Back to Journals » Infection and Drug Resistance » Volume 17

The Effect of Rifapentine and Rifampicin on Serum Voriconazole Levels Persist for 5 Days and 7 Days or More After Discontinuation in Tuberculosis Patients with Chronic Pulmonary Aspergillosis

Authors Lu H , Mao Y, Zeng Y, Li P, Yan P , Shi Q, Liu L

Received 29 January 2024

Accepted for publication 24 June 2024

Published 8 July 2024 Volume 2024:17 Pages 2853—2862

DOI https://doi.org/10.2147/IDR.S461785

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Professor Suresh Antony



Hong Lu,* Yanmei Mao,* Ying Zeng, Pengyu Li, Pan Yan, Qunzhi Shi, Lin Liu

Department of Pharmacy, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, People’s Republic of China

*These authors have contributed equally to this work

Correspondence: Hong Lu, Department of Pharmacy, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, People’s Republic of China, Email [email protected]

Purpose: Voriconazole, a first-line therapeutic agent for chronic pulmonary aspergillosis, is metabolized by the cytochrome 450 enzymes, specifically CYP2C19 and CYP3A4. Rifampicin and rifapentine act as inducers of the cytochrome P450 enzyme. The current study explored the potential drug interactions arising from the co-administration of voriconazole with either rifampicin or rifapentine, as well as the duration of this effect on serum voriconazole levels after discontinuation of rifampicin or rifapentine.
Patients and Methods: A retrospective study was conducted in tuberculosis patients with chronic pulmonary aspergillosis. These patients underwent a combination therapy involving voriconazole and rifampicin or rifapentine, or they were treated with voriconazole after discontinuation of rifampicin or rifapentine. The serum concentrations of voriconazole at steady-state were monitored. Data on demographic characteristics and the serum voriconazole levels were used for statistical analyses.
Results: A total of 124 serum voriconazole concentrations from 109 patients were included in the study. The average serum concentration of voriconazole fell below the effective therapeutic range in patients treated with both voriconazole and rifampicin or rifapentine. Notably the co-administration of rifapentine led to a substantial (> 70%) decrease in serum voriconazole levels in two patients. Moreover, this interfering effect persisted for at least 7 days following rifampicin discontinuation, while it endured for 5 days or more after discontinuation of rifapentine.
Conclusion: Concomitant use of voriconazole and rifampicin or rifapentine should be avoided, and it is not recommended to initiate voriconazole therapy within 5 or 7 days after discontinuation of rifapentine or rifampicin. Therapeutic drug monitoring not only provides a basis for the adjustment of clinical dose, but also serves as a valuable tool for identifying drug interactions.

Keywords: voriconazole, rifampicin, rifapentine, serum concentration, discontinuation

Introduction

Tuberculosis (TB) poses a serious threat to global health, ranking as the leading cause of death from a single infectious agent since in 2014. According to the global TB report 2023, 7.5 million people worldwide were newly diagnosed with TB in 2022, marking the highest figure since WHO began global TB monitoring in 1995.1 China continues to grapple with a high burden of TB. Even worse, TB is a significant risk factor for fungal infection, as cavitary lesions post TB infection can provide good reservoirs for fungal colonization.2 Chronic pulmonary aspergillosis (CPA), a destructive pulmonary disease caused by Aspergillus species, was estimated to affect approximately 1.2 million people in the world as a sequel to TB.3 Persistent respiratory symptoms associated with CPA are observed in about 20% of patients after two months of intensive anti-TB treatment.4 Voriconazole (VOR) is recommended as the first-line long-term treatment for Aspergillus, and its serum concentration at a steady state is monitored to evaluate the efficacy and safety of the treatment.5,6 But it has been found that a few Aspergillus isolates were resistant to azoles, which correlates with poor therapeutic outcome of zole. The studies indicated that nonsynonymous mutations in the cyp51A and overexpression of mdr1 and mfs genes resulted in azole-resistant phenotypes of Aspergillus.7,8 VOR is metabolized in the liver via the cytochrome P-450 (CYP450) enzyme family, which is mainly metabolized by CYP2C19 and CYP3A4, with CYP2C9 involvement being minimal. As a result, inducers or inhibitors of CYP2C19 and CYP3A4 and genotype status of CYP2C19 may give rise to variations in the serum concentration of voriconazole.9–11 A number of studies have demonstrated a relationship between VOR plasma concentration and clinical efficacy and toxicity, and the rate of treatment success was associated with VOR trough concentration of >0.5 mg/L.12–14 Therefore therapeutic drug monitoring (TDM) of VOR is important to improve the treatment response and reduce adverse events.15,16

Rifampicin (RFP) is a first-line drug for the treatment of drug-susceptible TB. RFP is a potent inducer of both the hepatic and intestinal CYP-450 enzyme system and P-glycoprotein (P-gp) transport system.17 Rifapentine (RFT) is a semi-synthetic rifamycin derivative from the piperazinyl hydrazone class with a microbiologic profile similar to that of RFP, which induces CYP-450 enzyme less than RFP, but RFT has a longer-duration action than RFP.18 In our hospital, treatment with RFT is substituted when patients have adverse drug reactions suspected to be caused by RFP. The medication package insert for VOR (Zyvox™; Pfizer Pharmaceuticals, New York, NY, USA) states that administration of VOR with RFP results in 93% and 96% reduction in the peak serum concentration (Cmax) and area under the drug concentration-time curve (AUC) for VOR, respectively, hence VOR combination with RFP is forbidden. Moreover concomitant administration of VOR with rifabutin results in 69% and 78% reduction in the Cmax and AUC of VOR, respectively. But the effect of RFT on serum VOR level is not mentioned in the package insert for VOR. Research concerning the drug interaction between VOR and RFT is rare. When the serum concentration of VOR can reach the therapeutic range after discontinuation of RFP or RFT was unclear.

The objective of the present study was to investigate the effect of combination of RFP or RFT on VOR serum concentration, and the duration of effect on VOR serum trough concentration after discontinuation of RFP or RFT in TB patients with CPA.

Patients and Methods

Patients

This single retrospective study included TB patients with CPA who were admitted at the TB Diagnosis and Treatment Center of Affiliated Changsha Central Hospital, the University of South China and who underwent therapeutic drug monitoring (TDM) of VOR trough concentration (Cmin) from April 2016 to July 2023. The study was approved by the Ethics Committee of the Affiliated Changsha Central Hospital (2020103).

The inclusion criteria were patients: (1) aged ≥18 years; (2) receiving continuous VOR at a maintenance dose of 200 mg per body every 12 h (Q12h) for treatment >5 days; (3) for whom TDM results of VOR at steady state were available; (4) who were treated by co-administration of VOR with RFP or RFT; (5) who were treated with VOR after discontinuation of RFP or RFT.

The exclusion criteria were patients: (1) who were pregnant or lactating; (2) who were administered other drugs that strongly reduced VOR serum concentration (eg carbamazepine, phenytoin, phenobarbital, rifabutin, efavirenz, ritonavir, St. John’s wort) simultaneously; (3) undergoing blood purification or other forms of kidney-replacement therapy. The flow chart of the study procedure is shown in Figure 1.

Figure 1 The flow chart of study procedure.

Determination of VOR Serum Concentration

When VOR had reached a steady-state concentration (after at least 4 days), blood was collected 30 min before the next administration to monitor the Cmin. VOR serum concentrations were analyzed by high-performance liquid chromatography using a photodiode array (HPLC-PDA, Shimadzu LC-20AT). The analytical method met the requirements for determination of biological samples, with absolute recovery >90% and a linear range of 0.12–20.64 mg/L (R2 = 0.9997). The limit of quantification was 0.12 mg/L, and values that were below this level were recorded as 0. The intra-day precision and inter-day precision were <5%. The Cmin of VOR in the range 1.0–5.5 mg/L was considered to be in the therapeutic range.19

Data Collection

The electronic medical record system (EMRS) of ACCH was used to retrieve and collect patient information. The data collected were organized as: (1) demographics (age, sex); (2) detection time and results of VOR Cmin; (3) laboratory data for liver function (albumin, total bilirubin, direct bilirubin, alanine aminotransferase, aspartate aminotransferase) and renal function (creatinine, urea nitrogen); and (4) the presence or absence of co-medication with proton pump inhibitor (PPI).

Statistical Analysis

Continuous variables are presented as the mean ± SD according to the normality (Kolmogorov–Smirnov) test. To estimate differences between variables, the chi-square test and Student´s t-test were used for categorical variables and continuous parametric variables, respectively. Statistical analyses were performed using GraphPad Prism 8 (GraphPad Software, Inc., San Diego, USA). The P-value <0.05 was statistically significant.

Results

Demographic Characteristics of the Patients

A total of 109 TB patients with CPA for whom VOR serum concentrations were monitored, amounting to 124 measured concentrations, were included in the study: 22 patients were in VOR + RFP group, and 32 patients were in VOR + RFT group; 32 patients were in after discontinuation of RFP group, while 23 patients were in after discontinuation of RFT group. The serum concentration of VOR was monitored twice in 11 patients, and VOR Cmin was monitored three times in 2 cases. A total of 100 patients were treated with VOR injection, 8 patients used VOR tablets, and 1 case used tablets for three days then changed to injection. A total of 50 patients were treated with RFP 450 mg every day (qd), 3 patients with 600 mg qd, and 1 case with 300 mg qd. Forty-eight patients were administered with RFT 450 mg twice a week (biw), 3 cases with 600 mg biw, 3 cases with 300 mg biw, and 1 case with 750 mg biw. RFP or RFT was taken in the morning on an empty stomach. All patients took bland diet in hospitalization. The characteristics of patients are displayed in Table 1. Except for patient age between after discontinuation of RFP/RFT groups, there were no significant differences in the basic characteristics of patients.

Table 1 Characteristics of Patients in the Two Groups

Serum Concentrations of VOR in Co-Administration with RFP or RFT

The serum concentrations of VOR were lower than the therapeutic range in all 22 patients who received VOR + RFP, with 68.18% (15/22) exhibiting serum VOR concentrations below the quantification limit of 0.12 mg/L. In contrast, serum VOR levels within the reference range of 1.00 to 5.50 mg/L were observed in 6 patients who received VOR + RFT, although 21.88% (7/32) of VOR Cmin was under the limit of quantification of 0.12 mg/L. As shown in Figure 2, the mean value of serum VOR concentration was significantly lower in patients who received VOR with RFP (0.14 ± 0.26 mg/L) than in those who received VOR with RFT (0.63 ± 0.97 mg/L).

Figure 2 VOR Cmin in patients who had co-administration with RFP or RFT.

The drug interaction between RFT and VOR is not mentioned in the medication package insert for VOR (Pfizer Pharmaceuticals, New York, NY, USA). The serum VOR concentrations of two patients who were treated with VOR injection for 7 days and 5 days were 1.38 mg/L and 2.52 mg/L, respectively, while they decreased to 0.25 mg/L and 0.72 mg/L after co-administration with RFT for 10 days and 7 days, respectively, which suggested that the serum concentration of VOR reduced by 71.4%–81.9% when VOR was combined with RFT.

Recovery of the Serum VOR Concentration After Discontinuation of RFP

A total of 32 patients, contributing to 41 VOR TDM records, were included to investigate the recovery of serum VOR concentration after discontinuation of RFP. The results were categorized into three different time periods following RFP discontinuation: 1–3 days (n=12), 4–6 days (n=20), and more than 7 days (n=9). The mean Cmin of VOR were 0.36 ± 0.34 mg/L and 0.49 ± 0.56 mg/L during the periods of 1–3 days and 4–6 days after RFP discontinuation. As shown in Figure 3, the average Cmin of VOR measured ≥7 days after discontinuation of RFP was 1.34 ± 0.72 mg/L, significantly higher than those measured during 1–3 days and 4–6 days after RFP discontinuation (P<0.05). The distribution of serum VOR concentration after RFP discontinuation is displayed in Table 2.

Table 2 Distribution of VOR Cmin After RFP Discontinuation

Figure 3 VOR Cmin in patients after RFP discontinuation.

Repeat measurements were carried out for 8 patients in whom serum VOR concentrations were lower than 1.0 mg/L after RFP discontinuation within 5 days. Apart from the eighth patient in whom the serum VOR levels were 0.72 mg/L and 0.91 mg/L on day 7 and day 10 after discontinuation of RFP, the VOR concentrations in the remaining 7 patients clearly increased to the target range after discontinuation of RFP for more than 6 days. Detailed results are displayed in Table 3.

Table 3 Several VOR TDM After Discontinuation of RFP in 8 Patients

Recovery of the Serum VOR Concentration After Discontinuation of RFT

A total of 23 patients, yielding 29 VOR TDM records, were included to assess the recovery of serum VOR concentration following the discontinuation of RFT. The time intervals were defined as 1–3 days (n=9), 4–5 days (n=12), and more than 6 days (n=8) after RFT discontinuation. The average Cmin of VOR were 0.45 ± 0.41 mg/L and 0.49 ± 0.33 mg/L during 1–3 days and 4–5 days after RFT discontinuation. As shown in Figure 4, the mean Cmin of VOR measured ≥6 days after RFT discontinuation was 2.31 ± 0.91 mg/L, significantly higher than those measured during 1–5 days after discontinuation of RFT (P<0.0001). The distribution of VOR Cmin after RFT discontinuation is displayed in Table 4.

Table 4 Distribution of VOR Cmin After RFT Discontinuation

Figure 4 VOR Cmin in patients after RFT discontinuation.

The serum concentrations of VOR were repeatedly measured in 5 patients after RFT discontinuation. As shown in Table 5, the VOR concentrations remained below 1.0 mg/L in 4 patients within the initial 4 days after RFT discontinuation, but exhibited a notable increase beyond 6 days. The serum VOR concentrations in all patients reached the target range from the sixth day after RFT discontinuation.

Table 5 Several VOR TDM After Discontinuation of RFT in 5 Patients

Discussion

Previous experiments have revealed that VOR undergoes transformation into N-oxide and is metabolized by liver CYP2C19, CYP3A4, and CYP2C9 enzymes,10 and thus drugs that inhibit or induce these activities probably affect the serum concentration of VOR. RFP is an inducer of CYP-450 oxidative enzymes, including CYP3A4, CYP2C19, CYP2C9, CYP1A2, and CYP2D6.17,20 Consequently, the combination of VOR and RFP leads to a loss of the therapeutic efficacy of VOR due to the underexposure. The Cmax of VOR and AUC in one patient was 3.92 mg/L and 27.4 h mg/L after the starting dose (400 mg), but the VOR exposure was decreased by 99% after 36 days of VOR therapy and 30 days of RFP therapy, with a Cmax of 0.038 mg/L and an AUC of 0.145 h mg/L. Meanwhile the plasma concentrations of three main metabolites were similar or even increased compared to the concentrations following the first dose without RFP.21 Co-administration of RFP was associated with a significant reduction in VOR exposure.13,22 Research has indicated that the total apparent clearances increased twofold in CYP2C9 activity, irrespective of the CYP2C9 genotypes in healthy volunteers after RFP administration.23 The average serum concentrations of VOR in all 22 patients co-administered with RFP was only 0.14 mg/L in the present study, which is consistent with the effect of RFP on serum itraconazole levels.24

RFT induces CYP3A4 and CYP2C8/9 as shown in human hepatocyte study,25 but the relative enzyme induction of RFT is less potent than that of RFP.20 Studies about the drug interaction with RFT are rare,26 and RFT was not included in clinically significant drug interactions with VOR.27,28 Therefore RFT is commonly substituted for RFP in order to minimize the drug interaction between VOR with RFP in the hospital. A serum concentration of VOR in the target range of 1.0–5.5 mg/L was only achieved in 18.75% of measurements in patients who were treated combination with VOR and RFT in the study. The average serum VOR concentration in the VOR + RFT group was 0.66 mg/L, lower than the therapeutic range, but was significantly higher than that in the VOR + RFP group. The serum concentrations of VOR in two patients reduced by more than 70% after administration with RFT. Previous study reported that the steady-state serum Cmax and AUC of indinavir were decreased by 55% and 70%, respectively,18 which was consistent with the present study.

Enzyme induction activity and the pharmacodynamic effects of the affected drug gradually return to baseline levels within 9–14 days after discontinuation of RFP.29,30 Studies about duration of effect of RFP on serum VOR concentration after discontinuation of RFP therapy were scarce. One case reported that the inductive effect of RFP on plasma VOR concentration was notable for at least 13 days in a 32-year-old patient with invasive central nervous system aspergillus.31 In our study a total of 9 Cmin measurements of VOR were in the therapeutic range of 1.0–5.5 mg/L after RFP discontinuation. Only 9.38% (3/32) patients' serum VOR level reached the target range after discontinuation of RFP for 6 days, but the percentage of VOR Cmin >1.0 mg/L was 66.67% (6/9) after RFP discontinuation for 7 days or more. Meanwhile the serum VOR levels in 8 repeated monitoring patients were gradually increased. RFT is approved for intermittent dosing in the treatment of TB, with a serum half-life several times higher than RFP.32 However the duration of induced enzyme activity after discontinuation of RFT is unclear. Enzyme activity of RFT returns to baseline within 2 weeks after the last dose of RFT.33 The serum concentrations of VOR failed to reach the effective treatment range in an 85-year-old man within 10 days of RFT discontinuation, which suggested the induction of hepatic enzymes may exceed 10 days after stopping RFT.34 There were 9 serum concentrations of VOR that reached the target range after RFT discontinuation in the present study, but all serum VOR concentrations measured ≥6 days after discontinuation of RFT were in the therapeutic range. As the elimination half-life of RFT is 14.8 h to 18.5 h,26 the effect of RFT on the serum concentration of VOR receded markedly after discontinuation for 5 days. Except for the enzyme-induced activity, the difference of effect duration on serum VOR concentrations after discontinuation of RFP or RFT may be related to the age of patients. In the present study, the median age of the patients in the after RFT discontinuation group was 69 (range 34–93), and 69.56% were >60 years of age. In addition, the age of patients in the after discontinuation of RFT group was markedly older than the age of patients in the after RFP discontinuation group. The previous studies had demonstrated the relationship between patient age with serum level of VOR, and the VOR concentrations in patients older than 60 years were higher than in younger patients.35,36 In older patients, the hepatic drug clearance is decreased, and the apparent volume of distribution of lipophilic drugs (such as VOR) is increased with a prolonged half-life.37 Although VOR is regarded as the first-line drug for the treatment of Aspergillus, its use is limited in TB patients by a wide range of drug interactions, especially rifamycin. Therefore, consideration should be given to starting with liposomal amphotericin B (L-AmB) or echinocandins with close monitoring of liver and kidney function to avoid this significant drug interaction. Once the patient is ready for discharge, oral VOR or isavuconazole may be used as maintenance therapy.38–41

There are a few limitations in this study. The serum concentration of VOR is lower than the effective range when VOR is combined with RFT, and the question remains whether doubling the dose of VOR to 400 mg Q12h would make the steady-state plasma VOR concentration reach the target range. As CYP2C19 gene variants had been well established to influence the VOR pharmacokinetics, we should assess the gene polymorphisms in the future research. This is a single-center study, and we will utilize a larger study cohort to confirm the finding.

Conclusion

The present study demonstrated that the serum concentrations of VOR were lower than the effective treatment range when combined with RFP or RFT. The average Cmin of VOR in VOR + RFT patients was significantly higher than the mean value of VOR in VOR + RFP group. The serum VOR levels were decreased by more than 70% when combined with RFT. Concomitant use of RFT and VOR should be avoided. The duration of inductive effect on serum VOR concentration was at least 7 days after RFP discontinuation, while it lasted for 5 days or longer after discontinuation of RFT. VOR TDM is an important tool for identifying the drug interactions of clinical significance and to improve the treatment response in TB patients with CPA.

Ethics Approval and Informed Consent

This study was approved by the ethics committee of the Affiliated Changsha Central Hospital, the University of South China (approval number 2020103). This study complied with the Declaration of Helsinki. Waiving of informed consent was given due to the retrospective, observational study. All patient data were collected anonymously and ensured the confidentiality of their information.

Author Contributions

All authors made substantial contributions to conception and design, acquisition of data, analysis of data; took part in drafting the article or revising it critically for important content; agreed to submit to the current journal; gave final approval of the version to be published; and agree to be accountable for all aspects of the work.

Funding

This work was supported by Hunan Medical Association of China with a fund number [MHA202001007].

Disclosure

The authors report no conflicts of interest in this work.

References

1. World Health Organization. Global Tuberculosis Report 2023. 2023; Available from: https://iris.who.int/bitstream/handle/10665/373828/9789240083851-eng.pdf?sequence=1. Accessed June 26 2024.

2. Jhun B, Jeon K, Eom JS, et al. Clinical characteristics and treatment outcomes of chronic pulmonary aspergillosis. Med Mycol. 2013;51(8):811–817. doi:10.3109/13693786.2013.806826

3. Denning D, Pleuvry A, Cole D. Global burden of chronic pulmonary aspergillosis as a sequel to pulmonary tuberculosis. Bull World Health Organ. 2011;89(12):864–872. doi:10.2471/BLT.11.089441

4. Namusobya M, Bongomin F, Mukisa J, et al. Chronic pulmonary aspergillosis in patients with active pulmonary tuberculosis with persisting symptoms in Uganda. Mycoses. 2022;65(6):625–634. doi:10.1111/myc.13444

5. Denning D, Cadranel J, Beigelman-Aubry C, et al. Chronic pulmonary aspergillosis: rationale and clinical guidelines for diagnosis and management. Eur Respir J. 2016;47(1):45–68. doi:10.1183/13993003.00583-2015

6. Patterson T, Thompson G, Denning D, et al. Practice guidelines for the diagnosis and management of aspergillosis: 2016 update by the infectious disease society of America. Clin Infec Dis. 2016;63(4):433–442. doi:10.1093/cid/ciw326

7. Chowdhary A, Sharma C, Meis JF. Azole-resistant aspergillosis: epidemiology, molecular mechanisms, and treatment. J Infect Dis. 2017;216(suppl_3):S436–S444. doi:10.1093/infdis/jix210

8. Sen P, Vijay M, Kamboj H, et al. cyp51A mutations, protein modeling, and efflux pump gene expression reveals multifactorial complexity towards understanding aspergillus section Nigri azole resistance mechanism. Sci Rep. 2024;14(1):6156. doi:10.1038/s41598-024-55237-9

9. Johnson L, Kauffman C. Voriconazole: a new triazole antifungal agent. Clin Infec Dis. 2003;36(5):630–637. doi:10.1086/367933

10. Hyland R, Jones BC, Smith DA. Identification of the cytochrome P450 enzymes involved in the N-oxidation of voriconazole. Drug Metab Dispos. 2003;31(5):540–547. doi:10.1124/dmd.31.5.540

11. Scholz I, Oberwittler H, Riedel KD, et al. Pharmacokinetics, metabolism and bioavailability of the triazole antifungal agent voriconazole in relation to CYP2C19 genotype. Br J Clin Pharmacol. 2009;68(6):906–915. doi:10.1111/j.1365-2125.2009.03534.x

12. Troke PF, Hockey HP, Hope WW. Observational study of the clinical efficacy of voriconazole and its relationship to plasma concentrations in patients. Antimicrob Agents Chemother. 2011;55(10):4782–4788. doi:10.1128/AAC.01083-10

13. Dolton MJ, Ray JE, Chen SC, et al. Multicenter study of voriconazole pharmacokinetics and therapeutic drug monitoring. Antimicrob Agents Chemother. 2012;56(9):4793–4799. doi:10.1128/AAC.00626-12

14. Jin H, Wang T, Falcione BA, et al. Trough concentration of voriconazole and its relationship with efficacy and safety: a systematic review and meta-analysis. J Antimicrob Chemother. 2016;71(7):1772–1785. doi:10.1093/jac/dkw045

15. Matsumoto K, Ikawa K, Abematsu K, et al. Correlation between voriconazole trough plasma concentration and hepatotoxicity in patients with different CYP2C19 genotypes. Int J Antimicrob Agents. 2009;34(1):91–94. doi:10.1016/j.ijantimicag.2009.01.008

16. Park WB, Kim NH, Kim KH, et al. The effect of therapeutic drug monitoring on safety and efficacy of voriconazole in invasive fungal infections: a randomized controlled trial. Clin Infect Dis. 2012;55(8):1080–1087.

17. Baciewicaz A, Chrisman C, Finch C, et al. Update of rifampin, rifabutin, and rifapentine drug interactions. Curr Med Res Opin. 2013;29(1):1–12. doi:10.1185/03007995.2012.747952

18. Temple ME, Nahata NC. Rifapentine: its role in the treatment of Tuberculosis. Ann Pharmacother. 1999;33(11):1203–1210. doi:10.1345/aph.18450

19. Ashbee HR, Barnes RA, Johnson EM, et al. Therapeutic drug monitoring (TDM) of antifungal agents: guidelines from the British society for medical mycology. J Antimicrob Chemother. 2014;69(5):1162–1176. doi:10.1093/jac/dkt508

20. Burman WJ, Gallicano K, Peloquin C. Comparative pharmacokinetics and pharmacodynamics of the rifamycin antibacterials. Clin Pharmacokinet. 2001;40(5):327–341. doi:10.2165/00003088-200140050-00002

21. Geist MJP, Egerer G, Burhenne J, et al. Induction of voriconazole metabolism by rifampin in a patient with acute myeloid leukemia: importance of interdisciplinary communication to prevent treatment errors with complex medications. Antimicrob Agents Chemother. 2007;51(9):3455–3456. doi:10.1128/AAC.00579-07

22. Cojutti P, Candoni A, Forghieri F, et al. Variability of voriconazole trough levels in haematological patients: influence of comedications with cytochrome P450 (CYP) inhibitors and/or with CYP inhibitors plus CYP inducers. Basic Clin Pharmacol Toxicol. 2016;118(6):474–479. doi:10.1111/bcpt.12530

23. Vormfelde SV, Brockmöller J, Bauer S, et al. Relative impact of genotype and enzyme induction on the metabolic capacity of CYP2C9 in healthy volunteers. Clin Pharmacol Ther. 2009;86(1):54–61. doi:10.1038/clpt.2009.4

24. Moon SM, Park HY, Jeong BH, et al. Effect of rifampin and rifabutin on serum itraconazole levels in patients with chronic pulmonary aspergillosis and coexisting nontuberculous mycobacterial infection. Antimicrob Agents Chemother. 2015;59(1):663–665. doi:10.1128/AAC.04075-14

25. Li AP, Reith MK, Rasmussen A, et al. Primary human hepatocytes as a tool for the evaluation of structure-activity relationship in cytochrome P450 induction potential of xenobiotics: evaluation of rifampin, rifapentine and rifabutin. Chem Biol Interact. 1997;107(1–2):17–30. doi:10.1016/s0009-2797(97)00071-9

26. Dooley K, Flexner C, Hackman J, et al. Repeated administration of high-dose intermittent rifapentine reduces rifapentine and moxifloxacin plasma concentrations. Antimicrob Agents Chemother. 2008;52(11):4037–4042. doi:10.1128/AAC.00554-08

27. Theuretzbacher U, Ihle F, Derendorf H. Pharmacokinetic/pharmacodynamic profile of voriconazole. Clin Pharmacokinet. 2006;45(7):649–663. doi:10.2165/00003088-200645070-00002

28. Li TY, Liu W, Chen K, et al. The influence of combination use of CYP450 inducers on the pharmacokinetics of voriconazole: a system review. J Clin Pharm Ther. 2017;42(2):135–146. doi:10.1111/jcpt.12493

29. Gallicano KD, Sahai J, Shukla VK, et al. Induction of zidovudine glucuronidation and amination pathways by rifampicin in HIV-infected patients. Br J Clin Pharmacol. 1999;48(2):168–179. doi:10.1046/j.1365-2125.1999.00987.x

30. Nolan SR, Self TH, Norwood JM. Interaction between rifampin and levothyroxine. South Med J. 1999;92(5):529–531. doi:10.1097/00007611-199905000-00018

31. Farrokh S, Avdic E. Voriconazole autoinduction and saturable metabolism after cessation of rifampin in a patient with invasive central nervous system aspergillus: importance of therapeutic drug monitoring. J Pharm Pract. 2019;32(5):589–594. doi:10.1177/0897190018760621

32. Chan JGY, Bai XX, Traini D. An update on the use of rifapentine for tuberculosis therapy. Expert Opin Drug Deliv. 2014;11(3):421–431. doi:10.1517/17425247.2014.877886

33. Keung A, Reith K, Eller MG, et al. Enzyme induction observed in healthy volunteers after repeated administration of rifapentine and its lack of effect on steady-state rifapentine pharmacokinetics: part I. Int J Tuber Lung Dis. 1999;3(5):426–436.

34. Ling GM, Li GM, Jing YP, et al. Decreased serum voriconazole levels caused by hepatic enzyme induction after rifapentine discontinuation: a case report and literature review. Eur Rev Med Pharmacol Sci. 2023;27(8):3714–3718. doi:10.26355/eurrev_202304_32170

35. Mitsani D, Nguyen MH, Shields RK, et al. Prospective, observational study of voriconazole therapeutic drug monitoring among lung transplant recipients receiving prophylaxis: factors impacting levels of and associations between serum troughs, efficacy, and toxicity. Antimicrob Agents Chemother. 2012;56(5):2371–2377. doi:10.1128/AAC.05219-11

36. Wang T, Chen S, Sun J, et al. Identification of factors influencing the pharmacokinetics of voriconazole and the optimization of dosage regimens based on monte carlo simulation in patients with invasive fungal infections. J Antimicrob Chemother. 2014;69(2):463–470. doi:10.1093/jac/dkt369

37. Klotz U. Pharmacokinetics and drug metabolism in the elderly. Drug Metab Rev. 2009;41(2):67–76. doi:10.1080/03602530902722679

38. Li JM, Cai XJ, Xu YY, et al. Individualized pharmaceutical care for antifungal therapy in a patient with aspergillus tubingensis spondylitis after discontinuation of rifampicin: a case report. Infect Drug Resist. 2023;4(16):4349–4356. doi:10.2147/IDR.S417604

39. Wasko JA, Westholder JS, Jacobson PA. Rifampin-sirolimus-voriconazole interaction in a hematopoietic cell transplant recipient. J Oncol Pharm Pract. 2017;23(1):75–79. doi:10.1177/1078155215624263

40. Buonomo AR, Viceconte G, Compare D, et al. Invasive pulmonary aspergillosis and pulmonary tuberculosis in a patient treated with infliximab for Crohn’s disease. IDCases. 2019;17:e00537. doi:10.1016/j.idcr.2019.e00537

41. Ahmed AO, Ali GA, Goravey W. Concomitant pulmonary tuberculosis and invasive aspergillosis infection in an immunocompetent host. Eur J Case Rep Intern Med. 2022;9(3):003249. doi:10.12890/2022_003249

Creative Commons License © 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.