Abstract
To evaluate the relationship between exposure of apalutamide and its active metabolite, N-desmethyl-apalutamide, and selected clinical efficacy and safety parameters in men with high-risk nonmetastatic castration-resistant prostate cancer.
An exploratory exposure–response analysis was undertaken using data from the 1,207 patients (806 apalutamide and 401 placebo) enrolled in the SPARTAN study, including those who had undergone dose reductions and dose interruptions. Univariate and multivariate Cox regression models evaluated the relationships between apalutamide and N-desmethyl-apalutamide exposure, expressed as area under the concentration–time curve at steady state, and metastasis-free survival (MFS). Univariate and multivariate logistic regression models assessed the relationship between apalutamide and N-desmethyl-apalutamide exposure and common treatment-emergent adverse events including fatigue, fall, skin rash, weight loss, and arthralgia.
A total of 21% of patients in the apalutamide arm experienced dose reductions diminishing the average daily dose to 209 mg instead of 240 mg. Within the relatively narrow exposure range, no statistically significant relationship was found between MFS and apalutamide and N-desmethyl-apalutamide exposure. Within apalutamide-treated subjects, skin rash and weight loss had a statistically significant association with higher apalutamide exposure.
The use of apalutamide at the recommended dose of 240 mg once daily provided a similar delay in metastases across the SPARTAN patient population, regardless of exposure. The exploratory exposure–safety analysis supports dose reductions in patients experiencing adverse events.
This article is featured in Highlights of This Issue, p. 4427
Translational Relevance
Apalutamide is an orally administered selective androgen receptor inhibitor, which was the first approved therapy in the United States and European Union for treatment of patients with nonmetastatic castration-resistant prostate cancer (nmCRPC) based on the pivotal phase III SPARTAN study. Differences in apalutamide and N-desmethyl-apalutamide exposures after the administration of 240 mg once daily do not result in clinically relevant differences in metastasis-free survival. On the basis of the relatively narrow exposure range, apalutamide at 240 mg once daily provides similarly efficacious exposure in most patients with nmCRPC. Skin rash and weight loss were identified to have a statistically significant association with apalutamide exposure, with patients who had higher exposures being more likely to experience skin rash or weight loss. Because of the exposure–response relationship with these adverse events, patients who experience them may benefit from a dose reduction without compromising efficacy.
Introduction
Apalutamide is an orally administered, specific inhibitor of the androgen receptor (AR), which is approved in the United States and European Union for the treatment of patients with nonmetastatic castration-resistant prostate cancer (nmCRPC). Apalutamide acts via inhibition of AR nuclear translocation and of AR binding to androgen response elements (1). The efficacy and safety of apalutamide versus placebo in men with high-risk nmCRPC have been evaluated in a multicenter phase III study (ARN-509-003; SPARTAN; NCT01946204), as described previously (2). Addition of apalutamide 240 mg once daily to androgen deprivation therapy (ADT) resulted in a 72% decrease in the risk of developing metastatic disease or death [HR = 0.28, 95% confidence interval (CI), 0.23–0.35; P < 0.001], compared with placebo plus ADT (3). This study also confirmed the tolerability and acceptable safety profile of apalutamide observed in earlier studies.
An understanding of the relationship between dose, exposure, and response is important to assess the benefit–risk profile and individualize the appropriate dose (4). This exploratory exposure–response (ER) analysis was undertaken to explore the relationships between exposure to apalutamide and its active metabolite, N-desmethyl-apalutamide, and selected clinical efficacy and safety endpoints.
Patients and Methods
Clinical study design and patient population
The ER analysis was performed with data collected from all patients enrolled in the phase 3 SPARTAN study (2, 3). Patients with nmCRPC were randomized in a 2:1 ratio to receive apalutamide 240 mg or matched placebo administered orally on a continuous daily dosing regimen. After the first dose on cycle 1 day 1 0.5–4 hours postdose, pharmacokinetic samples were collected while predose samples were available for cycles 2, 3, 6, 12, 18, and 24. All patients who not had bilateral orchiectomy had to continue medical ADT with a gonadotropin-releasing hormone (GnRH) agonist or antagonist. Two formulations of apalutamide were used, leading to dosing of 240 mg first as (8 × 30 mg) capsules then as (4 × 60 mg) tablets, with no previously identified clinically relevant exposure differences between these two formulations (5).
For patients who experienced treatment-emergent adverse events (TEAE), dose interruptions and/or reductions were permitted if study discontinuation criteria had not been met. When patients experienced an AE of grade 3 or higher, apalutamide was to be held until symptoms improved to grade 1 or lower. Reinitiation of apalutamide therapy was allowed at the same or lower dose [maximum of 2 dose level reductions (240 mg–180 mg; 180 mg–120 mg)].
Metastasis-free survival (MFS) was the primary endpoint of the SPARTAN trial, and was defined as the time from randomization to first evidence of detectable bone or soft-tissue distant metastasis as identified by independent central review or death due to any cause (whichever occurred earlier). The MFS data for patients without metastasis or death were censored on the date of the last tumor assessment (or, if no tumor assessment was performed after the baseline visit, at the date of randomization). Patients continued with study treatment until blinded independent central review confirmed the detection of distant metastatic disease, development of unacceptable toxicity, or withdrawal of consent.
SPARTAN was conducted in accordance with principles for human experimentation as defined in the Declaration of Helsinki and was approved by the Human Investigational Review Board of each study center and by the Competent Authority of each country. Written informed consent was obtained from each patient before enrollment in the study, after the patient was advised of the potential risks and benefits, as well as the investigational nature of the study.
Statistical analysis
Exposure metrics for apalutamide, N-desmethyl-apalutamide, and active moiety
The observed pharmacokinetic data was used as input in the population pharmacokinetic model to derive model-based exposure metrics (6). Individual steady-state exposure metrics [area under the concentration curve after 24 hours (AUC0–24h,ss), predose concentration (Cmin,ss), and maximum concentration (Cmax,ss)] were computed at steady state for apalutamide and N-desmethyl-apalutamide. The active moiety was calculated as the sum of apalutamide and N-desmethyl-apalutamide exposures weighted by their relative potency, considering that N-desmethyl-apalutamide exhibits one-third of the activity of apalutamide in an in vitro transcriptional reporter assay (7). After 4 weeks of treatment, more than 95% of patients had reached steady-state exposure of apalutamide and N-desmethyl-apalutamide, well before the first visit scheduled for MFS assessment at 16 weeks after start of treatment. The exposure metrics were derived on the basis of the maximum a posteriori estimates of the individual pharmacokinetic model parameter for apalutamide and N-desmethyl-apalutamide, which were obtained from the plasma concentrations collected in the SPARTAN study and the reference population pharmacokinetic model, described in a separate article (6). Given the option of dose modifications for AEs, the average taken daily dose up to the event of interest (either MFS or first occurrence of AE) was used to compute the exposure metrics. The exposure metrics (AUC0–24h,ss, Cmin,ss, and Cmax,ss), adjusted by the average daily dose, reflect the average exposure of a subject up to the time of the first event of interest, considering possible dose reductions, dose interruptions, or missed doses prior to first time of the event.
Scatterplots and linear regression analyses were performed between individual steady-state AUC0–24h,ss, Cmin,ss, and Cmax,ss to select the exposure metrics for the ER analysis. If a variance inflation factor higher than 5 (r2 > 0.80) was detected among the exposure metrics, the ER analysis was performed with the steady-state AUC0–24h,ss only; if not, Cmin,ss and Cmax,ss were also considered. The assessment of the correlation between exposure metrics was performed separately for apalutamide and N-desmethyl-apalutamide.
Efficacy endpoint data
MFS was the primary endpoint included in the ER analysis. The effects of the stratification factors prostate-specific antigen doubling time (PSADT; >6 months vs. ≤6 months), the presence of locoregional disease (N0 vs. N1), and the coadministration of a bone-sparing agent (yes vs. no) on MFS were considered in the efficacy analysis (3). Because age and Eastern Cooperative Oncology Group (ECOG) performance status were identified to have a statistically significant (P < 0.05) association with overall survival in SPARTAN, these prognostic factors were also included in the exposure–MFS analysis.
Safety data
The exposure–safety analysis focused on TEAEs of clinical relevance (i.e., classified as TEAEs of special interest or related to the drug), with occurrence higher than 10%, at any grade. On the basis of these criteria, fatigue (30.4% apalutamide vs. 21.1% placebo), falls (15.6% apalutamide vs. 9.0% placebo), skin rash (23.8% apalutamide vs. 5.5% placebo), weight loss (16.1% apalutamide vs. 6.3% placebo), and arthralgia (15.9% apalutamide vs. 7.5% placebo) were included in the exposure safety analysis.
ER analysis
Software
Pharmacokinetic analysis and the maximum a posteriori estimates of the individual pharmacokinetic model parameters for apalutamide and N-desmethyl-apalutamide were obtained using NONMEM software (Icon Development Solutions; ref. 8). The data management, diagnostic graphics, postprocessing of NONMEM analysis results, and statistical analysis were carried out using R Project for Statistical Computing, Version 3.4.1 or higher for Windows (Comprehensive R Network, http://cran.r-project.org/; ref. 9).
Exposure–efficacy analysis
The relationships between apalutamide, N-desmethyl-apalutamide, and active moiety exposure and MFS were first evaluated using Kaplan–Meier curves of MFS by quartiles of exposure. Placebo was included as a separate group. In addition, a multivariate Cox regression analysis for apalutamide and N-desmethyl-apalutamide exposure was performed; covariates included were treatment arm (apalutamide vs. placebo), the prespecified stratification factors [PSADT (≤ 6 months vs. > 6 months), presence of locoregional disease (N1 vs. N0), and bone-sparing agent used (yes vs. no)], and prognostic factors including age and ECOG performance status (1 vs. 0).
Patients randomized to the placebo group had apalutamide and N-desmethyl-apalutamide exposure assigned to zero. The impact of apalutamide and N-desmethyl-apalutamide exposure on MFS, after adjusting for the covariates, was assessed by the HR and its 95% CI. The P values as well as the change in −2 loglikelihood (LL) after the inclusion of each exposure metric were used for model comparison.
Exposure–safety analysis
The selected safety endpoints (fatigue, fall, skin rash, weight loss, and arthralgia) were dichotomized into presence or absence of any TEAEs regardless of grade. Patients with multiple occurrences of events were only counted once, when the first event was experienced. As half of fractures were preceded by falls, and causality was hard to determine, falls (regardless if this was followed by a fracture or not), but not fractures, were included in the exposure–safety analysis.
Univariate and multivariate logistic regression analysis were conducted to assess any possible association between treatment (apalutamide vs. placebo) and apalutamide and N-desmethyl-apalutamide exposure on the selected safety endpoints. The corresponding odds ratio (OR), 95% CI, χ², and P values were calculated. The P values as well as the change in −2LL were used for model comparison. In addition, a sensitivity analysis was conducted by including and excluding placebo patients in the exposure–safety analysis.
The ER analyses described above were exploratory and hypothesis-generating and were performed under the assumption that there was a sufficient number of events for a meaningful analysis. P values lower than 0.05 were considered statistically significant. No correction for multiple statistical testing was implemented.
Results
Analysis dataset
Data from all 1,207 randomized patients (806 in the apalutamide group and 401 in the placebo group) were included in the exposure–safety analysis while 1 patient from the apalutamide treatment group was excluded in the exposure–efficacy analysis because of missing values in a significant prognostic factor (ECOG performance status).
Apalutamide and N-desmethyl-apalutamide pharmacokinetic exposure
Consistent with the long half-life of apalutamide, pharmacokinetic data showed that 240 mg once-daily dosing led to a relatively constant apalutamide plasma concentration during the dosing interval at steady state, with limited peak-to-trough fluctuation ranging from a mean Cmax,ss value of 5.81 μg/mL (range: 1.46–13.9) to a mean Cmin,ss value of 4.24 μg/mL (range: 0.60–11.1). The peak-to-trough fluctuation of N-desmethyl-apalutamide plasma concentration at steady state is negligible, with relatively constant N-desmethyl-apalutamide plasma concentrations over the dosing interval at a plasma concentration average value of 6.3 μg/mL (range: 1.8–12.5). The apalutamide and N-desmethyl-apalutamide exposures, measured as AUC0–24h,ss, were 115 (range: 19.8–291) and 152 (range: 44.1–299) μg·hour/mL, respectively. The interindividual variability in apalutamide and N-desmethyl apalutamide exposure, expressed as coefficient of variation of AUC0–24h,ss was considered low to moderate (≤27%). In addition, following once-daily oral apalutamide administration of 240 mg, apalutamide and N-desmethyl-apalutamide systemic exposure showed a 5.3- and 85.2-fold accumulation (calculated, based on AUC) in plasma, respectively.
The correlations between the individual exposure metrics at steady state (AUC0–24h,ss, Cmin,ss, Cmax,ss) for apalutamide and N-desmethyl-apalutamide were high (r2 > 0.95) between all parameters. Because of this high correlation, AUC0–24h,ss was selected as the unique exposure metric to conduct the ER analysis for efficacy and safety endpoints. In contrast, the correlation between AUC0–24h,ss of apalutamide and N-desmethyl-apalutamide was estimated to be below 0.80 (r2 = 0.646, P < 0.001), and therefore both apalutamide and N-desmethyl-apalutamide AUC0–24h,ss were evaluated in the ER analysis, and the collinearity due to these two variables was further monitored.
Given the dose modification rate in the phase III SPARTAN study (21% dose reductions in the apalutamide group and 15% in the placebo group), the average daily dose (instead of the start dose of 240 mg) up to the event of interest (either MFS or first occurrence of TEAEs) was used to compute the exposure metrics. An average daily dose of 209 mg (apalutamide group) versus 222 mg (placebo group) in patients with one or more dose reductions was observed (Table 1); however, the relative difference between the derived AUC0–24h,ss from the average daily dose and the dose-normalized 240 mg (assuming 100% treatment adherence) was relatively small (<13%). In addition, predicted dose-normalized AUC0–24h,ss was similar in patients with or without dose reductions.
Summary statistics for the individual exposure metric (AUC0–24h,ss) for apalutamide and N-desmethyl-apalutamide, expressed as mean (CV%).
Treatment group . | Dose reduction . | Percentage of subjects (% of totala) . | Average daily dose, mg (CV%)b . | Apalutamide AUC0–24h,ss μg × h/mL (CV%) . | N-desmethyl-apalutamide AUC0–24h,ss, μg × h/mL (CV%) . |
---|---|---|---|---|---|
Placebo | No | 85% | 235 (5.4) | — | — |
Yes | 15% | 222 (11.5) | — | — | |
Apalutamide | No | 79% | 234 (5.5) | 113 (23) | 149 (18) |
Yes | 21% | 209 (17.1) | 112 (25.2) | 148 (18) |
Treatment group . | Dose reduction . | Percentage of subjects (% of totala) . | Average daily dose, mg (CV%)b . | Apalutamide AUC0–24h,ss μg × h/mL (CV%) . | N-desmethyl-apalutamide AUC0–24h,ss, μg × h/mL (CV%) . |
---|---|---|---|---|---|
Placebo | No | 85% | 235 (5.4) | — | — |
Yes | 15% | 222 (11.5) | — | — | |
Apalutamide | No | 79% | 234 (5.5) | 113 (23) | 149 (18) |
Yes | 21% | 209 (17.1) | 112 (25.2) | 148 (18) |
Abbreviation: CV, coefficient of variation.
aFor placebo patients, the average daily dose is the equivalent in placebo.
bTotal number of patients is those with exposure metrics and average daily dose calculated (n = 771 and n = 384 for apalutamide and placebo treatment groups, respectively).
Exposure–efficacy analysis
A summary of the prognostic factor distribution across treatment groups and quartiles of apalutamide and N-desmethyl-apalutamide exposure is presented in Table 2. These results suggest that the relevant prognostic factors were balanced among the treatment and exposure quartiles evaluated, and the potential imbalances that may exist should be controlled by the multivariate analysis. The Kaplan–Meier curves of MFS stratified by the quartiles of apalutamide, N-desmethyl-apalutamide, and the active moiety AUC0–24h,ss are shown in Fig. 1. In all pairwise comparisons of the exposure subgroups in the apalutamide group versus the placebo group, a statistically significant (P < 0.0001) increase in MFS was observed for the apalutamide, N-desmethyl-apalutamide, and active moiety exposure subgroups.
Distribution of the prognostic factors across treatment arms and quartiles of exposure for apalutamide and N-desmethyl-apalutamide.
. | . | N . | PSADT (≤6 months) . | Bone-sparing agent use . | Loco-regional disease (N1) . | Age (< 65 years) . | Age (65–75 years) . | Age (>75 years) . | ECOG PS (Grade 1) . |
---|---|---|---|---|---|---|---|---|---|
Treatment group | Placebo | 400 | 283 (70.8) | 39 (9.75) | 65 (16.3) | 190 (47.5) | 43 (10.8) | 167 (41.8) | 89 (22.3) |
Apalutamide | 806 | 576 (71.5) | 82 (10.2) | 133 (16.5) | 341 (42.3) | 106 (13.1) | 359 (44.5) | 183 (22.7) | |
Apalutamide | Q1 | 202 | 140 (69.3) | 20 (9.90) | 39 (19.3) | 92 (45.5) | 34 (16.8) | 76 (37.6) | 49 (24.3) |
Q2 | 201 | 139 (69.2) | 19 (9.45) | 30 (14.9) | 105 (52.2) | 23 (11.4) | 73 (36.3) | 40 (19.9) | |
Q3 | 201 | 162 (80.6) | 18 (8.96) | 33 (16.4) | 80 (39.8) | 34 (16.9) | 87 (43.3) | 49 (24.4) | |
Q4 | 202 | 135 (66.8) | 25 (12.4) | 31 (15.3) | 64 (31.7) | 15 (7.43) | 123 (60.9) | 45 (22.3) | |
N-desmethyl-apalutamide | Q1 | 202 | 139 (68.8) | 17 (8.42) | 37 (18.3) | 97 (48.0) | 29 (14.4) | 76 (37.6) | 58 (28.7) |
Q2 | 201 | 151 (75.1) | 21 (10.5) | 35 (17.4) | 91 (45.3) | 36 (17.9) | 74 (36.8) | 43 (21.4) | |
Q3 | 201 | 141 (70.1) | 23 (11.4) | 25 (12.4) | 84 (41.8) | 16 (7.96) | 101 (50.2) | 44 (21.9) | |
Q4 | 202 | 145 (71.8) | 21 (10.4) | 36 (17.8) | 69 (34.7) | 25 (12.4) | 108 (53.5) | 38 (18.8) |
. | . | N . | PSADT (≤6 months) . | Bone-sparing agent use . | Loco-regional disease (N1) . | Age (< 65 years) . | Age (65–75 years) . | Age (>75 years) . | ECOG PS (Grade 1) . |
---|---|---|---|---|---|---|---|---|---|
Treatment group | Placebo | 400 | 283 (70.8) | 39 (9.75) | 65 (16.3) | 190 (47.5) | 43 (10.8) | 167 (41.8) | 89 (22.3) |
Apalutamide | 806 | 576 (71.5) | 82 (10.2) | 133 (16.5) | 341 (42.3) | 106 (13.1) | 359 (44.5) | 183 (22.7) | |
Apalutamide | Q1 | 202 | 140 (69.3) | 20 (9.90) | 39 (19.3) | 92 (45.5) | 34 (16.8) | 76 (37.6) | 49 (24.3) |
Q2 | 201 | 139 (69.2) | 19 (9.45) | 30 (14.9) | 105 (52.2) | 23 (11.4) | 73 (36.3) | 40 (19.9) | |
Q3 | 201 | 162 (80.6) | 18 (8.96) | 33 (16.4) | 80 (39.8) | 34 (16.9) | 87 (43.3) | 49 (24.4) | |
Q4 | 202 | 135 (66.8) | 25 (12.4) | 31 (15.3) | 64 (31.7) | 15 (7.43) | 123 (60.9) | 45 (22.3) | |
N-desmethyl-apalutamide | Q1 | 202 | 139 (68.8) | 17 (8.42) | 37 (18.3) | 97 (48.0) | 29 (14.4) | 76 (37.6) | 58 (28.7) |
Q2 | 201 | 151 (75.1) | 21 (10.5) | 35 (17.4) | 91 (45.3) | 36 (17.9) | 74 (36.8) | 43 (21.4) | |
Q3 | 201 | 141 (70.1) | 23 (11.4) | 25 (12.4) | 84 (41.8) | 16 (7.96) | 101 (50.2) | 44 (21.9) | |
Q4 | 202 | 145 (71.8) | 21 (10.4) | 36 (17.8) | 69 (34.7) | 25 (12.4) | 108 (53.5) | 38 (18.8) |
Note: Values are expressed as number of patients (proportion of patients).
Abbreviations: ECOG PS, Eastern Cooperative Oncology Group performance status; PSADT, prostate-specific antigen doubling time.
Kaplan–Meier plot for MFS as a function of placebo and the lowest exposure quartile (Q1) to highest exposure quartile (Q4) of apalutamide (A), N-desmethyl apalutamide (B), and the active moiety (C) plasma steady-state AUC0–24h,ss. Active moiety: sum of apalutamide and N-desmethyl-apalutamide exposures weighted by their relative potency.
Kaplan–Meier plot for MFS as a function of placebo and the lowest exposure quartile (Q1) to highest exposure quartile (Q4) of apalutamide (A), N-desmethyl apalutamide (B), and the active moiety (C) plasma steady-state AUC0–24h,ss. Active moiety: sum of apalutamide and N-desmethyl-apalutamide exposures weighted by their relative potency.
However, after excluding the patients randomized to placebo, none of the other pairwise comparisons among the exposure subgroups for apalutamide, N-desmethyl-apalutamide, and active moiety were identified as statistically significant. These findings suggest that all exposure levels in patients treated with apalutamide experienced benefit. Because the results of the active moiety are comparable with the results of apalutamide and N-desmethyl-apalutamide, only apalutamide and N-desmethyl-apalutamide were carried forward to the multivariate Cox regression analysis.
The multivariate Cox regression analysis found a statistically significant association between apalutamide treatment, PSADT, presence of locoregional disease, and ECOG performance status on MFS. The effect of apalutamide on MFS (HR = 0.30; 95% CI, 0.24–0.36) was found to be similar after adjusting for the prognostic factors described previously (ref. 3; HR = 0.28, 95% CI, 0.23–0.35), suggesting that inclusion of the prognostic factors in the model did not produce any treatment-effect modification, and consequently the prognostic factors can be considered independently associated with MFS.
The addition of N-desmethyl-apalutamide AUC0–24h,ss in the model on top of apalutamide AUC0–24h,ss led to a substantial change of the treatment effect and its uncertainty (HR = 0.205, 95% CI, 0.097–0.435), which may be explained by the collinearity between prognostic factors, apalutamide treatment, and the exposure variables. A multivariate Cox regression analysis evaluated in the 806 patients treated with apalutamide (i.e., excluding placebo patients), showed a decrease in the collinearity and confirmed the results of the univariate analysis, that neither apalutamide AUC0–24h,ss nor N-desmethyl-apalutamide AUC0–24h,ss were statistically associated with MFS.
Exposure-safety analysis
A summary of the incidence of the treatment-emergent events for placebo, apalutamide, and the quartiles of apalutamide and N-desmethyl-apalutamide exposure is presented in Table 3.
Summary of the incidence of treatment-emergent adverse events for the placebo group and the apalutamide group and the quartiles of exposure for apalutamide and N-desmethyl-apalutamide in the SPARTAN population.
. | . | N . | Fatigue (%) . | N . | Fall (%) . | N . | Skin rash (%) . | N . | Weight loss (%) . | N . | Arthralgia (%) . |
---|---|---|---|---|---|---|---|---|---|---|---|
Treatment group | Placebo | 401 | 84 (20.9) | 401 | 36 (8.98) | 401 | 22 (5.49) | 401 | 25 (6.23) | 401 | 30 (7.48) |
Apalutamide | 806 | 244 (30.3) | 806 | 125 (15.5) | 806 | 191 (23.7) | 806 | 129 (16.0) | 806 | 128 (15.9) | |
Apalutamide | Q1 | 202 | 55 (27.2) | 202 | 29 (14.4) | 202 | 29 (14.4) | 202 | 14 (6.93) | 202 | 24 (11.9) |
Q2 | 201 | 63 (31.3) | 201 | 30 (14.9) | 201 | 51 (25.4) | 201 | 32 (15.9) | 201 | 31 (15.4) | |
Q3 | 201 | 63 (31.3) | 201 | 32 (15.9) | 201 | 52 (25.9) | 201 | 31 (15.4) | 201 | 39 (19.4) | |
Q4 | 202 | 36 (31.2) | 202 | 34 (16.8) | 202 | 59 (29.2) | 202 | 52 (25.7) | 202 | 34 (16.8) | |
N-desmethyl-apalutamide | Q1 | 202 | 49 (24.3) | 202 | 29 (14.4) | 202 | 26 (12.9) | 199 | 17 (8.54) | 199 | 18 (9.05) |
Q2 | 201 | 64 (31.8) | 201 | 34 (16.9) | 201 | 43 (21.4) | 201 | 23 (11.4) | 201 | 40 (19.9) | |
Q3 | 201 | 68 (33.8) | 209 | 31 (14.8) | 201 | 56 (27.9) | 208 | 42 (20.2) | 205 | 33 (16.1) | |
Q4 | 202 | 63 (31.2) | 194 | 31 (16.0) | 202 | 66 (32.7) | 195 | 47 (24.1) | 198 | 37 (18.7) |
. | . | N . | Fatigue (%) . | N . | Fall (%) . | N . | Skin rash (%) . | N . | Weight loss (%) . | N . | Arthralgia (%) . |
---|---|---|---|---|---|---|---|---|---|---|---|
Treatment group | Placebo | 401 | 84 (20.9) | 401 | 36 (8.98) | 401 | 22 (5.49) | 401 | 25 (6.23) | 401 | 30 (7.48) |
Apalutamide | 806 | 244 (30.3) | 806 | 125 (15.5) | 806 | 191 (23.7) | 806 | 129 (16.0) | 806 | 128 (15.9) | |
Apalutamide | Q1 | 202 | 55 (27.2) | 202 | 29 (14.4) | 202 | 29 (14.4) | 202 | 14 (6.93) | 202 | 24 (11.9) |
Q2 | 201 | 63 (31.3) | 201 | 30 (14.9) | 201 | 51 (25.4) | 201 | 32 (15.9) | 201 | 31 (15.4) | |
Q3 | 201 | 63 (31.3) | 201 | 32 (15.9) | 201 | 52 (25.9) | 201 | 31 (15.4) | 201 | 39 (19.4) | |
Q4 | 202 | 36 (31.2) | 202 | 34 (16.8) | 202 | 59 (29.2) | 202 | 52 (25.7) | 202 | 34 (16.8) | |
N-desmethyl-apalutamide | Q1 | 202 | 49 (24.3) | 202 | 29 (14.4) | 202 | 26 (12.9) | 199 | 17 (8.54) | 199 | 18 (9.05) |
Q2 | 201 | 64 (31.8) | 201 | 34 (16.9) | 201 | 43 (21.4) | 201 | 23 (11.4) | 201 | 40 (19.9) | |
Q3 | 201 | 68 (33.8) | 209 | 31 (14.8) | 201 | 56 (27.9) | 208 | 42 (20.2) | 205 | 33 (16.1) | |
Q4 | 202 | 63 (31.2) | 194 | 31 (16.0) | 202 | 66 (32.7) | 195 | 47 (24.1) | 198 | 37 (18.7) |
The univariate logistic regression analysis for the treatment effect showed that the probability of experiencing one of the described TEAEs was statistically significantly higher in the apalutamide group than the placebo group. Moreover, the probability of experiencing one of the described TEAEs significantly increases as apalutamide AUC0–24h,ss increases (Fig. 2). The same effect was observed for N-desmethyl-apalutamide AUC0–24h,ss (Fig. 3).
Logistic regression representing the probability of experiencing fatigue, fall, skin rash, weight loss, and arthralgia as function of apalutamide steady-state AUC0–24h,ss. The upper and lower open circles represent the presence or absence of a given treatment-emergent event across the range of the predicted apalutamide AUC0–24h,ss exposure, respectively. The dots depict the observed incidence for the placebo and the quartiles of exposure for the apalutamide group, respectively, whereas the corresponding vertical bars represent the exact 95% CI. Finally, the middle line and its corresponding shaded area represent model-based exposure–safety relationship and the 95% CI, respectively.
Logistic regression representing the probability of experiencing fatigue, fall, skin rash, weight loss, and arthralgia as function of apalutamide steady-state AUC0–24h,ss. The upper and lower open circles represent the presence or absence of a given treatment-emergent event across the range of the predicted apalutamide AUC0–24h,ss exposure, respectively. The dots depict the observed incidence for the placebo and the quartiles of exposure for the apalutamide group, respectively, whereas the corresponding vertical bars represent the exact 95% CI. Finally, the middle line and its corresponding shaded area represent model-based exposure–safety relationship and the 95% CI, respectively.
Logistic regression representing the probability of experiencing fatigue, fall, skin rash, weight loss, and arthralgia as function of N-desmethyl-apalutamide steady-state AUC0–24h,ss. The upper and lower open circles represent the presence or absence of a given treatment-emergent event across the range of the predicted N-desmethyl-apalutamide AUC0–24h,ss exposure, respectively. The dots depict the observed incidence for the placebo and the quartiles of exposure for the N-desmethyl-apalutamide group, respectively, whereas the corresponding vertical bars represent the exact 95% CI. Finally, the middle line and its corresponding shaded area represent model-based exposure–safety relationship and the 95% CI, respectively.
Logistic regression representing the probability of experiencing fatigue, fall, skin rash, weight loss, and arthralgia as function of N-desmethyl-apalutamide steady-state AUC0–24h,ss. The upper and lower open circles represent the presence or absence of a given treatment-emergent event across the range of the predicted N-desmethyl-apalutamide AUC0–24h,ss exposure, respectively. The dots depict the observed incidence for the placebo and the quartiles of exposure for the N-desmethyl-apalutamide group, respectively, whereas the corresponding vertical bars represent the exact 95% CI. Finally, the middle line and its corresponding shaded area represent model-based exposure–safety relationship and the 95% CI, respectively.
The treatment effect was included together with apalutamide or N-desmethyl-apalutamide exposure in the multivariate regression analysis. On the basis of the 95% CI of the OR, the contribution of apalutamide or N-desmethyl-apalutamide AUC0–24h,ss, when adjusted by treatment effect differences, was significant for the probability of experiencing skin rash and weight loss, but not for the probability of experiencing fatigue, fall, or arthralgia at any grade.
The results of the multivariate logistic regression analysis were further confirmed in a univariate exposure–safety analysis in patients treated with apalutamide, which showed a statistically significant association between apalutamide treatment and skin rash and weight loss.
Discussion
In the phase III SPARTAN study, the addition of apalutamide to ADT resulted in significant improvement over placebo plus ADT in the primary endpoint, MFS, as well as in other secondary endpoints, including time to symptomatic progression. However, apalutamide plus ADT was also associated with higher rates of skin rash, fatigue, arthralgia, weight loss, falls, and fracture (3). Some of these adverse events led to dose adjustments, which resulted in decreased drug exposure, and which provided the opportunity to undertake exploratory analyses to elucidate the relationship (if any) between apalutamide and N-desmethyl-apalutamide exposure and efficacy (MFS) as well as adverse drug reactions of special interest associated with the use of apalutamide.
The observed high correlation between the three exposure parameters selected (Cmax,ss, Cmin,ss, and AUC0–24h,ss) can be expected considering that the apalutamide and N-desmethyl-apalutamide terminal half-life is substantially longer than the dosing interval; therefore, fluctuations in drug concentration during the dosing interval are low and drug concentrations collected at steady state are highly correlated with AUC. It is useful to correlate long-term drug effects with steady-state AUC0–24h (as was done in this analysis), because AUC0–24h,ss represents the average drug concentration over a longer time period following multiple dosing (10–12).
The 240 mg dose of apalutamide used in SPARTAN was selected on the basis of phase I data as well as on the inhibition of uptake of fluoro-5α dihydrotestosterone (13), a pharmacodynamic biomarker for AR inhibition. This dose level also ensured that exposures were within the range previously identified required for maximum tumor regression in murine CRPC models. The apalutamide exposure–efficacy analysis presented here revealed that in the relatively narrow observed exposure range (for both apalutamide and its metabolite), no statistically significant differences in MFS between exposure quartiles were determined, further confirming that 240 mg once-daily dose of apalutamide provides efficacious exposure in patients with nmCRPC, regardless of dose adjustments. Furthermore, the exposure was similar in patients with and without dose reductions and interruptions. Therefore, dose reductions/interruptions for the management of TEAEs are not expected to reduce the efficacy of apalutamide.
These results are in line with those obtained with another AR antagonist used for the treatment of metastatic CRPC, enzalutamide, which was evaluated in the AFFRIM trial (14). In this analysis, no apparent ER relationship was found for overall survival with an oral dose of 160 mg/day. The analysis was conducted using Ctrough plasma values as the exposure metric instead of AUC0–24h,ss, but the exposure–efficacy analysis was also conducted classifying the Ctrough plasma values into quartiles, as in this analysis, and no significant differences in overall survival were seen regarding the quartiles of exposure.
Fatigue, fall, fracture, skin rash, weight loss, and arthralgia were adverse drug reactions that occurred at an incidence greater than 10% in SPARTAN patients receiving apalutamide. The exposure–safety analysis demonstrated that within the observed exposure range in apalutamide-treated patients, the exposure–TEAE relationship was only statistically significant for skin rash and weight loss. Simulations based on the developed exposure–safety relationship demonstrate that dose reductions will likely improve apalutamide tolerability in patients who develop toxicity after starting apalutamide treatment at 240 mg once daily.
Although these results suggest that reductions and interruptions are appropriate to prevent patients from discontinuing apalutamide, this is a relatively small dataset, and further study is warranted in real world use regarding the effect of drug holidays and extended use of lower doses.
In summary, ER analyses demonstrated that the MFS benefit in patients with nmCRPC was similar across the range of apalutamide and N-desmethyl-apalutamide exposure following the recommended apalutamide dose of 240 mg once daily. A significant association with apalutamide exposure was observed for the incidence of skin rash and weight loss TEAEs, at any grade. The ER relationship with these AEs suggests that patients who experience AEs may benefit from reducing the dose of apalutamide without compromising efficacy.
Disclosure of Potential Conflicts of Interest
C. Perez-Ruixo reports personal fees from Janssen RD (employee) during as well as outside the submitted work. O. Ackaert reports personal fees from Janssen RD (employee) during as well as outside the submitted work and reports other from Johnson & Johnson (stock ownership) outside the submitted work. D. Ouellet reports other from Janssen (former employee) during the conduct of the study and other from Pfizer (current employee) outside the submitted work. C. Chien reports other from Janssen R&D (holds JNJ stocks) outside of the submitted work. D. Olmos reports grants, personal fees, non-financial support, and other from AstraZeneca [clinical trial steering committee (unpaid), travel support to scientific meetings]; grants and personal fees from Astellas; grants, personal fees, non-financial support, and other from Bayer Healthcare [clinical trial steering committee (unpaid), travel support to scientific meetings] and Janssen (clinical trial steering committees, travel support to scientific meetings); personal fees from Clovis Oncology and Daiichi-Sankyo; non-financial support and other from F-Hoffman-La Roche [clinical trial steering committee (unpaid), travel support to scientific meetings]; non-financial support from Genentech [clinical trial steering committee (unpaid)]; other from Ipsen (travel support to scientific meetings); and grants from Sanofi outside the submitted work. P. Mainwaring reports personal fees from Janssen (lectures, translational research) during the conduct of the study. M.K. Yu reports other from Janssen R& D (employee) during as well as outside the submitted work; in addition, M.K. Yu is listed as a co-inventor on a patent regarding a method of use of apalutamide for the treatment of patients with nmCRPC owned by Janssen. J.J. Perez-Ruixo reports personal fees from Janssen RD (employee) and other from Johnson & Johnson (stock owner) during the conduct of the study. M.R. Smith reports other from Janssen (clinical research support to institution) during the conduct of the study, and personal fees from Bayer, Astellas, and Janssen (consultant) outside the submitted work. E.J. Small reports personal fees from Janssen (advisory board, speakers fees) outside the submitted work. No potential conflicts of interest were disclosed by the other authors.
Authors' Contributions
C. Perez-Ruixo: Formal analysis, methodology, writing-original draft, project administration, writing-review and editing. O. Ackaert: Formal analysis, methodology, writing-original draft, project administration, writing-review and editing. D. Ouellet: Formal analysis, methodology, writing-review and editing. C. Chien: Conceptualization, formal analysis, writing-review and editing. H. Uemura: Investigation, writing-review and editing. D. Olmos: Investigation, writing-review and editing. P. Mainwaring: Investigation, writing-review and editing. J.Y. Lee: Investigation, writing-review and editing. M.K. Yu: Conceptualization, writing-review and editing. J.J. Perez-Ruixo: Formal analysis, methodology, writing-original draft, writing-review and editing. M.R. Smith: Investigation, writing-review and editing. E.J. Small: Investigation, writing-review and editing.
Acknowledgments
The authors would like to thank the patients, investigators, and their medical, nursing, and laboratory staff who participated in the clinical studies included in the present work. The authors acknowledge Jonás Samuel Pérez-Blanco for his support during the exposure–response analysis. This study was funded by Janssen Research and Development.
The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.