Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Cell Host Microbe ; 31(4): 634-649.e8, 2023 04 12.
Article in English | MEDLINE | ID: mdl-37003258

ABSTRACT

Drug platforms that enable the directed delivery of therapeutics to sites of diseases to maximize efficacy and limit off-target effects are needed. Here, we report the development of PROT3EcT, a suite of commensal Escherichia coli engineered to secrete proteins directly into their surroundings. These bacteria consist of three modular components: a modified bacterial protein secretion system, the associated regulatable transcriptional activator, and a secreted therapeutic payload. PROT3EcT secrete functional single-domain antibodies, nanobodies (Nbs), and stably colonize and maintain an active secretion system within the intestines of mice. Furthermore, a single prophylactic dose of a variant of PROT3EcT that secretes a tumor necrosis factor-alpha (TNF-α)-neutralizing Nb is sufficient to ablate pro-inflammatory TNF levels and prevent the development of injury and inflammation in a chemically induced model of colitis. This work lays the foundation for developing PROT3EcT as a platform for the treatment of gastrointestinal-based diseases.


Subject(s)
Colitis , Single-Domain Antibodies , Animals , Mice , Escherichia coli , Colitis/chemically induced , Colitis/therapy , Tumor Necrosis Factor-alpha/metabolism
2.
Lancet Oncol ; 23(6): 758-767, 2022 06.
Article in English | MEDLINE | ID: mdl-35588752

ABSTRACT

BACKGROUND: Treatment options for malignant pleural mesothelioma are scarce. Tazemetostat, a selective oral enhancer of zeste homolog 2 (EZH2) inhibitor, has shown antitumour activity in several haematological cancers and solid tumours. We aimed to evaluate the anti-tumour activity and safety of tazemetostat in patients with measurable relapsed or refractory malignant pleural mesothelioma. METHODS: We conducted an open-label, single-arm phase 2 study at 16 hospitals in France, the UK, and the USA. Eligible patients were aged 18 years or older with malignant pleural mesothelioma of any histology that was relapsed or refractory after treatment with at least one pemetrexed-containing regimen, an Eastern Cooperative Oncology Group performance status of 0 or 1, and a life expectancy of greater than 3 months. In part 1 of the study, participants received oral tazemetostat 800 mg once on day 1 and then twice daily from day 2 onwards. In part 2, participants received oral tazemetostat 800 mg twice daily starting on day 1 of cycle 1, using a two-stage Green-Dahlberg design. Tazemetostat was administered in 21-day cycles for approximately 17 cycles. The primary endpoint of part 1 was the pharmacokinetics of tazemetostat and its metabolite at day 15 after administration of 800 mg tazemetostat, as measured by maximum serum concentration (Cmax), time to Cmax (Tmax), area under the concentration-time curve (AUC) to day 15 (AUC0-t), area under the curve from time 0 extrapolated to infinity (AUC0-∞), and the half-life (t1/2) of tazemetostat, assessed in all patients enrolled in part 1. The primary endpoint of part 2 was the disease control rate (the proportion of patients with a complete response, partial response, or stable disease) at week 12 in patients with malignant pleural mesothelioma per protocol with BAP1 inactivation determined by immunohistochemistry. The safety population included all the patients who had at least one post-dose safety assessment. This trial is now complete and is registered with ClinicalTrials.gov, NCT02860286. FINDINGS: Between July 29, 2016, and June 2, 2017, 74 patients were enrolled (13 in part 1 and 61 in part 2) and received tazemetostat, 73 (99%) of whom had BAP1-inactivated tumours. In part 1, following repeat dosing of tazemetostat at steady state, on day 15 of cycle 1, the mean Cmax was 829 ng/mL (coefficient of variation 56·3%), median Tmax was 2 h (range 1-4), mean AUC0-twas 3310 h·ng/mL (coefficient of variation 50·4%), mean AUC0-∞ was 3180 h·ng/mL (46·6%), and the geometric mean t1/2 was 3·1 h (13·9%). After a median follow-up of 35·9 weeks (IQR 20·6-85·9), the disease control rate in part 2 in patients with BAP1-inactivated malignant pleural mesothelioma was 54% (95% CI 42-67; 33 of 61 patients) at week 12. No patients had a confirmed complete response. Two patients had a confirmed partial response: one had an ongoing partial response with a duration of 18 weeks and the other had a duration of 42 weeks. The most common grade 3-4 treatment-emergent adverse events were hyperglycaemia (five [7%] patients), hyponatraemia (five [7%]), and anaemia (four [5%]); serious adverse events were reported in 25 (34%) of 74 patients. Five (7%) of 74 patients died while on study; no treatment-related deaths occurred. INTERPRETATION: Further refinement of biomarkers for tazemetostat activity in malignant pleural mesothelioma beyond BAP1 inactivation could help identify a subset of tumours that are most likely to derive prolonged benefit or shrinkage from this therapy. FUNDING: Epizyme.


Subject(s)
Mesothelioma, Malignant , Mesothelioma , Neoplasms , Benzamides/adverse effects , Biphenyl Compounds , Enhancer of Zeste Homolog 2 Protein/genetics , Enzyme Inhibitors/therapeutic use , Humans , Mesothelioma/drug therapy , Mesothelioma/pathology , Morpholines/therapeutic use , Neoplasms/chemically induced , Pyridones , Tumor Suppressor Proteins , Ubiquitin Thiolesterase
3.
Clin Pharmacol Drug Dev ; 8(7): 850-860, 2019 10.
Article in English | MEDLINE | ID: mdl-31418538

ABSTRACT

Population pharmacokinetics of rolapitant and its active metabolite M19 were studied in 482 patients receiving this neurokinin-1 receptor antagonist in combination with a 5-hydroxytryptamine-3 receptor antagonist and dexamethasone for prevention of chemotherapy-induced nausea and vomiting (CINV). Patients received a single dose of rolapitant (range, 9-180 mg) before administration of moderately or highly emetogenic chemotherapy. Population pharmacokinetic analysis was performed via nonlinear mixed-effects modeling. Rolapitant pharmacokinetics was best characterized by a 2-compartment model. Population typical values were estimated to be 0.962 L/h for apparent oral clearance and 214 L for central compartment volume of distribution. The intercompartment clearance and peripheral compartment volume of distribution was estimated to be 2.79 L/h and 164 L, respectively. Metabolite M19 pharmacokinetics was described by a 1-compartment model with an apparent metabolite clearance of 1.83 L/h. Intersubject variability was moderate for pharmacokinetics parameters. Weight positively correlated with central compartment volume of distribution and peripheral compartment volume of distribution but not with apparent oral clearance. No other demographic, clinical, or pathophysiologic covariates, including liver and renal function, influenced rolapitant pharmacokinetics. A slight positive trend was observed between rolapitant exposure and efficacy (ie, complete response defined as no emesis and no use of rescue medication) in the delayed phase of CINV (>24-120 hours after chemotherapy). This further supports the 180-mg dose of rolapitant in CINV patients. In summary, this validated population pharmacokinetic model satisfactorily describes pharmacokinetics of rolapitant and M19 in patients with CINV. These results support the recommendation that no dose adjustment for patient variables investigated is necessary.


Subject(s)
Antineoplastic Agents/administration & dosage , Nausea/prevention & control , Spiro Compounds/pharmacokinetics , Vomiting/prevention & control , Administration, Oral , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Dexamethasone/administration & dosage , Dexamethasone/therapeutic use , Double-Blind Method , Female , Humans , Male , Metabolic Clearance Rate , Middle Aged , Nausea/chemically induced , Spiro Compounds/administration & dosage , Vomiting/chemically induced , Young Adult
4.
Clin Pharmacol Drug Dev ; 8(2): 152-159, 2019 02.
Article in English | MEDLINE | ID: mdl-30624856

ABSTRACT

Rolapitant, a selective and long-acting neurokinin-1 receptor antagonist, is approved in an oral formulation for prevention of delayed chemotherapy-induced nausea and vomiting in adults. This pivotal open-label, randomized, single-dose, multicenter, parallel-group study assessed the bioequivalence of a single oral dose of 180 mg of rolapitant administered in tablet (2 × 90-mg tablets) or capsule (4 × 45-mg capsules) form in healthy male and female subjects. Blood samples for pharmacokinetic analysis were collected predose and at times up to 912 hours postdose. The rolapitant tablet was considered bioequivalent to the rolapitant capsule if the 90% confidence intervals for the ratios of the geometric means for rolapitant, observed maximum plasma concentration (Cmax ), and area under the curve from time 0 extrapolated to infinity (AUC0-∞ ) were within the 0.80-1.25 range. The pharmacokinetic profiles of the capsule group (n = 43) and tablet group (n = 44) were similar. The geometric mean ratios of Cmax and AUC0-∞ were 0.99 (0.89-1.11) and 1.05 (0.92-1.19), respectively, establishing bioequivalence of the rolapitant tablet and capsule formulations. Both formulations were well tolerated, with a similar incidence of treatment-emergent adverse events in the 2 groups.


Subject(s)
Spiro Compounds/administration & dosage , Spiro Compounds/pharmacokinetics , Administration, Oral , Adult , Area Under Curve , Capsules , Chromatography, Liquid , Female , Healthy Volunteers , Humans , Male , Middle Aged , Tablets , Tandem Mass Spectrometry , Therapeutic Equivalency , Young Adult
5.
Invest New Drugs ; 37(1): 139-146, 2019 02.
Article in English | MEDLINE | ID: mdl-30032410

ABSTRACT

Rolapitant is a neurokinin-1 receptor antagonist that is approved in combination with other antiemetic agents in adults for the prevention of delayed nausea and vomiting (CINV) associated with initial and repeat courses of emetogenic cancer chemotherapy, including but not limited to highly emetogenic chemotherapy. Here, we assessed the absorption, metabolism, and excretion of 14C-labeled rolapitant in healthy male subjects. Rolapitant was administered as a single 180-mg oral dose containing approximately 100 µCi of total radioactivity, with plasma, urine, and fecal samples collected at defined intervals after dosing. Rolapitant had a large apparent volume of distribution, indicating that it is widely distributed into body tissues. Rolapitant was slowly metabolized and eliminated with a mean half-life of 186 h. Exposure to the major metabolite of rolapitant, C4-pyrrolidinyl hydroxylated rolapitant or M19, was approximately 50% of rolapitant exposure in plasma. Renal clearance was not a significant elimination route for rolapitant-related entities. Total radioactivity recovered in urine accounted for 14.2% of the dose, compared to 72.7% recovery in feces. Adverse events (AEs) were generally mild; there were no serious AEs, and no clinically significant changes in laboratory or electrocardiogram parameters were observed. The combination of rolapitant safety, its long half-life, extensive tissue distribution, and slow elimination via the hepatobiliary route (rather than renal excretion) suggest suitability that a single dose of rolapitant may provide protection against CINV beyond the first 24 h after chemotherapy administration.


Subject(s)
Antiemetics/administration & dosage , Nausea/metabolism , Neurokinin-1 Receptor Antagonists/administration & dosage , Spiro Compounds/administration & dosage , Vomiting/metabolism , Adult , Antiemetics/pharmacokinetics , Healthy Volunteers , Humans , Male , Middle Aged , Nausea/drug therapy , Neurokinin-1 Receptor Antagonists/pharmacokinetics , Spiro Compounds/pharmacokinetics , Tissue Distribution , Vomiting/drug therapy
6.
Clin Pharmacol Drug Dev ; 8(2): 160-171, 2019 02.
Article in English | MEDLINE | ID: mdl-29905976

ABSTRACT

Rolapitant is a selective and long-acting neurokinin-1 receptor antagonist approved in an oral formulation in combination with dexamethasone and a 5-hydroxytryptamine type 3 receptor antagonist for the prevention of delayed chemotherapy-induced nausea and vomiting in adults. The pharmacokinetic and safety profiles of intravenous (IV) rolapitant were evaluated in two open-label, phase 1 trials in healthy subjects. Single ascending dose (SAD) and multiple ascending dose studies were conducted in one trial (PR-11-5012-C), and a supratherapeutic SAD study was conducted in a separate trial (PR-11-5022-C). In the SAD and supratherapeutic studies, rolapitant maximum plasma concentration, area under the plasma drug concentration-time curve (AUC) from time zero to time of last measured concentration, and AUC from time zero to infinity increased dose-proportionally following single IV infusions of 18 to 270 mg. In the multiple ascending dose study, following 10 daily IV infusions of rolapitant 18, 36, or 54 mg, the mean day 10:day 1 maximum concentration ratio was 1.97, 1.52, and 2.07, respectively, and the mean day 10:day 1 ratio of AUC from 0 to 24 hours was 4.30, 4.59, and 5.38, respectively, indicating drug accumulation over time. Across all studies, rolapitant was gradually eliminated from plasma, with a half-life of 135-231 hours. Rolapitant was safe and well tolerated across all studies, with no serious or severe rolapitant-related treatment-emergent adverse events. The most common rolapitant-related treatment-emergent adverse events were headache, dry mouth, and dizziness, which were predominantly mild in severity. Overall, the pharmacokinetic and safety profiles of IV rolapitant were consistent with those of the oral formulation.


Subject(s)
Spiro Compounds/administration & dosage , Spiro Compounds/pharmacokinetics , Administration, Intravenous , Adult , Area Under Curve , Female , Half-Life , Healthy Volunteers , Humans , Male , Middle Aged , Spiro Compounds/adverse effects , Young Adult
7.
Clin Pharmacol Drug Dev ; 8(5): 603-611, 2019 07.
Article in English | MEDLINE | ID: mdl-30256537

ABSTRACT

This 2-part study evaluated the QT/QTc prolongation potential and safety and pharmacokinetics of the antiemetic rolapitant, a neurokinin-1 receptor antagonist. Part 1 was a randomized, placebo-controlled single-dose-escalation study assessing the safety of a single high dose of rolapitant. Part 2 was a randomized, placebo- and positive-controlled, double-blind parallel-group study including 4 treatment arms: rolapitant at the highest safe dose established in part 1, placebo, moxifloxacin 400 mg (positive control), and rolapitant at the presumed therapeutic dose (180 mg). Among 184 adults, rolapitant was absorbed following oral administration under fasting conditions, with a median Tmax of 4 to 6 hours (range, 2-8 hours) and was safe at all doses up to 720 mg. No differences in mean change in QTcF were observed between placebo and rolapitant from baseline or at any point. At any point, the upper bound of the confidence interval for the mean difference between placebo and rolapitant was no greater than 4.4 milliseconds, and the mean difference between placebo and rolapitant was no greater than 1.7 milliseconds, suggesting an insignificant change in QTc with rolapitant. Rolapitant is safe and does not prolong the QT interval at doses up to 720 mg relative to placebo in healthy adults.


Subject(s)
Antiemetics/pharmacology , Heart Rate/drug effects , Spiro Compounds/pharmacology , Adolescent , Adult , Antiemetics/pharmacokinetics , Double-Blind Method , Electrocardiography/drug effects , Female , Healthy Volunteers , Humans , Male , Middle Aged , Neoplasms/drug therapy , Palliative Care , Spiro Compounds/pharmacokinetics , Young Adult
8.
Support Care Cancer ; 27(3): 819-827, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30084103

ABSTRACT

PURPOSE: Rolapitant is a neurokinin-1 receptor antagonist indicated in combination with other antiemetic agents in adults for the prevention of delayed chemotherapy-induced nausea and vomiting. We evaluated the effects of rolapitant oral on the pharmacokinetics of probe substrates for cytochrome P450 (CYP) 2D6 (dextromethorphan), 2C9 (tolbutamide), 2C19 (omeprazole), 2B6 (efavirenz), and 2C8 (repaglinide) in healthy subjects. METHODS: This open-label, multipart, randomized, phase 1 study assessed cohorts of 20-26 healthy subjects administered dextromethorphan, tolbutamide plus omeprazole, efavirenz, or repaglinide with and without single, oral doses of rolapitant. Maximum plasma analyte concentrations (Cmax) and area under the plasma analyte concentration-time curves (AUC) were estimated using noncompartmental analysis, and geometric mean ratios (GMRs) and 90% confidence intervals for the ratios of test (rolapitant plus probe substrate) to reference (probe substrate alone) treatment were calculated. RESULTS: Rolapitant significantly increased the systemic exposure of dextromethorphan in terms of Cmax and AUC0-inf by 2.2- to 3.3-fold as observed in GMRs on days 7 and 14. Rolapitant did not affect systemic exposure of tolbutamide, and minor excursions outside of the 80-125% no effect limits were detected for omeprazole, efavirenz, and repaglinide. CONCLUSIONS: Inhibition of dextromethorphan by a single oral dose of rolapitant 180 mg is clinically significant and can last at least 7 days. No clinically significant interaction was observed between rolapitant and substrates of CYP2C9, CYP2C19, CYP2B6, or CYP2C8. CYP2D6 substrate drugs with a narrow therapeutic index may require monitoring for adverse reactions if given concomitantly with rolapitant.


Subject(s)
Antiemetics/pharmacology , Neurokinin-1 Receptor Antagonists/pharmacology , Spiro Compounds/pharmacology , Administration, Oral , Adolescent , Adult , Alkynes , Benzoxazines/pharmacokinetics , Carbamates/pharmacokinetics , Cyclopropanes , Cytochrome P-450 CYP2B6/drug effects , Cytochrome P-450 CYP2C19/drug effects , Cytochrome P-450 CYP2C8/drug effects , Cytochrome P-450 CYP2C9/drug effects , Cytochrome P-450 CYP2D6/drug effects , Dextromethorphan/pharmacokinetics , Drug Combinations , Drug Interactions , Female , Healthy Volunteers , Humans , Male , Middle Aged , Molecular Probes/pharmacokinetics , Neurokinin-1 Receptor Antagonists/administration & dosage , Omeprazole/pharmacokinetics , Piperidines/pharmacokinetics , Tolbutamide/pharmacokinetics , Young Adult
9.
J Clin Pharmacol ; 58(8): 1074-1083, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29693712

ABSTRACT

Rolapitant is a selective, long-acting neurokinin-1 receptor antagonist, approved in the United States and Europe for prevention of delayed chemotherapy-induced nausea and vomiting in adults. This open-label study evaluated the effects of a new intravenous formulation of rolapitant on cytochrome P450 (CYP) enzyme (CYP3A, CYP1A2, CYP2C9, CYP2C19, and CYP2D6) activity. On days 1 and 14, 36 healthy volunteers received a modified Cooperstown cocktail (midazolam 3 mg [CYP3A substrate], caffeine 200 mg [CYP1A2 substrate], S-warfarin 10 mg [CYP2C9 substrate] + vitamin K 10 mg, omeprazole 40 mg [CYP2C19 substrate], and dextromethorphan 30 mg [CYP2D6 substrate]). On day 7, subjects received the modified Cooperstown cocktail after 166.5-mg rolapitant infusion. On days 21, 28, and 35, subjects received oral dextromethorphan. Maximum plasma concentration (Cmax ) and area under the plasma concentration-time curve (AUC0-last ) of probe drugs post- vs pre-rolapitant administration were assessed using geometric least-squares mean ratios (GMRs) with 90%CIs. The 90%CIs of the GMRs were within the 0.80-1.25 no-effect limits for caffeine and S-warfarin Cmax and AUC0-last . For midazolam Cmax and AUC0-last and omeprazole Cmax , the 90%CIs of the GMRs were marginally outside these limits. Intravenous rolapitant coadministration increased dextromethorphan exposure, peaking 14 days post-rolapitant administration (GMRs: Cmax , 2.74, 90%CI 2.21-3.40; AUC0-last , 3.36, 90%CI 2.74-4.13). Intravenous rolapitant 166.5 mg and probe drugs were well tolerated when coadministered. These data suggest that intravenous rolapitant is not an inhibitor of CYP3A, CYP2C9, CYP2C19, or CYP1A2 but is a moderate inhibitor of CYP2D6.

10.
Cancer Chemother Pharmacol ; 81(3): 497-503, 2018 03.
Article in English | MEDLINE | ID: mdl-29322231

ABSTRACT

PURPOSE: Niraparib is a highly selective inhibitor of PARP-1 and PARP-2 approved in the United States for maintenance treatment of adult patients with recurrent ovarian cancer in complete or partial response to platinum-based chemotherapy. In this open-label crossover study, we evaluated the effects of food on niraparib pharmacokinetics (PK) and safety. METHODS: Patients received a single 300-mg dose of niraparib either after a high-fat meal or under fasting conditions. After a 7-day PK assessment, all patients received a second 300-mg dose of niraparib under the opposite condition, followed by 7-day PK assessment. Blood samples for PK analyses were collected at baseline (on days 1 and 8) and up to 168 h post-dose. Bioequivalence between conditions was defined by the 90% confidence intervals (CIs) for area under the plasma concentration-time curve (AUC) from 0 to last measurable concentration (AUC0-last) and from 0 to infinity (AUC0-∞) being within the 80-125% range. RESULTS: The high-fat meal/fasting ratios of geometric least-squares means for AUC0-last and AUC0-∞ were 106.8 (90% CI 97.8-116.6) and 110.1 (90% CI 99.7-121.6), respectively, indicating bioequivalence between conditions. Mean half-life, maximum plasma concentration (Cmax), and time to Cmax after the high-fat meal were similar to, 27% smaller than, and 128% greater than after fasting, respectively. Adverse events were similar between conditions. CONCLUSIONS: A high-fat meal did not impact the PK profile of niraparib, indicating that niraparib can be taken with or without food. Niraparib was safe and well-tolerated.


Subject(s)
Dietary Fats/metabolism , Indazoles , Neoplasm Recurrence, Local , Ovarian Neoplasms , Piperidines , Adult , Aged , Area Under Curve , Drug Monitoring/methods , Fasting/metabolism , Female , Food-Drug Interactions , Humans , Indazoles/administration & dosage , Indazoles/pharmacokinetics , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Outcome Assessment, Health Care , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Piperidines/administration & dosage , Piperidines/pharmacokinetics , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Poly(ADP-ribose) Polymerase Inhibitors/pharmacokinetics , Therapeutic Equivalency
11.
J Clin Pharmacol ; 58(5): 686-693, 2018 05.
Article in English | MEDLINE | ID: mdl-29329482

ABSTRACT

Rolapitant is a selective and long-acting neurokinin-1 receptor antagonist approved in an oral formulation in combination with other antiemetic agents for the prevention of delayed chemotherapy-induced nausea and vomiting in adults. This was a phase 1 open-label, parallel-group pharmacokinetic and safety study of a single oral dose of 180 mg of rolapitant and its major active metabolite, M19, in subjects with mild and moderate hepatic impairment compared with healthy matched controls. Pharmacokinetics were assessed by a mixed-model analysis of variance of log-transformed values for maximum observed plasma concentration (Cmax ), observed time at Cmax (tmax ), area under the plasma concentration-time curve (AUC) from time 0 to the time of the last quantifiable concentration (AUC0-t ), and AUC from time 0 to 120 hours (AUC0-120 ), with hepatic group as a fixed effect. Mean rolapitant Cmax , AUC0-t , and AUC0-120 were similar in the mild hepatic impairment and healthy control groups. In subjects with moderate hepatic impairment, AUC0-t was similar and Cmax was 25% lower than in healthy controls. Mean M19 Cmax and AUC0-t were similar in the mild hepatic impairment group and healthy controls, but <20% lower in those with moderate hepatic impairment versus healthy controls. Fraction of unbound rolapitant was comparable in all groups for rolapitant and M19. Rolapitant was well tolerated in all groups, without serious adverse events. Pharmacokinetic differences between healthy subjects and those with mild or moderate hepatic impairment are unlikely to pose a safety risk and do not warrant predefined dosage adjustment in the presence of hepatic impairment.


Subject(s)
Liver Diseases/physiopathology , Neurokinin-1 Receptor Antagonists/administration & dosage , Spiro Compounds/administration & dosage , Administration, Oral , Area Under Curve , Case-Control Studies , Female , Humans , Male , Middle Aged , Neurokinin-1 Receptor Antagonists/adverse effects , Neurokinin-1 Receptor Antagonists/pharmacokinetics , Severity of Illness Index , Spiro Compounds/adverse effects , Spiro Compounds/pharmacokinetics
12.
J Clin Pharmacol ; 58(2): 202-211, 2018 02.
Article in English | MEDLINE | ID: mdl-28906558

ABSTRACT

Rolapitant is a selective and long-acting neurokinin-1 receptor antagonist approved in an oral formulation in combination with other antiemetic agents for the prevention of delayed chemotherapy-induced nausea and vomiting in adults. Four open-label phase 1 studies evaluated the safety and drug-drug interactions of a single dose of rolapitant given intravenously (166.5 mg) or orally (180 mg) with oral digoxin (0.5 mg) or sulfasalazine (500 mg), probe substrates for the P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP), respectively. Administration of intravenous rolapitant with the substrates did not result in clinically significant effects on digoxin and sulfasalazine pharmacokinetics. In contrast, peak concentration and area under the curve for last quantifiable plasma concentrations increased by 71% (geometric mean ratio [GMR], 1.71; 90% confidence interval [CI], 1.49-1.95) and 30% (GMR, 1.30; 90%CI, 1.19-1.42), respectively, when rolapitant was coadministered orally with digoxin compared with digoxin alone; they increased by 140% (GMR, 2.40; 90%CI, 2.02-2.86) and 127% (GMR, 2.27; 90%CI, 1.94-2.65), respectively, when rolapitant was given orally with sulfasalazine compared with sulfasalazine alone. Adverse events were mild to moderate in severity in the absence or presence of rolapitant. There were no abnormal clinical laboratory or electrocardiogram findings. Thus, whether administered orally or intravenously, rolapitant was safe and well tolerated. Patients taking oral rolapitant with P-gp and BCRP substrates with a narrow therapeutic index should be monitored for potential adverse events; although increased plasma concentrations of these substrates may raise the risk of toxicity, they are not contraindicated.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Digoxin/pharmacokinetics , Neoplasm Proteins/metabolism , Neurokinin-1 Receptor Antagonists/administration & dosage , Spiro Compounds/administration & dosage , Sulfasalazine/pharmacokinetics , Administration, Intravenous , Administration, Oral , Adult , Drug Interactions , Female , Healthy Volunteers , Humans , Male , Middle Aged , Neurokinin-1 Receptor Antagonists/adverse effects , Spiro Compounds/adverse effects
13.
J Clin Pharmacol ; 57(12): 1600-1606, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28906561

ABSTRACT

Rolapitant, a selective and long-acting neurokinin-1 receptor antagonist, is approved in an oral formulation for the prevention of delayed chemotherapy-induced nausea and vomiting in adults. The objective of this pivotal study was to assess the bioequivalence of a single intravenous infusion of rolapitant versus a single oral dose of rolapitant. In this randomized, open-label phase 1 study, healthy volunteers were administered rolapitant as a 180-mg oral dose or a 30-minute 166.5-mg intravenous infusion. Blood samples for pharmacokinetic analysis were collected predose and at points up to 912 hours postdose. Criteria for bioequivalence of the intravenous dose versus the oral dose were met if the 90% confidence intervals (CIs) for the ratios of the geometric least-squares means (GLSMs) for the area under the plasma concentration-time curve (AUC) from time 0 to the time of the last quantifiable concentration (AUC0-t ) and AUC from time 0 extrapolated to infinity (AUC0-∞ ) for rolapitant were within 0.80-1.25. Mean rolapitant systemic exposure and half-lives were similar in the oral (n = 62) and intravenous (n = 61) rolapitant groups. The 90%CIs of the ratio of GLSMs were within the 0.80-1.25 range for AUC0-t (0.94-1.09) and AUC0-∞ (0.93-1.10). The incidence of treatment-emergent adverse events, all mild or moderate in severity, was similar in the intravenous and oral groups. A 166.5-mg intravenous infusion of rolapitant met the bioequivalence criteria based on AUC to a 180-mg oral dose and was well tolerated.


Subject(s)
Neurokinin-1 Receptor Antagonists/administration & dosage , Neurokinin-1 Receptor Antagonists/pharmacokinetics , Spiro Compounds/administration & dosage , Spiro Compounds/pharmacokinetics , Administration, Intravenous , Administration, Oral , Adolescent , Adult , Area Under Curve , Female , Half-Life , Humans , Male , Middle Aged , Therapeutic Equivalency , Young Adult
14.
Lancet Oncol ; 16(9): 1079-1089, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26272769

ABSTRACT

BACKGROUND: Highly emetogenic chemotherapy induces emesis in almost all patients in the absence of prophylaxis. Guidelines recommend use of a neurokinin-1 (NK-1) receptor antagonist in conjunction with a 5-HT3 receptor antagonist and corticosteroid in patients receiving highly emetogenic chemotherapy. We aimed to assess rolapitant, an NK-1 receptor antagonist, for prevention of chemotherapy-induced nausea and vomiting in patients with cancer after administration of cisplatin-based highly emetogenic chemotherapy. METHODS: We conducted two global, randomised, double-blind, active-controlled, phase 3 trials (HEC-1 and HEC-2) at 155 cancer centres (76 in HEC-1 and 79 in HEC-2) in 26 countries (17 in HEC-1 and 14 in HEC-2). We enrolled patients with cancer aged 18 years or older, who had not previously been treated with cisplatin, with a Karnofsky performance score of 60 or higher, and a predicted life expectancy of 4 months or longer. We used an interactive web-based randomisation system to randomly assign patients to treatment. Patients were stratified by sex and randomly allocated to either oral rolapitant (180 mg dose; rolapitant group) or a placebo that was identical in appearance (active control group) about 1-2 h before administration of highly emetogenic chemotherapy. All patients received granisetron (10 µg/kg intravenously) and dexamethasone (20 mg orally) on day 1, and dexamethasone (8 mg orally) twice daily on days 2-4. Every cycle was a minimum of 14 days. In up to five subsequent cycles, patients were allowed to receive the same study drug they were assigned in cycle 1, unless removed at the clinician's discretion. Patients could also choose to leave the study at any point. Efficacy analysis was done in the modified intention-to-treat population (comprising all patients who received at least one dose of study drug at a cancer centre compliant with Good Clinical Practice [GCP]). The primary endpoint was the proportion of patients achieving a complete response (no emesis or use of rescue medication) in the delayed phase (>24-120 h after initiation of chemotherapy) in cycle 1. These studies are registered with ClinicalTrials.gov, numbers NCT01499849 and NCT01500213. Both studies have been completed. FINDINGS: Between Feb 21, 2012, and March 12, 2014, 532 patients in HEC-1 and 555 patients in HEC-2 were randomly assigned to treatment. 526 patients in HEC-1 (264 rolapitant and 262 active control) and 544 in HEC-2 (271 rolapitant and 273 active control) received at least one dose of study drug at a GCP-compliant site and were included in the modified intention-to-treat population. A significantly greater proportion of patients in the rolapitant group had complete responses in the delayed phase than did patients in the active control group (HEC-1: 192 [73%] vs 153 [58%]; odds ratio 1·9, 95% CI 1·3-2·7; p=0·0006; HEC-2: 190 [70%] vs 169 [62%]; 1·4, 1·0-2·1; p=0·0426; pooled studies: 382 [71%] vs 322 [60%]; 1·6, 1·3-2·1; p=0·0001). The incidence of adverse events was similar across treatment groups. The most commonly reported treatment-related treatment-emergent adverse events in the rolapitant versus active control groups were headache (three [<1%] vs two [<1%]), hiccups (three [<1%] vs four [<1%]), constipation (two [<1%] vs three [<1%]), and dyspepsia (two [<1%] vs three [<1%]). For cycle 1, the most common grade 3-5 adverse events in patients allocated rolapitant versus active control were neutropenia (HEC-1: nine [3%] vs 14 [5%]; HEC-2: 16 [6%] vs 14 [5%]), anaemia (HEC-1: one [<1%] vs one [<1%]; HEC-2: seven [3%] vs two [<1%]), and leucopenia (HEC-1: six [2%] vs two [<1%]; HEC-2: two [<1%] vs two [<1%]). No serious treatment-emergent adverse events were treatment related, and no treatment-related treatment-emergent adverse events resulted in death. INTERPRETATION: Rolapitant in combination with a 5-HT3 receptor antagonist and dexamethasone is well-tolerated and shows superiority over active control for the prevention of chemotherapy-induced nausea and vomiting during the at-risk period (120 h) after administration of highly emetogenic cisplatin-based chemotherapy. FUNDING: TESARO, Inc.


Subject(s)
Nausea/drug therapy , Neoplasms/drug therapy , Spiro Compounds/administration & dosage , Vomiting/drug therapy , Adult , Aged , Aged, 80 and over , Cisplatin/adverse effects , Drug-Related Side Effects and Adverse Reactions , Female , Humans , Male , Middle Aged , Nausea/chemically induced , Nausea/pathology , Neoplasms/pathology , Vomiting/chemically induced , Vomiting/pathology
15.
J Clin Pharmacol ; 55(1): 73-80, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25042797

ABSTRACT

This is the second of two manuscripts detailing the pharmacodynamic derivation of peginterferon lambda-1a (Lambda) dosing and treatment durations for Phase 3 studies in hepatitis C virus (HCV) infection, based on Phase 2 data. Herein, we describe the derivation of regression models for 12-week on-treatment virologic response and safety outcomes at 120, 180, and 240 µg Lambda with ribavirin. In patients with HCV genotypes 1 or 4, there was a significant (P = 0.024) relationship between undetectable HCV-RNA at Week 4 and Lambda exposure (AUC or Cmax ), with the largest difference between adjacent dose levels between the 180 and 120 µg exposure ranges. Risk of Grade 3-4 aminotransferase or bilirubin elevations relative to a peginterferon alfa-2a/ribavirin control were related to Lambda exposure for all patients, and the largest increase between adjacent dose levels was seen for 240 versus 180 µg. Anemia and neutropenia events were lower than control across all doses and exposures. Based on these data and those in our previous manuscript, Phase 3 studies will evaluate fixed 180 µg doses of Lambda in combination with ribavirin and a direct-acting antiviral for 24-48 weeks in HCV genotypes 1 or 4 or 12-24 weeks in genotypes 2 or 3.


Subject(s)
Antiviral Agents , Hepatitis C, Chronic/drug therapy , Interleukins , Polyethylene Glycols , Adult , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , Antiviral Agents/therapeutic use , Clinical Trials, Phase III as Topic , Dose-Response Relationship, Drug , Double-Blind Method , Drug Therapy, Combination , Female , Genotype , Hepacivirus/drug effects , Hepacivirus/genetics , Hepacivirus/physiology , Hepatitis C, Chronic/genetics , Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/virology , Humans , Interferon-alpha/therapeutic use , Interferons , Interleukins/administration & dosage , Interleukins/adverse effects , Interleukins/genetics , Interleukins/pharmacokinetics , Interleukins/therapeutic use , Logistic Models , Male , Middle Aged , Models, Biological , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/adverse effects , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/therapeutic use , RNA, Viral/blood , Recombinant Proteins/therapeutic use , Ribavirin/therapeutic use , Treatment Outcome , Viral Load/drug effects
16.
J Clin Pharmacol ; 55(1): 63-72, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25043197

ABSTRACT

Peginterferon lambda-1a (Lambda) is under clinical development for the treatment of chronic hepatitis B and C virus (HBV, HCV, respectively) infection. This is the first of two manuscripts detailing the pharmacodynamic derivation of Lambda dosing and treatment durations for Phase 3 studies in HCV, based on Phase 2 data. We describe here the derivation of a population model of Lambda exposure; the adaptation of a previously published viral dynamic model for Lambda treatment and host genotype, and its use to simulate sustained virologic responses (SVR). Lambda population pharmacokinetics was described by a one-compartment model with first-order absorption, and 33.0 L per day clearance with 47% interindividual (36% intra-individual) variability. Weight explained a negligible proportion of the variability. Based on SVR predictions, optimum treatment durations were 48 weeks for HCV genotypes 1 or 4 (SVR estimates for 120, 180, and 240 µg Lambda: 58%, 54%, 47%, respectively) and 24 weeks for genotypes 2 or 3 (75%, 72%, 67%). SVR predictions for 240 µg were lower due to dropout predictions. The SVR model established the optimum treatment duration for Phase 3 studies but did not differentiate between 120 and 180 µg dosing. A companion manuscript describes dose selection based on exposure-response/safety modeling.


Subject(s)
Antiviral Agents , Hepatitis C, Chronic/drug therapy , Interleukins , Models, Biological , Polyethylene Glycols , Adult , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Antiviral Agents/therapeutic use , Clinical Trials, Phase III as Topic , Double-Blind Method , Drug Therapy, Combination , Female , Genotype , Hepacivirus/drug effects , Hepacivirus/genetics , Hepacivirus/physiology , Hepatitis C, Chronic/genetics , Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/virology , Humans , Interferon-alpha/therapeutic use , Interferons , Interleukins/administration & dosage , Interleukins/genetics , Interleukins/pharmacokinetics , Interleukins/therapeutic use , Male , Middle Aged , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/therapeutic use , RNA, Viral/analysis , Recombinant Proteins/therapeutic use , Ribavirin/therapeutic use , Viral Load/drug effects
17.
PLoS One ; 8(5): e63818, 2013.
Article in English | MEDLINE | ID: mdl-23717490

ABSTRACT

UNLABELLED: Expression of the programmed death 1 (PD-1) receptor and its ligands are implicated in the T cell exhaustion phenotype which contributes to the persistence of several chronic viral infections, including human hepatitis C virus (HCV). The antiviral potential of BMS-936558 (MDX-1106) - a fully human anti-PD-1 monoclonal immunoglobulin-G4 that blocks ligand binding - was explored in a proof-of-concept, placebo-controlled single-ascending-dose study in patients (N = 54) with chronic HCV infection. Interferon-alfa treatment-experienced patients (n = 42) were randomized 5∶1 to receive a single infusion of BMS-936558 (0.03, 0.1, 0.3, 1.0, 3.0 mg/kg [n = 5 each] or 10 mg/kg [n = 10]) or of placebo (n = 7). An additional 12 HCV treatment-naïve patients were randomized to receive 10 mg/kg BMS-936558 (n = 10) or placebo (n = 2). Patients were followed for 85 days post-dose. Five patients who received BMS-936558 (0.1 [n = 1] or 10 mg/kg) and one placebo patient achieved the primary study endpoint of a reduction in HCV RNA ≥0.5 log10 IU/mL on at least 2 consecutive visits; 3 (10 mg/kg) achieved a >4 log10 reduction. Two patients (10 mg/kg) achieved HCV RNA below the lower limit of quantitation (25 IU/mL), one of whom (a prior null-responder) remained RNA-undetectable 1 year post-study. Transient reductions in CD4(+), CD8(+) and CD19(+) cells, including both naïve and memory CD4(+) and CD8(+) subsets, were observed at Day 2 without evidence of immune deficit. No clinically relevant changes in immunoglobulin subsets or treatment-related trends in circulating cytokines were noted. BMS-936558 exhibited dose-related exposure increases, with a half-life of 20-24 days. BMS-936558 was mostly well tolerated. One patient (10 mg/kg) experienced an asymptomatic grade 4 ALT elevation coincident with the onset of a 4-log viral load reduction. Six patients exhibited immune-related adverse events of mild-to-moderate intensity, including two cases of hyperthyroidism consistent with autoimmune thyroiditis. Further investigation of PD-1 pathway blockade in chronic viral disease is warranted. TRIAL REGISTRATION: ClinicalTrials.gov NCT00703469.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antiviral Agents/therapeutic use , Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Adult , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Antiviral Agents/adverse effects , Double-Blind Method , Female , Half-Life , Hepacivirus/genetics , Humans , Interferon-alpha/therapeutic use , Male , Middle Aged , Nivolumab , Programmed Cell Death 1 Receptor/metabolism , RNA, Viral/genetics , Viral Load/drug effects , Viral Load/genetics
18.
Mol Cancer Ther ; 11(7): 1432-42, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22474168

ABSTRACT

The use of combination drug regimens has dramatically improved the clinical outcome for patients with multiple myeloma. However, to date, combination treatments have been limited to approved drugs and a small number of emerging agents. Using a systematic approach to identify synergistic drug combinations, combination high-throughput screening (cHTS) technology, adenosine A2A and ß-2 adrenergic receptor (ß2AR) agonists were shown to be highly synergistic, selective, and novel agents that enhance glucocorticoid activity in B-cell malignancies. Unexpectedly, A2A and ß2AR agonists also synergize with melphalan, lenalidomide, bortezomib, and doxorubicin. An analysis of agonists, in combination with dexamethasone or melphalan in 83 cell lines, reveals substantial activity in multiple myeloma and diffuse large B-cell lymphoma cell lines. Combination effects are also observed with dexamethasone as well as bortezomib, using multiple myeloma patient samples and mouse multiple myeloma xenograft assays. Our results provide compelling evidence in support of development of A2A and ß2AR agonists for use in multi-drug combination therapy for multiple myeloma. Furthermore, use of cHTS for the discovery and evaluation of new targets and combination therapies has the potential to improve cancer treatment paradigms and patient outcomes.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Adrenergic beta-2 Receptor Antagonists/pharmacology , Antineoplastic Agents/pharmacology , Multiple Myeloma/metabolism , Animals , Cell Line, Tumor , Cluster Analysis , Drug Screening Assays, Antitumor , Drug Synergism , Female , Gene Expression Profiling , High-Throughput Screening Assays , Humans , Male , Mice , Mice, SCID , Multiple Myeloma/genetics , Transcriptome , Xenograft Model Antitumor Assays
19.
J Pharm Biomed Anal ; 50(2): 200-9, 2009 Sep 08.
Article in English | MEDLINE | ID: mdl-19467820

ABSTRACT

CRx-102 is an oral synergistic combination drug which contains the cardiovascular agent, dipyridamole (DP) and a very low dose of the glucocorticoid, prednisolone (PRED). CRx-102 works through a novel mechanism of action in which DP selectively amplifies the anti-inflammatory activity of PRED without replicating its side effects. CRx-102 is in clinical trials for the treatment of osteoarthritis. Here we delineate the in vitro metabolism and explore the potential for a drug-drug interaction between the active agents in CRx-102. Our study using human hepatocyte suspensions showed that both DP and PRED were metabolized by CYP3A4 isozymes, resulting in the formation of diverse arrays of both oxidative and oxidative-reduced metabolites. Within phase 1 biotransformation, CYP3A4 was one of the pathways responsible for the metabolism of PRED, while phase 2 biotransformation played a significant role in the metabolism of DP. Glucuronidation of DP was substantial and was catalyzed by many UGT members, specifically those in the UGT1A subfamily. Based on the tandem mass (MS/MS) product ion spectra (PIS) acquired, the major metabolites of both agents, namely, monooxygenated, mono-N-deethanolaminated, dehydrogenated and O-glucuronidated metabolites of DP and the monooxygenated (e.g., 6-hydroxyl), dehydrogenated (prednisone) and reduced (20-hydroxyl) metabolites of PRED, were identified and elucidated. The affinities for DP biotransformation, including CYP3A4-mediated oxidative pathways and UGT-mediated O-glucuronidation, appeared high (K(m)<10 microM), as compared with the modest affinities of PRED biotransformation catalyzed by CYP3A4 (K(m) approximately 40-170 microM). DP, but not PRED, exerted a minimal inhibitory effect on the drug-metabolizing CYP isoforms, including CYP3A4, which was determined using a panel of CYP isoform-preferred substrate activities in pooled human liver microsomal (HLM) preparations and microsomal preparations containing the recombinant enzymes (K(i) approximately 2-12 microM). Using the DP maximal plasma concentration (C(max)) observed in the clinic and a predictive mathematical model for metabolism-associated drug-drug interaction (DDI), we have demonstrated that there is little likelihood of a pharmacokinetic interaction between the two active agents in CRx-102.


Subject(s)
Dipyridamole/pharmacology , Drug Interactions , Prednisolone/pharmacology , Biotransformation , Cells, Cultured , Chromatography, Liquid/methods , Cytochrome P-450 CYP3A , Cytochrome P-450 CYP3A Inhibitors , Dipyridamole/pharmacokinetics , Drug Combinations , Humans , In Vitro Techniques , Mass Spectrometry/methods , Prednisolone/pharmacokinetics , Spectrophotometry, Ultraviolet/methods
20.
J Pharm Biomed Anal ; 49(5): 1241-9, 2009 Jul 12.
Article in English | MEDLINE | ID: mdl-19304431

ABSTRACT

We have developed and validated an accurate, sensitive, and robust LC-MS/MS method that determines the concentration of CRx-102 (the combination of prednisolone and dipyridamole) in human plasma. In this method, prednisolone, dipyridamole, and the combined internal standards (IS) prednisolone-d(6) (IS for prednisolone) and dipyridamole-d(20) (IS for dipyridamole) were extracted from 100 microL human EDTA plasma using methylbutyl ether. Calibration curves were linear over a concentration range of 0.4-200 ng/mL for prednisolone and 5-3000 ng/mL for dipyridamole. The analytes were quantitatively determined using tandem mass spectrometry operated in positive electrospray ionization in a multiple reaction monitoring (MRM) mode. This validated method has been used successfully in clinical pharmacokinetic studies of CRx-102 in healthy volunteers.


Subject(s)
Chromatography, Liquid/methods , Dipyridamole/blood , Prednisolone/blood , Tandem Mass Spectrometry/methods , Area Under Curve , Calibration , Dipyridamole/chemistry , Dipyridamole/pharmacokinetics , Drug Combinations , Drug Stability , Freezing , Half-Life , Humans , Metabolic Clearance Rate , Molecular Structure , Prednisolone/chemistry , Prednisolone/pharmacokinetics , Quality Control , Reference Standards , Reproducibility of Results , Sensitivity and Specificity , Spectrometry, Mass, Electrospray Ionization/methods , Temperature , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...