Skip to main content

Appropriate use of triazolam in elderly patients considering a quantitative benefit-risk assessment based on the pharmacokinetic-pharmacodynamic modeling and simulation approach supported by real-world data

Abstract

Background

Triazolam is a typical drug commonly used in the elderly; however, there have been concerns about its adverse events resulting from age-related changes in physiological function and drug interactions with concomitant drugs. Thus, updated information contributing to the appropriate use based on the latest pharmacokinetic and post-marketing surveillance methods is needed. In this study, we evaluated the appropriate use of triazolam in the elderly by integrating real-world data with a modeling and simulation approach.

Methods

The occurrence risk of adverse events in the elderly was evaluated using the spontaneous adverse event reporting regulatory databases from Japan and the United States. Information on drug concentrations and reactions was extracted from previous publications to estimate the threshold for plasma triazolam concentrations that cause adverse events. The pharmacokinetic/pharmacodynamic (PK/PD) model was then constructed, and the dose and administration were evaluated in various situations anticipated in medical practice.

Results

Among all prescriptions, 25.4% were prescribed to individuals aged 80 years or above, and 51.8% were for those aged 70 years or above. A majority of cases involved CYP3A-metabolized drug combinations, accounting for 85.6%. Elderly individuals were at a higher risk of developing delirium and fall-fracture. Based on the constructed PK/PD model, the risk of adverse events increased when the plasma concentration of triazolam exceeded the calculated threshold of 0.44 ng/mL at approximately 6 h after administration. Administering 0.125 mg of triazolam, is half the approved dose for the elderly in Japan was deemed appropriate. Moreover, there was a substantial risk of adverse events even at a dosage of 0.0625 mg in combination with a moderate or strong inhibitor of cytochrome P450 3 A.

Conclusion

Analyzing large-scale databases and existing research publications on PK/PD can practically contribute to optimizing triazolam drug therapy for the elderly in the daily clinical setting.

Peer Review reports

Introduction

Population aging has become an issue in many countries, and drug therapy for the elderly is becoming increasingly important. The elderly are prone to potential safety issues resulting from many factors, such as age-related decline in liver and kidney function [1,2,3] and polypharmacy [4, 5]. To improve drug therapy for the elderly, guidelines on the appropriate use of pharmaceuticals have been issued in Japan [6, 7]; however, generally, the elderly do not participate in clinical studies and there is insufficient data on drug dosing, including dosage and administration adjustments for pharmaceuticals. Therefore, it is necessary to generate new information to improve drug therapy that considers the unique characteristics of the elderly.

Chronic insomnia is common in the elderly [8, 9], and triazolam, a short-acting benzodiazepine receptor agonist, is frequently prescribed [10, 11]. However, triazolam poses the risk of inducing geriatric syndromes, such as amnesia; thus, it is necessary to avoid using it as much as possible [12,13,14]. In addition, benzodiazepine hypnotics have a prolonged half-life in the elderly [15,16,17]. Greenblatt et al. observed an increase in plasma triazolam concentration with reduced clearance in the elderly when triazolam was administered to young people (mean, 30 years) and the elderly (mean, 69 years). They also reported that indicators of delirium and cognitive function correlated with changes in plasma concentration [18]. Therefore, when using triazolam in the elderly, there is a higher risk of developing adverse events associated with exposure to high drug concentrations; thus, the practice of more thorough drug treatments is required. However, triazolam has been on the market for a long time and is used based on the experience of healthcare professionals in many cases, so it is a major issue to update information contributing to its proper use.

Epidemiological studies that evaluate the appropriate use of pharmaceuticals and research that reexamine the available data have received a lot of attention recently. In particular, post-marketing pharmacovigilance has become important [19], and the use of large-scale databases is essential for analyzing the safety information of pharmaceuticals. In addition, spontaneous adverse event reporting databases are publicly available in the United States and Japan, which reflect the real-world. They have made it possible to gather data on age groups and lengths of therapy, such as data on elderly patients that is not often available through clinical trials. The pharmaceutical evaluation method of modeling and simulation is also widely used, from pharmaceutical development to actual clinical practice. It contributes significantly to the prediction of the efficacy and adverse events of pharmaceuticals [20]. These methodologies allow for the extraction and evaluation of previously unidentified issues and trends.

This study aimed to provide evidence that contributes to the optimization of triazolam drug therapy in the elderly by integrating spontaneous adverse event reporting databases with existing information on pharmacokinetics and pharmacodynamics.

Methods

Data sources

To analyze triazolam prescriptions and patient age distribution, we extracted claims data from April 2014 to March 2018 from Japan’s National Database of Health Insurance Claims and Specific Health Checkups (NDB), published by the Ministry of Health, Labor, and Welfare. To comprehensively assess the appropriate use of triazolam, we first evaluated adverse event spontaneous reporting data that reflected the medical practice in Japan and also considered information on similar databases in the United States. Both the Japanese Adverse Drug Event Report database (JADER) and the FDA Adverse Event Reporting System (FAERS) were utilized to analyze a wide range of adverse event data from both databases, covering the period from April 2004 to March 2019. Adverse event data in patients receiving triazolam were analyzed to evaluate concomitant drug use using information from JADER. For pharmacokinetic interactions, using the University of Washington Drug Interaction Database, drugs with an area under the blood concentration-time curve increase ratio (AUCR) after triazolam dosing of 1.25 times or greater were extracted and are listed in Table 1 [21, 22].

Table 1 Drug-drug interaction for triazolam

Definitions of adverse events

Fall-fracture, delirium, disturbed consciousness, liver disorders, renal disorders, gastrointestinal disorders, and hypoglycemia were extracted as the seven typical adverse events associated with triazolam in the elderly [23, 24]. Based on the terminology described in the Medical Dictionary for Regulatory Activities Japanese version (MedDRA/J, version 22.0), adverse events were classified using the preferred terms listed below the classification. To determine the occurrence risk of adverse events, the cut-off age was set at 70 and 80 years, and the evaluation was performed with reporting odds ratio (ROR) and 95% confidence interval (95% CI) in the elderly versus the non-elderly. The signal detection criteria for adverse event occurrence was determined as 95%CI > 1.

Pharmacokinetic-pharmacodynamic modeling

To obtain information on changes in triazolam blood concentration and drug susceptibility (sedation and cognitive function), we extracted the mean value of each from the report by Greenblatt et al. [18] using WebPlot Digitizer version 4.3 (https://automeris.io/WebPlotDigitizer/). Since the original literature used as a reference for the analysis provided only mean values of blood concentration time course obtained from information on young and elderly subjects (30 years old, 72 kg and 69 years old, 69 kg), the effect of age was incorporated in a general power law form in the pharmacokinetic-pharmacodynamic (PK/PD) model based on this literature information [18]:

$${\text{clearance}} = {{{\theta }}_{{\text{clearance}}}} \times {\left( {\frac{{{\text{age}}}}{{30}}} \right)^{{{\theta{\text{cov}}}}}}$$
(1)

where θ is the estimated value of parameters. The basic pharmacokinetic model used in this study is a one-compartment model; when the dose is constant, the inverse of AUCR was added as a covariate of clearance. We set the threshold for drug susceptibility at the value where the fluctuation was less than 10% compared to the value with the largest fluctuation. This threshold was based on the signal-to-noise ratio of 10, a predominantly used lower limit in quantification, and previous results [18] showing minimal PD fluctuation in placebo and baseline regression post-triazolam administration. We calculated the blood concentration during the development of drug susceptibility using receiver-operating characteristic (ROC) analysis for values below this threshold. A PK/PD model was developed using a nonlinear regression model to estimate changes in drug susceptibility versus blood concentration. Both direct response and effect compartment models were considered for the PD model, with testing conducted for linear and Emax models, respectively. In the setting of the error model, inter-individual variability was not estimated, and the proportional error model (Eq. 2) was used for the PK model. In contrast, the additive error model (Eq. 3) was used for the PD model as residual variability:

$${\text{Obserbatio}}{{\text{n}}_{\text{j}}}={\text{Predictio}}{{\text{n}}_{\text{j}}} \times \left( {1+{\varepsilon}} \right)$$
(2)
$${\text{Obserbatio}}{{\text{n}}_{\text{j}}}={\text{Predictio}}{{\text{n}}_{\text{j}}}+{\varepsilon}$$
(3)

Obserbationj and Predictionj are the jth observation and prediction, respectively, whereas ε denotes a random variable following a normal distribution with a mean of 0 and a variance of σ2. The validity of the model was determined using a coefficient of variation (CV%) of the estimated values, objective function value (OFV), goodness-of-fit plots and Akaike’s information criterion. The NLME software (version 8.1; Certara, St. Louis, MO, USA) was used for model construction, parameter estimation, and simulation.

Simulation

The following estimates in different age groups during the combined use of typical drugs were evaluated by simulation using the constructed final model:

I: Estimation of the time required for blood concentration after 0.25 mg of triazolam dosing to decrease to the threshold in healthy young people.

II: Estimation of acceptable triazolam dose and administration, in which the blood concentration reaches the threshold at the time estimated by I in the elderly.

III: Estimation of changes in blood concentration during the combined use of 0.0625 mg of triazolam and various cytochrome P450 (CYP) 3 A inhibitors and estimation of the acceptable AUCR range, in which the blood concentration reaches the threshold at the time estimated by I in the elderly.

Statistical analysis

The R Commander Plug-in for the EZR package version 4.0.2 (RcmdrPlugin.EZR) was used for statistical analyses [25].

Results

The quantity of triazolam prescribed in Japan from April 2014 to March 2018 was 181.6 − 275.9 million tablets per year, and no particular trend was observed in the age distribution of patients prescribed with triazolam between the years. Of all prescriptions, 25.4% were for individuals aged 80 years or above, and 51.8% were for those aged 70 years or above. In addition, only 2.3% of all prescriptions were for individuals under 30 years of age. Among all of the JADER data, there were 4116 entries containing triazolam, which was the 97th highest among all drugs registered in JADER, after the hypnotic sedatives brotizolam, zolpidem, etizolam, and flunitrazepam. There were many combinations containing benzodiazepines and cases of drug combinations involving CYP3A in metabolism, accounting for the majority at 3542 cases (85.6%). Fifteen drugs were extracted as drugs with an AUCR of 1.25-fold or greater (Table 1). Of these, there were 354 cases (8.6%) of combinations with triazolam. Among these, there were 78 cases of combinations with clarithromycin, 16 cases with itraconazole, and no case with ritonavir, respectively, which are strong CYP3A inhibitors [21, 22]. There were 78 cases of combination with diltiazem, 14 cases with fluconazole, and 8 cases with erythromycin, respectively, which are moderate CYP3A inhibitors [21, 22].

Table 2 shows the occurrence risk of adverse events in JADER and FAERS, and triazolam was reported as the suspected drug in 784 and 36,770 adverse events, respectively. Occurrence signals were detected in the elderly because of delirium, fall-fracture, and renal disorders. In particular, delirium exhibited a higher ROR for age 70 years or above compared with the other adverse events, which were 7.7 in JADER and 2.6 in FAERS. In addition, although JADER had a smaller number of cases and calculated ROR values tended to be higher compared with FAERS, the two regulatory databases demonstrated similar trends in terms of the categories of adverse events detected.

Table 2 Signals of adverse events of triazolam in patients

Table 3 lists the estimated parameters for the resulting PK/PD model. For the PK model, a one-compartment model including a first-order absorption process best representing the changes in blood concentration and age was incorporated into distribution volume and clearance, respectively, as a covariate. For the PD model, an anti-clockwise hysteresis loop was observed between blood concentration and drug efficacy; therefore, an effect compartment model was adopted to eliminate the time difference. Furthermore, a drug susceptibility against concentration in the effect compartment exhibited a positive linear correlation. The OFV values during the model building process were OFV 223.5 for the linear and OFV 225.2 for the Emax in the direct response model. Compared to the direct response model, OFV 193.7 in the effect compartment model showed a significantly lower OFV and, therefore, was determined as the more appropriate model. The parameters and structure of the PK/PD model had a low CV% of 1.05 − 21.8%, and the goodness-of-fit plots between values estimated from the model and extracted values indicated the appropriate model fitting and valid (Supplemental fig). The threshold for sedation and cognitive function deterioration were 0.44 and 0.58 ng/mL, with the resulting sensitivity values of 0.85 and 1.00 and specificity values of 0.90 and 1.00, respectively.

Table 3 Population pharmacokinetic/pharmacodynamic parameters of triazolam

Based on the results of a simulation, 5.95 h was required for blood concentration after a 0.25 mg dose of triazolam in healthy young people to fall below 0.44 ng/mL (Fig. 1). In contrast, when triazolam was administered to the elderly, 0.15 mg was required for blood concentration to fall below 0.44 ng/mL after 5.95 h. In addition, when 0.0625 mg (half of a 0.125 mg tablet) was administered to the elderly, the blood concentration was 0.44 ng/mL after 5.95 h, and a concomitant AUCR of 2.27 was observed (Fig. 2). The larger the AUCR of the drug, the more delayed the elimination. The half-life of triazolam with an AUCR of 1.0 was 2.9 h, whereas the half-life of itraconazole with an AUCR of 27.1 was 69.8 h.

Fig. 1
figure 1

Simulated Plasma Triazolam Concentration-time Profile in young and elderly patients. Dashed lines: 30 years old (dosage: 0.250 and 0.125 mg/body), solid lines: 69 years old (dosage: 0.250, 0.125, and 0.0625 mg/body). Dotted lines (horizontal axis): cut-off concentration (0.44 µg/mL), dotted lines (vertical axis): time required to reach cut-off concentration in young patients (5.95 h)

Fig. 2
figure 2

Plasma triazolam concentration, sedation, and cognitive function versus time profiles in elderly patients, when combined with CYP3A inhibitors. Cut-off concentration: 0.44 µg/mL, time required to reach cut-off concentration in young patients: 5.95 h. Dosage: 0.0625 mg/body (69 years old). Red line, AUCR = 27.1 (itraconazole); orange line, 5.26 (clarithromycin); green line, 3.38 (diltiazem); blue line (0.44 ng/mL after 5.95 h), 2.27; and purple line, 1. AUCR: area under the concentration-time curve ratio

Discussion

In this study, the appropriate use of triazolam in the elderly was evaluated using a modeling and simulation approach, supported by real-world data. Triazolam has been on the market for a long time, and more information regarding its efficacy and safety has been collected following its manufacturing and marketing, compared with the approval and application phase. The approach proposed in this study on prescription status, adverse event reporting, and dose adjustment using modeling and simulation proved particularly useful for such drugs.

Fall-related fractures in the elderly greatly impact their quality of life and pose a risk of inducing fatal diseases, such as bedriddenness and aspiration pneumonia; thus, prevention is particularly important [39]. As triazolam has a very short half-life of 2.9 h and the effect of carry-over to the next morning is considered small, it tends to be more commonly used on the elderly compared with long-acting benzodiazepines. However, it has also been reported that the risk of falls is the same regardless of half-life or the presence or absence of a benzodiazepine skeleton [40]. Thus, there is no consensus on the effects of factors such as half-life on falls. In the present study, the occurrence signals of fall-fracture after triazolam dosing were detected in both JADER and FAERS at 70 or 80 years and above. Therefore, the results supported that of previous reports, in which dosing of triazolam in the elderly above 70 years old poses a fall risk and the Guidance of Appropriate Medication for Elderly Patients notified in May 2018 [6] and Jun 2019 [7] in Japan.

We constructed a PK/PD model to determine the relationship between changes in blood concentration after triazolam dosing in the elderly and pharmacodynamic events such as sedation and cognitive function. Blood concentration and pharmacological events are highly correlated [18], and the transfer half-life from the blood to sites of action was also rapid at 6.9 min. Thus, using blood concentration to indicate pharmacological events is considered useful. Interestingly, pharmacological events rarely occur when the blood concentration falls below the threshold. If it falls below the threshold within the period from dosing to waking up, the occurrence risk of fall-fracture may be reduced. In the present study, the threshold was estimated to be 0.44 ng/mL. When 0.25 mg of triazolam was administered to healthy young people (30 years), it took 5.95 h to decrease below the threshold. In other words, the blood triazolam concentration falls below the threshold when an individual wakes up. In contrast, when 0.25 mg of triazolam was administered to the elderly, it took 7.80 h to reach a threshold and blood triazolam concentration was not sufficiently reduced at waking time, suggesting that the risk of adverse events resulting from carry-over is increased. Next, we estimated that a dose of 0.156-mg triazolam was required for the concentration to fall below the threshold in the elderly at the same time as in healthy young people (Fig. 1). In the package insert, a warning is included that states, “in the elderly, dosing should be started from a low dose.” However, the specific dose for the elderly is not mentioned. Based on the results of this study, we suggest that dosing at a half-dose (0.125 mg) significantly reduced the risk of carry-over.

Polypharmacy, aimed at treating comorbidities, is common in the elderly [21, 22], and changes in blood concentration tend to be affected by other pharmaceuticals. Itraconazole, a CYP3A inhibitor, and human immunodeficiency virus protease inhibitors are contraindicated for combined use, while diltiazem and clarithromycin are considered medications to be used with caution. [41], it is necessary to pay attention to potential drug-drug interactions when administering triazolam in clinical settings for drug treatment. In the present study, we simulated changes in blood triazolam concentration and pharmacological events in the elderly when used in combination with diltiazem, clarithromycin, and itraconazole, whose combined use was reported in JADER, which is the adverse event report from medical settings in Japan (Fig. 2). In addition, even though the dose was set at 0.0625 mg, which is 1/4 of the usual dose, blood concentration did not decrease sufficiently when combined with pharmaceuticals with an AUCR exceeding 2.27, even when administered at a low dose outside the approved dose range, which suggests the possibility of carry-over. Therefore, it is desirable to avoid using triazolam when administering medications with inhibition activity on CYP3A to the elderly and to consider alternative medications instead.

There were several limitations to our study. Firstly, the published adverse events and judgment on the involvement of pharmaceuticals are at the discretion of the reporters who judge the involvement of pharmaceuticals, and there is also missing information, such as age. Secondly, there were many confounding factors present in this study, such as concomitant drugs and dose and unavoidable selection bias. Thirdly, ROR was exploratorily used as a qualitative signal; thus, quantitative evaluations, such as occurrence frequency, are impossible. Finally, in the PK/PD model, the effect of age was assumed to be linear, and indications for ages other than 30 or 69 years, as described in previous reports, are unknown.

Conclusion

In this study, based on available information from medical settings in Japan, notable adverse events associated with triazolam dosing in the elderly (delirium, fall-fracture) were extracted. Constructing a PK/PD model using age as a covariate allowed for establishing a dosing strategy for elderly patients using concurrent medications. Triazolam is frequently prescribed for elderly patients, and the findings will help with the optimal dose.

Data availability

JADER, FAERS, NDB open data Japan, and DIDB data files can be downloaded from [https://www.pmda.go.jp/safety/info-services/drugs/adr-info/suspected-adr/0004.html (Accessed 2020-08-20, Japanese)], [https://www.fda.gov/drugs/development-approval-process-drugs/drug-approvals-and-databases (Accessed 2020-08-20]), [https://www.mhlw.go.jp/stf/seisakunitsuite/bunya/0000177182.html (Accessed 2020-08-20, Japanese)], and [https://www.druginteractionsolutions.org/ (Accessed 2020-08-20)].

Abbreviations

95% CI:

95% confidence interval

AUCR:

Area under the blood concentration-time curve increase ratio

CV:

Coefficient of variation

CYP:

Cytochrome P450

FAERS:

FDA Adverse Event Reporting System

JADER:

Japanese Adverse Drug Event Report database

NDB:

National Database of Health Insurance Claims and Specific Health Checkups of Japan

OFV:

Objective function value

PK/PD:

Pharmacokinetic-pharmacodynamic

ROR:

Reporting odds ratio

References

  1. Mangoni AA, Jackson SH. Age-related changes in pharmacokinetics and pharmacodynamics: basic principles and practical applications. Br J Clin Pharmacol. 2004;57(1):6–14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Tanigawara Y. Recent advances in pharmacokinetics in elderly. Nihon Ronen Igakkai Zasshi. 2003;40(2):109–19.

    Article  PubMed  Google Scholar 

  3. Schlender JF, Meyer M, Thelen K, Krauss M, Willmann S, Eissing T, et al. Development of a whole-body physiologically based Pharmacokinetic Approach to assess the pharmacokinetics of drugs in Elderly individuals. Clin Pharmacokinet. 2016;55(12):1573–89.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Gutiérrez-Valencia M, Izquierdo M, Malafarina V, Alonso-Renedo J, González-Glaría B, Larrayoz-Sola B, et al. Impact of hospitalization in an acute geriatric unit on polypharmacy and potentially inappropriate prescriptions: a retrospective study. Geriatr Gerontol Int. 2017;17(12):2354–60.

    Article  PubMed  Google Scholar 

  5. Kojima T, Akishita M, Kameyama Y, Yamaguchi K, Yamamoto H, Eto M, et al. High risk of adverse drug reactions in elderly patients taking six or more drugs: analysis of inpatient database. Geriatr Gerontol Int. 2012;12(4):761–2.

    Article  PubMed  Google Scholar 

  6. Ministry of Health, Labour and Welfare. Guidance of Appropriate Medication for Elderly Patients (General). 2018. https://www.pmda.go.jp/files/000232249.pdf. Accessed May 1 2022.

  7. Ministry of Health, Labour and Welfare. Guidance of Appropriate Medication for Elderly Patients [Particular (by Recuperation Environment)]. 2019. https://www.mhlw.go.jp/stf/newpage_05217.html. (only in Japanese) Accessed May 1 2022.

  8. Patel D, Steinberg J, Patel P. Insomnia in the Elderly: a review. J Clin Sleep Med. 2018;14(6):1017–24.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Bain KT. Management of chronic insomnia in elderly persons. Am J Geriatr Pharmacother. 2006;4(2):168–92.

    Article  PubMed  CAS  Google Scholar 

  10. Baker MI, Oleen MA. The use of benzodiazepine hypnotics in the elderly. Pharmacotherapy. 1988;8(4):241–7.

    Article  PubMed  CAS  Google Scholar 

  11. Shorr RI, Bauwens SF, Landefeld CS. Failure to limit quantities of benzodiazepine hypnotic drugs for outpatients: placing the elderly at risk. Am J Med. 1990;89(6):725–32.

    Article  PubMed  CAS  Google Scholar 

  12. Kojima T, Mizukami K, Tomita N, Arai H, Ohrui T, Eto M, et al. Screening Tool for older persons’ appropriate prescriptions for Japanese: report of the Japan Geriatrics Society Working Group on Guidelines for medical treatment and its safety in the elderly. Geriatr Gerontol Int. 2016;16(9):983–1001.

    Article  PubMed  Google Scholar 

  13. Zhong G, Wang Y, Zhang Y, Zhao Y. Association between Benzodiazepine Use and Dementia: a Meta-analysis. PLoS ONE. 2015;10(5):e0127836.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Glass J, Lanctôt KL, Herrmann N, Sproule BA, Busto UE. Sedative hypnotics in older people with insomnia: meta-analysis of risks and benefits. BMJ. 2005;331(7526):1169.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Greenblatt DJ, Divoll M, Abernethy DR, Moschitto LJ, Smith RB, Shader RI. Reduced clearance of triazolam in old age: relation to antipyrine oxidizing capacity. Br J Clin Pharmacol. 1983;15(3):303–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Klotz U, Avant GR, Hoyumpa A, Schenker S, Wilkinson GR. The effects of age and liver disease on the disposition and elimination of diazepam in adult man. J Clin Invest. 1975;55(2):347–59.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Greenblatt DJ, Harmatz JS, von Moltke LL, Wright CE, Shader RI. Age and gender effects on the pharmacokinetics and pharmacodynamics of triazolam, a cytochrome P450 3A substrate. Clin Pharmacol Ther. 2004;76(5):467–79.

    Article  PubMed  CAS  Google Scholar 

  18. Greenblatt DJ, Harmatz JS, Shapiro L, Engelhardt N, Gouthro TA, Shader RI. Sensitivity to triazolam in the elderly. N Engl J Med. 1991;324(24):1691–8.

    Article  PubMed  CAS  Google Scholar 

  19. Bihan K, Lebrun-Vignes B, Funck-Brentano C, Salem JE. Uses of pharmacovigilance databases: an overview. Therapie. 2020;75(6):591–8.

    Article  PubMed  Google Scholar 

  20. Kuemmel C, Yang Y, Zhang X, Florian J, Zhu H, Tegenge M, et al. Consideration of a credibility Assessment Framework in Model-Informed Drug Development: potential application to physiologically-based pharmacokinetic modeling and Simulation. CPT Pharmacometrics Syst Pharmacol. 2020;9(1):21–8.

    Article  PubMed  CAS  Google Scholar 

  21. 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.

    Article  PubMed  CAS  Google Scholar 

  22. Guideline on drug interaction for drug development and appropriate provision of information. https://www.pmda.go.jp/files/000228122.pdf. Accessed May 1 2022.

  23. HALCION (TRIAZOLAM). Label - FDA (https://www.accessdata.fda.gov/, Accessed 2021-10-22).

  24. Schou JS. Triazolam: adverse effects in relation to dosage. Pharmacol Toxicol. 1989;64(1):6–8.

    Article  PubMed  CAS  Google Scholar 

  25. Kanda Y. Investigation of the freely available easy-to-use software ‘EZR’ for medical statistics. Bone Marrow Transpl. 2013;48(3):452–8.

    Article  CAS  Google Scholar 

  26. Barbhaiya RH, Shukla UA, Kroboth PD, Greene DS. Coadministration of nefazodone and benzodiazepines: II. A pharmacokinetic interaction study with triazolam. J Clin Psychopharmacol. 1995;15(5):320–6.

    Article  PubMed  CAS  Google Scholar 

  27. Greenblatt DJ, von Moltke LL, Harmatz JS, Counihan M, Graf JA, Durol AL, et al. Inhibition of triazolam clearance by macrolide antimicrobial agents: in vitro correlates and dynamic consequences. Clin Pharmacol Ther. 1998;64(3):278–85.

    Article  PubMed  CAS  Google Scholar 

  28. Warot D, Bergougnan L, Lamiable D, Berlin I, Bensimon G, Danjou P, et al. Troleandomycin-triazolam interaction in healthy volunteers: pharmacokinetic and psychometric evaluation. Eur J Clin Pharmacol. 1987;32(4):389–93.

    Article  PubMed  CAS  Google Scholar 

  29. Ochs HR, Greenblatt DJ, Knüchel M. Differential effect of isoniazid on triazolam oxidation and oxazepam conjugation. Br J Clin Pharmacol. 1983;16(6):743–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Varhe A, Olkkola KT, Neuvonen PJ. Oral triazolam is potentially hazardous to patients receiving systemic antimycotics ketoconazole or itraconazole. Clin Pharmacol Ther. 1994;56(6 Pt 1):601–7.

    Article  PubMed  CAS  Google Scholar 

  31. Varhe A, Olkkola KT, Neuvonen PJ. Effect of fluconazole dose on the extent of fluconazole-triazolam interaction. Br J Clin Pharmacol. 1996;42(4):465–70.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Culm-Merdek KE, von Moltke LL, Gan L, Horan KA, Reynolds R, Harmatz JS, et al. Effect of extended exposure to grapefruit juice on cytochrome P450 3A activity in humans: comparison with ritonavir. Clin Pharmacol Ther. 2006;79(3):243–54.

    Article  PubMed  CAS  Google Scholar 

  33. Pourbaix S, Desager JP, Hulhoven R, Smith RB, Harvengt C. Pharmacokinetic consequences of long term coadministration of cimetidine and triazolobenzodiazepines, alprazolam and triazolam, in healthy subjects. Int J Clin Pharmacol Ther Toxicol. 1985;23(8):447–51.

    PubMed  CAS  Google Scholar 

  34. O’Connor-Semmes RL, Kersey K, Williams DH, Lam R, Koch KM. Effect of ranitidine on the pharmacokinetics of triazolam and alpha-hydroxytriazolam in both young (19–60 years) and older (61–78 years) people. Clin Pharmacol Ther. 2001;70(2):126–31.

    Article  PubMed  Google Scholar 

  35. Backman JT, Wang JS, Wen X, Kivistö KT, Neuvonen PJ. Mibefradil but not isradipine substantially elevates the plasma concentrations of the CYP3A4 substrate triazolam. Clin Pharmacol Ther. 1999;66(4):401–7.

    Article  PubMed  CAS  Google Scholar 

  36. Varhe A, Olkkola KT, Neuvonen PJ. Diltiazem enhances the effects of triazolam by inhibiting its metabolism. Clin Pharmacol Ther. 1996;59(4):369–75.

    Article  PubMed  CAS  Google Scholar 

  37. Lilja JJ, Kivistö KT, Backman JT, Neuvonen PJ. Effect of grapefruit juice dose on grapefruit juice-triazolam interaction: repeated consumption prolongs triazolam half-life. Eur J Clin Pharmacol. 2000;56(5):411–5.

    Article  PubMed  CAS  Google Scholar 

  38. Stoehr GP, Kroboth PD, Juhl RP, Wender DB, Phillips JP, Smith RB. Effect of oral contraceptives on triazolam, temazepam, alprazolam, and lorazepam kinetics. Clin Pharmacol Ther. 1984;36(5):683–90.

    Article  PubMed  CAS  Google Scholar 

  39. Kannus P, Sievänen H, Palvanen M, Järvinen T, Parkkari J. Prevention of falls and consequent injuries in elderly people. Lancet. 2005;366(9500):1885–93.

    Article  PubMed  Google Scholar 

  40. Masudo C, Ogawa Y, Yamashita N, Mihara K. Association between Elimination Half-life of benzodiazepines and falls in the Elderly: a Meta-analysis of Observational studies. Yakugaku Zasshi. 2019;139(1):113–22.

    Article  PubMed  CAS  Google Scholar 

  41. Wienkers LC, Heath TG. Predicting in vivo drug interactions from in vitro drug discovery data. Nat Rev Drug Discov. 2005;4(10):825–33.

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

This study was supported by a Japan Society for the Promotion of Science (JSPS) Grant-in-Aid for Young Scientists, Grant Number 19K20736 (AO). The authors thank Ms. Natsuko Takano from Musashino University for assisting in the collection of some data for this study.

Funding

This study was supported by a Japan Society for the Promotion of Science (JSPS) Grant-in-Aid for Young Scientists, Grant Number 19K20736 (AO).

Author information

Authors and Affiliations

Authors

Contributions

AO and NN contributed equally to the conception, design of the research, and acquisition of the data; AO and SS contributed to the analysis of the data. All the authors critically revised the manuscript, and read and approved the final manuscript, and agree to be fully accountable for ensuring the integrity and accuracy of the work.

Corresponding author

Correspondence to Akira Okada.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, 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 you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. 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-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Okada, A., Sera, S. & Nagai, N. Appropriate use of triazolam in elderly patients considering a quantitative benefit-risk assessment based on the pharmacokinetic-pharmacodynamic modeling and simulation approach supported by real-world data. BMC Pharmacol Toxicol 25, 60 (2024). https://doi.org/10.1186/s40360-024-00777-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40360-024-00777-z

Keywords