Rifampicin Alters Metformin Plasma Exposure but Not Blood Glucose Levels in Diabetic Tuberculosis Patients

Lindsey H M Te Brake, Vycke Yunivita, Resvi Livia, Nanny Soetedjo, Eleonora van Ewijk-Beneken Kolmer, Jan B Koenderink, David M Burger, Prayudi Santoso, Reinout van Crevel, Bachti Alisjahbana, Rob E Aarnoutse, Rovina Ruslami, TANDEM Consortium, Lindsey H M Te Brake, Vycke Yunivita, Resvi Livia, Nanny Soetedjo, Eleonora van Ewijk-Beneken Kolmer, Jan B Koenderink, David M Burger, Prayudi Santoso, Reinout van Crevel, Bachti Alisjahbana, Rob E Aarnoutse, Rovina Ruslami, TANDEM Consortium

Abstract

The pharmacokinetic (PK) and clinical implications of combining metformin with rifampicin are relevant to increasing numbers of patients with diabetic tuberculosis (TB) across the world and are yet unclear. We assessed the impact of rifampicin on metformin PKs and its glucose-lowering effect in patients with diabetic TB by measuring plasma metformin and blood glucose during and after TB treatment. Rifampicin increased metformin exposure: plasma area under the plasma concentration-time curve from time point 0 to the end of the dosing interval (AUC0-τ ) and peak plasma concentration (Cmax ) geometric mean ratio (GMR; during vs. after TB treatment) were 1.28 (90% confidence interval (CI) 1.13-1.44) and 1.19 (90% CI 1.02-1.38; n = 22). The metformin glucose-lowering efficacy did not change (Δglucose - Cmax ; P = 0.890; n = 18). Thus, we conclude that additional glucose monitoring in this population is not warranted. Finally, 57% of patients on metformin and rifampicin, and 38% of patients on metformin alone experienced gastrointestinal adverse effects. Considering this observation, we advise patients to take metformin and rifampicin with food and preferably separated in time. Clinicians could consider metoclopramide if gastrointestinal adverse effects occur.

Conflict of interest statement

The authors declared no competing interests for this work.

© 2018 The Authors Clinical Pharmacology & Therapeutics published by Wiley Periodicals, Inc. on behalf of American Society for Clinical Pharmacology and Therapeutics.

Figures

Figure 1
Figure 1
Schematic overview of the most important transporters involved in the absorption, hepatic uptake, and excretion of metformin.14, 38, 39, 40, 41, 42 The physiological significance of the transporters depicted in gray is unclear.14 MATE, multidrug and toxin extrusion; OCT, organic cation transporter; PMAT, plasma membrane monoamine transporter.
Figure 2
Figure 2
Individual changes in steady‐state metformin pharmacokinetic parameters, AUC 0–τ (a), Cmax (b) and tubular secretion (c), with and without co‐administration of rifampicin. Metformin plasma concentrations were extrapolated from 8 until 12–24 hours to calculate AUC 0–τ for patients taking metformin once or twice daily. Tubular secretion was based on observed urine collections up to 8 hours after metformin intake. Data were assessed with paired‐samples t test on the log‐transformed parameters. AUC0–τ, area under the plasma concentration‐time curve from time point 0 to the end of the dosing interval; Cmax, maximum plasma concentration. ETH, ethambutol; IC, informed consent; INH, isoniazid; MTF, metformin; PK‐GC, pharmacokinetic‐glucose curve sampling session; PZA, pyrazinamide; RIF, rifampicin.
Figure 3
Figure 3
Individual changes in G‐AUC (a) and Gmax (b), after a 75‐g glucose challenge, with and without co‐administration of rifampicin. Data were assessed with paired‐samples t test on log‐transformed blood glucose parameters (n = 18). AUC, area under the blood concentration‐time curve from; Gmax, maximum blood glucose concentration.
Figure 4
Figure 4
Schematic overview of the study design in weeks from the start of tuberculosis (TB) treatment (wk 0). The TB treatment period (week 0–24) is colored dark gray. Patients were enrolled if they were continuation phase of TB treatment. Daily intake of TB drugs (for > 7days) is indicated by dashed lines. In between sessions, there was at least a 1‐month washout period, after which any induction caused by rifampicin was expected to have dissipated.9 AUC, area under the plasma concentration‐time curve; Cmax, maximum plasma concentration.

References

    1. WHO . Global tuberculosis report 2018. <> (2018).
    1. Jeon, C.Y. & Murray, M.B. Diabetes mellitus increases the risk of active tuberculosis: a systematic review of 13 observational studies. PLoS Med. 5, e152 (2008).
    1. Riza, A.L. et al Clinical management of concurrent diabetes and tuberculosis and the implications for patient services. Lancet Diabetes Endocrinol. 2, 740–753 (2014).
    1. Lonnroth, K. , Roglic, G. & Harries, A.D. Improving tuberculosis prevention and care through addressing the global diabetes epidemic: from evidence to policy and practice. Lancet Diabetes Endocrinol. 2, 730–739 (2014).
    1. Baker, M.A. et al The impact of diabetes on tuberculosis treatment outcomes: a systematic review. BMC Med. 9, 81 (2011).
    1. Takasu, N. et al Rifampicin‐induced early phase hyperglycemia in humans. Am. Rev. Respir. Dis. 125, 23–27 (1982).
    1. Waterhouse, M. , Wilson, C. , White, V.L. & Chowdhury, T.A. Resolution of insulin‐requiring diabetes after cessation of chemotherapy for tuberculosis. J. R. Soc. Med. 98, 270–271 (2005).
    1. Ruslami, R. , Aarnoutse, R.E. , Alisjahbana, B. , van der Ven, A.J. & van Crevel, R. Implications of the global increase of diabetes for tuberculosis control and patient care. Trop. Med. Int. Health 15, 1289–1299 (2010).
    1. Niemi, M. , Backman, J.T. , Fromm, M.F. , Neuvonen, P.J. & Kivisto, K.T. Pharmacokinetic interactions with rifampicin: clinical relevance. Clin. Pharmacokinet. 42, 819–850 (2003).
    1. Burman, W.J. , Gallicano, K. & Peloquin, C. Comparative pharmacokinetics and pharmacodynamics of the rifamycin antibacterials. Clin. Pharmacokinet. 40, 327–341 (2001).
    1. IDF . Global Guideline for Type 2 Diabetes. <> (2012).
    1. Intensive blood‐glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). UK Prospective Diabetes Study (UKPDS) group. Lancet. 352, 837–853 (1998).
    1. Hermann, L.S. , Schersten, B. , Bitzen, P.O. , Kjellstrom, T. , Lindgarde, F. & Melander, A. Therapeutic comparison of metformin and sulfonylurea, alone and in various combinations. A double‐blind controlled study. Diabetes Care 17, 1100–1109 (1994).
    1. Graham, G.G. et al Clinical pharmacokinetics of metformin. Clin. Pharmacokinet. 50, 81–98 (2011).
    1. Summary of Product Characteristics Glucophage 500 mg and 850 mg. Updated January 23, 2015 by Merck. <>.
    1. Campbell, R.K. , Jr. White, J.R. & Saulie, B.A. Metformin: a new oral biguanide. Clin. Ther. 18, 360–371 (1996); discussion 359.
    1. Chen, J. & Raymond, K. Roles of rifampicin in drug‐drug interactions: underlying molecular mechanisms involving the nuclear pregnane X receptor. Ann. Clin. Microbiol. Antimicrob. 5, 3 (2006).
    1. Maeda, T. et al Effect of pregnane X receptor ligand on pharmacokinetics of substrates of organic cation transporter Oct1 in rats. Drug Metab. Dispos. 35, 1580–1586 (2007).
    1. Cho, S.K. et al Rifampin enhances the glucose‐lowering effect of metformin and increases OCT1 mRNA levels in healthy participants. Clin. Pharmacol. Ther. 89, 416–421 (2011).
    1. Xu, C. et al The altered renal and hepatic expression of solute carrier transporters (SLCs) in type 1 diabetic mice. PLoS One 10, e0120760 (2015).
    1. Atilano‐Roque, A. , Roda, G. , Fogueri, U. , Kiser, J.J. & Joy, M.S. Effect of disease pathologies on transporter expression and function. J. Clin. Pharmacol. 56(suppl. 7), S205–S221 (2016).
    1. Staudinger, J.L. Disease, drug metabolism, and transporter interactions. Pharm. Res. 30, 2171–2173 (2013).
    1. Williams, R.L. , Chen, M.L. & Hauck, W.W. Equivalence approaches. Clin. Pharmacol. Ther. 72, 229–237 (2002).
    1. Zhou, M. , Xia, L. & Wang, J. Metformin transport by a newly cloned proton‐stimulated organic cation transporter (plasma membrane monoamine transporter) expressed in human intestine. Drug Metab. Dispos. 35, 1956–1962 (2007).
    1. Gerich, J.E. Role of the kidney in normal glucose homeostasis and in the hyperglycaemia of diabetes mellitus: therapeutic implications. Diabet. Med. 27, 136–142 (2010).
    1. Gerich, J.E. Physiology of glucose homeostasis. Diabetes Obes. Metab. 2, 345–350 (2000).
    1. Spruiell, K. , Richardson, R.M. , Cullen, J.M. , Awumey, E.M. , Gonzalez, F.J. & Gyamfi, M.A. Role of pregnane X receptor in obesity and glucose homeostasis in male mice. J. Biol. Chem. 289, 3244–3261 (2014).
    1. Ihunnah, C.A. , Jiang, M. & Xie, W. Nuclear receptor PXR, transcriptional circuits and metabolic relevance. Biochim. Biophys. Acta 1812, 956–963 (2011).
    1. Lin, H.C. , Yu, M.C. , Liu, H.J. & Bai, K.J. Impact of food intake on the pharmacokinetics of first‐line antituberculosis drugs in Taiwanese tuberculosis patients. J. Formos. Med. Assoc. 113, 291–297 (2014).
    1. Ciarimboli, G. et al Proximal tubular secretion of creatinine by organic cation transporter OCT2 in cancer patients. Clin. Cancer Res. 18, 1101–1108 (2012).
    1. Breyer, M.D. & Qi, Z. Better nephrology for mice – and man. Kidney Int. 77, 487–489 (2010).
    1. Inker, L.A. et al Estimating glomerular filtration rate from serum creatinine and cystatin C. N. Engl. J. Med. 367, 20–29 (2012).
    1. Levey, A.S. et al A new equation to estimate glomerular filtration rate. Ann. Intern. Med. 150, 604–612 (2009).
    1. Diletti, E. , Hauschke, D. & Steinijans, V.W. Sample size determination for bioequivalence assessment by means of confidence intervals. Int. J. Clin. Pharmacol. Ther. Toxicol. 29, 1–8 (1991).
    1. Common Terminology Criteria for Adverse Events 4.0 (CTCAE) . <>.
    1. Devandla, A. , Yamsani, S.K. & Yamsani, M.R. Effect of rifampicin pretreatment on the oral bioavailability of domperidone in healthy human volunteers. Drug Metab. Pers. Ther. 30, 257–261 (2015).
    1. van Crevel, R. et al Bioavailability of rifampicin in Indonesian subjects: a comparison of different local drug manufacturers. Int. J. Tuberc. Lung Dis. 8, 500–503 (2004).
    1. Kusuhara, H. et al Effects of a MATE protein inhibitor, pyrimethamine, on the renal elimination of metformin at oral microdose and at therapeutic dose in healthy subjects. Clin. Pharmacol. Ther. 89, 837–844 (2011).
    1. Muller, J. , Lips, K.S. , Metzner, L. , Neubert, R.H. , Koepsell, H. & Brandsch, M. Drug specificity and intestinal membrane localization of human organic cation transporters (OCT). Biochem. Pharmacol. 70, 1851–1860 (2005).
    1. Nies, A.T. et al Expression of organic cation transporters OCT1 (SLC22A1) and OCT3 (SLC22A3) is affected by genetic factors and cholestasis in human liver. Hepatology 50, 1227–1240 (2009).
    1. Tzvetkov, M.V. et al The effects of genetic polymorphisms in the organic cation transporters OCT1, OCT2, and OCT3 on the renal clearance of metformin. Clin. Pharmacol. Ther. 86, 299–306 (2009).
    1. Motohashi, H. & Inui, K. Organic cation transporter OCTs (SLC22) and MATEs (SLC47) in the human kidney. AAPS J. 15, 581–588 (2013).

Source: PubMed

3
Abonner