首页 > 最新文献

Clinical Pharmacology & Therapeutics最新文献

英文 中文
Evolving Research and Development Landscape for Rare Diseases: Growing Concerns Over Orphan Drug Lag in Japan
IF 6.3 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-04 DOI: 10.1002/cpt.3553
Kazuaki Enya, Itsuki Kageyama, Yoshiyuki Kobayashi, Yeongjoo Lim, Shintaro Sengoku, Kota Kodama

Patients with rare diseases worldwide face substantial unmet therapeutic needs. In Japan, drug lag—delays in drug approval compared to other countries—has resurfaced as a pressing public health issue. This study analyzes orphan drugs (ODs) approved in the United States (US) from 2005 to 2021, examining OD lag trends, and research and development (R&D) models to streamline OD development in Japan. Despite increased OD approval in the United States since 2018, the number of unapproved ODs in Japan has substantially increased. Although OD lag decreased, it has resurged since 2017. This is largely due to changes in the R&D strategies of pharmaceutical companies, which are driven by the growing presence of US- and Europe-based small- to mid-sized enterprises (SMEs) and the evolving industry landscape. Large foreign pharmaceutical companies have shifted toward a global development strategy for Japan, aiming for more efficient development and competitive advantage. This has been propelled by a move toward in-licensing earlier-stage drug candidates with global exclusivity from SMEs. Japanese pharmaceutical companies have focused on in-licensing late-stage drug candidates for the Japanese market from SMEs without a business presence in Japan, which have not been developed locally, thereby employing a bridging strategy for Japan. With the increase in ODs developed in the United States by these SMEs, this practice has substantially exacerbated the OD lag. As these SMEs are unlikely to enter the Japanese market, it is crucial for Japanese pharmaceutical companies to proactively pursue earlier, more proactive global partnerships with SMEs.

全世界的罕见病患者都面临着大量未得到满足的治疗需求。在日本,药物滞后--与其他国家相比药物审批的延迟--已重新成为一个紧迫的公共卫生问题。本研究分析了 2005 年至 2021 年美国批准的孤儿药 (OD),研究了 OD 滞后趋势以及日本简化 OD 开发的研发 (R&D) 模式。尽管自2018年以来美国批准的OD有所增加,但日本未获批准的OD数量却大幅增加。虽然OD滞后现象有所减少,但自2017年以来又重新抬头。这主要是由于美国和欧洲的中小型企业(SMEs)越来越多,以及行业格局的不断变化推动了制药公司研发战略的变化。大型外国制药公司已转向日本的全球发展战略,旨在提高开发效率和竞争优势。这主要得益于向中小型企业内购具有全球独家代理权的早期候选药物。日本制药公司侧重于从在日本没有业务的中小型企业获得面向日本市场的后期候选药物的内部许可,这些候选药物尚未在日本本土开发,因此在日本采用了一种桥梁战略。随着这些中小企业在美国开发的 OD 的增加,这种做法大大加剧了 OD 的滞后性。由于这些中小型企业不太可能进入日本市场,因此日本制药公司必须积极寻求与中小型企业建立更早、更主动的全球伙伴关系。
{"title":"Evolving Research and Development Landscape for Rare Diseases: Growing Concerns Over Orphan Drug Lag in Japan","authors":"Kazuaki Enya,&nbsp;Itsuki Kageyama,&nbsp;Yoshiyuki Kobayashi,&nbsp;Yeongjoo Lim,&nbsp;Shintaro Sengoku,&nbsp;Kota Kodama","doi":"10.1002/cpt.3553","DOIUrl":"10.1002/cpt.3553","url":null,"abstract":"<p>Patients with rare diseases worldwide face substantial unmet therapeutic needs. In Japan, drug lag—delays in drug approval compared to other countries—has resurfaced as a pressing public health issue. This study analyzes orphan drugs (ODs) approved in the United States (US) from 2005 to 2021, examining OD lag trends, and research and development (R&amp;D) models to streamline OD development in Japan. Despite increased OD approval in the United States since 2018, the number of unapproved ODs in Japan has substantially increased. Although OD lag decreased, it has resurged since 2017. This is largely due to changes in the R&amp;D strategies of pharmaceutical companies, which are driven by the growing presence of US- and Europe-based small- to mid-sized enterprises (SMEs) and the evolving industry landscape. Large foreign pharmaceutical companies have shifted toward a global development strategy for Japan, aiming for more efficient development and competitive advantage. This has been propelled by a move toward in-licensing earlier-stage drug candidates with global exclusivity from SMEs. Japanese pharmaceutical companies have focused on in-licensing late-stage drug candidates for the Japanese market from SMEs without a business presence in Japan, which have not been developed locally, thereby employing a bridging strategy for Japan. With the increase in ODs developed in the United States by these SMEs, this practice has substantially exacerbated the OD lag. As these SMEs are unlikely to enter the Japanese market, it is crucial for Japanese pharmaceutical companies to proactively pursue earlier, more proactive global partnerships with SMEs.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":"117 5","pages":"1325-1337"},"PeriodicalIF":6.3,"publicationDate":"2025-02-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cpt.3553","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143187863","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CB1 Receptor Antagonist Selonabant (ANEB-001) Blocks Acute THC Effects in Healthy Volunteers: A Phase II Randomized Controlled Trial
IF 6.3 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-03 DOI: 10.1002/cpt.3581
Andriy A. Gorbenko, Jules A. A. C. Heuberger, Maria Juachon, Erica Klaassen, Michael Tagen, Joseph F. Lawler, Daniel Schneeberger, Kenneth C. Cundy, Linda E. Klumpers, Geert J. Groeneveld

Emergency department visits due to cannabinoid-induced toxicity, including acute cannabinoid intoxication (ACI) have increased worldwide as more states have liberalized cannabis policy. ACI symptoms include anxiety, panic attacks, tachycardia, and psychosis, primarily mediated through cannabinoid type 1 receptor (CB1) agonism by Δ9-tetrahydrocannabinol (THC). This phase II randomized, double-blind, placebo-controlled study assessed the potential of CB1 receptor antagonist selonabant (ANEB-001) to block THC-induced effects in healthy adults. In Part A of the study, 10.5 mg of THC was coadministered with 50 mg (N = 20) or 100 mg (N = 20) selonabant, or matching placebo (N = 20). In Part B, 21-mg THC was coadministered with 30 mg (N = 9) or 10 mg (N = 7) selonabant, or matching placebo (N = 9). THC-related effects were assessed using visual analogue scales (VAS) for feeling high and alertness, objective measures of postural stability, and heart rate and analyzed using a mixed effects model. Selonabant significantly reduced VAS “Feeling High” (up to −82.8% (95% CI: −91.0%, −67.2%, P < 0.0001) at 30-mg selonabant) and increased VAS “Alertness” (up to 10.8 mm (95% CI: 4.7, 16.8 mm, P = 0.001) at 30-mg selonabant) vs. placebo. Selonabant 10 and 30 mg significantly reduced body sway (up to −30.6% (95% CI: −44.1%, −13.9%, P = 0.002) at 30 mg selonabant) vs. placebo. Effects on heart rate were not significant. Selonabant was generally safe and no clinically meaningful changes in mood occurred. Nausea and vomiting occurred more frequently at high selonabant doses; 10-mg selonabant was both well tolerated and efficacious. Present results support further development of selonabant for emergency treatment of ACI.

{"title":"CB1 Receptor Antagonist Selonabant (ANEB-001) Blocks Acute THC Effects in Healthy Volunteers: A Phase II Randomized Controlled Trial","authors":"Andriy A. Gorbenko,&nbsp;Jules A. A. C. Heuberger,&nbsp;Maria Juachon,&nbsp;Erica Klaassen,&nbsp;Michael Tagen,&nbsp;Joseph F. Lawler,&nbsp;Daniel Schneeberger,&nbsp;Kenneth C. Cundy,&nbsp;Linda E. Klumpers,&nbsp;Geert J. Groeneveld","doi":"10.1002/cpt.3581","DOIUrl":"10.1002/cpt.3581","url":null,"abstract":"<p>Emergency department visits due to cannabinoid-induced toxicity, including acute cannabinoid intoxication (ACI) have increased worldwide as more states have liberalized cannabis policy. ACI symptoms include anxiety, panic attacks, tachycardia, and psychosis, primarily mediated through cannabinoid type 1 receptor (CB<sub>1</sub>) agonism by Δ<sup>9</sup>-tetrahydrocannabinol (THC). This phase II randomized, double-blind, placebo-controlled study assessed the potential of CB<sub>1</sub> receptor antagonist selonabant (ANEB-001) to block THC-induced effects in healthy adults. In Part A of the study, 10.5 mg of THC was coadministered with 50 mg (<i>N</i> = 20) or 100 mg (<i>N</i> = 20) selonabant, or matching placebo (<i>N</i> = 20). In Part B, 21-mg THC was coadministered with 30 mg (<i>N</i> = 9) or 10 mg (<i>N</i> = 7) selonabant, or matching placebo (<i>N</i> = 9). THC-related effects were assessed using visual analogue scales (VAS) for feeling high and alertness, objective measures of postural stability, and heart rate and analyzed using a mixed effects model. Selonabant significantly reduced VAS “Feeling High” (up to −82.8% (95% CI: −91.0%, −67.2%, <i>P</i> &lt; 0.0001) at 30-mg selonabant) and increased VAS “Alertness” (up to 10.8 mm (95% CI: 4.7, 16.8 mm, <i>P</i> = 0.001) at 30-mg selonabant) vs. placebo. Selonabant 10 and 30 mg significantly reduced body sway (up to −30.6% (95% CI: −44.1%, −13.9%, <i>P</i> = 0.002) at 30 mg selonabant) vs. placebo. Effects on heart rate were not significant. Selonabant was generally safe and no clinically meaningful changes in mood occurred. Nausea and vomiting occurred more frequently at high selonabant doses; 10-mg selonabant was both well tolerated and efficacious. Present results support further development of selonabant for emergency treatment of ACI.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":"117 5","pages":"1427-1436"},"PeriodicalIF":6.3,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cpt.3581","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143078089","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Machine Learning for Prediction of Drug Concentrations: Application and Challenges
IF 6.3 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-03 DOI: 10.1002/cpt.3577
Shuqi Huang, Qihan Xu, Guoping Yang, Junjie Ding, Qi Pei

With the advancements in algorithms and increased accessibility of multi-source data, machine learning in pharmacokinetics is gaining interest. This review summarizes studies on machine learning-based pharmacokinetics analysis up to September 2024, identified from the PubMed and IEEE Xplore databases. The main focus of this review is on the use of machine learning in predicting drug concentration. This review provides a comprehensive summary of the advances in the machine learning algorithms for pharmacokinetics analysis. Specifically, we describe the common practices in data preprocessing, the application scenarios of various algorithms, and the critical challenges that require attention. Most machine learning models show comparable performance to those of population pharmacokinetics models. Tree-based algorithms and neural networks have the most applications. Furthermore, the use of ensemble modeling techniques can improve the accuracy of these models' predictions of drug concentrations, especially the ensembles of machine learning and pharmacometrics.

{"title":"Machine Learning for Prediction of Drug Concentrations: Application and Challenges","authors":"Shuqi Huang,&nbsp;Qihan Xu,&nbsp;Guoping Yang,&nbsp;Junjie Ding,&nbsp;Qi Pei","doi":"10.1002/cpt.3577","DOIUrl":"10.1002/cpt.3577","url":null,"abstract":"<p>With the advancements in algorithms and increased accessibility of multi-source data, machine learning in pharmacokinetics is gaining interest. This review summarizes studies on machine learning-based pharmacokinetics analysis up to September 2024, identified from the PubMed and IEEE Xplore databases. The main focus of this review is on the use of machine learning in predicting drug concentration. This review provides a comprehensive summary of the advances in the machine learning algorithms for pharmacokinetics analysis. Specifically, we describe the common practices in data preprocessing, the application scenarios of various algorithms, and the critical challenges that require attention. Most machine learning models show comparable performance to those of population pharmacokinetics models. Tree-based algorithms and neural networks have the most applications. Furthermore, the use of ensemble modeling techniques can improve the accuracy of these models' predictions of drug concentrations, especially the ensembles of machine learning and pharmacometrics.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":"117 5","pages":"1236-1247"},"PeriodicalIF":6.3,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143121864","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A Guide for Implementing DPYD Genotyping for Systemic Fluoropyrimidines into Clinical Practice
IF 6.3 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-01-31 DOI: 10.1002/cpt.3567
Teresa T. Ho, D. Max Smith, Christina L. Aquilante, Emily J. Cicali, Nihal El Rouby, Daniel L. Hertz, Iman Imanirad, Jai N. Patel, Stuart A. Scott, Sandra M. Swain, Sony Tuteja, J. Kevin Hicks, the Pharmacogenomics Global Research Network Publication Committee

The safety of systemic fluoropyrimidines (e.g., 5-fluorouracil, capecitabine) is impacted by germline genetic variants in DPYD, which encodes the dihydropyrimidine dehydrogenase (DPD) enzyme that functions as the rate-limiting step in the catabolism of this drug class. Genetic testing to identify those with DPD deficiency can help mitigate the risk of severe and life-threatening fluoropyrimidine-induced toxicities. Globally, the integration of DPYD genetic testing into patient care has varied greatly, ranging from being required as the standard of care in some countries to limited clinical use in others. Thus, implementation strategies have evolved differently across health systems and countries. The primary objective of this tutorial is to provide practical considerations and best practice recommendations for the implementation of DPYD-guided systemic fluoropyrimidine dosing. We adapted the Exploration, Preparation, Implementation, and Sustainment (EPIS) framework to cover topics including the clinical evidence supporting DPYD genotyping to guide fluoropyrimidine therapy, regulatory guidance for DPYD genotyping, key stakeholder engagement, logistics for DPYD genotyping, development of point-of-care clinical decision support tools, and considerations for the creation of sustainable and scalable DPYD genotype-integrated workflows. This guide also describes approaches to counseling patients about DPYD testing and result disclosure, along with examples of patient and provider educational resources. Together, DPYD testing and clinical practice integration aim to promote safe prescribing of fluoropyrimidine therapy and decrease the risk of severe and life-threatening fluoropyrimidine toxicities.

{"title":"A Guide for Implementing DPYD Genotyping for Systemic Fluoropyrimidines into Clinical Practice","authors":"Teresa T. Ho,&nbsp;D. Max Smith,&nbsp;Christina L. Aquilante,&nbsp;Emily J. Cicali,&nbsp;Nihal El Rouby,&nbsp;Daniel L. Hertz,&nbsp;Iman Imanirad,&nbsp;Jai N. Patel,&nbsp;Stuart A. Scott,&nbsp;Sandra M. Swain,&nbsp;Sony Tuteja,&nbsp;J. Kevin Hicks,&nbsp;the Pharmacogenomics Global Research Network Publication Committee","doi":"10.1002/cpt.3567","DOIUrl":"10.1002/cpt.3567","url":null,"abstract":"<p>The safety of systemic fluoropyrimidines (e.g., 5-fluorouracil, capecitabine) is impacted by germline genetic variants in <i>DPYD,</i> which encodes the dihydropyrimidine dehydrogenase (DPD) enzyme that functions as the rate-limiting step in the catabolism of this drug class. Genetic testing to identify those with DPD deficiency can help mitigate the risk of severe and life-threatening fluoropyrimidine-induced toxicities. Globally, the integration of <i>DPYD</i> genetic testing into patient care has varied greatly, ranging from being required as the standard of care in some countries to limited clinical use in others. Thus, implementation strategies have evolved differently across health systems and countries. The primary objective of this tutorial is to provide practical considerations and best practice recommendations for the implementation of <i>DPYD</i>-guided systemic fluoropyrimidine dosing. We adapted the Exploration, Preparation, Implementation, and Sustainment (EPIS) framework to cover topics including the clinical evidence supporting <i>DPYD</i> genotyping to guide fluoropyrimidine therapy, regulatory guidance for <i>DPYD</i> genotyping, key stakeholder engagement, logistics for <i>DPYD</i> genotyping, development of point-of-care clinical decision support tools, and considerations for the creation of sustainable and scalable <i>DPYD</i> genotype-integrated workflows. This guide also describes approaches to counseling patients about <i>DPYD</i> testing and result disclosure, along with examples of patient and provider educational resources. Together, <i>DPYD</i> testing and clinical practice integration aim to promote safe prescribing of fluoropyrimidine therapy and decrease the risk of severe and life-threatening fluoropyrimidine toxicities.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":"117 5","pages":"1194-1208"},"PeriodicalIF":6.3,"publicationDate":"2025-01-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cpt.3567","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143062845","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Intracellular Penetration of Atazanavir, Ritonavir and Dolutegravir With Concomitant Rifampicin: A Dose Escalation Study
IF 6.3 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-01-31 DOI: 10.1002/cpt.3572
Amedeo De Nicolò, Alice Palermiti, Henry Mugerwa, Shamim Nakabuye, Josephine Namusanje, Josephine Kobusingye, Denis Odoch, Mohammed Lamorde, Allan Kengo, Paolo Denti, Kamunkhwala Gausi, Gary Maartens, Helen McIlleron, Lubbe Wiesner, Saye Khoo, Catriona Waitt, Antonio D'Avolio

Ritonavir-boosted atazanavir is a victim of drug–drug interaction with rifampicin, a key component of antitubercular treatment. In a recent dose escalation clinical trial, we showed that increasing atazanavir/ritonavir to 300/100 mg b.i.d. compensates for reduced drug exposure in plasma due to rifampicin, but the intracellular effects remained unexplored. This sub-study investigated the intracellular penetration of atazanavir/ritonavir and dolutegravir into peripheral blood mononuclear cells (PBMC). Twenty-six healthy volunteers living with HIV, virologically suppressed, and taking atazanavir/ritonavir containing regimens were enrolled. The trial consisted of four sequential periods: PK1, participants were on atazanavir/ritonavir 300/100 mg q.d.; at PK2, rifampicin 600 mg q.d. and dolutegravir 50 mg b.i.d. were added (2 weeks); at PK3, atazanavir/ritonavir dose was increased to 300/100 mg b.i.d. (1 week); at PK4, rifampicin dose was doubled (1 week). Atazanavir, ritonavir, and dolutegravir were quantified in plasma and PBMC using LC–MS/MS methods to evaluate steady-state concentrations at the end of each period. Atazanavir/ritonavir dose escalation successfully restored intracellular concentrations comparable to those observed without rifampicin, with a geometric mean ratio of 0.99 (CI90 0.72–1.41) for atazanavir at PK3 compared with PK1. The intracellular concentration of dolutegravir increased significantly with atazanavir/ritonavir dose escalation, similar to plasma. Finally, further, increasing the rifampicin dose did not show an additional impact on atazanavir/ritonavir concentrations in PBMC. The study confirms that increasing the ATV/r dose can be an effective strategy for compensating rifampicin effects even at the intracellular level, supporting its use in clinical settings.

利托那韦增强型阿扎那韦是与利福平(抗结核治疗的关键成分)发生药物相互作用的受害者。在最近的一项剂量递增临床试验中,我们发现将阿扎那韦/利托那韦的剂量提高到 300/100 毫克(b.i.d.)可以弥补利福平导致的血浆中药物暴露量的减少,但细胞内效应仍有待探索。本子研究调查了阿扎那韦/利托那韦和多鲁曲韦在外周血单核细胞(PBMC)中的细胞内渗透。试验招募了 26 名感染艾滋病毒、病毒学抑制并服用含阿扎那韦/利托那韦治疗方案的健康志愿者。试验分为四个阶段:PK1 期,参与者服用阿扎那韦/利托那韦 300/100 mg q.d.;PK2 期,加入利福平 600 mg q.d. 和多罗替拉韦 50 mg b.i.d.(2 周);PK3 期,阿扎那韦/利托那韦剂量增至 300/100 mg b.i.d.(1 周);PK4 期,利福平剂量加倍(1 周)。采用 LC-MS/MS 方法对血浆和 PBMC 中的阿扎那韦、利托那韦和多罗替拉韦进行定量,以评估每个阶段结束时的稳态浓度。阿扎那韦/利托那韦剂量递增成功地恢复了细胞内浓度,与不使用利福平时观察到的浓度相当,与 PK1 相比,PK3 时阿扎那韦的几何平均比值为 0.99(CI90 为 0.72-1.41)。随着阿扎那韦/利托那韦剂量的增加,多鲁曲韦的细胞内浓度显著增加,与血浆相似。最后,增加利福平剂量对阿扎那韦/利托那韦在白细胞介质中的浓度没有额外影响。这项研究证实,即使在细胞内水平,增加阿托伐韦/利托那韦的剂量也是补偿利福平效应的有效策略,支持在临床中使用。
{"title":"Intracellular Penetration of Atazanavir, Ritonavir and Dolutegravir With Concomitant Rifampicin: A Dose Escalation Study","authors":"Amedeo De Nicolò,&nbsp;Alice Palermiti,&nbsp;Henry Mugerwa,&nbsp;Shamim Nakabuye,&nbsp;Josephine Namusanje,&nbsp;Josephine Kobusingye,&nbsp;Denis Odoch,&nbsp;Mohammed Lamorde,&nbsp;Allan Kengo,&nbsp;Paolo Denti,&nbsp;Kamunkhwala Gausi,&nbsp;Gary Maartens,&nbsp;Helen McIlleron,&nbsp;Lubbe Wiesner,&nbsp;Saye Khoo,&nbsp;Catriona Waitt,&nbsp;Antonio D'Avolio","doi":"10.1002/cpt.3572","DOIUrl":"10.1002/cpt.3572","url":null,"abstract":"<p>Ritonavir-boosted atazanavir is a victim of drug–drug interaction with rifampicin, a key component of antitubercular treatment. In a recent dose escalation clinical trial, we showed that increasing atazanavir/ritonavir to 300/100 mg b.i.d. compensates for reduced drug exposure in plasma due to rifampicin, but the intracellular effects remained unexplored. This sub-study investigated the intracellular penetration of atazanavir/ritonavir and dolutegravir into peripheral blood mononuclear cells (PBMC). Twenty-six healthy volunteers living with HIV, virologically suppressed, and taking atazanavir/ritonavir containing regimens were enrolled. The trial consisted of four sequential periods: PK1, participants were on atazanavir/ritonavir 300/100 mg q.d.; at PK2, rifampicin 600 mg q.d. and dolutegravir 50 mg b.i.d. were added (2 weeks); at PK3, atazanavir/ritonavir dose was increased to 300/100 mg b.i.d. (1 week); at PK4, rifampicin dose was doubled (1 week). Atazanavir, ritonavir, and dolutegravir were quantified in plasma and PBMC using LC–MS/MS methods to evaluate steady-state concentrations at the end of each period. Atazanavir/ritonavir dose escalation successfully restored intracellular concentrations comparable to those observed without rifampicin, with a geometric mean ratio of 0.99 (CI<sub>90</sub> 0.72–1.41) for atazanavir at PK3 compared with PK1. The intracellular concentration of dolutegravir increased significantly with atazanavir/ritonavir dose escalation, similar to plasma. Finally, further, increasing the rifampicin dose did not show an additional impact on atazanavir/ritonavir concentrations in PBMC. The study confirms that increasing the ATV/r dose can be an effective strategy for compensating rifampicin effects even at the intracellular level, supporting its use in clinical settings.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":"117 5","pages":"1393-1402"},"PeriodicalIF":6.3,"publicationDate":"2025-01-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cpt.3572","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143072953","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Clinical Evaluation of Drug–Drug Interactions with Obeldesivir, an Orally Administered Antiviral Agent
IF 6.3 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-01-31 DOI: 10.1002/cpt.3575
Chi-Chi Peng, Rita Humeniuk, Anuja Raut, Anna Kwan, Lily Mak, Caitlin Stacom, Deqing Xiao, Shuguang Chen, Santosh Davies, Sharline Madera, Yiannis Koullias, Amos Lichtman, Joe Llewellyn, Elham Amini, Helen Winter, Luzelena Caro

Obeldesivir is an oral nucleoside analog prodrug inhibitor of SARS-CoV-2 RNA-dependent RNA polymerase and other viral polymerases. Here, two Phase I studies evaluated potential drug–drug interactions between obeldesivir and substrates or inhibitors of cytochrome P450 and drug transporters in healthy participants. When obeldesivir was tested as a precipitant, pharmacokinetic parameter point estimates for midazolam (CYP3A4 inhibition/induction), caffeine (CYP1A2 inhibition), and metformin (organic cation transporter 1 inhibition) exposures were within 80–125% no-effect bounds representing the interval within which a systemic exposure change does not warrant clinical action based on EMA/FDA guidance. Dabigatran (P-glycoprotein substrate) and pitavastatin (organic anion transporting polypeptide 1B1/1B3 substrate) exposures decreased by approximately 25% and 30%, respectively, with obeldesivir coadministration; these were considered not clinically relevant, as these exposure changes are not associated with dose changes or precautions in the US prescribing information for these drugs. When obeldesivir was evaluated as an object, exposures of GS-441524, the parent nucleoside metabolite of obeldesivir, were within the 80–125% no-effect bounds for ritonavir (P-glycoprotein inhibition) and cyclosporin A (breast cancer resistance protein inhibition) coadministration [Correction added on 7 February 2025, after first online publication: The word monophosphate has been removed in this version.]. Famotidine (gastric acid suppression) coadministration decreased GS-441524 exposure by approximately 26%; this was within the range of exposures observed in previous Phase III studies and was considered not clinically relevant. Obeldesivir was well tolerated, and adverse events were mild to moderate. These findings indicate that obeldesivir has low potential for drug–drug interactions. Obeldesivir remains a promising treatment against a broad spectrum of viruses given its antiviral activity and favorable safety profile.

{"title":"Clinical Evaluation of Drug–Drug Interactions with Obeldesivir, an Orally Administered Antiviral Agent","authors":"Chi-Chi Peng,&nbsp;Rita Humeniuk,&nbsp;Anuja Raut,&nbsp;Anna Kwan,&nbsp;Lily Mak,&nbsp;Caitlin Stacom,&nbsp;Deqing Xiao,&nbsp;Shuguang Chen,&nbsp;Santosh Davies,&nbsp;Sharline Madera,&nbsp;Yiannis Koullias,&nbsp;Amos Lichtman,&nbsp;Joe Llewellyn,&nbsp;Elham Amini,&nbsp;Helen Winter,&nbsp;Luzelena Caro","doi":"10.1002/cpt.3575","DOIUrl":"10.1002/cpt.3575","url":null,"abstract":"<p>Obeldesivir is an oral nucleoside analog prodrug inhibitor of SARS-CoV-2 RNA-dependent RNA polymerase and other viral polymerases. Here, two Phase I studies evaluated potential drug–drug interactions between obeldesivir and substrates or inhibitors of cytochrome P450 and drug transporters in healthy participants. When obeldesivir was tested as a precipitant, pharmacokinetic parameter point estimates for midazolam (CYP3A4 inhibition/induction), caffeine (CYP1A2 inhibition), and metformin (organic cation transporter 1 inhibition) exposures were within 80–125% no-effect bounds representing the interval within which a systemic exposure change does not warrant clinical action based on EMA/FDA guidance. Dabigatran (P-glycoprotein substrate) and pitavastatin (organic anion transporting polypeptide 1B1/1B3 substrate) exposures decreased by approximately 25% and 30%, respectively, with obeldesivir coadministration; these were considered not clinically relevant, as these exposure changes are not associated with dose changes or precautions in the US prescribing information for these drugs. When obeldesivir was evaluated as an object, exposures of GS-441524, the parent nucleoside metabolite of obeldesivir, were within the 80–125% no-effect bounds for ritonavir (P-glycoprotein inhibition) and cyclosporin A (breast cancer resistance protein inhibition) coadministration [Correction added on 7 February 2025, after first online publication: The word monophosphate has been removed in this version.]. Famotidine (gastric acid suppression) coadministration decreased GS-441524 exposure by approximately 26%; this was within the range of exposures observed in previous Phase III studies and was considered not clinically relevant. Obeldesivir was well tolerated, and adverse events were mild to moderate. These findings indicate that obeldesivir has low potential for drug–drug interactions. Obeldesivir remains a promising treatment against a broad spectrum of viruses given its antiviral activity and favorable safety profile.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":"117 5","pages":"1403-1412"},"PeriodicalIF":6.3,"publicationDate":"2025-01-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143062847","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
IQ Survey Results on Current Industry Practices-Part 1: Immunogenicity Risk Assessment.
IF 6.3 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-01-28 DOI: 10.1002/cpt.3568
Christine Grimaldi, Susan Richards, Daniel Baltrukonis, Shelley Sims Belouski, Kelly Coble, Sanjay L Dholakiya, Joanna Grudzinska-Goebel, Gerry Kolaitis, Jocelyn H Leu, Linlin Luo, Stephen Lowe, Tao Niu, Henrik Toft-Hansen, Jianning Yang, Benjamin Wu

An immunogenicity risk assessment (IRA) is a relatively new expectation of health authorities that is increasingly incorporated into the drug development process across the pharmaceutical/biotech industry. The guiding principle for an IRA includes a comprehensive evaluation of product- and patient-related factors that may influence the immunogenic potential of a biotherapeutic drug and a potential action plan. The Immunogenicity Working Group from the IQ Consortium (Clinical Pharmacology Leadership Group) has conducted a survey to understand the current practices for conducting IRAs and relevant aspects of bioanalysis. Survey results were provided by 19 IQ member companies participating in the Clinical Pharmacology Leadership Group (CPLG) and the Translational and ADME Sciences Leadership Group (TALG). Nearly all the respondents reported experience with monoclonal antibodies (mAb), with 10 other drug modalities including bioengineered protein therapeutics such as fusion and multi-domain proteins, peptides, oligonucleotides as well as gene and cell therapies. The survey results demonstrate that most companies have a defined IRA process, and there was a common understanding that the IRA may need to be revised as more information becomes available or the drug development strategy changes. Some differences found across the respondents are related to the time frame for implementation of IRA document, the types of preclinical data and computational methods used to assess risk, and how the IRA informs clinical plans and documentation practices. These results highlight that while there have been widespread insights gained with performing IRA for mAbs, more experience is needed to perform IRAs for the novel modalities.

{"title":"IQ Survey Results on Current Industry Practices-Part 1: Immunogenicity Risk Assessment.","authors":"Christine Grimaldi, Susan Richards, Daniel Baltrukonis, Shelley Sims Belouski, Kelly Coble, Sanjay L Dholakiya, Joanna Grudzinska-Goebel, Gerry Kolaitis, Jocelyn H Leu, Linlin Luo, Stephen Lowe, Tao Niu, Henrik Toft-Hansen, Jianning Yang, Benjamin Wu","doi":"10.1002/cpt.3568","DOIUrl":"https://doi.org/10.1002/cpt.3568","url":null,"abstract":"<p><p>An immunogenicity risk assessment (IRA) is a relatively new expectation of health authorities that is increasingly incorporated into the drug development process across the pharmaceutical/biotech industry. The guiding principle for an IRA includes a comprehensive evaluation of product- and patient-related factors that may influence the immunogenic potential of a biotherapeutic drug and a potential action plan. The Immunogenicity Working Group from the IQ Consortium (Clinical Pharmacology Leadership Group) has conducted a survey to understand the current practices for conducting IRAs and relevant aspects of bioanalysis. Survey results were provided by 19 IQ member companies participating in the Clinical Pharmacology Leadership Group (CPLG) and the Translational and ADME Sciences Leadership Group (TALG). Nearly all the respondents reported experience with monoclonal antibodies (mAb), with 10 other drug modalities including bioengineered protein therapeutics such as fusion and multi-domain proteins, peptides, oligonucleotides as well as gene and cell therapies. The survey results demonstrate that most companies have a defined IRA process, and there was a common understanding that the IRA may need to be revised as more information becomes available or the drug development strategy changes. Some differences found across the respondents are related to the time frame for implementation of IRA document, the types of preclinical data and computational methods used to assess risk, and how the IRA informs clinical plans and documentation practices. These results highlight that while there have been widespread insights gained with performing IRA for mAbs, more experience is needed to perform IRAs for the novel modalities.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":" ","pages":""},"PeriodicalIF":6.3,"publicationDate":"2025-01-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143057415","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Integrated Longitudinal Population Dose-Hemoglobin Response of Daprodustat Following Dose Titration in Patients With Anemia in Chronic Kidney Disease
IF 6.3 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-01-28 DOI: 10.1002/cpt.3544
Kelly M. Mahar, Martijn van Noort, Paul van den Berg, Shuying Yang, Sandra A. G. Visser, Teun M. Post

Daprodustat, a novel oral hypoxia-inducible factor prolyl hydroxylase inhibitor is approved in the United States for the treatment of anemia due to chronic kidney disease (CKD) in adults receiving dialysis for at least 4 months. Pharmacodynamic dose-hemoglobin (Dose-Hgb) models were developed as daprodustat progressed through development. To support global phase III development, a dose-titration algorithm, guided by simulations from the initial Dose-Hgb model based on phase II clinical data, was implemented. This work was to update and re-calibrate this model to support the dose titration algorithm. Data from five pivotal phase III studies in CKD patients with anemia treated with daprodustat once daily (q.d.) and/or three times a week (t.i.w.) using a titration dosing schedule were included. The data comprised 2,770 CKD patients with anemia providing 53,535 Hgb observations over a period of 6 months up to 4 years. This final Dose-Hgb model consisted of a precursor cell compartment and 12 transit compartments to describe the red blood cell (RBC) lifespan. Treatment increased the precursor cell production rate (Kin) by a power of allometrically scaled dose. Disease progression, as an exponential decline of Hgb production rate over time, varied with dialysis status. The dose-titration algorithm resulted in comparable response for t.i.w. dosing relative to q.d. dosing. Titration-based visual predictive checks for Hgb target criteria for the analysis dataset and the prediction dataset showed that the model adequately predicted the observed data. This re-calibrated Dose-Hgb model will provide further support for the individualized dosing strategy in CKD patients with anemia treated with daprodustat.

{"title":"Integrated Longitudinal Population Dose-Hemoglobin Response of Daprodustat Following Dose Titration in Patients With Anemia in Chronic Kidney Disease","authors":"Kelly M. Mahar,&nbsp;Martijn van Noort,&nbsp;Paul van den Berg,&nbsp;Shuying Yang,&nbsp;Sandra A. G. Visser,&nbsp;Teun M. Post","doi":"10.1002/cpt.3544","DOIUrl":"10.1002/cpt.3544","url":null,"abstract":"<p>Daprodustat, a novel oral hypoxia-inducible factor prolyl hydroxylase inhibitor is approved in the United States for the treatment of anemia due to chronic kidney disease (CKD) in adults receiving dialysis for at least 4 months. Pharmacodynamic dose-hemoglobin (Dose-Hgb) models were developed as daprodustat progressed through development. To support global phase III development, a dose-titration algorithm, guided by simulations from the initial Dose-Hgb model based on phase II clinical data, was implemented. This work was to update and re-calibrate this model to support the dose titration algorithm. Data from five pivotal phase III studies in CKD patients with anemia treated with daprodustat once daily (q.d.) and/or three times a week (t.i.w.) using a titration dosing schedule were included. The data comprised 2,770 CKD patients with anemia providing 53,535 Hgb observations over a period of 6 months up to 4 years. This final Dose-Hgb model consisted of a precursor cell compartment and 12 transit compartments to describe the red blood cell (RBC) lifespan. Treatment increased the precursor cell production rate (<i>K</i><sub>in</sub>) by a power of allometrically scaled dose. Disease progression, as an exponential decline of Hgb production rate over time, varied with dialysis status. The dose-titration algorithm resulted in comparable response for t.i.w. dosing relative to q.d. dosing. Titration-based visual predictive checks for Hgb target criteria for the analysis dataset and the prediction dataset showed that the model adequately predicted the observed data. This re-calibrated Dose-Hgb model will provide further support for the individualized dosing strategy in CKD patients with anemia treated with daprodustat.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":"117 3","pages":"846-856"},"PeriodicalIF":6.3,"publicationDate":"2025-01-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cpt.3544","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143057412","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Toward Developing Alternative Opioid Antagonists for Treating Community Overdose: A Model-Based Evaluation of Important Pharmacological Attributes
IF 6.3 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-01-28 DOI: 10.1002/cpt.3527
Anik Chaturbedi, John Mann, Shilpa Chakravartula, Bradlee Thrasher, Ghazal Arabidarrehdor, Joel Zirkle, Hamed Meshkin, Srikanth C. Nallani, Jeffry Florian, Zhihua Li

In response to increased illicit use of synthetic opioids, various μ-receptor antagonist formulations, with varied pharmacological characteristics, have been and are being developed. To understand how pharmacologic characteristics such as absorption rate and clearance rate affect reversal in treating community opioid overdose, we used our previously published translational opioid model. We adapted this model with in vitro receptor binding data and clinical pharmacokinetic data of three intranasal nalmefene formulations along with an intranasal naloxone formulation to study the reversal of fentanyl and carfentanil-induced respiratory depression in chronic opioid users. Nalmefene has a longer plasma half-life and slower unbinding from the μ-receptor compared to naloxone. For a more rapid reversal of acute overdose-induced respiratory depression, a fast-absorbing antagonist formulation may be of greater utility than a slow-absorbing one containing the same dosage of the antagonist. For preventing renarcotization caused by a long opioid exposure, a slow-clearing antagonist with slow unbinding from the receptor may be of value. While a more potent antagonist with a longer half-life may have the potential to facilitate recovery from respiratory depression for overdose with synthetic opioids, such interventions may also lead to longer and more pronounced withdrawal. This emphasizes the need for a nuanced consideration of several facets while choosing a μ-receptor antagonist, dose, and formulation to treat community opioid overdose cases.

{"title":"Toward Developing Alternative Opioid Antagonists for Treating Community Overdose: A Model-Based Evaluation of Important Pharmacological Attributes","authors":"Anik Chaturbedi,&nbsp;John Mann,&nbsp;Shilpa Chakravartula,&nbsp;Bradlee Thrasher,&nbsp;Ghazal Arabidarrehdor,&nbsp;Joel Zirkle,&nbsp;Hamed Meshkin,&nbsp;Srikanth C. Nallani,&nbsp;Jeffry Florian,&nbsp;Zhihua Li","doi":"10.1002/cpt.3527","DOIUrl":"10.1002/cpt.3527","url":null,"abstract":"<p>In response to increased illicit use of synthetic opioids, various μ-receptor antagonist formulations, with varied pharmacological characteristics, have been and are being developed. To understand how pharmacologic characteristics such as absorption rate and clearance rate affect reversal in treating community opioid overdose, we used our previously published translational opioid model. We adapted this model with <i>in vitro</i> receptor binding data and clinical pharmacokinetic data of three intranasal nalmefene formulations along with an intranasal naloxone formulation to study the reversal of fentanyl and carfentanil-induced respiratory depression in chronic opioid users. Nalmefene has a longer plasma half-life and slower unbinding from the μ-receptor compared to naloxone. For a more rapid reversal of acute overdose-induced respiratory depression, a fast-absorbing antagonist formulation may be of greater utility than a slow-absorbing one containing the same dosage of the antagonist. For preventing renarcotization caused by a long opioid exposure, a slow-clearing antagonist with slow unbinding from the receptor may be of value. While a more potent antagonist with a longer half-life may have the potential to facilitate recovery from respiratory depression for overdose with synthetic opioids, such interventions may also lead to longer and more pronounced withdrawal. This emphasizes the need for a nuanced consideration of several facets while choosing a μ-receptor antagonist, dose, and formulation to treat community opioid overdose cases.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":"117 3","pages":"836-845"},"PeriodicalIF":6.3,"publicationDate":"2025-01-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143057417","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Investigation of Genomic and Transcriptomic Risk Factors of Clopidogrel Response in African Americans
IF 6.3 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-01-27 DOI: 10.1002/cpt.3552
Guang Yang, Cristina Alarcon, Catherine Chanfreau, Norman H. Lee, Paula Friedman, Edith Nutescu, Matthew Tuck, Travis O'Brien, Li Gong, Teri E. Klein, Kyong-Mi Chang, Philip S. Tsao, David O. Meltzer, Julie A. Lynch, Million Veteran Program, Sony Tuteja, Minoli A. Perera

Clopidogrel, an anti-platelet drug, is used to prevent thrombosis after percutaneous coronary intervention. Clopidogrel resistance results in recurring ischemic events, with African Americans (AA) suffering disproportionately. The aim of this study was to discover novel biomarkers of clopidogrel resistance in African Americans using genome and transcriptome data. We conducted a genome-wide association study (GWAS), including local ancestry adjustment, in 141 AA on clopidogrel to identify genetic associations with high on-treatment platelet reactivity (HTPR), with validation of genome-wide significant and suggestive loci in an independent cohort of AA clopidogrel patients (N = 823) from the Million Veteran's Program (MVP) along with in vitro functional analysis. We performed differential gene expression (DGE) analysis in whole blood to identify transcriptomic predictors of response, followed by functional validation in MEG-01 cells. GWAS identified one signal on Chromosome 7 as significantly associated with increasing risk of HTPR. The lead single-nucleotide polymorphism (SNP), rs7807369, within thrombospondin 7A (THSD7A) was associated with an increased risk of HTPR (odds ratio (OR) = 4.02, P = 4.56 × 10−9). Higher THSD7A gene expression was associated with HTPR in an independent cohort of clopidogrel-treated patients (P = 0.004) and carrying a risk allele showed increased gene expression in primary human endothelial cells. Notably, the CYP2C19*2 variants showed no association with clopidogrel response in the discovery or MVP cohorts. DGE analysis identified an association with decreased LAIR1 and AP3B2 expression to HTPR. LAIR1 knockdown in MEG-01 cells resulted in increased expression of SYK and AKT1, suggesting an inhibitory role of LAIR1 in the Glycoprotein VI pathway. In summary, these findings suggest that other variants and genes outside of CYP2C19 star alleles play an important role in clopidogrel response in AA.

氯吡格雷是一种抗血小板药物,用于预防经皮冠状动脉介入治疗后的血栓形成。氯吡格雷耐药性会导致反复发生缺血性事件,而非裔美国人(AA)所受的影响尤为严重。本研究旨在利用基因组和转录组数据发现非裔美国人对氯吡格雷耐药的新型生物标志物。我们在 141 名服用氯吡格雷的 AA 患者中进行了全基因组关联研究(GWAS),包括当地血统调整,以确定与治疗中血小板高反应性(HTPR)的遗传关联,并在来自 "百万退伍军人计划"(MVP)的独立 AA 氯吡格雷患者队列(N = 823)中验证了全基因组显著和提示性位点,同时进行了体外功能分析。我们在全血中进行了差异基因表达 (DGE) 分析,以确定反应的转录组预测因子,然后在 MEG-01 细胞中进行了功能验证。GWAS 确定了 7 号染色体上的一个信号与 HTPR 风险的增加显著相关。单核苷酸多态性(SNP)rs7807369 与 HTPR 风险增加有关(比值比 (OR) = 4.02,P = 4.56 × 10-9)。在一个独立的氯吡格雷治疗患者队列中,较高的 THSD7A 基因表达与 HTPR 相关(P = 0.004),携带风险等位基因的患者在原代人类内皮细胞中的基因表达增加。值得注意的是,在发现队列或 MVP 队列中,CYP2C19*2 变异与氯吡格雷反应无关。DGE 分析发现,LAIR1 和 AP3B2 的表达减少与 HTPR 有关。在 MEG-01 细胞中敲除 LAIR1 会导致 SYK 和 AKT1 表达增加,这表明 LAIR1 在糖蛋白 VI 通路中起抑制作用。总之,这些研究结果表明,CYP2C19星形等位基因以外的其他变体和基因在AA患者对氯吡格雷的反应中起着重要作用。
{"title":"Investigation of Genomic and Transcriptomic Risk Factors of Clopidogrel Response in African Americans","authors":"Guang Yang,&nbsp;Cristina Alarcon,&nbsp;Catherine Chanfreau,&nbsp;Norman H. Lee,&nbsp;Paula Friedman,&nbsp;Edith Nutescu,&nbsp;Matthew Tuck,&nbsp;Travis O'Brien,&nbsp;Li Gong,&nbsp;Teri E. Klein,&nbsp;Kyong-Mi Chang,&nbsp;Philip S. Tsao,&nbsp;David O. Meltzer,&nbsp;Julie A. Lynch,&nbsp;Million Veteran Program,&nbsp;Sony Tuteja,&nbsp;Minoli A. Perera","doi":"10.1002/cpt.3552","DOIUrl":"10.1002/cpt.3552","url":null,"abstract":"<p>Clopidogrel, an anti-platelet drug, is used to prevent thrombosis after percutaneous coronary intervention. Clopidogrel resistance results in recurring ischemic events, with African Americans (AA) suffering disproportionately. The aim of this study was to discover novel biomarkers of clopidogrel resistance in African Americans using genome and transcriptome data. We conducted a genome-wide association study (GWAS), including local ancestry adjustment, in 141 AA on clopidogrel to identify genetic associations with high on-treatment platelet reactivity (HTPR), with validation of genome-wide significant and suggestive loci in an independent cohort of AA clopidogrel patients (<i>N</i> = 823) from the Million Veteran's Program (MVP) along with <i>in vitro</i> functional analysis. We performed differential gene expression (DGE) analysis in whole blood to identify transcriptomic predictors of response, followed by functional validation in MEG-01 cells. GWAS identified one signal on Chromosome 7 as significantly associated with increasing risk of HTPR. The lead single-nucleotide polymorphism (SNP), rs7807369, within thrombospondin 7A (<i>THSD7A</i>) was associated with an increased risk of HTPR (odds ratio (OR) = 4.02, <i>P</i> = 4.56 × 10<sup>−9</sup>). Higher <i>THSD7A</i> gene expression was associated with HTPR in an independent cohort of clopidogrel-treated patients (<i>P</i> = 0.004) and carrying a risk allele showed increased gene expression in primary human endothelial cells. Notably, the <i>CYP2C19*2</i> variants showed no association with clopidogrel response in the discovery or MVP cohorts. DGE analysis identified an association with decreased <i>LAIR1</i> and <i>AP3B2</i> expression to HTPR. <i>LAIR1</i> knockdown in MEG-01 cells resulted in increased expression of SYK and AKT1, suggesting an inhibitory role of <i>LAIR1</i> in the Glycoprotein VI pathway. In summary, these findings suggest that other variants and genes outside of <i>CYP2C19</i> star alleles play an important role in clopidogrel response in AA.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":"117 5","pages":"1313-1324"},"PeriodicalIF":6.3,"publicationDate":"2025-01-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cpt.3552","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143045107","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Clinical Pharmacology & Therapeutics
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1