Xiuqi Li, Xiaofei Wu, Aihong Huo, Guanghuai Zeng, Richard Jones, Rui Chen, Hongyun Wang
Phosphodiesterase 4 (PDE4) is a branch of the phosphodiesterase isoenzyme family and plays a crucial role in maintaining intracellular cAMP homeostasis. Mufemilast, a novel PDE4 inhibitor, has demonstrated anti-inflammatory effects in preclinical studies and holds promise for treating inflammatory diseases. The pharmacokinetics, safety, and tolerability profiles of mufemilast were evaluated in healthy participants. In the single ascending dose study, 68 healthy male subjects were randomized to receive single oral doses of mufemilast ranging from 10 to 125 mg. In the multiple ascending dose study, 24 healthy subjects received mufemilast at doses of 15, 30, and 60 mg twice daily for 7 consecutive days. In the two-period, crossover food effect study, 12 healthy subjects were administered a 52.5-mg dose of mufemilast both in the fasted and fed state. The results showed that mufemilast was rapidly absorbed and the exposure increased with dose. Following multiple doses, the mean accumulation ratios indicated some accumulations of mufemilast. The Tmax was 3 and 5 h under fasted and fed conditions, while the geometric mean ratio and 90% CIs for AUClast, AUCinf, and Cmax were 105.76 [92.69%,120.66%], 105.60 [92.52%,120.52%], and 92.85 [78.60%,109.68%], respectively. Most AEs were grade 1 or 2, with positive occult blood test as the most common. Mufemilast was safe and tolerated by healthy participants across all dose groups (10–125 mg). PK analysis revealed that mufemilast exhibited linear PK characteristics. These results support the further evaluation of its efficacy.
{"title":"Safety, Tolerability, and Pharmacokinetics of Mufemilast, a PDE4 Inhibitor, in Healthy Participants: A First-in-Human Phase 1 Study","authors":"Xiuqi Li, Xiaofei Wu, Aihong Huo, Guanghuai Zeng, Richard Jones, Rui Chen, Hongyun Wang","doi":"10.1002/cpdd.70005","DOIUrl":"https://doi.org/10.1002/cpdd.70005","url":null,"abstract":"<p>Phosphodiesterase 4 (PDE4) is a branch of the phosphodiesterase isoenzyme family and plays a crucial role in maintaining intracellular cAMP homeostasis. Mufemilast, a novel PDE4 inhibitor, has demonstrated anti-inflammatory effects in preclinical studies and holds promise for treating inflammatory diseases. The pharmacokinetics, safety, and tolerability profiles of mufemilast were evaluated in healthy participants. In the single ascending dose study, 68 healthy male subjects were randomized to receive single oral doses of mufemilast ranging from 10 to 125 mg. In the multiple ascending dose study, 24 healthy subjects received mufemilast at doses of 15, 30, and 60 mg twice daily for 7 consecutive days. In the two-period, crossover food effect study, 12 healthy subjects were administered a 52.5-mg dose of mufemilast both in the fasted and fed state. The results showed that mufemilast was rapidly absorbed and the exposure increased with dose. Following multiple doses, the mean accumulation ratios indicated some accumulations of mufemilast. The T<sub>max</sub> was 3 and 5 h under fasted and fed conditions, while the geometric mean ratio and 90% CIs for AUC<sub>last</sub>, AUC<sub>inf</sub>, and C<sub>max</sub> were 105.76 [92.69%,120.66%], 105.60 [92.52%,120.52%], and 92.85 [78.60%,109.68%], respectively. Most AEs were grade 1 or 2, with positive occult blood test as the most common. Mufemilast was safe and tolerated by healthy participants across all dose groups (10–125 mg). PK analysis revealed that mufemilast exhibited linear PK characteristics. These results support the further evaluation of its efficacy.</p>","PeriodicalId":10495,"journal":{"name":"Clinical Pharmacology in Drug Development","volume":"15 1","pages":""},"PeriodicalIF":1.8,"publicationDate":"2026-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145987223","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Obesity has emerged as a global health crisis requiring innovative therapeutic strategies beyond conventional approaches. While glucagon-like peptide-1 (GLP-1) and dual GIP/GLP-1 receptor agonists have redefined pharmacological management, their limitations necessitate further innovation. Retatrutide (LY3437943), a novel triple agonist targeting GLP-1, glucose-dependent insulinotropic polypeptide (GIP), and glucagon receptors, represents a transformative advance in obesity pharmacotherapy. Phase 2 trials report unprecedented weight reductions, comparable to bariatric surgery, with additional benefits for metabolic comorbidities such as NASH and cardiovascular disease. Retatrutide exemplifies rational multi-agonist peptide engineering and signals a paradigm shift in systems pharmacology. This perspective underscores the urgent need for scientific engagement, equity considerations, and policy preparedness, positioning retatrutide as a watershed in obesity treatment and a blueprint for future poly-agonist therapies.
{"title":"The Triple-Agonist Revolution: Retatrutide and the Paradigm Shift in Multi-Hormonal Pharmacotherapy for Obesity and Cardiometabolic Comorbidities","authors":"Nila Ganamurali, Sarvesh Sabarathinam","doi":"10.1002/cpdd.70001","DOIUrl":"https://doi.org/10.1002/cpdd.70001","url":null,"abstract":"<p>Obesity has emerged as a global health crisis requiring innovative therapeutic strategies beyond conventional approaches. While glucagon-like peptide-1 (GLP-1) and dual GIP/GLP-1 receptor agonists have redefined pharmacological management, their limitations necessitate further innovation. Retatrutide (LY3437943), a novel triple agonist targeting GLP-1, glucose-dependent insulinotropic polypeptide (GIP), and glucagon receptors, represents a transformative advance in obesity pharmacotherapy. Phase 2 trials report unprecedented weight reductions, comparable to bariatric surgery, with additional benefits for metabolic comorbidities such as NASH and cardiovascular disease. Retatrutide exemplifies rational multi-agonist peptide engineering and signals a paradigm shift in systems pharmacology. This perspective underscores the urgent need for scientific engagement, equity considerations, and policy preparedness, positioning retatrutide as a watershed in obesity treatment and a blueprint for future poly-agonist therapies.</p>","PeriodicalId":10495,"journal":{"name":"Clinical Pharmacology in Drug Development","volume":"15 1","pages":""},"PeriodicalIF":1.8,"publicationDate":"2026-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145983715","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Angela Jeong, Kathleen Weisel, Dessislava Dimitrova, Brianna Donnelly, Daniel Wang, An Vermeulen, Zhenhua Xu
Guselkumab is approved for the treatment of psoriasis, psoriatic arthritis, as well as ulcerative colitis and Crohn's disease. Two delivery devices for subcutaneous (SC) injection previously had been approved for the administration of 100 mg guselkumab. This study was designed to demonstrate the bioequivalence and tolerability of guselkumab following a single-dose SC administration of 100 mg guselkumab using a 1 mL prefilled syringe (PFS) assembled with an Ypsomate autoinjector (i.e., 1 mL PFS-Y, Test device) as compared to the approved 1 mL PFS assembled with the UltraSafe Plus needle safety guard (i.e., 1 mL PFS-U, Reference device). Mean serum guselkumab concentration–time profiles were nearly superimposable for both devices following a single SC injection. The geometric mean ratios and the corresponding 90% confidence interval [CI] for Cmax and AUCinf were 102.28% (94.85%–110.30%) and 102.10% (95.19%–109.51%), respectively. There were no significant differences in the incidence of treatment-emergent adverse events between the two treatment groups, and the incidence of treatment-emergent anti-drug antibodies was low and comparable between the two groups. Overall, these results suggest that the pharmacokinetics, safety/tolerability, and immunogenicity of guselkumab are comparable when administered via the 1 mL PFS-Y and 1 mL PFS-U.
{"title":"A Phase 1 Bioequivalence Study to Assess the Pharmacokinetics, Safety and Tolerability of Guselkumab After a Single-Dose Administration via Two Subcutaneous Injection Devices in Healthy Volunteers","authors":"Angela Jeong, Kathleen Weisel, Dessislava Dimitrova, Brianna Donnelly, Daniel Wang, An Vermeulen, Zhenhua Xu","doi":"10.1002/cpdd.70007","DOIUrl":"https://doi.org/10.1002/cpdd.70007","url":null,"abstract":"<p>Guselkumab is approved for the treatment of psoriasis, psoriatic arthritis, as well as ulcerative colitis and Crohn's disease. Two delivery devices for subcutaneous (SC) injection previously had been approved for the administration of 100 mg guselkumab. This study was designed to demonstrate the bioequivalence and tolerability of guselkumab following a single-dose SC administration of 100 mg guselkumab using a 1 mL prefilled syringe (PFS) assembled with an Ypsomate autoinjector (i.e., 1 mL PFS-Y, Test device) as compared to the approved 1 mL PFS assembled with the UltraSafe Plus needle safety guard (i.e., 1 mL PFS-U, Reference device). Mean serum guselkumab concentration–time profiles were nearly superimposable for both devices following a single SC injection. The geometric mean ratios and the corresponding 90% confidence interval [CI] for C<sub>max</sub> and AUC<sub>inf</sub> were 102.28% (94.85%–110.30%) and 102.10% (95.19%–109.51%), respectively. There were no significant differences in the incidence of treatment-emergent adverse events between the two treatment groups, and the incidence of treatment-emergent anti-drug antibodies was low and comparable between the two groups. Overall, these results suggest that the pharmacokinetics, safety/tolerability, and immunogenicity of guselkumab are comparable when administered via the 1 mL PFS-Y and 1 mL PFS-U.</p>","PeriodicalId":10495,"journal":{"name":"Clinical Pharmacology in Drug Development","volume":"15 1","pages":""},"PeriodicalIF":1.8,"publicationDate":"2026-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145983689","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Non-compartmental analysis (NCA) is commonly used to estimate pharmacokinetic (PK) parameters in individual participants during Phase 1 studies. PK sampling schedules are important for accurately characterizing a drug's PK profile. However, collecting blood samples on time is often challenging, even in Phase 1 studies involving healthy participants, due to tightly scheduled study activities. To address these constraints, sampling windows—defined as permissible time intervals for blood sample collection—are often implemented. These windows provide operational flexibility at clinical study sites while maintaining a reasonable level of accuracy in parameter estimation. To date, no published literature has described the construction of sampling windows for NCA, and the general practice of constructing sampling windows remains unstandardized. In this study, we evaluated the impact of varying sampling window lengths on NCA-derived PK parameters, leveraging bioequivalence criteria as an indicator to assess the effects and provide insights into the construction of sampling windows for NCA. As a result, we were able to quantitatively evaluate how changes in sampling window length affect NCA-based PK parameters. Based on these findings, we provide recommendations for appropriate sampling windows.
{"title":"Impact of Sampling Window Variability on Pharmacokinetic Parameters Estimated by Non-Compartmental Analysis: Case Studies of Various Types of Drugs","authors":"Kyosuke Takahashi, Kazuhiko Asari, Kazuhiko Hanada","doi":"10.1002/cpdd.70004","DOIUrl":"https://doi.org/10.1002/cpdd.70004","url":null,"abstract":"<p>Non-compartmental analysis (NCA) is commonly used to estimate pharmacokinetic (PK) parameters in individual participants during Phase 1 studies. PK sampling schedules are important for accurately characterizing a drug's PK profile. However, collecting blood samples on time is often challenging, even in Phase 1 studies involving healthy participants, due to tightly scheduled study activities. To address these constraints, sampling windows—defined as permissible time intervals for blood sample collection—are often implemented. These windows provide operational flexibility at clinical study sites while maintaining a reasonable level of accuracy in parameter estimation. To date, no published literature has described the construction of sampling windows for NCA, and the general practice of constructing sampling windows remains unstandardized. In this study, we evaluated the impact of varying sampling window lengths on NCA-derived PK parameters, leveraging bioequivalence criteria as an indicator to assess the effects and provide insights into the construction of sampling windows for NCA. As a result, we were able to quantitatively evaluate how changes in sampling window length affect NCA-based PK parameters. Based on these findings, we provide recommendations for appropriate sampling windows.</p>","PeriodicalId":10495,"journal":{"name":"Clinical Pharmacology in Drug Development","volume":"15 1","pages":""},"PeriodicalIF":1.8,"publicationDate":"2026-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145987225","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Thomas M. Polasek, Kushal J. Paneliya, Tom Lin, Nathan L. Mata, Irene Wang, Hendrik P. Scholl, Jessyca Lin
Tinlarebant is an oral retinol binding protein 4 antagonist in clinical development for geographic atrophy, an advanced stage of dry age-related macular degeneration, and Stargardt disease, an inherited juvenile-onset macular degeneration. A randomized, open-label, two-period, interaction study in healthy adults was conducted in four parts to determine the effects of gastric acid suppression (omeprazole 40 mg QD), cytochrome P4503A (CYP3A) inhibition (itraconazole 200 mg BD) and induction (rifampin 600 mg QD), and food on the pharmacokinetics of tinlarebant (5 mg single dose). The effects on tinlarebant exposure were quantified by geometric least squares (GLS) mean Cmax and AUCinf ratios, where GLS mean Cmax or AUCinf with potential perpetrator is divided by GLS mean Cmax or AUCinf without potential perpetrator. Steady-state dosing of omeprazole had no effect on tinlarebant exposure (Cmax ratio = 1.16 and AUCinf ratio = 1.03). The Cmax and AUCinf ratios of tinlarebant following itraconazole dosing were 1.29 and 2.42, respectively. Rifampin co-administration decreased tinlarebant Cmax and AUCinf ratios to 0.53 and 0.19, respectively. Compared with the fasting state, taking tinlarebant with food gave Cmax and AUCinf ratios in the range 1.08–1.22. No unexpected safety signals occurred and tinlarebant was well tolerated in all participants. These data show that the pharmacokinetics of tinlarebant is not significantly altered by gastric acid suppression or food. Dosing patients with tinlarebant and strong CYP3A inhibitors is unlikely to compromise safety based on its pharmacokinetic–pharmacodynamic relationships, but tinlarebant should be contraindicated with strong CYP3A inducers due to potential treatment failure.
{"title":"Effects of Gastric Acid Suppression, Cytochrome P4503A Inhibition and Induction, and Food on the Pharmacokinetics of Tinlarebant in Healthy Adults","authors":"Thomas M. Polasek, Kushal J. Paneliya, Tom Lin, Nathan L. Mata, Irene Wang, Hendrik P. Scholl, Jessyca Lin","doi":"10.1002/cpdd.70009","DOIUrl":"https://doi.org/10.1002/cpdd.70009","url":null,"abstract":"<p>Tinlarebant is an oral retinol binding protein 4 antagonist in clinical development for geographic atrophy, an advanced stage of dry age-related macular degeneration, and Stargardt disease, an inherited juvenile-onset macular degeneration. A randomized, open-label, two-period, interaction study in healthy adults was conducted in four parts to determine the effects of gastric acid suppression (omeprazole 40 mg QD), cytochrome P4503A (CYP3A) inhibition (itraconazole 200 mg BD) and induction (rifampin 600 mg QD), and food on the pharmacokinetics of tinlarebant (5 mg single dose). The effects on tinlarebant exposure were quantified by geometric least squares (GLS) mean C<sub>max</sub> and AUC<sub>inf</sub> ratios, where GLS mean C<sub>max</sub> or AUC<sub>inf</sub> with potential perpetrator is divided by GLS mean C<sub>max</sub> or AUC<sub>inf</sub> without potential perpetrator. Steady-state dosing of omeprazole had no effect on tinlarebant exposure (C<sub>max</sub> ratio = 1.16 and AUC<sub>inf</sub> ratio = 1.03). The C<sub>max</sub> and AUC<sub>inf</sub> ratios of tinlarebant following itraconazole dosing were 1.29 and 2.42, respectively. Rifampin co-administration decreased tinlarebant C<sub>max</sub> and AUC<sub>inf</sub> ratios to 0.53 and 0.19, respectively. Compared with the fasting state, taking tinlarebant with food gave C<sub>max</sub> and AUC<sub>inf</sub> ratios in the range 1.08–1.22. No unexpected safety signals occurred and tinlarebant was well tolerated in all participants. These data show that the pharmacokinetics of tinlarebant is not significantly altered by gastric acid suppression or food. Dosing patients with tinlarebant and strong CYP3A inhibitors is unlikely to compromise safety based on its pharmacokinetic–pharmacodynamic relationships, but tinlarebant should be contraindicated with strong CYP3A inducers due to potential treatment failure.</p>","PeriodicalId":10495,"journal":{"name":"Clinical Pharmacology in Drug Development","volume":"15 1","pages":""},"PeriodicalIF":1.8,"publicationDate":"2026-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145983687","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-01Epub Date: 2025-07-03DOI: 10.1002/cpdd.1571
Hegui Yan, Yu Peng, Zhixiang Pan, Yiyi Wang, Quan Li, Guan Liu
Aildenafil citrate is a novel and potent phosphodiesterase type 5 inhibitor for the treatment of erectile dysfunction (ED). This study evaluates the pharmacokinetics and bioequivalence of 2 formulations of aildenafil citrate tablets (30 and 60 mg) in healthy Chinese male subjects under both fasting and fed conditions. A single-center, randomized, open-label, 2-period crossover design was employed, with 78 participants enrolled, including 30 in the fasting condition and 48 in the fed condition. Blood samples were collected at multiple time points for pharmacokinetic analysis, which included Cmax, AUC0-t, and AUC0-∞ as the primary parameters. The results demonstrated that the pharmacokinetic profiles of the test and reference formulations were comparable in both the fasting and fed states, with 90% confidence intervals for the geometric mean ratios of Cmax, AUC0-t, and AUC0-∞ falling within the 80%-125% range, confirming bioequivalence. Although food intake slightly delayed the time to peak concentration and reduced the absorption rate, it did not significantly affect the overall bioavailability. Both formulations were well tolerated, with adverse events being mild and resolving spontaneously. This study provides evidence supporting the bioequivalence of the 2 aildenafil citrate formulations and their interchangeability for clinical use in treating ED.
{"title":"Pharmacokinetics and Bioequivalence Evaluation of Two Different Aildenafil Citrate Tablet Formulations in Healthy Chinese Male Subjects Under Fasting and Fed Conditions.","authors":"Hegui Yan, Yu Peng, Zhixiang Pan, Yiyi Wang, Quan Li, Guan Liu","doi":"10.1002/cpdd.1571","DOIUrl":"10.1002/cpdd.1571","url":null,"abstract":"<p><p>Aildenafil citrate is a novel and potent phosphodiesterase type 5 inhibitor for the treatment of erectile dysfunction (ED). This study evaluates the pharmacokinetics and bioequivalence of 2 formulations of aildenafil citrate tablets (30 and 60 mg) in healthy Chinese male subjects under both fasting and fed conditions. A single-center, randomized, open-label, 2-period crossover design was employed, with 78 participants enrolled, including 30 in the fasting condition and 48 in the fed condition. Blood samples were collected at multiple time points for pharmacokinetic analysis, which included C<sub>max</sub>, AUC<sub>0-t</sub>, and AUC<sub>0-∞</sub> as the primary parameters. The results demonstrated that the pharmacokinetic profiles of the test and reference formulations were comparable in both the fasting and fed states, with 90% confidence intervals for the geometric mean ratios of C<sub>max</sub>, AUC<sub>0-t</sub>, and AUC<sub>0-∞</sub> falling within the 80%-125% range, confirming bioequivalence. Although food intake slightly delayed the time to peak concentration and reduced the absorption rate, it did not significantly affect the overall bioavailability. Both formulations were well tolerated, with adverse events being mild and resolving spontaneously. This study provides evidence supporting the bioequivalence of the 2 aildenafil citrate formulations and their interchangeability for clinical use in treating ED.</p>","PeriodicalId":10495,"journal":{"name":"Clinical Pharmacology in Drug Development","volume":" ","pages":"e1571"},"PeriodicalIF":1.8,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144552508","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Gerard Greig, Justin Hay, Patricia Valencia, Mena Boules, Tomasz Masior
Intestinal failure-associated liver disease occurs in 20% to 30% of patients with short bowel syndrome and intestinal failure (SBS-IF). Apraglutide is a glucagon-like peptide-2 (GLP-2) analog in clinical development for the treatment of patients with SBS-IF. This study assessed the potential for changes in exposure of apraglutide in individuals with impaired hepatic function versus healthy volunteers. In this Phase 1, open-label, nonrandomized, single-dose trial, apraglutide 3.5 mg was administered to participants with moderate hepatic impairment (Child-Pugh B) or normal hepatic function. Primary pharmacokinetic endpoints were area under the plasma concentration-time curve (AUC) from time 0 to infinity (AUCinf) or AUC from time 0 to last quantifiable concentration (AUClast) if AUCinf could not be reliably estimated, AUC0-168 h, and maximum observed plasma concentration (Cmax). Secondary endpoints included safety and tolerability. Each group comprised eight participants. No increased apraglutide exposure was observed in individuals with moderate hepatic impairment. A lower Cmax and AUCinf of apraglutide was observed in individuals with moderate hepatic impairment versus those with normal hepatic function (Cmax = 58.7 vs 71.3 ng/mL; AUCinf = 4086 vs 5351 h ng/mL, respectively). The respective geometric mean ratios were 0.835 and 0.936 for Cmax and AUCinf, and the upper bounds of their 90% confidence intervals indicate that participants with moderate hepatic impairment were not overexposed to apraglutide versus those with normal hepatic function. Adverse events were mild or moderate in severity. The results of this trial suggest that apraglutide does not require dose alteration in patients with mild and moderate hepatic impairment.
20%至30%的短肠综合征和肠衰竭(SBS-IF)患者发生肠衰竭相关肝病。Apraglutide是临床开发用于治疗SBS-IF患者的胰高血糖素样肽-2 (GLP-2)类似物。本研究评估了肝功能受损个体与健康志愿者在阿普鲁肽暴露方面的潜在变化。在这项开放标签、非随机、单剂量的1期临床试验中,阿普鲁肽3.5 mg被给予中度肝功能损害(Child-Pugh B)或肝功能正常的参与者。主要药代动力学终点为0-∞时间血浆浓度-时间曲线下面积(AUC) (AUCinf)或0-∞时间至最后可量化浓度(AUClast)(如果AUCinf不能可靠估计)、AUC0-168 h和最大观察血浆浓度(Cmax)。次要终点包括安全性和耐受性。每组由8名参与者组成。中度肝功能损害患者未观察到阿普鲁肽暴露增加。中度肝功能损害患者与肝功能正常者相比,阿普拉鲁肽的Cmax和AUCinf较低(Cmax = 58.7 vs 71.3 ng/mL; AUCinf = 4086 vs 5351 h ng/mL)。Cmax和AUCinf的几何平均比值分别为0.835和0.936,其90%置信区间的上界表明,中度肝功能损害的参与者与肝功能正常的参与者相比,并未过度暴露于阿拉格鲁肽。不良事件的严重程度为轻度或中度。该试验的结果表明,阿普鲁肽在轻度和中度肝功能损害患者中不需要改变剂量。
{"title":"Pharmacokinetics and Safety of Single-Dose Apraglutide in Individuals with Normal and Impaired Hepatic Function: A Phase 1, Open-Label Trial.","authors":"Gerard Greig, Justin Hay, Patricia Valencia, Mena Boules, Tomasz Masior","doi":"10.1002/cpdd.70006","DOIUrl":"https://doi.org/10.1002/cpdd.70006","url":null,"abstract":"<p><p>Intestinal failure-associated liver disease occurs in 20% to 30% of patients with short bowel syndrome and intestinal failure (SBS-IF). Apraglutide is a glucagon-like peptide-2 (GLP-2) analog in clinical development for the treatment of patients with SBS-IF. This study assessed the potential for changes in exposure of apraglutide in individuals with impaired hepatic function versus healthy volunteers. In this Phase 1, open-label, nonrandomized, single-dose trial, apraglutide 3.5 mg was administered to participants with moderate hepatic impairment (Child-Pugh B) or normal hepatic function. Primary pharmacokinetic endpoints were area under the plasma concentration-time curve (AUC) from time 0 to infinity (AUC<sub>inf</sub>) or AUC from time 0 to last quantifiable concentration (AUC<sub>last</sub>) if AUC<sub>inf</sub> could not be reliably estimated, AUC<sub>0-168 h</sub>, and maximum observed plasma concentration (C<sub>max</sub>). Secondary endpoints included safety and tolerability. Each group comprised eight participants. No increased apraglutide exposure was observed in individuals with moderate hepatic impairment. A lower C<sub>max</sub> and AUC<sub>inf</sub> of apraglutide was observed in individuals with moderate hepatic impairment versus those with normal hepatic function (C<sub>max</sub> = 58.7 vs 71.3 ng/mL; AUC<sub>inf</sub> = 4086 vs 5351 h ng/mL, respectively). The respective geometric mean ratios were 0.835 and 0.936 for C<sub>max</sub> and AUC<sub>inf</sub>, and the upper bounds of their 90% confidence intervals indicate that participants with moderate hepatic impairment were not overexposed to apraglutide versus those with normal hepatic function. Adverse events were mild or moderate in severity. The results of this trial suggest that apraglutide does not require dose alteration in patients with mild and moderate hepatic impairment.</p>","PeriodicalId":10495,"journal":{"name":"Clinical Pharmacology in Drug Development","volume":"15 1","pages":"e70006"},"PeriodicalIF":1.8,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145988559","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Zavegepant is the only calcitonin gene-related peptide antagonist approved as a nasal spray in the United States for acute treatment of migraine in adults with or without aura. This Phase 1, open-label study evaluated the pharmacokinetics, safety, and tolerability of a single intranasal dose of zavegepant 10 mg in 12 healthy Chinese adults. Blood samples were collected for pharmacokinetic assessment prior to dosing and from 5 min to 24 h post dose. Geometric mean values for the primary pharmacokinetic parameters were 53.53 ng h/mL for area under the plasma concentration-time curve (AUC) from time zero to infinity, 44.25 ng h/mL for AUC from time zero to time of last quantifiable concentration, and 20.32 ng/mL for maximum plasma concentration (Cmax). Secondary parameters included time to Cmax (median, 0.58 h), apparent clearance (geometric mean, 186.8 L/h), apparent volume of distribution (geometric mean, 2943 L), and terminal half-life (arithmetic mean, 11.0 h). Results were comparable to exposures observed previously in non-Asian healthy participants following a single intranasal dose of zavegepant 10 mg. Zavegepant demonstrated a favorable safety profile, with no serious or severe adverse events and no clinically relevant findings regarding laboratory tests, vital signs, or electrocardiograms observed.
Zavegepant是唯一一种降钙素基因相关肽拮抗剂,在美国被批准作为鼻喷雾剂用于有或无先兆的成人偏头痛的急性治疗。这项1期开放标签研究评估了12名健康中国成年人单次鼻内给药10mg zavegepent的药代动力学、安全性和耐受性。在给药前和给药后5分钟至24小时采集血样进行药代动力学评估。主要药代动力学参数从时间0到无穷远的血药浓度-时间曲线下面积(AUC)几何平均值为53.53 ng h/mL,从时间0到最后可量化浓度时间(AUC)几何平均值为44.25 ng h/mL,最大血药浓度(Cmax)几何平均值为20.32 ng/mL。次要参数包括到达Cmax的时间(中位数,0.58 h)、表观间隙(几何平均值,186.8 L/h)、表观分布体积(几何平均值,2943 L)和终末半衰期(算术平均值,11.0 h)。结果与先前在非亚洲健康参与者中观察到的单次鼻内给药10mg zavegepent的暴露相当。Zavegepant显示出良好的安全性,没有严重或严重的不良事件,也没有在实验室检查、生命体征或心电图方面观察到临床相关发现。
{"title":"A Phase 1 Open-Label Study to Evaluate the Pharmacokinetics, Safety, and Tolerability of a Single Intranasal Dose of Zavegepant in Healthy Chinese Adults.","authors":"Qiong Wei, Jufang Wu, Yangyi Dai, Jiaye Lin, Beikang Ge, Yanhui Sun, Jing Zhang, Jing Wang, Chen Li, Ding Ding, Jing Liu, Mohamed H Shahin","doi":"10.1002/cpdd.1617","DOIUrl":"10.1002/cpdd.1617","url":null,"abstract":"<p><p>Zavegepant is the only calcitonin gene-related peptide antagonist approved as a nasal spray in the United States for acute treatment of migraine in adults with or without aura. This Phase 1, open-label study evaluated the pharmacokinetics, safety, and tolerability of a single intranasal dose of zavegepant 10 mg in 12 healthy Chinese adults. Blood samples were collected for pharmacokinetic assessment prior to dosing and from 5 min to 24 h post dose. Geometric mean values for the primary pharmacokinetic parameters were 53.53 ng h/mL for area under the plasma concentration-time curve (AUC) from time zero to infinity, 44.25 ng h/mL for AUC from time zero to time of last quantifiable concentration, and 20.32 ng/mL for maximum plasma concentration (C<sub>max</sub>). Secondary parameters included time to C<sub>max</sub> (median, 0.58 h), apparent clearance (geometric mean, 186.8 L/h), apparent volume of distribution (geometric mean, 2943 L), and terminal half-life (arithmetic mean, 11.0 h). Results were comparable to exposures observed previously in non-Asian healthy participants following a single intranasal dose of zavegepant 10 mg. Zavegepant demonstrated a favorable safety profile, with no serious or severe adverse events and no clinically relevant findings regarding laboratory tests, vital signs, or electrocardiograms observed.</p>","PeriodicalId":10495,"journal":{"name":"Clinical Pharmacology in Drug Development","volume":" ","pages":"e1617"},"PeriodicalIF":1.8,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145318192","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
<p>There are some beliefs in science that, as practicing pharmacokineticists and statisticians, we do not question too much. This is particularly true if those beliefs are written in a guidance.<span><sup>1</sup></span> We are busy, there are not enough hours in the working day to tilt at windmills, and no one else seems to be complaining, so we accept that belief. If that belief is codified in a guidance, then we must accept it, even though there are times when a fleeting doubt as to that belief's validity enters our minds.</p><p>One of these beliefs that we, the authors, would like to challenge is how the limit of quantitation (LOQ) is currently used for the reporting of drug concentration data. The LOQ is the lowest concentration in a validated drug assay with acceptable accuracy and precision (typically within ±15%–20% of the nominal concentration)<span><sup>1</sup></span>. On its face, this does not seem too unreasonable. But it is the way that the LOQ is subsequently used to report data that is the real problem.</p><p>Let us look at an example. Suppose we have an assay with an LOQ of 0.05 ng/mL. The CV% at this concentration is 15%, so this is a reasonable LOQ. Figure 1 shows the distribution of many repeated measurements (>30) from a spiked QC sample of 0.05 ng/mL. Because of the 15% variability, each measured sample will differ from the nominal concentration—it will either be above 0.05 or below, and if normally distributed (as we have assumed) these measurements will be distributed 50:50 above and below the mean. The problem comes when the analytical chemist reports these values. Those values ≥0.05 ng/mL are reported (Figure 1 in green), but any values <0.05 ng/mL are not reported (Figure 1 in red), even though they come from the exact same distribution! The samples <LOQ are given the designation “BQL” (below quantitation limit), and the actual values will never be reported no matter how much one pleads, upon threat of receiving a Form 483 from the FDA. In effect, the analytical chemist censors all data <LOQ, even though an acceptable LOQ accuracy is <b><i>plus or minus</i></b> 15%–20% of the nominal concentration.</p><p>It gets worse when we pharmacokineticists receive the data. If we are performing a non-compartmental analysis, we change all the BQLs to zeros as we are instructed,<span><sup>2</sup></span> even though we know that there are actual values lurking there somewhere. We know this because “real” zero concentrations are reported as such. But we pretend that the BQLs are zero anyway because the guidance or white paper tells us to do so. If we are performing a population PK analysis, and there are enough BQLs to concern us (typically 10% or more of the total number of samples<span><sup>3</sup></span>), then we use a sophisticated imputation method for those BQLs. We sleep well at night knowing that our data are safe from any sordid contamination by BQL samples, and we (secretly) pat ourselves on the back for knowing
{"title":"It Is Time to Re-Think the Limit of Quantitation","authors":"Michael J. Fossler, Brian Smith","doi":"10.1002/cpdd.70000","DOIUrl":"https://doi.org/10.1002/cpdd.70000","url":null,"abstract":"<p>There are some beliefs in science that, as practicing pharmacokineticists and statisticians, we do not question too much. This is particularly true if those beliefs are written in a guidance.<span><sup>1</sup></span> We are busy, there are not enough hours in the working day to tilt at windmills, and no one else seems to be complaining, so we accept that belief. If that belief is codified in a guidance, then we must accept it, even though there are times when a fleeting doubt as to that belief's validity enters our minds.</p><p>One of these beliefs that we, the authors, would like to challenge is how the limit of quantitation (LOQ) is currently used for the reporting of drug concentration data. The LOQ is the lowest concentration in a validated drug assay with acceptable accuracy and precision (typically within ±15%–20% of the nominal concentration)<span><sup>1</sup></span>. On its face, this does not seem too unreasonable. But it is the way that the LOQ is subsequently used to report data that is the real problem.</p><p>Let us look at an example. Suppose we have an assay with an LOQ of 0.05 ng/mL. The CV% at this concentration is 15%, so this is a reasonable LOQ. Figure 1 shows the distribution of many repeated measurements (>30) from a spiked QC sample of 0.05 ng/mL. Because of the 15% variability, each measured sample will differ from the nominal concentration—it will either be above 0.05 or below, and if normally distributed (as we have assumed) these measurements will be distributed 50:50 above and below the mean. The problem comes when the analytical chemist reports these values. Those values ≥0.05 ng/mL are reported (Figure 1 in green), but any values <0.05 ng/mL are not reported (Figure 1 in red), even though they come from the exact same distribution! The samples <LOQ are given the designation “BQL” (below quantitation limit), and the actual values will never be reported no matter how much one pleads, upon threat of receiving a Form 483 from the FDA. In effect, the analytical chemist censors all data <LOQ, even though an acceptable LOQ accuracy is <b><i>plus or minus</i></b> 15%–20% of the nominal concentration.</p><p>It gets worse when we pharmacokineticists receive the data. If we are performing a non-compartmental analysis, we change all the BQLs to zeros as we are instructed,<span><sup>2</sup></span> even though we know that there are actual values lurking there somewhere. We know this because “real” zero concentrations are reported as such. But we pretend that the BQLs are zero anyway because the guidance or white paper tells us to do so. If we are performing a population PK analysis, and there are enough BQLs to concern us (typically 10% or more of the total number of samples<span><sup>3</sup></span>), then we use a sophisticated imputation method for those BQLs. We sleep well at night knowing that our data are safe from any sordid contamination by BQL samples, and we (secretly) pat ourselves on the back for knowing ","PeriodicalId":10495,"journal":{"name":"Clinical Pharmacology in Drug Development","volume":"14 12","pages":"900-902"},"PeriodicalIF":1.8,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://accp1.onlinelibrary.wiley.com/doi/epdf/10.1002/cpdd.70000","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145652416","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Lucy Lee, Amy-Lee Richards, Nageswara Reddy, Brian Beers, Lee Golden, Ronald Kong
Votoplam is a novel, orally bioavailable, small molecule HTT gene splicing modifier that is being developed for the treatment of Huntington's disease. This was a single dose, open-label, two-period, crossover food effect study that evaluated the effect of high- and low-fat meals on 20 mg votoplam in healthy participants. There was a washout of 21 days between the two periods. Twenty-six participants completed the study. There were minimal changes in the bioavailability and pharmacokinetics of votoplam following administration of a single dose of 20 mg votoplam when taken with low-fat and high-fat meals relative to fasted condition. The mean Cmax, AUC0-last, and AUC0-inf for votoplam following administration of a single dose of 20 mg votoplam were 1.4-fold, 1.2-fold, and 1.1-fold higher, respectively, in high-fat fed conditions, and were 1.3-fold, 1.1-fold, and 1.1-fold higher, respectively, in the low-fat fed conditions, when compared to fasted conditions. There were no relevant safety findings in any of the treatment groups. Votoplam can be administered with or without food in patients.
{"title":"Effects of High- and Low-Fat Meals on the Bioavailability and Pharmacokinetics of Votoplam, a HTT Gene Splicing Modifier.","authors":"Lucy Lee, Amy-Lee Richards, Nageswara Reddy, Brian Beers, Lee Golden, Ronald Kong","doi":"10.1002/cpdd.1626","DOIUrl":"https://doi.org/10.1002/cpdd.1626","url":null,"abstract":"<p><p>Votoplam is a novel, orally bioavailable, small molecule HTT gene splicing modifier that is being developed for the treatment of Huntington's disease. This was a single dose, open-label, two-period, crossover food effect study that evaluated the effect of high- and low-fat meals on 20 mg votoplam in healthy participants. There was a washout of 21 days between the two periods. Twenty-six participants completed the study. There were minimal changes in the bioavailability and pharmacokinetics of votoplam following administration of a single dose of 20 mg votoplam when taken with low-fat and high-fat meals relative to fasted condition. The mean C<sub>max</sub>, AUC<sub>0-last</sub>, and AUC<sub>0-inf</sub> for votoplam following administration of a single dose of 20 mg votoplam were 1.4-fold, 1.2-fold, and 1.1-fold higher, respectively, in high-fat fed conditions, and were 1.3-fold, 1.1-fold, and 1.1-fold higher, respectively, in the low-fat fed conditions, when compared to fasted conditions. There were no relevant safety findings in any of the treatment groups. Votoplam can be administered with or without food in patients.</p>","PeriodicalId":10495,"journal":{"name":"Clinical Pharmacology in Drug Development","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145647661","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}