CLINICAL PHARMACOLOGY
Mechanism Of Action
Rolapitant is a selective and competitive antagonist of human substance P/NK1 receptors. Rolapitant does not have significant affinity for the NK2 or NK3 receptors or for a battery of other receptors, transporters, enzymes and ion channels. Rolapitant is also active in animal models of chemotherapy-induced emesis.
Pharmacodynamics
NK1 Receptor Occupancy
A human Positron Emission Tomography (PET) study with rolapitant demonstrated that rolapitant crosses the blood brain barrier and occupies brain NK1 receptors. A dose-dependent increase in mean NK1 receptor occupancy was observed in the oral dose range from 4.5 mg to 180 mg of rolapitant. At the 180 mg oral dose of rolapitant, the mean NK1 receptor occupancy was 73% in the striatum at 120 hours after a single dose administration in healthy subjects. The relationship between NK1 receptor occupancy and the clinical efficacy of rolapitant has not been established.
Cardiac Electrophysiology
At an oral dose of 720 mg (4 times the recommended oral dose), VARUBI does not prolong the QT interval to any clinically relevant extent. The maximum rolapitant concentration after a single 720 mg dose of oral rolapitant was 30% to 50% higher than that achieved with the recommended dose of
166.5 mg of intravenous rolapitant.
Pharmacokinetics
Absorption
Oral tablet
Following a single oral dose administration of 180 mg VARUBI tablets under fasting conditions to
healthy subjects, rolapitant was measurable in plasma between 30 minutes and the peak plasma
concentration (Cmax) for rolapitant which was reached in about 4 hours and mean Cmax was
968 ng/mL (%CV:28%).
Following multiple oral doses of 9 to 45 mg once daily of rolapitant (5% to 25% of the recommended dose) for 10 days, accumulation of rolapitant (ratio of AUC0-24hr) ranged from 5.0 to 5.3 fold.
The systemic exposures (Cmax and AUC) to rolapitant increased in a dose-proportional manner when single oral doses of rolapitant increased from 4.5 mg to 180 mg. With 4 times the recommended clinical oral dose of 180 mg, the Cmax and AUC of rolapitant increased 3.1 fold and 3.7 fold, respectively.
Concomitant administration of a high-fat meal did not significantly affect the pharmacokinetics of rolapitant after administration of 180 mg VARUBI tablets [see DOSAGE AND ADMINISTRATION].
Injectable emulsion
Following a 30-minute infusion of a single intravenous dose of 166.5 mg VARUBI injectable emulsion to healthy subjects, the mean Cmax at the end of the infusion (30 min) was 1986 ng/mL (%CV:39%).
Following multiple intravenous doses of 18 to 54 mg once daily of rolapitant (approximately 10% to 33% of the recommended dose) for 10 days, accumulation of rolapitant (ratio of AUC0-24hr) ranged from 4.3 fold to 5.4 fold.
The systemic exposures (Cmax and AUC) to rolapitant increased in a dose-proportional manner when single intravenous doses of rolapitant increased from 18 mg to 180 mg. The Cmax and AUC of rolapitant increased 11.6 fold and 10.1 fold, respectively.
At the recommended dose, the absolute bioavailability of rolapitant is 91%.
Distribution
Rolapitant was highly protein bound to human plasma (99.8%). The volume of distribution (Vd) was 392 L in healthy subjects following a single intravenous dose of 166.5 mg rolapitant. The apparent volume of distribution (Vd/F) following a single oral dose of 180 mg rolapitant was 460 L in healthy subjects. The large Vd indicated an extensive tissue distribution of rolapitant. In a population pharmacokinetic analysis of oral rolapitant, the Vd/F was 387 L in cancer patients.
Elimination
Following single intravenous doses (18 to 180 mg) and single oral doses (4.5 to 180 mg) of rolapitant, the mean terminal half-life (t1/2) of rolapitant ranged from 138 to 205 hours and 169 to 183 hours (approximately 7 days), respectively, and was independent of dose. In a population pharmacokinetic analysis, the apparent total clearance (CL/F) of oral rolapitant was 0.96 L/hour in cancer patients. The total clearance following intravenous administration of 166.5 mg rolapitant in healthy subjects was 1.65 L/hour.
Metabolism
Rolapitant is metabolized primarily by CYP3A4 to form a major active metabolite, M19 (C4-pyrrolidine-hydroxylated rolapitant). In a mass balance study, the metabolite M19 was the major circulating metabolite. The formation of M19 was significantly delayed with the median Tmax of 120 hours (range: 24-168 hours) with Cmax of 183 ng/mL. The mean half-life of M19 was 158 hours.
The exposure ratio of M19 to rolapitant was approximately 50% in plasma.
The Cmax and AUC of M19 following single intravenous administration of 166.5 mg of rolapitant were 149 ng/mL and 68,600 ng·h/mL, respectively. The median Tmax was 168 hours. The mean half-life was 182 hours.
Excretion
Rolapitant is eliminated primarily through the hepatic/biliary route. Following administration of a single oral 180-mg dose of [14C]-rolapitant, on average 14.2% (range 9% to 20%) and 73% (range 52% to 89%) of the dose was recovered in the urine and feces, respectively over 6 weeks. In pooled samples collected over 2 weeks, 8.3% of the dose was recovered in the urine primarily as metabolites and 37.8% of the dose was recovered in the feces primarily as unchanged rolapitant. Unchanged rolapitant or M19 were not found in pooled urine sample.
Specific Populations
Age, Male And Female Patients And Racial Or Ethnic Groups
Population pharmacokinetic analyses indicated that age, sex and race had no significant impact on the pharmacokinetics of rolapitant.
Patients With Hepatic Impairment
Following administration of a single oral dose of 180 mg rolapitant to patients with mild hepatic impairment (Child-Pugh Class A), the pharmacokinetics of rolapitant were comparable with those of healthy subjects. In patients with moderate hepatic impairment (Child-Pugh Class B), the mean Cmax was 25% lower while mean AUC of rolapitant was similar compared to those of healthy subjects. The median Tmax for M19 was delayed to 204 hours in patients with mild or moderate hepatic impairment compared to 168 hours in healthy subjects. The pharmacokinetics of rolapitant were not studied in patients with severe hepatic impairment (Child-Pugh Class C) [see Use In Specific Populations].
Patients With Renal Impairment
In population pharmacokinetic analyses, creatinine clearance (CLcr) at baseline did not show a significant effect on rolapitant pharmacokinetics in cancer patients with mild (CLcr: 60 to 90 mL/min) or moderate (CLcr: 30 to 60 mL/min) renal impairment compared to cancer patients with normal kidney function. Information is insufficient for the effect of severe renal impairment. The pharmacokinetics of rolapitant was not studied in patients with end-stage renal disease requiring hemodialysis.
Drug Interaction Studies
Effect Of Other Drugs On Rolapitant
Rolapitant is a substrate for CYP3A4.
CYP3A4 inducers
When 600 mg rifampin was administered once daily for 7 days before and 7 days after administration of a single oral dose of 180 mg rolapitant, the mean Cmax of rolapitant was reduced by 30% and the mean AUC was reduced by 85% compared to administration of rolapitant alone. The mean half-life of rolapitant decreased from 176 hours without rifampin to 41 hours with concurrent rifampin [see DRUG INTERACTIONS].
CYP3A4 inhibitors
Concurrent administration of 400 mg ketoconazole, a strong CYP3A4 inhibitor, once daily for 21 days following a single 90 mg oral dose of rolapitant, did not affect the Cmax of rolapitant while the AUC increased by 21%. These pharmacokinetic differences are not clinically significant.
Effect Of Rolapitant On Other Drugs
The effect of VARUBI on CYP450 enzymes and transporters is summarized below.
CYP3A4 substrates
Rolapitant is neither an inhibitor nor an inducer of CYP3A4.
Midazolam: A single oral dose of 180 mg rolapitant had no significant effects on the pharmacokinetics of midazolam when oral midazolam 3 mg was co-administered on Day 1 and administered alone on Days 6 and 9. A single intravenous dose of 166.5 mg rolapitant had no significant effects on the pharmacokinetics of midazolam when oral midazolam 3 mg was co-administered on Day 1 and administered alone on Day 8.
Ondansetron: Rolapitant had no significant effects on the pharmacokinetics of intravenous ondansetron when concomitantly administered with a single 180 mg oral dose of rolapitant on the same day.
Dexamethasone: Rolapitant had no significant effects on the pharmacokinetics of dexamethasone when oral dexamethasone was administered on Days 1 to 3 after a single 180 mg oral dose of rolapitant was co-administered on Day 1 [see DOSAGE AND ADMINISTRATION].
CYP2D6 substrates
Rolapitant is a moderate inhibitor of CYP2D6 [see CONTRAINDICATIONS, WARNINGS AND PRECAUTIONS, and DRUG INTERACTIONS]. See Table 4 and Table 5 for a summary of the effects of VARUBI on the pharmacokinetics of co-administered dextromethorphan, a CYP2D6 substrate.
Table 4: Effect of Oral Rolapitant on the Systemic Exposure of Co-administered CYP2D6 Substrate (Dextromethorphan)*
Rolapitant Dose |
% Change for Dextromethorphan |
Day 1 with rolapitant |
Day 8 without rolapitant |
Change in Cmax |
Change in AUC |
Change in Cmax |
Change in AUC |
180 mg |
120% ↑ |
160% ↑ |
180% ↑ |
230% ↑ |
*A single oral dose of 180 mg rolapitant was administered on Day 1; the interacting drug (dextromethorphan 30 mg) was administered orally on Day 1 with rolapitant and alone on Day 8.
↑ Denotes a mean increase in exposure by the percentage indicated. |
Table 5: Effect of Intravenous Rolapitant on the Systemic Exposure of Co-administered CYP2D6 Substrate (Dextromethorphan)*
Rolapitant Dose |
% Change for Dextromethorphan |
Day 1 with rolapitant |
Day 8 without rolapitant |
Day 15 without rolapitant |
Day 22 without rolapitant |
Day 29 without rolapitant |
Change in Cmax |
Change in AUC |
Change in Cmax |
Change in AUC |
Change in Cmax |
Change in AUC |
Change in Cmax |
Change in AUC |
Change in Cmax |
Change in AUC |
166.5 mg |
75% ↑ |
110% ↑ |
140% ↑ |
220% ↑ |
170% ↑ |
220% ↑ |
120% ↑ |
180% ↑ |
96% ↑ |
130% ↑ |
*A single intravenous dose of 166.5 mg rolapitant was administered on Day 1; the interacting drug (dextromethorphan 30 mg) was administered orally on Day 1 with rolapitant and alone on Days 8, 15, 22, and 29.
↑ Denotes a mean increase in exposure by the percentage indicated. |
BCRP transporter
In vitro, rolapitant is a BCRP transporter inhibitor.
When sulfasalazine (BCRP substrate) was administered with a single intravenous dose of 166.5 mg rolapitant on Day 1 and without rolapitant on Day 11, no effect on Cmax and AUC of sulfasalazine 500 mg was observed on Day 1, an 18% decrease in Cmax and an 11% decrease in AUC were observed on Day 11. These differences in systemic exposures are not clinically significant.
When sulfasalazine was administered with a single oral dose of 180 mg rolapitant on Day 1 and without rolapitant on Day 8, a 140% increase in Cmax and a 130% increase in AUC of sulfasalazine 500 mg was observed on Day 1, a 17% increase in Cmax and a 32% increase in AUC was observed on Day 8 [see DRUG INTERACTIONS].
P-glycoprotein substrates
In vitro, rolapitant is a P-gp inhibitor.
When digoxin (P-gp substrate) was administered with a single oral dose of 180 mg rolapitant, a 70% increase in Cmax and a 30% increase in AUC of digoxin 0.5 mg were observed [see DRUG INTERACTIONS].
When digoxin was administered with a single intravenous dose of 166.5 mg rolapitant, no effect on AUC and a 21% increase in the Cmax of digoxin 0.5 mg was observed [see DRUG INTERACTIONS].
Warfarin
When warfarin was administered with a single intravenous dose of 166.5 mg rolapitant, 3% and 18% increases in Cmax and AUC of S-warfarin were observed on Day 1, respectively. On Day 8, the increases were 3% for Cmax and 21% for AUC. The effect on INR or prothrombin time was not measured [see DRUG INTERACTIONS].
Substrates for other CYP enzymes
In vitro studies suggest that rolapitant is not an inhibitor of CYP1A2 and CYP2E1. In vitro studies suggest that rolapitant inhibits CYP2A6; however, a clinically meaningful drug interaction via an inhibition of CYP2A6 appears unlikely.
No clinically significant interaction was seen on the systemic exposures of the following drugs when administered with a single intravenous dose of 166.5 mg rolapitant on Day 1 and without rolapitant on Day 8: caffeine (CYP1A2 substrate; no effect on caffeine 200 mg on Days 1 and 8), and omeprazole (CYP2C19 substrate; 14% increase in Cmax and no effect on AUC of omeprazole 40 mg on Day 1; on Day 8: 15% increase in Cmax and no effect on AUC).
No clinically significant interaction was seen on the systemic exposures of the following drugs when administered with a single oral dose of 180 mg rolapitant on Day 1: repaglinide (CYP2C8 substrate; no effect on repaglinide 0.25 mg on Day 1; on Day 8: 29% and 24% increase in Cmax and AUC, respectively), efavirenz (CYP2B6 substrate; 18% decrease in Cmax and no effect on AUC of efavirenz 600 mg on Day 1; on Day 8: no effect on Cmax and 28% increase in AUC), tolbutamide (CYP2C9 substrate; no effect on tolbutamide 500 mg on Day 1 and on Day 8), or omeprazole (CYP2C19 substrate; 44% increase in Cmax and 23% increase in AUC of omeprazole 40 mg on Day 1; on Day 8: 37% and 15% increase in Cmax and AUC, respectively).
Substrates for other transporters
In vitro studies suggest that oral rolapitant is unlikely to inhibit organic anion transporting polypeptides 1B1 and 1B3 (OATP1B1 and OATP1B3), organic anion transporters 1 and 3 (OAT1 and OAT3), organic cation transporter 2 (OCT2), and multidrug and toxin extrusion proteins 1 and 2K (MATE1 and MATE2K) in vivo.
In vitro studies suggest that intravenous rolapitant is unlikely to inhibit OATP1B3, OAT1, OAT3, OCT2, and MATE2K in vivo. However, the potential for intravenous rolapitant to inhibit OATP1B1 or MATE1 in vivo cannot be ruled out.
Clinical Studies
Cisplatin-Based Highly Emetogenic Chemotherapy (HEC)
In two multicenter, randomized, double-blind, parallel group, controlled clinical studies (Study 1 and Study 2), the VARUBI regimen (VARUBI tablets, granisetron and dexamethasone) was compared with control therapy (placebo, granisetron and dexamethasone) in patients receiving a chemotherapy regimen that included cisplatin >60 mg/m2. See Table 6 for the treatment regimens.
Table 6: Treatment Regimens in Studies 1 and 2
|
Day 1 |
Day 2 to 4 |
VARUBI Regimen |
Oral VARUBI† |
180 mg |
None |
Oral Dexamethasone |
20 mg‡ |
8 mg twice daily |
Intravenous Granisetron |
10 mcg/kg§ |
None |
Control Regimen* |
Oral Dexamethasone |
20 mg‡ |
8 mg twice daily |
Intravenous Granisetron |
10 mcg/kg§ |
None |
VARUBI placebo was used to maintain blinding.
† VARUBI was administered 1 to 2 hours prior to chemotherapy treatment on Day 1.
‡ Dexamethasone was administered 30 minutes prior to chemotherapy on Day 1. There is no drug interaction between VARUBI and dexamethasone, so no dosage adjustment for dexamethasone is required [see CLINICAL PHARMACOLOGY].
§ The dose of granisetron was administered 30 minutes prior to chemotherapy on Day 1. |
Study 1
A total of 532 patients were randomized to either the VARUBI regimen (N=266) or control therapy (N=266). A total of 526 patients were included in the evaluation of efficacy. Of those randomized 42% were women, 58% men, 67% White, 23% Asian, 1% Black, and 9% multi-racial/other/unknown. The proportion of patients from North America was 16%. Patients in this clinical study ranged from 20 to 90 years of age, with a mean age of 57 years. In Study 1, 26% of patients were 65 years or older, with 3% of patients being 75 years or older. The mean cisplatin dose was 77 mg/m2.
During this study, 82% of the patients received a concomitant chemotherapeutic agent in addition to protocol-mandated cisplatin. The most common concomitant chemotherapeutic agents administered during Cycle 1 were: gemcitabine (17%), paclitaxel (12%), fluorouracil (11%), etoposide (10%), vinorelbine (9%), docetaxel (9%), pemetrexed (7%), doxorubicin (6%) and cyclophosphamide (5%).
Study 2
A total of 555 patients were randomized to either the VARUBI regimen (N=278) or control therapy (N=277). A total of 544 patients were included in the evaluation of efficacy. Of those randomized 32% were women, 68% men, 81% White, 14% Asian, 1% Black, and 5% multi-racial/other/unknown. The proportion of patients from North America was 7%. Patients in this clinical study ranged from 18 to 83 years of age, with a mean age of 58 years. In this study, 27% of patients were 65 years or older, with 3% of patients being 75 years or older. The mean cisplatin dose was 76 mg/m2.
During this study, 85% of the patients received a concomitant chemotherapeutic agent in addition to protocol-mandated cisplatin. The most common concomitant chemotherapeutic agents administered during Cycle 1 were: vinorelbine (16%), gemcitabine (15%), fluorouracil (12%), etoposide (11%), pemetrexed (9%), docetaxel (7%), paclitaxel (7%), epirubicin (5%) and capecitabine (4%).
The primary endpoint in both studies was complete response (defined as no emetic episodes and no rescue medication) in the delayed phase (25 to 120 hours) of chemotherapy-induced nausea and vomiting.
Moderately Emetogenic Chemotherapy (MEC) And Combinations Of Anthracycline And Cyclophosphamide Chemotherapy
Study 3
In Study 3, a multicenter, randomized, double-blind, parallel group, controlled clinical study in moderately emetogenic chemotherapy, the VARUBI regimen (VARUBI tablets, granisetron and dexamethasone) was compared with control therapy (placebo, granisetron and dexamethasone) in patients receiving a moderately emetogenic chemotherapy regimen that included at least 50% of patients receiving a combination of anthracycline and cyclophosphamide. The percentage of patients who received carboplatin in Cycle 1 was 30%. Treatment regimens for the VARUBI and control arms are summarized in Table 7.
Table 7: Treatment Regimens in Study 3
|
Day 1 |
Day 2 to 3 |
VARUBI Regimen |
Oral VARUBI† |
180 mg |
none |
Oral Dexamethasone |
20 mg‡ |
none |
Oral Granisetron |
2 mg§ |
2 mg once daily |
Control Regimen* |
Oral Dexamethasone |
20 mg‡ |
none |
Oral Granisetron |
2 mg§ |
2 mg once daily |
* VARUBI placebo was used to maintain blinding.
† VARUBI was administered 1 to 2 hours prior to chemotherapy treatment on Day 1.
‡ Dexamethasone was administered 30 minutes prior to chemotherapy on Day 1. There is no drug interaction between VARUBI and dexamethasone, so no dosage adjustment for dexamethasone is required [see CLINICAL PHARMACOLOGY].
§ The dose of granisetron was administered 30 minutes prior to chemotherapy on Day 1. |
A total of 1369 patients were randomized to either the VARUBI regimen (N=684) or control therapy (N=685). A total of 1332 patients were included in the evaluation of efficacy. Of those randomized 80% were women, 20% men, 77% White, 13% Asian, 4% Black, and 6% multi-racial/other/unknown. The proportion of patients from North America was 33%. Patients in this clinical study ranged from 22 to 88 years of age, with a mean age of 57 years. In this study, 28% of patients were 65 years or older, with 7% of patients being 75 years or older.
The primary endpoint was complete response (defined as no emetic episodes and no rescue medication) in the delayed phase (25 to 120 hours) of chemotherapy-induced nausea and vomiting.
A summary of the study results from HEC Studies 1 and 2 and for the MEC Study 3 is shown in Table 8.
Table 8: Percent of Patients Receiving Emetogenic Chemotherapy Responding by Treatment Group for the HEC Studies 1 and 2 and for the MEC Study 3
Endpoint |
HEC Study 1 |
HEC Study 2 |
MEC Study 3 |
|
VARUBI†
(N=264) Rate (%) |
Control†
(N=262) Rate (%) |
P-Value Treatment Difference
(95% C.I.) |
VARUBI†
(N=271) Rate (%) |
Control†
(N=273) Rate (%) |
P-Value Treatment Difference
(95% C.I.) |
VARUBI†
(N=666) Rate (%) |
Control†
(N=666) Rate (%) |
P-Value Treatment Difference
(95% C.I.) |
Primary Endpoint: Complete Response in the Delayed Phase |
72.7 |
58.4 |
<0.001* 14.3 (6.3, 22.4) |
70.1 |
61.9 |
0.043* 8.2 (0.3, 16.1) |
71.3 |
61.6 |
<0.001* 9.8 (4.7, 14.8) |
† Granisetron and dexamethasone were used as concomitant drugs.
* Results were obtained based on the Cochran-Mantel-Haenszel test stratified by gender. |
Multiple-Cycle Extension: In Studies 1, 2, and 3, patients had the option of continuing into a multiple-cycle extension for up to 5 additional cycles of chemotherapy receiving the same treatment as assigned in cycle 1. At Day 6 to 8 following initiation of chemotherapy, patients were asked to recall whether they had any episode of vomiting or retching or nausea that interfered with normal daily life. The results are summarized by study and treatment group in Figure 1 below.
Figure 1: No Emesis and No Nausea Interfering with Daily Life over Cycles 2-6
Study 1
Study 2
Study 3