resistant tuberculosis not due to non-compliance but due to between-patient pharmacokinetic variability. J Infect Dis. 204(12):1951-9.

7. Alffenaar J.C., Migliori G.B., and Gumbo T. 2017.Multidrug-resistant tuberculosis: pharmacokinetic and pharmacodynamic science. Lancet Infect Dis. 17(9):898.

8. Sharma A., Hill A., Kurbatova E., van der Walt M., Kvasnovsky C., Tupasi T.E., Caoili J.C., Gler M.T., Volchenkov G.V., Kazennyy B.Y., Demikhova O.V., Bayona J., Contreras C., Yagui M., Leimane V., Cho S.N., Kim H.J., Kliiman K., Akksilp S., Jou R., Ershova J., Dalton T., Cegielski P.; Global Preserving Effective TB Treatment Study Investigators. 2017. Estimating the future burden of multidrug-resistant and extensively drug-resistant tuberculosis in India, the Philippines, Russia, and South Africa: a mathematical modelling study. Lancet Infect Dis. 17(7):707-715.

9. Pranger A.D., Alffenaar J.W., and Aarnoutse R.E. 2011. Fluoroquinolones, the cornerstone of treatment of drug-resistant tuberculosis: a pharmacokinetic and pharmacodynamic approach. Curr Pharm Des. 17(27):2900-30.

10. World Health Organization (WHO) (ed.). 2016. WHO treatment guidelines for drug-resistant tuberculosis. World Health Organization, Geneva, Switzerland.

11. Gumbo T., Louie A., Deziel M.R., Parsons L.M., Salfinger M., and Drusano G.L. 2004. Selection of a moxifloxacin dose that suppresses drug resistance in Mycobacterium tuberculosis, by use of an in vitro pharmacodynamic infection model and mathematical modeling. J Infect Dis. 190(9):1642-51.

12. Pranger A.D., van Altena R., Aarnoutse R.E., van Soolingen D., Uges D.R., Kosterink J.G., van der Werf T.S., and Alffenaar J.W. 2011. Evaluation of moxifloxacin for the treatment of tuberculosis: 3 years of experience. Eur Respir J. 38(4):888-94.

13. Nijland H.M., Ruslami R., Suroto A.J., Burger D.M., Alisjahbana B., van Crevel R., and Aarnoutse R.E. 2007. Rifampicin reduces plasma concentrations of moxifloxacin in patients with tuberculosis. Clin Infect Dis. 45(8): 1001-7.

(14)

Chapter

4

b

Conclusions

In conclusion, the MFX AUC0-24h on 400mg/day orally is highly variable. Low MFX exposure

was found in male TB patients and in patients with a RIF-based TB regimen. The AUC0-24h

tended to be lower in HIV-TB patients and at the early stage of TB disease.

References

1. World Health Organization (WHO) (ed.). 2017. Global tuberculosis report 2017. World Health Organization, Geneva, Switzerland.

2. Migliori G.B., De laco G., Besozzi G., Centis R., and Cirillo D.M. 2007. First tuberculosis cases in Italy resistant to all tested drugs. Eurosurveillance. 12(5):E070517.1.

3. Velyati A.A., Masjedi M.R., Farnia P., Tabarsi P., Ghanavi J., Ziazarifi A.H., and Hoffner S.E. 2009. Emergence of new forms of totally drug-resistant tuberculosis bacilli: super extensively drug-resistant tuberculosis or totally drug-resistant strains in iran. Chest. 136(2):420-5. 4. Udwadia Z.F., Amale R.A., Ajbani K.K., and Rodrigues C. 2012. Totally drug-resistant

tuberculosis in India. Clin Infect Dis. 54(4):579-81.

5. Stop TB Partnership (ed.). Drug pipeline. Stop TB Partnership. Accessed: March 18th 2018

via www.newtbdrugs.org.

6. Srivastava S., Pasipanodya J.G., Meek C., Leff R., and Gumbo T. 2011.Multidrug-resistant tuberculosis not due to non-compliance but due to between-patient pharmacokinetic variability. J Infect Dis. 204(12):1951-9.

7. Alffenaar J.C., Migliori G.B., and Gumbo T. 2017.Multidrug-resistant tuberculosis: pharmacokinetic and pharmacodynamic science. Lancet Infect Dis. 17(9):898.

8. Sharma A., Hill A., Kurbatova E., van der Walt M., Kvasnovsky C., Tupasi T.E., Caoili J.C., Gler M.T., Volchenkov G.V., Kazennyy B.Y., Demikhova O.V., Bayona J., Contreras C., Yagui M., Leimane V., Cho S.N., Kim H.J., Kliiman K., Akksilp S., Jou R., Ershova J., Dalton T., Cegielski P.; Global Preserving Effective TB Treatment Study Investigators. 2017. Estimating the future burden of multidrug-resistant and extensively drug-resistant tuberculosis in India, the Philippines, Russia, and South Africa: a mathematical modelling study. Lancet Infect Dis. 17(7):707-715.

9. Pranger A.D., Alffenaar J.W., and Aarnoutse R.E. 2011. Fluoroquinolones, the cornerstone of treatment of drug-resistant tuberculosis: a pharmacokinetic and pharmacodynamic approach. Curr Pharm Des. 17(27):2900-30.

10. World Health Organization (WHO) (ed.). 2016. WHO treatment guidelines for drug-resistant tuberculosis. World Health Organization, Geneva, Switzerland.

11. Gumbo T., Louie A., Deziel M.R., Parsons L.M., Salfinger M., and Drusano G.L. 2004. Selection of a moxifloxacin dose that suppresses drug resistance in Mycobacterium tuberculosis, by use of an in vitro pharmacodynamic infection model and mathematical modeling. J Infect Dis. 190(9):1642-51.

12. Pranger A.D., van Altena R., Aarnoutse R.E., van Soolingen D., Uges D.R., Kosterink J.G., van der Werf T.S., and Alffenaar J.W. 2011. Evaluation of moxifloxacin for the treatment of tuberculosis: 3 years of experience. Eur Respir J. 38(4):888-94.

13. Nijland H.M., Ruslami R., Suroto A.J., Burger D.M., Alisjahbana B., van Crevel R., and Aarnoutse R.E. 2007. Rifampicin reduces plasma concentrations of moxifloxacin in patients with tuberculosis. Clin Infect Dis. 45(8): 1001-7.

(15)

14. Weiner M., Burman W., Luo C.C., Peloquin C.A., Engle M., Goldberg S., Agarwal V., and Vernon A. 2007. Effects of rifampin and multidrug resistance gene polymorphism on concentrations of moxifloxacin. Antimicrob Agents Chemother. 51(8):2861-6.

15. Manika K., Chatzika K., Zaroqoulidis K., and Kioumis I. 2012. Moxifloxacin in multidrug-resistant tuberculosis: is there any indication for therapeutic drug monitoring? Eur Respir J. 40(4):1051-3.

16. Daskapan A., de Lange W.C., Akkerman O.W., Kosterink J.G., van der Werf T.S., Stienstra Y., and Alffenaar J.W. 2015. The role of therapeutic drug monitoring in individualised drug dosage and exposure measurement in tuberculosis and HIV co-infection. Eur Respir J. 45(2): 569-71.

17. Macallan D.C. 1999. Malnutrition in tuberculosis. Diagn Microbiol Infect Dis. 34(2):153-7. 18. Stass H., Kubitza D., and Schühly U. 2001. Pharmacokinetics, safety and tolerability of

moxifloxacin, a novel 8-methoxyfluoroquinolone, after repeated oral administration. Clin Pharmacokinet. 40 Suppl 1:1-9.

19. Pletz M.W., Bloos F., Burkhardt O., Brunkhorst F.M., Bode-Böger S.M., Martens-Lobenhoffer J., Greer M.W., Stass H, and Welte T. 2010. Pharmacokinetics of moxifloxacin in patients with severe sepsis or septic shock. Intensive Care Med. 36(6):979-83.

20. National Institute of Health (NIH) (ed.). ClinicalTrials.gov Id. NCT01329250. U.S. National Library of Health, Bethesda, United States of America. Accessed: March 24th 2018 via

clinicaltrials.gov.

21. Vu D.H., Koster R.A., Alffenaar J.W., Brouwers J.R., and Uges D.R. 2011. Determination of moxifloxacin in dried blood spots using LC-MS/MS and the impact of hematocrit and blood volume. J Chromatogr B Analyt Technol Biomed Life Sci. 879(15-16):1063-70.

22. Alffenaar J.W., van Altena R., Bökkerink H.J., Luijckx G.J., van Soolingen D., Aarnoutse R.E., and van der Werf T.S. 2009. Pharmacokinetics of moxifloxacin in cerebrospinal fluid and plasma in patients with tuberculous meningitis. Clin Infect Dis. 49(7):1080-2.

23. Magis-Escurra C., Later-Nijland H.M., Alffenaar J.W., Broeders J., Burger D.M., van Crevel R., Boeree M.J., Donders A.R., van Altena R., van der Werf T.S., and Aarnoutse R.E. 2014. Population pharmacokinetics and limited sampling strategy for first-line tuberculosis drugs and moxifloxacin. Int J Antimicrob Agents. 44(3):229-34.

24. Medicines Evaluation Board (MEB) (ed.). 2017. Summary of product characteristics. Rifampicin. Medicines Evaluation Board, Utrecht, The Netherlands.

25. Pranger A.D., Alffenaar J.W., Wessels A.M., Greijdanus B., and Uges D.R. 2010.

Determination of moxifloxacin in human plasma, plasma ultrafiltrate, and cerebrospinal fluid by a rapid and simple liquid chromatography-tandem mass spectrometry method. J Anal Toxicol. 34(3):135-41.

26. Pranger A.D., Kosterink J.G., van Altena R., Aarnoutse R.E., van der Werf T.S., Uges D.R., and Alffenaar J.W. 2011. Limited-sampling strategies for therapeutic drug monitoring of moxifloxacin in patients with tuberculosis. Ther Drug Monit. 33(3):350-4.

27. Stass H. and Kubitza D. 1999. Pharmacokinetics and elimination of moxifloxacin after oral and intravenous administration in man. J Antimicrob Chemother. 43 Suppl B:83-90.

28. Ray J., Gardiner I., and Marriott D. 2003. Managing antituberculosis drug therapy by therapeutic drug monitoring of rifampicin and isoniazid. Intern Med J. 33(5-6):229-34. 29. Mcllleron H., Wash P., Burger A., Norman J., Folb P.I., and Smith P. 2006. Determinants of

rifampin, isoniazid, pyrazinamide, and ethambutol pharmacokinetics in a cohort of tuberculosis patients. Antimicrob Agents Chemother. 50(4):1170-7.

30. Verbeeck R.K, Günther G., Kibuule D., Hunter C., and Rennie T.W. 2016. Optimizing treatment outcome of first-line anti-tuberculosis drugs: the role of therapeutic drug monitoring. Eur J Clin Pharmacol. 72(8):905-16.

31. Cello J.P. and Day L.W. 2009. Idiopathic AIDS enteropathy and treatment of gasterointestinal oppertunistic pathogens. Gastroenterology. 136(6):1952-65.

32. Naidoo A., Chirehwa M., Mcllleron H., Naidoo K., Essack S., Yende-Zuma N., Kimba-Phongi E., Adamson J., Govender K., Padayatchi N., and Denti P. 2017. Effect of rifampicin and efavirenz on moxifloxacin concentrations when co-administered in patients with drug-susceptible TB. J Antimicrob Chemother. 72(5):1441-1449.

(16)

Chapter

4

b

14. Weiner M., Burman W., Luo C.C., Peloquin C.A., Engle M., Goldberg S., Agarwal V., and Vernon A. 2007. Effects of rifampin and multidrug resistance gene polymorphism on concentrations of moxifloxacin. Antimicrob Agents Chemother. 51(8):2861-6.

15. Manika K., Chatzika K., Zaroqoulidis K., and Kioumis I. 2012. Moxifloxacin in multidrug-resistant tuberculosis: is there any indication for therapeutic drug monitoring? Eur Respir J. 40(4):1051-3.

16. Daskapan A., de Lange W.C., Akkerman O.W., Kosterink J.G., van der Werf T.S., Stienstra Y., and Alffenaar J.W. 2015. The role of therapeutic drug monitoring in individualised drug dosage and exposure measurement in tuberculosis and HIV co-infection. Eur Respir J. 45(2): 569-71.

17. Macallan D.C. 1999. Malnutrition in tuberculosis. Diagn Microbiol Infect Dis. 34(2):153-7. 18. Stass H., Kubitza D., and Schühly U. 2001. Pharmacokinetics, safety and tolerability of

moxifloxacin, a novel 8-methoxyfluoroquinolone, after repeated oral administration. Clin Pharmacokinet. 40 Suppl 1:1-9.

19. Pletz M.W., Bloos F., Burkhardt O., Brunkhorst F.M., Bode-Böger S.M., Martens-Lobenhoffer J., Greer M.W., Stass H, and Welte T. 2010. Pharmacokinetics of moxifloxacin in patients with severe sepsis or septic shock. Intensive Care Med. 36(6):979-83.

20. National Institute of Health (NIH) (ed.). ClinicalTrials.gov Id. NCT01329250. U.S. National Library of Health, Bethesda, United States of America. Accessed: March 24th 2018 via

clinicaltrials.gov.

21. Vu D.H., Koster R.A., Alffenaar J.W., Brouwers J.R., and Uges D.R. 2011. Determination of moxifloxacin in dried blood spots using LC-MS/MS and the impact of hematocrit and blood volume. J Chromatogr B Analyt Technol Biomed Life Sci. 879(15-16):1063-70.

22. Alffenaar J.W., van Altena R., Bökkerink H.J., Luijckx G.J., van Soolingen D., Aarnoutse R.E., and van der Werf T.S. 2009. Pharmacokinetics of moxifloxacin in cerebrospinal fluid and plasma in patients with tuberculous meningitis. Clin Infect Dis. 49(7):1080-2.

23. Magis-Escurra C., Later-Nijland H.M., Alffenaar J.W., Broeders J., Burger D.M., van Crevel R., Boeree M.J., Donders A.R., van Altena R., van der Werf T.S., and Aarnoutse R.E. 2014. Population pharmacokinetics and limited sampling strategy for first-line tuberculosis drugs and moxifloxacin. Int J Antimicrob Agents. 44(3):229-34.

24. Medicines Evaluation Board (MEB) (ed.). 2017. Summary of product characteristics. Rifampicin. Medicines Evaluation Board, Utrecht, The Netherlands.

25. Pranger A.D., Alffenaar J.W., Wessels A.M., Greijdanus B., and Uges D.R. 2010.

Determination of moxifloxacin in human plasma, plasma ultrafiltrate, and cerebrospinal fluid by a rapid and simple liquid chromatography-tandem mass spectrometry method. J Anal Toxicol. 34(3):135-41.

26. Pranger A.D., Kosterink J.G., van Altena R., Aarnoutse R.E., van der Werf T.S., Uges D.R., and Alffenaar J.W. 2011. Limited-sampling strategies for therapeutic drug monitoring of moxifloxacin in patients with tuberculosis. Ther Drug Monit. 33(3):350-4.

27. Stass H. and Kubitza D. 1999. Pharmacokinetics and elimination of moxifloxacin after oral and intravenous administration in man. J Antimicrob Chemother. 43 Suppl B:83-90.

28. Ray J., Gardiner I., and Marriott D. 2003. Managing antituberculosis drug therapy by therapeutic drug monitoring of rifampicin and isoniazid. Intern Med J. 33(5-6):229-34. 29. Mcllleron H., Wash P., Burger A., Norman J., Folb P.I., and Smith P. 2006. Determinants of

rifampin, isoniazid, pyrazinamide, and ethambutol pharmacokinetics in a cohort of tuberculosis patients. Antimicrob Agents Chemother. 50(4):1170-7.

30. Verbeeck R.K, Günther G., Kibuule D., Hunter C., and Rennie T.W. 2016. Optimizing treatment outcome of first-line anti-tuberculosis drugs: the role of therapeutic drug monitoring. Eur J Clin Pharmacol. 72(8):905-16.

31. Cello J.P. and Day L.W. 2009. Idiopathic AIDS enteropathy and treatment of gasterointestinal oppertunistic pathogens. Gastroenterology. 136(6):1952-65.

32. Naidoo A., Chirehwa M., Mcllleron H., Naidoo K., Essack S., Yende-Zuma N., Kimba-Phongi E., Adamson J., Govender K., Padayatchi N., and Denti P. 2017. Effect of rifampicin and efavirenz on moxifloxacin concentrations when co-administered in patients with drug-susceptible TB. J Antimicrob Chemother. 72(5):1441-1449.

(17)

Supplemental table 1: Tuberculosis treatment (n = 39). Parameter Value Tuberculosis Culture-positive 38 (97) Localization Pulmonary 28 (72) Extrapulmonary 11 (28) Resistance pattern Drug-sensitive 11 (28)

Intermediate INH resistant 1 (3)

INH resistant 2 (5) MDR 23 (59) XDR 1 (3) Co-treatment for TB1 First-line agents2 Isoniazid 13 (33) Rifampicin 15 (39) Pyrazinamide 17 (44) Ethambutol 24 (62) Rifabutin 1 (3)

Data are presented as n (%). 1TB regimen based on drug-tolerance and

drug susceptibility tests. 2WHO classification.

Supplemental table 1: Tuberculosis treatment (n = 39) (continued).

Co-treatment for TB1

Second-line agents2

Group A. Fluoroquinolones

Levofloxacin 1 (3)

Group B. Second-line injectable agents

Amikacin 19 (49)

Kanamycin 5 (13)

Capreomycin 1 (3)

Group C. Other core second-line agents

Prothionamide 8 (21)

Cycloserine 2 (5)

Linezolid 25 (64)

Clofazimine 17 (44)

Group D. Add-on agents

p-Aminosalicylic acid 1 (3) Thioacetazone 1 (3) Other drugs Clarithromycin 3 (8) Doxycycline 1 (3) Co-trimoxazole 7 (18) Azithromycin 2 (5)

Data are presented as n (%). 1TB regimen based on drug-tolerance and

(18)

Chapter

4

b

Supplemental table 1: Tuberculosis treatment (n = 39).

Parameter Value Tuberculosis Culture-positive 38 (97) Localization Pulmonary 28 (72) Extrapulmonary 11 (28) Resistance pattern Drug-sensitive 11 (28)

Intermediate INH resistant 1 (3)

INH resistant 2 (5) MDR 23 (59) XDR 1 (3) Co-treatment for TB1 First-line agents2 Isoniazid 13 (33) Rifampicin 15 (39) Pyrazinamide 17 (44) Ethambutol 24 (62) Rifabutin 1 (3)

Data are presented as n (%). 1TB regimen based on drug-tolerance and

drug susceptibility tests. 2WHO classification.

Supplemental table 1: Tuberculosis treatment (n = 39) (continued).

Co-treatment for TB1

Second-line agents2

Group A. Fluoroquinolones

Levofloxacin 1 (3)

Group B. Second-line injectable agents

Amikacin 19 (49)

Kanamycin 5 (13)

Capreomycin 1 (3)

Group C. Other core second-line agents

Prothionamide 8 (21)

Cycloserine 2 (5)

Linezolid 25 (64)

Clofazimine 17 (44)

Group D. Add-on agents

p-Aminosalicylic acid 1 (3) Thioacetazone 1 (3) Other drugs Clarithromycin 3 (8) Doxycycline 1 (3) Co-trimoxazole 7 (18) Azithromycin 2 (5)

Data are presented as n (%). 1TB regimen based on drug-tolerance and

(19)

4C

Chapter

Rifampicin and moxifloxacin

for tuberculous meningitis

O.W. Akkerman, A.D. Pranger, R. van Altena, T.S. van der Werf, and J.W.C. Alffenaar

Referenties

GERELATEERDE DOCUMENTEN

For both studies several factors that could have influenced the pharmacokinetics of vorico- nazole and hence the voriconazole trough concentration were not included in the

Publication of this thesis was financially supported by the Graduate School of Medical Sciences and the University Medical Center Groningen.

tuberculosis isolate settled in an individual patient might be as important for treatment success, especially, when factors like the pathophysiology of other diseases or

Concentrations in plasma, urinary excretion and bactericidal activity of levofloxacin (500 mg) versus ciprofloxacin (500 mg) in healthy volunteers receiving a single oral

In addition, despite limited data on GFX PK, in one of the Phase III trials (OFLOTUB), the 4-month GFX containing regimen was not associated with treatment success for the

(prepared with precipitation reagent without internal control), five samples of controlled human plasma (prepared with precipitation reagent with internal control cyanoimipramine),

Based on the pharmacokinetic curves of both patients with and without concomitant use of MFX and RIF, two separate one-compartmental pharmacokinetic population models with

Their findings that a treatment regimen containing a higher dose of rifampicin and standard- dose or high-dose moxifloxacin during the first 2 weeks is safe in patients with