Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
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
2.
Clin Pharmacol Drug Dev ; 12(8): 810-818, 2023 08.
Article in English | MEDLINE | ID: mdl-36942507

ABSTRACT

Peroxisome proliferator-activated receptor δ (PPARδ) plays a central role in modulating mitochondrial function in ischemia-reperfusion injury. ASP1128, a potent and selective modulator of PPARδ, is currently under investigation for treating acute kidney injury. This randomized, first-in-human study assessed the safety, tolerability, pharmacokinetics, and pharmacodynamics of ASP1128 administered intravenously in healthy participants. Forty-nine participants received a single dose of ASP1128 0.3-10 mg (n = 37) or placebo (n = 12) and 53 received daily (7 days) doses of ASP1128 3-100 mg (n = 39) or placebo (n = 14), including a cohort aged ≥65 years (ASP1128 100 mg, n = 3; placebo, n = 2). Treatment-emergent adverse events occurred in 37.8%, 59.0%, and 33.3%-35.7% of participants in the single ASP1128, multiple ASP1128, and placebo groups, respectively. All were mild in severity, and the frequency of adverse events did not appear to be dose-related. One participant (multiple ASP1128 3 mg group) withdrew with an infusion site erythema, possibly related to study drug. Exposure was roughly dose-proportional, and elimination was generally consistent across doses (mean t½ 14.6-17.4 hours in the 10, 30, and 100 mg groups on day 7). There was little accumulation in plasma following multiple dosing; steady state was reached after ∼4 days. ASP1128 treatment led to rapid and dose-related upregulation of six fatty acid oxidation-related PPARδ target genes at ≥10 mg, which lasted >24 hours postdose. In conclusion, single and multiple intravenous doses of ASP1128 were generally well tolerated, with dose-dependent pharmacokinetics and target gene engagement in healthy participants.


Subject(s)
PPAR delta , Humans , Healthy Volunteers , Dose-Response Relationship, Drug , Area Under Curve , Administration, Oral
3.
J Pers Med ; 12(5)2022 Apr 21.
Article in English | MEDLINE | ID: mdl-35629092

ABSTRACT

Advances in biotechnology have enabled us to assay human tissue and cells to a depth and resolution that was never possible before, redefining what we know as the "biomarker", and how we define a "disease". This comes along with the shift of focus from a "one-drug-fits-all" to a "personalized approach", placing the drug development industry in a highly dynamic landscape, having to navigate such disruptive trends. In response to this, innovative clinical trial designs have been key in realizing biomarker-driven drug development. Regulatory approvals of cancer genome sequencing panels and associated targeted therapies has brought personalized medicines to the clinic. Increasing availability of sophisticated biotechnologies such as next-generation sequencing (NGS) has also led to a massive outflux of real-world genomic data. This review summarizes the current state of biomarker-driven drug development and highlights examples showing the utility and importance of the application of real-world data in the process. We also propose that all stakeholders in drug development should (1) be conscious of and efficiently utilize real-world evidence and (2) re-vamp the way the industry approaches drug development in this era of personalized medicines.

4.
Muscle Nerve ; 65(1): 110-120, 2022 01.
Article in English | MEDLINE | ID: mdl-34642949

ABSTRACT

INTRODUCTION/AIMS: ASP0367, or bocidelpar sulfate, is an orally administered small molecule that potently and selectively modulates peroxisome proliferator-activated receptor δ (PPARδ) to address mitochondrial dysfunction occurring in diseases including primary mitochondrial myopathy and Duchenne muscular dystrophy. The objectives of this first-in-human trial were to evaluate the safety/tolerability, pharmacokinetics, and pharmacodynamics of ASP0367 in healthy participants. METHODS: In this double-blind phase 1 study, adult participants were randomized to single or multiple ascending oral doses of ASP0367 or placebo. The study duration was 1 and 14 days, respectively. Pharmacokinetic parameters under fed conditions were also evaluated. RESULTS: A total of 64 (single-dose cohort) and 37 (multiple-dose cohort) participants were included in the study. After single doses of 1 to 120 mg, ASP0367 was rapidly absorbed, with median time to maximum plasma concentration (tmax ) of 1.50 to 2.24 hours under fasting conditions; ASP0367 concentrations declined in a multiphasic manner after reaching maximum plasma concentration. Under fed conditions, tmax was delayed 1.7 hours. After multiple once-daily doses, mean half-life of ASP0367 10 to 75 mg ranged from 14.1 to 17.5 hours; steady state was reached after 4 days. Negligible accumulation was observed after repeated dosing. No participants receiving ASP0367 discontinued treatment, and all treatment-emergent adverse events were mild to moderate in severity; none were considered drug-related. No clinically significant changes were observed on laboratory or electrocardiographic evaluation. Treatment- and dose-dependent upregulation of six PPARδ target genes was observed with single and multiple doses of ASP0367. DISCUSSION: ASP0367, or bocidelpar sulfate, was well tolerated; rapid absorption, roughly dose-proportional bioavailability, and effects on PPARδ target genes were demonstrated in healthy adult participants.


Subject(s)
Caproates , Imidazoles , PPAR delta , Sulfates , Administration, Oral , Adult , Area Under Curve , Caproates/therapeutic use , Dose-Response Relationship, Drug , Double-Blind Method , Healthy Volunteers , Humans , Imidazoles/therapeutic use , Sulfates/therapeutic use
5.
Biol Pharm Bull ; 41(3): 440-444, 2018.
Article in English | MEDLINE | ID: mdl-29491221

ABSTRACT

Liver sinusoidal endothelial cells (LSECs), which are specialized endothelial cells that line liver sinusoids, have been reported to participate in a variety of liver functions, such as blood macromolecule clearance and factor VIII production. In addition, LSECs play crucial roles in liver regeneration following acute liver injury, as well as the development and progression of liver diseases or drug-induced hepatotoxicity. However, the molecular mechanisms underlying their roles remain mostly unknown. Therefore, in order to contribute to the clarification of those mechanisms, herein we report on the development of a new immortalized human LSEC (HLSEC) line. To produce this cell line, two immortalized genes were introduced into the primary HLSECs, which eventually resulted in the establishment of the HLSEC/conditionally immortalized, clone-J (HLSEC/ciJ). Consistent with the two-immortalized gene expression, HLSEC/ciJ showed excellent proliferation activity. Additionally, the results of gene expression analyses showed that several LSEC (as well as pan-endothelial) marker mRNAs and proteins were clearly expressed in HLSEC/ciJ. Furthermore, we found that adherence junction proteins were localized at the cell border in the HLSEC/ciJ monolayer, and that the cells exhibited a tube-like structure formation property. Taken together, the results obtained thus far indicate that we have successfully immortalized HLSECs, resulting in creation of HLSEC/ciJ, a cell line that possesses infinite proliferation ability while retaining possession of at least some HLSEC features. We believe that the HLSEC/ciJ have the potential to provide a valuable and unlimited alternative source of HLSECs for use in liver/LSEC physiology/pathophysiology, pharmacology, and toxicology studies.


Subject(s)
Endothelial Cells/drug effects , Liver/cytology , Cell Culture Techniques , Cell Line , Cell Proliferation , Cryopreservation , DNA, Complementary/biosynthesis , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Gene Expression , Hepatocytes , Humans , Liver/metabolism , RNA/biosynthesis
6.
Drug Metab Pharmacokinet ; 31(1): 102-105, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26825849

ABSTRACT

Ornithine carbamoyltransferase (OCT) is a mitochondrial protein expressed primarily in the liver. It has been shown that, like alanine aminotransferase (ALT), OCT is released from damaged hepatocytes in rats and humans, which has given rise to the possibility that OCT might provide a diagnostic biomarker of various forms of liver damage, including drug-induced liver injury (DILI). However, OCT release characteristics in DILI, as well as their diagnostic advantages, remain elusive. Therefore, this study aimed at clarifying whether and how OCT is released from rat primary hepatocytes in vitro using seven potentially hepatotoxic drugs. The results showed that OCT releases from damaged hepatocytes were observed for all tested drugs, and that those releases were not associated with mitochondrial membrane proteins. It should be underscored that the release dynamics were significantly larger than those of ALT. Furthermore, unlike ALT, the maximum OCT release levels showed differences depending on the drug being tested, suggesting that OCT release was susceptible to toxicity mechanisms. Taken together, these unique release characteristics highlight the possibility that OCT could provide a promising DILI biomarker that might contribute not only to diagnostic accuracy improvements, but also to a better understanding of toxicity types in clinical and drug development settings.


Subject(s)
Biomarkers/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Hepatocytes/metabolism , Ornithine Carbamoyltransferase/metabolism , Alanine Transaminase/metabolism , Animals , Liver/metabolism , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL