Zum Hauptinhalt springen

Virtual twins for model-informed precision dosing of clozapine in patients with treatment-resistant schizophrenia.

Mostafa, S ; Rafizadeh, R ; et al.
In: CPT: pharmacometrics & systems pharmacology, Jg. 13 (2024-03-01), Heft 3, S. 424-436
Online academicJournal

Virtual twins for model‐informed precision dosing of clozapine in patients with treatment‐resistant schizophrenia  Study highlights

Model‐informed precision dosing using virtual twins (MIPD‐VTs) is an emerging strategy to predict target drug concentrations in clinical practice. Using a high virtualization MIPD‐VT approach (Simcyp version 21), we predicted the steady‐state clozapine concentration and clozapine dosage range to achieve a target concentration of 350 to 600 ng/mL in hospitalized patients with treatment‐resistant schizophrenia (N = 11). We confirmed that high virtualization MIPD‐VT can reasonably predict clozapine concentrations in individual patients with a coefficient of determination (R2) ranging between 0.29 and 0.60. Importantly, our approach predicted the final dosage range to achieve the desired target clozapine concentrations in 73% of patients. In two thirds of patients treated with fluvoxamine augmentation, steady‐state clozapine concentrations were overpredicted two to four‐fold. This work supports the application of a high virtualization MIPD‐VT approach to inform the titration of clozapine doses in clinical practice. However, refinement is required to improve the prediction of pharmacokinetic drug–drug interactions, particularly with fluvoxamine augmentation.

• WHAT IS THE CURRENT KNOWLEDGE ON THE TOPIC?

Clozapine pharmacokinetics are influenced by various genetic and environmental factors including drug interactions (CYP1A2 inhibitors or inducers, including smoking), obesity, inflammation, geriatric age, and pregnancy.

• WHAT QUESTION DID THIS STUDY ADDRESS?

Can model‐informed precision dosing using virtual twins (MIPD‐VTs) reliably identify the clozapine dosage range to achieve target concentrations of 350–600 ng/mL?

• WHAT DOES THIS STUDY ADD TO OUR KNOWLEDGE?

The study verified that the MIPD‐VT can reasonably predict clozapine plasma concentrations in patients with treatment‐resistant schizophrenia and has the potential to be utilized in identifying the required clozapine dose range to achieve therapeutic concentrations.

• HOW MIGHT THIS CHANGE DRUG DISCOVERY, DEVELOPMENT, AND/OR THERAPEUTICS?

The use of MIPD‐VT may be utilized to avoid toxicity during clinical optimization of narrow therapeutic index drugs within inpatient and outpatient settings.

INTRODUCTION

Not all patients respond to the same drug equally. Variability in drug response results from differences between patients in demographic, genetic, and/or environmental factors.[[1], [3], [5]] The combined effect of these factors can significantly impact drug concentrations (pharmacokinetics [PKs]), resulting in adverse drug reactions (ADRs) or nonresponse to treatment. Model‐informed precision dosing (MIPD), in which computer modeling and simulation combines patient‐specific factors to optimize dose recommendations, is an attractive solution to achieve target drug concentrations in clinical practice. This is especially important for drugs with a well‐defined concentration‐response relationship and a narrow therapeutic index that carry an increased risk of nonresponse or life‐threatening adverse effects if drug concentrations occur outside the therapeutic range. In recent years, physiologically‐based PK (PBPK) approaches have been used to construct virtual twins (VTs) of real patients for MIPD.[[3], [7], [9]] Clozapine, the most effective antipsychotic medication in treatment‐resistant schizophrenia, is suggested to have the narrowest therapeutic index among second‐generation antipsychotic medications, and has potentially life‐threatening adverse effects, such as myocarditis, agranulocytosis, and seizures. Furthermore, its metabolism is influenced by various genetic and environmental factors, including drug interactions (CYP1A2 inhibitors or inducers, including smoking), obesity, inflammation (C‐reactive protein [CRP] elevations), geriatric age, and pregnancy.[10] Consequently, it is an ideal candidate to evaluate the potential of the MIPD‐VT approach.

In our previous study,[11] we built VTs of 42 clozapine‐treated patients using Simcyp and evaluated the impact of systematically increasing the virtualization of covariates on predicted clozapine PKs. Three evaluation scenarios were simulated: low (demographic), medium (demographic and environmental), and high (demographic, environmental, and genetic) covariate virtualization. The high covariate virtualization model provided the best prediction of the observed clozapine plasma concentrations, supporting the idea that high‐level covariate virtualization is required for clinical utilization of the MIPD‐VT approach.

In the present study, the high covariate virtualization approach is applied to a novel data set of patients with treatment‐resistant schizophrenia who had received clozapine therapy and had repeated clozapine plasma concentrations. Unlike the previous study, there was high accuracy of the timing of clozapine dosing and blood samples for clozapine concentration quantification (due to these patients being hospitalized). This new data set was used to evaluate: (i) the performance of MIPD‐VT in predicting steady‐state plasma concentrations at multiple timepoints, (ii) whether MIPD‐VT can identify lower and upper dose ranges for clozapine to achieve target concentrations (350 to 600 ng/mL) in each patient,[12] and (iii) the performance of MIPD‐VT in predicting plasma concentrations with fluvoxamine augmentation.

METHODS

Participants

Eleven patients diagnosed with treatment‐resistant schizophrenia who were enrolled in the Metabolic and Genetic Explorations in Refractory Schizophrenia (MAGERS) study were selected to construct VTs in Simcyp. Briefly, MAGERS study patients were recruited and genotyped during their hospitalizations at the British Columbia Psychosis Program (BCPP), a 25‐bed tertiary inpatient resource for treatment‐resistant psychosis in the Canadian province of British Columbia. Adult patients (aged 18–65) can only be admitted to the BCPP after failing outpatient trials with two or more antipsychotics, and typically, multiple inpatient hospitalizations in general and/or regional psychosis‐specialized psychiatric units. To be eligible for the MAGERS study, BCPP inpatients had to meet Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition criteria for schizophrenia or schizoaffective disorder (with or without a history of catatonia), or catatonia associated with another mental disorder. Caffeine intake was monitored and controlled in the study patients, who consumed less than three to four cups of coffee per day. This intake of caffeine is not considered to cause clinically significant inhibition of clozapine metabolism. Informed consent, or assent plus surrogate consent, was obtained by a research assistant. The study was conducted under approval of the University of British Columbia Clinical Research Ethics Board (study H14‐02657).

Blood samples were collected at least 6 days after any clozapine or fluvoxamine/bupropion dose adjustments and ~12 h after the last clozapine dose to measure clozapine plasma concentrations.

Quantification of clozapine plasma concentrations

Clozapine and N‐desmethylclozapine concentrations were quantified by an accredited clinical laboratory (British Columbia Provincial Toxicology Centre) by liquid chromatography–tandem spectrometry, using a commercial assay kit (Chromsystems, Munchen, Germany). As per the manufacturer's instructions, samples were extracted by combining 0.05 mL of patient serum with 0.025 mL of Chromsystems extraction buffer (#92005) and 0.25 mL of internal standard (ISTD) working solution. The ISTD working solution is a mix of 4 mL of internal standard mix—Chromsystems product (#92046/AN1/XT) MassTox antidepressants 1/extended and neuroleptics 1/extended internal standard set (ISTD 5 for clozapine and ISTD 22 for N‐desmethylclozapine)—and 50 mL of Chromsystems Precipitation Reagent (#92003). The high‐performance liquid chromatography column used is the Chromsystems MassTox TDM MasterColumn Series A (#92110), and the mobile phase consists of Chromsystems MassTox TDM series A Mobile Phase 1 (#92001) and Chromsystems MassTox TDM series A Mobile phase II (#92002). The samples were measured using the mass spectrometry analyzer Agilent QQQ 6470, operated in the multiple reaction monitoring (MRM) positive ionization mode. The MRMs are clozapine MRM1 327>270, clozapine MRM2 327>192, N‐desmethylclozapine MRM1 313>192, N‐desmethylclozapine MRM2 313>270, ISTD5 (clozapine ISTD) MRM1 335>275, and ISTD22 (N‐desmethylclozapine ISTD) MRM1 321>192. The limits of sensitivity are as follows: clozapine analytical measuring range 1 to 3600 nmol/L, and N‐desmethylclozapine analytical measuring range 1 to 1600 nmol/L. The precision is as follows: clozapine quality control level 1 (QC1) concentration was 964.9 nmol/L and within run standard deviation/coefficient of variation (SD/CV) 7.4/0.8 (total SD/CV 10.5/1.1), clozapine QC2 concentration was 1649.1 nmol/L and within run SD/CV 15.7/1.0 (total SD/CV 45.11/2.7), N‐desmethylclozapine QC1 concentration was 528.4 nmol/L and within run SD/CV 7.1/1.4 (total SD/CV 9.1/1.2), N‐desmethylclozapine QC2 concentration was 739.5 nmol/L and within run SD/CV 9.1/1.2 (total SD/CV 21.7/2.9), clozapine QC1 between day SD/CV 7.8/0.8, clozapine QC2 between day SD/CV 42.2/2.6, N‐desmethylclozapine QC1 between day SD/CV was not performed, and N‐desmethylclozapine QC2 between day SD/CV 16.7/2.7.

Whole genome sequencing

High molecular weight genomic DNA was extracted in the Fraser Health Molecular Cytogenetics Laboratory at Royal Columbian Hospital from whole blood collected in BD Vacutainer EDTA tubes. After rapid thawing, micro‐fluidic partitioned DNA libraries for each sample were created in Canada's Michael Smith Genome Sciences Centre at BC Cancer, using the Chromium system from 10× Genomics (10× Genomics). Gel beads‐in‐Emulsion (GEMs) were produced by combining DNA, Master Mix, and partitioning oil in the 10× Genomics Chromium Controller instrument with the micro‐fluidic Genome Chip (PN‐120216; 10× Genomics). The DNA in each GEM then underwent isothermic amplification with barcoding of each fragment. Barcoded fragments then underwent Illumina library construction (as described in the Chromium Genome Reagent Kits Version 2 User Guide, PN‐120229). Each resulting library was then assessed for quality using an Agilent 2100 Bioanalyzer and a DNA 1000 assay. Libraries were then sequenced at 40× average coverage (effective coverage was ~32× due to the overhead of the DNA "bar codes" tagging the gDNA reads, which were removed in software) in 150 base paired end tag lanes on an Illumina HiSeqX sequencer.[13] De‐identified FASTQ files were transferred over a secure link to the Pavlidis laboratory in UBC's Michael Smith Laboratories. Alignment of linked reads produced by whole‐genome sequencing (WGS) to the hg19 reference genome was performed using the 10× Genomics Long Ranger (version 2.2.2) pipeline.[14] Sequence variants were called with GATK (version 4.0.3.0), and filtered for quality using settings recommended by the GATK community.

Pharmacogenomic analysis and reporting

Variant Call Format (VCF) and Binary Alignment Map (BAM) files were analyzed, utilizing Stargazer version 1.08[15] to call diplotypes for CYP2D6, CYP2C9, CYP2C19, CYP3A4, and CYP3A5. Stargazer uses WGS data to identify phased haplotypes comprised of single‐nucleotide polymorphisms, small indels, and large structural variants, and then maps these haplotypes to star alleles based on translation tables maintained by the Pharmacogene Variation Consortium. Genotype to phenotype translation followed procedures developed by the Pharmacogene Variation Consortium[16] and the Clinical Pharmacogenetics Implementation Consortium (CPIC).[17]

Simulation and comparison workflow

Simulations were performed blinded to the actual observed clozapine concentrations. Once the VT simulations were completed for all patients, the predicted clozapine concentrations and the clozapine dosage range to achieve the target clozapine concentrations (350–600 ng/mL) were matched to the observed clozapine concentrations and the final clinical dose of each patient. This combined data set was used to test the high covariate virtualization model in terms of its ability to (i) predict each steady‐state clozapine concentration versus the observed clozapine concentration and (ii) the predicted dosage range to achieve the nominal therapeutic range of 350 to 600 ng/mL for each patient.

Individual VT predictions

All the VT predictions of clozapine plasma concentration were performed using the high covariate virtualization model described in our previous study.[11]

Construction of VTs

Individual VTs were constructed in Simcyp version 21, as previously described by Mostafa et al.[11] In short, each participant was matched for demographics, clozapine dose, CYP phenotypes, and the presence of inhibitors and inducers (see Table 1). Fluvoxamine and bupropion were the only inhibitors with validated inhibitor drug profiles in Simcyp and were accounted for when co‐prescribed with clozapine. The Simcyp population of North European Whites (Sim‐NEurCaucasian) was used for participants with a body mass index (BMI) of less than 30 and assigned as having White, Mixed White, or Middle Eastern Ancestry, as per demographic data in Table 1. The obese population in Simcyp was used for participants with a BMI between 30 and 40, whereas the morbidly obese population was used for those with a BMI greater than 40, irrespective of ancestry. The Simcyp population of Sim‐Chinese healthy volunteers was used for one participant who was assigned as having East Asian ancestry with a BMI less than 30.

1 TABLE Patient characteristics in the clinical study.

Study IDAge (years)AncestrySexClozapine dose (mg)Smoking (C/D)CYP1A2CYP2D6CYP2C19CYP2C9CYP3A4CYP3A5Ht (cm)Wt (kg)BMIConcomitant Inh/ind
132NAEMA: 150 @ 12 p.m. & 5 p.m. & 350 @ 9 p.m.B: 150 @ 12 p.m. & 350 @ 9 p.m.C: 500 @ 9 p.m.D & E: 50 @ 5 p.m. & 500 @ 9 p.m.10*1F/*1F*41/*68 + *4*1/*1*1/*1*1/*1*1/*317310635
232EAMA: 50 @ 9 p.m.B: 100 @ 9 p.m.C: 150 @ 9 p.m.UNK*1A/*1F*10/*36 + *10*2/*2*1/*1*1/*1x2*3/*31706522
332NAEMA: 350 @ 9 p.m.B: 200 @ 9 p.m.C: 200 @ 9 p.m.NS*1F/*1F*1/*4*1/*1*1/*2*1/*1*3/*317110531B: FLV 37.5 mg/dayC: FLV 50 mg/day
436MCMA: 100 @ 08 a.m. & 300 @ 9 p.m.B: 50 @ 08 a.m. & 150 @5 p.m.C: 450 @ 5 p.m.8–9*1A/*1F*13C/*21*1/*1*1/*1*1/*1*3/*31676423B: FLV 25 mg/dayC: FLV 50 mg/day
521EAMA: 75 @ 12 p.m. & 125 @ 9 p.m.B, C & D: 75 @ 12 p.m. & 175 @ 9 p.m.UNK*1A/*1F*1/*10*1/*1*1/*3*1/*1*3/*31727525
632NAEMA & B: 50 @ 12 p.m. & 300 @ 9 p.m.NS*1F/*1F*4/*13C*1/*2*1/*1*1/*1*3/*318311233
721MEMA: 100 @12 p.m. & 200 @ 9 p.m.B & C: 150 @ 12 p.m. & 250 @ 9 p.m.NS*1A/*1F*13B/*21x2*1/*1*1/*1*1B/*1B*1/*618911131
843NAEFA: 100 @ 12 p.m. & 200 @ 9 p.m.B: 75 @ 6 p.m.C: 150 @ 6 p.m.NS*1A/*1F*1/*35*1/*17*1/*1*1/*1*3/*31678029A: BPR 450 mg/dayB: FLV 25 mg/dayC: FLV 50 mg/day
935NAEMA: 150 @ 08 a.m. & 350 @ 5 p.m.2*1F/*1F*1/*68 + *4*1/*1*1/*2*1/*22*3/*31718027
1058NAEMA: 325 @ 9 p.m.B & C: 50 @ 12 p.m. & 300 @ 9 p.m.1–2*1A/*1F*2/*21*1/*1*1/*1*1/*1*3/*31686724
1135NAEMA: 100 @ 12 p.m. & 400 @ 9 p.m.NS*1A/*1F*4/*4*17/*17*1/*1*1/*1*3/*31807924

1 Abbreviations: A, fidosing period A; B, dosing period B; BPR, bupropion; C, dosing period C; C/D, cigarettes per day; D, dosing period D; E, dosing period E; EA, East Asian; F, female; FLV, fluvoxamine; Ht, height; Ind, inducer; Inh, inhibitor; M, male; MC, mixed Caucasian; ME, Middle Eastern; NAE, North American/European; NS, nonsmoker; UNK, unknown quantity; Wt, weight.

  • 2 a No study participants had elevated CRP around the time of blood sample collection for clozapine measurement. CRP elevations have been associated with CYP1A2 phenoconversion.
  • 3 b Participants with unknown smoking quantity were assumed to smoke one to five cigarettes/day for simulations.

The Simcyp validated drug profile for clozapine was used. Participants with known CYP1A2 genotypes (CYP1A2*1A/*1A, *1A/*1F, or *1F/*1F) were selected to assess the importance of customizing the CYP1A2 enzyme abundance based on the inducible CYP1A2*1F/*1F genotype. Participants carrying CYP1A2 alleles with unknown function (e.g., *1L or *1V) were excluded. Smokers with a CYP1A2*1F/*1F genotype were assigned a CYP1A2 enzyme abundance based on the number of cigarettes smoked per day, as described previously by Plowchalk et al.[18] The enzyme abundance values were adjusted using the following groupings of cigarette consumption: 1‐5 cigarettes/day (64 pmol/mg protein), 6‐10 cigarettes/day (79 pmol/mg protein), 11–20 cigarettes/day (90 pmol/mg protein), and greater than 20 cigarettes/day (94 pmol/mg protein). All other participants were assigned the nonsmoker CYP1A2 enzyme abundance of 52 pmol/mg microsomal protein. Smokers with the genotype CYP1A2*1A/*1F were additionally simulated using enzyme abundance values based on the number of cigarettes smoked given the potential inducibility by smoking as per available literature.[19] In two participants, the quantity of cigarettes smoked per day was unknown and thus a conservative quantity of 1‐5 cigarettes/day was assumed. The inbuilt Simcyp enzyme abundance data for the corresponding CYP2D6, CYP2C19, CYP2C9, CYP3A4, and CYP3A5 enzyme phenotypes were applied without alteration. One participant with a CYP3A4*1/*1x2 genotype was treated as a normal metabolizer (NM) in Simcyp, as little is known about CYP3A4*1 allele duplication and its effect on CYP3A4 phenotype. Carriers of the CYP3A5*1/*3 genotype were treated as NMs, as CYP3A5 is thought to play a minor role in the metabolism of clozapine and the corresponding enzyme abundance for the intermediate metabolizer phenotype is not available.

Estimation of target clozapine dosage range to achieve clozapine therapeutic drug monitoring...

The high covariate virtualization model was also used to perform VT simulation patients to predict the lowest clozapine dose needed to reach a clozapine plasma concentration greater than or equal to 350 ng/mL and the maximum dose to reach a plasma clozapine concentration less than or equal to 600 ng/mL. Initially, each VT was simulated using a standard clozapine dose titration schedule (see Table 2) to the maximum dose of 200 mg per day. To ensure the predicted clozapine concentrations were always at steady‐state, the predicted clozapine plasma concentration was simulated at least 6 days after the last dosage adjustment (i.e., day 20). If the VT had not achieved a steady‐state concentration of greater than or equal to 350 ng/mL by the end of this dose titration period, further dose increases in increments of 25–50 mg were added to the dosing schedule. Once the minimum dose required to achieve the lower target clozapine concentration (350 ng/mL) was identified, the clozapine titration schedule was then further adjusted with additional dose titrations (25–50 mg day) to attain the target dose capable of achieving the upper clozapine concentration of 600 ng/mL. For Study IDs 3, 4, and 8, the last dose prior to the introduction of fluvoxamine was used for this analysis, as fluvoxamine augmentation is expected to result in CYP1A2 phenoconversion, reducing clozapine clearance and dose requirements.

2 TABLE Initial standard clozapine dosage titration schedule used in the initial simulation.

Dose numberDayTimeDose time (h)Dose (mg)
119:00012.5
229:002425
339:004825
449:007225
5421:008425
659:009625
7521:0010825
869:0012025
9621:0013250
1079:0014425
11721:0015675
1289:0016825
13821:00180100
1499:0019250
15921:00204100
16109:0021650
171021:00228100
18119:0024050
191121:00252125
20129:0026450
211221:00276125
22139:0028850
231321:00300125
24149:0031250
251421:00324150
26159:0033650
271521:00348150
28169:0036050
291621:00372150
30179:0038450
311721:00396150
32189:0040850
331821:00420150
34199:0043250
351921:00444150
36209:0045650
372021:00468150

Interpretation of data comparisons – population versus individual

We have undertaken this comparison in two steps: (i) a whole population approach for the first observation only and (ii) at an individual level (i.e., a comparison between observed vs. predicted across repeated measures). The former provides a direct comparison against our previous paper,[11] whereas the latter allows better visualization and interpretation of our MIPD‐VT approach to the individual patient, which is consistent with the proposed approaches undertaken in precision medicine.

Statistical analysis

Linear regression was used to compare the first measured clozapine plasma concentration against the first predicted clozapine concentration from their VT. Given MIPD‐VT is an individual optimization approach rather than a population optimization, we have opted to display each patient individually to show their trajectory (repeated measures) with no regression approaches. GraphPad Prism (version 9.0.1 (151) for Windows; GraphPad Software, www.graph pad.com) was used to perform descriptive analysis and produce the simple linear regression graph. No statistical approach was undertaken to compare the VT dose range to achieve the nominal therapeutic range for clozapine against the final dose, as this was simple count data only. The metric for success was if the final observed clozapine dose was within the predicted lower and upper dosage range for their VT.

RESULTS

VT predictions

Figure 1a shows the predicted versus the first observed trough (dosing period A) clozapine concentrations for the full cohort of 11 patients using the high covariate virtualization model. None of the patients were receiving fluvoxamine during dosing period A. Similar to our previous work, the linear regression analysis confirmed a good relationship between the observed versus predicted plasma clozapine trough concentrations with a correlation of determination (R2) value of 0.29. When the VTs of the four smokers with CYP1A2*1A/*1F genotypes were re‐assigned to a high rather than low CYP1A2 enzyme abundance, to account for potential inducibility, predictions were visually and statistically improved, that is, R2 value increased from 0.29 to 0.60 (see Figure S1).

psp413093-fig-0001.jpg

During the VT building process, we observed that two out of three patients who received fluvoxamine augmentation had clozapine plasma concentrations that were poorly predicted. Indeed, the clozapine concentrations were predicted to be two to four times higher than the observed clozapine concentrations. This observation was consistent on all consecutive clozapine blood concentrations taken (Figure 1b) after the co‐administration of fluvoxamine.

Individual patient VT predictions

Figure 2 shows the predicted versus observed clozapine concentrations per patient with one to four repeated concentrations. The MIPD‐VT approach appears to predict the overall trajectory of the observed plasma clozapine concentrations and was consistent with any prescribed dose changes made by the medical team. For example, for Study ID 1, the first clozapine concentration (see

GRAPH

concentration point) was well above the nominal therapeutic range at 871 ng/mL. Following subsequent dosage reductions, the observed and predicted clozapine concentrations tracked back into the desired therapeutic range (see

GRAPH

,

GRAPH

, and

GRAPH

concentration points).

psp413093-fig-0002.jpg

Predicted concentrations for Study IDs 3, 4, and 8 are grouped together at the bottom of Figure 2, as these were the patients who were augmented with fluvoxamine after an initial clozapine concentration taken on clozapine alone. For Study IDs 3 and 4, the predicted clozapine concentration in the presence of fluvoxamine were consistently overpredicted by a two to four‐fold increase (see

GRAPH

and

GRAPH

concentration points). In contrast, Study ID 8 predictions were more accurate. A full concentration‐time curve of the simulated clozapine plasma concentrations with and without fluvoxamine interactions is illustrated for Study IDs 4 and 8 in Figure 3a,b. When the clozapine and fluvoxamine doses were relatively low (150 mg or less for clozapine and 50 mg or less for fluvoxamine), MIPD‐VT appears to describe the observed concentrations well (see Table S1). At higher doses of clozapine, our MIPD‐VT significantly overpredicted the extent of the PK drug interaction (i.e., 2–4‐fold higher predicted clozapine concentrations).

psp413093-fig-0003.jpg

Estimation of a clozapine dosage range to reach a target plasma concentration between 350 and...

Table 3 illustrates the clozapine dose ranges predicted by the high covariate virtualization model to get into the suggested therapeutic range of 350–600 ng/mL for efficacy. The simulation predicted dosage range captured the actual clinical dose utilized by the medical team in 8 out of 11 patients (73%). Although the dose taken by patient 5 was captured in the predicted range, the observed clozapine plasma concentration was 137.91 ng/mL, which is well below 350 ng/mL. It was noted that the patient was clinically stable at this measured concentration, thus further titration was not undertaken by the medical team. Thus, highlighting that perhaps a target concentration intervention approach rather than a TDM approach may be possible with clozapine dose optimization even in this treatment‐resistant schizophrenia population.

3 TABLE Estimation of the clozapine dosage range to reach a target plasma concentration between 350 and 600 ng/mL.

Study IDLowest dose (mg)to reach ≥350 ng/mLVT ‐ Predicted clozapine concentration (ng/mL)Highest dose (mg) to reach ≤600 ng/mLVT‐ Predicted clozapine concentration (ng/mL)Individual Target dose range (mg)Last clozapine dose (mg) without fluvoxamineObserved clozapine concentration at last dose (ng/mL)Is dose within predicted range?Predicted average clozapine dose (mg)Deviation from last clozapine dose (%)
1425356.1725590.4425–725550669.0Yes575+4.5
2225383.5325593.8225–325150166.0No275+83.3
3350358.1575582.6350–575350610.1Yes462.5+32.1
4350356.4575579.1350–575400253.6Yes462.5+15.6
5250392.1350582.7250–350250137.9Yes300+20.0
6375355.5625587.4375–625350594.4No500+42.9
7400370.0650595.3400–650400514.1Yes525+31.3
8400374.0625575.1400–625300453.6No512.5+70.8
9450368.1725587.5450–725500513.4Yes587.5+17.5
10275352.0425586.5275–425350443.5Yes3500.0
11450386.5725597.7450–725500518.0Yes587.5+17.5

  • 4 Abbreviation: VT, virtual twin.
  • 5 a Target dose range to achieve a lower concentration of ≥350 ng/mL and an upper concentration of ≤600 ng/mL.
  • 6 b The observed plasma concentration in these two patients were slightly above the recommended range of 600 ng/mL.
  • 7 c The observed plasma concentration in these two patients were below the desired therapeutic range. Note both patients were of East Asian ancestry.
  • 8 d This patient was taking bupropion conomitantly, which is a strong CYP2D6 inhibitor. VT simulations were performed with the addition of bupropion as an inhibitor drug, however, this did not make a significant impact on the predicted clozapine plasma concentration.
DISCUSSION

The performance of high virtualization MIPD‐VT was evaluated by predicting clozapine concentrations from a cohort of hospitalized patients with treatment‐resistant schizophrenia. These results confirm our previous work demonstrating that MIPD‐VT can reasonably predict clozapine steady‐state concentrations in real patients (R2 = 0.29 compared with R2 = 0.37 previously).[11] A particular strength of the study was that all individual simulations were undertaken without knowledge of the final clinical outcome and concentration (blinded).

There are several noteworthy findings of this study. First, predictions were greatly improved when the four smokers with CYP1A2*1A/*1F genotypes were assigned the induced CYP1A2 enzyme abundance in Simcyp (from R2 = 0.29 to R2 = 0.60). There is poor consensus in the literature regarding the influence of CYP1A2*1F on the inducibility of the CYP1A2 enzyme in smokers. However, our finding suggests that increased CYP1A2 abundance may not be specific to homozygous CYP1A2*1F, and that those who are heterozygous for this allele may similarly be susceptible to smoking induction.[19] Second, there was significant overprediction of clozapine concentrations in two of the three cases with fluvoxamine augmentation. Both patients were on a fluvoxamine dose of 50 mg/day and clozapine doses of 200 mg/day (Study ID 3) and 450 mg/day (Study ID 4), suggesting that extrapolation of inhibitory fluvoxamine concentrations at hepatic CYP1A2 may be overestimated in simulations with increasing clozapine dose. This highlights the need for further validation of inhibitor and inducer compound files within PBPK software to cater for phenoconversion, including weak and moderate inhibitors and inducers of CYP enzymes.[20]

It is important to note that the drug inhibitor compound file within Simcyp for fluvoxamine has been extensively validated from a PK‐drug–drug interaction (PK‐DDI) perspective for regulatory purposes. Our observations in this study suggests that PBPK compound libraries developed for early phase drug development (phase I–IIb and clinical pharmacology studies) may not always be suitable for PK‐DDI predictions in real patients. Third, we predicted the a priori dose range in 8 out of 11 patients (73%) in a difficult to treat patient population. In two patients (Study IDs 2 and 5), the therapeutic clozapine concentrations were well below the minimum target concentration of 350 ng/mL, which reinforces the importance of patient response and clinical judgment in clozapine dosing. Indeed, the target therapeutic range for clozapine is a useful guide for clinicians, however, some patients respond to clozapine concentrations outside of this range.[12]

Because of unpredictable PKs and a high burden of severe adverse effects that require resource intensive close monitoring, many psychiatrists are hesitant to prescribe clozapine and use it only as the last resort in seriously sick patients.[21] The MIPD‐VT approach could be used in clinics without rapid access to plasma clozapine concentrations or to change the titration schedule. Ideally, pharmacogenomic results should be available within the first week of commencing clozapine. Importantly, the proposed MIPD‐VT approach does not supplant TDM when it is available or clinical judgment regarding patient response. Instead, it is used to understand the factors driving the PK variability of clozapine and assist in predicting optimal starting doses. This approach also provides re‐assurance to the clinician in cases where rapid titration to a higher clozapine dose may be required, and then TDM is used to nuance this dose to achieve the desired clinical response. In other words, the MIPD‐VT approach could be used to predict optimal starting doses of clozapine, and then TDM used with standard dose proportional adjustments or Bayesian methods to nuance the dose. We therefore propose it should be possible to further reduce pseudo‐clozapine resistance caused by subtherapeutic concentrations and avoid ADRs linked to supratherapeutic clozapine concentrations by using the proposed MIPD‐VT approach.

There are several limitations of this study. First, given the retrospective real‐world nature of the clinical data, there may be some inaccuracies, such as the time of administration, patient adherence,[22] and the actual quantity of cigarettes smoked per day. However, these issues were mitigated as much as possible, as all patients were institutionalized and under direct supervision for dosing, blood sample collection, and day‐to‐day function. Second, PBPK drug profile validation is limited by access to sufficient number and quality of independent clinical data sets. This is illustrated in the overprediction of clozapine concentrations following fluvoxamine augmentation in this study, as validation of this specific DDI is difficult due to lack of adequate clinical data sets. We acknowledge that the observed overprediction with fluvoxamine augmentation in this study is limited by the small sample size of three patients. Further validation of this observed overprediction is warranted in a larger cohort of patients. Third, the Simcyp interface is not designed for clinical practice, including how diseases change physiological parameters important in PK predictions. For example, CRP elevations can result in CYP1A2 phenoconversion influencing clozapine PK.[10] Fourth, assumptions were made when making assignments of CYP1A2 enzyme abundance in smokers based on CYP1A2 genotype. Future clinical work to quantify CYP1A2 induction at varying levels of cigarette consumption by CYP1A2 genotype using CYP1A2‐selective probe substrate studies are warranted.

In summary, our previously described MIPD‐VT approach to predict clozapine PKs[11] was verified by comparing its predictions with the observations in a new cohort of patients with treatment‐resistant schizophrenia in a mental healthcare tertiary setting. Subsequently, our approach has the potential to be utilized on admission of patients with schizophrenia to proactively identify the required clozapine dose range to achieve the therapeutic threshold concentrations and help avoid toxicity during the clinical optimization of these patients. Prior to clinical implementation, however, pragmatic prospective trials will be needed to demonstrate clinical validity and utility within inpatient or outpatient settings.

AUTHOR CONTRIBUTIONS

S.M., R.R., T.M.P., C.M.J.K., C.B., A.R.‐H., and L.J.S. wrote the manuscript. S.M., R.R., T.M.P., C.M.J.K., R.S., and P.C. designed the research. S.M. performed the research. S.M., R.R., T.M.P., and C.M.J.K. analyzed the data.

ACKNOWLEDGMENTS

The authors wish to acknowledge the contributions of the MAGERS study project team, study participants, clinicians at recruitment services, and staff at BC Psychosis Program (BCPP). The authors would also like to acknolwedge the staff at BC provincial toxicology centre for performing the clozapine assay and Sandrine Merette for assisting with questions on the assay. The MAGERS study received funding from the BC Schizophrenia Society and Foundation, UBC Development office (donations from Robert Baker and Elizabeth Carter), the Djavad Mowafaghian Centre for Brain Health, VGH, and UBC Hospital Foundation (donations from the James Family Foundation), UBC Neuropsychiatry ERIN Research Fund; and by in‐kind support from the UBC Institutes of Mental Health and Department of Psychiatry. Certara UK Limited (Simcyp Division) granted access to the Simcyp Simulators through a sponsored academic license. Open access publishing facilitated by Monash University, as part of the Wiley ‐ Monash University agreement via the Council of Australian University Librarians.

FUNDING INFORMATION

No funding was received for this work.

CONFLICT OF INTEREST STATEMENT

S.M. and L.S. are employees and shareholders of myDNA Inc., a pharmacogenomic testing and interpretation company. T.M.P. provides a consultancy service to Sonic Genetics for the interpretation of pharmacogenomic test results. T.M.P. and A.R.‐H. are employees of Certara, a company that provides modeling and simulation software and services to the pharmaceutical industry, including a population‐based PBPK simulator (Simcyp). C.M.J.K. was the academic lead on the Certara‐Monash Fellowship program funded by MTPConnect. C.A.B. is founder and equity holder of Sequence2Script Inc. and a member of the Clinical Pharmacogenetics Implementation Consortium and the Genetic Testing Committee of the International Society of Psychiatric Genetics. He has also received material support from Assurex, CNSDose, Genomind, and AB‐Biotics for research purposes and has ongoing research collaborations with myDNA but does not have equity, stocks, or options in these companies or any other pharmacogenetic companies. All other authors declared no competing interests for this work.

psp413093-sup-0001-FigureS1.jpg

GRAPH: Table S1.

REFERENCES 1 Darwich AS, Polasek TM, Aronson JK, et al. Model‐informed precision dosing: background, requirements, validation, implementation, and forward trajectory of individualizing drug therapy. Annu Rev Pharmacol Toxicol. 2021 ; 61 : 225 ‐ 245. 2 Polasek TM, Rostami‐Hodjegan A. Virtual twins: understanding the data required for model‐informed precision dosing. Clin Pharmacol Ther. 2020 ; 107 (4): 742 ‐ 745. 3 Polasek TM, Tucker GT, Sorich MJ, et al. Prediction of olanzapine exposure in individual patients using physiologically based pharmacokinetic modelling and simulation. Br J Clin Pharmacol. 2018 ; 84 (3): 462 ‐ 476. 4 Rowland A, van Dyk M, Hopkins AM, et al. Physiologically based pharmacokinetic modeling to identify physiological and molecular characteristics driving variability in drug exposure. Clin Pharmacol Ther. 2018 ; 104 (6): 1219 ‐ 1228. 5 McLachlan AJ, Hilmer SN, Le Couteur DG. Variability in response to medicines in older people: phenotypic and genotypic factors. Clin Pharmacol Ther. 2009 ; 85 (4): 431 ‐ 433. 6 Lin JH. Pharmacokinetic and pharmacodynamic variability: a daunting challenge in drug therapy. Curr Drug Metab. 2007 ; 8 (2): 109 ‐ 136. 7 Fendt R, Hofmann U, Schneider ARP, et al. Data‐driven personalization of a physiologically based pharmacokinetic model for caffeine: a systematic assessment. CPT Pharmacometrics Syst Pharmacol. 2021 ; 10 : 782 ‐ 793. doi: 10.1002/psp4.12646 8 Lee J, Kim MG, Jeong HC, Shin KH. Physiologically‐based pharmacokinetic model for clozapine in Korean patients with schizophrenia. Transl Clin Pharmacol. 2021 ; 29 (1): 33 ‐ 44. 9 Gerhart JG, Carreño FO, Loop MS, et al. Use of real‐world data and physiologically‐based pharmacokinetic modeling to characterize enoxaparin disposition in children with obesity. Clin Pharmacol Ther. 2022 ; 112 (2): 391 ‐ 403. de Leon J, Schoretsanitis G, Smith RL, et al. An international adult guideline for making clozapine titration safer by using six ancestry‐based personalized dosing titrations, CRP, and clozapine levels. Pharmacopsychiatry. 2022 ; 55 (2): 73 ‐ 86. Mostafa S, Polasek TM, Bousman C, et al. Delineating gene‐environment effects using virtual twins of patients treated with clozapine. CPT Pharmacometrics Syst Pharmacol. 2023 ; 12 (2): 168 ‐ 179. Siskind D, Sharma M, Pawar M, et al. Clozapine levels as a predictor for therapeutic response: a systematic review and meta‐analysis. Acta Psychiatr Scand. 2021 ; 144 (5): 422 ‐ 432. Illumina. HiSeq X System: Lab setup and site prep guide. Illumina, Inc. ; 2017. Marks P, Garcia S, Barrio AM, et al. Resolving the full spectrum of human genome variation using linked‐reads. Genome Res. 2019 ; 29 (4): 635 ‐ 645. Lee SB, Wheeler MM, Thummel KE, Nickerson DA. Calling star alleles with stargazer in 28 pharmacogenes with whole genome sequences. Clin Pharmacol Ther. 2019 ; 106 (6): 1328 ‐ 1337. Pharmacogene Variation Consortium (PharmVar). at www.PharmVar.org ; (Gaedigk et al. 2018, CPT 103:399; Gaedigk et al. 2019, CPT 105:29). [Accessed April 11, 2023] Available from: https://www.pharmvar.org/genes Clinical Pharmacogenetics Implementation Consortium (CPIC). [cited APR 11 2023]. https://cpicpgx.org/alleles/ Plowchalk DR, Rowland Yeo K. Prediction of drug clearance in a smoking population: modeling the impact of variable cigarette consumption on the induction of CYP1A2. Eur J Clin Pharmacol. 2012 ; 68 (6): 951 ‐ 60. Koonrungsesomboon N, Khatsri R, Wongchompoo P, Teekachunhatean S. The impact of genetic polymorphisms on CYP1A2 activity in humans: a systematic review and meta‐analysis. Pharmacogenomics J. 2018 ; 18 (6): 760 ‐ 768. Mostafa S, Polasek TM, Bousman CA, et al. Pharmacogenomics in psychiatry ‐ the challenge of cytochrome P450 enzyme phenoconversion and solutions to assist precision dosing. Pharmacogenomics. 2022 ; 23 (15): 857 ‐ 867. Bachmann CJ, Aagaard L, Bernardo M, et al. International trends in clozapine use: a study in 17 countries. Acta Psychiatr Scand. 2017 ; 136 (1): 37 ‐ 51. Couchman L, Morgan PE, Spencer EP, Flanagan RJ. Plasma clozapine, norclozapine, and the clozapine:norclozapine ratio in relation to prescribed dose and other factors: data from a therapeutic drug monitoring service, 1993‐2007. Ther Drug Monit. 2010 ; 32 (4): 438 ‐ 447.

By Sam Mostafa; Reza Rafizadeh; Thomas M. Polasek; Chad A. Bousman; Amin Rostami‐Hodjegan; Robert Stowe; Prescilla Carrion; Leslie J. Sheffield and Carl M. J. Kirkpatrick

Reported by Author; Author; Author; Author; Author; Author; Author; Author; Author

Titel:
Virtual twins for model-informed precision dosing of clozapine in patients with treatment-resistant schizophrenia.
Autor/in / Beteiligte Person: Mostafa, S ; Rafizadeh, R ; Polasek, TM ; Bousman, CA ; Rostami-Hodjegan, A ; Stowe, R ; Carrion, P ; Sheffield, LJ ; Kirkpatrick, CMJ
Link:
Zeitschrift: CPT: pharmacometrics & systems pharmacology, Jg. 13 (2024-03-01), Heft 3, S. 424-436
Veröffentlichung: 2015- : Hoboken, NJ : Wiley ; <i>Original Publication</i>: New York, NY : Nature Pub. Group, 2024
Medientyp: academicJournal
ISSN: 2163-8306 (electronic)
DOI: 10.1002/psp4.13093
Schlagwort:
  • Humans
  • Fluvoxamine
  • Schizophrenia, Treatment-Resistant
  • Clozapine pharmacokinetics
  • Clozapine therapeutic use
  • Antipsychotic Agents pharmacokinetics
  • Schizophrenia drug therapy
Sonstiges:
  • Nachgewiesen in: MEDLINE
  • Sprachen: English
  • Publication Type: Journal Article
  • Language: English
  • [CPT Pharmacometrics Syst Pharmacol] 2024 Mar; Vol. 13 (3), pp. 424-436. <i>Date of Electronic Publication: </i>2024 Jan 19.
  • MeSH Terms: Clozapine* / pharmacokinetics ; Clozapine* / therapeutic use ; Antipsychotic Agents* / pharmacokinetics ; Schizophrenia* / drug therapy ; Humans ; Fluvoxamine ; Schizophrenia, Treatment-Resistant
  • References: Pharmacogenomics. 2022 Oct;23(15):857-867. (PMID: 36169629) ; Clin Pharmacol Ther. 2020 Apr;107(4):742-745. (PMID: 32056199) ; Genome Res. 2019 Apr;29(4):635-645. (PMID: 30894395) ; Pharmacopsychiatry. 2022 Mar;55(2):73-86. (PMID: 34911124) ; Transl Clin Pharmacol. 2021 Mar;29(1):33-44. (PMID: 33854999) ; CPT Pharmacometrics Syst Pharmacol. 2021 Jul;10(7):782-793. (PMID: 34053199) ; Annu Rev Pharmacol Toxicol. 2021 Jan 6;61:225-245. (PMID: 33035445) ; Br J Clin Pharmacol. 2018 Mar;84(3):462-476. (PMID: 29194718) ; CPT Pharmacometrics Syst Pharmacol. 2023 Feb;12(2):168-179. (PMID: 36424701) ; Curr Drug Metab. 2007 Feb;8(2):109-36. (PMID: 17305491) ; Pharmacogenomics J. 2018 Dec;18(6):760-768. (PMID: 29282363) ; CPT Pharmacometrics Syst Pharmacol. 2024 Mar;13(3):424-436. (PMID: 38243630) ; Eur J Clin Pharmacol. 2012 Jun;68(6):951-60. (PMID: 22258279) ; Clin Pharmacol Ther. 2022 Aug;112(2):391-403. (PMID: 35451072) ; Clin Pharmacol Ther. 2009 Apr;85(4):431-3. (PMID: 19225449) ; Acta Psychiatr Scand. 2017 Jul;136(1):37-51. (PMID: 28502099) ; Acta Psychiatr Scand. 2021 Nov;144(5):422-432. (PMID: 34374073) ; Clin Pharmacol Ther. 2018 Dec;104(6):1219-1228. (PMID: 29574693) ; Clin Pharmacol Ther. 2019 Dec;106(6):1328-1337. (PMID: 31206625) ; Ther Drug Monit. 2010 Aug;32(4):438-47. (PMID: 20463634)
  • Substance Nomenclature: J60AR2IKIC (Clozapine) ; 0 (Antipsychotic Agents) ; O4L1XPO44W (Fluvoxamine)
  • Entry Date(s): Date Created: 20240120 Date Completed: 20240318 Latest Revision: 20240522
  • Update Code: 20240522
  • PubMed Central ID: PMC10941576

Klicken Sie ein Format an und speichern Sie dann die Daten oder geben Sie eine Empfänger-Adresse ein und lassen Sie sich per Email zusenden.

oder
oder

Wählen Sie das für Sie passende Zitationsformat und kopieren Sie es dann in die Zwischenablage, lassen es sich per Mail zusenden oder speichern es als PDF-Datei.

oder
oder

Bitte prüfen Sie, ob die Zitation formal korrekt ist, bevor Sie sie in einer Arbeit verwenden. Benutzen Sie gegebenenfalls den "Exportieren"-Dialog, wenn Sie ein Literaturverwaltungsprogramm verwenden und die Zitat-Angaben selbst formatieren wollen.

xs 0 - 576
sm 576 - 768
md 768 - 992
lg 992 - 1200
xl 1200 - 1366
xxl 1366 -