Skip to main content

Induction of CYP3A activity by dexamethasone may not be strong, even at high doses: insights from a case of tacrolimus co-administration

Abstract

Background

Dexamethasone (DEX) induces CYP3A activity in a concentration-dependent manner. However, no study has examined changes in the blood concentration of CYP3A substrate drugs when DEX is administered at high doses. Herein, we present a case in which tacrolimus (TAC), a typical CYP3A substrate drug, was co-administered with a chemotherapy regimen that included high-dose DEX.

Case presentation

A 71-year-old woman underwent liver transplantation for hepatocellular carcinoma 18 years prior to her inclusion in this case study. She was receiving TAC orally at 2 mg/day and had a stable trough blood concentration of approximately 4 ng/mL and a trough blood concentration/dose (C/D) ratio of approximately 2. The patient was diagnosed with post-transplant lymphoproliferative disease (histological type: Burkitt's lymphoma) after admission. Thereafter, the patient received cyclophosphamide-prednisolone (CP), followed by two courses of R-HyperCVAD (rituximab, cyclophosphamide, doxorubicin, vincristine, and DEX) and R-MA (rituximab, methotrexate, and cytarabine) replacement therapy. DEX (33 mg/day) was administered intravenously on days 1–4 and days 11–14 of R-HyperCVAD treatment, and aprepitant (APR) was administered on days 1–5 in both courses. The TAC C/D ratio decreased to approximately 1 on day 11 during both courses, and then increased. Furthermore, a decreasing trend in the TAC C/D ratio was observed after R-MA therapy. The decrease in the TAC C/D ratio was attributed to APR administration rather than to DEX.

Conclusion

The induction of CYP3A activity by a high dose of DEX may not be strong. The pharmacokinetic information on DEX and in vitro enzyme activity induction studies also suggested that CYP3A activity induction is not prominent under high-dose DEX treatment.

Background

Dexamethasone (DEX) is a steroid used to treat various diseases, and its dosage and duration of use vary widely depending on the indication and purpose for prescription [1]. DEX reportedly induces cytochrome P450 (CYP) 3A activity in vitro in a concentration-dependent manner, [2] and its label describes interactions with CYP3A substrate drugs [3, 4]. CYP3A is a major metabolizing enzyme in the human liver and intestine, and is involved in the metabolism of over half of all marketed drugs [5]. There are two types of clinically important drug–drug interactions (DDIs) [6]. One type is mediated by the inhibition of metabolic enzyme and transporter activities, whereas the other is mediated by the induction of these protein activities [6]. The therapeutic and adverse effects of drugs are often intensified by an increase in their blood concentrations caused by inhibition-based DDIs; however, these effects are generally diminished by a decrease in the blood concentrations caused by induction-based DDIs. Therefore, it is important to assess the extent of induction of CYP3A activity by DEX to ensure that the therapeutic effects of the combination drugs are maintained during co-administration.

McCune et al. reported that the increase in CYP3A activity, determined by measuring testosterone 6-β-hydroxylation level, in human hepatocytes treated with 2, 10, 50, 100, and 250 µM DEX was 1.7-, 1.9-, 3.9-, 6.9-, and 6.6-fold, respectively [2]. This finding indicates that DEX exerts a concentration-dependent effect on CYP3A activity induction [2]. In a study in which triazolam, a CYP3A substrate drug, was administered following low-dose oral DEX (1.5 mg/day) administration, no significant decrease in the blood concentration of triazolam was observed [7]. In an erythromycin breath test conducted on 12 healthy adults, 5-day oral administration of medium-dose DEX (16 mg/day) reportedly increased CYP3A activity by 25.7% [2]. However, the range of increase varied from -8% to 70%, indicating significant individual differences. In addition, the CYP3A activity increased by an average of 55% in patients undergoing the erythromycin breath test 2–9 days after the oral administration of 16–24 mg/day DEX [8]. However, it is important to note that this study included a limited number of participants, with a total of just five patients. Among them, 2 patients received 16 mg/day oral DEX, 1 received 18 mg/day oral DEX, 1 received 24 mg/day oral DEX, and 1 received 175 mg/day intravenous hydrocortisone for 2 days. However, the specific values were not provided for each participant.

DEX is often used at high doses (20–40 mg) for the treatment of multiple myeloma and in palliative care settings [1]. No study has examined changes in the blood levels of CYP3A substrate drugs when DEX is administered at high doses, and the impact of these changes remains unclear. Herein, we report a case in which tacrolimus (TAC), a typical CYP3A substrate drug, was combined with a chemotherapy regimen including high-dose DEX. We discuss the effects of this combination on the blood concentration of TAC and the strength of high-dose DEX in terms of its effect to induce CYP3A activity.

Case presentation

A 71-year-old Japanese woman underwent a living-donor liver transplant for hepatocellular carcinoma 18 years prior to her inclusion in this case study. She was receiving TAC orally at 2 mg/day (twice daily), with a trough blood concentration of approximately 4 ng/mL and stable trough blood concentration/dose (C/D) ratio (ng/mL/mg) of approximately 2. The patient presented night sweats, anorexia, and swelling on the left side of the neck. Owing to the presence of atypical cells in her peripheral blood and a substantial decrease in her platelet count (20,000/µL), she was urgently referred to our hospital for admission, thorough examination, and treatment. The patient was diagnosed with post-transplant lymphoproliferative disease (histological type: Burkitt's lymphoma) after admission. She was managed in the intensive care unit with continuous hemodiafiltration (CHDF) and positive airway pressure (CPAP) for 2 weeks because of tumor collapse syndrome, left subdural hematoma, and acute kidney injury (AKI). Figure 1 illustrates the course of this case. Upon admission, the patient presented with elevated lactate dehydrogenase (LDH; 11,232 U/L) and alanine aminotransferase (ALT; 164 U/L) levels, indicating potential liver dysfunction. Consequently, the TAC dose was reduced from 2 to 1 mg/day. Furthermore, owing to the presence of AKI, TAC administration was temporarily discontinued. As oxygenation improved and the patient's condition progressed, cyclophosphamide–prednisolone (CP) therapy was initiated. Following CP therapy, the LDH and ALT levels decreased. Subsequently, TAC administration was resumed at 2 mg/day. The patient received two cycles of R-HyperCVAD (rituximab, cyclophosphamide, doxorubicin, vincristine, and DEX) and R-MA (rituximab, methotrexate, cytarabine, and methylprednisolone) alternating therapy. DEX (33 mg/day) was administered intravenously on days 1–4 and days 11–14 of the R-HyperCVAD course. Additionally, aprepitant (APR) was administered on days 1–5 in both courses. The TAC dosage was adjusted to maintain a trough blood concentration of approximately 2 ng/mL. Fluconazole 200 mg/day was administered orally throughout the TAC-administration period.

Fig. 1
figure 1

Clinical course of the present patient. The doses of APR, DEX and TAC; TAC C/D ratio; and laboratory parameters are shown. APR, aprepitant; DEX, dexamethasone; TAC, tacrolimus; TAC C/D ratio: TAC trough blood concentration/dose ratio; LDH, lactate dehydrogenase; ALT, alanine aminotransferase

The C/D ratio of TAC during the first course of R-HyperCVAD was initially low (1.07) on day 11, but then increased, reaching 2.57 on day 25. Similarly, during the second course of R-HyperCVAD, the C/D ratio on day 3 (in Fig. 1, day 57) was 2.42, but subsequently decreased to 0.92 on day 11. However, it then exhibited an upward trend, reaching 1.8 on day 18 and 2.86 on day 32. The C/D ratio of TAC on day 4 (in Fig. 1, day 93) of the second course of R-MA was 4.4, but decreased to 1.25 on day 11. Notably, the C/D ratio started to increase during DEX administration on days 11–14 of R-HyperCVAD treatment and did not decrease after 2 weeks, whereas it exhibited a decreasing trend during R-MA treatment. Therefore, the decrease in the C/D ratio was primarily attributed to APR rather than to DEX.

Discussion and conclusions

TAC is a well-known CYP3A substrate, and its concentration can be influenced by both function of the liver and presence of CYP3A inhibitors and inducers [9]. Additionally, TAC is a P-glycoprotein (P-gp) substrate and may therefore be affected by P-gp inhibitors/inducers [10]. In our case, no significant changes were observed in liver function following the initiation of DEX treatment. In addition to DEX, concomitant medications such as APR and fluconazole can influence CYP3A activity. The patient consistently took 200 mg/day fluconazole while on TAC. Fluconazole is indeed classified as a moderate CYP3A inhibitor [6]. However, considering that the patient was taking the medication concurrently for an extended period, the impact of any fluctuations in TAC concentration was expected to be minimal. APR exhibits both inhibitory and inductive effects on CYP3A activity. In a study involving 12 healthy adults, oral administration of APR at 125 mg on day 1 and 80 mg on days 2–3 was followed by intravenous administration of 2 mg midazolam on days 4, 8, and 15. The area under the blood concentration curve (AUC) ratio for midazolam was 1.25 on day 4, 0.81 on day 8, and 0.96 on day 15 [11]. As these data were obtained after intravenous midazolam administration, it is expected that both inhibition and induction would be affected more when midazolam is administered orally because of the influence of the first-pass effect. APR is also an inducer of CYP2C9 [11], and Ohno et al. reported that its induction effect was observed approximately 2 weeks after the start of APR administration and generally recovered after 3 weeks [12, 13]. The mechanisms of induction of CYP3A and CYP2C activities are generally considered to be similar, and the duration of their induction depends on the turnover rate of the CYP enzymes [14, 15]. In the present case, the TAC C/D ratio decreased approximately 2 weeks after APR administration, with both R-HyperCVAD and R-MA treatments, and then began to increase. These findings suggest that the variation in the TAC C/D ratio in the present case can be attributed mainly to the CYP3A activity-inducing effect of APR. The possibility that the induction effect of DEX also contributes to this phenomenon cannot be ruled out. However, the fact that the TAC C/D ratio increased during DEX administration on days 11–14 of R-HyperCVAD treatment suggests that the effect, if any, is not substantial. Using Horn et al.'s Drug Interaction Probability Scale (DIPS), the interaction with DEX was rated as doubtful. However, the interaction with APR was rated as probable [16].

This case study had some limitations. Induction of CYP activity by DEX is mediated by the pregnane X receptor (PXR/NR1I2), whereas PXR/NR1I2 polymorphisms have been reported to affect DDIs of steroids and TAC [17]. Additionally, CYP3A5 polymorphisms are known to influence the pharmacokinetics of TAC [10, 18]. These findings imply that the extent of CYP activity induction by DEX may vary depending on the genetic background; however, information regarding these genetic polymorphisms was not available in this case.

Steroids other than DEX used in this case were methylprednisolone (40 mg/day on days 2–4) during R-MA therapy and methylprednisolone 2 mg/day throughout the duration of TAC administration, but the former was only administered for 3 days and the latter at a lower dose. Therefore, the effect on TAC concentration was considered to be minimal.

Doxorubicin and vincristine, administered in this case as part of the chemotherapy regimen, have been reported to be substrates for CYP3A and P-gp in in vitro studies [19,20,21]. Doxorubicin has also been reported to mildly increase the AUC of docetaxel, a substrate for both CYP3A4 and P-gp, by 50–75% [22]. On the contrary, in a study comprising nine patients treated with chemotherapy regimens containing doxorubicin or vincristine in combination with verapamil, a typical substrate for both CYP3A4 and P-gp, a decrease in AUC was reported in all but one patient, which might have been due to gastrointestinal mucosal damage [23]. No other drug interaction trials have been conducted with these anticancer drugs and CYP3A or P-gp substrate probe. Hence, the impact of these anticancer agents as CYP3A/P-gp inhibitors on the in vivo pharmacokinetics of tacrolimus remains unknown.

Furthermore, although hematocrit levels and inflammatory responses are known to influence the pharmacokinetics of TAC [24,25,26], no association was observed between the C/D ratio of TAC and the course of hematocrit and C-reactive protein levels (an inflammatory response marker) in the present case.

Recently, Hibino et al. reported that CYP3A activity increases on days 4–8 in patients taking DEX and APR [27]. As mentioned earlier, APR has both CYP3A activity-inhibitory and -inducing effects. In an interaction study on midazolam, the AUC of midazolam increased on day 4 and decreased on day 8 [11]. Therefore, the relatively early increase in CYP3A activity reported by Hibino et al. [27] may be partially attributed to DEX. On the contrary, one of the possible reasons for the increase in TCR C/D to 4.4 on day 4 of the second course of R-MA therapy in the present case may be that inhibition was more significant than induction, as APR was administered for only 3 days.

McCune et al. reported the dose-dependent induction of CYP3A activity in human hepatocytes treated with 2–250 µM DEX; however, CYP3A activity-inducing effect was not observed at levels below 1 µM. Considering that the average blood concentration of DEX at 0.5–3.0 h after 16 mg/day oral administration is 0.1 µM, [2] the blood concentration of DEX intravenously administered at 33 mg/day (oral bioavailability of approximately 80% [28], equivalent to 40 mg/day orally) would be approximately 0.25 µM. Moreover, the concentration of the unbound form would be even lower, which is consistent with the fact that the CYP3A activity-inducing effect was not strong in this case.

Although DEX at moderate doses has been reported to result in mild induction of CYP3A activity in erythromycin breath tests, these reports are highly variable [2, 8]. Moreover, no clinical trial has examined the effect of moderate or higher doses of DEX on blood concentrations of typical CYP3A substrate drugs, and conducting such a trial in the future would be ethically unlikely. Therefore, this case, in which high-dose DEX was combined with TAC, out of clinical necessity, is valuable as it suggests that the CYP3A activity-inducing effect of high-dose DEX is not significant.

In conclusion, the C/D ratio trend of TAC in this case suggested that the CYP3A activity-inducing effect of high-dose DEX was not strong. The pharmacokinetic information on DEX and the results of an in vitro enzyme activity-induction study also support the notion that the CYP3A activity-inducing effect of high-dose DEX is not substantial.

Availability of data and materials

Data used in this case study will not be shared owing to the risk of identifying the patient.

Abbreviations

DEX:

Dexamethasone

TAC:

Tacrolimus

C/D:

Trough blood concentration/dose

APR:

Aprepitant

CYP:

Cytochrome P450

P-gp:

P-glycoprotein

LDH:

Lactate dehydrogenase

ALT:

Alanine aminotransferase

References

  1. Dexamethasone. Available from: https://www.uptodate.com/contents/dexamethasone-systemic-drug-information. Accessed 1 Jul 2023.

  2. McCune JS, Hawke RL, LeCluyse EL, Gillenwater HH, Hamilton G, Ritchie J, et al. In vivo and in vitro induction of human cytochrome P4503A4 by dexamethasone. Clin Pharmacol Ther. 2000;68(4):356–66. https://doi.org/10.1067/mcp.2000.110215.

    Article  CAS  PubMed  Google Scholar 

  3. Dexametasone(HEMADY), Labeling-Package Insert, Revised: 6/2021. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/211379s003lbl.pdf. Accessed 1 Jul 2023.

  4. Dexamethasone sodium phosphate(DECADRON), Labeling-Package Insert, Revised: 5/2022. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/211379s003lbl.pdf. Accessed 1 Jul 2023.

  5. Rogers JF, Nafziger AN, Bertino JS Jr. Pharmacogenetics affects dosing, efficacy, and toxicity of cytochrome P450-metabolized drugs. Am J Med. 2002;113(9):746–50. https://doi.org/10.1016/s0002-9343(02)01363-3.

    Article  CAS  PubMed  Google Scholar 

  6. Maeda K, Hisaka A, Ito K, Ohno Y, Ishiguro A, Sato R, et al. Classification of drugs for evaluating drug interaction in drug development and clinical management. Drug Metab Pharmacokinet. 2021;41:100414. https://doi.org/10.1016/j.dmpk.2021.100414.

    Article  CAS  PubMed  Google Scholar 

  7. Villikka K, Kivisto KT, Neuvonen PJ. The effect of dexamethasone on the pharmacokinetics of triazolam. Pharmacol Toxicol. 1998;83(3):135–8. https://doi.org/10.1111/j.1600-0773.1998.tb01457.x.

    Article  CAS  PubMed  Google Scholar 

  8. Watkins PB, Murray SA, Winkelman LG, Heuman DM, Wrighton SA, Guzelian PS. Erythromycin breath test as an assay of glucocorticoid-inducible liver cytochromes P-450. Studies in rats and patients. J Clin Invest. 1989;83(2):688–97. https://doi.org/10.1172/JCI113933.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Venkataramanan R, Swaminathan A, Prasad T, Jain A, Zuckerman S, Warty V, et al. Clinical pharmacokinetics of tacrolimus. Clin Pharmacokinet. 1995;29(6):404–30. https://doi.org/10.2165/00003088-199529060-00003.

    Article  CAS  PubMed  Google Scholar 

  10. Haufroid V, Mourad M, Van Kerckhove V, Wawrzyniak J, De Meyer M, Eddour DC, Malaise J, Lison D, Squifflet JP, Wallemacq P. The effect of CYP3A5 and MDR1 (ABCB1) polymorphisms on cyclosporine and tacrolimus dose requirements and trough blood levels in stable renal transplant patients. Pharmacogenetics. 2004;14:147–54.

    Article  CAS  PubMed  Google Scholar 

  11. Shadle CR, Lee Y, Majumdar AK, Petty KJ, Gargano C, Bradstreet TE, et al. Evaluation of potential inductive effects of aprepitant on cytochrome P450 3A4 and 2C9 activity. J Clin Pharmacol. 2004;44(3):215–23. https://doi.org/10.1177/0091270003262950.

    Article  CAS  PubMed  Google Scholar 

  12. Ohno Y, Yamada M, Yamaguchi R, Hisaka A, Suzuki H. Persistent drug interaction between aprepitant and warfarin in patients receiving anticancer chemotherapy. Int J Clin Pharm. 2014;36(6):1134–7. https://doi.org/10.1007/s11096-014-0022-y.

    Article  CAS  PubMed  Google Scholar 

  13. Takaki J, Ohno Y, Yamada M, Yamaguchi R, Hisaka A, Suzuki H. Assessment of drug-drug Interaction between warfarin and aprepitant and its effects on PT-INR of patients receiving anticancer chemotherapy. Biol Pharm Bull. 2016;39(5):863–8. https://doi.org/10.1248/bpb.b16-00014.

    Article  CAS  PubMed  Google Scholar 

  14. Hewitt NJ, Lecluyse EL, Ferguson SS. Induction of hepatic cytochrome P450 enzymes: methods, mechanisms, recommendations, and in vitro-in vivo correlations. Xenobiotica. 2007;37(10–11):1196–224. https://doi.org/10.1080/00498250701534893.

    Article  CAS  PubMed  Google Scholar 

  15. Honma M, Kozawa M, Suzuki H. Methods for the quantitative evaluation and prediction of CYP enzyme induction using human in vitro systems. Expert Opin Drug Discov. 2010;5(5):491–511. https://doi.org/10.1517/17460441003762717.

    Article  CAS  PubMed  Google Scholar 

  16. Horn JR, Hansten PD, Chan LN. Proposal for a new tool to evaluate drug interaction cases. Ann Pharmacother. 2007;41(4):674–80. https://doi.org/10.1345/aph.1H423.

    Article  PubMed  Google Scholar 

  17. Stifft F, van Kuijk SMJ, Bekers O, Christiaans MHL. Increase in tacrolimus exposure after steroid tapering is influenced by CYP3A5 and pregnane X receptor genetic polymorphisms in renal transplant recipients. Nephrol Dial Transplant. 2018;33:1668–75. https://doi.org/10.1093/ndt/gfy096.

    Article  CAS  PubMed  Google Scholar 

  18. Hesselink DA, van Schaik RH, van der Heiden IP, van der Werf M, Gregoor PJ, Lindemans J, et al. Genetic polymorphisms of the CYP3A4, CYP3A5, and MDR-1 genes and pharmacokinetics of the calcineurin inhibitors cyclosporine and tacrolimus. Clin Pharmacol Ther. 2003;74:245–54. https://doi.org/10.1016/S0009-9236(03)00168-1.

    Article  CAS  PubMed  Google Scholar 

  19. Gianni L, Vigano L, Locatelli A, Capri G, Giani A, Tarenzi E, et al. Human pharmacokinetic characterization and in vitro study of the interaction between doxorubicin and paclitaxel in patients with breast cancer. J Clin Oncol. 1997;15:1906–15. https://doi.org/10.1200/JCO.1997.15.5.1906.

    Article  CAS  PubMed  Google Scholar 

  20. Szakacs G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM. Targeting multidrug resistance in cancer. Nat Rev Drug Discov. 2006;5:219–34. https://doi.org/10.1038/nrd1984.

    Article  CAS  PubMed  Google Scholar 

  21. Dennison JB, Kulanthaivel P, Barbuch RJ, Renbarger JL, Ehlhardt WJ, Hall SD. Selective metabolism of vincristine in vitro by CYP3A5. Drug Metab Dispos. 2006;34:1317–27. https://doi.org/10.1124/dmd.106.009902.

    Article  CAS  PubMed  Google Scholar 

  22. D’Incalci M, Schuller J, Colombo T, Zucchetti M, Riva A. Taxoids in combination with anthracyclines and other agents: pharmacokinetic considerations. Semin Oncol. 1998;25:16–20.

    CAS  PubMed  Google Scholar 

  23. Kuhlmann J, Woodcock B, Wilke J, Welling H, Rietbrock N. Verapamil plasma concentrations during treatment with cytostatic drugs. J Cardiovasc Pharmacol. 1985;7:1003–6. https://doi.org/10.1097/00005344-198509000-00030.

    Article  CAS  PubMed  Google Scholar 

  24. Kirubakaran R, Stocker SL, Hennig S, Day RO, Carland JE. Population pharmacokinetic models of tacrolimus in adult transplant recipients: A systematic review. Clin Pharmacokinet. 2020;59:1357–92. https://doi.org/10.1007/s40262-020-00922-x.

    Article  CAS  PubMed  Google Scholar 

  25. Enokiya T, Nishikawa K, Hamada Y, Ikemura K, Sugimura Y, Okuda M. Temporary decrease in tacrolimus clearance in cytochrome P450 3A5 non-expressors early after living donor kidney transplantation: Effect of interleukin 6-induced suppression of the cytochrome P450 3A gene. Basic Clin Pharmacol Toxicol. 2021;128:525–33. https://doi.org/10.1111/bcpt.13539.

    Article  CAS  PubMed  Google Scholar 

  26. Chavant A, Fonrose X, Gautier-Veyret E, Hilleret MN, Roustit M, Stanke-Labesque F. Variability of tacrolimus trough concentration in liver transplant patients: Which role of inflammation? Pharmaceutics. 2021;13:1960. https://doi.org/10.3390/pharmaceutics13111960.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Hibino H, Sakiyama N, Makino Y, Makihara-Ando R, Horinouchi H, Fujiwara Y, et al. Evaluation of hepatic CYP3A enzyme activity using endogenous markers in lung cancer patients treated with cisplatin, dexamethasone, and aprepitant. Eur J Clin Pharmacol. 2022;78:613–21. https://doi.org/10.1007/s00228-022-03275-5.

    Article  CAS  PubMed  Google Scholar 

  28. Duggan DE, Yeh KC, Matalia N, Ditzler CA, McMahon FG. Bioavailability of oral dexamethasone. Clin Pharmacol Ther. 1975;18:205–9. https://doi.org/10.1002/cpt1975182205.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank all staff who were involved in this study.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Author information

Authors and Affiliations

Authors

Contributions

YO and TO conceptualized and designed the study. YO drafted the manuscript. YO and TO acquired patient data. TT critically reviewed the manuscript. AH and MK are the primary doctors and supervised the treatment. The authors read and approved the final manuscript.

Corresponding author

Correspondence to Yoshiyuki Ohno.

Ethics declarations

Ethics approval and consent to participate

This study was approved by the institutional review board of the Graduate School of Medicine and Faculty of Medicine, The University of Tokyo (Approval numbers: 2654-11. This study received general consent. Nevertheless, as this was a case study performed using data obtained from routine medical care, the study was conducted with full consideration of protecting patient’s personal information according to the guidelines on privacy policy.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ohno, Y., Oriyama, T., Honda, A. et al. Induction of CYP3A activity by dexamethasone may not be strong, even at high doses: insights from a case of tacrolimus co-administration. J Pharm Health Care Sci 9, 39 (2023). https://doi.org/10.1186/s40780-023-00310-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40780-023-00310-0

Keywords