Abstract
There are a limited number of studies that guide dosing of posaconazole delayed-release (DR) tablets for the pediatric population. Current FDA-approved doses are only recommended for patients 13 years and older. For younger patients, providers are faced with the challenge of recommending posaconazole doses extrapolated from adult studies or choosing an alternative agent. We report on a case of a 10-year-old patient who experienced a supratherapeutic trough serum concentration and transaminitis after receiving the extrapolated adult dosage of posaconazole DR tablets (300 mg twice daily for the first day, followed by 300 mg daily) for 7 days. In the end, the patient required a smaller dose of 200 mg daily to achieve the desired trough target concentration for the treatment of a Rhizopus neck infection. Our findings highlight the need for additional studies to determine the optimal dosing of posaconazole DR tablets for children.
Keywords: invasive fungal infections, posaconazole, Rhizopus, supratherapeutic
Introduction
Posaconazole is a broad-spectrum antifungal agent used to treat invasive fungal infections. In comparison to the oral suspension formulation, the delayed-release (DR) oral tablet is preferred because of improved bio-availability, fewer food interactions, and decreased interindividual variability associated with this tablet.1,2 However, a limitation of the DR tablet is the lack of pediatric data associated with its use. The dosing regimen of 300 mg twice daily for the first day, followed by 300 mg daily is FDA approved for adolescents ≥13 years of age, on the basis of extrapolated data from adults receiving the oral tablets and children receiving the oral suspension.3 For patients younger than 13 years of age, the most appropriate dosing strategy remains unclear. We describe a case using posaconazole DR tablets in a 10-year-old patient, which resulted in a supratherapeutic trough serum concentration and transaminitis after 7 days of treatment.
Case Report
A 10-year-old female patient (31 kg) with a history of small bowel transplant for short gut syndrome and diabetes was admitted with a fever (38.7°C) and severe erythema and swelling around a neck lesion. A biopsy was performed, with histopathologic findings of hyphal elements. Because of concern for a possible invasive fungal infection, combination therapy with liposomal amphotericin B and voriconazole was promptly initiated.
On day 11 of therapy, tissue cultures from the neck biopsy indicated the presence of Rhizopus species. The decision was made to discontinue voriconazole, to continue liposomal amphotericin B, and to add caspofungin for potential synergistic activity.4 The patient then underwent 2 extensive surgical debridements for source control, which successfully led to repeat negative biopsies. After 5 weeks (day 36) of therapy and multiple wound washouts, the patient was clinically stable and signs of progressive infection were absent; the decision was made to step down to an oral antifungal regimen. Caspofungin was discontinued, and posaconazole DR tablets were initiated at a dosage of 300 mg twice daily for the first day, followed by 300 mg daily. During this time, the patient was also taking 2.5 mg of oral tacrolimus twice daily. The dose of tacrolimus was preemptively reduced to 1.5 mg twice daily because of the CYP3A4 drug-drug interaction with posaconazole. The patient was also taking other medications that did not interact with posaconazole: vitamin D3, vitamin B6, ferrous sulfate, magnesium oxide, montelukast, cetirizine, albuterol, sodium citrate, aspirin, insulin glargine, amlodipine, ranitidine, lactulose, and rifaximin. Lactulose was being used to enhance gut motility, and rifaximin was prescribed to inhibit bacterial overgrowth in the patient's transplanted bowel.
At our institution, we typically target posaconazole trough serum concentrations between 1 and 2.5 mg/L for the treatment of mucormycosis.5 However, because higher minimum inhibitory concentrations for posaconazole have been reported for Rhizopus spp., a target trough of >2 mg/L was chosen.6,7 A week after posaconazole was initiated, a trough concentration was measured using ultraperformance liquid chromatography–tandem mass spectrometry. Because the specimen was processed by an outside laboratory, it took 4 days for the result to return at 5.5 mg/L. The following day, we reduced the posaconazole dose to 200 mg daily and discontinued liposomal amphotericin B. We also requested daily monitoring of liver function tests.
The Table summarizes the timeline of posaconazole administration, drug serum concentrations, and relevant laboratory studies. Following the supratherapeutic trough concentration of 5.5 mg/L, liver function tests increased from baseline: alanine aminotransferase (ALT) 16 to 119 U/L, aspartate aminotransferase (AST) 30 to 72 U/L, and alkaline phosphatase (ALP) 132 to 187 U/L (See Table footnotes for normal reference ranges). We predict that these measurements were even higher on the actual day of the supratherapeutic trough concentration (day 7), although the patient never showed signs of acute liver injury. After a week of close monitoring and continuation of posaconazole at a reduced dose, the repeat trough returned therapeutic at 2.2 mg/L, and the patient's liver function tests improved. We did not identify any other adverse effects from posaconazole, such as QTc prolongation or electrolyte abnormalities (calcium, magnesium, potassium). A few days later, the patient was discharged home and given instructions to continue the same dosage until outpatient clinic follow-up occurred. Two months later, a repeat posaconazole trough remained therapeutic at 2.5 mg/L.
Table.
Clinical and Laboratory Parameters on Posaconazole (Initiated on Day 0)
| Day | Posaconazole Dose | Posaconazole Concentration* | Tacrolimus Dose | Tacrolimus Concentration† | Laboratory Findings‡ | ||||||||
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| ALT | AST | ALP | Total Bilirubin | Albumin | Ca | Mg | K | SCr | |||||
| -1 | 2.5 mg every 12 hr | 6.4 | 16 | 30 | 132 | 0.2 | 2.8 | 9.0 | — | 5.0 | 1.35 | ||
| 0 | 300 mg every 12 hr | — | 1.5 mg every 12 hr | — | — | — | — | — | — | — | — | — | — |
| 1 | 300 mg every 24 hr | — | 1.5 mg every 12 hr | 4.5 | — | — | — | — | — | 9.0 | 2.2 | 4.7 | 1.35 |
| 2 | 300 mg every 24 hr | — | 1 mg every 12 hr | 19 | — | — | — | — | — | 2.1 | 5 | 1.48 | |
| 3 | 300 mg every 24 hr | — | Hold | 13.5 | — | — | — | — | — | 8.5 | 2.1 | 4.7 | 1.51 |
| 4 | 300 mg every 24 hr | — | Hold | 14 | — | — | — | — | — | 8.2 | 1.7 | 3.3 | 1.5 |
| 5 | 300 mg every 24 hr | — | Hold | 18.2 | — | — | — | — | — | 8.5 | 2.3 | 3.1 | 1.99 |
| 6 | 300 mg every 24 hr | — | Hold | 13.8 | — | — | — | — | — | 8.1 | 2.1 | 2.8 | 1.46 |
| 7 | 300 mg every 24 hr | 5.5 (inpatient) | 0.5 mg every 24 hr | 12.1 | — | — | — | — | — | 8.6 | 2.1 | 3.3 | 1.13 |
| 8 | 300 mg every 24 hr | — | 0.5 mg every 24 hr | 7.6 | — | — | — | — | — | 8.4 | 1.9 | 3.3 | 1.1 |
| 9 | 300 mg every 24 hr | — | 0.5 mg every 24 hr | 8 | — | — | — | — | — | 8.7 | 1.7 | 3.9 | 0.99 |
| 10 | 300 mg every 24 hr | — | 0.5 mg every 24 hr | 5.6 | — | — | — | — | — | 0.99 | 1.7 | 3.9 | 0.99 |
| 11 | 300 mg every 24 hr | Notified of supra-therapeutic concentration | 0.5 mg every 24 hr | 5.6 | — | — | — | — | — | 8.7 | 1.7 | 3.8 | 0.93 |
| 12 | 200 mg every 24 hr | — | 0.5 mg every 24 hr | 8.2 | 119 | 72 | 187 | 0.2 | 2.6 | 8.4 | 1.7 | 5.1 | 0.98 |
| 13 | 200 mg every 24 hr | — | 0.5 mg every 24 hr | 8 | 97 | 37 | 220 | 0.3 | 3.1 | 8.7 | 4.4 | 0.83 | |
| 14 | 200 mg every 24 hr | — | 0.5 mg every 24 hr | 8.2 | 111 | 86 | 169 | 0.3 | 2.8 | 8.7 | 1.8 | 4.2 | 0.69 |
| 15 | 200 mg every 24 hr | — | 0.5 mg every 24 hr | 4.7 | 98 | 61 | 158 | 0.2 | 2.5 | 8.3 | 3.7 | 0.69 | |
| 16 | 200 mg every 24 hr | — | 0.5 mg every 12 hr | 5.7 | 80 | 55 | 182 | 0.5 | 2.4 | 8.3 | 1.6 | 3.9 | 0.71 |
| 17 | 200 mg every 24 hr | — | 1 mg every 12 hr | 10.1 | 67 | 30 | 198 | 0.5 | 2.8 | 9.4 | 1.7 | 4.3 | 0.65 |
| 18 | 200 mg every 24 hr | 2.2 (inpatient) | 0.5 mg every 12 hr | 10.7 | 56 | 41 | 76 | 0.5 | 2.7 | 9 | 1.9 | 4.2 | 0.57 |
| 19 | 200 mg every 24 hr | — | 0.5 mg every 12 hr | — | — | — | — | — | — | — | — | — | — |
| 20 | 200 mg every 24 hr | — | 0.5 mg every 12 hr | 10.7 | 68 | 46 | 159 | 0.5 | 2.5 | 8.9 | 1.7 | 5.6 | 0.56 |
| 80 | 200 mg every 24 hr | 2.5 (outpatient) | 0.5 mg every 12 hr | 11.3 | 22 | 39 | 133 | 0.2 | 2 | 8.2 | 1.9 | 4.8 | 0.6 |
ALP, alkaline phosphatase; ALT, alanine aminotransferase; AST, aspartate aminotransferase; Ca, calcium; K, potassium; Mg, magnesium; SCr, serum creatinine; —, laboratory test not performed
*Posaconazole trough serum concentration goal 2–3.75 mg/L.
† Tacrolimus trough serum concentration goal 6–8 ng/mL.
‡Age-related normal values: ALT (1–45 U/L); AST (1–35 U/L); ALP (38–126 U/L); total bilirubin (0.1–1.2 mg/dL); albumin (3.5–4.9 g/dL); Ca (8.5–10.5 mg/dL); Mg (1.5–2.5 mg/dL); K (3.5–5.2 mEq/L); SCr (0.5–1.1 mg/dL).
Discussion
We report our case of a 10-year-old patient with supratherapeutic posaconazole serum concentrations after receiving the extrapolated adult dosage of the DR tablets for 7 days. Despite guideline recommendations, the tablet formulation was chosen over the suspension to maximize drug bioavailability, especially because our patient had a history of small bowel transplant and was able to swallow pills. After achieving adequate source control through 2 surgical debridements, posaconazole was determined to be an appropriate step-down oral agent based on in vitro data8 demonstrating potent activity against Rhizopus species. We chose to prescribe an extrapolated adult dosing regimen of 300 mg twice daily for the first day, followed by 300 mg daily, based on the severity of infection, limited data mentioned below, and lack of information regarding the minimum inhibitory concentration for our particular Rhizopus species.
In 2013, posaconazole DR tablets were approved by the FDA for pediatric patients between the ages of 13 and 17 years. Approval was based on extrapolated data from adults receiving oral tablets and studies in pediatric patients receiving the oral suspension.3 One study3 conducted on 10 pediatric patients receiving the oral suspension for prophylaxis of invasive fungal infections showed similar mean steady-state posaconazole average concentrations among adolescents (13–17 years of age) and adults greater than 18 years of age. Another study by Krishna et al9 compared posaconazole concentrations from the oral suspension between children (ranging from 8 to 17 years of age) and adults; both groups received 800 mg/day, either 200 mg 4 times daily or 400 mg twice daily, and the patients had similar mean serum posaconazole concentrations (0.776 mg/L vs 0.817 mg/L). There was no difference in overall success rate or adverse events, suggesting that adults and children older than 8 years can be given the same dose. However, the study was limited by the disproportionate number of children (n = 12) compared with adults (n = 194) and included only one patient who was the same age as our patient. Furthermore, it is difficult to extrapolate these results to the DR tablets because the posaconazole suspension formulation is not interchangeable due to bioavailability differences.
In 2017, Döring et al10 conducted a study on posaconazole DR tablets for antifungal prophylaxis in patients with hematopoietic stem cell transplantation. A total of 32 patients were included, the ages for which were as follows: ≤6 years (n = 4), 7 to 12 years (n = 9), and 13 to 17 years (n = 19). Patients were administered a non–FDA-approved weight-based dosing regimen of 5 to 7 mg/kg twice a day as the initial loading dose, followed by 5 to 7 mg/kg daily. This translated to 100 mg (for weight of 15–21 kg), 150 mg (for weight of 22–30 kg), 200 mg (for weight of 31–35 kg), 250 mg (for weight of 36–40 kg), and 300 mg (for weight of >40 kg or >13 years of age). At the end of the study, no proven or possible fungal infections and no deaths were reported. When drug serum concentrations were compared, the posaconazole concentrations of patients younger than 6 years were not significantly different from those in the older age groups. Median concentrations were 0.874 mg/L at 7 days of therapy, 0.978 mg/L after 14 ± 1 days of therapy, and 1.541 mg/L after 28 ± 2 days of therapy. Most patients achieved prophylaxis targets of >0.7 mg/L and treatment targets of >1 to 1.25 mg/L for invasive fungal infection.9,11–18 Only 2 of 32 (6.3%) patients experienced drug-related events. One patient reported nausea, and the other patient experienced facial exanthema. Increases in ALT of >1.5 and >2.5 times the upper limits of normal were observed in 23 out of 32 patients (71.9%), and an increase in AST of >1.5 and >2.5 times the upper normal limit was observed in 21 of 32 patients (65.6%). Direct and total bilirubin concentrations were not affected. Our patient that we report on had increases in both ALT (>2.5 times the upper limit of normal [>112.5 U/L]) and AST (>1.5 times the upper limit of normal [AST >52.5 U/L]). Posaconazole has been associated with both transient aminotransferase elevations and acute drug-induced liver injury so it remains unclear whether our patient's transaminitis was due to the latter or would have resolved independently on its own.9,15,19 However, we did observe improvements in transaminitis shortly after the posaconazole dose was reduced to 200 mg daily.
Currently, there are a very limited number of studies20–22 investigating the relationship between posaconazole serum concentration and toxicity. In 2015, Martino et al22 reported a case of a 13-year-old female with suspected posaconazole toxicity after being inadvertently prescribed posaconazole DR tablets 400 mg twice daily. The trough serum concentration was 9.5 mg/L, and the patient suffered from nausea, fatigue, decreased appetite, musculoskeletal pain, hypokalemia, and anemia. Our patient's serum concentration was not as high, nor did she show similar signs of toxicity; however, we speculated that this could have occurred if the dose was continued at 300 mg daily. The study by Döring et al9 revealed that posaconazole concentrations can rise even after steady state is achieved. We chose to reduce the dose to 200 mg daily in the setting of transaminitis and to prevent even higher serum concentrations, especially because we considered 3.75 mg/L to be the upper limit for our trough target.23
We considered various factors that could have contributed to the supratherapeutic trough concentration. It is known that posaconazole absorption can be affected by diet or nutritional status. Absorption can be enhanced with a fatty meal, particularly following administration of the oral suspension formulation.24 However, this was not the case for our patient because she was known to be a selective eater and nutritionally deficient. Another consideration was our patient's hypoalbuminemia, because posaconazole is highly bound to serum albumin (>98%), which would result in more unbound drug freely available in the serum. Posaconazole absorption can also be enhanced with decreased gut motility, increased gastrointestinal surface area, or altered intestinal pH.25,26 Our patient had a history of small bowel transplant, so it is unclear how this could have affected her drug absorption. She was also taking lactulose, which enhances gut motility. In terms of drug interactions, ranitidine was discussed but dismissed after learning that the interaction occurs primarily with the suspension formulation and not the DR tablet formulation. Posaconazole suspension relies on low pH in the stomach to dissolve, whereas the DR tablets are released from their polymer in the small intestine.25
Conclusion
We report on a case of a 10-year-old patient with a supratherapeutic posaconazole serum concentration and transaminitis after receiving the extrapolated adult dosage of posaconazole DR tablets for 7 days. In the end, this patient required a smaller dose of 200 mg daily to achieve the desired trough target concentrations for the treatment of a Rhizopus infection. This case report highlights the need for additional studies to investigate dosing of posaconazole DR tablets in children.
Acknowledgments
We would like to thank the Pediatric Infectious Disease physicians, surgeons, pharmacists, medical teams, and nurses at Mount Sinai Hospital for their contributions in caring for this patient.
ABBREVIATIONS
- ALP
alkaline phosphatase
- ALT
alanine aminotransferase
- AST
aspartate aminotransferase
- DR
delayed-release
- FDA
US Food and Drug Administration
Footnotes
Disclosures. The authors declare no conflicts or financial interest in any product or service mentioned in the manuscript, including grants, equipment, medications, employment, gifts, and honoraria. The authors had full access to all patient information in this report and take responsibility for the integrity and accuracy of the report.
Ethical Approval and Informed Consent. Given the nature of this study, the institution review board/ethics committee review was not required and informed consent was not obtained.
References
- 1.Stockmann C, Constance JE, Roberts FK et al. Pharmacokinetics and pharmacodynamics of antifungals in children and their clinical implications. Clin Pharmacokinet. 2014;53(5):429–454. doi: 10.1007/s40262-014-0139-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Krishna G, Ma L, Martinho M, O'Mara E. Single-dose phase I study to evaluate the pharmacokinetics of posaconazole in new tablet and capsule formulations relative to oral suspension. Antimicrob Agents Chemother. 2012;56(8):4196–4201. doi: 10.1128/AAC.00222-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Noxafil posaconazole prescribing information. Merck and Co, Inc, September 2017. Accessed January 12, 2020. https://www.merck.com/product/usa/pi_circulars/n/noxafil/noxafil_pi.pdf.
- 4.Reed C, Bryant R, Ibrahim AS et al. Combination polyenecaspofungin treatment of rhino-orbital-cerebral mucormycosis. Clin Infect Dis. 2008;47(3):364–371. doi: 10.1086/589857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Cornely OA, Alastruey-Izquierdo A, Arenz D et al. Global guideline for the diagnosis and management of mucormycosis: an initiative of the European Confederation of Medical Mycology in cooperation with the Mycoses Study Group Education and Research Consortium. Lancet Infect Dis. 2019;19(12):e405–e421. doi: 10.1016/S1473-3099(19)30312-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Alastruey-Izquierdo A, Castelli MV, Cuesta I et al. In vitro activity of antifungals against Zygomycetes. Clin Microbiol Infect. 2009;15(suppl 5):71–76. doi: 10.1111/j.1469-0691.2009.02984.x. [DOI] [PubMed] [Google Scholar]
- 7.Bradley JS, Nelson JD, Barnett ED, editors. 2018 Nelson's Pediatric Antimicrobial Therapy. 24th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2018. [Google Scholar]
- 8.Arikan S, Sancak Bm, Alp S et al. Comparative in vitro activities of posaconazole, voriconazole, itraconazole, and amphotericin B against Aspergillus and Rhizopus and synergy testing for Rhizopus. Med Mycol. 2008;46(6):567–573. doi: 10.1080/13693780801975576. [DOI] [PubMed] [Google Scholar]
- 9.Krishna G, Sansone-Parsons A, Martinho M et al. Posaconazole plasma concentrations in juvenile patients with invasive fungal infection. Antimicrob Agents Chemother. 2007;51(3):812–810. doi: 10.1128/AAC.00454-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Döring M, Cabanillas Stanchi KM, Queudeville M et al. Efficacy, safety and feasibility of antifungal prophylaxis with posaconazole tablet in paediatric patients after haemotopoietic stem cell transplantation. J Cancer Res Clin Oncol. 2017;143(7):1281–1292. doi: 10.1007/s00432-017-2369-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Walsh TJ, Raad I, Patterson TF et al. Treatment of invasive aspergillosis with posaconazole in patients who are refractory to or intolerant of conventional therapy: an externally controlled trial. Clin Infect Dis. 2007;44(1):2–12. doi: 10.1086/508774. [DOI] [PubMed] [Google Scholar]
- 12.Jang SH, Colangelo PM, Gobburu JV. Exposure-response of posaconazole used for prophylaxis against invasive fungal infections: evaluating the need to adjust doses based on drug concentrations in plasma. Clin Pharmacol Ther. 2010;88(1):115–119. doi: 10.1038/clpt.2010.64. [DOI] [PubMed] [Google Scholar]
- 13.Dolton MJ, Ray JE, Chen SC et al. Multicenter study of posaconazole therapeutic drug monitoring: exposure-response relationship and factors affecting concentration. Antimicrob Agents Chemother. 2012;56(11):5503–5510. doi: 10.1128/AAC.00802-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Döring M, Müller C, Johann PD et al. Analysis of posaconazole as oral antifungal prophylaxis in pediatric patients under 12 years of age following allogeneic stem cell transplantation. BMC Infect Dis. 2012;12:263. doi: 10.1186/1471-2334-12-263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bernardo VA, Cross SJ, Crews KR et al. Posaconazole therapeutic drug monitoring in pediatric patients and young adults with cancer. Ann Pharmacother. 2013;47(7–8):979–983. doi: 10.1345/aph.1R775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Dekkers BGJ, Bakker M, van der Elst KCM et al. Therapeutic drug monitoring of posaconazole: an update. Curr Fungal Infect Rep. 2016;10:51–61. doi: 10.1007/s12281-016-0255-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Fleming S, Yannakou CK, Haeusler GM et al. Consensus guidelines for antifungal prophylaxis in haematological malignancy and haemopoietic stem cell transplantation, 2014. Intern Med J. 2014;44(12b):1283–1297. doi: 10.1111/imj.12595. [DOI] [PubMed] [Google Scholar]
- 19.Lehrnbecher T, Attarbaschi A, Duerken M et al. Posaconazole salvage treatment in paediatric patients: a multi-center survey. Eur J Clin Microbiol Infect Dis. doi: 10.1007/s10096-010-0957-4. 12 Use. Assessment report noxafil. EMA/159150/2014. Accessed on July 26, 2021. http//www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Assessment_Report_-_Variation/human/000610/WC500168187.pdf. [DOI] [PubMed]
- 20.Nguyen MH, Davis MR, Wittenberg R Clin Infect Dis. 2019. Posaconazole serum drug levels associated with pseudohyperaldosteronism. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Parkes LO, Cheng MP, Sheppard DC. Visual hallucinations associated with high posaconazole concentrations in serum. Antimicrob Agents Chemother. 2016;60:1170–1. doi: 10.1128/AAC.02739-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Martino J, Fisher BT, Boss KR et al. Suspected posaconazole toxicity in a pediatric oncology patient. Pediatr Blood Cancer. 2015;62(9):1682. doi: 10.1002/pbc.25568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.European Medicines Agency, Committee for Medicinal Products for Human Use Assessment report noxafil. EMA/159150/2014. Available from http//www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Assessment_Report_-_Variation/human/000610/WC500168187.pdf.
- 24.Kersemaekers WM, Dogterom P, Xu J et al. Effect of a high-fat meal on the pharmacokinetics of 300-milligram posaconazole in a solid oral tablet formulation. Antimicrob Agents Chemother. 2015;59(6):3385–3389. doi: 10.1128/AAC.05000-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kraft WK, Chang PS, van Iersel MLPS et al. Posaconazole tablet pharmacokinetics: lack of effect of concomitant medications altering gastric pH and gastric motility in healthy subjects. Antimicrob Agents Chemother. 2014;58(7):4020–4025. doi: 10.1128/AAC.02448-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Hens B, Corsetti M, Brouwers J et al. Gastrointestinal and systemic monitoring of posaconazole in humans after fasted and fed state administration of a solid dispersion. J Pharm Sci. 2016;105(9):2904–2912. doi: 10.1016/j.xphs.2016.03.027. [DOI] [PubMed] [Google Scholar]
