Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Article in English | MEDLINE | ID: mdl-30745392

ABSTRACT

The combination of the hepatitis C virus (HCV) nonstructural protein 5A (NS5A) inhibitor elbasvir and the NS3/4A protease inhibitor grazoprevir is a potent, once-daily therapy indicated for the treatment of chronic HCV infection in individuals coinfected with human immunodeficiency virus (HIV). We explored the pharmacokinetic interactions of elbasvir and grazoprevir with ritonavir and ritonavir-boosted HIV protease inhibitors in three phase 1 trials. Drug-drug interaction trials with healthy participants were conducted to evaluate the effect of ritonavir on the pharmacokinetics of grazoprevir (n = 10) and the potential two-way pharmacokinetic interactions of elbasvir (n = 30) or grazoprevir (n = 39) when coadministered with ritonavir-boosted atazanavir, lopinavir, or darunavir. Coadministration of ritonavir with grazoprevir increased grazoprevir exposure; the geometric mean ratio (GMR) for grazoprevir plus ritonavir versus grazoprevir alone area under the concentration-time curve from 0 to 24 h (AUC0-24) was 1.91 (90% confidence interval [CI]; 1.31 to 2.79). Grazoprevir exposure was markedly increased with coadministration of atazanavir-ritonavir, lopinavir-ritonavir, and darunavir-ritonavir, with GMRs for grazoprevir AUC0-24 of 10.58 (90% CI, 7.78 to 14.39), 12.86 (90% CI, 10.25 to 16.13), and 7.50 (90% CI, 5.92 to 9.51), respectively. Elbasvir exposure was increased with coadministration of atazanavir-ritonavir, lopinavir-ritonavir, and darunavir-ritonavir, with GMRs for elbasvir AUC0-24 of 4.76 (90% CI, 4.07 to 5.56), 3.71 (90% CI, 3.05 to 4.53), and 1.66 (90% CI, 1.35 to 2.05), respectively. Grazoprevir and elbasvir had little effect on atazanavir, lopinavir, and darunavir pharmacokinetics. Coadministration of elbasvir-grazoprevir with atazanavir-ritonavir, lopinavir-ritonavir, or darunavir-ritonavir is contraindicated, owing to an increase in grazoprevir exposure. Therefore, HIV treatment regimens without HIV protease inhibitors should be considered for HCV/HIV-coinfected individuals who are being treated with elbasvir-grazoprevir.


Subject(s)
Antiviral Agents/pharmacokinetics , HIV Infections/drug therapy , HIV Protease Inhibitors/pharmacokinetics , Hepatitis C/drug therapy , Adult , Amides , Antiviral Agents/pharmacology , Atazanavir Sulfate/pharmacokinetics , Atazanavir Sulfate/pharmacology , Benzofurans/pharmacokinetics , Benzofurans/pharmacology , Carbamates , Cyclopropanes , Darunavir/pharmacokinetics , Darunavir/pharmacology , Drug Interactions , Female , HIV Protease Inhibitors/pharmacology , HIV-1/drug effects , Healthy Volunteers , Hepacivirus/drug effects , Humans , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Lopinavir/pharmacokinetics , Lopinavir/pharmacology , Male , Middle Aged , Quinoxalines/pharmacokinetics , Quinoxalines/pharmacology , Ritonavir/pharmacokinetics , Ritonavir/pharmacology , Sulfonamides , Viral Nonstructural Proteins/antagonists & inhibitors , Young Adult
2.
Ophthalmology ; 126(8): 1141-1154, 2019 08.
Article in English | MEDLINE | ID: mdl-30946888

ABSTRACT

PURPOSE: To evaluate the safety and efficacy of the Port Delivery System with ranibizumab (PDS) for neovascular age-related macular degeneration (nAMD) treatment. DESIGN: Phase 2, multicenter, randomized, active treatment-controlled clinical trial. PARTICIPANTS: Patients diagnosed with nAMD within 9 months who had received 2 or more prior anti-vascular endothelial growth factor intravitreal injections and were responsive to treatment. METHODS: Patients were randomized 3:3:3:2 to receive the PDS filled with ranibizumab 10 mg/ml, 40 mg/ml, 100 mg/ml, or monthly intravitreal ranibizumab 0.5-mg injections. MAIN OUTCOME MEASURES: Time to first implant refill assessed when the last enrolled patient completed the month 9 visit (primary efficacy end point), improvement in best-corrected visual acuity (BCVA) and central foveal thickness (CFT), and safety. RESULTS: The primary analysis population was 220 patients, with 58, 62, 59, and 41 patients in the PDS 10-mg/ml, PDS 40-mg/ml, PDS 100-mg/ml, and monthly intravitreal ranibizumab 0.5-mg arms, respectively. Median time to first implant refill was 8.7, 13.0, and 15.0 months in the PDS 10-mg/ml, PDS 40-mg/ml, and PDS 100-mg/ml arms, respectively. At month 9, the adjusted mean BCVA change from baseline was ‒3.2 Early Treatment Diabetic Retinopathy Study (ETDRS) letters, ‒0.5 ETDRS letters, +5.0 ETDRS letters, and +3.9 ETDRS letters in the PDS 10-mg/ml, PDS 40-mg/ml, PDS 100-mg/ml, and monthly intravitreal ranibizumab 0.5-mg arms, respectively. At month 9, the adjusted mean CFT change from baseline was similar in the PDS 100-mg/ml and monthly intravitreal ranibizumab 0.5-mg arms. The optimized PDS implant insertion and refill procedures were generally well tolerated. After surgical procedure optimization, postoperative vitreous hemorrhage rate was 4.5% (7/157; 1 event classified as serious). There was no evidence of implant clogging. CONCLUSIONS: In the phase 2 Ladder trial, the PDS was generally well tolerated and demonstrated a dose response across multiple end points in patients with nAMD. The PDS 100-mg/ml arm showed visual and anatomic outcomes comparable with monthly intravitreal ranibizumab 0.5-mg injections but with a reduced total number of ranibizumab treatments. The PDS has the potential to reduce treatment burden in nAMD while maintaining vision.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Delayed-Action Preparations/administration & dosage , Drug Implants , Macular Degeneration/drug therapy , Ranibizumab/administration & dosage , Aged , Aged, 80 and over , Female , Humans , Intravitreal Injections , Male , Middle Aged
3.
Article in English | MEDLINE | ID: mdl-29133549

ABSTRACT

RG7667, a novel combination of two anticytomegalovirus (anti-CMV) monoclonal IgG1 antibodies (MCMV5322A and MCMV3068A), was designed to block CMV entry into host cells. It was developed as a potential therapy for preventing CMV infection and disease in transplant recipients. RG7667 was assessed for preventing CMV infection in a phase 2a trial with CMV-seronegative recipients of kidney transplants from CMV-seropositive donors. The patients received 4 intravenous doses of RG7667 (10 mg/kg of body weight of each antibody, n = 60) or placebo (n = 60) at the time of the transplant and at 1, 4, and 8 weeks after the transplant. Serum samples were collected for pharmacokinetic (PK) analysis and antidrug antibody (ADA) evaluation. To guide future dose selection, the relationships between RG7667 exposure and pharmacological activity were evaluated. MCMV5322A and MCMV3068A exposures were confirmed in all RG7667-treated patients. Mean clearances for MCMV5322A and MCMV3068A were 2.97 and 2.65 ml/day/kg, respectively, and the terminal half-lives of MCMV5322A and MCMV3068A were 26.9 and 27.4 days, respectively. The ADA incidence was low and was not associated with lower drug exposure. Patients with RG7667 or component antibody exposures greater than the respective median values had a lower incidence of viremia at 12 weeks and 24 weeks after transplantation and a longer delayed time to detectable CMV viremia than patients with exposures less than the median values. MCMV5322A and MCMV3068A exhibited expected IgG1 PK profiles in high-risk kidney transplant recipients, consistent with the earlier PK behavior of RG7667 in healthy subjects. Higher drug exposure was associated with better anti-CMV pharmacological activity. (This study has been registered at ClinicalTrials.gov under identifier NCT01753167.).


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antibodies, Viral/pharmacology , Antiviral Agents/pharmacokinetics , Cytomegalovirus Infections/prevention & control , Kidney Transplantation , Viremia/prevention & control , Adult , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/pharmacology , Antibodies, Viral/blood , Antiviral Agents/blood , Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , Cytomegalovirus/physiology , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Half-Life , Humans , Injections, Intravenous , Male , Middle Aged , Transplant Recipients
4.
Int J Clin Pharmacol Ther ; 55(9): 746-752, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28679468

ABSTRACT

AIMS: Sugammadex rapidly reverses moderate and deep rocuronium- or vecuronium-induced neuromuscular blockade at doses of 4 mg/kg and 2 mg/kg, respectively. Sugammadex is renally eliminated. This study evaluated the pharmacokinetics of sugammadex in subjects with renal impairment versus those with normal renal function. METHODS: This open-label, two-part, phase 1 study included adults with moderate (creatinine clearance (CLcr) 30 - < 50 mL/min) and severe (CLcr < 30 mL/min) renal impairment and healthy controls (CLcr ≥ 80 mL/min). A single intravenous (IV) bolus injection of sugammadex 4 mg/kg was administered into a peripheral vein over 10 seconds directly by straight needle in part 1 (n = 24; 8/group), and via an IV catheter followed by a saline flush in part 2 (n = 18; 6/group). Plasma concentrations of sugammadex were collected after drug administration. Due to dosing issues in part 1, pharmacokinetic parameters were determined for part 2 only. Safety was assessed throughout the study. RESULTS: Pharmacokinetic data were obtained from 18 subjects. Mean sugammadex exposure (AUC0-∞) in subjects with moderate and severe renal impairment was 2.42- and 5.42-times, respectively, that of healthy controls. Clearance decreased and apparent terminal half-life was prolonged with increasing renal dysfunction. Similar Cmax values were observed in subjects with renal impairment and healthy controls. There were no serious adverse events. CONCLUSIONS: Sugammadex exposure is increased in subjects with moderate and severe renal insufficiency due to progressively decreased clearance as a function of worsening renal function. Sugammadex 4 mg/kg was well tolerated in subjects with renal impairment, with a safety profile similar to that of healthy subjects. These results indicate that dose adjustment of sugammadex is not required in patients with moderate renal impairment; however, current safety experience is insufficient to support the use of sugammadex in patients with CLcr < 30 mL/min.
.


Subject(s)
Kidney/metabolism , Renal Insufficiency/metabolism , gamma-Cyclodextrins/pharmacokinetics , Aged , Case-Control Studies , Female , Half-Life , Humans , Male , Middle Aged , Neuromuscular Blockade/methods , Sugammadex
5.
Retina ; 37(10): 1847-1858, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28106709

ABSTRACT

PURPOSE: To evaluate the systemic pharmacokinetics (PKs) of aflibercept, bevacizumab, and ranibizumab in patients with neovascular age-related macular degeneration (AMD), diabetic macular edema (DME), or retinal vein occlusion (RVO). METHODS: Prospective, open-label, nonrandomized clinical trial of patients with AMD, DME, or RVO who were antivascular endothelial growth factor (VEGF) naïve or had not received anti-VEGF for ≥4 months. Patients received 3 monthly intravitreal injections of aflibercept 2.0 mg, bevacizumab 1.25 mg, or ranibizumab (0.5 mg for AMD/RVO, 0.3 mg for DME). The main outcome measures were serum PKs and plasma free-VEGF concentrations after the first and third injections. RESULTS: A total of 151 patients were included. In AMD/DME/RVO, systemic exposure to each drug was highest with bevacizumab, then aflibercept, and lowest with ranibizumab. Ranibizumab cleared from the bloodstream more quickly than bevacizumab or aflibercept. Aflibercept treatment resulted in the greatest reductions in plasma free-VEGF relative to baseline levels, whereas ranibizumab treatment resulted in the smallest decreases in plasma free-VEGF. CONCLUSION: The three anti-VEGF treatments examined in this analysis demonstrated notable differences in systemic PKs. Generally, the reduction in plasma free-VEGF levels correlated with elevated levels of circulating anti-VEGF agents, with the reduction in free-VEGF levels greatest with aflibercept and least with ranibizumab.


Subject(s)
Bevacizumab/pharmacokinetics , Diabetic Retinopathy/drug therapy , Macular Edema/drug therapy , Ranibizumab/pharmacokinetics , Recombinant Fusion Proteins/pharmacokinetics , Wet Macular Degeneration/drug therapy , Aged , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacokinetics , Bevacizumab/administration & dosage , Diabetic Retinopathy/blood , Diabetic Retinopathy/complications , Enzyme-Linked Immunosorbent Assay , Female , Follow-Up Studies , Humans , Intravitreal Injections , Macular Edema/blood , Macular Edema/etiology , Male , Middle Aged , Prospective Studies , Ranibizumab/administration & dosage , Receptors, Vascular Endothelial Growth Factor/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/blood , Wet Macular Degeneration/blood , Wet Macular Degeneration/diagnosis
6.
J Pharmacol Exp Ther ; 355(2): 288-96, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26359312

ABSTRACT

Lampalizumab is an antigen-binding fragment of a humanized monoclonal antibody against complement factor D (CFD), a rate-limiting enzyme in the activation and amplification of the alternative complement pathway (ACP), which is in phase III clinical trials for the treatment of geographic atrophy. Understanding of the pharmacokinetics, pharmacodynamics, and biodistribution of lampalizumab following intravitreal administration in the ocular compartments and systemic circulation is limited but crucial for selecting doses that provide optimal efficacy and safety. Here, we sought to construct a semimechanistic and integrated ocular-systemic pharmacokinetic-pharmacodynamic model of lampalizumab in the cynomolgus monkey to provide a quantitative understanding of the ocular and systemic disposition of lampalizumab and CFD inhibition. The model takes into account target-mediated drug disposition, target turnover, and drug distribution across ocular tissues and systemic circulation. Following intravitreal administration, lampalizumab achieves rapid equilibration across ocular tissues. Lampalizumab ocular elimination is relatively slow, with a τ1/2 of approximately 3 days, whereas systemic elimination is rapid, with a τ1/2 of 0.8 hours. Target-independent linear clearance is predominant in the eye, whereas target-mediated clearance is predominant in the systemic circulation. Systemic CFD synthesis was estimated to be high (7.8 mg/day); however, the amount of CFD entering the eye due to influx from the systemic circulation was small (<10%) compared with the lampalizumab dose and is thus expected to have an insignificant impact on the clinical dose-regimen decision. Our findings support the clinical use of intravitreal lampalizumab to achieve significant ocular ACP inhibition while maintaining low systemic exposure and minimal systemic ACP inhibition.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Complement Factor D/antagonists & inhibitors , Geographic Atrophy/metabolism , Immunoglobulin Fab Fragments/pharmacology , Administration, Intravenous , Animals , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Aqueous Humor/metabolism , Female , Geographic Atrophy/drug therapy , Immunoglobulin Fab Fragments/therapeutic use , Intravitreal Injections , Macaca fascicularis , Male , Models, Biological , Retina/metabolism , Vitreous Body/metabolism
7.
Clin Pharmacokinet ; 63(4): 423-438, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38609704

ABSTRACT

Enfortumab vedotin is an antibody-drug conjugate comprised of a human monoclonal antibody directed to Nectin-4 and monomethyl auristatin E (MMAE), a microtubule-disrupting agent. The objectives of this review are to summarize the clinical pharmacology of enfortumab vedotin monotherapy and demonstrate that the appropriate dose has been selected for clinical use. Pharmacokinetics (PK) of enfortumab vedotin (antibody-drug conjugate and total antibody) and free MMAE were evaluated in five clinical trials of patients with locally advanced or metastatic urothelial carcinoma (n = 748). Intravenous enfortumab vedotin 0.5-1.25 mg/kg on days 1, 8, and 15 of a 28-day cycle showed linear, dose-proportional PK. No significant differences in exposure or safety of enfortumab vedotin and free MMAE were observed in mild, moderate, or severe renal impairment versus normal renal function. Patients with mildly impaired versus normal hepatic function had a 37% increase in area under the concentration-time curve (0-28 days), a 31% increase in maximum concentration of free MMAE, and a similar adverse event profile. No clinically significant PK differences were observed based on race/ethnicity with weight-based dosing, and no clinically meaningful QT prolongation was observed. Concomitant use with dual P-glycoprotein and strong cytochrome P450 3A4 inhibitors may increase MMAE exposure and the risk of adverse events. Approximately 3% of patients developed antitherapeutic antibodies against enfortumab vedotin 1.25 mg/kg. These findings support enfortumab vedotin 1.25 mg/kg monotherapy on days 1, 8, and 15 of a 28-day cycle. No dose adjustments are required for patients with renal impairment or mild hepatic impairment, or by race/ethnicity.


Subject(s)
Antibodies, Monoclonal , Immunoconjugates , Nectins , Humans , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Immunoconjugates/pharmacokinetics , Immunoconjugates/administration & dosage , Immunoconjugates/pharmacology , Immunoconjugates/adverse effects , Immunoconjugates/therapeutic use , Oligopeptides/pharmacokinetics , Oligopeptides/administration & dosage , Oligopeptides/therapeutic use , Oligopeptides/pharmacology , Oligopeptides/adverse effects , Urologic Neoplasms/drug therapy , Urologic Neoplasms/pathology , Dose-Response Relationship, Drug , Carcinoma, Transitional Cell/drug therapy , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology
8.
CPT Pharmacometrics Syst Pharmacol ; 12(9): 1262-1273, 2023 09.
Article in English | MEDLINE | ID: mdl-37496366

ABSTRACT

The antibody-drug conjugate (ADC) tisotumab vedotin (TV) received accelerated approval from the US Food and Drug Administration for treatment of adults with recurrent or metastatic cervical cancer (r/mCC) with disease progression on or after chemotherapy. A population pharmacokinetic (PK) model, developed using dosing data from four clinical TV studies, was used to estimate individual exposure and explore safety and efficacy exposure-response (ER) relationships. Because PK analysis showed no appreciable accumulation of TV and monomethyl auristatin E (MMAE) with repeated dosing, cycle 1 exposure metrics and predicted average concentrations from time zero until end of the cycle in which an event occurred (CavgLast ) were used for ER analyses. The probability of achieving objective response increased significantly as the ADC cycle 1 maximum serum concentration (Cmax ) increased. The probability of treatment-related adverse events (AEs) leading to dose modification increased significantly as ADC cycle 1 area under the concentration-time curve (AUC) increased. Number of grade 2+ ocular AEs increased significantly as ADC cycle 1 AUC, Cmax , and ADC CavgLast increased. MMAE cycle 1 AUC predicted risk of serious treatment-related AEs. The relationship between ADC exposure and efficacy end points suggests ADC treatment was associated with clinically meaningful response across the observed exposures; greater exposure was associated with increased efficacy. The relationship between ADC and MMAE exposure and safety end points suggests increased exposure was associated with increased AE risk. These results align with clinical findings showing TV 2 mg/kg (≤200 mg for patients ≥100 kg) every 3 weeks is efficacious and tolerable for patients with r/mCC.


Subject(s)
Immunoconjugates , Neoplasm Recurrence, Local , Adult , Humans , Antibodies, Monoclonal, Humanized/adverse effects , Immunoconjugates/adverse effects
9.
CPT Pharmacometrics Syst Pharmacol ; 11(10): 1358-1370, 2022 10.
Article in English | MEDLINE | ID: mdl-35932175

ABSTRACT

Tisotumab vedotin is an investigational antibody-drug conjugate (ADC) for treatment of solid tumors expressing tissue factor with accelerated approval from the US Food and Drug Administration for treatment of recurrent or metastatic cervical cancer with disease progression during or after chemotherapy. This study describes development of a population pharmacokinetic (PK) model to assess the PK profile of tisotumab vedotin and microtubule-disrupting agent monomethyl auristatin E (MMAE) using data from 399 patients with solid tumors across four phase I/II trials. The ADC-MMAE model describes ADC and MMAE concentrations following intravenous administration of tisotumab vedotin. This four-compartment model comprises a two-compartment ADC model with parallel linear and Michaelis-Menten elimination, a delay compartment, and a one-compartment MMAE model. Nonspecific linear clearance of ADC was 1.42 L/day, central volume of distribution (Vc ) was 3.10 L, and median terminal half-life of ADC was 4.04 days. Apparent clearance of MMAE was 42.8 L/day, and apparent volume of distribution was 2.09 L. Terminal slope of the MMAE concentration-time curve was defined by the delay compartment rate with a half-life of 2.56 days. Patients with higher body weight and lower albumin concentration had faster ADC clearance. Male patients and those with higher body weight and lower albumin concentration had higher Vc . Body weight was the most influential covariate influencing distribution and elimination of ADC and MMAE, thus supporting weight-based dosing of tisotumab vedotin. Presence of antidrug antibodies (detected in 3.3% of patients) did not affect key PK parameters or exposures for ADC and MMAE.


Subject(s)
Immunoconjugates , Neoplasms , Albumins , Antibodies, Monoclonal, Humanized/pharmacokinetics , Body Weight , Clinical Trials as Topic , Female , Humans , Male , Neoplasms/drug therapy , Oligopeptides/pharmacokinetics , Thromboplastin/therapeutic use
10.
Br J Clin Pharmacol ; 71(4): 616-20, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21395656

ABSTRACT

AIMS: Oral contraceptives such as norgestimate-ethinyl estradiol (Ortho Tri-Cyclen®) are commonly prescribed in the HIV-infected patient population. A placebo-controlled, randomized, two-period crossover study in healthy HIV-seronegative subjects was conducted to assess the effect of raltegravir on the pharmacokinetics of the estrogen and progestin components of norgestimate-ethinyl estradiol [ethinyl estradiol (EE) and norelgestromin (NGMN), an active metabolite of norgestimate (NGT)]. METHODS: In each of two periods, nineteen healthy women established on norgestimate-ethinyl estradiol contraception (21 days of active contraception; 7 days of placebo) received either 400 mg raltegravir or matching placebo twice daily on days 1-21. Pharmacokinetics were analysed on day 21 of each period. RESULTS: The geometric mean ratio (GMR) and 90% confidence interval (CI) for the EE component of norgestimate-ethinyl estradiol when co-administrated with raltegravir relative to EE alone was 0.98 (0.93-1.04) for the area under the concentration-time curve from 0 to 24 h (AUC(0-24 h) ) and 1.06 (0.98-1.14) for the maximum concentration of drug in the plasma (C(max) ); the GMR (90% CI) for the NGMN component of norgestimate-ethinyl estradiol when co-administered with raltegravir relative to NGMN alone was 1.14 (1.08-1.21) for AUC(0-24 h) and 1.29 (1.23-1.37) for C(max) . There were no discontinuations due to a study drug-related adverse experience, nor any serious clinical or laboratory adverse experience. CONCLUSIONS: Raltegravir has no clinically important effect on EE or NGMN pharmacokinetics. Co-administration of raltegravir and an oral contraceptive containing EE and NGT was generally well tolerated; no dose adjustment is required for oral contraceptives containing EE and NGT when co-administered with raltegravir.


Subject(s)
Contraceptives, Oral, Synthetic/pharmacokinetics , Estradiol/pharmacokinetics , Estrogens/pharmacokinetics , Norgestrel/analogs & derivatives , Peptide Fragments/antagonists & inhibitors , Pyrrolidinones/pharmacokinetics , Adolescent , Adult , Drug Interactions , Estradiol/blood , Female , Humans , Middle Aged , Norgestrel/blood , Norgestrel/pharmacokinetics , Peptide Fragments/pharmacokinetics , Raltegravir Potassium , Statistics as Topic , Young Adult
11.
J Clin Pharmacol ; 60(11): 1509-1518, 2020 11.
Article in English | MEDLINE | ID: mdl-32621543

ABSTRACT

MHAA4549A is a human anti-influenza A monoclonal antibody developed to treat influenza A. We report MHAA4549A serum, nasopharyngeal, and tracheal aspirate pharmacokinetics from a phase 2b study in hospitalized patients with severe influenza A. Patients were randomized 1:1:1 into 3 groups receiving single intravenous doses of 3600 mg (n = 55) or 8400 mg (n = 47) MHAA4549A or placebo (n = 56). Patients also received oral oseltamivir twice daily for ≥5 days. Serum, nasopharyngeal, and tracheal aspirate pharmacokinetic samples were collected on days 1-60 from MHAA4549A-treated groups. Day 5 plasma samples from all groups were collected for assessing the pharmacokinetics of oseltamivir and its active metabolite, oseltamivir carboxylate. Noncompartmental pharmacokinetic analysis was performed using Phoenix WinNonlin. Data were collected during a preplanned interim analysis that became final when the trial terminated because of a lack of efficacy. Serum MHAA4549A concentrations were dose-proportional and biphasic. Mean MHAA4549A clearance was 288-350 mL/day, and mean half-life was 17.8-19.0 days. Nasopharyngeal MHAA4549A concentrations were non-dose-proportional. We detected MHAA4549A in tracheal aspirate samples, but intersubject variability was high. MHAA4549A serum and nasopharyngeal exposures were confirmed in all MHAA4549A-treated patients. Serum MHAA4549A had faster clearance and a shorter half-life in influenza A-infected patients compared with healthy subjects. MHAA4549A detection in tracheal aspirate samples indicated exposure in the lower respiratory tract. Oseltamivir and oseltamivir carboxylate exposures were similar between MHAA4549A-treated and placebo groups, suggesting a lack of MHAA4549A interference with oseltamivir pharmacokinetics.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Influenza, Human/drug therapy , Oseltamivir/administration & dosage , Oseltamivir/pharmacokinetics , Administration, Oral , Aged , Antibodies, Monoclonal, Humanized/blood , Antibodies, Neutralizing/blood , Antiviral Agents/blood , Drug Therapy, Combination/methods , Half-Life , Humans , Influenza A virus/drug effects , Infusions, Intravenous , Inpatients , Middle Aged , Nasopharynx/metabolism , Oseltamivir/analogs & derivatives , Oseltamivir/blood , Trachea/metabolism
12.
Clin Infect Dis ; 48(4): 489-92, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19143531

ABSTRACT

Raltegravir, a human immunodeficiency virus type 1 (HIV-1) integrase inhibitor, has pH-dependent solubility. Raltegravir plasma concentration increases with omeprazole coadministration in healthy subjects; this is likely secondary to an increase in bioavailability attributable to increased gastric pH. Increased gastric pH has been reported in HIV-1-infected individuals, and the effects of omeprazole in this intended population may be diminished. Further investigation is necessary.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Drug Interactions , Omeprazole/pharmacokinetics , Plasma/chemistry , Pyrrolidinones/pharmacokinetics , Adolescent , Adult , Anti-HIV Agents/therapeutic use , Female , HIV Infections/drug therapy , Humans , Male , Middle Aged , Omeprazole/therapeutic use , Pyrrolidinones/therapeutic use , Raltegravir Potassium , Young Adult
13.
Antimicrob Agents Chemother ; 53(7): 2752-5, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19398643

ABSTRACT

Raltegravir (RAL) is a novel and potent human immunodeficiency virus type 1 integrase inhibitor that is predominantly metabolized via glucuronidation. The protease inhibitor combination tipranavir (TPV) at 500 mg and ritonavir (RTV) at 200 mg (TPV-RTV) has inhibitory and inductive effects on metabolic enzymes, which includes the potential to induce glucuronosyltransferase. Because RAL may be coadministered with TPV-RTV, there is the potential for the induction of RAL metabolism. Consequently, we assessed the effect of TPV-RTV on the pharmacokinetics of RAL and the safety and tolerability of this combination. Eighteen healthy adults were enrolled in this open-label study. The participants received RAL at 400 mg twice daily for 4 days (period 1) and TPV-RTV twice daily for 7 days (period 2), followed immediately by 400 mg RAL with TPV-RTV twice daily for 4 days (period 3). Under steady-state conditions, the RAL concentration at 12 h (C(12)) was decreased when RAL was administered with TPV-RTV (geometric mean ratio [GMR], 0.45; 90% confidence interval [CI] 0.31, 0.66; P = 0.0021); however, the area under the concentration-time curve from time zero to 12 h (GMR, 0.76; 90% CI, 0.49, 1.19; P = 0.2997) and the maximum concentration in serum (GMR, 0.82; 90% CI, 0.46, 1.46; P = 0.5506) were not substantially affected. There were no serious adverse experiences or discontinuations due to study drug-related adverse experiences, and RAL coadministered with TPV-RTV was generally well tolerated. Although the RAL C(12) was decreased with TPV-RTV in this study, favorable efficacy data collected in phase III studies substantiate that TPV-RTV may be coadministered with RAL without dose adjustment.


Subject(s)
HIV Protease Inhibitors/pharmacology , Pyridines/pharmacology , Pyrones/pharmacology , Pyrrolidinones/pharmacokinetics , Ritonavir/pharmacology , Adolescent , Adult , Female , Humans , Male , Middle Aged , Pyrrolidinones/blood , Raltegravir Potassium , Sulfonamides , Young Adult
14.
Antimicrob Agents Chemother ; 53(7): 2852-6, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19433563

ABSTRACT

Raltegravir is a human immunodeficiency virus type 1 integrase strand transfer inhibitor that is metabolized by glucuronidation via UGT1A1 and may be affected by inducers of UGT1A1, such as rifampin (rifampicin). Two pharmacokinetic studies were performed in healthy subjects: study 1 examined the effect of administration of 600-mg rifampin once daily on the pharmacokinetics of a single dose of 400-mg raltegravir, and study 2 examined the effect of 600-mg rifampin once daily on the pharmacokinetics of 800-mg raltegravir twice daily compared to 400-mg raltegravir twice daily without rifampin. Raltegravir coadministered with rifampin resulted in lower plasma raltegravir concentrations: in study 1, the geometric mean ratios (GMRs) and 90% confidence intervals (90% CIs) for the plasma raltegravir concentration determined 12 h postdose (C(12)), area under the concentration-time curve from 0 h to infinity (AUC(0-infinity)), and maximum concentration of drug in plasma (C(max)) (400-mg raltegravir plus rifampin/400-mg raltegravir) were 0.39 (0.30, 0.51), 0.60 (0.39, 0.91), and 0.62 (0.37, 1.04), respectively. In study 2, the GMRs and 90% CIs for raltegravir C(12), AUC(0-12), and C(max) (800-mg raltegravir plus rifampin/400-mg raltegravir) were 0.47 (0.36, 0.61), 1.27 (0.94, 1.71), and 1.62 (1.12, 2.33), respectively. Doubling the raltegravir dose to 800 mg when coadministered with rifampin therefore compensates for the effect of rifampin on raltegravir exposure (AUC(0-12)) but does not overcome the effect of rifampin on raltegravir trough concentrations (C(12)). Coadministration of rifampin and raltegravir is not contraindicated; however, caution should be used, since raltegravir trough concentrations in the presence of rifampin are likely to be at the lower limit of clinical experience.


Subject(s)
Antibiotics, Antitubercular/pharmacology , HIV Integrase Inhibitors/pharmacokinetics , Pyrrolidinones/pharmacokinetics , Rifampin/pharmacology , Adolescent , Adult , Drug Administration Schedule , Drug Interactions , Female , Humans , Male , Middle Aged , Raltegravir Potassium , Young Adult
15.
Antimicrob Agents Chemother ; 53(5): 1747-52, 2009 May.
Article in English | MEDLINE | ID: mdl-19223645

ABSTRACT

Raltegravir is a human immunodeficiency virus type 1 integrase strand transfer inhibitor with potent activity in vitro and in vivo. Raltegravir is primarily cleared by hepatic metabolism via glucuronidation (via UDP glucuronosyltransferase 1A1), with a minor component of elimination occurring via the renal pathway. Since the potential exists for raltegravir to be administered to patients with hepatic or renal insufficiency, two studies were conducted to evaluate the influence of moderate hepatic insufficiency (assessed by using the Child-Pugh criteria) and severe renal insufficiency (creatinine clearance, <30 ml/min/1.73 m(2)) on the pharmacokinetics of raltegravir. Study I evaluated the pharmacokinetics of 400 mg raltegravir in eight patients with moderate hepatic insufficiency and eight healthy, matched control subjects. Study II evaluated the pharmacokinetics of 400 mg raltegravir in 10 patients with severe renal insufficiency and 10 healthy, matched control subjects. All participants received a single 400-mg dose of raltegravir in the fasted state. In study I, the geometric mean ratios (GMR; mean value for the group with moderate hepatic insufficiency/mean value for the healthy controls) and 90% confidence intervals (CIs) for the area under the concentration-time curve from time zero to infinity (AUC(0-infinity)), the maximum concentration of drug in plasma (C(max)), and the concentration at 12 h (C(12)) were 0.86 (90% CI, 0.41, 1.77), 0.63 (90% CI, 0.23, 1.70), and 1.26 (90% CI, 0.65, 2.43), respectively. In study II, the GMRs (mean value for the group with renal insufficiency/mean value for the healthy controls) and 90% CIs for AUC(0-infinity), C(max), and C(12) were 0.85 (90% CI, 0.49, 1.49), 0.68 (90% CI, 0.35, 1.32), and 1.28 (90% CI, 0.79, 2.06), respectively. Raltegravir was generally well tolerated by patients with moderate hepatic or severe renal insufficiency, and there was no clinically important effect of moderate hepatic or severe renal insufficiency on the pharmacokinetics of raltegravir. No adjustment in the dose of raltegravir is required for patients with mild or moderate hepatic or renal insufficiency.


Subject(s)
Anti-HIV Agents/pharmacokinetics , HIV Integrase Inhibitors/pharmacokinetics , Hepatic Insufficiency/physiopathology , Pyrrolidinones/pharmacokinetics , Renal Insufficiency/physiopathology , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/adverse effects , HIV Integrase Inhibitors/administration & dosage , HIV Integrase Inhibitors/adverse effects , Hepatic Insufficiency/metabolism , Humans , Pyrrolidinones/administration & dosage , Pyrrolidinones/adverse effects , Raltegravir Potassium , Renal Insufficiency/metabolism , Treatment Outcome
16.
Clin Pharmacol Drug Dev ; 8(7): 962-970, 2019 10.
Article in English | MEDLINE | ID: mdl-31173674

ABSTRACT

Treatment of individuals coinfected with hepatitis C virus (HCV) and human immunodeficiency virus (HIV) requires careful consideration of potential drug-drug interactions. We evaluated the pharmacokinetic interaction of the direct-acting antiviral agents elbasvir and grazoprevir coadministered with the nucleotide reverse transcriptase inhibitor tenofovir disoproxil fumarate (TDF). Three open-label, multidose studies in healthy adults were conducted. In the first study (N = 10), participants received TDF 300 mg once daily, elbasvir 50 mg once daily, and elbasvir coadministered with TDF. In the second study (N = 12), participants received TDF 300 mg once daily, grazoprevir 200 mg once daily, and grazoprevir coadministered with TDF. In the third study (N = 14), participants received TDF 300 mg once daily and TDF 300 mg coadministered with coformulated elbasvir/grazoprevir 50 mg/100 mg once daily. Pharmacokinetics and safety were evaluated. Following coadministration, the tenofovir area under the plasma concentration-time curve to 24 hours and maximum plasma concentration geometric mean ratios (90% confidence intervals) for tenofovir and coadministered drug(s) versus tenofovir were 1.3 (1.2, 1.5) and 1.5 (1.3, 1.6), respectively, when coadministered with elbasvir; 1.2 (1.1, 1.3) and 1.1 (1.0, 1.2), respectively, when coadministered with grazoprevir; and 1.3 (1.2, 1.4) and 1.1 (1.0, 1.4), respectively, when coadministered with the elbasvir/grazoprevir coformulation. TDF had minimal effect on elbasvir and grazoprevir pharmacokinetics. Elbasvir and/or grazoprevir coadministered with TDF resulted in no clinically meaningful tenofovir exposure increases and was generally well tolerated, with no deaths, serious adverse events (AEs), discontinuations due to AEs, or laboratory AEs reported. No dose adjustments for elbasvir/grazoprevir or TDF are needed for coadministration in HCV/HIV-coinfected people.


Subject(s)
Antiviral Agents/pharmacokinetics , Benzofurans/pharmacokinetics , Imidazoles/pharmacokinetics , Quinoxalines/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacokinetics , Tenofovir/pharmacokinetics , Adult , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Area Under Curve , Benzofurans/administration & dosage , Benzofurans/adverse effects , Drug Administration Schedule , Drug Combinations , Drug Interactions , Female , HIV/drug effects , Healthy Volunteers , Hepacivirus/drug effects , Humans , Imidazoles/administration & dosage , Imidazoles/adverse effects , Male , Middle Aged , Quinoxalines/administration & dosage , Quinoxalines/adverse effects , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/adverse effects , Tenofovir/administration & dosage , Tenofovir/adverse effects , Young Adult
17.
J Clin Pharmacol ; 48(2): 209-14, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18077730

ABSTRACT

Raltegravir is a novel HIV-1 integrase inhibitor with potent in vitro activity (95% inhibitory concentration = 33 nM in 50% human serum). In vitro characterization of raltegravir inhibition potential was assessed against a panel of cytochrome P450 (CYP) enzymes. An open-label, 2-period study was conducted to assess the effect of raltegravir on the pharmacokinetics of midazolam, a sensitive CYP 3A4 probe substrate: period 1, 2.0 mg of midazolam; period 2, 400 mg of raltegravir every 12 hours for 14 days with 2.0 mg of midazolam on day 14. There was no meaningful in vitro effect of raltegravir on inhibition of a panel of CYP enzymes and induction of CYP 3A4. In the presence of raltegravir, midazolam area under the curve extrapolated to infinity (AUC(0-infinity)) and maximum plasma concentration (C(max)) geometric mean ratios were similar (geometric mean ratios and 90% confidence intervals: 0.92 [0.82, 1.03] (P = .208) and 1.03 [0.87, 1.22] (P = .751), respectively). No substantial differences were observed in T(max) (P = .750) or apparent half-life (P = .533) of midazolam. Plasma levels of midazolam were not substantially affected by raltegravir, which implies that raltegravir is not a clinically important inducer or inhibitor of CYP 3A4 and that raltegravir would not be expected to affect the pharmacokinetics of other drugs metabolized by CYP 3A4 to a clinically meaningful extent.


Subject(s)
Hepatocytes/drug effects , Midazolam/pharmacokinetics , Organic Chemicals/pharmacology , Area Under Curve , Cells, Cultured , Chromatography, Liquid , Cross-Over Studies , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Female , HIV Integrase Inhibitors/pharmacology , Half-Life , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Male , Mass Spectrometry , Metabolic Clearance Rate , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Midazolam/administration & dosage , Midazolam/blood , Pyrrolidinones , Raltegravir Potassium , Reverse Transcriptase Polymerase Chain Reaction
18.
Clin Pharmacokinet ; 57(3): 367-377, 2018 03.
Article in English | MEDLINE | ID: mdl-28639229

ABSTRACT

BACKGROUND AND OBJECTIVES: MHAA4549A, a human anti-influenza immunoglobulin (Ig) G1 monoclonal antibody, is being developed to treat patients hospitalized for influenza A infection. This study examined the pharmacokinetics (PKs) of MHAA4549A in a phase IIa, randomized, double-blind, dose-ranging trial in healthy volunteers challenged with influenza A virus. METHODS: Serum PK data were collected from 60 subjects in three single-dose groups (400, 1200, or 3600 mg) who received MHAA4549A intravenously 24-36 h after inoculation with the influenza A virus. Nasopharyngeal swab MHAA4549A concentration data were collected on days 1-8, and all subjects, including the placebo group, received 75 mg oseltamivir twice daily from days 7 to 11. Plasma samples were collected 4 h postdose on day 8 for oseltamivir and its active metabolite oseltamivir carboxylate (OC) (all subjects, n = 100), including subjects treated with oseltamivir alone and placebo. Noncompartmental analysis was performed for both nasal and serum PKs. RESULTS: MHAA4549A showed dose-proportional serum PKs with a long terminal half-life (approximately 21.9-24.6 days) and slow clearance (approximately 152-240 mL/day); however, nasopharyngeal swab PKs were not dose proportional. No differences in mean plasma concentrations of oseltamivir and OC at 4 h postdose on day 8 were observed between the MHAA4549A treatment and placebo groups. No subjects who received MHAA4549A developed anti-drug antibodies. CONCLUSION: MHAA4549A serum PKs were consistent with that of a human IgG1antibody without known endogenous targets. MHAA4549A showed nonlinear PKs in nasopharyngeal swab samples, which will guide future dose selection to achieve the high drug concentrations needed at the site of action for efficacy. These data demonstrate no PK interactions between MHAA4549A and oseltamivir, and support flat dosing. TRIAL REGISTRATION: ClinicalTrials.gov identifier, NCT01980966.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antiviral Agents/pharmacokinetics , Influenza A virus/drug effects , Influenza, Human/drug therapy , Adolescent , Adult , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Antiviral Agents/administration & dosage , Dose-Response Relationship, Drug , Double-Blind Method , Female , Half-Life , Humans , Influenza, Human/virology , Male , Middle Aged , Time Factors , Young Adult
19.
Clin Cancer Res ; 24(10): 2276-2284, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29506988

ABSTRACT

Purpose: Duligotuzumab is a dual-action antibody directed against EGFR and HER3.Experimental Design: Metastatic colorectal cancer (mCRC) patients with KRAS ex2 wild-type received duligotuzumab or cetuximab and FOLFIRI until progression or intolerable toxicity. Mandatory tumor samples underwent mutation and biomarker analysis. Efficacy analysis was conducted in patients with RAS exon 2/3 wild-type tumors.Results: Of 134 randomly assigned patients, 98 had RAS ex2/3 wild-type. Duligotuzumab provided no progression-free survival (PFS) or overall survival (OS) benefit compared with cetuximab, although there was a trend for a lower objective response rate (ORR) in the duligotuzumab arm. No relationship was seen between PFS or ORR and ERBB3, NRG1, or AREG expression. There were fewer skin rash events for duligotuzumab but more diarrhea. Although the incidence of grade ≥3 AEs was similar, the frequency of serious AEs was higher for duligotuzumab.Conclusions: Duligotuzumab plus FOLFIRI did not appear to improve the outcomes in patients with RAS exon 2/3 wild-type mCRC compared with cetuximab + FOLFIRI. Clin Cancer Res; 24(10); 2276-84. ©2018 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers, Tumor , Camptothecin/adverse effects , Camptothecin/therapeutic use , Cetuximab/administration & dosage , Cetuximab/pharmacokinetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Female , Fluorouracil/adverse effects , Fluorouracil/therapeutic use , Genes, ras , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/pharmacology , Leucovorin/adverse effects , Leucovorin/therapeutic use , Male , Middle Aged , Molecular Targeted Therapy , Mutation , Neoplasm Metastasis , Neoplasm Staging , Progression-Free Survival , Retreatment , Treatment Outcome , Young Adult
20.
Curr Biol ; 13(15): R611-3, 2003 Aug 05.
Article in English | MEDLINE | ID: mdl-12906816

ABSTRACT

Leukocyte recruitment to sites of inflammation is initiated by the selectin family of adhesion receptors. Recent research reveals that P-selectin binding to its ligand exhibits 'catch' to 'slip' bond transition that may help explain the shear threshold phenomenon.


Subject(s)
Ligands , Models, Molecular , P-Selectin/metabolism , Receptors, Leukocyte-Adhesion/metabolism , Cell Adhesion , Membrane Glycoproteins/metabolism , Shear Strength
SELECTION OF CITATIONS
SEARCH DETAIL