Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Sep 1.
Published in final edited form as: J Clin Pharmacol. 2024 Apr 29;64(9):1130–1140. doi: 10.1002/jcph.2446

Assessing Pharmacogenomic loci Associated with the Pharmacokinetics of Vamorolone in Boys with Duchenne Muscular Dystrophy

Xiaonan Li 1, Daniele Sabbatini 2, Elena Pegoraro 2, Luca Bello 2, Paula Clemens 3, Michela Guglieri 4, John van den Anker 5,6, Jesse Damsker 6, John McCall 6, Utkarsh J Dang 7, Eric P Hoffman 6,8, William J Jusko 1,*
PMCID: PMC11357888  NIHMSID: NIHMS1982133  PMID: 38682893

Abstract

Human genetic variation (polymorphisms) in genes coding proteins involved in the absorption, distribution, metabolism, and elimination (ADME) of drugs can have a strong effect on drug exposure and downstream efficacy and safety outcomes. Vamorolone, a dissociative steroidal anti-inflammatory drug for treating Duchenne Muscular Dystrophy (DMD), primarily undergoes oxidation by CYP3A4 and CYP3A5 and glucuronidation by UDP-glucuronosyltransferases. This work assesses the pharmacokinetics (PK) of vamorolone and sources of inter-individual variability (IIV) in 81 steroid-naïve boys with DMD aged 4 to <7 years old considering the genetic polymorphisms of CYPS3A4 (CYP3A4*22, CYP3A4*1B), CYP3A5 (CYP3A5*3) and UGT1A1 (UGT1A1*60) utilizing population pharmacokinetic (PopPK) modeling. A one-compartment model with zero-order absorption (Tk0, duration of absorption), linear clearance (CL/F) and volume (V/F) describes the plasma PK data for boys with DMD receiving a wide range of vamorolone doses (0.25~6 mg/kg/day). The typical CL/F and V/F values of vamorolone were 35.8 L/hr and 119 L, with modest IIV. The population Tk0 was 3.14 hr yielding an average zero-order absorption rate (k0) of 1.16 mg/kg/hr with similar absorption kinetics across subjects at the same vamorolone dose (i.e., no IIV on Tk0). The covariate analysis showed that none of the genetic covariates had any significant impact on the PK of vamorolone in boys with DMD. Thus, the PK of vamorolone is very consistent in these young boys with DMD.

Keywords: Duchenne Muscular Dystrophy, vamorolone, pharmacogenomics, population pharmacokinetic modeling

Introduction

Duchenne Muscular Dystrophy (DMD) is a genetic muscle-wasting disease caused by variants of the DMD gene that leads to deficient or defective dystrophin production in muscle1, resulting in progressive muscle deterioration and later loss of ambulation, and ultimately dilated cardiomyopathy, the need for assisted ventilation and premature death2. It mainly affects males with symptom onset in early childhood (around 2–5 years old)3. Corticosteroids (CS) serve as the current standard of care in patients with DMD to help alleviate symptoms and slow disease progression. However, time- and dose-dependent side effects associated with CS, such as loss of bone density, weight gain, behavior changes, and stunting of growth, seriously affect patients’ quality of life4.

Vamorolone is a novel steroidal anti-inflammatory drug that is structurally differentiated from other corticosteroids by lacking the 11β oxygen moiety; the 11β position is a hydroxyl in active hormones and drugs (cortisol; active corticosteroid drugs), or a ketone in inactive hormones and drugs (cortisone; corticosteroid pro-drugs). As a result of loss of the 11β oxygen (replaced by a Δ9–11 carbon double bond), vamorolone uniquely loses substrate activity for the two drug/pro-drug modulatory 11β-hydroxysteroid dehydrogenase enzymes (HSD11B1, HSD11B2)5. The loss of the 11β oxygen also removes a contact site with the glucocorticoid receptor, changing the 3-dimensional structure of GR/ligand complexes at the obligatory co-factor binding site, with resulting changes in co-repressor (increased) and co-activator (decreased) binding6. Vamorolone is an antagonist of the mineralocorticoid receptor, with activities similar to eplerenone and spironolactone in pre-clinical models – this is also unique among the steroidal anti-inflammatory drugs7. Thus, while vamorolone binds to the same receptors as corticosteroids (i.e., glucocorticoid and mineralocorticoid receptors), vamorolone modifies the downstream activity of the receptors and shows beneficial efficacy/safety profiles versus other corticosteroids as seen in pre-clinical (mouse) studies7, 8, and human clinical trials.

Vamorolone received regulatory approval for use in Duchenne muscular dystrophy in 2023 by both EMA (dissociative steroid label; age 4 years and older), and FDA (corticosteroid label; age 2 years and older). In a double-blind clinical trial of vamorolone vs. placebo and prednisone, vamorolone showed robust evidence of clinical benefit for all motor outcomes studied, and reduced safety concerns relative to prednisone (no stunting of growth, no deleterious changes in bone biomarkers)9, 10. These findings were further supported by long-term open label studies vs. natural history comparators11, 12.

Vamorolone is primarily metabolized by oxidation (CYP3A4, CYP3A5) and glucuronidation (UDP-glucuronosyltransferases). Corresponding genes contain polymorphisms that result in different enzymatic functionalities and thereby contribute to variability in pharmacokinetics (PK) of their substrates. One of the key single-nucleotide polymorphisms (SNPs) related to the functional variation of CYP3A5 is 6986A>G (rs776746), with the wild-type A nucleotide defined as *1 allele and the variant G nucleotide defined as *3 allele. Homozygous carriers of G (GG or CYP3A5*3/*3) produce very low or non-detectable levels of functional CYP3A5 protein13, and were reported to have significantly higher systemic exposure of tacrolimus and lower dose requirements as compared to CYP3A5*1/*1 (AA) or CYP3A5*1/*3 (AG) carriers among solid-organ transplant recipients14. CYP3A4*1B (rs2740574, −392A>G) is one of the most extensively studied CYP3A4 SNPs, which is linked to enhanced CYP3A4 activity15, 16. Another relevant CYP3A4 SNP is CYP3A4*22 (rs35599367), with the homozygous GG being the wild type and the presence of AG or AA associated with decreased enzyme activity of CYP3A417, 18. The c.−3279 T > G mutation of UGT1A1 (UGT1A1*60, rs4124874) is more common in Caucasians than other ethnic groups19, 20. This SNP results in reduced UGT1A1 activity and is associated with the toxic effects of irinotecan due to decreased glucuronidation of its active metabolite SN-38 by UGT1A1. A previous case report showed that the reduced methylprednisolone clearance in a male subject could not be explained by the CYP3A5*3/*3 genotype21.

Despite the established relationships of drug exposure and these pharmacogenomic gene loci in other disease states that have led to genotype-guided dosing of certain drugs17, 2225, no studies of pharmacogenomics (PGx) have been done with vamorolone. The PK of vamorolone and sources of variability were assessed by population PK (PopPK) analysis previously using data from parallel-group studies in 86 healthy men (fed and fasted, phase I, NCT02415439) and 48 boys with DMD (phase IIa, NCT02415439)26. A one-compartment model with zero-order absorption, linear clearances (CL/F) and volumes (V/F) was applied, and covariates found to significantly affect the PK were food (high fat meal) and body weight (BW). This work builds upon this previous PopPK analysis to study 81 boys with DMD (27 from a phase IIa dose-ranging study [NCT02760264], and 54 from a more recent phase IIb study [NCT03439670]). We assess the impact of gene polymorphisms on vamorolone PK in boys with DMD (i.e., male, steroid-naïve, 4 to <7 years of age) undergoing these clinical trials with vamorolone.

Methods

Subjects and PK Sampling

Ethical standards committee approvals on the VBP15–002, VBP15–003, and VBP15–004 clinical trials were obtained for each of the 33 participating clinical trial recruitment sites, either for individual institutional or regional approvals. Written informed consent was obtained from all parents/guardians of participants in each trial (consent for research). The studies were conducted in accordance with the International Conference on Harmonization Guideline on Good Clinical Practice and applicable national or regional regulations or guidelines.

The patient populations assessed in the current PopPK analysis were previously steroid-naïve boys with DMD aged 4 to <7 years old from two vamorolone clinical trials (i.e., VBP15–002 and VBP15–004). VBP15–002 (NCT02760264) is a first-in-patient 2-week, open-label Phase IIa multiple ascending dose (0.25, 0.75, 2.0 and 6.0 mg/kg/day) study with 12 boys being assigned to each of the dose groups. Serial PK samples were collected on days 1 and 14 at 0 (pre-dose), 1, 2, 4, 6, and 8 hours post-dose. VBP15–004 (NCT03439670) is a randomized, double-blind, placebo- and prednisone-controlled Phase IIb trial carried out in 121 boys with DMD that included two 24-week treatment periods. For treatment period 1 (Weeks 1–24), participants were randomly assigned to the placebo (tablets or oral suspension), prednisone (0.75 mg/kg/day as tablets), and vamorolone (2 or 6 mg/kg/day as oral suspension) groups in a 1:1:1:1 ratio. A 4-week transition period (Weeks 25–28) followed the end of treatment period 1 during which all subjects continued to receive the same oral suspension (vamorolone or matching placebo) that they received during treatment period 1 and were tapered off their study medication tablets (prednisone or matching placebo). During treatment period 2 (Weeks 28–48), all participants received vamorolone at one of two dose levels (2 or 6 mg/kg/day) and blood samples were collected at 2 hours post-dose at the Week 2 Visit. Details of these clinical studies have been published9, 27.

Single-Nucleotide Polymorphism (SNP) Genotyping and Metabolizer Phenotyping

All clinical trial participants in this current study provided informed consent (by their parents) for genetic studies of candidate gene loci. Blood samples for the genetic analyses were collected at clinical trial sites in EDTA tubes, then frozen after collection, and sent to the central laboratory. For the VBP15–002 trial, the blood samples were then sent to AGADA BioSciences (Halifax, Nova Scotia, Canada) where DNA isolation was carried out using QIAamp DNA Blood Maxi or Midi Kits. For VBP15–004, DNA isolation from frozen samples was carried out by the central laboratory, and DNA shipped to AGADA BioSciences. DNA samples were quantitated and adjusted to 100–350 ng/μl, and aliquoted by AGADA BioSciences, gridded onto 96-well microtiter plates. SNP genotyping was carried out by the Genomics Core of Icahn School of Medicine at Mt Sinai, NY using the Illumina GDA-PGx platform (GlobalDiversity Array with enhanced pharmacogenomics content). A data mask was utilized to only retrieve information related to the candidate genetic loci, thus obscuring possible identification of participants through their genetic profiles.

The specific SNPs assessed in this study included CYP3A4*22 C>T (rs35599367, C=CYP3A4*1, T=CYP3A4*22), CYP3A4*1B −392A>G (rs2740574; A=CYP3A4*1, G=CYP3A4*1B), CYP3A5*3 6986A > G (rs776746; A= CYP3A5*1, G= CYP3A5*3), and UGT1A1*60 c.−3279 T > G (rs4124874, T= UGT1A1*1, G=UGT1A1*60). For CYP3A4*22, GG allele genotype (CYP3A4*1/*1) is considered as expresser allele while AG (CYP3A4*1/*22) and AA (CYP3A4*22/*22) allele genotypes are considered as a decrease of function (DOF) alleles. For CYP3A4*1B, AA (CYP3A4*1/*1) is the wild type while GA (CYP3A4*1/*B) and GG (CYP3A4*B/*B) are DOF variants. For CYP3A5*3, individuals carrying at least one functional allele, i.e., AA (CYP3A5*1/*1) or AG (CYP3A5*1/*3), are considered as CYP3A5 expressers. UGT1A1*60 expressers carry at least one functional allele, i.e., AA (*1/*1) and CA (*1/*60) and nonexpressers are homozygous carriers of the mutant allele C (*60/*60).

As CYP3A4 and CYP3A5 exhibit substrate overlaps and comparable expression levels in some individuals13, 28, the CYP3A4*22 and CYP3A5*3 genotypes are commonly combined to assess the interaction between CYP3A4 and CYP3A5 polymorphisms23, 2931:

Poor metabolizers (PM): individuals who are CYP3A5 nonexpressers (*3/*3) and carriers of at least one DOF allele of CYP3A4 (*1/*22; *22/*22). Normal metabolizers (NM): individuals who are CYP3A4 normal expressers (*1/*1) and CYP3A5 nonexpressers (*3/*3) or those who carry at least one CYP3A5 expresser allele (*1/*1; *1/*3) and at least one DOF allele of CYP3A4*22 (*1/*22; *22/*22). Ultra-metabolizers (UM): individuals with normal CYP3A4 activity (*1/*1) and at least one CYP3A5 functional allele (*1/*1; *1/*3).

The enzyme phenotype assignments based on the expresser status for the genotypes of individual or combined SNPs are listed in Table 1.

Table 1.

Summary of subject covariates assessed in this PopPK analysis.

Covariate VBP15–002
(n=27)
VBP15–004
(n=54)
Total
(n=81)
Body weight (kg)
 Mean (CV%) 20.14 (13.75) 20.19 (16.42) 20.18 (15.5)
 Median [Min, Max] 19.97 [15.1, 26.2] 20.15 [14.2, 26.2] 20.1 [14.2, 26.2]
Individual SNP genotype Phenotypea n (%)
 CYP3A4*22 (C>T, rs35599367)
  GG (*1/*1) NM 23 (85) 44 (81) 67 (83)
  AG (*1/*22) PM 4 (15) 10 (19) 14 (17)
 CYP3A4*1B (392A>G, rs2740574)
  AA (*1/*1) NM 23 (85) 50 (93) 73 (90)
  GA; GG (*1/*1B; *1B/*1B) UM 4 (15) 4 (7) 8 (10)
 CYP3A5*3 (6986A > G, rs776746)
  AA; AG (*1/*1; *1/*3) NM 4 (15) 9 (17) 13 (16.1)
  GG (*3/*3) PM 23 (85) 45 (83) 68 (83.9)
 UGT1A1*60 (c.−3279 T > G, rs4124874) a
  AA; CA (*1/*1; *1/*60) NM 22 (81.5) 42 (77.8) 64 (79)
  CC (*60/*60) PM 5 (18.5) 12 (22.2) 17 (21)
Combined SNP genotype
 CYP3A4
  *1/*1 NM 19 (70.4) 40 (74.1) 59 (72.8)
  *1/*22; *1B/*22 PM 4 (14.8) 10 (18.5) 14 (17.3)
  *1B/*1B; *1/*1B UM 4 (14.8) 4 (7.4) 8 (9.9)
 CYP3A4/5 b
  3A4*1/*1 & 3A5*3/*3; 3A4 (*1/*22; *22/*22) & 3A5 (*1/*1; *1/*3) NM 19 (70.4) 35 (64.8) 54 (66.7)
  3A4 (*1/*22; *22/*22) & 3A5*3/*3 PM 4 (14.8) 10 (18.5) 14 (17.3)
  3A4*1/*1 & 3A5 (*1/*1; *1/*3) UM 4 (14.8) 9 (16.7) 13 (16.0)
a

In NONMEM, NM, PM, and UM were indicated by 0, 1, and 2.

Population PK Analysis

The PopPK analysis was carried out using the first-order conditional estimation method with interaction (FOCEI) implemented in the nonlinear mixed effects modeling program (NONMEM) version 7.5.0 (ICON Development Solutions, San Antonio, TX, USA). NONMEM was used with Pirana (version 21.11.1), Perl-speaks-NONMEM (PsN, version 5.3.0), and Xpose (version 4). Data manipulation, plotting, and descriptive statistics calculation were performed using R (version 4.3.0).

All PK data were simultaneously described by a one-compartment model with zero-order absorption as was done previously26:

V/FdCdt=Input-CL/FC (1)
Input=DoseTk0,      tTk00,          t>Tk0 (2)

where V/F is the apparent volume of distribution, CL/F is the apparent clearance, C is plasma concentration, Input is the zero-order absorption rate (k0), and Tk0 is the duration of Input.

For individual i, the model parameters are:

Tk0,i=Tk0~eηTk0,i (3)
V/Fi=V/F~eηv,i (4)
CL/Fi=CL/F~eηCL,i (5)

where bars indicate typical values of parameters; the IIV of model parameters were described using an exponential function and η values are the random-effect parameters that were assumed to be normally distributed with mean 0 and variance ω2; individual-specific parameters were assumed to have a log-normal distribution.

Residual variability was described using an additive plus constant coefficient of variation model:

Yij=Y^ij+Y^ijε1ij+ε2ij (6)

where Yij and Y^ij are the jth observed and predicted concentrations for the ith individual; ε1ij and ε2ij are the proportional and additive random residual effects. Both ε were assumed to be normally distributed (ε1ij~N(0,σ2prop), ε2ij~N(0,σ2add)).

The relationships between individual parameter estimates obtained by the base PopPK model and the genetic covariates listed in Table 1 were initially assessed by graphical evaluation. Next, potential relationships were evaluated using the Stepwise Covariate Modeling (SCM) automated method in PsN implemented by Pirana with the forward selection and backward elimination. For the forward selection, statistical significance was considered a decrease of the objective function value (OFV) of at least 3.84 (χ2, p < 0 .05, df=1) and for backward elimination an increase of OFV of at least 10.83 (χ2, p < 0.001, df=1). Functional forms used for the covariate analysis included a power model for continuous covariates, i.e., BW (Eq.7) and a linear function for categorical covariates, i.e., pharmacogenomic loci (Eq.8):

Pi=PpopBWiMedian BWθBWPi (7)
Pi=Ppop,                             NMs=0Ppop(1+θPMs),     PMs=1Ppop(1+θUMs),     UMs=2 (8)

where Pi represents the individual PK parameter; Ppop indicates the typical value of the parameter; θBWPi is the power coefficient reflecting the change in the natural log of individual parameter per unit change in the natural log of the ratio of individual BWi and median BW (20.1 kg); θPMs and θUMs reflect the fold-changes in parameters for PM and UM as compared to those for NM.

The evaluation of model performance was based on goodness-of-fit (GOF) plots, precision of parameter estimates, the OFV and visual predictive check (VPC). Typical GOF diagnostic plots consist of observed concentration (DV) versus individual predicted concentration (IPRED), DV versus population predicted concentration (PRED), conditional weighted residuals (CWRES) versus PRED, and CWRES versus time since the first dose (TIME). The VPC was performed with 1000 simulated datasets that were obtained based on the parameter values from the final model, with the 5th, 50th, and 95th percentiles of the predicted concentration-time data plotted and compared to those of the observed data to assess the agreement graphically.

Individual PK exposure values of vamorolone in boys with DMD, i.e., area under the concentration-time curve from time 0 to infinity (AUCinf), were further obtained from dose divided by the PopPK model predicted individual CL/F.

Statistical Analysis

The statistical analysis was performed using R (version 4.3.0). Linear regression was applied to assess the relationships between individual PK parameters and continuous covariates. The Wilcoxon rank sum test was applied to compare the categorical data between two groups and the Kruskal-Wallis test was used when there were more than two groups. Statistically significant differences were concluded when p <0.05.

Results

A total of 169 participants with DMD were enrolled in the VBP15–002/003 (n=48) and VBP15–004 clinical trials (n=121). A total of 123 boys with DMD (47 from VBP15–002 and 76 from VBP15–004) who had evaluable vamorolone PK data are displayed in Figure 1. Missing PK data from VBP15–004 (n=45 missing) was due to missed visits due to the COVID pandemic, or refusal of the participant for PK sampling. Among these, 81 boys (27 from VBP15–002 and 54 from VBP15–004) with available PGx data were included. DNA samples that were missing were due to national ethical considerations (Israel) or refusal of the participant for blood draws for DNA. Concentrations below the lower limit of quantification (LOQ) (i.e., 2 ng/mL) were also included in the final modeling dataset. The value and feasibility of incorporating concentration data below the LOQ in PK analysis such as PopPK modeling have been demonstrated previously32, 33. Covariates BW and PGx are summarized in Table 1. The BW distributions of boys with DMD from the two clinical studies are similar, with an overall median value of 20.1 kg (range: 14.2~26.2 kg). Among all included subjects, UGT1A1 NMs (*1/*1; *1/*60) and PMs (*60/*60) accounted for 79% and 21%. For the remaining CYP SNPs, the variant allele frequencies of CYP3A4*22 (A) and CYP3A4*1B (G) are comparable (17% and 10%) with more than 80% of boys carrying the wild-type alleles (*1/*1). In contrast, the majority (84%) of boys (68 out of 81) were CYP3A5 PMs carrying the non-functional homozygote GG (CYP3A5*3/*3) whereas 13 (16%) were NMs with at least one functional allele (i.e., AG or AA). For the combined CYP3A4 and CYP3A4/5 genotypes, 72.8% and 66.7% were designated as NMs, 9.9% and 16% were defined as UMs, and PMs accounted for 17.3% in both categories among the 81 patients with available CYP genotypes. White is the predominant race (~98%) in the clinical trials; the allele frequencies of the studied variants of CYP3A4, CYP3A5 and UGT1A1 in boys with DMD were generally consistent with those reported for the white population.3437

Figure 1.

Figure 1.

Plasma vamorolone concentration-time profiles for boys with DMD from VBP15–002 and VBP15–004 studies. Closed circles are individual observations color coded by the indicated doses (unit: mg/kg/day). Lines connect mean values at each dose. Note: 2-mg/kg/day values from the VBP15–004 study are moved from 2 to 1.8 hr for better visualization.

In our preliminary assessments, all 123 boys with DMD with evaluable vamorolone PK data were included to examine the influence of BW on PK parameters applying a power function (Eq. 7). The BW of the 123 boys were summarized in Supplementary Table S1. The estimated power coefficients for CL/F and V/F were close to 0.75 and 1.0, which is consistent with the typical allometric relationship between BW and these PK parameters38. Therefore, the power coefficients for CL/F and V/F in Eq. 7 were fixed to 0.75 and 1 (Figure 2) in the subsequent PopPK analysis involving the 81 boys with DMD who had PGx data, with the primary goal to assess the impact of genetic covariates on vamorolone PK.

Figure 2.

Figure 2.

Individual estimates of clearances (CL/F) and volumes (V/F) versus BW. The solid red line indicates nonlinear regression line applying Eq. 7 with fixed power coefficients of 0.75 for CL/F and 1.0 for V/F.

The plasma PK profiles of vamorolone in the 81 boys with DMD receiving the 0.25~6 mg/kg/day doses were reasonably described by a one-compartment model with zero-order absorption, the same base PK model as previously26. As shown by the PK plots from the 002 study (Figure 3 and Supplementary Figure S1), both individual and population predicted concentrations are in good agreement with the observed data. All model parameters were well estimated with low CV% values (Table 2). The duration of the zero-order absorption process (Tk0) was estimated to be 3.14 hr, yielding an average zero-order absorption rate (k0) of 1.16 mg/kg/hr. No IIV was estimated on Tk0, indicating that the absorption kinetics of vamorolone in boys with DMD are similar at the same vamorolone dose given the comparable PK data. The apparent clearance (CL/F) and apparent volume of distribution (V/F) were estimated to be 35.8 L/hr (34.9 % IIV) and 119 L (42.8% IIV), which is 1.78 L/hr/kg and 5.92 L/kg given a median BW of 20.1 kg (Table 1). These PK parameter estimates are comparable to those obtained previously where the PK data for healthy men were included26.

Figure 3.

Figure 3.

Individual plasma concentration versus time plots for boys receiving 2 or 6 mg/kg of vamorolone from the 002 study. Closed circles are observations; solid and dotted lines indicate individual and population predictions by the PopPK model. The data for study day 1 and day 14 are indicated by red and blue.

Table 2.

Final pharmacokinetic parameter estimates obtained in the population analysis.

Parameter [Units] Definition Final Model Bootstrap
Estimate RSE% Median 95%CI
Population typical values
Tk0 [hr] Duration of zero-order absorption process 3.14 4.4 3.14 (2.66, 3.68)
CL/F [L/hr] Apparent clearance 35.8 17 35.83 (31.82, 40.51)
V/F [L] Apparent volume of distribution 119 13.2 118 (98, 145)
Inter-individual variability
ω2-CL/F Interindividual variability of CL/F 0.122 38.9 0.114 (0.043, 0.262)
CV% 34.9 33.8
ω2-V/F Interindividual variability of V/F 0.183 37 0.184 (0.083, 0.312)
CV% 42.8 42.9
Residual variability
σ 2 - Prop Proportional residual error variance 0.298 13.3 0.291 (0.215, 0.382)
σ 2 - Add Additive residual error variance 7.02E-08 38.2 6.83E-08 (1.07E-08, 3.81E-07)

Typical GOF diagnostic plots (Supplementary Figure S3) revealed no significant systemic deviation or major bias, indicating the robustness of the model fits. There is a slight bias for individual plasma drug concentration greater than about 500 ng/mL (Supplementary Figure S3 panel A). The IIV on Tk0 was tested in our preliminary analysis, however, neither the IIV can be identified, nor the indicated bias can be improved. This might be either due to the lack of data at early time points or may be explained by similar absorption kinetics across subjects at the same vamorolone dose. Therefore, the IIV on Tk0 was not included in the current model. Supplementary Figure S4 displays the VPC plots stratified by dose. As shown, the 5th, 50th and 95th percentiles of the observed data generally fall within their corresponding 90% prediction intervals of the simulated data generated by 1000 model simulations, representing a good predictive performance of the PopPK model.

For the covariate analysis, the individual PK parameter estimates obtained by the base PK model were plotted against the genetic covariates listed in Table 1 and statistical analysis was performed to examine any potential significant trends. As was shown, no statistically significant covariate-parameter relationship was identified for CL/F (Figure 4) or V/F (Figure S2). In line with the graphical assessment results, the automated SCM also showed that none of the genetic covariates exhibited any significant impact on either the CL/F or V/F of vamorolone in these subjects and thus the base PK model was accepted as the final model.

Figure 4.

Figure 4.

Individual estimates of clearances (CL/F) versus genetic covariates. The boxes represent the 25th to 75th percentiles, with the whiskers extending to the 5th and 95th percentiles of the data and the asterisks representing outliers. The boxes are joined at the median values.

Bootstrap analysis was performed on the final model yielding a 96.4% convergence rate (i.e., the model converged in 482 out of 500 bootstrap runs). The bootstrap results are presented in Table 2. The original final model parameter estimates are close to the bootstrap medians and are included in the 95%CI based on the bootstrap procedure, suggesting good qualification of the final PopPK model in characterizing the PK data of vamorolone in boys with DMD.

The individual AUCinf values of vamorolone in boys with DMD calculated from Dose divided by (CL/F) are summarized in Table 3. The predicted AUCinf values in this work are comparable to those obtained by non-compartmental analysis (NCA) using the day-14 PK data for boys with DMD from the 002 study39.

Table 3.

Summary of individual AUCinf (ng×hr/mL) values predicted by the PopPK model.

Dose [mg/kg/day] No. of Subjects Mean (CV%) Median [Min, Max]
VBP15–002 VBP15–004
0.25 9 NA 132 (30.5) 120 [81, 230]
0.75 6 NA 608 (43.2) 589 [270, 1059]
2 6 21 1130 (13.0) 1150 [720, 1340]
6 6 33 3361 (8.8) 3340 [2320, 3990]

NA: not applicable

Discussion

It is increasingly recognized that human genetic variation (polymorphisms) in gene coding proteins involved in ADME processes can have a strong effect on drug exposure. In a study of over 1000 patients seen at the Mayo Clinic, 99% were found to have actionable variants (observable after drug dosing) in key PGx gene loci40. A recent study of routine genotyping of 9 PGx loci (including CYP3A4, CYP3A5) found that preemptive screening offered medication improvement opportunities in 56% of participants for commonly used medications.41

Vamorolone has had ADME aspects defined through an extensive series of in vivo (mouse, dog, human) and in vitro (human liver cells and homogenates, cell lines) studies (https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/215239s000lbl.pdf; https://www.ema.europa.eu/en/documents/product-information/agamree-epar-product-information_en.pdf). Additional aspects of tying drug dose to drug exposure includes drug-drug interactions (DDI), where metabolic pathways of vamorolone might be altered by concomitant medications leading to altered vamorolone exposure for a given dose, or vice versa (a strong inhibitor of CYP3A4, itraconazole, increased vamorolone Cmax and AUC by 8% and 44%) (https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/215239s000lbl.pdf). As demonstrated by these studies, CYP3A4 and CYP3A5 are the major CYP isoforms involved in the metabolism of vamorolone. In addition, this nonionized drug is made more hydrophilic (as are all steroid hormones) by glucuronidation. Indeed, mass balance and metabolite studies of vamorolone in both adult volunteers and boys with DMD show that inactive glucuronides are at high concentrations in serum (72% of total drug-related product in sera). Glucuronidation is carried out by UGTs, and this is a large family of drug metabolism loci (as are the CYPs). The most important UGT loci for corticosteroids are UGT1A1 and UGT1A442. Both these loci have impactful and relatively common PGx alleles. The most common genetic variant for UGT1A1 seen in the Caucasian population is UGT1A1*28 (rs8175347, (TA)7TAA), with approximately 10% carrying 2 copies of the UGT1A1*28 allele. The presence of the defective variant results in reduced UGT enzyme activity and is significantly associated with the risk of irinotecan toxicity due to accumulation of its active metabolites in the blood. Carriers of the homozygous allele UGT1A1*28/*28 are at increased risk for neutropenia following the treatment of irinotecan, and a reduction in the starting dose is recommended for these individuals43, 44. Evidence has shown that there is a highly significant linkage disequilibrium that exists between UGT1A1*60 c.−3279 T > G and UGT1A1*28 in Caucasians19. For UGT1A4, there are key haplotypes that have been shown to have strong impact on UGT activity42, but these are relatively rare alleles making it challenging to develop PGx associations.

In this work, the gene polymorphisms of CYP3A5*3, CYP3A4*22, CYP3A4*1B, and UGT1A1*60 were examined and their impact on vamorolone PK were assessed in steroid naïve boys with DMD aged 4 to <7 years old utilizing PopPK modeling. The previous PopPK analysis utilized extensive data in healthy men to establish the base PK model (zero-order absorption, one-compartment PK, and linear clearances (CL/F) and volumes (V/F)26, where the significant covariates were found to be food effect for all PK parameters (Tk0, CL/F and V/F) and BW for Tk0. Here, the same base PK model was also applicable for describing the more limited PK data in boys with DMD, where the IIV was estimated on CL/F and V/F but not Tk0. According to the results of the previous work26 and given the relatively homogenous DMD patient population with a narrow age range assessed herein, only BW and gene polymorphisms of metabolizing enzymes relevant to vamorolone PK were included as potential covariates (Table 1) in this analysis. Preliminary assessments revealed typical allometric relationships between BW and PK parameters (i.e., CL/F and V/F) when all 123 boys with DMD who had evaluable vamorolone PK data were included. As such, this piece of information was incorporated into the PopPK model to solely assess the impact of gene polymorphisms on the PK for 81 boys who had both PK and PGx data. Based on the pre-defined criteria for covariate inclusion, our analysis indicated that none of the genetic covariates was considered to significantly affect the PK of vamorolone in these boys with DMD.” It is noteworthy that the PM genotypes of each of the genetic loci showed the expected decreased values of both vamorolone clearance (CL/F) and estimates of volumes (V/F) relative to NM genotypes. However, the wide variance in PK estimates, in part due to single sample estimates (2 hours) in the VBP15–004 study, which the majority (2/3rd) of participants were from, make these findings preliminary.

While the multiple blood sampling from the −002 studies provides a robust basis for the PopPK, one limitation of this study and analysis is the single blood sampling in the −004 study at the 2 and 6 mg/kg vamorolone doses. This is not uncommon in PopPK studies, especially in children. Another limitation of this study is the relatively small number of participants (n=81) from the two trials (n=169 participants total) where both PK and DNA samples were available. This was due, in part, to some countries limiting or prohibiting DNA studies of trial participants, and limitations on patient study visits and missed sampling on participants due to the COVID-19 pandemic.

Conclusions

The gene polymorphisms of CYP3A5*3, CYP3A4*22, CYP3A4*1B, and UGT1A1*60 were examined and their impact on vamorolone PK were found to be negligible in steroid naïve boys with DMD aged 4 to <7 years old utilizing PopPK modeling. The results of this work adds to the evidence that vamorolone exhibits favorable PK profiles (linear and stationary with moderate variability in PK parameters) in the DMD population studied.

Supplementary Material

Supinfo

Acknowledgements

The authors thank the clinical trial participants and their families for their time and effort in participating in the vamorolone clinical trials, and all participating clinical site investigators and their staff.

Funding

Supported by the US Department of Defense, CDMRP award W81XWH-22-1-0668 (Drs. Hoffman, Jusko, and Li). Additional support for the conduct of the vamorolone clinical trials was provided by the following grants: National Institutes of Health, National Institute of Neurological Disorders and Stroke R44NS095423 (Drs Hoffman and Clemens); National Institute of Child Health and Human Development 5U54HD090254 (Dr. van den Anker), and National Institute of Arthritis and Musculoskeletal and Skin Diseases U34AR068616 (Dr. Clemens), and grant agreement number 667078 from the European Commission Horizons 2020 (Dr. Guglieri). Also supported by NIH grant R35-GM131800 (Drs. Jusko and Li).

Conflicts of Interest

E.H., J.V.D.A. and J.M.D. are employees of ReveraGen BioPharma. E.H., J.V.D.A., J.M.D. and J.M.M. hold stock or stock options in ReveraGen BioPharma. U.D. has a consulting history with ReveraGen BioPharma. The following has/had financial support: D.S. Fellowship from the University of Padua; E.P. Cariparo Foundation, Biogen, Roche, Alexion, UCB Biopharma, and Sanofi; L.B. Telethon Foundation, PTC Therapeutics, Edgewise Therapeutics, UCB, Pfizer, and Roche; P.R.C. FDA, Foundation to Eradicate Duchenne, and NS Pharma; M.G. Duchenne UK, Edgewise, PTC Therapeutics, Berkley University, NIH, Pfizer, Dyne, NS Pharma, Italfarmaco, Sarepta, Roche, Novartis, and Antisense Therapeutics; J.V.D.A. Pfizer, J&J, Shionogi, and Eli-Lilly; J.M.D. NIH; J.M.M. Santhera PharmaNIH, NIH NINDS, MycRx, Carawy, Sandos, Evommune, Capacity, Sanofi, Bright Minds, Various Patents, Boca Grande Art Alliance, and Nimbus; U.D. Foundation to Eradicate Duchenne, NIH NINDS, NIH NIAMS, NIH NCATS, NSERC, Lupin Neurosciences, and Iuvo BioScience; E.H. NIH, Santhera Pharmaceuticals, Catalyst, NS Pharma, Various Patents, Foundation to Eradicate Duchenne, C3 Foundation, DuchenneUK, and AGADA BioSciences.

Data Sharing and Data Availability

Data are fully displayed in the manuscript and supplemental files and will be available to qualified individuals upon written request.

References

  • 1.Hoffman EP, Brown RH Jr., Kunkel LM. Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell. 1987;51(6): 919–928. [DOI] [PubMed] [Google Scholar]
  • 2.Duan D, Goemans N, Takeda S, Mercuri E, Aartsma-Rus A. Duchenne muscular dystrophy. Nat Rev Dis Primers. 2021;7(1): 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Mercuri E, Bonnemann CG, Muntoni F. Muscular dystrophies. Lancet. 2019;394(10213): 2025–2038. [DOI] [PubMed] [Google Scholar]
  • 4.Bello L, Gordish-Dressman H, Morgenroth LP, et al. Prednisone/prednisolone and deflazacort regimens in the CINRG Duchenne Natural History Study. Neurology. 2015;85(12): 1048–1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Hoffman EP, Riddle V, Siegler MA, et al. Phase 1 trial of vamorolone, a first-in-class steroid, shows improvements in side effects via biomarkers bridged to clinical outcomes. Steroids. 2018;134: 43–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Liu X, Wang Y, Gutierrez JS, et al. Disruption of a key ligand-H-bond network drives dissociative properties in vamorolone for Duchenne muscular dystrophy treatment. Proc Natl Acad Sci U S A. 2020;117(39): 24285–24293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Heier CR, Yu Q, Fiorillo AA, et al. Vamorolone targets dual nuclear receptors to treat inflammation and dystrophic cardiomyopathy. Life Sci Alliance. 2019;2(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Heier CR, Damsker JM, Yu Q, et al. VBP15, a novel anti-inflammatory and membrane-stabilizer, improves muscular dystrophy without side effects. EMBO Mol Med. 2013;5(10): 1569–1585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Guglieri M, Clemens PR, Perlman SJ, et al. Efficacy and Safety of Vamorolone vs Placebo and Prednisone Among Boys With Duchenne Muscular Dystrophy: A Randomized Clinical Trial. JAMA Neurol. 2022;79(10): 1005–1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Dang U, Damsker JM, Guglieri M, Clemens PR. Efficacy and safety of vamorolone over 48 weeks in boys with Duchenne muscular dystrophy: Randomized controlled trial. Neurology, in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Smith EC, Conklin LS, Hoffman EP, et al. Efficacy and safety of vamorolone in Duchenne muscular dystrophy: An 18-month interim analysis of a non-randomized open-label extension study. PLoS Med. 2020;17(9): e1003222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Mah JK, Clemens PR, Guglieri M, et al. Efficacy and Safety of Vamorolone in Duchenne Muscular Dystrophy: A 30-Month Nonrandomized Controlled Open-Label Extension Trial. JAMA Netw Open. 2022;5(1): e2144178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Kuehl P, Zhang J, Lin Y, et al. Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression. Nat Genet. 2001;27(4): 383–391. [DOI] [PubMed] [Google Scholar]
  • 14.Hesselink DA, Bouamar R, Elens L, van Schaik RH, van Gelder T. The role of pharmacogenetics in the disposition of and response to tacrolimus in solid organ transplantation. Clin Pharmacokinet. 2014;53(2): 123–139. [DOI] [PubMed] [Google Scholar]
  • 15.Tamashiro EY, Felipe CR, Genvigir FDV, et al. Influence of CYP3A4 and CYP3A5 polymorphisms on tacrolimus and sirolimus exposure in stable kidney transplant recipients. Drug Metab Pers Ther. 2017;32(2): 89–95. [DOI] [PubMed] [Google Scholar]
  • 16.Shi WL, Tang HL, Zhai SD. Effects of the CYP3A4*1B Genetic Polymorphism on the Pharmacokinetics of Tacrolimus in Adult Renal Transplant Recipients: A Meta-Analysis. PLoS One. 2015;10(6): e0127995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Albekairy A, Alkatheri A, Fujita S, et al. Cytochrome P450 3A4FNx011B as pharmacogenomic predictor of tacrolimus pharmacokinetics and clinical outcome in the liver transplant recipients. Saudi J Gastroenterol. 2013;19(2): 89–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hannachi I, Ben Fredj N, Chadli Z, et al. Effect of CYP3A4*22 and CYP3A4*1B but not CYP3A5*3 polymorphisms on tacrolimus pharmacokinetic model in Tunisian kidney transplant. Toxicol Appl Pharmacol. 2020;396: 115000. [DOI] [PubMed] [Google Scholar]
  • 19.Innocenti F, Grimsley C, Das S, et al. Haplotype structure of the UDP-glucuronosyltransferase 1A1 promoter in different ethnic groups. Pharmacogenetics. 2002;12(9): 725–733. [DOI] [PubMed] [Google Scholar]
  • 20.Kitagawa C, Ando M, Ando Y, et al. Genetic polymorphism in the phenobarbital-responsive enhancer module of the UDP-glucuronosyltransferase 1A1 gene and irinotecan toxicity. Pharmacogenet Genomics. 2005;15(1): 35–41. [DOI] [PubMed] [Google Scholar]
  • 21.Lee SJ, Jusko WJ, Salaita CG, et al. Reduced methylprednisolone clearance causing prolonged pharmacodynamics in a healthy subject was not associated with CYP3A5*3 allele or a change in diet composition. J Clin Pharmacol. 2006;46(5): 515–526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Wang D, Guo Y, Wrighton SA, Cooke GE, Sadee W. Intronic polymorphism in CYP3A4 affects hepatic expression and response to statin drugs. Pharmacogenomics J. 2011;11(4): 274–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Elalem EG, Jelani M, Khedr A, et al. Association of cytochromes P450 3A4*22 and 3A5*3 genotypes and polymorphism with response to simvastatin in hypercholesterolemia patients. PLoS One. 2022;17(7): e0260824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Marsh S, Hoskins JM. Irinotecan pharmacogenomics. Pharmacogenomics. 2010;11(7): 1003–1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.van Gelder T, van Schaik RH, Hesselink DA. Pharmacogenetics and immunosuppressive drugs in solid organ transplantation. Nat Rev Nephrol. 2014;10(12): 725–731. [DOI] [PubMed] [Google Scholar]
  • 26.Mavroudis PD, van den Anker J, Conklin LS, et al. Population Pharmacokinetics of Vamorolone (VBP15) in Healthy Men and Boys With Duchenne Muscular Dystrophy. J Clin Pharmacol. 2019;59(7): 979–988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Conklin LS, Damsker JM, Hoffman EP, et al. Phase IIa trial in Duchenne muscular dystrophy shows vamorolone is a first-in-class dissociative steroidal anti-inflammatory drug. Pharmacol Res. 2018;136: 140–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Williams JA, Ring BJ, Cantrell VE, et al. Comparative metabolic capabilities of CYP3A4, CYP3A5, and CYP3A7. Drug Metab Dispos. 2002;30(8): 883–891. [DOI] [PubMed] [Google Scholar]
  • 29.Hernandez S, Aquilante C, Deininger K, et al. Composite CYP3A (CYP3A4 and CYP3A5) phenotypes and influences on tacrolimus dose adjusted concentration in adult heart transplant recipients. Res Sq. 2023. [DOI] [PubMed] [Google Scholar]
  • 30.Kitzmiller JP, Sullivan DM, Phelps MA, Wang D, Sadee W. CYP3A4/5 combined genotype analysis for predicting statin dose requirement for optimal lipid control. Drug Metabol Drug Interact. 2013;28(1): 59–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Sanchez Spitman AB, Moes D, Gelderblom H, Dezentje VO, Swen JJ, Guchelaar HJ. Effect of CYP3A4*22, CYP3A5*3, and CYP3A combined genotypes on tamoxifen metabolism. Eur J Clin Pharmacol. 2017;73(12): 1589–1598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Jusko WJ. Use of pharmacokinetic data below lower limit of quantitation values. Pharm Res. 2012;29(9): 2628–2631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Keizer RJ, Jansen RS, Rosing H, et al. Incorporation of concentration data below the limit of quantification in population pharmacokinetic analyses. Pharmacol Res Perspect. 2015;3(2): e00131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Elens L, van Gelder T, Hesselink DA, Haufroid V, van Schaik RH. CYP3A4*22: promising newly identified CYP3A4 variant allele for personalizing pharmacotherapy. Pharmacogenomics. 2013;14(1): 47–62. [DOI] [PubMed] [Google Scholar]
  • 35.Hiratsuka M, Takekuma Y, Endo N, et al. Allele and genotype frequencies of CYP2B6 and CYP3A5 in the Japanese population. Eur J Clin Pharmacol. 2002;58(6): 417–421. [DOI] [PubMed] [Google Scholar]
  • 36.Wang D, Sadee W. CYP3A4 intronic SNP rs35599367 (CYP3A4*22) alters RNA splicing. Pharmacogenet Genomics. 2016;26(1): 40–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Borucki K, Weikert C, Fisher E, et al. Haplotypes in the UGT1A1 gene and their role as genetic determinants of bilirubin concentration in healthy German volunteers. Clin Biochem. 2009;42(16–17): 1635–1641. [DOI] [PubMed] [Google Scholar]
  • 38.Anderson BJ, Holford NH. Mechanism-based concepts of size and maturity in pharmacokinetics. Annu Rev Pharmacol Toxicol. 2008;48: 303–332. [DOI] [PubMed] [Google Scholar]
  • 39.Li X, Conklin LS, van den Anker J, Hoffman EP, Clemens PR, Jusko WJ. Exposure-Response Analysis of Vamorolone (VBP15) in Boys With Duchenne Muscular Dystrophy. J Clin Pharmacol. 2020;60(10): 1385–1396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ji Y, Skierka JM, Blommel JH, et al. Preemptive Pharmacogenomic Testing for Precision Medicine: A Comprehensive Analysis of Five Actionable Pharmacogenomic Genes Using Next-Generation DNA Sequencing and a Customized CYP2D6 Genotyping Cascade. J Mol Diagn. 2016;18(3): 438–445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Matey ET, Ragan AK, Oyen LJ, et al. Nine-gene pharmacogenomics profile service: The Mayo Clinic experience. Pharmacogenomics J. 2022;22(1): 69–74. [DOI] [PubMed] [Google Scholar]
  • 42.Vaillancourt J, Turcotte V, Caron P, et al. Glucuronidation of Abiraterone and Its Pharmacologically Active Metabolites by UGT1A4, Influence of Polymorphic Variants and Their Potential as Inhibitors of Steroid Glucuronidation. Drug Metab Dispos. 2020;48(2): 75–84. [DOI] [PubMed] [Google Scholar]
  • 43.Fujita K, Sparreboom A. Pharmacogenetics of irinotecan disposition and toxicity: a review. Curr Clin Pharmacol. 2010;5(3): 209–217. [DOI] [PubMed] [Google Scholar]
  • 44.Liu X, Cheng D, Kuang Q, Liu G, Xu W. Association of UGT1A1*28 polymorphisms with irinotecan-induced toxicities in colorectal cancer: a meta-analysis in Caucasians. Pharmacogenomics J. 2014;14(2): 120–129. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supinfo

Data Availability Statement

Data are fully displayed in the manuscript and supplemental files and will be available to qualified individuals upon written request.

RESOURCES