Table 1 Background Information for the Analyzed Data
Metrics
|
Category
|
Number of Cases (%)
|
Total
|
-
|
322 (100%)
|
Drug Modality
|
Small Molecule
|
169 (52.5%)
|
Monoclonal Antibody
|
78 (24.2%)
|
Other Biologics1
|
54 (16.8%)
|
Others
|
21 (6.52%)
|
First-in-class
|
Yes
|
83 (25.8%)
|
No
|
239 (74.2%)
|
First Indication
|
Yes
|
127 (39.4%)
|
No
|
195 (60.6%)
|
Therapeutic Area
|
Oncology (Solid)
|
77 (23.9%)
|
Oncology (Non-solid)
|
30 (9.32%)
|
Immunology
|
43 (13.4%)
|
Infectious Disease
|
26 (8.07%)
|
Central Nervous System Disease
|
23 (7.14%)
|
Cardiovascular/renal Disease
|
20 (6.21%)
|
Metabolic Disease
|
17 (5.28%)
|
Respiratory Disease
|
4 (1.24%)
|
Others
|
82 (25.5%)
|
Special Notices
|
Orphan Drug Designation
|
73 (22.7%)
|
Accelerated Review
|
15 (4.66%)
|
MRCTs in Package
|
Yes
|
142 (44.1%)
|
No
|
180 (55.9%)
|
Sponsor
|
Japanese Domestic Company
|
157 (48.8%)
|
Multi-national Company
|
165 (51.2%)
|
MRCT = multi-regional clinical trial
1 Other biologics include biologics other than monoclonal antibodies such as enzymes and peptides.
Table 2 Summary of Percentage of Modeling Analyses Performed (PMAP) and Percentage of Dose Selection and Dosage Modification using the Modeling Analysis (PDSM) Stratified by Categories
Metrics
|
Category
|
PMAP
|
PDSM
|
Total
|
n = 322
|
154 (47.8%)
|
78 (24.2%)
|
First-in-class
|
Yes (n = 83)
|
50 (60.2%)
|
26 (31.3%)
|
No (n = 239)
|
104 (43.5%)
|
52 (21.8%)
|
First Indication
|
Yes (n = 127)
|
80 (63.0%)
|
39 (30.7%)
|
No (n = 195)
|
74 (37.9%)
|
39 (20.0%)
|
Special Notices
|
Orphan Drug Designation (n = 73)
|
43 (58.9%)
|
21 (28.8%)
|
Accelerated Review (n = 15)
|
9 (60.0%)
|
3 (20.0%)
|
MRCTs in Package
|
Yes (n = 142)
|
104 (73.2%)
|
57 (40.1%)
|
No (n = 180)
|
50 (27.8%)
|
21 (11.7%)
|
Sponsor
|
Japanese Domestic Company (n = 157)
|
56 (35.7%)
|
28 (17.8%)
|
Multi-National Company (n = 165)
|
98 (59.4%)
|
50 (30.3%)
|
MRCT = multi-regional clinical trial. shown as number of cases (%)
Table 3 Cases of model-informed dose selection for proposed dose regimens for phase III studies different from those in phase II studies
Compound name
[Therapeutic area]
|
Phase II doses
|
Phase III doses
|
Point of MIDD
|
Semaglutide
[Metabolic disease]
|
2.5, 5, 10, 20, and 40 mg
|
3, 7, and 14 mg
|
Based on the data from a phase II study investigating five doses of oral semaglutide (2.5, 5, 10, 20, and 40 mg) versus placebo, E-R model was developed to describe the relationship between semaglutide Cavg and the change in HbA1c levels. Based on the simulated E-R curve, three doses of oral semaglutide as 3, 7, and 14 mg were selected for the phase III study.
|
Ravulizumab
[Others]
|
400, 600, 800, 900, 1400, 1600, 1800, and 3000 mg
|
Initial dose of 2400 mg and maintenance dose of 3000 mg for body weight range of 40 to <60 kg; Initial dose of 2700 mg and maintenance dose 3300 mg for body weight range of 60 to <100 kg; Initial dose of 3000 mg and maintenance dose of 3600 mg for body weight of ≥100 kg
|
Population PK analysis was performed which showed that body weight affected PK of ravulizumab, with higher exposures in lighter-weight patients than those in heavier-weight patients, suggesting the need for weight-based dose adjustment. A PK-LDH model was also established for the inhibitory response to terminal complements (measured as the largest clinically relevant LDH response) and set a weight-based dosage regimen (weight band dose) in a phase III trial which was expected to be above target thresholds in all individuals by simulations.
|
Galcanezumab
[CNS]
|
5, 50, 120, and 300 mg (no loading dose)
|
120 mg (240 mg once as a loading dose), 240 mg
|
Simulations using population PK modeling showed that the serum steady-state levels may be attained rapidly by setting 240 mg as a loading dose; hence, two groups were selected in the phase III study [120-mg group (240 mg once as a loading dose) and 240-mg group].
|
Belotralstat hydrochloride
[Others]
|
55, 110, 218, and 300 mg
|
110 and 150 mg
|
Simulations were performed at each dose-level from 125–250 mg to estimate the percentage of participants meeting the same EC50 target levels. Parameter estimates with linear regression were used to impute trough estimates for the selected dose. The simulations supported the selection of 150 mg.
|
Gefapixant citrate
[Others]
|
7.5, 20, and 50 mg
|
15 and 45 mg
|
From E-R simulation (efficacy and safety), 15 mg was expected to provide a clinically meaningful effect, and approximately 80% of the participants may not exhibit adverse events. A dose level of 45 mg was predicted to maintain a low rate of discontinuation and achieve the greatest effect.
|
Nemolizumab
[Others]
|
0.1, 0.5, and 2.0 mg/kg
|
60 mg
|
A dose of 60 mg/body was expected to be included within the range as those provided at 0.5–2.0 mg/kg, to be as effective as or better than this dosage in patients with a body weight of 30–120 kg. Therefore, the fixed dose was chosen in the phase III study considering convenience as well.
|
CNS, central nervous system; E-R, exposure-response; HbA1c, glycated hemoglobin; PK, pharmacokinetic; LDH, lactate dehydrogenase; MIDD, Model-Informed Drug Development
The therapeutic areas were categorized as oncology (solid), oncology (non-solid), immunology, infectious disease, central nervous system, cardiovascular/renal disease, metabolic disease, and respiratory disease.
Table 4 Cases of model-informed dose selection for proposed different dose regimens for new drug applications from those in the phase III study
(a) Cases accepted by the Japanese authority
Compound name
[Therapeutic area]
|
Phase III study dose 1
|
Proposed dose at submission
|
MIDD use points
|
Acceptance in USPI
|
Fremanezumab
[CNS]
|
225 mg once every 4 weeks with a loading dose of 675 mg
|
225 mg once every 4 weeks without loading dose
|
Population PK and E-R analyses conducted at the time of review in Europe and the United States were analyzed again with additional data from the Japan-Korea joint studies (Studies 406-102-00001 and 406-102-00002), suggesting that Japanese patients with chronic migraine (chronic migraine) have similar efficacy 225 mg dose once every 4 weeks with and without the loading dose of 675 mg.
|
Yes
|
Durvalumab
[Oncology (solid)]
|
In a phase III study (dose: 1500 mg Q3W/Q4W), participants with body weight ≤30 kg were excluded.
|
A single dose is 20 mg/kg if the body weight is below 30 kg
|
Exposure in patients with body weight of 30 kg at 20 mg/kg dose Q3W or Q4W was lower than that in patients with body weight of more than 30 kg at 1500 mg dose Q3W or Q4W, but was shown to overlap. There was no clear relationship between drug exposure and the occurrence of overall survival or adverse events in the exposure range at 1500 mg.
|
Yes
|
Pembrolizumab
[Oncology (solid)]
|
200 mg Q3W
|
400 mg Q6W
|
The E-R relationship showed that efficacy and safety were consistent across doses and AUC ranging from 2 mg/kg Q3W to 10 mg/kg Q2W (>5-fold higher exposures), regardless of cancer type. In addition, exposures at 200 mg Q3W were covered within the range of those provided by the dose range above, where both efficacy and safety have been confirmed. The exposure (AUC) following 400 mg Q6W was predicted to be as efficacious and safe as 200 mg Q3W.
|
Yes
|
Nivolumab
[Oncology (solid)]
|
Nivolumab 3 mg/kg and ipilimumab 1 mg/kg
|
Nivolumab 240 mg and ipilimumab 1 mg/kg
|
Although exposure to 240 mg nivolumab was expected to be higher than that to nivolumab 3 mg/kg, it was also expected to be lower than that of nivolumab at the dosage and administration (10 mg/kg Q2W), which was well tolerated in Japanese patients.
|
Yes
|
Nivolumab
[Oncology (solid)]
|
240 mg Q2W
|
240 mg Q2W or 480 mg Q4W
|
Simulations using population PK and E-R models predicted that there would be no clear difference in efficacy and safety between nivolumab 480 mg Q4W and 240 mg Q2W, which is the approved dosage and administration of nivolumab. Overall survival and response rates of nivolumab 3 mg/kg treated with Q2W, 240 mg treated with Q4W, 480 mg treated with Q2W were not expected to differ clearly. Similarly, no clear difference was predicted between the dose regimens for the adverse events studied.
|
Yes
|
Adalimumab
[Immunology]
|
0.6–2.4 mg/kg in a phase III study of moderate to severe ulcerative colitis patients aged from 4–17 years old
|
Setting the dosage from 20–160 mg based on weight category in pediatric patients
|
Simulations of fixed doses by body weight category and weight equivalent doses used in phase III studies showed similar pharmacokinetic profiles.
|
Yes2
|
Selpercatinib
[Oncology (solid)]
|
160 mg BID in Study 17001 for patients aged ≥12 years
|
The following dose is set for patients aged ≥12 years according to body surface area category (approximately 92 mg/m2 per dose):
80 mg BID for <2 m2, 120 mg BID for 1.2 to <1.6 m2, 160 mg BID for ≥1.6 m2..
|
There was an identified risk of QT prolongation with increase in selpercatinib exposure, and 160 mg fixed dose can result in excess exposure in children. When doses were selected for each body surface area category in the simulation, the exposure of drug in each body surface area category was similar. In addition, a preliminary study of a multi-regional phase I/II study in patients with advanced solid cancers aged 6 months to 21 years with RET genetic aberrations showed that responders were observed in patients with thyroid cancer (including patients with medullary thyroid cancer) who received 92 mg/m2 BID, and that the exposure of drug in pediatric patients who received 92 mg/m2 BID was similar to that in adult patients who received 160 mg BID, supporting the appropriateness of dose-setting by body surface area category.
|
Partially yes3
|
RET, rearranged during transfection; Q3W, every 3 weeks; Q6W, every 6 weeks; BID, twice daily; USPI, US prescribing information
The therapeutic areas were categorized as oncology (solid), oncology (non-solid), immunology, infectious disease, central nervous system, cardiovascular/renal disease, metabolic disease, and respiratory disease.
1 Among the doses and administrations established in the phase III study, only those related to the changed or added dosage and administration at the time of application for approval are described.
2 The weight category of USPI differs from that of the domestic package insert.
3 For USPI, the dose is set according to body weight category rather than body surface area.
(b) Cases not accepted by the Japanese authority
Compound name
[Therapeutic area]
|
Phase III dosage and administration 1
|
Proposed dosage and administration
|
MIDD use points
|
Comment from Japan regulatory agency
|
Acceptance in USPI
|
Paliperidone palmitate
[CNS]
|
12-week intervals
|
Once every 3 months
|
Exposure with Q13W treatment was similar to that with Q12W treatment.
|
Although the predicted PK parameters were similar, a dosing interval of 12 weeks was used in two phase III studies, and the efficacy and safety of continuous dosing other than 12-week intervals have not been investigated. In addition, a 12-week interval was considered appropriate as it adheres to the dosing interval in terms of promoting optimum use.
|
Yes
|
Somapacitan
[Others]
|
The initial dosage was set at 1.5 mg for patients aged ≥60 years, 1.0 mg for over 60 years old, and 2.0 mg for female patients who were taking oral estrogen.
|
When switching from human somatotropic preparations to somapacitan in women, the Initial dose was set at 2.0 mg for ≤60 years, 1.5 mg for >60 years old, and 4.0 mg for female patients who were taking oral estrogen.
|
Based on the findings using population PK/PD analysis, a dose higher than the starting dose used in the phase III study was selected.
|
The starting dose used in the phase III studies is appropriate in terms of efficacy and safety.
|
No
|
Molidustat sodium
[Metabolic disease]
|
Postprandial administration
|
No dietary restrictions
|
A simulated Hb level was determined, assuming that BA levels increase by 30% in the fasting state compared with those in the postprandial state. Changes in BA did not appear to significantly impact Hb levels. Hence, no restriction on the meal in the administration was observed.
|
The postprandial administration was appropriate owing to the following reasons:
-PK of the phase I study showed an effect similar to that of food.
-The effect of the exposure variation on Hb level was unclear.
-The postprandial administration was set in the domestic phase III study.
|
-2
|
Adalimumab
[Immunology]
|
For patients with moderate-to-severe ulcerative colitis aged 4–17 years, adalimumab was administered at Weeks 0, 1, 2, 4, and 6 during the run-in period (up to Week 8).
|
The dose at Week 1 was not set in pediatric patients. The dosing was set once every 2 weeks after the first dose. The same dose as that at Week 2 was administered at Weeks 4 and 6
|
Population PK indicated that the exposure did not change significantly in the absence of treatment at Week 1. In addition, E-R analysis showed that the same level of efficacy could be achieved by administering the fixed dosage and administration according to body weight category, regardless of the administration at Week 1.
|
Early induction of remission is important in active ulcerative colitis. To increase the possibility of remission, high serum levels must be maintained after the initiation of treatment with drug, and it is appropriate to set the dosage at Week 1.
|
No
|
The therapeutic areas were categorized as oncology (solid), oncology (non-solid), immunology, infectious disease, central nervous system, cardiovascular/renal disease, metabolic disease, and respiratory disease.
1 Among the doses and administrations established in the phase III study, only those related to the changed or added dosage and administration at the time of application for approval are described.
2 It is not approved in US.
BA = bioavailability; Hb = hemoglobin; USPI = US prescribing information
Table 5 Cases of model-informed dose selection without conducting proof-of-concept (PoC) and/or dose-finding studies
Compound name
[Therapeutic area]
|
MIDD use points
|
Special Remarks
|
Inebilizumab
[Others]
|
PK/PD modeling was constructed based on the outcome of a phase I study (0.1–10 mg/kg single intravenous administration) in patients with other indications, without conducting a phase I study in patients who could be evaluated for PoM/PoC, and applied to the dosage regimen selection in the following study.
|
Orphan drug
|
Nirmatrelvir/ Ritonavir
[Infectious diseases]
|
Though dose-finding clinical studies were not performed, the validity of dose selection was explained
The phase II/III dose was selected as a dose of PK reaching in vitro EC90 for severe acute respiratory syndrome coronavirus 2
Duration of phase II/III studies was set using QSP modeling.
|
Products subject to special approval
|
PoM, proof of mechanism; PoC, proof of concept; QSP, quantitative system pharmacology
The therapeutic areas were categorized as oncology (solid), oncology (non-solid), immunology, infectious disease, central nervous system, cardiovascular/renal disease, metabolic disease, and respiratory disease.
Table 6 Cases of model-informed dose adjustments for special populations
Generic name [Disease area]
|
Alectinib hydrochloride [oncology (non-solid)]
|
Dose modulator
|
Pediatrics and body weight
|
MIDD use points
|
Dose-setting in pediatric phase II studies supported by the results of adult phase I studies and simulation
|
Analysis method
|
PK for pediatrics was predicted without the use of pediatric actual data. PK predictions for pediatrics were performed using the equation of allometry, which mathematically expresses the relationship of body weight to PK parameters in adults.1
|
Analysis results
|
It was estimated that the trough levels of patients with body weight of 20 kg corresponding to pediatrics aged 6-years old were approximately 2.3 times higher than those of patients with body weight of 60 kg. The trough concentrations at 150 mg BID in pediatrics aged 6 years old were predicted to be similar to those of 300 mg BID in patients with body weight of 60 kg and decreased with increasing body weight. Based on E-R relationship and in vitro growth inhibitory activity, it was predicted that 150 mg BID may be effective in patients who may have relatively low trough concentration levels.
|
Japanese authority's view
|
Unknown because CTD and review report did not include Japanese authority’s opinion on the setting of dosage regimen for phase II studies.
|
Generic name [Therapeutic area]
|
Glucagon [metabolic disease]
|
Dose modulator
|
Japanese pediatrics
|
MIDD use points
|
Dose adequacy of Japanese pediatric patients was explained by model analysis (data from non-Japanese pediatric patients were utilized. No domestic clinical studies were conducted).
Cases in which Japanese pediatrics could be extrapolated using population PK models and E-R models for Japanese adults, foreign adults, and foreign children
|
Analysis method
|
Data from patient populations including Japanese adult patients and non-Japanese pediatric patients aged 4 years or older were available for analysis. In population PK, allometric scaling was used, with body weight as a covariate. E-R analysis described the relationship between glucagon levels in blood and elevated glucose using an indirect response model. In the simulations, three different age groups were assumed (4 to <8 years, 8 to <12 years, and 12 to <18 years), and glucagon exposure and glucose response were simulated after 3 mg intranasal or intramuscular glucagon administration based on body weight (1 mg for body weight >25 kg, and 0.5 mg for body weight of ≤25 kg) in each age group.
|
Analysis results
|
The predicted BGmax of intranasal 3 mg glucagon administered to pediatrics and adults in each age-group resembled those predicted for intramuscular glucagon injections based on body weight. Simulations also predicted that in children and adolescents of three age-groups, the treatment success rate after administration of 3 mg glucagon nasal spray was more than 99%, which was similar to that after administration of glucagon injection.
|
Japanese authority's view
|
Although no clinical trials have been conducted in Japanese pediatric patients, successful treatment can be expected when 3 mg drug is administered nasally to Japanese pediatric patients based on the simulations. In the simulation, it was predicted that exposure with 3 mg intranasal glucagon in Japanese pediatric patients is higher than that observed in adult patients but is below Cmax for intravenous glucagon 2 mg, the largest approved dose in the United States. In addition, considering that the safety profile of 3 mg nasal administration in adults was not significantly different between Japan and foreign countries, 3 mg may be selected as the intranasal administration dose of the drug in pediatrics, similar to that in foreign countries. However, the intranasal administration of 3 mg glucagon to Japanese pediatric patients with diabetes mellitus has not been investigated. Therefore, collecting post-marketing information on the safety and efficacy of drug administration to these patients is necessary.
|
Generic name [Therapeutic area]
|
Mepolizumab [immunology]
|
Dose modulator
|
Pediatrics and body weight
|
MIDD use points
|
Establishment of dosage and administration for pediatrics based on population PK:
In a multi-national phase II study in pediatrics aged 6–11 years, doses were 40 mg for body weight <40 kg, 100 mg for body weight ≥ 40 kg and the doses were administered subcutaneously at 4-week intervals. Based on the subsequent population PK, 40 mg dose every 4 weeks was selected for pediatrics aged 6–11 years with severe asthma, regardless of body weight.
|
Analysis method
|
In population PK, the effect of body weight on PK was incorporated allometrically. In PK/PD (blood eosinophil count), an indirect response model, which included the production inhibition rate constant of eosinophils, was utilized.
|
Analysis results
|
Population PK analysis estimated that the expected dosage and administration in pediatrics aged 6–11 years with severe asthma was 40 mg every 4 weeks regardless of body weight to correspond exposure levels in patients aged ≥12 years with severe asthma. In clinical studies, drug exposure and the percentage reduction in blood eosinophil count after subcutaneous administration of the pediatric dose in patients with body weight of 40 kg were not significantly different from those after subcutaneous administration of 100 mg in patients aged 12 years or older with severe asthma. Simulated dose-response relationships between blood drug levels and blood eosinophil counts suggested that the exposure of drug and the rate of reduction in blood eosinophil counts after subcutaneous administration of 40 mg every 4 weeks in pediatric patients with severe asthma and body weight ≥40 kg were similar to those after subcutaneous administration of 100 mg in patients with severe asthma aged ≥12 years.
|
Japanese authority's view
|
It was considered acceptable to use 40 mg every 4 weeks subcutaneously. In the absence of results for this dosage and administration to pediatric patients with severe asthma and body weight ≥ 40 kg, the safety and efficacy of drug administration should be determined through post-marketing surveillance.
|
Generic name [Therapeutic area]
|
Azilsartan [metabolic disease]
|
Dose modulator
|
Pediatrics and body weight
|
MIDD use points
|
Dose selection for the phase III study (OCT-101 study)
|
Analysis method
|
A population PK analysis was performed using plasma drug concentrations in Japanese hypertensive patients aged from 6 to less than 16 years and healthy Japanese adults. The effects of body weight on CL/F and Vc/F were incorporated in an allometric manner.
|
Analysis results
|
Population PK analysis showed that the exposure in pediatrics with body weight of 50–80 kg to once-daily dose of 2.5–40 mg was similar to that in adults at the same dose, and exposure with the same dose in pediatrics with body weight of 20–49 kg was marginally less than twice compared to that of adults at the same dose. To ensure safety of the patients, the starting dose for OCT-101 study was 5 mg once daily, which was one fourth of the adult dose for body weight ≥ 50 kg and 2.5 mg once daily for body weight of <50 kg. Based on this estimation by population PK analysis, the maximum dose for pediatrics was 40 mg once daily for body weight ≥ 50 kg and 20 mg once daily for body weight <50 kg, which were doses that did not result in exposure at 40 mg once daily, which is the maximum dose for adults.
|
Japanese authority's view
|
The dosage and administration of drug established in OCT-101 study was considered appropriate from the viewpoint of obtaining exposures similar to or lower in Japanese pediatrics aged ≥6 years than those obtained when the approved dosage and administration of drug is administered to Japanese adults.
|
Generic name [Therapeutic area]
|
Gefapixant citrate [others]
|
Dose modulator
|
Renal function
|
MIDD use points
|
Dosage and administration in patients with renal impairment was selected based on population PK analysis.
|
Analysis method
|
Population PK analysis
|
Analysis results
|
A 1.46-fold increase in patients with moderate renal impairment and a 1.89-fold increase in patients with severe renal impairment were estimated, relative to patients with normal renal function. In addition, the steady-state Cmax and AUC at 45 mg once daily in patients with severe renal impairment were predicted to be similar to those at 45 mg twice daily in patients with normal renal function. Based on these findings, the dosing regimen of 45 mg once daily was proposed in patients with severe renal impairment.
|
Authority's view
|
The explanation was accepted.
|
Generic name [Therapeutic area]
|
Sodium tolvaptan phosphate [others]
|
Dose modulator
|
Patients with difficulty in oral fluid intake
|
MIDD use points
|
Population PK and simulations to explain the starting dose in the special population at the proposed dose for NDA.
|
Analysis method
|
Population PK analysis
|
Analysis results
|
Estimated exposures to tolvaptan after intravenous administration of 2, 4, 8, and 16 mg and oral administration of 7.5 and 15 mg indicated that the dose of 8 mg provided similar exposure to that of the oral tolvaptan 7.5 mg and was half of the recommended drug dosing regimen. To reduce the risk due to excessive water diuresis immediately after the initiation of drug administration, it was recommended that drug administration should be started at a half-dose (8 mg) of the standard dose in patients with difficulty in oral intake.
|
Japanese authority's view
|
The applicant's policy to advise that it is desirable to start administration of drug at half-dose (8 mg) of the standard dosage is justified.
|
Generic name [Therapeutic area]
|
Nirmatrelvir and ritonavir [infectious diseases]
|
Dose modulator
|
Pediatrics and body weight
|
MIDD use points
|
Dosage regimen for pediatrics based on predictions using adult population PK model.
Cases in which pediatric patients aged ≥12 years and body weight ≥ 40 kg could be extrapolated using a population PK model in adults and a distribution of body weight in pediatrics.
|
Analysis method
|
Prediction of exposure differences by body weight in population PK analysis
|
Analysis results
|
Plasma drug exposures to 300 mg nirmatrelvir and 100 mg ritonavir co-administered twice daily were predicted to be similar in pediatrics with body weight ≥ 40 kg and adults. The dosage and administration in pediatrics aged ≥12 years and body weight ≥ 40 kg was set at the same dosage and administration as that in adults.
|
Authority’s view
|
It is reasonable that the dosage and administration in pediatrics aged ≥ 12 years and body weight ≥40 kg should be set to the same dosage and administration as in adults. However, it is necessary to continue to collect post-marketing information on the efficacy and safety in pediatrics, including the results of clinical studies in pediatrics under planning, and to provide information to medical institutions as soon as new findings are obtained.
|
Generic name [Therapeutic area]
|
Valbenazine tosilate [central nervous system]
|
Dose modulator
|
Deficiency in CYP2D6 activity
|
MIDD use points
|
Dose selection for phase III studies and explaining the need for dose adjustments in patients with factors that increase exposure
|
Analysis method
|
Population PK analysis
|
Analysis results
|
In CYP2D6 poor metabolizer, the exposure of the metabolite NBI-98782 after drug administration was considered to increase approximately twofold. E-R analysis showed that the percentage change from baseline in the Abnormal Involuntary Movement Rating Scale total score and the change from baseline as well as the global impression-improvement score of tardive dyskinesia tended to improve with increasing exposure. The incidences of somnolence-related adverse events, parkinsonism-related adverse events, and akathisia-related adverse events were related to the exposure levels (Cmax and AUC) of unchanged drug and NBI-98782.
|
Japanese authority’s view
|
The applicant’s belief that 40 mg should be administered to patients with genetically defective CYP2D6 activity and those with risk of the increased exposure, and that the dose should not be increased to 80 mg is acceptable.
|
Generic name [Therapeutic area]
|
Filgotinib maleate [immunology]
|
Dose modulator
|
Renal function
|
MIDD use points
|
Selection of proposed dose based on renal function
|
Analysis method
|
Population PK analysis
|
Analysis results
|
Dose adjustment for patients with ulcerative colitis (UC) with mild and moderate renal impairment was considered unnecessary for the following reasons:
- As a consequence of population PK, exposures (Cmax and AUCtau) of unchanged drug and major metabolite (GS-829845) in patients with UC and mild renal impairment were similar to those in patients with normal renal function.
- Exposure of unchanged drug was similar between patients with UC and moderate renal impairment and patients with normal renal function, and exposure of GS-829845 was 20–30% higher in those with UC and moderate renal impairment than in patients with normal renal function.
|
Japanese authority's view
|
Caution should be exercised for half the dose of drug in patients with moderate renal impairment for the following reasons: 2
- PK parameters between the approved indication (rheumatoid arthritis: RA) and the proposed indication (UC) were similar.
- The results of predictions for renal dysfunction in patients with UC were similar as those for renal dysfunction in patients with RA.
Dedicated study assessing the effect of renal function suggested that exposure to GS-829845 in moderate renal impairment may be increased more than 2-fold.
|
CL/F, apparent total clearance; Vc/F, apparent volume of distribution in central compartment
The therapeutic areas were categorized as oncology (solid), oncology (non-solid), immunology, infectious disease, central nervous system, cardiovascular/renal disease, metabolic disease, and respiratory disease.
1 Biological rules for the relationship between organism function and size, represented by the equation Y = a × Wb (Y: variable representing organism function; W: variable representing organism size; a, b: constants). To predict pharmacokinetics, Y is used as a PK parameter, such as clearance and volume of distribution, and W is used as body weight. The constants a and b are derived from the relationship between PK parameters and body weight in existing data (often 0.75, clearance and 1.0 for volume of distribution, respectively) and predict the PK parameter at the desired body weight.
2 It was not acceptable to the authorities to propose that no dosage adjustment of drug is required for UC with moderate renal impairment.
BGmax, maximum plasma glucose level; CTD, common technical document.
Table 7 Summary of Cases without Japanese Data in Which Modeling Analysis Provided Major Rationale for Dosage Selection
Compound Name
|
Approval Year in Japan
|
Indication
|
Development Phase
|
Application of Modeling Analysis
|
Ambrisentan
|
2021
|
Pulmonary arterial hypertension (Orphan)
|
End of Phase I
|
- For selecting dose/dosage for multi-regional (including Japan) phase II study in pediatric patients using data from phase II/III study in adults conducted outside Japan.
|
Inebilizumab
|
2021
|
Neuromyelitis optica spectrum disorder (first indication, orphan)
|
End of Phase I
|
- For selecting dose/dosage for multi-regional (including Japan) phase II/III study using data from phase I study conducted outside Japan.
|
Pemigatinib
|
2021
|
Cholangiocarcinoma with FGFR2 fusion or rearrangement (first indication, orphan)
|
End of Phase I
|
- For selecting dose/dosage for multi-regional (including Japan) phase II study using data from phase I study conducted outside Japan.
|
Ixazomib
|
2021
|
Relapsed/Refractory Multiple myeloma
|
End of Phase II
|
- For selecting dose/dosage for multi-regional (including Japan) phase III study using data from phase I/II study conducted outside Japan.
|
Perampanel
|
2020
|
Epilepsy
|
End of Phase II
|
- For selecting dose/dosage for multi-regional (including Japan) phase III study in pediatric patients using data from phase II in pediatric patients and part of phase III in adults and adolescents conducted outside Japan.
|
Osilodorostat
|
2021
|
Cushing’s disease (first indication)
|
End of Phase II
|
- For selecting dose/dosage for multi-regional (including Japan) phase III study using data from early-phase clinical studies conducted outside Japan.
|
Ofatumumab
|
2021
|
Multiple sclerosis (Orphan)
|
End of Phase II
|
- For selecting dose/dosage for phase III (outside Japan) and multi-regional phase II (including Japan) study using data from phase II study conducted outside Japan.
|
Valbenazine
|
2022
|
Tardive dyskinesia (first indication)
|
End of Phase II
|
- For selecting dose/dosage for phase III study (outside Japan) using data from phase II study conducted outside Japan.
- Domestic phase II/III study was separately conducted in Japan with the same dose as the phase III study determined by the analysis.
|
Lasmiditan
|
2022
|
Migraine (first indication)
|
End of Phase III
|
- For selecting dose for approval using data from early-phase clinical trials conducted outside Japan.
- No PK sampling was done in late-phase clinical trials owing to its dosage (only administered for migraine attack).
|
Clomiphene
|
2022
|
Oligozoospermia
|
After launch
|
- For demonstrating exposure similarity among different dose/dosage (25 mg once daily vs. 50 mg once in two days).
- Public knowledge-based application.
- No additional clinical study was conducted.
|